Archives

Normal and Abnormal Puberty

ABSTRACT

 

Puberty is a biological process that represents the development of secondary sexual characteristics and attainment of reproductive capacity, influenced by genetic, metabolic, environmental, ethnic, geographic, and economic factors. Pubertal onset is characterized by the increased kisspeptin and neurokinin B secretion leading to re-emergence of pulsatile gonadotropin releasing hormone signaling from the hypothalamus which stimulates increased pituitary secretion of luteinizing hormone and follicle stimulating hormone, which in turn stimulate gonadal sex hormone production. Precocious puberty refers to secondary sexual development occurring earlier than the lower end of normal age and delayed puberty refers to secondary sexual development occurring later than the upper end of normal age for the onset of puberty. These changes may represent a serious underlying condition or signify a common variation of normal for which treatment may not be necessary. Clinical evaluation should include a detailed history and physical examination, including anthropometric measurements, calculation of linear growth velocity, and evaluation of secondary sexual characteristics. This chapter summarizes the physiology of pubertal development, variations in pubertal development, and recent developments regarding human puberty.

 

INTRODUCTION

 

Puberty is the process through which reproductive competence is achieved (1). Physical characteristics associated with this process include the development of secondary sex characteristics, acceleration in linear growth velocity, and the occurrence of menarche in women and spermatogenesis in men. The sex chromosome karyotype of the embryo, XX or XY, determines the trajectory for differentiation of the gonads and development of the internal and external genital structures. This complex process, beginning in utero, depends on neuroendocrine signaling and gonadal components. Ultimately, integrated communication between the reproductive and metabolic systems is critical for timely pubertal development (2).

 

Pubertal development and neuroendocrine system maturation involve the ontogeny, activity, and interactions of the gonadotropin releasing hormone (GnRH) neurons. The onset of puberty is accompanied by increased kisspeptin and neurokinin B secretion causing the GnRH neurons to secrete GnRH in a pulsatile manner. Increased GnRH secretion stimulates pulsatile pituitary luteinizing hormone (LH) and follicle stimulating hormone (FSH) secretion (3). LH and FSH stimulate gonadal sex steroid secretion which promotes development of secondary sex characteristics and influences hypothalamic-pituitary function via negative feedback inhibition. This chapter summarizes the physiology of pubertal development, variations in pubertal development, and recent developments regarding human puberty.

 

CLINICAL FEATURES OF NORMAL PUBERTAL DEVELOPMENT

 

Children typically demonstrate a predictable sequence of physical changes during pubertal maturation. Within the chronologic age ranges for pubertal development, individual variations regarding age at onset and tempo of pubertal development are expected.

 

In humans, two physiological processes, gonadarche and adrenarche, govern pubertal transition. Gonadarche reflects the reactivation of the hypothalamic GnRH pulse generator which has been quiescent since late infancy. Increasing pulsatile GnRH secretion stimulates pulsatile gonadotropin secretion which, in turn, stimulates the growth and maturation of the gonads and gonadal sex steroid secretion. Increased estrogen secretion promotes breast development, cornification of the vaginal mucosa, and uterine growth in girls. Increased testosterone secretion promotes penile enlargement. The increased HPG axis activity culminates in folliculogenesis, ovulation, and menses in the female and spermatogenesis in the male.

 

In addition to gonadal sex steroid secretion, humans experience adrenarche signifying adrenal pubertal maturation. Adrenarche typically begins prior to the first visible physical manifestation of gonadarche, breast development, or testicular enlargement. Pubarche, the physical manifestation of adrenarche, is characterized by the development of pubic hair, axillary hair, apocrine odor, and acne. Adrenarche indicates increased adrenal cortical zona reticularis activity and is accompanied by increased secretion of dehydroepiandrosterone sulfate (DHEAS), dehydroepiandrosterone (DHEA), androstenedione, and 11-hydroxyandrostenedione (4, 5). These so-called “adrenal androgens” are C19 steroids which do not bind directly to the androgen receptor and can be peripherally converted to more potent androgens. Circulating concentrations of two adrenal 11-oxyandrogens, 11-hydroxyandrostenedione and 11-ketotestosterone increase with adrenarche. Whereas 11-hydroxyandrostenedione has minimal androgenic activity, 11-ketotestosterone is almost as potent as testosterone. During adrenarche, 11-ketotestosterone appears to be the major bioactive adrenal C19 steroid and may be responsible for the physical changes associated with pubarche (6).

 

Gonadarche and adrenarche are dissociated such that the absence of adrenarche does not prevent gonadarche or the attainment of fertility (7). Curiously, the phenomenon of adrenarche appears to be limited to humans and a few species of non-human primates (8, 9). The factors that drive the dynamic changes within a strictly defined zona reticularis within the adrenal cortex, are still poorly defined. How adrenarche and increased adrenal C19 steroids impact brain development during human adolescence is indeterminate (10). Urinary steroid hormone profiling suggest that adrenarche may be a gradual process that likely begins earlier than previously believed (11).

 

STAGING OF PUBERTY

 

Tanner and colleagues followed the pubertal development of children living in an orphanage in the UK. Their five-stage classification system continues to be commonly utilized for clinical assessments (12, 13, 14). For girls, Tanner staging is used to describe breast and pubic hair development (See Figure 1). For boys, Tanner staging is used to describe testicular volume, penile development, and pubic hair development (See Figure 2). Tanner and his colleagues also described that the tempo of puberty varies between individuals.

 

Figure 1. Tanner Staging for pubertal development in girls. In girls, breast development is rated from 1 (preadolescent) to 5 (mature), and stage 2 (appearance of the breast bud) marks the onset of pubertal development. Pubic hair stages are rated from 1 (preadolescent, no pubic hair) to 5 (adult), and stage 2. Although pubic hair and genital or breast development are represented as synchronous in the illustration, they do not necessarily track together and should be scored separately. Figure 1 Adapted with permission from Carel JC, Léger J. Clinical practice. Precocious puberty. N Engl J Med. 2008;358(22):2367 and Klein DA, Emerick JE, Sylvester JE, Vogt KS. Disorders of Puberty: An Approach to Diagnosis and Management. Am Fam Physician. 2017 Nov 1;96(9):590-599. PMID: 29094880

Figure 2. Tanner Staging for pubertal development in boys. In boys, genital development is rated from 1 (preadolescent) to 5 (adult); stage 2 marks the onset of pubertal development and is characterized by an enlargement of the scrotum and testis and by a change in the texture and a reddening of the scrotal skin. Pubic hair stages are rated from 1 (preadolescent, no pubic hair) to 5 (adult), and stage 2 marks the onset of pubic hair development. Although pubic hair and genital or breast development are represented as synchronous in the illustration, they do not necessarily track together and should be scored separately. In normal boys, stage 2 pubic hair develops at an average of 12 to 20 months after stage 2 genital development. Figure 2 Adapted with permission from Carel JC, Léger J. Clinical practice. Precocious puberty. N Engl J Med. 2008;358(22):2367 and Klein DA, Emerick JE, Sylvester JE, Vogt KS. Disorders of Puberty: An Approach to Diagnosis and Management. Am Fam Physician. 2017 Nov 1;96(9):590-599. PMID: 29094880

 

Girls

 

The typical first clinical sign of puberty in girls is the appearance of breast tissue with elevation of the breast and papilla; this is considered to be Tanner Stage 2 (Figure 1). Initially, breast development (thelarche) may be unilateral. Many girls complain of mild breast tenderness or discomfort during this stage that subsequently resolves. Tanner stage 3 breast development is considered to be additional enlargement of the breast and areola. During Tanner stage 4, the papilla forms a secondary mound above the breast; this stage is often very rapid. Tanner stage 5 represents mature breast development due to recession of the areola to the contour of the breast. Palpation of the breast is obligatory to differentiate breast tissue from lipomastia. In children with obesity without breast development, a palpable depression beneath the nipple in the center of the areola when examined in the supine position gives the impression of a donut and is referred to as the ‘donut’ sign.  Breast ultrasound correlates reasonably well with Tanner staging by palpation and can detect breast development slightly earlier than physical exam (15). In most instances, breast development is evident before pubic hair development. Typically, the pubertal growth spurt in girls occurs concurrently with the onset of breast development with only 4-6 cm of linear growth occurring after menarche, however this may be variable.

 

The appearance of sexual hair including pubic hair (pubarche) signifies the onset of adrenarche. In girls, Tanner stage 2 pubic hair is characterized by sparse, coarse, lightly pigmented hairs along the labia majora. For Tanner stage 3, pubic hair becomes progressively darker, coarser, and spreads over the mons pubis. For Tanner stage 4, pubic hair continues to extend to become an inverse triangle, with spread to the medial aspects of the thighs being considered Tanner stage 5.

 

With the onset of ovarian estrogen secretion, the vaginal mucosa changes from shiny bright red to pale pink appearance due to cornification of the vaginal mucosa. Increased estrogen secretion promotes uterine growth and causes physiologic leukorrhea, a thin, white, non-foul-smelling vaginal discharge that typically begins 6 to 12 months before menarche. Menarche occurs, on average, 2 to 2.5 years after the onset of breast development (See figure 3A). During the first-year post-menarche, menses are usually irregular and anovulatory. These early years are characterized by inconsistent ovulation and varying lengths of follicular and luteal phases. Ultimately, coordinated maturation of the hypothalamic, pituitary, and ovarian components occurs culminating in cyclic monthly ovulation (16). Although full HPG axis maturation generally occurs over several years, by three years post-menarche, most cycles are between 21-35 days.

 

Figure 3A and 3B. Average ages and sequence of pubertal development. Panel A: girls; Panel B: boys.

 

Boys

 

For boys, increased testicular volume is the first physical finding indicating onset of gonadarche (See Figure 2 and Figure 3B). Palpation of the testes and use of a Prader orchidometer is essential for accurate assessment. A Prader orchidometer is a collection of 3-D ellipsoids ranging in volume from 1 to 25 mL (See Figure 4). During gonadarche, testicular volume increases, and the penis increases in length and diameter. Flaccid penile length can be measured using a straight edge on the dorsal surface from the pubic ramus to the tip of the glans while compressing the suprapubic fat pad and applying gentle traction to stretch to penis.

 

Figure 4. Prader Orchidometer.

 

Increased testicular volume represents Sertoli cell proliferation, differentiation, and eventually, the initiation of spermatogenesis. The onset of puberty is defined as a testicular volume > 4 ml and a testicular length > 2.5 cm. The volume of mature human testis is approximately 20-25 ml. Spermatozoa (spermaturia) can be found in early morning urine samples beginning during genital stage 3 (16). Nocturnal sperm emissions may also begin around this time.

 

For boys, Tanner stage 2 pubic hair consists of downy hairs at the base of the penis. During pubic hair stage 3, the hair becomes longer, darker, and extends over the junction of the pubic bones. For pubic hair stage 4, the extent of hair has increased, but has not yet achieved the adult male escutcheon with spread to the medial aspects of the thighs that would be considered Tanner stage 5. Additional features include axillary hair, increased size of the larynx, voice break with deepening of the voice, increased bone mass, and increased muscle strength. Terminal hair develops in androgen-dependent regions on the face and trunk approximately three years after appearance of pubic hair. The distribution and density of beard, chest, abdominal, and back hair varies among individuals.

 

Peak height velocity is both age and sex-dependent. It occurs earlier in girls, between Tanner breast stages 2 and 3, and later in boys, between Tanner testis stages 3 and 4.

 

Approximately 50% of boys experience pubertal gynecomastia (17). Typically, pubertal gynecomastia is transient and most prominent in mid-puberty when the ratio of circulating estradiol to testosterone concentrations is relatively higher.

 

DISCOVERY OF THE HYPOTHALAMIC-PITUITARY-GONADAL (HPG) AXIS

 

Since ancient times, it was known that castration of animals and humans interfered with development of secondary sex characteristics and fertility (14). In 1935, Ernst Laquer and colleagues isolated testosterone from several tons of steer testes (18). Later that year, Adolf Butenandt, Gunter Hanisch, Leopold Ruzicka, and A. Wettstein published the chemical synthesis of testosterone (19, 20). After showing that follicular fluid obtained from a sow ovary was able to induce cornification of vaginal mucosa, Edgar Allen and Edward Doisy isolated the active substance, estrone (21). Donald MacCorquodale, Stanley Thayer, and Edward Doisy isolated estradiol from 8000 pounds of sow ovaries in 1935 (22). Philip Smith, Bernhard Zondek, Hermann Zondek, H.L. Fevold and colleagues, and Geoffrey Harris established the functional relationships involved in HPG axis function (23, 24, 25, 26). Roger Guillemin and Andrew Schally engaged in a vigorous competition to identify hypothalamic releasing hormones including GnRH (27, 28, 29). Ernst Knobil and his colleagues identified that pulsatile GnRH secretion was essential for sustained pituitary gonadotropin secretion (28, 30). Fred Karsch and Ernst Knobil independently developed the concept of the “GnRH pulse generator” (31). In the 1970s, Melvin Grumbach and colleagues measured circulating gonadotropin concentrations in the human fetus (32). Around the same time, Charles Faiman and Jeremy Winter also reported gonadotropin concentrations in normal and agonadal children (33). Their collective findings led to recognition of early postnatal HPG axis activity followed by quiescence of the HPG axis during childhood until resumption of GnRH pulse generator activity at the onset of puberty.

 

Ontogeny of GnRH Neurons

 

Reproductive competence depends on the meticulous development of the GnRH neuron system. In the human fetus, GnRH neurons initially develop in the olfactory placode outside the central nervous system. The olfactory placodes invaginate at approximately 39 days of gestation in the human. Based on the appearance of immunoreactive GnRH protein, the GnRH neuron specification occurs between 39-44 days of gestation (34). The developing GnRH neurons are associated with the embryonic vomeronasal organ. Available data suggest that the GnRH neuron precursors are distinct from those giving rise to the vomeronasal neurons (35).

 

Subsequently, the GnRH neurons migrate accompanied by olfactory-derived axons, olfactory epithelial sheath cells, and blood vessels towards the cribriform plate (36). Migration of the GnRH neurons seems to pause at the nasal/forebrain junction prior to crossing the cribriform plate (37). During this “pause” phase, multiple tissues, chemokines, growth factors, and neurotransmitters appear to form gradients influencing movement of GnRH neurons. Upon reaching the hypothalamus, the GnRH neurons disperse to their final locations sending projections to the median eminence to release GnRH into the hypophyseal portal vasculature.

 

The precise origin and particular factors responsible for the specification and differentiation of GnRH neuron precursors remain enigmatic. Inaccessibility of developing human GnRH neurons has led to development of alternative approaches to elucidate the history of GnRH neurons. One approach has involved a protocol to generate GnRH neurons from human pluripotent stem cells (38). With this approach, Yellapragada et al. demonstrated that dose- and time-dependent FGF8 signaling via FGFR1 is indispensable for human GnRH neuron ontogeny (39). Using a differentiation trajectory analysis approach, DLX family of transcription factors have been reported to promote in vitro human GnRH neuron differentiation (40).

 

Components of the HPG Axis

 

Gonadotropin-releasing hormone is a decapeptide (pGlu-His-Trp-Ser-Trp-Gly-Leu-Arg-Pro-Gly-NH2) derived from a 92-amino acid precursor, preproGnRH, that was characterized in 1984 (41). LH and FSH are synthesized in the same gonadotroph cell located in the anterior pituitary. LH and FSH are glycoproteins consisting of two subunits. The alpha subunits are identical whereas the beta subunits confer hormone specificity. Each GnRH pulse stimulates an LH pulse.

 

During human gestation, human chorionic gonadotropin (hCG) drives fetal testicular testosterone secretion in the developing male fetus early during gestation. The pituitary gland begins to secrete gonadotropins with LH and FSH becoming detectable in fetal blood after 14 weeks of gestation (42, 43). Initially, pituitary gonadotropin secretion appears to be GnRH-independent with progressive transition to kisspeptin-GnRH regulation of pituitary gonadotropin secretion during the third trimester (44). Peak gonadotropin concentrations occur around the midpoint of gestation followed by a progressive decline attributed to suppression by placental estrogens (45). In the male fetus, testicular testosterone secretion is essential for normal development of internal and external male genital structures. Comparatively, the fetal ovary is quiescent.

 

As noted above, GnRH stimulates pituitary LH and FSH secretion. LH and FSH signal through their cognate receptors which are G-protein coupled receptors (46).

 

GONADS

 

The gonads synthesize sex steroids from cholesterol. In the testis, LH acting through the LH receptor stimulates conversion of cholesterol to testosterone in the Leydig cell. In specific target tissues such as external genital skin and the prostate, testosterone is converted to dihydrotestosterone by the enzyme, 5α-reductase type 2 encoded by the SRD5A2gene. Testosterone influences pituitary LH secretion through negative feedback either via direct actions or indirectly after conversion to estradiol. FSH acting through the FSH receptor promotes growth of seminiferous tubules and supports sperm development. Growth of the seminiferous tubules and increasing numbers of germ cells accounts for increasing testicular volume during puberty.

 

In females, the two cell-two gonadotropin model applies to ovarian steroidogenesis. LH stimulates the theca cell to synthesize androstenedione which diffuses to the granulosa cell where FSH-stimulated aromatase activity stimulates estradiol synthesis. Estradiol has both negative feedback and positive feedback. Estradiol mediated positive feedback is required to elicit the LH surge responsible for ovulation.

 

Activin and inhibin are heterodimeric glycoproteins secreted by the gonads. Inhibins consist of an alpha subunit and one of two homologous yet distinct beta subunits, βA or βB. Inhibin B is composed of an alpha subunit and a βB subunit whereas inhibin A consists of an alpha subunit and a βA subunit. Inhibins are secreted by Sertoli cells in the testes and granulosa cells in the ovary. Inhibin B influences pituitary FSH secretion by negative feedback. In prepubertal boys, inhibin B concentrations reflect Sertoli cell mass and function. After puberty, inhibin B concentrations reflect spermatogenesis (47). Inhibin B correlates inversely with FSH levels in adult men. Activins are dimers of inhibin β subunits, βA, βB and βC; the best characterized are activin A (βAβA) and activin B (βBβB). Activin A stimulates pituitary FSH secretion(48, 49). Follistatin is a monomeric protein that modulates activin activity and can irreversibly inhibit activin activity.

 

Leydig cells secrete insulin-like peptide 3 (INSL3), a small peptide that, in utero, acts through the relaxin-like family peptide receptor 2 (RXFP2) to promote trans-abdominal testicular descent. INSL3 concentrations increase in boys during puberty (50).

 

HYPOTHALAMUS

 

The hypothalamus serves as the rheostat for many physiological functions especially reproduction and growth. The adult human hypothalamus contains approximately 2000 GnRH neurons with cell bodies diffusely distributed in a rostro-caudal continuum (34). The GnRH neurons send projections to the median eminence that terminate in close association with the capillaries of the primary plexus of the hypophyseal portal circulation. Synchronized activity of the GnRH neurons leads to episodic GnRH release into the median eminence with consequent pulsatile pituitary gonadotropin secretion.

 

An extrinsic hypothalamic neuronal network, known as the GnRH pulse generator, governs GnRH neuron function. This network is located within the infundibular nucleus (known as the arcuate nucleus in non-human species). In the human, the GnRH pulse generator is responsible for tonic gonadotropin secretion; pulsatile LH and FSH secretion regulate testicular function in men and modulate ovarian function, especially folliculogenesis in women. In women, the developing follicle secretes increasing amounts of estradiol ultimately triggering an LH surge followed by ovulation. In adult men, pulse frequency is relatively constant at approximately one pulse every 90-120 minutes. Among women, pulse frequency varies across the menstrual cycle from approximately one pulse per hour during the follicular phase and one pulse every 180 minutes during the luteal phase.

 

Among GnRH deficient women, pulsatile GnRH administered at a frequency simulating the follicular phase led to ovulatory menstrual cycles (51). In a preclinical model, administration of pulsatile GnRH to prepubertal rhesus female monkeys initiated pubertal development including ovulatory menstrual cycles (52). Thus, puberty in girls and boys is entirely dependent on resumption of pulsatile GnRH release.

 

Although the GnRH pulse generator was conceptualized by Fred Karsch and Ernst Knobil, the anatomic location of the pulse generator was indeterminant. Identification of loss of function variants in the kisspeptin receptor (KISS1R) gene in patients with congenital hypogonadotropic hypogonadism launched the investigations establishing kisspeptin, neurokinin B, dynorphin, and their cognate receptors as major components of the pulse generator (53, 54). Kisspeptin signals through its receptor, KISS1R, expressed on GnRH cells. Neurokinin B is a decapeptide encoded by the TAC3 (Tac2 in rodents) gene and its cognate receptor encoded by NK3R gene. Both the kisspeptin and neurokinin B receptors are G-protein coupled receptors. Dynorphin is an opioid peptide that signals through a kappa-opioid receptor which is also a G-protein coupled receptor.

 

Due to the inaccessibility of human brain, especially the pubertal brain, the contemporary model of the GnRH pulse generator has been delineated by preclinical studies performed in rodents, sheep, and non-human primates (55). This model predicts that reciprocal interactions within a network of kisspeptin neurons in the infundibular nucleus leads to synchronous intermittent activation transmitted to GnRH neurons by kisspeptin fibers that project to the median eminence. These kisspeptin fibers are closely associated with GnRH projections targeting the portal capillaries (56).

 

Based on the detection of kisspeptin, neurokinin B, and dynorphin in the arcuate kisspeptin neurons of mice and sheep, these neurons have been labeled as KNDy neurons (57). Preclinical data suggest that KNDy neurons serve as the intrinsic GnRH pulse generator (58). Kisspeptin and neurokinin B stimulate GnRH release whereas dynorphin appears to be inhibitory. Coordinated interactions of these neuropeptides within the arcuate kisspeptin neuronal network are ostensibly central to the neurobiology of the GnRH pulse generator resulting in pulsatile kisspeptin output. However, the applicability of these findings to human biology remains to be confirmed.

 

In humans, the HPG axis is active during gestation and the early neonatal period followed by the quiescent years of childhood until the onset of puberty occurs. This pattern suggests that diverse mechanisms integrate the hierarchical activation and deactivation of various stimulatory and inhibitory neuronal pathways ultimately regulating pubertal onset and progression towards reproductive maturity. Thus, a central inhibition of the axis occurs during childhood. For puberty to occur, increased expression of the key factors, KISS1, NKB3, and GnRH, must begin along with decreased expression of the various inhibitory factors. In other words, during the pubertal transition, the balance between inhibitory and stimulatory factors shifts to favor the re-activation of the HPG axis, onset of pubertal changes, and reproductive competence.

 

Identifying the proximate factors and specific interactions responsible for the on-off-on pattern of HPG axis activity in humans has been a longstanding enigma. Starting with clinical findings, the availability of more sophisticated tools and preclinical models have begun to identify pieces of the puzzle to elucidate the fine details of HPG axis functioning. One factor involved in the suppression of puberty was identified in families with paternally inherited GnRH-dependent/central precocious puberty (CPP). Exome sequencing analyses in multiple families with CPP identified loss of function variants in the makorin 3 (MKRN3) gene (59). This gene, mapped to the Prader Willi region at chromosome 15q11.2, is exclusively expressed from the paternal allele. Consistent with the hypothesis that MKRN3 suppresses the GnRH pulse generator, circulating MKRN3 concentrations decline during puberty (60, 61, 62). 

 

The MKRN3 protein is an E3 ubiquitin ligase consisting of 507 amino acids. It is expressed in KNDy neurons. The protein has five zinc finger domains. Regarding its function, the protein can ubiquitinate substrates and can undergo auto-ubiquitination (63). MKRN3 ubiquitinates methyl-CpG-DNA binding protein 3 (MBP3) interfering with GnRH1transcription (64). Available preclinical data suggest that MKRN3 functions as a brake on neuronal GnRH release (65). One potential factor influencing MKRN3 expression is microRNA (miRNA) miR-30. Using a rat model, hypothalamic miR-30 expression increased while Mkrn3 expression decreased during puberty. In addition, treatment with agents that interfered with the binding of miR-30 to Mkrn3 were associated with delayed puberty in female rats (66). Using proteomics, MKRN3 targets include insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) and several members of the polyadenylate-binding protein family (67). The decline of hypothalamic Mkrn3 expression in mice and serum MKRN3 protein levels in females prior to the onset of puberty support the hypothesis that MKRN3 suppresses pubertal initiation possibly through effects on prepubertal hypothalamic development and plasticity (61, 67)

 

Preclinical studies have provided persuasive evidence regarding the regulatory role of epigenetic modifications in pubertal maturation. Epigenetics refers to changes in gene expression and/or activity independent of changes in the primary nucleotide sequence (68). Epigenetic changes include DNA modifications such as methylation/demethylation and histone post-translational modifications such as acetylation/deacetylation. Other post-translational protein modifications such as ubiquitination may also influence protein function. Ubiquitination involves the transfer of ubiquitin to a protein altering its function typically by interfering with protein actions or by promoting protein degradation. As noted above, the MKRN3 protein can function as a ubiquitin ligase. Noncoding RNAs such as miRNAs provide yet another regulatory mechanism.

 

Another example of epigenetic regulation of pubertal maturation involves two mutually antagonistic histone methylating complexes, the Poly-comb and Trithorax groups. The Poly-comb group represses gene transcription while the Trithorax group appears to function as a gene activator. Preclinical studies performed in rats showed that the Poly-comb group effectively silenced Kiss1 expression until the onset of puberty when increased methylation of the Eed and Cbx7 genes occurred leading to decreased Eed and Cbx7 expression and increased Kiss1 expression (69). Recruitment of the Trithorax activity group further enhanced.  Kiss1 expression (70, 71). Genome wide association studies have implicated zinc finger (ZNF) genes. In nonhuman primates, expression of two ZNFs, GATAD1 and ZNF573, decreases upon pubertal reactivation of the GnRH pulse generator (71).

 

Clinically, it has long been recognized that extremes of body energy status such as chronic malnutrition or severe obesity influence the HPG axis especially in girls and women. The hypothalamic kisspeptin neurons integrate various peripheral and central metabolic signals reflecting energy intake, energy expenditure, and environmental circumstances. Signal coordination between reproductive and metabolic neurons can be direct or indirect. For example, leptin does not directly regulate kisspeptin neurons yet acts as a permissive factor for the onset of puberty (72). Cellular energy and metabolic sensors include mammalian target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), and sirtuin 1 (SIRT1) (73). Depending on energy status, mTOR and AMPK promote or repress puberty, respectively, by activating or inhibiting Kiss 1 neurons in the arcuate nucleus. Other factors such as melanocortin and agouti-related peptides also interact with kisspeptin pathway (74). In the hypothalamus, neuronal nitric oxide (NO) appears to act on GnRH neurons to integrate metabolic and gonadal information (75, 76). Detailed reviews regarding the neurobiology of the GnRH pulse generator are beyond the scope of this chapter and are available elsewhere (77, 78, 79, 80, 81, 82).

 

MINI-PUBERTY

 

Facilitated by the availability of more sensitive hormone assays, Forest and her colleagues described a transient period of increased HPG axis activity in early infancy (83, 84). Following the low gonadotropin concentrations at birth, gonadotropin concentrations were found to rise in both boys and girls within weeks of birth (85). This period of transient gonadotropin secretion has been designated as “minipuberty”. Gonadotropin concentrations in the immediate neonatal period are likely low due to in-utero suppression by placental estrogen. With removal of the placental estrogen suppression, the HPG axis is functional. Relevantly, physical findings typical of pubertal sex steroid secretion are absent with the rare exception of vaginal bleeding attributed to decreased exposure to placental estrogen.

 

Over the first few years of life, sexual dimorphism in gonadotropin concentrations occurs (86) Boys have higher LH concentrations which peak between 2-10 weeks of age and decline by 4-6 months of age. Girls have higher FSH concentrations which may remain elevated until 2-4 years of age.

 

In boys, LH stimulates testicular testosterone secretion with testosterone concentrations typically peaking around 1 month of age followed by a decline to prepubertal concentrations by 7-12 months of age. During this phase, the number of germ cells and Sertoli cells increase and penile size increases (87, 88). The proliferation of Sertoli cells leads to a transient increase in testicular volume (89). Sertoli cells secrete Anti-Mullerian Hormone (AMH) and inhibin B. Since Sertoli cells do not express androgen receptors during this stage, spermatogenesis does not occur and AMH secretion remains high (90, 91). A temporary increase in the number of Leydig cells also occurs, but subsequent fetal Leydig cell apoptosis reduces fetal Leydig cell number (92). Longitudinal data obtained from healthy boys suggests a temporal dissociation of Leydig and Sertoli cell activity during minipuberty (93). These data suggest that single blood sample may be insufficient to assess HPG axis activity during early infancy and that obtaining several consecutive samples may be more informative. Curiously, gonadotropin and testosterone concentrations are higher among preterm boys. In addition, increases in testicular volume and penile length are greater in preterm boys compared to full term boys essentially enabling catch-up for testicular volume and penile length (94). Some small studies have documented an exaggerated physiologic hormonal response in extremely premature infants (95).

 

In girls, the gonadotropins promote granulosa cell proliferation and ovarian estrogen and AMH secretion (96). As would be anticipated, AMH concentrations remain much lower in girls compared to boys (97). A longitudinal study involving healthy full-term infant girls demonstrated two gonadotropin peaks in early infancy with one peak occurring around days 15 to 27 and a later peak occurring at days 164-165 (98). Again, collecting several consecutive samples may be more informative than a single blood sample to assess for minipuberty in infancy.

 

This transient time period of an active HPG axis, provides an opportunity to diagnose individuals with differences/variants of sex development (DSD/VSD). In a series including both healthy infants and infants with DSD, testosterone measured by LC-MS/MS, AMH concentration, and LH/FSH ratio provided the best discrimination between sexes. The cut-point for LH/FSH ratio was 0.32. Inhibin B and AMH levels were higher in boys with minimal overlap in girls (99). Infants with Turner Syndrome usually have elevated FSH concentrations. Surprisingly, gonadotropin concentrations are typically not elevated in patients with complete androgen insensitivity.

 

This brief interval of HPG axis activity can also help diagnose congenital hypogonadotropic hypogonadism in boys who present with micropenis accompanied by low gonadotropin and testosterone concentrations (100). Testosterone, LH, FSH, AMH, and inhibin B concentrations may provide information regarding the functionality of testicular tissue in infant boys (101).

 

As noted above, the human HPG axis displays an “on-off-on” pattern. The biological basis and rationale for transient post-natal HPG axis activity during the first few months of life are enigmatic. At birth, the brain is still plastic with ongoing development. Most axon and synapse formations are completed during the first year of life. Does this transient HPG axis activity imprint specific areas in the brain? Does minipuberty influence future patterns for female and male reproductive function with cyclic gonadotropin patterns in females and not in males? Are gonadal hormones during infancy able to affect future fertility, gender identity, sexual orientation, behaviors, and risk for autism spectrum dysfunction? Data are accruing regarding patterns of hormone secretion during the first six months of life. However, the factors that initiate and terminate this transient period of HPG axis activity and maintain the quiescence of the HPG axis until the onset of puberty are still unknown.

 

 

Over the past few decades, several studies have observed that puberty is beginning at a younger age. Clinical studies examining ages of the onset of puberty depend on the criteria used to denote puberty. Onset of breast development and age at menarche are the conventional indicators of puberty in girls. Prospective observations and retrospective questioning of parents and young girls through in-person questioning has been used to record age at menarche; shorter recall intervals provide the greatest accuracy regarding the details of menarche (102, 103). For boys, age at voice change has been used as a surrogate marker because accurate ascertainment of pubertal onset in boys requires testicular exams using an orchidometer, thus, effectively excluding large-scale epidemiologic clinical studies (104).

 

During medieval times, available evidence suggests that puberty began around 10-12 years of age. However, the tempo of puberty was slow with menarche occurring closer to 15 years in rural areas and 17 years in London (105). Presumably, undernutrition, increased infections, and greater physical exertion impacted both the timing and tempo of puberty during medieval times (106). The age of menarche declined from 16 to 17 years in the early 19th century to 13 years of age in the late 20th century in Europe and North America. Similarly, the age at menarche has declined in the Yunnan Province in China (107). This decline has been attributed to the improvement in socioeconomic conditions. Currently, the dialogue continues as to whether the trend towards earlier puberty is persisting and, if so, what are the factors driving this process. 

 

Data regarding pubertal milestones in American girls were obtained through the cross-sectional Third National Health and Nutrition Examination Survey (NHANES III) between 1988 and 1994.  Among these American girls, mean ages in years for breast development, pubic hair development, and menarche were 9.5, 9.5, and 12.1 for non-Hispanic black girls; 9.8, 10.3, and 12.2 for Mexican-American girls; and 10.3, 10.5, and 12.7 years for non-Hispanic white girls, respectively (108). In 1997, the Pediatric Research in Office Settings (PROS) study reported earlier onset of thelarche with the caveat that breast palpation was not performed (109). The Copenhagen Puberty Study reported that mean age at breast development was lower in the 2006 cohort compared to the 1991 cohort whereas mean age at menarche was similar in both cohorts. Independent of BMI, gonadotropin concentrations were comparable between these cohorts while estradiol concentrations were lower in the 2006 cohort (110).

 

Beginning in 2004, the Breast Cancer and Environment Research Program (BCERP) prospectively recruited three cohorts of girls aged 6-8 years. This program recruited non-Hispanic white, Hispanic, non-Hispanic black girls, and Asian girls living in New York, Ohio, and California. The overall median age at menarche was 12.25 years with ethnic background median ages as follows: Hispanic girls 11.6 years, black girls at 11.8 years, white girls at 12.5 years, and Asian girls at 12.0 years (111). This cohort differed from the NHANES III study because Hispanic girls experienced menarche earlier than the black girls. These studies, all performed in the United States, report race and ethnicity-related differences in onset of pubertal milestones. Detailed assessment of the potential impact of socio-economic factors was not performed. Notably, differences noted in pubertal timing are smaller than the overall variation among individuals in the population. Most importantly, clinical decision-making should reflect an individual patient's characteristics and family history with less dependence on racial or ethnic backgrounds.

 

Comparable studies from Spain and Greece have also reported earlier onset of breast development and slower pubertal tempo (112, 113). Thus, available data including a systemic review of international studies largely confirm the ongoing trend for earlier breast development with minimal decline in age at menarche (114).

 

Several questions regarding this earlier onset of puberty, predominantly earlier thelarche, need to be considered. Does this earlier breast development reflect earlier resumption of GnRH pulse generator activity, extragonadal estrogen production, or environmental exposures? What, if any, is the relationship of BMI to puberty? Another consideration is that race/ethnicity are socio-political constructs and are not fully representative of biology. While genetic ancestry likely influences the onset of puberty, nutritional factors and environmental exposures play important roles. Hence, should cut-off points based primarily on race/ethnicity continue to be utilized?

 

Based on single unstimulated gonadotropin concentrations, data from the Copenhagen puberty in girls study suggest that gonadotropin concentrations are not obviously increased in girls with early thelarche. Thus, the phenomenon of early thelarche appears to be independent of gonadotropin secretion and may not signify early resumption of GnRH pulse generator activity (115).

 

The possibility that exposure to endocrine-disrupting chemicals (EDCs) can induce early thelarche has been questioned. EDCs are defined as exogenous chemicals that interfere with hormone action. EDCs include phthalates, phenols, phytoestrogens, organochlorine pesticides, polybrominated flame retardants, diphenyl ethers, heavy metals, and perfluorochemicals. In addition to pesticides, these chemicals can be found in common household products such as hair products, soaps, toothpaste, perfumes, plastics, essential oils, and cleaning products (116). Valid assessment of the consequences of EDCs on puberty is problematic because exposure may occur in utero and generally involves a mixture of assorted EDCs with differing half-lives and activities. Differences in the duration and route of the exposure(s), methodology to detect EDCs, and potential sample contamination further confound analyses. One potential example regarding EDCs involved transient past exposure to organochlorine pesticides among internationally adopted girls in Belgium who subsequently developed precocious puberty (117). Animal models suggest that EDCs can affect puberty through epigenetic mechanisms (118). Nevertheless, most data available regarding the consequences of EDCs on human puberty are inconclusive (119).

 

Relationship with BMI

 

Observational data has shown a relationship between BMI and age at puberty in girls (120, 121). The BCERP study found that girls who were overweight or obese at baseline experienced menarche 0.3 years earlier with age at thelarche being inversely correlated with BMI. The BCERP also concluded that BMI had a greater effect than ethnic background on age at menarche (111). Limited data exist regarding the relationship of BMI to pubertal onset in boys. The Puberty Cohort of the Danish National Birth Cohort reported that increased BMI was associated with earlier onset of puberty in boys and girls (122). Among boys, pubertal milestones, testicular enlargement, voice break, and testosterone concentrations showed inverse correlation with BMI (104). Hence, available evidence strongly indicates an inverse relationship between BMI and the onset of puberty in both boys and girls.  

 

Yet, investigating the relationship between puberty and BMI is confounded by potential hormonal and genetic influences (123). Obesity may be associated with hyperinsulinemia and lower sex hormone binding globulin concentrations with consequent higher free sex steroid concentrations. In addition, some genes influence both BMI and pubertal timing (124, 125). The pro-opiomelanocortin (POMC) and central melanocortin systems provide one example of the intricate interrelationships between nutrient signaling and reproductive function. Neurons expressing POMC, producing α-MSH (melanocyte-stimulating hormone), have been suggested to stimulate puberty onset and gonadotropin secretion via modulation of arcuate Kiss1 neurons (126, 127).

 

Genetic Factors

 

Genetic  factors influence pubertal timing as evidenced by twin studies demonstrating > 50% hereditability for menarche (128). Skeletal maturation, age at pubertal growth spurt, and Tanner staging also show greater concordance between monozygotic twins compared to dizygotic twins emphasizing the relevance of genetic variation in the timing of puberty. Thus, 50-80% of variation in the timing of puberty onset may reflect genetic variation (129). Parental self-reports regarding pubertal timing are associated with timing of specific pubertal milestones in offspring of the concordant sex (130, 131). Genome-wide association studies (GWAS) have detected loci associated with age at menarche (132). Some loci appear to be common and independent of ancestry. A large-scale trans-ethnic GWAS, involving 38,546 women of diverse and predominantly non-European ancestry or ethnicity, identified a novel locus in chromosome 10p15 that is associated with early menarche. This region maps to intron 7 of the aldo-keto reductase Family 1, member C4 (AKR1C4) gene, a member of family of enzymes involved in steroid metabolism and action (133).

 

To summarize, the secular trends suggesting an earlier onset of puberty appear to be persistent although the age at menarche appears to be relatively static. Likely contributing factors include the rising prevalence of obesity, exposure to potential EDCs, specific dietary influences, and decreased physical activity.

 

VARIATIONS IN PUBERTAL DEVELOPMENT

 

Timing of the onset of puberty reflects complex interactions between hormonal and neuronal signals with genetic, metabolic, and environmental factors. These interactions presumably begin early in development and ultimately lead to the re-activation of the HPG axis concomitant with the onset of puberty. Multiple factors, both known and unknown, influence the reactivation of the GnRH pulse generator modulating pubertal onset. As noted above, familial patterns of pubertal development and twin studies highlight the role of genetic factors. Studies of families with either delayed or precocious puberty led to discovery of genes relevant to pubertal onset. In addition, genetic factors including single nucleotide polymorphisms (SNPs) have been associated with pubertal timing in both sexes and across ethnic groups. Epigenetic mechanisms have been suggested to affect the development and function of the GnRH neuronal network ultimately influencing HPG axis function. How confounders such as socioeconomic, environmental, and nutritional status influence pubertal development is unclear. These factors can influence puberty timing, HPG axis function, and fertility.

 

Precocious puberty is defined as the development of puberty prior to age 8 in girls, and age 9 in boys (134, 135). In girls, delayed puberty is defined as the absence of breast development by age 13 years, absence of menarche by age 15 or lack of menses after 3 years since breast development. In boys, delayed puberty is defined as absence of pubertal development by age 14 (136). Evaluation of a child with abnormal timing of puberty entails thorough knowledge of normal pubertal development, typical variations of normal pubertal development, and causes of abnormal pubertal development. The next section focuses on the evaluation of a patient presenting with a variation in pubertal development.

 

PRECOCIOUS PUBERTY  

 

Traditionally, the diagnosis of precocious puberty is considered when signs of puberty develop prior to 8 years of age in girls and 9 years in boys (137). These ages are based on Tanner’s original observations on English children regarding typical ages at specific pubertal stages. However, these age criteria should be used as guidelines to complement the evaluation of individual patients. Precocious puberty can be categorized as central or gonadotropin-dependent precocious puberty (CPP) or non-gonadotropin-dependent or peripheral precocious puberty (PPP). Additionally precocious puberty can be further classified as familial or sporadic and syndromic or non-syndromic. The specific etiologies and management differ between the two broad categories of CPP or PPP. Potential consequences of early puberty and menarche in girls include increased risks for breast cancer and diabetes as adults (138, 139).

 

Central Precocious Puberty or Gonadotrophin Dependent Precocious Puberty

 

Central precocious puberty (CPP) is associated with early maturation of the HPG with premature reactivation of the GnRH pulse generator and sequential maturation of breasts and pubic hair in females. In males, sequential maturation of testicular volume, penile enlargement, and pubic hair is observed. Typically, the pubertal characteristics are appropriate for the child's sex (isosexual). Despite the earlier onset of puberty, the sequence of pubertal events is usually normal. CPP is due to organic lesions in approximately 40-100 percent of boys whereas idiopathic precocious puberty is the most common diagnosis in girls (69-98%) (140). These children have accelerated linear growth for age, advanced bone age, and pubertal levels of LH and FSH. A Spanish observational report described an annual incidence of CPP ranging between 0.02 and 1.07 new cases per 100,000 (141) while a Korean study reported an incidence of 15.3 per 100,000 girls, and 0.6 per 100,000 boys (142). Distinguishing among CPP, isolated premature thelarche, and premature adrenarche is important because the pathophysiology and therapeutic interventions differ.

 

CNS LESIONS/INSULTS

 

CPP can be associated with central nervous system lesions. Hamartomas of the tuber cinereum are congenital benign lesions comprised of heterotopic gray matter, neurons, and glial cells. The prevalence is approximately 1 in 200,000 children (143). Hamartomas are the most commonly recognized CNS lesions associated with CPP in very young children. Hamartomas can be categorized as para-hypothalamic, attached or suspended from the floor of the third ventricle, or as intrahypothalamic, in which the mass is enveloped by the hypothalamus and distorts the third ventricle. The lesions do not grow over time, do not metastasize, and do not produce β-human chorionic gonadotropin-(β-hCG). In some instances, hamartomas are associated with gelastic (laughing or crying) seizures. Yet, most patients with hypothalamic hamartomas do not display neurological symptoms (144, 145). Most hypothalamic hamartomas are sporadic and appear to be idiopathic. Hypothalamic hamartomas can also occur in Pallister-Hall Syndrome (PHS) and oral-facial-digital syndrome (OFD) types I and VI (146). Genetic variants in the sonic hedgehog pathway have been associated with hypothalamic hamartoma (147, 148). The mechanism(s) through which hypothalamic hamartomas lead to CPP is unknown. Hamartoma located close to the infundibulum or tuber cinereum are often associated with CPP whereas those functionally connected to the mammillary bodies and limbic circuit are typically associated with epilepsy without CPP (149, 150). As discussed below, medical treatment is usually indicated for hypothalamic hamartomas associated with CPP. Surgical treatment should be limited to large hamartomas complicated by severe refractory drug-resistant epilepsy (151).

 

CNS tumors such as astrocytomas, ependymomas, and pinealomas have rarely been associated with CPP. Among girls, factors associated with CNS lesions include: (1) age younger than 6 years; (2) absence of pubic hair; and (3) estradiol concentrations greater than 30 pg/ml (110 pmol/L) (152, 153). As noted above, suspicion for CNS lesions is higher for boys than for girls.

 

Neurofibromatosis type 1 (NF1) is an autosomal dominant multi-system neurocutaneous disorder due to loss-of-function variants in the neurofibromin-1 (NF1) gene located at chromosome 17q11.2. NF1 is often associated with CPP typically due to optic glioma. The glioma is usually a benign pilocytic astrocytoma that can occur anywhere along the optic tract; the most common locations are within the optic nerve or chiasm. CPP has also been described in NF1 in the absence of optic glioma (154). Children with meningomyelocele and spina bifida also have an increased incidence of CPP. Although the precise mechanism responsible for CPP in these children is unclear, associated factors may include increased perinatal intracranial pressure and brainstem malformations such as Chiari II malformations (155). The mechanistic link between CPP and Rathke cleft cysts, Chiari malformation, and pineal and arachnoid cysts is unclear.

 

Septo-optic dysplasia (SOD) is a heterogeneous congenital condition characterized by presence of at least two features of the classic triad which include optic nerve hypoplasia, anterior pituitary hormone deficiencies, and midline brain anomalies. SOD is associated with genetic variants in HESX1, SOX2, SOX3, and OTX2 genes. Although SOD is typically associated with delayed puberty, CPP can occur (156, 157).

 

CNS tumors may be treated with CNS irradiation (158). In some instances, CNS irradiation is associated with acquired CPP (159). In this situation, concurrent growth hormone (GH) deficiency may be present. The linear growth spurt of CPP may mask the decreased linear growth velocity due to GH deficiency. Hence, in this setting, consideration should be given to evaluating the GH axis by provocative GH testing. If testing shows GH deficiency, the patient may benefit from treatment with GH combined with GnRH agonist therapy. Rarely, CPP occurs following head trauma and can develop many years after the injury(160, 161).

 

SECONDARY CPP

 

Some children exposed to elevated circulating high sex steroid concentrations occurring in other disorders such as McCune-Albright syndrome, congenital adrenal hyperplasia, and virilizing adrenocortical tumors may develop a secondary CPP (163). These individuals typically have accelerated bone age maturation. The precise mechanism responsible for development of the secondary CPP is unclear. The secondary CPP may represent a priming effect of sex steroids on the hypothalamus or potentially as the consequence of the acute decrease in sex steroid concentrations with treatment of the underlying etiology (164) (165).

 

NON-SYNDROMIC CPP

 

Specific genetic variants have been associated with non-syndromic CPP (See Table 1) (166). Loss of function MKRN3variants are the most reported genetic cause of familial CPP. Paternally inherited loss-of-function MKRN3 variants have been reported in up to 33-46 percent of familial cases of CPP and nearly 0-20% percent of sporadic cases (167) . To date, at least 70 deleterious MKRN3 variants have been identified in patients with CPP. These variants lead to diminished inhibition of puberty results in early onset of puberty. Differing ubiquitination patterns suggests that MKRN3 has multiple molecular mechanisms associated with CPP (168). Curiously, a GWAS study investigating parental effects on pubertal development reported that the paternal allele of a specific SNP (rs12148769, G>A) in MKRN3 was associated with age at menarche in healthy girls suggesting that variants in this region affect pubertal timing within the normal range (132). Although circulating MKRN3 concentrations decrease with onset of puberty, peripheral blood MKRN3 concentrations are not adequately sensitive to distinguish CPP (169).

 

TABLE 1. Genes Associated with Central Precocious Puberty (175, 461)

 

Gene

(Reference/s)

Protein encoded

Genetic locus

Comments

MKRN3

(59, 63, 167, 462)

Makorin ring finger protein 3

15q11-q13

Loss-of-function mutation

KISS1R (previously named GPR54)

(463, 464, 465)

Kisspeptin receptor

19p13.3

Gain-of-function mutation

KISS1

(465)

 

Kisspeptin

1q32

Gain-of-function mutation

DLK1

(466, 467, 468)

Delta-like homolog 1

14q32

-Loss-of-function mutation

-Metabolic abnormalities (obesity, type 2 diabetes, hyperlipidemia)

ESR1

(469, 470)

Estrogen receptor 1

6q25.1-q25.2

Mutations/polymorphisms, epigenetic change

CYP19A1

(471)

 

Aromatase

15q21

(TTTA)n polymorphism, epigenetic change

 

Evaluation of another family with CPP led to identification of a loss of function variant in the delta-like 1 homologue (DLK1) gene. DLK1, also known as preadipocyte factor 1, plays a role in the Notch signaling pathway. DLK1 is a paternally expressed gene located at chromosome 14q32.2. Two differentially methylated regions influence the DLK1 imprinting pattern. DLK is located within the genetic locus associated with Temple syndrome. Temple syndrome is characterized by prenatal growth retardation, hypotonia in infancy, motor delay, small hands, CPP, and short stature. In addition to DLK1 loss, two other genes from the paternally inherited chromosome, RTL1 and DIO3, results in Temple Syndrome. Genetic findings associated with Temple syndrome include maternal uniparental disomy, paternal deletion, or loss of differential methylation at the DLK1/MEG3 region on chromosome 14 (170). Women with DLK1 variants also have a metabolic phenotype characterized by overweight/obesity and insulin resistance (171).

 

Gain-of-function variants in the kisspeptin 1 gene (KISS1) and its cognate receptor, KISS1R, gene have been identified in children with CPP. A heterozygous variant in the KISS1 gene, p.Pro74Ser, was identified in a boy who developed CPP at one year of age; in vitro studies suggested that this variant was more stable than the normal protein leading to a prolonged duration of action (172). A girl with precocious puberty was found to have a variant in the KISS1R gene; in vitro studies of this p.Arg386Pro variant showed prolonged activation of the intracellular signaling pathways following kisspeptin stimulation (173, 174).

 

Among a series of 586 children with familial CPP, both maternal and paternal inheritance patterns were found. Variants in MKRN3 were the most common cause in paternally inherited CPP. Among the maternally inherited cases, genetic analysis detected rare variants of unknown significance (175).

 

SYNDROMIC CPP

 

In addition to genetic and idiopathic CPP, CPP can occur as a feature in specific syndromes. Pallister-Hall and Temple Syndrome are described above. Other syndromes associated with CPP include Cowden and Cowden-like cancer predisposition syndromes associated with PTEN, SDHB-D and KLLN gene variants. These disorders are characterized by multiple multisystemic hamartomas which may be associated with CPP when the skull base, infundibulum, or hypothalamus are affected. Although Prader-Willi syndrome is typically associated with delayed puberty, CPP has also been reported (176). Other genetic syndromes associated with CPP include tuberous sclerosis and Williams-Beuren (See Table 2). Williams-Beuren is associated with genetic variant at chromosome 7q11.23 (177). Rare cases of precocious puberty have also been reported in Russell Silver syndrome (178).

 

Table 2. Syndromic Causes of Central Precocious Puberty Without CNS Lesions (CPP)

Gene (Reference/s)

Genetic locus

Comments

MECP2

methyl-CpG-binding protein 2

(472)

 

Xq28

Rare forms of Rett syndrome

X-linked dead-box helicase 3

(461)

Xp11.4

Neurodevelopmental delay

Xp22.33 deletion, SHOX region

(473)

Xp22.33

Body disproportion, short stature, Madelung deformity

Xp11.23-p.11.22 duplication

(474)

Xp11.23-p11.22

Intellectual disability, speech delay, electroencephalogram abnormalities, excessive weight, skeletal anomalies

Temple syndrome

-DLK1

Maternal uniparental disomy or paternal deletion

(170, 473)

14q32.2

Imprinting defect, act via DLK1,

Prenatal and postnatal growth failure, hypotonia, small hands and/or feet, obesity, motor delay

Prader-Willi syndrome

- MKRN3

Paternal deletion or maternal uniparental disomy of chromosome 15q11-q13

(475)

15q11-q13

Changes to the imprinted MKRN3 and/or MAGEL2genes

Hypotonia, obesity, growth failure, cognitive disabilities, hypogonadism

Silver-Russell syndrome

Hypomethylation of chromosome 11p15 or maternal uniparental disomy of chromosome 7       

(476)

 

11p15.5

Possible imprinted or recessive factors, not well elucidated,

Prenatal and postnatal growth retardation, relative macrocephaly, prominent forehead, body asymmetry, feeding difficulties

Williams-Beuren

(177, 477, 478)

7q11.23

Distinct face, cardiovascular disease, short stature, intellectual disability, hyper-sociability

Kabuki syndrome

(479)

12q13.12

Downregulation of estrogen receptor activation

Neurodevelopmental phenotypes, typical distinct face, short stature

Mucopolysaccharidosis type IIIA or Sanfilippo disease

(480)

17q25.3

Severe neurologic deterioration, visceromegaly, skeletal abnormalities

 

NONPROGRESSIVE PRECOCIOUS GONADARCHE

 

Some children experience a nonprogressive (or slowly progressing) CPP (179). Typically, basal gonadotropin concentrations are prepubertal. In general, children with nonprogressive CPP show no or minimal pubertal responsiveness to GnRH stimulation. Height potential is generally unaffected. Typically, these individuals do not usually benefit from GnRH-Ra therapy. Physical findings alone cannot distinguish between progressive and nonprogressive CPP. Presumably this early pubertal development reflects a transient premature activation of the GnRH pulse generator. Longitudinal follow-up to assure that puberty is not progressive is the most appropriate management.

 

GONADOTROPH ADENOMA

 

The anterior pituitary gland consists of highly differentiated ectoderm-derived cells expressing specific hormones such as LH, FSH, GH, prolactin, and ACTH. LH and FSH are secreted by gonadotrophs which are derived from the steroidogenesis factor 1(SF-1) lineage. Gonadotroph adenomas, a type of pituitary adenoma, account for approximately 40% of pituitary adenomas  (180, 181) in adults. In children, gonadotroph adenomas can very rarely cause central precocious puberty (182). Though, most gonadotroph adenomas are nonfunctional and benign, rare cases of functional adenomas have been reported. Hormone profiles of functioning adenomas most commonly show elevated FSH concentrations with or without increase in LH concentrations. Elevated TSH secretion resulting in hyperthyroidism may occur concurrently (180, 181).

 

GUT MICROBIOME AND PUBERTY

 

Microbiota interact with a variety of metabolic and endocrine pathways of the host through genetic expression of more than 100 times the human genome. The gut microbiome variety, composition and impact on health depend on a vast number of variables, both internal, such as age, genetic factors, gender, and endocrine and immune systems, as well as external factors, such as diet, environment, drugs, and pathogens. The relationship between sex hormones and gut microbiome is complex. Sex steroids may directly or indirectly influence the sex-specific gut microbiome that develops during puberty (183). One study reported several gut microbiome alterations in girls with CPP including Ruminococcus bromii, Ruminococcus callidus, Roseburia inulinivorans, Coprococcus eutactus, Clostridium sporosphaeroides, Clostridium lactatifermentans, Alistipes, Klebsiella and Sutterella (176). Although the evidence of the interaction between microbiota and sex hormones remains limited, evidence of diversity of the gut microbiota at different pubertal stages and that alterations may occur in girls with CPP represents an area for potential future development in the prediction and prevention of precocious puberty (184).

 

Treatment of central precocious puberty

 

GONADOTROPIN-RELEASING HORMONE ANALOGS

 

Long-acting Gonadotropin-releasing hormone analogs (GnRHa) have been the standard treatment of CPP since the mid-1980s (185, 186). The GnRHa are super-agonists that bind to the pituitary GnRH receptor downregulating the endogenous pituitary GnRH receptor resulting in decreased gonadotropin and sex steroid secretion. These medications are modified preparations of the native GnRH decapeptide engineered to increase potency and duration of action by substituting a D-isomer amino acid for the naturally occurring L-glycine at position 6. In some analogs, the tenth amino acid is deleted with modification of the naturally occurring L-proline at position 9 (14).

 

Several distinct GnRHa preparations are available differing in route of administration and duration of action (See Table 3) (28). The choice of a specific GnRHa depends on patient, caregiver, and physician preference and on insurance coverage/payment/authorization. Treatment with GnRHa leads to regression or stabilization of pubertal symptoms, deceleration of linear growth velocity, and slowing of skeletal maturation. Some girls experience estrogen withdrawal bleeding about 2-3 weeks following the first injection. Parents and the patient should be counseled to expect this episode of vaginal bleeding (187).  

 

 Table 3. Currently Available GnRHa Therapeutic Options

GnRHa Preparations                         

Dose  

Frequency       

Route

Goserelin

3.6mg

Once a month

intramuscular

Leuprolide

7.5mg

Once a month

intramuscular

 

11.25mg

Once a month

intramuscular

 

15mg

Once a month

intramuscular

 

11.25mg

Every 3 months

intramuscular

 

30mg

Every 3 months

intramuscular

 

45mg

Every 6 months

intramuscular

Leuprolide

45mg

Every 6 months

subcutaneous

Triptorelin

22.5mg

Every 6 months

intramuscular

Nafarelin

800mcg

Twice daily

intranasal

Histrelin

50mg

Annually *

Subdermal implant

*May be used up to 2 years (481).

 

Adverse Effects

 

In general, GnRHas are safe and effective. Adverse events include injection site reactions and sterile abscesses at the site of the injection or implant (188, 189, 190) which may result in loss of efficacy. Minor reported side effects include headaches, hot flashes, vaginal withdrawal bleeding, and mood swings (191). Extremely rare side effects include hypersensitivity reactions, seizures, slipped capital femoral epiphysis, idiopathic intracranial hypertension, and anaphylaxis. One concern regarding the histrelin implant is possible device fracture during extraction; ultrasound-guided removal of the remaining fragments may be necessary (192).

 

GnRHas, specifically only leuprolide and degarelix, have been associated with prolonged QT interval. A prolonged QT interval increases the risk of developing torsades de pointes (TdP) which is a ventricular arrhythmia associated with sudden cardiac death. Low serum potassium or magnesium may exacerbate the risk for prolonged QT interval. Individuals also taking anti-psychotics (typical and atypical), anxiolytics, and anti-depressants may have an increased risk for prolonged QT intervals when taking leuprolide. Hence, providers should inquire regarding other medications, history of congenital heart disease, and family history of Long QT Syndrome or sudden death. If positive, the provider should obtain screening and follow-up EKGs.

 

Studies conflict regarding how GnRHa treatment impacts weight gain and BMI. Some studies have reported weight gain during treatment (193, 194, 195, 196) whereas others have not found any significant change in weight or BMI (197, 198).As with all patients, counseling patients regarding the pre-treatment weight trajectory and healthy lifestyle is beneficial. Women with a history of CPP have been reported to have similar adult weight to the general population (199).

 

Bone mineral density is typically elevated at diagnosis with deceleration in bone mineral accrual during treatment. However, follow-up several years after treatment shows normal bone mineral density compared to population norms (200). Available outcome data suggest that fertility is not compromised for women or men with histories of CPP (192, 201, 202, 203, 204).

 

Despite suggestions that CPP is associated with subsequent development of PCOS, available data are inconsistent. Prospective longitudinal studies are needed to adequately address this concern (205, 206).

 

Who to Treat?

 

For patients less than 7 years of age with a confirmed diagnosis of CPP, the benefit of GnRHa treatment is generally unequivocal. However, the value of GnRHa treatment may be unclear for the peripubertal child (typically a girl) with onset of puberty between 7-9 years of age especially when treatment is unlikely to improve the predicted adult height (PAH) (207). Some girls and their families are comfortable with early pubertal onset and early menarche. In contrast, some girls and their families are distraught when even contemplating early puberty and premature menarche. Consistent evidence-based data regarding negative psychosocial consequences in children with CPP are lacking (208). Further, it may be challenging to justify the medical benefits of GnRHa therapy for early puberty due to the accompanying burdens of increased physician office visits and financial impact. Shared decision-making involving the patient, parents, and medical staff is indispensable to address the benefits and risks of GnRHa in the individual patient (209) . 

 

Goals of Treatment

 

Goals of GnRHa treatment include prevention of pubertal progression and height preservation (210). Growth velocity can significantly decline in some children during GnRHa treatment particularly in those with a markedly advanced bone age (211). The use of other height augmenting medications including recombinant human growth hormone (GH) (212, 213, 214, 215), stanozolol (216, 217), and oxandrolone (218) have been explored but none are recommended for sole use or as an adjunct to GnRHa therapy (219, 220). 

 

Increasing adult height must be judged considering the financial and psychological burdens of this intensive treatment regimen (221). Several recent studies have recommended treatment beyond a bone age of 12 years, however more rigorous studies are needed before such treatment is endorsed (222, 223).

 

Another goal of CPP treatment is to mitigate psychosocial distress and prevent adverse mental health outcomes. One epidemiological study of over 7000 women showed that adolescents with early age of menarche had higher rates of depression and antisocial behavior, which persisted into adulthood (224). Adverse psychosocial experiences reported in girls with early age at menarche include increased likelihood of teenage pregnancy and childbearing, sexual and physical assault, and reduced likelihood of high school graduation (225). However, studies thus far do not show that GnRHa therapy can mitigate these effects. One small study of 36 girls with CPP treated with GnRHas evaluated behavioral health diagnosis and health-related quality of life and found no abnormalities in psychological functioning (226). In a small study of 15 girls with CPP treated with GnRHa and 15 age-matched controls, comprehensive test batteries revealed similar scores in cognitive performance, behavioral, and psychosocial problems (227). A review of 15 studies evaluating the psychosocial impact of CPP showed an increased psychosocial and health-related quality of life burdens with CPP compared with controls (228). The same study showed qualitative data demonstrating emotional lability in patients with CPP and that physical differences associated with sexual precocity could increase feelings of shame and embarrassment which further increase isolation and social withdrawal (228). Again, larger studies are needed to better establish if and how GnRHas influences the psychosocial issues associated with CPP.

 

Monitoring of Treatment

 

Treatment efficacy can be monitored by repeat clinical exams assessing pubertal progression, ultrasensitive LH, FSH and sex hormone concentrations (estradiol in girls, testosterone in boys), rate of progression of bone maturation, estimates of PAH and change in PAH, and patient satisfaction. No uniform consensus exists regarding the optimal strategy for monitoring treatment efficacy in children with CPP. Progression of breast or testicular development may indicate poor adherence, treatment failure, or incorrect diagnosis (188).

 

Random basal LH concentrations to confirm treatment efficacy may be unhelpful because random LH levels often fail to revert to a prepubertal range even when the HPG axis is fully suppressed (229, 230). Therefore, random LH concentrations cannot be used to indicate treatment failure. To confirm gonadotropin suppression, a GnRH stimulation test with short-acting GnRH or, alternatively, a single LH sample 30–120 min after long-acting GnRH analog administration may be performed (231, 232) and different protocols exist regarding the specific timing and number of LH and FSH measurements (233). Some clinicians prefer to utilize clinical indices particularly in areas where hormone determinations are costly.

 

During treatment, breast tissue usually becomes softer with variable changes in size. The rate of bone maturation typically slows with adequate treatment resulting in a decline in BA/CA or a change in BA divided by time. Recent data show that the decline in BA/CA is non-linear and that larger declines are seen in the first 18 months of treatment (222). Thereafter, a slower rate of decrease suggests maintenance of suppression rather than treatment failure.

 

Height velocity is typically rapid prior to treatment and decreases on treatment. The height deceleration is most apparent during the first 18 months of treatment, similar to the deceleration in skeletal maturation. Subsequently, a prepubertal growth rate is often evident (222). Ideally, the rate of bone maturation decelerates resulting in a net gain in height potential. Therefore, calculating PAH during treatment helps assess efficacy. It is also important to understand that mid-parental height (MPH) influences height outcome. GnRHa treatment for CPP may restore genetic potential but rarely causes PAH to surpass genetic potential. Therefore, treatment efficacy by PAH assessment is always in comparison to MPH.

 

Discontinuation of Therapy

 

The decision to discontinue GnRHa treatment needs to be tailored to meet the patient’s specific needs. Factors influencing the decision-making process include synchronizing pubertal progression with peers, patient readiness for resumption of puberty, recent linear growth velocity, bone age X-ray results, and adult height prediction (234). Specific considerations for the developmentally delayed child may be reviewed with the caregivers (137, 234, 235). Pubertal manifestations generally reappear within months of discontinuation of GnRHa treatment; the mean time to menarche is approximately 16 months (217, 218). Several studies have reported that ovulatory function and menstrual cycles are normal once they resume (137, 236).

 

Eripheral Precocious Puberty or Gonadotropin-Independent Precocious Puberty

 

Peripheral precocious puberty (PPP) is due to either excessive endogenous gonadal or adrenal sex steroid secretion (estrogens or androgens) or from exogenous exposure to sex steroids. Ectopic gonadotropin secretion typically from a germ-cell tumor often located in the CNS can also lead to PPP. PPP may be appropriate for the child's sex (isosexual) or inappropriate, with virilization of females and feminization of males (heterosexual). In most instances, pubertal development is incomplete, and fertility is not attained. Etiologies of PPP include:

 

MCCUNE-ALBRIGHT SYNDROME

 

McCune-Albright syndrome (MAS) is an uncommon disorder characterized by the triad of gonadotropin-independent precocious puberty, irregular café-au-lait skin pigmentation and fibrous dysplasia of bone (237, 238).  It has been recognized more recently that MAS may exist as a “form fruste” with only one or two features (239). MAS affects both boys and girls. Importantly, precocious puberty is not observed in all affected individuals and tends to be more common among girls.

 

MAS is due to a somatic cell (post-zygotic) variant arising early during embryogenesis in the GNAS1 gene which is located at chromosome 20q13.3. This gene encodes the Gsα protein coupled to the G-protein membrane receptors for glycoprotein hormones. Vertical transmission has not been reported suggesting that germline variants are embryonic lethal. Variability in post-zygotic expression of the deleterious variant results in a mosaic pattern of tissue expression and inconsistent clinical manifestations between affected individuals (237).

 

Two missense variants, Arg201His and Arg201Cys, are the most frequently identified variants. These variants lead to loss of the α-subunit’s intrinsic GTPase activity resulting in inappropriate cyclic AMP production and constitutive receptor activation. The net result is autonomous ligand-independent signaling by LH, FSH, TSH, GHRH, and ACTH receptors leading to the associated hyperfunctioning endocrinopathies(237).

 

The café-au-lait lesions are generally large with irregular “coast of Maine” borders and typically do not cross the midline. The café-au-lait lesions result from increased tyrosinase gene expression and melanin production in affected melanocytes (240).

 

Bone manifestations are characterized by dysplastic lesions with abnormal bone turnover and inadequate mineralization. These lesions can be associated with pain, malformations, fractures, or nerve compression. The somatic cell gain-of-function variants alter the differentiation of multi-potent skeletal stem cells resulting in the replacement of normal bone and marrow with immature woven bone and fibrotic stroma. The dysplastic tissue is characterized by abundant osteoclast-like cells. Although the somatic Gsα skeletal variants arise during embryogenesis, bone development appears to be normal in utero.

 

Bony lesions become apparent during early childhood typically reaching the maximal burden in young adulthood. The variability in the somatic cell expression accounts for the variability in the location and extent of the fibrous dysplasia.  To date, an accurate ascertainment of risk to develop bone disease is unavailable. However, younger age and higher skeletal burden score derived from scintigraphic bone scans appear to predict longitudinal progression of bone disease. Importantly, evolution of bony lesions is not associated with the extent of endocrine manifestations (241).

 

Overproduction of fibroblast growth factor 23 (FGF23) by skeletal cells bearing the GNAS1 variant can lead to increased urinary phosphate excretion and decreased renal 1-α-hydroxylase activity (242). Although overt hypophosphatemic rickets is uncommon due to compensatory mechanisms, affected individuals often manifest increased serum FGF23 levels and renal phosphate wasting (243).

 

In the gonads, these variants induce ligand independent activation of gonadotropin receptors resulting in subsequent autonomous ovarian estrogen and testicular testosterone secretion in affected prepubertal girls and boys, respectively.

 

Girls may develop recurrent estrogen-secreting cysts accompanied by breast development and linear growth acceleration. Spontaneous resolution of a cyst decreases the estrogen concentration resulting in withdrawal vaginal bleeding. The sequence of pubertal development may be atypical with vaginal bleeding preceding breast development. Hence, MAS should be considered in females with recurrent ovarian cysts and vaginal withdrawal bleeding. Ovarian torsion rarely occurs. Bloodwork may reveal elevated estradiol concentrations with suppressed gonadotropin concentrations. Pelvic ultrasound typically shows one or more ovarian cysts and uterine enlargement. Nevertheless, serum estradiol concentrations and pelvic ultrasound results may be unremarkable following spontaneous involution of an ovarian cyst. Estrogen exposure may lead to accelerated skeletal maturation with adverse consequences on final adult height. In some instances, a secondary gonadotropin-dependent precocious puberty develops. In adult women, the persistent autonomous ovarian activity can lead to abnormal uterine bleeding, menometrorrhagia, which may be so severe as to require blood transfusion. Spontaneous pregnancies can occur, but relative infertility is common (244).

 

Among boys, autonomous GNAS1 activation in the testes leads to Leydig and Sertoli cell hyperplasia which can be associated with macro-orchidism. Scrotal ultrasound may show focal masses, diffuse heterogeneity, and microlithiasis. Differing from typical pubertal progression, testicular volume in boys with MAS does not accurately indicate pubertal status. Substantial autonomous testosterone production is uncommon. Approximately 15% of boys manifest clinical signs of excessive androgen secretion (239). Leydig cell hyperplasia, the most common histologic finding of the testes, carries a low risk of malignant transformation. Thus, conservative management with periodic scrotal ultrasound imaging is appropriate for follow-up of testicular masses detected in boys with MAS (245).

 

Other features associated with MAS include thyrotoxicosis, growth hormone excess (gigantism or acromegaly), and Cushing syndrome. Hypercortisolism is uncommon, typically occurs during the first year of life, and is associated with higher mortality attributed to secondary infections (246). Specific laboratory evaluation and treatment for associated endocrine features should be obtained. Genetic variants can be found in other nonendocrine organs (liver, intestines, and heart) resulting in cholestasis and/or hepatitis, intestinal polyps, and cardiac arrhythmias, respectively (247, 248). Since GNAS1 variants are considered to be weak oncogenes, the risk for malignant transformation is slightly higher than for the general population (239). In addition, women with MAS have an increased risk for breast cancer attributed to earlier estrogen exposure (249).

 

The diagnosis of MAS is usually based on the characteristic clinical features. Due to GNAS1 variant mosaicism, only 20-30% of peripheral blood lymphocytes are positive for the variant using traditional PCR-based testing. However, variant detection is greater than 80% in the affected tissues (250). Newer circulating cell free DNA testing offers a potential methodology to assess for MAS variants (251) . Importantly, negative testing, especially of peripheral blood lymphocytes, does not exclude the diagnosis of MAS.

 

Therapeutic goals focus on treating specific clinical manifestations. For manifestations related to puberty, current medications either inhibit sex steroid biosynthesis or block their actions at the level of end organs. Minimal evidence-based data are available because of the low prevalence of MAS.  Ketoconazole, an anti-fungal medication, has been used because it inhibits the steroidogenic cytochrome P450 enzymes decreasing adrenal and gonadal steroidogenesis (252). However, ketoconazole may interfere with cortisol synthesis; patients need to be monitored for possible adrenal insufficiency and may benefit from use of stress dose hydrocortisone treatment. Rarely hepatic toxicity can occur. 

 

Aromatase inhibitors prevent conversion of androgens to estrogens. Initial reports for testolactone, fadrozole, and anastrozole were disheartening because no enduring beneficial effects on skeletal growth and bone maturation were observed. Letrozole has been used and showed sustained beneficial effects on skeletal maturation and predicted final height in one small series (253).

 

Selective estrogen receptor modulators such as tamoxifen and fulvestrant have been used. Tamoxifen has both agonist and antagonist activity at the estrogen receptor. Despite reports regarding the efficacy of tamoxifen to reduce vaginal bleeding accompanied by positive effects on bone, this medication has been reported to increase risk of endometrial disease in adult women (254). In view of potential risks for endometrial cancer, tamoxifen should be used with great caution in women with MAS (255).

 

Fulvestrant is a pure estrogen receptor blocker administered by intramuscular injections at monthly intervals. In one small series, vaginal bleeding was reduced with complete cessation of vaginal bleeding in only 8/25 girls. The rate of skeletal maturation decreased without any significant change in linear growth velocity or predicted adult height. Fulvestrant was reported to be well tolerated; additional studies are needed to supplement these initial findings (256).

 

In the past, surgery cystectomy or oophorectomy had been performed in girls with MAS (257). Since cyst recurrence is common, cystectomy should be avoided if possible. Women with MAS have the potential for fertility and spontaneous pregnancy; hence, oophorectomy should be avoided (258).

 

For boys with MAS associated precocious puberty, therapeutic interventions include androgen receptor blockers, aromatase inhibitors, and ketoconazole to interfere with testosterone synthesis (258). Combination therapy with bicalutamide and anastrozole was successfully utilized in one boy with PPP due to MAS (259). Bicalutamide is a potent nonsteroidal antiandrogen that binds to and inhibits the androgen receptor and increases the receptor’s degradation. Surgical intervention should only be considered for rapidly enlarging palpable testicular masses due to the risk of malignancy (245).

 

PREMATURE MENARCHE AND OVARIAN CYSTS  

 

Functioning ovarian follicular cysts can secrete estradiol resulting in isolated premature vaginal bleeding or peripheral precocious puberty (260, 261). Additional signs of puberty may be absent in girls with isolated premature menarche. Although some girls may present with slight breast development followed by vaginal bleeding. The bleeding typically lasts only a few days and is usually attributed to spontaneous resolution/regression of an estrogen-secreting ovarian cyst. By the time a pelvic ultrasound can be obtained, the cyst has resolved, and the ultrasound shows no abnormalities. Isolated premature menarche may be limited to a single episode or may be recurrent. In most instances, linear growth velocity, onset of cyclic menstrual cycles, and final adult height are unaltered.

 

Differential diagnosis includes sexual abuse, vaginal foreign body, vaginal infections, MAS, or primary hypothyroidism(262). Due to the intermittent nature of these cysts, conservative medical management is usually appropriate (263). Large cysts may predispose to ovarian torsion (264, 265, 266, 267). Patients with ovarian torsion usually present with short duration of pain and systemic symptoms such as vomiting. Given the low frequency of malignancy in such an ovarian, detorsion with or without cystectomy is generally preferred (268). Gonadectomy should be avoided to preserve fertility. Rarely, rhabdomyosarcoma or sclerosing stromal tumors can present with vaginal bleeding.

 

OVARIAN TUMORS  

 

Estrogen-secreting ovarian tumors are a rare cause of peripheral precocious puberty. Specific types of tumors include granulosa cell, gonadal stromal cell, ovarian sex cord stromal, and theca cell tumors.

 

Juvenile granulosa cell tumors (JGCT) are the most common ovarian tumors. Typically, these tumors present with rapidly progressive isosexual precocity (269). Most JGCT are large enough to be palpated during an examination and are typically limited to the ovary at the time of diagnosis. Circulating estradiol concentrations may be extremely elevated with suppressed gonadotropin concentrations. Circulating tumor markers including α-fetoprotein (AFP), lactate dehydrogenase (LDH), β-human chorionic gonadotropin (β-hCG), cancer antigen 125 (CA-125), and inhibin B can be identified. Genetic somatic variants have been identified in juvenile granulosa cell tumors. Over 60% of JGCT carry in frame duplications in the AKT1 gene; this gene codes for a kinase involved in ovarian mitogenic signaling (270). Other identified variants include KMT2C-truncating and the ribonuclease III domain of DICER1 variants.  In contrast to adult granulosa cell tumors of the ovary, variants in the FOXL2 gene are generally not found in JGCT. Ollier and Maffucci syndromes, rare disorders associated with benign cartilaginous enchondroma, have been associated with JGCT (271). Surgical excision with peritoneal cytology for staging is the primary treatment.

 

Rarely, other tumors including gonadoblastoma, lipid tumors, cystadenomas, and ovarian carcinomas can secrete sex steroids. Finding elevated serum inhibin and AMH concentrations suggest that the tumor cells are derived from granulosa or Sertoli cells.

 

Sex cord tumors with annular tubules can occur in patients with Peutz-Jeghers Syndrome. Peutz-Jeghers Syndrome is an autosomal dominant disorder associated with mucocutaneous pigmentation, gastrointestinal polyposis, and genetic variants in the STK11 gene located at chromosome 19p13.3. (272) The gonadal tumors can be multi-focal, bilateral, and can differentiate into granulosa cell or large cell calcifying Sertoli cell tumors with the potential to secrete estrogen. Thus, girls may present with precocious puberty whereas boys may present with gynecomastia.

 

Sertoli-Leydig cell tumors are rare ovarian tumors often associated with somatic or germline DICER1 variants (273). Most are unilateral, but bilateral tumors have been described.  These tumors contain testicular structures, Sertoli and Leydig cells, and can rarely secrete androgens. Hence, girls can virilize with pubic hair development (274, 275). Girls known to carry germline DICER1 variants should undergo regular pelvic ultrasounds to screen for ovarian tumors (276).

 

LEYDIG CELL TUMORS  

 

Leydig cell tumors are a subtype of testicular stromal tumors that arise from testosterone producing Leydig cells. In prepubertal boys, presenting features include penile enlargement, acne, development of pubic and axillary hair, and accelerated linear growth velocity. Examination of the testes typically show asymmetric testicular volume due to a unilateral testicular tumor. Leydig cell tumors are usually benign. Bloodwork shows elevated circulating testosterone concentrations and suppressed gonadotropin concentrations. Ultrasound is useful to assess testicular volume and morphology.

 

Treatment involves surgical removal of the tumor. When possible, testis-sparing enucleation is preferred to radical orchiectomy to preserve testicular function and fertility. The surgical approach is dictated by the intraoperative assessment of tumor size, location, and the amount of remaining normal testicular parenchyma (277, 278).

 

HUMAN CHORIONIC GONADOTROPIN SECRETING GERM CELL TUMORS  

 

During early gestation, primordial germ cells migrate from the hindgut to the gonads. In some instances, the germ cells can migrate to locations outside of the gonad, fail to undergo apoptosis, proliferate in these atypical locations, and ultimately become hCG-secreting germ cell tumors (279). Common locations for germ cell tumors include the CNS, lung, or liver (280). Boys and men with Klinefelter syndrome are at higher risk to develop extra-gonadal GCTs particularly in the mediastinum (281). In addition, hepatoblastoma secreting hCG and α-fetoprotein can also present with precocious puberty (282). Due to the similarity between hCG and LH which have identical α-subunits and related β-subunits, tumor-derived hCG stimulates testicular LH receptors resulting in testosterone secretion. In the prepubertal boy, the aberrant hormone exposure can result in precocious puberty (27, 28). Testicular volume may not increase since seminiferous tubule growth does not occur in the absence of FSH stimulation. Bloodwork shows elevated hCG and testosterone concentrations with suppressed/variable LH and FSH concentrations.

 

Prepubertal girls generally do not develop isosexual precocious puberty with hCG-secreting germ cell tumors because in the absence of FSH, the granulosa cells do not express aromatase and are unable to synthesize estradiol.

 

GERM CELL TUMORS

 

Chromosomal aneuploidy or genetic variants can interfere with gonadal development resulting in dysgenetic gonads. In this situation, the appropriate microenvironment for normal germ cell maturation is absent, thereby disrupting the normal maturational progression for germ cells. This situation may result in the development of gonadal germ cell tumors. Precursor lesions of germ cell tumors include germ cell neoplasia in situ (GCNIS, formerly termed carcinoma in situ – CIS) and gonadoblastoma (283). Subsequently, dysgerminoma, seminoma, or non-seminoma may develop. Usually, such germ cell tumors do not secrete significant amounts of sex steroids.

 

FAMILIAL MALE LIMITED PRECOCIOUS PUBERTY (FMPP)

 

Familial male-limited precocious puberty (also known as testotoxicosis) is due to an autosomal dominant activating germline variant in the LH/choriogonadotropin receptor (LHCGR) gene located at chromosome 2p21. The LH/CG receptor is a G-protein coupled receptor (284). The variant is associated with autonomous ligand-independent receptor signaling leading to Leydig cell hyperplasia and premature testosterone secretion in prepubertal boys. Pathogenic missense variants associated with FMPP tend to congregate in an apparent hot spot located in the 6th transmembrane segment and in the 3rd intracellular loop (285).

 

Affected males typically present between two to six years of age with penile enlargement, linear growth acceleration, advanced skeletal maturation, acne, and pubarche. The testes are usually symmetrically enlarged due to the Leydig cell hyperplasia, but the size is disproportionately smaller compared to the testosterone levels (286, 287). A large portion of the testicular volume is formed by Sertoli cells which are not stimulated in this condition (286, 287). Circulating testosterone concentrations are elevated with suppressed gonadotropin concentrations. Although adult height is generally compromised, fertility has been reported (288). Longitudinal follow-up with testicular self-examination and scrotal ultrasound is recommended because malignant testicular germ cell tumors have been described in a few individuals (289).

 

Therapeutic goals include decreasing autonomous testicular testosterone secretion and slowing epiphyseal maturation. To date, several medications including ketoconazole, spironolactone, bicalutamide, and aromatase inhibitors have been used with varying efficacy (286). To date, the most effective therapy is combination treatment with an anti-androgen and an aromatase inhibitor (290, 291). If secondary GnRH-dependent precocious puberty develops, GnRH agonist therapy can be added to the therapeutic regimen. Abiraterone, a selective CYP17A1 inhibitor, was utilized in a young boy with bilateral Leydig cell tumors and resistance to the usual combination regimen of an anti-androgen and aromatase inhibitor. He required glucocorticoid replacement therapy and monitoring for possible excessive mineralocorticoid action because of abiraterone treatment-associated iatrogenic 17-hydroxylase/17,20-lyase deficiency (292).

 

Although inherited as an autosomal dominant disorder, girls do not develop precocious puberty (293). Since only the LHCGR gene is affected, the presumably minimally increased theca cell androgens cannot be aromatized to estradiol in the absence of FSH stimulation. Importantly, asymptomatic women can transmit the affected allele to their sons.   

 

PRIMARY HYPOTHYROIDISM

 

Children with profound chronic primary hypothyroidism may present with precocious puberty. Van Wyk and Grumbach described this association in 1960 (294). Clinical features in girls include early breast development, vaginal bleeding, and galactorrhea. Boys present with testicular enlargement. Pubic and axillary hair are absent. Typical features associated with hypothyroidism such as short stature, impaired linear growth, puffy face, dry skin, constipation, and delayed skeletal maturation (despite pubertal changes) are usually evident. Pituitary imaging shows an enlarged pituitary gland. Abdominal ultrasound may show ovarian enlargement with or without ovarian cysts. Labs show mild elevation in FSH levels but LH levels usually remain prepubertal.

 

Levothyroxine therapy induces regression of pubertal symptoms, stops vaginal bleeding, and decreases pituitary volume. However, final height may often be compromised due to accelerated skeletal maturation upon initiation of thyroxine treatment. One potential mechanism for the precocious puberty is cross-reactivity of TSH at the ovarian FSH receptor. TSH and FSH share a common α-subunit with hormone specificity due to the differing β-subunits (295). This mechanism was tested using recombinant human TSH in an in vitro bioassay which, at high concentrations, was able to stimulate human FSH receptors (296, 297, 298, 299).

 

VIRILIIZING CONGENITAL ADRENAL HYPERPLASIAS

 

The virilizing congenital adrenal hyperplasias (CAHs) are autosomal recessive disorders associated with impaired adrenal steroidogenesis due to genetic variants in steroidogenic enzyme genes. The most common is 21-hydroxylase deficiency due to variants in the 21-hydroxylase gene (CYP21A2) located at chromosome 6p21.33. Clinically, congenital adrenal hyperplasias reflect a phenotypic spectrum ranging from presentation in neonatal period with classic salt-losing CAH to presentation during infancy/todder age with classic simple virilizing CAH to later presentations with non-classic CAH. Milder or non-classic forms have been described for 11-β-hydroxylase deficiency and 3β-hydroxysteroid dehydrogenase deficiency (300).

 

Children with non-classic CAH typically present with premature pubarche characterized by pubic/axillary hair development, acne, accelerated linear growth velocity, and advanced skeletal maturation. Girls may have clitoromegaly whereas boys may have phallic enlargement with prepubertal testicular volume. Adult women with non-classic CAH usually present with irregular menses, hirsutism, and infertility.

 

The diagnostic test for 21-hydroxylase deficiency is an elevated 17-hydroxyprogesterone (17-OHP) concentration. Early morning basal 17-OHP values have been suggested as an effective screening test with reports of 100% sensitivity and 99% specificity with a threshold value of 200 ng/dl (6 nmol/L) to diagnose NCAH in children who present with premature pubarche (301). If the diagnosis is highly suspected despite relatively normal 17-OHP concentrations, an ACTH stimulation test may be indicated to exclude the diagnosis of 21-hydroxylase deficiency. For an ACTH stimulation test, following collection of a basal blood sample, 0.25 mg synthetic ACTH (Cortrosyn) is administered by intravenous or intramuscular routes; a second blood sample is collected at 30 and/or 60 minutes. Physician preference governs the timing of the ACTH-stimulated 17-OHP concentration. In the future, 21-deoxycortisol and 11-oxyandrogens may be increasingly utilized in the diagnosis and management of 21-hydroxylase deficiency (302, 303). The reader is referred to more extensive discussion of the virilizing CAH (304, 305).

 

ADRENOCORTICAL TUMORS

 

Androgen-secreting adrenocortical tumors are extremely rare causes of PPP accounting for less than 1% of all childhood malignancies. Most tumors occur in children younger than 4 years of age with a second smaller peak in adolescents. Pediatric adrenocortical tumors are categorized as adenomas or carcinomas based on histological features. However, histopathologic differentiation may be challenging, and biologic behavior of the tumor may help with this categorization (306).

 

Pediatric adrenocortical carcinoma is more common in girls than boys and has a bimodal pattern with peaks under age 5 and over 10 years of age (307). Adrenocortical tumors are associated with several genetic syndromes such as Li-Fraumeni syndrome and Beckwith-Wiedemann syndrome (BWS). Li-Fraumeni Syndrome is an autosomal dominant familial cancer syndrome associated with germline p53 gene variants. The p53 gene (or TP53 gene) is a tumor suppressor gene located at chromosome 17p13.1, and codes for the protein p53. Malignancies associated with Li-Fraumeni syndrome include adrenocortical carcinomas, breast cancer, brain tumors, and sarcoma. The incidence of adrenocortical tumors is 10-15 times higher in southern Brazil; this has been attributed to the higher prevalence of the R337H variant of the TP53 gene (308).

 

Beckwith-Wiedemann syndrome is characterized by macroglossia, macrosomia, organomegaly, neonatal hypoglycemia due to hyperinsulinism, and abdominal wall defects. This disorder is associated with uniparental disomy in the 11p15 chromosomal region leading to IGF2 growth factor overexpression. Although only 1% of children with Beckwith-Wiedemann Syndrome will develop adrenocortical carcinomas, these adrenal tumors account for approximately 20% of the neoplasms in children with this disorder (309). Other disorders associated with adrenal tumors include Multiple Endocrine Neoplasia Syndrome Type 1 (MEN1) and Carney complex (310).

 

The next section reviews variants of puberty associated with early pubertal changes and are important differentials to consider in the evaluation of CPP.

.

PREMATURE THELARCHE

 

Premature thelarche is the premature development of glandular breast development. The breast development may be unilateral or bilateral. Typically, premature thelarche develops in otherwise healthy girls between 12-24 months of age and is self-limited. No other pubertal changes are evident; linear growth velocity is normal and pubic/axillary hair are absent. On physical examination, the areolae and vaginal mucosa are prepubertal. The diagnosis can usually be made on a clinical basis without bloodwork or bone age X-rays (311). Pelvic ultrasound showed increased prevalence of ovarian microcysts in girls with premature thelarche compared to age-matched controls; no correlation between ovarian cysts, gonadotropin concentrations, and estradiol concentrations has been found (312). Longitudinal follow-up is appropriate to confirm the diagnosis and assess for the unlikely possibility of progression to CPP.

 

PREMATURE ADRENARCHE

 

Pubarche refers to the appearance of pubic/axillary hair, increased apocrine odor, and acne due to the onset of adrenarche. Adrenarche refers to the pubertal maturation of the adrenal zona reticularis. Adrenarche which normally occurs in children between 6-8 years of age and is characterized by increased secretion of the adrenal androgen precursors DHEA, DHEAS, and androstenedione.

 

Premature adrenarche is characterized by premature pubarche, which is defined as the development of pubic or axillary hair before 8 years in girls or 9 years in boys. There is no breast development in females and no testicular enlargement in males. Bone age is usually not advanced. Premature adrenarche is a diagnosis of exclusion. Thus, exclusion of other disorders such as CAH, androgen-secreting tumors, exogenous androgen exposures, and other rare genetic disorders such as apparent cortisone reductase and PAPS synthase 2 (PAPSS2) deficiencies is essential (313).

 

Children with premature adrenarche and early androgen excess may be at a higher risk to develop the metabolic syndrome. Waist circumference (WC), waist/hip ratio, and total and truncal fat mass increase are detected in premature adrenarche. Increases in systolic and diastolic blood pressure (BP), total cholesterol (TC), very low-density lipoprotein (VLDL), TC/high density lipoprotein (HDL), low density lipoprotein (LDL)/HDL ratio, and atherogenic index (AI) have been reported. Increased insulin concentrations starting from prepubertal ages may occur suggesting that premature adrenarche may be one of the first symptom of insulin resistance (IR) in childhood (314, 315). T2DM may occur in a subset of these cases. Ovarian hyperandrogenism, hirsutism, ovulatory dysfunction, and polycystic ovaries may be more frequent in girls with premature adrenarche during post pubertal ages than normal population. Although, early retrospective data in a homogenous population suggests an association between premature adrenarche and adolescent hyperandrogenism (316), more recent longitudinal data suggests that premature adrenarche was not associated with adolescent ovarian dysfunction and was only associated with lower SHBG concentrations (317).

 

EXPOSURE TO EXOGENOUS SEX STERIODS  

 

Feminization, including gynecomastia in males, has been attributed to excess estrogen exposure from creams, ointments, and sprays. Other possible sources of estrogen exposure include contamination of food with hormones, phytoestrogens (e.g., in soy), and over-the-counter remedies such as lavender oil and tea tree oil (318, 319, 320, 321). Similarly, virilization of young children has been described following inadvertent exposure to androgen-containing creams/gels (322).

 

Endocrine-Disrupting Chemicals 

 

Various endocrine-disrupting chemicals (EDCs) are found in the environment  (323, 324, 325, 326).  Most EDCs have chemical structures similar to those of endogenous sex steroids. These chemicals can disrupt steroid hormone receptor binding and hormone metabolism altering hormone concentrations or changing hormone synthesis/degradation (327). EDCs can act beyond steroid hormone receptors by affecting transcriptional modulators and direct effects on genes. Some EDCs have mixed activities, and most EDCs include several different chemicals. The patient’s age and duration of exposure modulate the consequences of EDC exposure. In addition, EDCs can be classified as persistent (long-lasting) or non-persistent (short half-lives). Environmental EDC exposures can be transgenerational such that future generations could be affected (328). Mechanisms for EDC exposures include ingestion, topical use, inhalation, and transfer across the placenta (329).

 

The consequences of mixed “cocktail” EDC exposures on pubertal development are indeterminate. A systematic review with a stringent meta-analysis found no consistent association between xenobiotic EDCs and pubertal timing apart from an insinuation that, in girls, postnatal exposure to phthalates could be associated with earlier thelarche and later pubarche, consistent with their anti-androgenic properties. Methodological heterogeneity, limited number of studies, and variability in statistical analyses constrained the conclusions of this systematic review. Hence, future longitudinal epidemiologic studies to clarify the specific EDCs, age at exposure, and duration of exposure will be valuable (327, 330).

 

 

DELAYED PUBERTY

 

Gonadarche associated with the reactivation of the GnRH pulse generator, is signified by breast development in girls and testicular enlargement in boys. Delayed puberty is defined as absence or delayed onset of gonadarche at a chronologic age >2 standard deviations later than the population mean. In girls, delayed puberty is defined as absence of breast development by age 13 years or lack of menarche by age 15 years (331) or 3 years from onset of thelarche (332). In boys, delayed puberty is defined as the lack of testicular enlargement to a volume >= 4 ml by age 14 years (333). Delayed puberty is more common in boys than in girls.

 

Four main categories of delayed puberty have been described (See Table 4).

  • transient hypogonadotropic hypogonadism associated with delayed maturation of the HPG axis also known as constitutional delay of growth and puberty (CDGP)
  • hypergonadotropic hypogonadism characterized by primary gonadal dysfunction with consequent elevated LH and FSH concentrations.
  • hypogonadotropic hypogonadism with low LH and FSH concentrations due to congenital (CHH) or acquired causes
  • functional hypogonadotropic hypogonadism (FHH), as seen in chronic health disorders such as cystic fibrosis, renal failure, inflammatory bowel disease, restrictive eating disorders etc.

 

Table 4. Etiologies of Delayed Puberty (458)

Condition

Etiology

Constitutional Delay of Growth and Puberty

Genetic basis has infrequently been described in HS6ST1, FTO, IGSF10, EAP1 genes

Hypergonadotropic Hypogonadism

Congenital:

-Klinefelter’s syndrome

-Turner syndrome

-Gonadal dysgenesis

-Anorchia

-Primary ovarian insufficiency

-Testicular regression syndrome

-Genetic causes: FMR1, STAG3, NR0B1, NR5A1, FOXL2, WT1 and others

-Galactosemia

Acquired:

-Infectious (mumps)

-Autoimmune (polyglandular syndromes)

-Surgery (torsion, trauma)

-Chemotherapy (alkylating agents)

-Radiation

-Gonadal tumor

Hypogonadotropic Hypogonadism

Congenital:

-Isolated HH: Over 50 genes have been identified; notable are ANOS1, FGFR1, FGF8, PROK2, CHD7, KISS1, KISS1R, GNRH, GNRHR and others

-Prader Willi

-CHARGE syndrome

-Noonan

-Bardet-Biedl

- Panhypopituitarism associated with genetic variants in PROP1, HESX1, LHX, LHB, FSHB and others.

Acquired:

-Central nervous system tumors (e.g., craniopharyngiomas, germinomas), cysts,

-Cranial surgeries,

-Cranial radiation therapy greater than

 30 Gy

-Other inflammatory, autoimmune (hypophysitis), and infiltrative (Langerhans cell histiocytosis) diseases of the pituitary gland

Functional Hypogonadotropic Hypogonadism

-intense physical stress (competitive gymnastics, ballerina syndrome)

-emotional stress (elevated glucocorticoids)

-caloric deficit (anorexia nervosa)

-chronic systemic illness (celiac, inflammatory bowel disease, CF, renal disease)

-endocrinopathies (hypothyroidism, excess glucocorticoids, hyperprolactinemia)

-medication adverse effects

-pituitary iron deposits in chronic transfusion dependent children

 

Transient Hypogonadotropic Hypogonadism Associated with Delayed Maturation of the HPG Axis/ Constitutional Delay of Growth and Puberty (CDGP)

 

Transient hypogonadotropic hypogonadism also known as constitutional delay in growth and puberty (CDGP) is the most common etiology of delayed puberty occurring in 70% of boys and 32% of girls with delayed puberty (334). In both sexes, CDGP is self-limited and is considered to represent a variant of normal pubertal timing. CDGP has a strong genetic component, with a positive family history of delayed puberty reported in 50% to 75% of cases (335).

 

Distinguishing between CDGP and congenital hypogonadotropic hypogonadism (CHH) may be challenging because these conditions share clinical features, hormone levels, and radiological findings. Inhibin B and LH levels tend to be lower in boys with CDGP, but the overlap in values precludes the use of these hormones to distinguish between CDGP and CHH.

 

For boys, 3-4 months of steroid priming with testosterone followed by 3-4 months of observation is commonly used to discriminate CHH from CDGP (336). It has been suggested that this sex steroid exposure stimulates resumption of the HPG axis activity leading to secondary sex characteristics typical of male puberty (337). Individuals who show no pubertal progression during the observation period should be evaluated for CHH or another disorder affecting the HPG axis. Estradiol priming has been used similarly in girls to distinguish CHH from CDGP (338). Due to the differences in the long-term outcomes, accurate diagnosis is essential, with CDGP being largely a diagnosis of exclusion (339).

 

CDGP occurs more commonly in family members of individuals with CHH compared to the general population (340). Individuals with CDGP appear to have higher prevalence of pathogenic variants compared to unaffected family members or controls (341). Some genetic variants have been detected in both individuals with CDGP and CHH; these genes include HS6ST1, PROKR2, TAC3, TAC3R, and IL17RD (342). Genetic variants associated primarily with CDGP include IGFS10, EAP1, and FTO (338).

 

Hypergonadotropic Hypogonadism

 

Pubertal delay associated with hypergonadotropic hypogonadism is usually associated with disorders affecting gonadal function, specifically gonadal steroidogenesis. With the onset of gonadarche and increased GnRH and gonadotropin secretion, inadequate gonadal steroid secretion and lack of negative feedback leads to increasing gonadotropin secretion. These conditions may be present at birth or acquired.

 

TURNER SYNDROME  

 

Turner Syndrome refers to deletions or structural rearrangements of the X chromosome. The

reported incidence is around 1 in 2500 liveborn female births (343). The initial in utero process of ovarian differentiation proceeds normally with migration of the primordial germ cells into the developing ovary during the fourth week of gestation. By 18 weeks of gestation, premature degeneration of ovarian follicles has begun. The ovarian follicles are typically replaced by connective tissue resulting in the characteristic streak gonad. This accelerated follicular atresia usually leads to premature ovarian insufficiency. Girls with Turner syndrome have gonadal dysgenesis or “streak gonads” in 85% of cases at birth. However, because adrenal androgen secretion is not impaired, the onset of pubarche usually occurs at a normal time. Typical clinical features of girls with Turner syndrome include short stature, short/webbed neck, shield shaped chest with the appearance of widely spaced nipples, cubitus valgus, and Madelung deformity of the forearm and wrist, shortened fourth metacarpals/metatarsals, horseshoe kidneys, coarctation of the aorta, increased risk for autoimmune conditions, and aberrant development of the lymphatic system. Many girls with Turner syndrome may remain undiagnosed until later in childhood or adolescence when they present with short stature and/or delayed puberty. With increased utilization of noninvasive prenatal screening (NIPS), many girls with Turner Syndrome are detected prenatally. The reader is referred to other publications for more extensive discussion regarding the features and approach to multidisciplinary health care management (344, 345, 346).

 

KLINEFELTER SYNDROME

 

Klinefelter Syndrome is a chromosomal aneuploidy characterized by 47, XXY karyotype and premature testicular insufficiency. Increased NIPS utilization has led to detection of many boys in utero and is estimated to occur in 1 in 667 males based on prenatal cytogenetic analysis (347). However, many men remain underdiagnosed, with less than 10% of patients being diagnosed prior to puberty. Men with Klinefelter syndrome typically present with tall stature, incomplete puberty, or gynecomastia. Generally, the onset of puberty is not delayed. Klinefelter syndrome is associated with small firm testes, Sertoli cell dysgenesis, impaired spermatogenesis, and variable degrees of testosterone deficiency (348). Learning disabilities, language and visuospatial processing defects, and neuropsychiatric conditions such as attention-deficit/hyperactivity disorder and depression are common (349). If a tumor is found in the anterior mediastinum, a karyotype should be performed to evaluate for Klinefelter syndrome because of its association with mediastinal germinoma (350). Despite normal BMI, the body fat percentage, and the ratio between android fat percentage and gynoid fat percentage are significantly higher than normal. They may also have an impaired bone metabolism starting during childhood and adolescence. Systematic studies are needed to evaluate whether testosterone replacement therapy during puberty will improve these parameters (351). The reader is referred to other publications for more extensive discussion regarding the features and approach to multidisciplinary health care management (352, 353, 354, 355, 356).

 

DIFFERENCES OF SEX DEVELOPMENT  

 

Differences of sex development (DSDs) are a group of conditions where external genital development is atypical. These disorders are associated with chromosomal anomalies, genetic variants, and environmental influences (357). Gonadal function is impaired in some types of DSDs resulting in primary gonadal failure and hypergonadotropic hypogonadism. Detailed review of DSDs is beyond the scope of this chapter. The interested reader is referred to other Endotext chapters for more extensive review of DSDs.

 

GENETIC CAUSES OF PREMATURE OVARIAN INSUFFICIENCY (POI)

 

POI can present with primary or secondary amenorrhea.  Fragile X-associated premature ovarian insufficiency is among a family of disorders caused by the expansion of a CGG trinucleotide repeat sequence located in the 5′ untranslated region (UTR) of the fragile X messenger ribonucleoprotein 1 (FMR1) gene on the X chromosome. One etiology is premutation of the FMR1 gene associated with 55-200 CGG repeats without abnormal methylation of the neighboring CpG island and promoter, responsible for both fragile X associated premature ovarian insufficiency in females and fragile X associated tremor ataxia syndrome in males and females where patients may present with mild to moderate intellectual disability, intentional tremor and cerebellar ataxia, peripheral neuropathy, Parkinsonism, and urinary and bowel incontinence.

 

The X chromosome carries many genes that govern follicular maturation and overall ovarian function, and numerical and structural changes in this chromosome, as in Turner syndrome or triple X syndrome, are associated with POI.

 

Multiple genes are involved in ovarian differentiation, oocyte development, and, ultimately, folliculogenesis and variants in these genes may be associated with premature ovarian insufficiency (358, 359). The clinical phenotype ranges from delayed puberty to secondary amenorrhea (360). 

 

GALACTOSEMIA

 

Galactosemia is a rare cause of delayed puberty. Classic galactosemia is a rare inborn error of galactose metabolism due to a defect in the gene encoding the galactose-1-phospate uridyltransferase enzyme (GALT). The prevalence is approximately 1/30,000–60,000 (361). Early manifestations include lactose intolerance, jaundice, failure to thrive, lethargy, hepatocellular damage, renal tubular disease, and cataracts. A galactose-free diet can reverse the neonatal symptoms. However, some long-term complications such as developmental delay, intellectual disability, epilepsy, osteoporosis, and premature ovarian insufficiency may still develop. In females, hypergonadotropic hypogonadism resulting in delayed puberty, primary or secondary amenorrhea, and infertility may occur (362, 363). Available data from patients with classic galactosemia suggest that the primary ovarian insufficiency is due to dysregulation of pathways essential for folliculogenesis culminating in premature ovarian insufficiency (364). Several previous cohort studies in males showed delayed puberty and below-target final height (365, 366, 367), however a recent study with 47 males showed that puberty and fertility were normal and in contrast to earlier reports, AMH, testosterone and Inhibin B levels were normal (361).

 

Hypogonadotropic Hypogonadism

 

Pubertal delay associated with hypogonadotropic hypogonadism is usually associated with disorders affecting the neurons that secrete GnRH or the pituitary gonadotrophs that secrete the FSH and LH. These conditions may be present at birth or acquired as described below.

 

CONGENITAL HYPOGONADOTROPIC HYPOGONADISM

 

The initiation and maintenance of reproductive capacity in humans depends on pulsatile GnRH secretion. Congenital hypogonadotropic hypogonadism (CHH) results from the absence of the normal pulsatile GnRH secretion or deficient pituitary gonadotropin secretion leading to delayed puberty and infertility. The number of genetic loci associated with CHH continues to expand (Table 4). CHH may be associated with variants in genes involved in the development or migration of GnRH neurons as well as genes involved in the secretion or action of GnRH (368). Autosomal recessive, autosomal dominant, X-linked, and oligogenic inheritance have been described (369, 370). Additional genetic influences include epigenetic factors (371). Clinical heterogeneity has been described between and within families (372).

 

Given the developmental origins of GnRH neurons in the olfactory placode, CHH can be associated with anosmia or hyposmia. The association of CHH and anosmia is known as Kallmann syndrome. Classic Kallmann syndrome is associated with variants in the ANOS1 gene which is mapped to the X chromosome. Other features of Kallmann syndrome due to ANOS1 variants include unilateral renal agenesis, sensorineural hearing loss, dental agenesis, synkinesia (alternating mirror movements), and cleft lip/palate (373).

 

In syndromic CHH, associated clinical features may help identify the possible gene(s). For example, clinical features associated with FGFR1 variants include anosmia/hyposmia, cleft lip/cleft palate, dental agenesis, and skeletal anomalies. CHH can also occur in the CHARGE syndrome, which is characterized by coloboma, congenital heart disease, choanal atresia, genital anomalies, ear anomalies, and development delay. CHH can occur with impaired pituitary development associated with PROP1, HESX1, or LHX variants. CHH is also associated with variants in the GnRH, GnRHR, LHB, and FSHB genes (See Table 5). Although some genetic loci are common to both CDGP and CHH, the genetic architectures of these two conditions are largely distinct (374).

 

Table 5. Genes Associated with Delayed Puberty (338, 482, 483)

Gene

(Reference/s)

Protein encoded

Genetic locus

Associated features/syndromes

                                         SYNDROMIC CAUSES

FGFR1/FGF8

 

(484, 485)

Fibroblast Growth Factor Receptor 1/fibroblast growth factor 8

8p11.23

Hartsfield syndrome

LEPR/LEP

(486, 487, 488)

Leptin receptor and Leptin

1p31.3

Severe obesity syndromes

PCSK1

(489)

Prohormone convertase 1 gene

5q15

Obesity, ACTH deficiency, diabetes

DMXL2

(490)

DmX-like protein 2

15q21

Polyendocrinopathy, Polyneuropathy syndrome

RNF216/

OTUD4

(491)

Ring finger protein 216/ OTU domain-containing protein 4

4q31.21

Gordon Holmes

PNPLA6

(492, 493)

Patatin-like phospholipase domain-containing protein 6

19p13.2

Gordon Holmes, Oliver McFarlane,

Lawrence Moon, Boucher-Neuhauser syndrome

SOX10

(494)

Sex determining region Y-Box transcription factor 10

22q13.1  

Wardenburg syndrome

SOX2

(495)

Sex determining region Y-Box transcription factor 2

3q26.33

Optic nerve hypoplasia, CNS abnormalities

SOX3

(496)

Sex determining region Y-Box transcription factor 3

Xq27.1

Intellectual disability, craniofacial abnormalities, multiple pituitary hormone deficiencies

IGSF1

(497, 498)

Immunoglobulin superfamily member 1

Xq26

Associated with X-linked central hypothyroidism, macro-orchidism

HESX1

(499)

HESX homeobox 1

3p14.3

Hypopituitarism, septo-optic dysplasia

CHD7

(500, 501, 502)

Chromodomain helicase DNA binding protein 7

8q12.2

CHARGE syndrome

POLR3A/

POLR3B

(503, 504, 505)

 

RNA polymerase III

12q23.3

Hypomyelination, hypodontia

NROB1

(DAX-1)

 

(506, 507)

Nuclear Receptor Subfamily 0 Group B Member 1/ dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1

Xp21

Adrenal hypoplasia

REV3L/

PLXND1

(508)

 

Catalytic subunit of DNA polymerase zeta

6q21

Möbius syndrome

PWS

(509, 510)

 

15q11.2

Prader-Willi syndrome

BBS1, BBS2, ARL6, BBS4, BBS5, MKKS, BBS7, TTC8, BBS9, BBS10,

TRIM32, BBS12

(511, 512)

Encoded protein may play a role in eye, limb, cardiac and reproductive system development

11q13.2, 20p12, 16q21, 15q22.3‐23, 14q32.1

(multiple loci)

Bardet-Biedl syndrome

PHF6

(513)

Plant homeodomain (PHD)-like finger protein 6 

Xp26.2

Borjeson‐Forssman‐Lehmann syndrome

SMCHD1

 

(514)

Structural maintenance of chromosomes flexible hinge domain containing 1

18p11.32

Bosma arhinia microphthalmia syndrome

TBC1D20/

RAB18

(459, 515)

TBC1 Domain Family Member 20, GTPase activator proteins of Rab-like small GTPases

 

20p13

Warburg micro syndrome

HDAC8

(516)

 

Histone deacetylase 8

Xq13.1

Cornelia de Lange syndrome

                                          NON-SYNDROMIC CAUSES

FGF17

(517)

Fibroblast Growth Factor 17

8p21.3

 

ANOS1 (KAL1)

(518, 519)

Kallmann syndrome protein, which is now known as Anosmin 1

Xp22.31 

involved in fibroblast growth factor (FGF) signaling

GNRHR/

GNRH1

(520, 521, 522)

 

Gonadotropin-releasing hormone receptor/ gonadotropin-releasing hormone 1

4q13.2

 

KISS1R/

KISS1

(54, 523)

Kisspeptin-1 receptor/ kisspeptin-1

19p13.3

 

 KLB

(524)

Klotho Beta

4p14

Metabolic defects

TAC3/TACR3

(342, 525)

Tachykinin 3, Tachykinin 3 receptor

Encodes neurokinin b

4q24

 

IL17RD

(517)

 

Interleukin 17 Receptor D

3p14.3

 

DUSP6

(517)

 

Dual specificity phosphatase 6

12q22–q23

 

SEMA3A/

SEMA3E/

SEMA7A

(526)

Semaphorin 3A

7q21.11

 

SPRY4

(517)

Sprouty homolog 2

5q31.3

 

FLRT3

(517)

Fibronectin leucine rich transmembrane protein 3

20p11

 

PROKR2/

PROK2

(527, 528)

Prokineticin-2 and Prokineticin receptor 2

3p13

 

WDR11

(529)

 

WD repeat domain 11

10p26.12

 

 

CCDC141

(530)

Coiled-Coil Domain Containing 141

2q31.2

 

FEZF1

(531)

FEZ family zinc finger 1

7q31.32

 

LHB

(532)

Luteinizing hormone

19q13.33

 

FSHB

(533, 534)

Follicle-stimulating hormone

11p14.1

 

AXL

(458)

 

AXL receptor tyrosine kinase

19q13.2

 

EAP1

(535)

Enhanced at puberty 1

14q24

Trans-activates the GnRH promoter

LGR4

(536)

Receptor for R-spondins which, once activated, potentiates the canonical Wnt signaling pathway

11p14.1

 

TUBB3

(483)

Microtubule protein β-III-tubulin

16q24.3

Congenital fibrosis of the extraocular muscles

WDR11, PROP1, PROK2, PROKR2

(529)

Bromodomain and WD repeat-containing protein 2, Homeobox protein 

prophet of PIT-1, prokinectin 2

10q26.12,

5q35.3,

3p13

Combined pituitary hormone deficiency

FTO

(482, 537)

Fat mass and obesity-associated protein

16q12.2

Mice lacking FTO had significantly delay in pubertal onset

 

ACQUIRED HYPOGONADOTROPIC HYPOGONADISM

 

Several conditions are associated with primary gonadal insufficiency. These conditions include auto-immune disorders, trauma, neoplasia, vascular events, and infection. Autoimmune disorders can be associated with premature ovarian and testicular insufficiency. Biallelic mutations in the autoimmune regulator (AIRE) gene are associated with autoimmune polyendocrine syndrome type 1 which is also known as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. Associated features include mucocutaneous candidiasis, hypoparathyroidism, and adrenal insufficiency (375). The detection of autoantibodies directed against tissue-specific antigens suggests an autoimmune diagnosis.   

 

Antineoplastic chemotherapy with alkylating agents, as well as localized ionizing radiation, may permanently damage germ cells leading to infertility. In males, Sertoli cells are more susceptible to such toxicity than Leydig cells such that testosterone production may remain intact despite Sertoli and germ cell injury. Mumps orchitis should be considered, especially in unvaccinated males. Decreased blood flow to the gonads from surgical injury (e.g., orchiopexy in boys), torsion, or trauma can lead to ischemia and atrophy, with resultant primary testicular insufficiency.

 

The presence of otherwise normal male external genitalia associated with nonpalpable gonads indicates that the testes were present and functioning at least early in gestation. The “vanishing testes syndrome” also known as testicular regression is associated with atrophy or regression of testicular tissue initially formed during early embryonic development. Potential etiologies of the testicular regression include in utero vascular disruption or testicular torsion. Pathogenic variants of the DEAH-box RNA helicase DHX37 (DHX37) gene have been identified in boys with testicular regression and in association with gonadal dysgenesis (376).

 

In addition to autoimmune etiologies, premature ovarian insufficiency can be associated with ovarian/pelvic tumors, chemotherapy, especially alkylating agents, and radiation therapy. The location of the pelvic tumor and treatments influence the ovarian reserve and risk for premature ovarian insufficiency. Low or declining serum AMH levels provide an indirect measure of ovarian reserve. However, due to much variability and lack of diagnostic thresholds, measuring AMH values does not accurately predict ovarian insufficiency in cancer survivors (377).

 

Acquired hypogonadotropic hypogonadism (HH) can be due to central nervous system tumors such as craniopharyngiomas and germ cell tumors. Such tumors can disrupt the hypothalamic-pituitary stalk or can impact pituitary function producing decreased gonadotropin production. Hyperprolactinemia due to prolactin-secreting adenomas can cause acquired HH (378). Other central nervous system disorders associated with acquired HH include hypophysitis, histiocytosis, and hemochromatosis. Intracranial surgeries and/or cranial radiation therapy greater than 30 Gy are known risk factors for HH. Moderate to severe trauma to the brain is associated with injuries to the hypothalamus, stalk (infundibulum), or pituitary gland itself; the consequences of traumatic brain injury may not manifest for many years. Chronic steroid treatment can be associated with acquired HH in boys with Duchenne muscular dystrophy (379, 380). Inflammatory and infiltrative diseases of the pituitary gland are other rare causes of acquired HH.

 

FUNCTIONAL HYPOGONADOTROPIC HYPOGONADISM

 

Functional HH is the hypothalamic response to intense physical or emotional stress, caloric deficit, or chronic systemic illness (381). In this situation, the otherwise normal HPG axis fails to function due to the concomitant stress. Puberty can be delayed or stalled until the underlying condition has been adequately addressed. The finding of hypercortisolemia in women with functional HH associated with restrictive eating disorders highlights the relevance of HPA axis function in FHH (382). Importantly, functional HH can have long lasting adverse consequences on bone health (383).

 

The hypothalamus receives numerous inputs regarding body energy status and subsequently modulates reproductive status based on this information. Hence, nutritional status and energy output influence HPG axis activity in part via leptin signaling which regulates the sensitivity of the pituitary to GnRH (384). Energy deficits may occur due to weight loss, excessive energy expenditure (rigorous physical activity, renal disease, cystic fibrosis, congenital heart disease), decreased caloric intake or malabsorption (disordered eating behaviors, bowel disorders such as celiac, Crohn’s, and ulcerative colitis) are associated with delayed or stalled puberty and  functional hypothalamic amenorrhea (385, 386, 387). Elevated circulating levels of cytokines (as seen in some acute or inflammatory conditions) may also inhibit the HPG axis. Elevated prolactin levels, due to prolactinoma or severe primary hypothyroidism may inhibit gonadotropin release.

 

Some boys with obesity have low gonadotropin and testosterone levels and manifest delayed puberty (388). It is important to recognize that certain medications such as antipsychotics (typical and atypical), certain antidepressants, and opioids can alter menses (364). 

 

Treatment of Delayed Puberty

 

A variety of therapeutic regimens for pubertal induction have been described for both boys and girls. However, large, randomized trials providing evidence-based data regarding the optimum regimen are lacking (389). Sex steroid replacement therapy remains a mainstay of treatment. The type and route of administration of the sex steroids is dependent on patient preference, insurance coverage, and health care provider practices. Importantly, the specific treatment regimen depends on the underlying etiology of the pubertal delay. Future novel therapies could include kisspeptin and neurokinin B analogs (390).

 

BOYS

 

Pulsatile GnRH therapy is the most physiological method and can induce adult secondary sex characteristics, achieving normal adult testosterone concentrations, and spermatogenesis (370) in boys with HH. However, the inconvenience of wearing a mini-pump and conflicting outcome data limits its usefulness. Other approaches include hCG, FSH, hMG, and/or GnRH treatments. Despite much heterogeneity, a systematic study reported that treatment with hCG and FSH induced greater increase in testicular volume and rate of spermatogenesis compared to hCG alone (391)370). Importantly, available limited data suggest that testosterone administration prior to gonadotropin treatments does not interfere with the beneficial effects on testicular growth and spermatogenesis. Based on the physiologic roles of LH and FSH, pubertal induction should begin with FSH to promote testicular maturation followed by combined FSH and hCG treatment. The subsequent hCG treatment will promote testicular testosterone secretion leading to virilization, growth spurt, and psychosocial development.

 

Still, at the present time, testosterone is the most established treatment for pubertal induction in boys with delayed puberty. Traditionally, IM testosterone esters, primarily testosterone enanthate or testosterone cypionate, have been used. A subcutaneous testosterone enanthate auto-injector has recently been approved, but this approach requires more weekly injections and is more expensive. However, no evidence-based guidelines exist for testosterone-induced pubertal initiation. Potential adverse consequences of testosterone therapy include erythrocytosis, premature epiphyseal closure especially with excessive doses which may result in aggressive behavior, mood swings, and priapism.

 

Other testosterone formulations include testosterone gels, pills, and pellets. Limitations of testosterone gels include difficulties in accurately titrating low doses, potential testosterone exposure to household members, and the cost. Oral methyltestosterone and its 17α-derivatives have been associated with hepatic dysfunction and should be avoided. Oral testosterone undecanoate was approved by the FDA in 2019 to treat hypogonadal adult men. However, due to its short half-life, multiple daily doses are necessary, and no data are available regarding use for puberty induction. Testosterone pellets require surgical placement every 3-4 months, are expensive, and often spontaneously extrude (392).

 

For the younger adolescent boy with a strong family history of CDGP, reassurance and continued clinical monitoring may be adequate. However, discerning CDGP from CHH is essential because the treatment, genetics, and psychosocial implications differ. Hence, low dose testosterone for 3-4 months followed by a similar period of observation may be helpful to distinguish CDGP from CHH. Individuals with CHH will show persistently low gonadotropin and sex steroid hormone levels after the 3–4-month period of observation whereas individuals with CDGP will usually show spontaneous pubertal progression. Curiously, testosterone exposure apparently activates GnRH production and secretion leading to “reversal” with onset of HPG axis activity in some boys with CHH. This reversal is associated with specific genetic variants and may be transient (393, 394).

 

GIRLS

 

Timely induction of pubertal development is fundamental. Two major goals of estrogen therapy are mimicking typical pubertal progression with breast development and promoting adequate uterine growth (395). Although pulsatile GnRH treatment can be used, this approach has no advantage over estrogen for pubertal induction in girls. Though all therapeutic approaches utilize estrogens, details regarding specific formulations and methods of administration vary. Transdermal estradiol is preferred for replacement therapy because this approach avoids the first pass through the liver and the potential for adverse effects on clotting factors.

 

Typically, low transdermal estradiol doses are used for the initial phase of pubertal induction. Transdermal estradiol doses of 3-7 mcg/day can be achieved by cutting matrix patches (0.014-0.025 mg/24 h) into quarters or eighths. Subsequently, the dose can be increased approximately every six months until adult replacement dosage is achieved taking about 24-36 months to do so. High initial estrogen doses should be avoided due to increased likelihood for atypical breast development characterized by prominent nipples with little supporting breast tissue. High estrogen doses should also be avoided as premature epiphyseal fusion could impair additional linear growth.

 

Oral micronized 17β-estradiol can be used for those with severe skin irritation or aversion to the use of a patch. Oral preparations containing conjugated equine estrogens or ethinyl estradiol should be avoided for both pubertal induction and maintenance therapy. Most combined oral contraceptives contain ethinyl estradiol at doses higher than appropriate for induction of puberty. Approximately 18-24 months after initiation of unopposed estrogen therapy, progestogens can be added to induce withdrawal bleeding and to reduce the risk for endometrial hyperplasia. Progestogens can be introduced earlier if breakthrough vaginal bleeding occurs. Pelvic ultrasounds before and during pubertal induction can be planned to assess uterine size and shape as well as to evaluate endometrial thickness to ascertain optimal timing to introduce progestins. Progestins vary in potency and can be administered by transdermal, oral, or uterine routes. Although increased potency may have beneficial effects on withdrawal bleeding, greater progestogenic side effects may develop.

 

No evidence-based data exist, and no single regimen has been demonstrated to be superior. Pubertal induction therapy should be individualized based on clinical response and other auxologic parameters.

 

Oral contraceptive pills may be used for convenience but should be limited to after completion of pubertal development. Since some girls may experience sporadic ovulation, contraception should be utilized by those at risk of undesired pregnancies.

 

EVALUATION OF A CHILD WITH A VARIATION IN PUBERTAL DEVELOPMENT

 

The diagnostic tools are comparable for the evaluation of either precocious or delayed puberty. Detailed medical history and physical examination provide the preliminary information to guide the differential diagnosis for a child with a variation in pubertal development (165, 396) (see Figures 5-8). Laboratory, imaging, and genetic studies are subsequently utilized to ascertain the specific diagnosis. The tools for evaluation of a child with a variation in pubertal development are described below. The tools are comparable, but the interpretation of test results differs for precocious and delayed puberty.

 

Figure 5. Algorithm to evaluate a girl presenting with precocious puberty. *follow clinical progression every 3-6 months. FSH: Follicle Stimulating Hormone; LH: Luteinizing Hormone; CAH: Congenital Adrenal Hyperplasia; ACTH: Adrenocorticotrophic Hormone; GnRH: Gonadotropin Releasing Hormone; DHEAS: Dehydroepiandrosterone Sulfate; 17OHP: 17-hydroxy progesterone; NF-1: Neurofibromatosis-1.

Figure 6. Algorithm to evaluate a boy presenting with precocious puberty. FSH: Follicle Stimulating Hormone; LH: Luteinizing Hormone; MRI: Magnetic Resonance Imaging.

Figure 7. Algorithm to evaluate a girl presenting with delayed puberty. FSH: Follicle Stimulating Hormone; LH: Luteinizing Hormone; CDGP: Constitutional Delay in Growth and Puberty; CHH: Congenital Hypogonadotropic Hypogonadism; MRI: Magnetic Resonance Imaging.

Figure 8. Algorithm to evaluate a boy presenting with delayed puberty. FSH: Follicle Stimulating Hormone; LH: Luteinizing Hormone; CDGP: Constitutional Delay in Growth and Puberty; CHH: Congenital Hypogonadotropic Hypogonadism; MRI: Magnetic Resonance Imaging.

 

History and Physical Examination

 

The medical history focuses on the timing and sequence of the pubertal changes in the patient as well as parents, grandparents, and siblings. Review of past medical history and medications (including chemotherapy and nutritional supplements) is essential. Inquiry regarding exposures (tea tree/lavender oils, sex steroids, radiation) may help to identify potential environmental factors (162). Obtaining birth history, length and weight, history of SGA (397), prematurity, or CNS insult at birth or later provide relevant information (162).

 

A history of gelastic seizures may point to a hypothalamic hamartoma. Inquiry regarding use of transdermal testosterone by a family member may identify the cause of premature virilization. Pubertal delay associated with micropenis, anosmia, cryptorchidism, deafness, choanal atresia, hearing loss, and/or digital abnormalities suggests congenital hypogonadotropic hypogonadism (CHH). A family history of anosmia, subfertility, and deafness should be sought for those with pubertal delay. Multiple syndromes are associated with CHH (see Table 3); suggestive features include absent/reduced sense of smell, choanal atresia, hearing loss, morbid obesity, visual impairment. Family history of precocious or delayed puberty in close relatives may be discovered (398). Behavioral difficulties or learning disabilities may be associated with specific syndromes such as Turner or Klinefelter syndromes.

 

A complete physical examination including height, weight, arm span, and sitting height is essential. Review of the child’s growth curves provides valuable information regarding changes in linear growth and weight gain. Acceleration in linear growth and upward crossing of centiles may be seen in precocious puberty. A gradual downward crossing of centiles may be noted in constitutional delay in growth and puberty (CDGP) as linear growth slows compared to peers who are entering puberty (399). Pubic hair development (pubarche) may also be delayed in CDGP as opposed to CHH where adrenarche occurs at the normal age for population (372).

 

Physical exam includes ascertainment of the sexual maturity rating for breast, pubic hair, and testicular volume based on the scoring system derived by Tanner and colleagues (Figure 1). Due to challenges in discriminating lipomastia from true glandular breast development, palpation of the breasts is important. Firm glandular tissue under the areolae is indicative of thelarche. Accurate measurement of testicular volume using an orchidometer is essential (see Figure 2). A testicular volume of ≤ 1.1 mL has a reported sensitivity and specificity of 100% and 91%, respectively, for CHH (400).

 

The physical examination should assess for midline defects, dysmorphic features, visual field abnormalities, and features characteristic for specific syndrome. For example, short stature, cubitus valgus, low hair line, widely spaced nipples, and delayed puberty suggest Turner Syndrome. The physical examination needs to include palpation of the thyroid gland, skin examination for acne or café-au-lait macules (which would suggest neurofibromatosis or McCune-Albright syndrome) and a visual field exam. Melanocytic macules typical of Peutz-Jeghers syndrome could point to the presence of a sex cord tumor causing gonadotropin independent (peripheral) sexual precocity.

 

Laboratory Evaluation

 

Laboratory evaluation assists the diagnostic process to identify the etiology of “off-time puberty.” Circulating gonadotropin and sex steroid concentrations reflect HPG axis status (187, 236). Most current gonadotropin assays are sandwich assays specific to the β-subunit. Ultrasensitive FSH and LH assays should be used when available. For LH, samples should preferably be obtained in the morning. The lower limit of detection for most ultrasensitive immunochemiluminescent assays (ICMA) is ≤0.1 mIU/mL (230, 401, 402).

 

When the clinical concern is precocious puberty, LH concentrations greater than 0.3-0.5 mIU/mL suggest central precocious puberty (CPP) with higher cut-points increasing the sensitivity and specificity of the LH determination (403). Elevated basal LH levels show high sensitivity and specificity for boys when high quality immunochemiluminometric assays (ICMA) is used (404). Different cut-points need to be used to interpret LH concentrations in girls under two years of age because LH concentrations may be elevated at this age leading to misdiagnosis of CPP followed by inappropriate treatment during this phase of development (405). For the child with physical signs of premature puberty, LH concentrations in the prepubertal range are consistent with either peripheral precocity or a benign pubertal variant such as premature thelarche. Typically, LH and FSH concentrations are suppressed in children with peripheral precocious puberty (406).

 

In the evaluation for delayed puberty, low gonadotropin concentrations suggest a central etiology such as CDGP or hypogonadotropic hypogonadism while elevated gonadotropin concentrations suggest primary gonadal insufficiency. Random gonadotropin concentrations may provide only limited information because gonadotropin secretion is pulsatile. Distinguishing hypogonadotropic hypogonadism from CDGP is often challenging because LH, FSH and sex hormone reference intervals vary widely even in healthy adolescents (407). Similarly, due to significant overlap in hormone reference intervals, GnRH agonist and human chorionic gonadotropin (hCG) stimulated gonadotropin (408) and sex steroid concentrations fail to distinguish youth with CHH from those with CDGP (407, 409).

 

Due to the small structural differences between steroid molecules, immunoassays are confounded by cross-reactivity issues. Assay issues are amplified in children because commercial immunoassays for estradiol and testosterone are usually designed to measure hormone concentrations within the normal adult reference interval. Hence, most estradiol immunoassays have low sensitivity and specificity to quantify the low concentrations (< 30 pg/ml) typically found in prepubertal children and individuals with hypogonadism. Similar issues occur with testosterone immunoassays. Hence, steroid hormone concentrations should be measured by liquid chromatographic separation followed by mass spectrometry (LC-MS/MS). Serum testosterone is best measured using LC-MS/MS technology to limit cross-reactivity and increase sensitivity and specificity especially when low hormone concentrations might be anticipated. LC-MS/MS is also the optimal technique to measure circulating concentrations of other steroids including 17-hydroxyprogesterone, DHEA, androstenedione, and the 11-oxy androgens. It offers greater sensitivity and specificity and allows simultaneous measurement of multiple hormone concentrations (410).

 

Sex steroids such as estradiol and testosterone circulate bound to sex hormone binding globulin (SHBG). Tissue availability of the free hormone, presumed to be the active moiety, is regulated by SHBG. Direct free testosterone concentrations should be avoided because direct immunoassays have poor reproducibility and reliability. When free testosterone concentrations need to be determined, equilibrium dialysis should be performed despite known potential limitations including increased expense, reliance on total testosterone accuracy, temperature control, and sample dilution (411).

 

Another confounding factor is biotin (vitamin B7) which is an over-the counter supplement by itself or as an addition to many preparations used to strengthen nails and hair. Biotin interferes with the technical aspects of immunoassays and can lead to either falsely elevated or falsely low result when streptavidin binding is utilized in the assay detection system. When immunoassay results seem incongruous, use of biotin-containing products should be queried. Biotin does not interfere with LC-MS/MS assays (412).

 

GnRH or GnRH AGONIST STIMULATION TEST

 

Historically, the established gold standard to diagnosis CPP was the LH and FSH response to a standard bolus of native GnRH. With decreased availability of native GnRH, most stimulation tests are now performed with the GnRH agonist (GnRHa) leuprolide acetate, a synthetic nonapeptide with much greater potency. The timing and peak values of FSH and LH levels differ between GnRH and leuprolide acetate. Following native GnRH administration, LH levels peak after 20–40 minutes, followed by a decline. With leuprolide acetate, peak LH occurs between 0.5 - 4 hours followed by sustained LH elevation.

 

The optimal cutoff value of peak stimulated LH for identifying children with CPP is unclear due to assay variability. For most LH assays, a value of 3.3 to 5 mIU/mL defines the upper limit of normal for stimulated LH values in prepubertal children. Stimulated LH concentrations above this range suggest CPP (232). Children with progressive CPP tend to have a high stimulated LH:FSH ratios compared with those with non- or intermittently progressive precocious puberty. Measuring the appropriate sex steroid 24 hours following GnRHa administration can help confirm a CPP diagnosis (413). However, obtaining this second sample may burden the family because of the need for a second venipuncture, expense of another hormone determination, and missed school and work.

 

As noted above regarding basal LH levels, care must be taken in interpreting the results of GnRH stimulation test in females under the age of two years, as both basal and stimulated LH levels can be elevated as part of the normal hormonal changes associated with mini-puberty (405).

 

To assess GnRH production by the hypothalamus, kisspeptin-stimulated LH response has been proposed to identify individuals with GnRH deficiency and thus CHH. Kisspeptin stimulates GnRH secretion, thus promoting LH, and to a lesser extent FSH, secretion.

 

One study found that maximal LH rise after kisspeptin administration was more accurate for diagnosis of men with GnRH deficiency than GnRH stimulation testing (414). A similar study in adolescents with pubertal delay (3 females and 13 males), peak LH post kisspeptin stimulation was demonstrated to be superior to GnRH stimulation testing for predicting capacity to progress through puberty (noting that the LH cut off values were different and an ideal cutoff value still needs to be determined) (346). Further research is required to better define the parameters of using kisspeptin stimulation in clinical practice (404).

 

In children with precocious pubarche, measurement of adrenal steroids may be necessary to help distinguish between peripheral precocity and benign premature adrenarche. Children with premature adrenarche can have mild elevation in adrenal hormones (415). Since premature adrenarche is a diagnosis of exclusion, further investigation for congenital adrenal hyperplasia and virilizing adrenal tumors may be indicated. In children, an early-morning 17-hydroxyprogesterone (17-OHP) value >200 ng/dL (6 nmol/L) has a high sensitivity and specificity for non-classic congenital adrenal hyperplasia secondary to 21-hydroxylase deficiency. An adrenocorticotropic hormone (ACTH) stimulation test is needed to confirm the diagnosis (313, 416). An ACTH stimulation test involves administration of 0.25 mg synthetic ACTH (1-24) or 15 mcg/kg for children up to 2 years of age with blood samples obtained at baseline and either 30 or 60 minutes after synthetic ACTH administration. Although 21-hydroxylase deficiency is the most common virilizing form of CAH, 17-hydroxypregnenolone, DHEA, and 11-deoxycortisol determinations may be necessary to assess for 3β-hydroxysteroid dehydrogenase or 11β-hydroxylase deficiencies.

 

Boys with hypogonadotropic hypogonadism tend to have lower inhibin B values compared to boys with CDGP. However, a validated cut-point for inhibin B concentrations remains to be established (417, 418, 419). FSH stimulated inhibin B concentrations < 116 pmol/L have been demonstrated in a study of adolescents with delayed puberty to have more accurate diagnostic discrimination and a promising test for prediction of onset of puberty (414).

 

Human chorionic gonadotropin concentrations can be measured in males to evaluate for the possibility of an hCG-secreting tumor leading to peripheral precocity (280). A thyroid-stimulating hormone (TSH) concentration should be measured if chronic primary hypothyroidism is suspected as the underlying cause for the sexual precocity, known as the Van-Wyk-Grumbach syndrome (296, 298) .

 

Targeted diagnostic tests are warranted in some cases to investigate for specific causes of apparent functional hypogonadotropic hypogonadism, such as anti-transglutaminase IgA for celiac disease. Despite promising data, measurement of AMH and INSL3 in addition to testosterone, as endocrine markers to guide the differential diagnosis (418, 420), need additional studies  

 

The testosterone response to long-term hCG stimulation and peak serum FSH response to GnRH were found to be significantly different in CHH patients (421). However, there are potential long-term drawbacks to prolonged hCG therapy in males who are FSH-naïve regarding premature stimulation of Sertoli and germ cell differentiation prior to FSH exposure (338). 

 

Imaging Studies  

 

BONE AGE

 

Assessing the skeletal maturation based on a radiograph of the left hand and wrist is an important diagnostic tool in pubertal evaluation. For the commonly utilized Greulich and Pyle method, the patient’s bone age radiograph is compared with an atlas of radiographs from children of known ages (422). For the Tanner-Whitehouse 2 method, 20 different hand and wrist bones are scored. Bone age standards are largely based on hand and wrist radiographs obtained from children of European ancestry between the 1930s to the 60s (423). Despite this limitation, the bone age radiograph is a valuable indicator regarding sex steroid exposure and epiphysial (growth plate) maturation. Additional factors such as other hormones, obesity, genetics, nutritional status, various disease states, and certain medications can influence the rate of epiphyseal maturation (424) (425).

 

Bone age has been used to predict adult height using the tables of Bayley and Pinneau (426), but reliability is low with a tendency toward overestimation (427). The use of automated measurement systems with artificial intelligence has increased, mitigating previous limitations due to intra- and inter-observer variability (428, 429). Bone age readings within two standard deviations of the chronologic age are considered to be within normal limits. A delayed bone age is usually observed in patients with delayed puberty and an advanced bone age is observed with precocious puberty. One exception is patients with precocious puberty associated with hypothyroidism (Van Wyk Grumbach syndrome) where the bone age is delayed despite pubertal changes. In some instances, monitoring the predicted adult height (PAH) during the course of treatment of pubertal disorders helps to assess treatment efficacy.

 

ULTRASOUND IMAGING

 

In females, pelvic ultrasound is a rapid, non-invasive, and relatively low-cost method to ascertain the anatomy of the ovaries and uterus, ovarian volume, and uterine development. This imaging is generally readily accessible and does not require sedation, radiation, or use of contrast material. However, the quality of the device and operator experience influence the analysis.

 

During puberty, increased gonadotropin secretion promotes ovarian growth, increased estradiol secretion, and increased uterine volume (430). Girls with CPP have increased uterine size and ovarian volumes compared to prepubertal girls or those with premature thelarche. However, the overlap between prepubertal and early pubertal girls for ovarian volume and uterine size confounds interpretation of the ultrasound findings (431) (432). In a prepubertal patient with isolated vaginal bleeding, a normal pelvic ultrasound does not exclude the diagnosis of a functional ovarian cyst because the cyst may have regressed prior to imaging. Pelvic ultrasounds should be obtained in girls with primary amenorrhea who fail progesterone withdrawal to assess for Mullerian duct and renal anomalies. For patients with rapid development of secondary sex characteristics, pelvic ultrasound studies may be needed to assess for gonadal tumors.

 

The use of Doppler ultrasound to assess utero-ovarian blood flow may also provide helpful information. With increased estradiol secretion and stimulation of the estrogen receptors, vascular resistance of the uterine arteries is reduced. The pulsatility index (PI) is defined as the difference between peak systolic flow and end-diastolic flow divided by the mean flow velocity; the PI reflects impedance to blood flow distal to the sampling point. A review showed that PI is lower among pubertal girls. However, definitive cut-points for PI values have not been established. In addition, testing is operator dependent (433, 434, 435, 436).

 

Ultrasound examination of the testes, especially if asymmetric in size, should be performed in males with peripheral precocity to evaluate for the possibility of a Leydig cell tumor (437, 438). Testicular ultrasound imaging should be performed regularly to assess for testicular rest tissue in boys with congenital adrenal hyperplasia (439).

 

MR AND CT IMAGING

 

Brain MR or CT imaging is performed to define brain and pituitary anatomy. Brain and pituitary MR is helpful to assess for intracranial pathology among those with CNS symptoms. Most studies recommend a contrast-enhanced brain MRI for girls with onset of secondary sexual characteristics before six years of age because of higher rates of CNS abnormalities in these patients (137). In a 2018 meta-analysis (440), the prevalence of intracranial lesions was 3 percent among girls presenting with CPP after six years of age, compared with 25 percent among those presenting before six years. Thus, girls with pubertal onset between six and eight years of age may not need the MRI in the absence of clinical evidence of CNS pathology (441, 442, 443). MRI should be limited to high-risk individuals (younger age, neurologic symptoms) (444). Current guidelines recommend that in otherwise asymptomatic girls with CPP, a discussion occur with the parents regarding the pros and cons of brain imaging and assist in informed decision making (137, 445, 446). While contrast-enhanced brain MRIs are recommended for all boys presenting with CPP (412), one study found that these rates may be overestimated. The prevalence of intracranial lesions among boys who were healthy, did not have neurological symptoms, and were diagnosed with CPP was lower than that previously reported and none of the identified lesions necessitated treatment, suggesting the need to globally reevaluate the prevalence of pathological brain lesions among boys with CPP (447).

 

For children with delayed puberty, MR imaging of the pituitary gland and olfactory structures can assess for features of CHH such as absence of the olfactory bulbs (448, 449, 450).

 

Pelvic MRI is helpful to characterize and stage pediatric ovarian masses due to excellent soft tissue contrast. In addition, MR imaging does not involve the use of ionizing radiation and allows better assessment of the abdomen and kidneys. Disadvantages of MRI include that it is time-consuming, expensive, and may require sedation.

 

In both girls and boys, adrenal tumors can cause peripheral precocious puberty, progressive virilization, and/or markedly elevated serum adrenal androgens (e.g., DHEAS). If diagnoses such as congenital adrenal hyperplasia and exogenous androgen or testosterone exposure have been excluded, such patients should have an imaging study of the adrenal glands (451, 452, 453). CT may be preferable for evaluation, staging, surgical planning for adrenal tumors (454). Despite radiation exposure, CT can be readily performed in emergent situations.

 

Genetic Testing  

 

A karyotype can help with a diagnosis of Turner or Klinefelter syndrome (455).Newer sequencing technologies along with increased knowledge regarding genes involved in puberty has advanced the usefulness of genetic testing (456). The known genetic causes of CPP and HH have increased exponentially over the past five years. Genetic testing could therefore precede brain MRI, at least in familial CPP cases (167, 457).

 

Patients with delayed puberty associated with phenotypic features such as anosmia/hyposmia, synkinesia, or hearing loss, the probability of detecting a pathogenic variant on genetic testing for HH is increased (458) (459, 460). Consideration should be given to using genetic testing early in the diagnostic process while recognizing the limitations of genetic testing. Challenges in using genetic testing as a discriminatory test between CHH and CDGP remain, and more research is needed in this area.

 

SUMMARY

 

Pubertal development and maturation of the neuroendocrine system involve the ontogeny, activity, and interactions of the GnRH neurons. Pubertal onset is accompanied by an increase in kisspeptin and neurokinin B secretion regulating the pulsatile GnRH secretion that stimulates pulsatile pituitary LH and FSH secretion. LH and FSH stimulate gonadal sex steroid secretion promoting development of secondary sex characteristics and influencing hypothalamic-pituitary function via negative feedback inhibition.

 

Alterations of gut microbiome at different pubertal stages may present an area for future development in the prediction and prevention of precocious puberty. Use of genetic testing including targeted next generation sequencing and whole exome sequencing may have increasing utility as diagnostic tools early on in the evaluation of pubertal disorders.

 

Discovery regarding the details of normal reproductive physiology followed by identification of the genetic basis for disorders of pubertal timing established our current knowledge base for the evaluation and management of children with disorders affecting the timing of puberty. Despite the vast expansion of our knowledge, much remains to be learned about the physiology and regulation of the HPG axis from the fetus to the young adult.

 

REFERENCES

 

  1. Springfield: Mass, G. & C. Merriam Co; 1961. Webster’s third new international dictionary of the English language, unabridged.
  2. Argente J, Dunkel L, Kaiser UB, Latronico AC, Lomniczi A, Soriano-Guillen L, et al. Molecular basis of normal and pathological puberty: from basic mechanisms to clinical implications. Lancet Diabetes Endocrinol. 2023;11(3):203-16.
  3. Abreu AP. Unveiling the central regulation of pubertal development. J Clin Endocrinol Metab. 2023.
  4. Reiter EO, Fuldauer VG, Root AW. Secretion of the adrenal androgen, dehydroepiandrosterone sulfate, during normal infancy, childhood, and adolescence, in sick infants, and in children with endocrinologic abnormalities. J Pediatr. 1977;90(5):766-70.
  5. Witchel SF BA, Oberfield SE. Adrenal Androgens, Adrenarche and Adrenopause. 8th Edition ed. Robertson RP GL, Grossman A, Hammer GD, Jensen MD, Kahaly GJ, Swerdloff R, Thakker RV (eds), editor: Elsevier; 2021.
  6. Rege J, Turcu AF, Kasa-Vubu JZ, Lerario AM, Auchus GC, Auchus RJ, et al. 11-Ketotestosterone Is the Dominant Circulating Bioactive Androgen During Normal and Premature Adrenarche. J Clin Endocrinol Metab. 2018;103(12):4589-98.
  7. Counts DR, Pescovitz OH, Barnes KM, Hench KD, Chrousos GP, Sherins RJ, et al. Dissociation of adrenarche and gonadarche in precocious puberty and in isolated hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 1987;64(6):1174-8.
  8. Conley AJ, Moeller BC, Nguyen AD, Stanley SD, Plant TM, Abbott DH. Defining adrenarche in the rhesus macaque (Macaca mulatta), a non-human primate model for adrenal androgen secretion. Mol Cell Endocrinol. 2011;336(1-2):110-6.
  9. Cutler GB, Jr., Glenn M, Bush M, Hodgen GD, Graham CE, Loriaux DL. Adrenarche: a survey of rodents, domestic animals, and primates. Endocrinology. 1978;103(6):2112-8.
  10. Cumberland AL, Hirst JJ, Badoer E, Wudy SA, Greaves RF, Zacharin M, et al. The Enigma of the Adrenarche: Identifying the Early Life Mechanisms and Possible Role in Postnatal Brain Development. Int J Mol Sci. 2021;22(9).
  11. Remer T, Boye KR, Hartmann MF, Wudy SA. Urinary markers of adrenarche: reference values in healthy subjects, aged 3-18 years. J Clin Endocrinol Metab. 2005;90(4):2015-21.
  12. Marshall WA, Tanner JM. Variations in pattern of pubertal changes in girls. Arch Dis Child. 1969;44(235):291-303.
  13. Marshall WA, Tanner JM. Variations in the pattern of pubertal changes in boys. Arch Dis Child. 1970;45(239):13-23.
  14. Fuqua JS, Eugster EA. History of Puberty: Normal and Precocious. Horm Res Paediatr. 2022;95(6):568-78.
  15. Bruserud IS, Roelants M, Oehme NHB, Madsen A, Eide GE, Bjerknes R, et al. References for Ultrasound Staging of Breast Maturation, Tanner Breast Staging, Pubic Hair, and Menarche in Norwegian Girls. J Clin Endocrinol Metab. 2020;105(5):1599-607.
  16. Pedersen JL, Nysom K, Jorgensen M, Nielsen CT, Muller J, Keiding N, et al. Spermaturia and puberty. Arch Dis Child. 1993;69(3):384-7.
  17. Mieritz MG, Raket LL, Hagen CP, Nielsen JE, Talman ML, Petersen JH, et al. A Longitudinal Study of Growth, Sex Steroids, and IGF-1 in Boys With Physiological Gynecomastia. J Clin Endocrinol Metab. 2015;100(10):3752-9.
  18. David K DE, Freud J, Laqueur E. Crystalline male hormone from the testes (Testosterone) is more effective than androsterone derived from urine or cholesterin. Hoppe-Seyler Z Physiol Chem. 1935:233:81–2.
  19. Butenandt A HG. Umwandlung des Dehydroandrosterons in Androstendiol und Testosteron; ein Weg zur Darstellung des Testosterons aus Cholesterin. Hoppe-Seyler Z Physiol Chem. 1935:237:89–98.
  20. Ruzicka L WA. Synthetische Darstellung des Testikelhormons Testosteron (Androsten 3-on-17-ol). Helv Chim Acta. 1935:18:1264–75.
  21. Doisy EA TS, Veler CD The Crystals of the Follicular Ovarian Hormone. Proceedings of the Society for Experimental Biology and Medicine. 1930(27(5):417-419).
  22. Maccorquodale DW, Thayer SA, Doisy EA. The Crystalline Ovarian Follicular Hormone. Proceedings of the Society for Experimental Biology and Medicine. 1935;32:1182 -
  23. Smith PE. Ablation and transplantation of the hypophysis in the rat. Anat Rec. 1926;32(221):15-6.
  24. Zondek B. Uber die hormone des hypophysenvorderlappes. I. Wachstums Hormon, follikelreifungshormon (Prolan A), Luteinisierungshormon (Prolan B) Stoffwechselhormon. Klin Wochschr. 1930(9(6):245–8 ).
  25. H. L. Fevold FLH, and S. L. Leonard. THE GONAD STIMULATING AND THE LUTEINIZING HORMONES OF THE ANTERIOR LOBE OF THE HYPOPHESI. The American Journal of Physiology. 1931:97.2.291.
  26. Harris GW. The Induction of Ovulation in the Rabbit, by Electrical Stimulation of the Hypothalamo-hypophysial Mechanism. Proceedings of The Royal Society B: Biological Sciences. 1937;122:374-94.
  27. Schally AV, Arimura A, Kastin AJ, Matsuo H, Baba Y, Redding TW, et al. Gonadotropin-releasing hormone: one polypeptide regulates secretion of luteinizing and follicle-stimulating hormones. Science. 1971;173(4001):1036-8.
  28. Belchetz PE, Plant TM, Nakai Y, Keogh EJ, Knobil E. Hypophysial responses to continuous and intermittent delivery of hypopthalamic gonadotropin-releasing hormone. Science. 1978;202(4368):631-3.
  29. Burgus R, Butcher M, Ling N, Monahan M, Rivier J, Fellows R, et al. [Molecular structure of the hypothalamic factor (LRF) of ovine origin monitoring the secretion of pituitary gonadotropic hormone of luteinization (LH)]. C R Acad Hebd Seances Acad Sci D. 1971;273(18):1611-3.
  30. Plant TM. Recognition that sustained pituitary gonadotropin secretion requires pulsatile GnRH stimulation: a Pittsburgh Saga. F S Rep. 2023;4(2 Suppl):3-7.
  31. Plant TM, Steiner RA. The fifty years following the discovery of gonadotropin-releasing hormone. J Neuroendocrinol. 2022;34(5):e13141.
  32. Kaplan SL, Grumbach MM. The ontogenesis of human foetal hormones. II. Luteinizing hormone (LH) and follicle stimulating hormone (FSH). Acta Endocrinol (Copenh). 1976;81(4):808-29.
  33. Winter JS, Faiman C, Hobson WC, Prasad AV, Reyes FI. Pituitary-gonadal relations in infancy. I. Patterns of serum gonadotropin concentrations from birth to four years of age in man and chimpanzee. J Clin Endocrinol Metab. 1975;40(4):545-51.
  34. Casoni F, Malone SA, Belle M, Luzzati F, Collier F, Allet C, et al. Development of the neurons controlling fertility in humans: new insights from 3D imaging and transparent fetal brains. Development. 2016;143(21):3969-81.
  35. Taroc EZM, Naik AS, Lin JM, Peterson NB, Keefe DL, Jr., Genis E, et al. Gli3 Regulates Vomeronasal Neurogenesis, Olfactory Ensheathing Cell Formation, and GnRH-1 Neuronal Migration. J Neurosci. 2020;40(2):311-26.
  36. Duittoz AH, Forni PE, Giacobini P, Golan M, Mollard P, Negron AL, et al. Development of the gonadotropin-releasing hormone system. J Neuroendocrinol. 2022;34(5):e13087.
  37. Fueshko SM, Key S, Wray S. GABA inhibits migration of luteinizing hormone-releasing hormone neurons in embryonic olfactory explants. J Neurosci. 1998;18(7):2560-9.
  38. Lund C, Pulli K, Yellapragada V, Giacobini P, Lundin K, Vuoristo S, et al. Development of Gonadotropin-Releasing Hormone-Secreting Neurons from Human Pluripotent Stem Cells. Stem Cell Reports. 2016;7(2):149-57.
  39. Yellapragada V, Eskici N, Wang Y, Madhusudan S, Vaaralahti K, Tuuri T, et al. FGF8-FGFR1 signaling regulates human GnRH neuron differentiation in a time- and dose-dependent manner. Dis Model Mech. 2022;15(8).
  40. Wang Y, Madhusudan S, Cotellessa L, Kvist J, Eskici N, Yellapragada V, et al. Deciphering the Transcriptional Landscape of Human Pluripotent Stem Cell-Derived GnRH Neurons: The Role of Wnt Signaling in Patterning the Neural Fate. Stem Cells. 2022;40(12):1107-21.
  41. Seeburg PH, Adelman JP. Characterization of cDNA for precursor of human luteinizing hormone releasing hormone. Nature. 1984;311(5987):666-8.
  42. Hagen C, McNeilly AS. The gonadotrophins and their subunits in foetal pituitary glands and circulation. J Steroid Biochem. 1977;8(5):537-44.
  43. Clements JA, Reyes FI, Winter JS, Faiman C. Studies on human sexual development. III. Fetal pituitary and serum, and amniotic fluid concentrations of LH, CG, and FSH. J Clin Endocrinol Metab. 1976;42(1):9-19.
  44. Guimiot F, Chevrier L, Dreux S, Chevenne D, Caraty A, Delezoide AL, et al. Negative fetal FSH/LH regulation in late pregnancy is associated with declined kisspeptin/KISS1R expression in the tuberal hypothalamus. J Clin Endocrinol Metab. 2012;97(12):E2221-9.
  45. Kaplan SL, Grumbach MM. Pituitary and placental gonadotrophins and sex steroids in the human and sub-human primate fetus. Clin Endocrinol Metab. 1978;7(3):487-511.
  46. Althumairy D, Zhang X, Baez N, Barisas G, Roess DA, Bousfield GR, et al. Glycoprotein G-protein Coupled Receptors in Disease: Luteinizing Hormone Receptors and Follicle Stimulating Hormone Receptors. Diseases. 2020;8(3).
  47. Andersson AM, Skakkebaek NE. Serum inhibin B levels during male childhood and puberty. Mol Cell Endocrinol. 2001;180(1-2):103-7.
  48. Vale W, Rivier J, Vaughan J, McClintock R, Corrigan A, Woo W, et al. Purification and characterization of an FSH releasing protein from porcine ovarian follicular fluid. Nature. 1986;321(6072):776-9.
  49. Wijayarathna R, de Kretser DM. Activins in reproductive biology and beyond. Hum Reprod Update. 2016;22(3):342-57.
  50. Abbara A, Koysombat K, Phylactou M, Eng PC, Clarke S, Comninos AN, et al. Insulin-like peptide 3 (INSL3) in congenital hypogonadotrophic hypogonadism (CHH) in boys with delayed puberty and adult men. Front Endocrinol (Lausanne). 2022;13:1076984.
  51. Crowley WF, Jr., McArthur JW. Simulation of the normal menstrual cycle in Kallman's syndrome by pulsatile administration of luteinizing hormone-releasing hormone (LHRH). J Clin Endocrinol Metab. 1980;51(1):173-5.
  52. Wildt L, Marshall G, Knobil E. Experimental induction of puberty in the infantile female rhesus monkey. Science. 1980;207(4437):1373-5.
  53. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Jr., Shagoury JK, et al. The GPR54 gene as a regulator of puberty. N Engl J Med. 2003;349(17):1614-27.
  54. de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A. 2003;100(19):10972-6.
  55. Plant TM. The neurobiological mechanism underlying hypothalamic GnRH pulse generation: the role of kisspeptin neurons in the arcuate nucleus. F1000Res. 2019;8.
  56. Herbison AE. The Gonadotropin-Releasing Hormone Pulse Generator. Endocrinology. 2018;159(11):3723-36.
  57. Cheng G, Coolen LM, Padmanabhan V, Goodman RL, Lehman MN. The kisspeptin/neurokinin B/dynorphin (KNDy) cell population of the arcuate nucleus: sex differences and effects of prenatal testosterone in sheep. Endocrinology. 2010;151(1):301-11.
  58. Nagae M, Uenoyama Y, Okamoto S, Tsuchida H, Ikegami K, Goto T, et al. Direct evidence that KNDy neurons maintain gonadotropin pulses and folliculogenesis as the GnRH pulse generator. Proc Natl Acad Sci U S A. 2021;118(5).
  59. Abreu AP, Dauber A, Macedo DB, Noel SD, Brito VN, Gill JC, et al. Central precocious puberty caused by mutations in the imprinted gene MKRN3. N Engl J Med. 2013;368(26):2467-75.
  60. Busch AS, Hagen CP, Almstrup K, Juul A. Circulating MKRN3 Levels Decline During Puberty in Healthy Boys. J Clin Endocrinol Metab. 2016;101(6):2588-93.
  61. Hagen CP, Sorensen K, Mieritz MG, Johannsen TH, Almstrup K, Juul A. Circulating MKRN3 levels decline prior to pubertal onset and through puberty: a longitudinal study of healthy girls. J Clin Endocrinol Metab. 2015;100(5):1920-6.
  62. Varimo T, Dunkel L, Vaaralahti K, Miettinen PJ, Hero M, Raivio T. Circulating makorin ring finger protein 3 levels decline in boys before the clinical onset of puberty. Eur J Endocrinol. 2016;174(6):785-90.
  63. Palumbo S, Cirillo G, Aiello F, Papparella A, Miraglia Del Giudice E, Grandone A. MKRN3 role in regulating pubertal onset: the state of art of functional studies. Front Endocrinol (Lausanne). 2022;13:991322.
  64. Li C, Lu W, Yang L, Li Z, Zhou X, Guo R, et al. MKRN3 regulates the epigenetic switch of mammalian puberty via ubiquitination of MBD3. Natl Sci Rev. 2020;7(3):671-85.
  65. Abreu AP, Toro CA, Song YB, Navarro VM, Bosch MA, Eren A, et al. MKRN3 inhibits the reproductive axis through actions in kisspeptin-expressing neurons. J Clin Invest. 2020;130(8):4486-500.
  66. Heras V, Sangiao-Alvarellos S, Manfredi-Lozano M, Sanchez-Tapia MJ, Ruiz-Pino F, Roa J, et al. Hypothalamic miR-30 regulates puberty onset via repression of the puberty-suppressing factor, Mkrn3. PLoS Biol. 2019;17(11):e3000532.
  67. Naule L, Mancini A, Pereira SA, Gassaway BM, Lydeard JR, Magnotto JC, et al. MKRN3 inhibits puberty onset via interaction with IGF2BP1 and regulation of hypothalamic plasticity. JCI Insight. 2023;8(8).
  68. Lomniczi A, Loche A, Castellano JM, Ronnekleiv OK, Bosch M, Kaidar G, et al. Epigenetic control of female puberty. Nat Neurosci. 2013;16(3):281-9.
  69. Lomniczi A, Ojeda SR. The Emerging Role of Epigenetics in the Regulation of Female Puberty. Endocr Dev. 2016;29:1-16.
  70. Toro CA, Wright H, Aylwin CF, Ojeda SR, Lomniczi A. Trithorax dependent changes in chromatin landscape at enhancer and promoter regions drive female puberty. Nat Commun. 2018;9(1):57.
  71. Lomniczi A, Wright H, Castellano JM, Matagne V, Toro CA, Ramaswamy S, et al. Epigenetic regulation of puberty via Zinc finger protein-mediated transcriptional repression. Nat Commun. 2015;6:10195.
  72. Barker-Gibb ML, Sahu A, Pohl CR, Plant TM. Elevating circulating leptin in prepubertal male rhesus monkeys (Macaca mulatta) does not elicit precocious gonadotropin-releasing hormone release, assessed indirectly. J Clin Endocrinol Metab. 2002;87(11):4976-83.
  73. Vazquez MJ, Toro CA, Castellano JM, Ruiz-Pino F, Roa J, Beiroa D, et al. SIRT1 mediates obesity- and nutrient-dependent perturbation of pubertal timing by epigenetically controlling Kiss1 expression. Nat Commun. 2018;9(1):4194.
  74. Manfredi-Lozano M, Roa J, Tena-Sempere M. Connecting metabolism and gonadal function: Novel central neuropeptide pathways involved in the metabolic control of puberty and fertility. Front Neuroendocrinol. 2018;48:37-49.
  75. Chachlaki K, Messina A, Delli V, Leysen V, Maurnyi C, Huber C, et al. NOS1 mutations cause hypogonadotropic hypogonadism with sensory and cognitive deficits that can be reversed in infantile mice. Sci Transl Med. 2022;14(665):eabh2369.
  76. Constantin S, Reynolds D, Oh A, Pizano K, Wray S. Nitric oxide resets kisspeptin-excited GnRH neurons via PIP2 replenishment. Proc Natl Acad Sci U S A. 2021;118(1).
  77. Witchel SF, Plant TM. Neurobiology of puberty and its disorders. Handb Clin Neurol. 2021;181:463-96.
  78. Goodman RL, Herbison AE, Lehman MN, Navarro VM. Neuroendocrine control of gonadotropin-releasing hormone: Pulsatile and surge modes of secretion. J Neuroendocrinol. 2022;34(5):e13094.
  79. Terasawa E. The mechanism underlying the pubertal increase in pulsatile GnRH release in primates. J Neuroendocrinol. 2022;34(5):e13119.
  80. Avendano MS, Vazquez MJ, Tena-Sempere M. Disentangling puberty: novel neuroendocrine pathways and mechanisms for the control of mammalian puberty. Hum Reprod Update. 2017;23(6):737-63.
  81. Aylwin CF, Lomniczi A. Sirtuin (SIRT)-1: At the crossroads of puberty and metabolism. Curr Opin Endocr Metab Res. 2020;14:65-72.
  82. Lhomme T, Clasadonte J, Imbernon M, Fernandois D, Sauve F, Caron E, et al. Tanycytic networks mediate energy balance by feeding lactate to glucose-insensitive POMC neurons. J Clin Invest. 2021;131(18).
  83. Forest MG, Cathiard AM. Pattern of plasma testosterone and delta4-androstenedione in normal newborns: Evidence for testicular activity at birth. J Clin Endocrinol Metab. 1975;41(5):977-80.
  84. Forest MG, Sizonenko PC, Cathiard AM, Bertrand J. Hypophyso-gonadal function in humans during the first year of life. 1. Evidence for testicular activity in early infancy. J Clin Invest. 1974;53(3):819-28.
  85. Grumbach MM. The neuroendocrinology of human puberty revisited. Horm Res. 2002;57 Suppl 2:2-14.
  86. Bizzarri C, Cappa M. Ontogeny of Hypothalamus-Pituitary Gonadal Axis and Minipuberty: An Ongoing Debate? Front Endocrinol (Lausanne). 2020;11:187.
  87. Cortes D, Muller J, Skakkebaek NE. Proliferation of Sertoli cells during development of the human testis assessed by stereological methods. Int J Androl. 1987;10(4):589-96.
  88. Muller J, Skakkebaek NE. Fluctuations in the number of germ cells during the late foetal and early postnatal periods in boys. Acta Endocrinol (Copenh). 1984;105(2):271-4.
  89. Kuijper EA, van Kooten J, Verbeke JI, van Rooijen M, Lambalk CB. Ultrasonographically measured testicular volumes in 0- to 6-year-old boys. Hum Reprod. 2008;23(4):792-6.
  90. Chemes HE, Rey RA, Nistal M, Regadera J, Musse M, Gonzalez-Peramato P, et al. Physiological androgen insensitivity of the fetal, neonatal, and early infantile testis is explained by the ontogeny of the androgen receptor expression in Sertoli cells. J Clin Endocrinol Metab. 2008;93(11):4408-12.
  91. Boukari K, Meduri G, Brailly-Tabard S, Guibourdenche J, Ciampi ML, Massin N, et al. Lack of androgen receptor expression in Sertoli cells accounts for the absence of anti-Mullerian hormone repression during early human testis development. J Clin Endocrinol Metab. 2009;94(5):1818-25.
  92. Codesal J, Regadera J, Nistal M, Regadera-Sejas J, Paniagua R. Involution of human fetal Leydig cells. An immunohistochemical, ultrastructural and quantitative study. J Anat. 1990;172:103-14.
  93. Busch AS, Ljubicic ML, Upners EN, Fischer MB, Raket LL, Frederiksen H, et al. Dynamic Changes of Reproductive Hormones in Male Minipuberty: Temporal Dissociation of Leydig and Sertoli Cell Activity. J Clin Endocrinol Metab. 2022;107(6):1560-8.
  94. Kuiri-Hanninen T, Seuri R, Tyrvainen E, Turpeinen U, Hamalainen E, Stenman UH, et al. Increased activity of the hypothalamic-pituitary-testicular axis in infancy results in increased androgen action in premature boys. J Clin Endocrinol Metab. 2011;96(1):98-105.
  95. Santos MC, Limao S, Ferreira P. Exacerbated mini-puberty of infancy in an ex-extreme preterm girl. BMJ Case Rep. 2020;13(9).
  96. Kuiri-Hanninen T, Kallio S, Seuri R, Tyrvainen E, Liakka A, Tapanainen J, et al. Postnatal developmental changes in the pituitary-ovarian axis in preterm and term infant girls. J Clin Endocrinol Metab. 2011;96(11):3432-9.
  97. Hagen CP, Aksglaede L, Sorensen K, Main KM, Boas M, Cleemann L, et al. Serum levels of anti-Mullerian hormone as a marker of ovarian function in 926 healthy females from birth to adulthood and in 172 Turner syndrome patients. J Clin Endocrinol Metab. 2010;95(11):5003-10.
  98. Ljubicic ML, Busch AS, Upners EN, Fischer MB, Petersen JH, Raket LL, et al. A Biphasic Pattern of Reproductive Hormones in Healthy Female Infants: The COPENHAGEN Minipuberty Study. J Clin Endocrinol Metab. 2022;107(9):2598-605.
  99. Johannsen TH, Main KM, Ljubicic ML, Jensen TK, Andersen HR, Andersen MS, et al. Sex Differences in Reproductive Hormones During Mini-Puberty in Infants With Normal and Disordered Sex Development. J Clin Endocrinol Metab. 2018;103(8):3028-37.
  100. Swee DS, Quinton R. Congenital Hypogonadotrophic Hypogonadism: Minipuberty and the Case for Neonatal Diagnosis. Front Endocrinol (Lausanne). 2019;10:97.
  101. Jespersen K, Ljubicic ML, Johannsen TH, Christiansen P, Skakkebaek NE, Juul A. Distinguishing between hidden testes and anorchia: the role of endocrine evaluation in infancy and childhood. Eur J Endocrinol. 2020;183(1):107-17.
  102. Koo MM, Rohan TE. Accuracy of short-term recall of age at menarche. Ann Hum Biol. 1997;24(1):61-4.
  103. Dorn LD, Sontag-Padilla LM, Pabst S, Tissot A, Susman EJ. Longitudinal reliability of self-reported age at menarche in adolescent girls: variability across time and setting. Dev Psychol. 2013;49(6):1187-93.
  104. Busch AS, Hollis B, Day FR, Sorensen K, Aksglaede L, Perry JRB, et al. Voice break in boys-temporal relations with other pubertal milestones and likely causal effects of BMI. Hum Reprod. 2019;34(8):1514-22.
  105. Lewis ME, Shapland F, Watts R. The influence of chronic conditions and the environment on pubertal development. An example from medieval England. Int J Paleopathol. 2016;12:1-10.
  106. Arthur NA, Gowland RL, Redfern RC. Coming of age in Roman Britain: Osteological evidence for pubertal timing. Am J Phys Anthropol. 2016;159(4):698-713.
  107. Liu W, Yan X, Li C, Shu Q, Chen M, Cai L, et al. A secular trend in age at menarche in Yunnan Province, China: a multiethnic population study of 1,275,000 women. BMC Public Health. 2021;21(1):1890.
  108. Wu T, Pauline M, Germaine BM. Ethnic differences in the Presence of Secondary Sex Characteristics and Menarche Among US Girls:The Third National Health and Nutrition Examination Survey,1988-1994. Pediatrics. 2002;110(4).
  109. Herman-Giddens ME, Slora EJ, Wasserman RC, Bourdony CJ, Bhapkar MV, Koch GG, et al. Secondary sexual characteristics and menses in young girls seen in office practice: a study from the Pediatric Research in Office Settings network. Pediatrics. 1997;99(4):505-12.
  110. Aksglaede L, Sorensen K, Petersen JH, Skakkebaek NE, Juul A. Recent decline in age at breast development: the Copenhagen Puberty Study. Pediatrics. 2009;123(5):e932-9.
  111. Biro FM, Pajak A, Wolff MS, Pinney SM, Windham GC, Galvez MP, et al. Age of Menarche in a Longitudinal US Cohort. J Pediatr Adolesc Gynecol. 2018;31(4):339-45.
  112. Marti-Henneberg C, Vizmanos B. The duration of puberty in girls is related to the timing of its onset. J Pediatr. 1997;131(4):618-21.
  113. Pantsiotou S, Papadimitriou A, Douros K, Priftis K, Nicolaidou P, Fretzayas A. Maturational tempo differences in relation to the timing of the onset of puberty in girls. Acta Paediatr. 2008;97(2):217-20.
  114. Eckert-Lind C, Busch AS, Petersen JH, Biro FM, Butler G, Brauner EV, et al. Worldwide Secular Trends in Age at Pubertal Onset Assessed by Breast Development Among Girls: A Systematic Review and Meta-analysis. JAMA Pediatr. 2020;174(4):e195881.
  115. Sorensen K, Mouritsen A, Aksglaede L, Hagen C, Mogensen S, Juul A. Recent secular Trends in Pubertal Timing: Implications for Evaluation and Diagnosis of Precocious Puberty. Hormone Research in Paediatrics. 2012;77:137-45.
  116. Predieri B, Iughetti L, Bernasconi S, Street ME. Endocrine Disrupting Chemicals' Effects in Children: What We Know and What We Need to Learn? Int J Mol Sci. 2022;23(19).
  117. Krstevska-Konstantinova M, Charlier C, Craen M, Du Caju M, Heinrichs C, de Beaufort C, et al. Sexual precocity after immigration from developing countries to Belgium: evidence of previous exposure to organochlorine pesticides. Hum Reprod. 2001;16(5):1020-6.
  118. Lopez-Rodriguez D, Franssen D, Heger S, Parent AS. Endocrine-disrupting chemicals and their effects on puberty. Best Pract Res Clin Endocrinol Metab. 2021;35(5):101579.
  119. Fudvoye J, Lopez-Rodriguez D, Franssen D, Parent AS. Endocrine disrupters and possible contribution to pubertal changes. Best Pract Res Clin Endocrinol Metab. 2019;33(3):101300.
  120. Kaplowitz PB, Slora EJ, Wasserman RC, Pedlow SE, Herman-Giddens ME. Earlier onset of puberty in girls: relation to increased body mass index and race. Pediatrics. 2001;108(2):347-53.
  121. Kaplowitz PB. Link between body fat and the timing of puberty. Pediatrics. 2008;121 Suppl 3:S208-17.
  122. Brix N, Ernst A, Lauridsen LLB, Parner ET, Arah OA, Olsen J, et al. Childhood overweight and obesity and timing of puberty in boys and girls: cohort and sibling-matched analyses. Int J Epidemiol. 2020;49(3):834-44.
  123. Ahmed ML, Ong KK, Dunger DB. Childhood obesity and the timing of puberty. Trends Endocrinol Metab. 2009;20(5):237-42.
  124. Day FR, Perry JR, Ong KK. Genetic Regulation of Puberty Timing in Humans. Neuroendocrinology. 2015;102(4):247-55.
  125. Shalitin S, Gat-Yablonski G. Associations of Obesity with Linear Growth and Puberty. Horm Res Paediatr. 2022;95(2):120-36.
  126. Reid RL, Ling N, Yen SS. Gonadotropin-releasing activity of alpha-melanocyte-stimulating hormone in normal subjects and in subjects with hypothalamic-pituitary dysfunction. J Clin Endocrinol Metab. 1984;58(5):773-7.
  127. Israel DD, Sheffer-Babila S, de Luca C, Jo YH, Liu SM, Xia Q, et al. Effects of leptin and melanocortin signaling interactions on pubertal development and reproduction. Endocrinology. 2012;153(5):2408-19.
  128. Morris DH, Jones ME, Schoemaker MJ, Ashworth A, Swerdlow AJ. Familial concordance for age at menarche: analyses from the Breakthrough Generations Study. Paediatr Perinat Epidemiol. 2011;25(3):306-11.
  129. Palmert MR, Hirschhorn JN. Genetic approaches to stature, pubertal timing, and other complex traits. Mol Genet Metab. 2003;80(1-2):1-10.
  130. Busch AS, Hagen CP, Juul A. Heritability of pubertal timing: detailed evaluation of specific milestones in healthy boys and girls. Eur J Endocrinol. 2020;183(1):13-20.
  131. Wohlfahrt-Veje C, Mouritsen A, Hagen CP, Tinggaard J, Mieritz MG, Boas M, et al. Pubertal Onset in Boys and Girls Is Influenced by Pubertal Timing of Both Parents. J Clin Endocrinol Metab. 2016;101(7):2667-74.
  132. Day FR, Thompson DJ, Helgason H, Chasman DI, Finucane H, Sulem P, et al. Genomic analyses identify hundreds of variants associated with age at menarche and support a role for puberty timing in cancer risk. Nat Genet. 2017;49(6):834-41.
  133. Sarnowski C, Cousminer DL, Franceschini N, Raffield LM, Jia G, Fernandez-Rhodes L, et al. Large trans-ethnic meta-analysis identifies AKR1C4 as a novel gene associated with age at menarche. Hum Reprod. 2021;36(7):1999-2010.
  134. Klein KO. Precocious puberty: who has it? Who should be treated? J Clin Endocrinol Metab. 1999;84(2):411-4.
  135. Parent AS, Teilmann G, Juul A, Skakkebaek NE, Toppari J, Bourguignon JP. The timing of normal puberty and the age limits of sexual precocity: variations around the world, secular trends, and changes after migration. Endocr Rev. 2003;24(5):668-93.
  136. Kaplowitz PB. Delayed puberty. Pediatr Rev. 2010;31(5):189-95.
  137. Bangalore Krishna K, Fuqua JS, Rogol AD, Klein KO, Popovic J, Houk CP, et al. Use of Gonadotropin-Releasing Hormone Analogs in Children: Update by an International Consortium. Horm Res Paediatr. 2019;91(6):357-72.
  138. Goldberg M, D'Aloisio AA, O'Brien KM, Zhao S, Sandler DP. Pubertal timing and breast cancer risk in the Sister Study cohort. Breast Cancer Res. 2020;22(1):112.
  139. Mueller NT, Duncan BB, Barreto SM, Chor D, Bessel M, Aquino EML, et al. Earlier age at menarche is associated with higher diabetes risk and cardiometabolic disease risk factors in Brazilian adults: Brazilian Longitudinal Study of Adult Health (ELSA-Brasil). Cardiovasc Diabetol. 2014;13.
  140. Partsch CJ, Heger S, Sippell WG. Management and outcome of central precocious puberty. Clin Endocrinol (Oxf). 2002;56(2):129-48.
  141. Soriano-Guillen L, Corripio R, Labarta JI, Canete R, Castro-Feijoo L, Espino R, et al. Central precocious puberty in children living in Spain: incidence, prevalence, and influence of adoption and immigration. J Clin Endocrinol Metab. 2010;95(9):4305-13.
  142. Kim SH, Huh K, Won S, Lee KW, Park MJ. A Significant Increase in the Incidence of Central Precocious Puberty among Korean Girls from 2004 to 2010. Plos One. 2015;10(11).
  143. Brandberg G, Raininko R, Eeg-Olofsson O. Hypothalamic hamartoma with gelastic seizures in Swedish children and adolescents. Eur J Paediatr Neurol. 2004;8(1):35-44.
  144. Cukier P, Castro LH, Banaskiwitz N, Teles LR, Ferreira LR, Adda CC, et al. The benign spectrum of hypothalamic hamartomas: infrequent epilepsy and normal cognition in patients presenting with central precocious puberty. Seizure. 2013;22(1):28-32.
  145. Harrison VS, Oatman O, Kerrigan JF. Hypothalamic hamartoma with epilepsy: Review of endocrine comorbidity. Epilepsia. 2017;58 Suppl 2(Suppl 2):50-9.
  146. Yang Y, Shen F, Jing XP, Zhang N, Xu SY, Li DD, et al. Case Report: Whole-Exome Sequencing of Hypothalamic Hamartoma From an Infant With Pallister-Hall Syndrome Revealed Novel de novo Mutation in the GLI3. Front Surg. 2021;8:734757.
  147. Fujita A, Higashijima T, Shirozu H, Masuda H, Sonoda M, Tohyama J, et al. Pathogenic variants of DYNC2H1, KIAA0556, and PTPN11 associated with hypothalamic hamartoma. Neurology. 2019;93(3):e237-e51.
  148. Hildebrand MS, Griffin NG, Damiano JA, Cops EJ, Burgess R, Ozturk E, et al. Mutations of the Sonic Hedgehog Pathway Underlie Hypothalamic Hamartoma with Gelastic Epilepsy. Am J Hum Genet. 2016;99(2):423-9.
  149. Chan YM, Fenoglio-Simeone KA, Paraschos S, Muhammad L, Troester MM, Ng YT, et al. Central precocious puberty due to hypothalamic hamartomas correlates with anatomic features but not with expression of GnRH, TGFalpha, or KISS1. Horm Res Paediatr. 2010;73(5):312-9.
  150. Harrison VS, Oatman O, Kerrigan JF. Hypothalamic hamartoma with epilepsy: Review of endocrine comorbidity. Epilepsia. 2017;58:50-9.
  151. Bourdillon P, Ferrand-Sorbet S, Apra C, Chipaux M, Raffo E, Rosenberg S, et al. Surgical treatment of hypothalamic hamartomas. Neurosurg Rev. 2021;44(2):753-62.
  152. Chalumeau M, Chemaitilly W, Trivin C, Adan L, Breart G, Brauner R. Central precocious puberty in girls: an evidence-based diagnosis tree to predict central nervous system abnormalities. Pediatrics. 2002;109(1):61-7.
  153. Stephen MD, Zage PE, Waguespack SG. Gonadotropin-dependent precocious puberty: neoplastic causes and endocrine considerations. Int J Pediatr Endocrinol. 2011;2011(1):184502.
  154. Pinheiro SL, Maciel J, Cavaco D, Figueiredo AA, Damasio IL, Donato S, et al. Precocious and accelerated puberty in children with neurofibromatosis type 1: results from a close follow-up of a cohort of 45 patients. Hormones. 2023;22(1):79-85.
  155. Almutlaq N, O'Neil J, Fuqua JS. Central precocious puberty in spina bifida children: Guidelines for the care of people with spina bifida. J Pediatr Rehabil Med. 2020;13(4):557-63.
  156. Cerbone M, Guemes M, Wade A, Improda N, Dattani M. Endocrine morbidity in midline brain defects: Differences between septo-optic dysplasia and related disorders. EClinicalMedicine. 2020;19:100224.
  157. Oatman OJ, McClellan DR, Olson ML, Garcia-Filion P. Endocrine and pubertal disturbances in optic nerve hypoplasia, from infancy to adolescence. Int J Pediatr Endocrinol. 2015;2015(1):8.
  158. Siegel DA, King JB, Lupo PJ, Durbin EB, Tai E, Mills K, et al. Counts, incidence rates, and trends of pediatric cancer in the United States, 2003-2019. J Natl Cancer Inst. 2023.
  159. Chemaitilly W, Merchant TE, Li Z, Barnes N, Armstrong GT, Ness KK, et al. Central precocious puberty following the diagnosis and treatment of paediatric cancer and central nervous system tumours: presentation and long-term outcomes. Clin Endocrinol (Oxf). 2016;84(3):361-71.
  160. De Sanctis V, Soliman AT, Elsedfy H, Soliman NA, Elalaily R, El Kholy M. Precocious Puberty Following Traumatic Brain Injury in Early Childhood: A Review of the Literature. Pediatr Endocrinol Rev. 2015;13(1):458-64.
  161. Dassa Y, Crosnier H, Chevignard M, Viaud M, Personnier C, Flechtner I, et al. Pituitary deficiency and precocious puberty after childhood severe traumatic brain injury: a long-term follow-up prospective study. Eur J Endocrinol. 2019;180(5):281-90.
  162. Partsch CJ, Sippell WG. Pathogenesis and epidemiology of precocious puberty. Effects of exogenous oestrogens. Hum Reprod Update. 2001;7(3):292-302.
  163. Stecchini MF, Braid Z, More CB, Aragon DC, Castro M, Moreira AC, et al. Gonadotropin-dependent pubertal disorders are common in patients with virilizing adrenocortical tumors in childhood. Endocr Connect. 2019;8(5):579-89.
  164. Chen H, Mo CY, Zhong LY. Central precocious puberty secondary to peripheral precocious puberty due to a pineal germ cell tumor: a case and review of literature. BMC Endocr Disord. 2023;23(1):237.
  165. Maione L, Bouvattier C, Kaiser UB. Central precocious puberty: Recent advances in understanding the aetiology and in the clinical approach. Clin Endocrinol (Oxf). 2021;95(4):542-55.
  166. Moise-Silverman J, Silverman LA. A review of the genetics and epigenetics of central precocious puberty. Front Endocrinol (Lausanne). 2022;13:1029137.
  167. Valadares LP, Meireles CG, De Toledo IP, Santarem de Oliveira R, Goncalves de Castro LC, Abreu AP, et al. MKRN3 Mutations in Central Precocious Puberty: A Systematic Review and Meta-Analysis. J Endocr Soc. 2019;3(5):979-95.
  168. Magnotto JC, Mancini A, Bird K, Montenegro L, Tutunculer F, Pereira SA, et al. Novel MKRN3 Missense Mutations Associated With Central Precocious Puberty Reveal Distinct Effects on Ubiquitination. J Clin Endocrinol Metab. 2023;108(7):1646-56.
  169. Li M, Chen Y, Liao B, Tang J, Zhong J, Lan D. The role of kisspeptin and MKRN3 in the diagnosis of central precocious puberty in girls. Endocr Connect. 2021;10(9):1147-54.
  170. Ioannides Y, Lokulo-Sodipe K, Mackay DJ, Davies JH, Temple IK. Temple syndrome: improving the recognition of an underdiagnosed chromosome 14 imprinting disorder: an analysis of 51 published cases. J Med Genet. 2014;51(8):495-501.
  171. Gomes LG, Cunha-Silva M, Crespo RP, Ramos CO, Montenegro LR, Canton A, et al. DLK1 Is a Novel Link Between Reproduction and Metabolism. J Clin Endocrinol Metab. 2019;104(6):2112-20.
  172. Silveira LG, Noel SD, Silveira-Neto AP, Abreu AP, Brito VN, Santos MG, et al. Mutations of the KISS1 gene in disorders of puberty. J Clin Endocrinol Metab. 2010;95(5):2276-80.
  173. Teles MG, Bianco SD, Brito VN, Trarbach EB, Kuohung W, Xu S, et al. A GPR54-activating mutation in a patient with central precocious puberty. N Engl J Med. 2008;358(7):709-15.
  174. Bianco SD, Vandepas L, Correa-Medina M, Gereben B, Mukherjee A, Kuohung W, et al. KISS1R intracellular trafficking and degradation: effect of the Arg386Pro disease-associated mutation. Endocrinology. 2011;152(4):1616-26.
  175. Tinano FR, Canton APM, Montenegro LR, de Castro Leal A, Faria AG, Seraphim CE, et al. Clinical and Genetic Characterization of Familial Central Precocious Puberty. J Clin Endocrinol Metab. 2023;108(7):1758-67.
  176. Nicoara DM, Scutca AC, Mang N, Juganaru I, Munteanu AI, Vitan L, et al. Central precocious puberty in Prader-Willi syndrome: a narrative review. Front Endocrinol (Lausanne). 2023;14:1150323.
  177. Spielmann S, Partsch CJ, Gosch A, Pankau R. Treatment of central precocious puberty and early puberty with GnRH analog in girls with Williams-Beuren syndrome. J Pediatr Endocrinol Metab. 2015;28(11-12):1363-7.
  178. Patti G, Malerba F, Calevo MG, Schiavone M, Scaglione M, Casalini E, et al. Pubertal timing in children with Silver Russell syndrome compared to those born small for gestational age. Front Endocrinol (Lausanne). 2022;13:975511.
  179. Palmert MR, Malin HV, Boepple PA. Unsustained or slowly progressive puberty in young girls: initial presentation and long-term follow-up of 20 untreated patients. J Clin Endocrinol Metab. 1999;84(2):415-23.
  180. Cote DJ, Smith TR, Sandler CN, Gupta T, Bale TA, Bi WL, et al. Functional Gonadotroph Adenomas: Case Series and Report of Literature. Neurosurgery. 2016;79(6):823-31.
  181. Ntali G, Capatina C. Updating the Landscape for Functioning Gonadotroph Tumors. Medicina (Kaunas). 2022;58(8).
  182. Uhing A, Ahmed A, Salamat S, Chen M. A Rare Case of Precocious Puberty Secondary to an LH-secreting Pituitary Adenoma. JCEM Case Rep. 2023;1(3):luad055.
  183. Sisk-Hackworth L, Kelley ST, Thackray VG. Sex, puberty, and the gut microbiome. Reproduction. 2023;165(2):R61-R74.
  184. Calcaterra V, Rossi V, Massini G, Regalbuto C, Hruby C, Panelli S, et al. Precocious puberty and microbiota: The role of the sex hormone-gut microbiome axis. Front Endocrinol (Lausanne). 2022;13:1000919.
  185. Chaussain JL, Roger M, Couprie C, Lahlou N, Canlorbe P. Treatment of precocious puberty with a long-acting preparation of D-Trp6-LHRH. Horm Res. 1987;28(2-4):155-63.
  186. Lahlou N, Roger M, Chaussain JL, Feinstein MC, Sultan C, Toublanc JE, et al. Gonadotropin and alpha-subunit secretion during long term pituitary suppression by D-Trp6-luteinizing hormone-releasing hormone microcapsules as treatment of precocious puberty. J Clin Endocrinol Metab. 1987;65(5):946-53.
  187. Carel JC, Léger J. Clinical practice. Precocious puberty. N Engl J Med. 2008;358(22):2366-77.
  188. Chen M, Eugster EA. Central Precocious Puberty: Update on Diagnosis and Treatment. Pediatr Drugs. 2015;17(4):273-81.
  189. Johnson SR, Nolan RC, Grant MT, Price GJ, Siafarikas A, Bint L, et al. Sterile abscess formation associated with depot leuprorelin acetate therapy for central precocious puberty. J Paediatr Child Health. 2012;48(3):E136-9.
  190. Miller BS, Shukla AR. Sterile abscess formation in response to two separate branded long-acting gonadotropin-releasing hormone agonists. Clin Ther. 2010;32(10):1749-51.
  191. Pienkowski C, Tauber M. Gonadotropin-Releasing Hormone Agonist Treatment in Sexual Precocity. Endocr Dev. 2016;29:214-29.
  192. De Sanctis V, Soliman AT, Di Maio S, Soliman N, Elsedfy H. Long-term effects and significant Adverse Drug Reactions (ADRs) associated with the use of Gonadotropin-Releasing Hormone analogs (GnRHa) for central precocious puberty: a brief review of literature. Acta Biomed. 2019;90(3):345-59.
  193. van der Sluis IM, Boot AM, Krenning EP, Drop SL, de Muinck Keizer-Schrama SM. Longitudinal follow-up of bone density and body composition in children with precocious or early puberty before, during and after cessation of GnRH agonist therapy. J Clin Endocrinol Metab. 2002;87(2):506-12.
  194. Paterson WF, McNeill E, Young D, Donaldson MD. Auxological outcome and time to menarche following long-acting goserelin therapy in girls with central precocious or early puberty. Clin Endocrinol (Oxf). 2004;61(5):626-34.
  195. Traggiai C, Perucchin PP, Zerbini K, Gastaldi R, De Biasio P, Lorini R. Outcome after depot gonadotrophin-releasing hormone agonist treatment for central precocious puberty: effects on body mass index and final height. Eur J Endocrinol. 2005;153(3):463-4.
  196. Wolters B, Lass N, Reinehr T. Treatment with gonadotropin-releasing hormone analogues: different impact on body weight in normal-weight and overweight children. Horm Res Paediatr. 2012;78(5-6):304-11.
  197. Palmert MR, Mansfield MJ, Crowley WF, Crigler JF, Crawford JD, Boepple PA. Is obesity an outcome of gonadotropin-releasing hormone agonist administration? Analysis of growth and body composition in 110 patients with central precocious puberty. J Clin Endocrinol Metab. 1999;84(12):4480-8.
  198. Boot AM, De Muinck Keizer-Schrama S, Pols HA, Krenning EP, Drop SL. Bone mineral density and body composition before and during treatment with gonadotropin-releasing hormone agonist in children with central precocious and early puberty. J Clin Endocrinol Metab. 1998;83(2):370-3.
  199. Lazar L, Lebenthal Y, Yackobovitch-Gavan M, Shalitin S, de Vries L, Phillip M, et al. Treated and untreated women with idiopathic precocious puberty: BMI evolution, metabolic outcome, and general health between third and fifth decades. J Clin Endocrinol Metab. 2015;100(4):1445-51.
  200. Guaraldi F, Beccuti G, Gori D, Ghizzoni L. MANAGEMENT OF ENDOCRINE DISEASE: Long-term outcomes of the treatment of central precocious puberty. Eur J Endocrinol. 2016;174(3):R79-87.
  201. Lee JW, Kim HJ, Choe YM, Kang HS, Kim SK, Jun YH, et al. Significant adverse reactions to long-acting gonadotropin-releasing hormone agonists for the treatment of central precocious puberty and early onset puberty. Ann Pediatr Endocrinol Metab. 2014;19(3):135-40.
  202. Omar AA, Nyaga G, Mungai LNW. Pseudotumor cerebri in patient on leuprolide acetate for central precocious puberty. Int J Pediatr Endocrinol. 2020;2020(1):22.
  203. Lazar L, Meyerovitch J, de Vries L, Phillip M, Lebenthal Y. Treated and untreated women with idiopathic precocious puberty: long-term follow-up and reproductive outcome between the third and fifth decades. Clin Endocrinol (Oxf). 2014;80(4):570-6.
  204. Bertelloni S, Baroncelli GI, Ferdeghini M, Menchini-Fabris F, Saggese G. Final height, gonadal function and bone mineral density of adolescent males with central precocious puberty after therapy with gonadotropin-releasing hormone analogues. Eur J Pediatr. 2000;159(5):369-74.
  205. Thornton P, Silverman LA, Geffner ME, Neely EK, Gould E, Danoff TM. Review of outcomes after cessation of gonadotropin-releasing hormone agonist treatment of girls with precocious puberty. Pediatr Endocrinol Rev. 2014;11(3):306-17.
  206. Arcari AJ, Freire AV, Ballerini MG, Escobar ME, Diaz Marsiglia YM, Bergada I, et al. Prevalence of Polycystic Ovarian Syndrome in Girls with a History of Idiopathic Central Precocious Puberty. Horm Res Paediatr. 2023:1-6.
  207. Eugster EA. Treatment of Central Precocious Puberty. J Endocr Soc. 2019;3(5):965-72.
  208. Schoelwer MJ, Donahue KL, Didrick P, Eugster EA. One-Year Follow-Up of Girls with Precocious Puberty and Their Mothers: Do Psychological Assessments Change over Time or with Treatment? Horm Res Paediatr. 2017;88(5):347-53.
  209. Kaplowitz PB, Backeljauw PF, Allen DB. Toward More Targeted and Cost-Effective Gonadotropin-Releasing Hormone Analog Treatment in Girls with Central Precocious Puberty. Horm Res Paediatr. 2018;90(1):1-7.
  210. Popovic J, Geffner ME, Rogol AD, Silverman LA, Kaplowitz PB, Mauras N, et al. Gonadotropin-releasing hormone analog therapies for children with central precocious puberty in the United States. Frontiers in Pediatrics. 2022;10.
  211. Eugster EA. Treatment of Central Precocious Puberty. Journal of the Endocrine Society. 2019;3(5):965-72.
  212. Pasquino AM, Municchi G, Pucarelli I, Segni M, Mancini MA, Troiani S. Combined treatment with gonadotropin-releasing hormone analog and growth hormone in central precocious puberty. J Clin Endocrinol Metab. 1996;81(3):948-51.
  213. Shi Y, Ma Z, Yang X, Ying Y, Luo X, Hou L. Gonadotropin-releasing hormone analogue and recombinant human growth hormone treatment for idiopathic central precocious puberty in girls. Front Endocrinol (Lausanne). 2022;13:1085385.
  214. Cho AY, Shim YS, Lee HS, Hwang JS. Effect of gonadotropin-releasing hormone agonist monotherapy and combination therapy with growth hormone on final adult height in girls with central precocious puberty. Sci Rep. 2023;13(1):1264.
  215. Pucarelli I, Segni M, Ortore M, Arcadi E, Pasquino AM. Effects of combined gonadotropin-releasing hormone agonist and growth hormone therapy on adult height in precocious puberty: a further contribution. J Pediatr Endocrinol Metab. 2003;16(7):1005-10.
  216. Li YH, Zhu SY, Ma HM, Su Z, Chen HS, Chen QL, et al. [Effect of gonadotropin-releasing hormone analog combined with stanazolol on final height in girls with idiopathic central precocious puberty and apparent decrease of linear growth]. Zhonghua Er Ke Za Zhi. 2013;51(11):807-12.
  217. Zhu S, Long L, Hu Y, Tuo Y, Li Y, Yu Z. GnRHa/Stanozolol Combined Therapy Maintains Normal Bone Growth in Central Precocious Puberty. Front Endocrinol (Lausanne). 2021;12:678797.
  218. Vottero A, Pedori S, Verna M, Pagano B, Cappa M, Loche S, et al. Final height in girls with central idiopathic precocious puberty treated with gonadotropin-releasing hormone analog and oxandrolone. J Clin Endocrinol Metab. 2006;91(4):1284-7.
  219. Liu S, Liu Q, Cheng X, Luo Y, Wen Y. Effects and safety of combination therapy with gonadotropin-releasing hormone analogue and growth hormone in girls with idiopathic central precocious puberty: a meta-analysis. J Endocrinol Invest. 2016;39(10):1167-78.
  220. Wit JM. Should Skeletal Maturation Be Manipulated for Extra Height Gain? Front Endocrinol (Lausanne). 2021;12:812196.
  221. Dotremont H, France A, Heinrichs C, Tenoutasse S, Brachet C, Cools M, et al. Efficacy and safety of a 4-year combination therapy of growth hormone and gonadotropin-releasing hormone analogue in pubertal girls with short predicted adult height. Front Endocrinol (Lausanne). 2023;14:1113750.
  222. Trujillo MV, Lee PA, Reifschneider K, Backeljauw PF, Dragnic S, Van Komen S, et al. Using change in predicted adult height during GnRH agonist treatment for individualized treatment decisions in girls with central precocious puberty. J Pediatr Endocrinol Metab. 2023;36(3):299-308.
  223. Vargas Trujillo M, Dragnic S, Aldridge P, Klein KO. Importance of individualizing treatment decisions in girls with central precocious puberty when initiating treatment after age 7 years or continuing beyond a chronological age of 10 years or a bone age of 12 years. J Pediatr Endocrinol Metab. 2021;34(6):733-9.
  224. Mendle J, Ryan RM, McKone KMP. Age at Menarche, Depression, and Antisocial Behavior in Adulthood. Pediatrics. 2018;141(1).
  225. Mendle J, Ryan RM, McKone KMP. Early Menarche and Internalizing and Externalizing in Adulthood: Explaining the Persistence of Effects. J Adolescent Health. 2019;65(5):599-606.
  226. Schoelwer MJ, Donahue KL, Didrick P, Eugster EA. One-Year Follow-Up of Girls with Precocious Puberty and Their Mothers: Do Psychological Assessments Change over Time or with Treatment? Hormone Research in Paediatrics. 2017;88(5):347-53.
  227. Wojniusz S, Callens N, Sütterlin S, Andersson S, De Schepper J, Gies I, et al. Cognitive, Emotional, and Psychosocial Functioning of Girls Treated with Pharmacological Puberty Blockage for Idiopathic Central Precocious Puberty. Front Psychol. 2016;7:1053.
  228. Williams VSL, Soliman AM, Barrett AM, Klein KO. Review and evaluation of patient-centered psychosocial assessments for children with central precocious puberty or early puberty. J Pediatr Endocrinol Metab. 2018;31(5):485-95.
  229. Lewis KA, Eugster EA. Random luteinizing hormone often remains pubertal in children treated with the histrelin implant for central precocious puberty. J Pediatr. 2013;162(3):562-5.
  230. Neely EK, Silverman LA, Geffner ME, Danoff TM, Gould E, Thornton PS. Random unstimulated pediatric luteinizing hormone levels are not reliable in the assessment of pubertal suppression during histrelin implant therapy. Int J Pediatr Endocrinol. 2013;2013(1):20.
  231. Klein KO, Lee PA. Gonadotropin-Releasing Hormone (GnRHa) Therapy for Central Precocious Puberty (CPP): Review of Nuances in Assessment of Height, Hormonal Suppression, Psychosocial Issues, and Weight Gain, with Patient Examples. Pediatr Endocrinol Rev. 2018;15(4):298-312.
  232. Houk CP, Kunselman AR, Lee PA. The diagnostic value of a brief GnRH analogue stimulation test in girls with central precocious puberty: a single 30-minute post-stimulation LH sample is adequate. J Pediatr Endocrinol Metab. 2008;21(12):1113-8.
  233. Yazdani P, Lin Y, Raman V, Haymond M. A single sample GnRHa stimulation test in the diagnosis of precocious puberty. Int J Pediatr Endocrinol. 2012;2012(1):23.
  234. Ohyama K, Tanaka T, Tachibana K, Niimi H, Fujieda K, Matsuo N, et al. Timing for discontinuation of treatment with a long-acting gonadotropin-releasing hormone analog in girls with central precocious puberty. TAP-144SR CPP Study Group. Endocr J. 1998;45(3):351-6.
  235. Carel JC, Roger M, Ispas S, Tondu F, Lahlou N, Blumberg J, et al. Final height after long-term treatment with triptorelin slow release for central precocious puberty: importance of statural growth after interruption of treatment. French study group of Decapeptyl in Precocious Puberty. J Clin Endocrinol Metab. 1999;84(6):1973-8.
  236. Carel JC, Eugster EA, Rogol A, Ghizzoni L, Palmert MR, Antoniazzi F, et al. Consensus statement on the use of gonadotropin-releasing hormone analogs in children. Pediatrics. 2009;123(4):e752-62.
  237. Boyce AM, Collins MT. Fibrous Dysplasia/McCune-Albright Syndrome: A Rare, Mosaic Disease of Galpha s Activation. Endocr Rev. 2020;41(2):345-70.
  238. Tufano M, Ciofi D, Amendolea A, Stagi S. Auxological and Endocrinological Features in Children With McCune Albright Syndrome: A Review. Front Endocrinol (Lausanne). 2020;11:522.
  239. Spencer T, Pan KS, Collins MT, Boyce AM. The Clinical Spectrum of McCune-Albright Syndrome and Its Management. Horm Res Paediatr. 2019;92(6):347-56.
  240. Kim IS, Kim ER, Nam HJ, Chin MO, Moon YH, Oh MR, et al. Activating mutation of GS alpha in McCune-Albright syndrome causes skin pigmentation by tyrosinase gene activation on affected melanocytes. Horm Res. 1999;52(5):235-40.
  241. Szymczuk V, Taylor J, Michel Z, Sinaii N, Boyce AM. Skeletal Disease Acquisition in Fibrous Dysplasia: Natural History and Indicators of Lesion Progression in Children. J Bone Miner Res. 2022;37(8):1473-8.
  242. Collins MT, Chebli C, Jones J, Kushner H, Consugar M, Rinaldo P, et al. Renal phosphate wasting in fibrous dysplasia of bone is part of a generalized renal tubular dysfunction similar to that seen in tumor-induced osteomalacia. J Bone Miner Res. 2001;16(5):806-13.
  243. Bhattacharyya N, Wiench M, Dumitrescu C, Connolly BM, Bugge TH, Patel HV, et al. Mechanism of FGF23 processing in fibrous dysplasia. J Bone Miner Res. 2012;27(5):1132-41.
  244. Boyce AM, Casey RK, Ovejero Crespo D, Murdock CM, Estrada A, Guthrie LC, et al. Gynecologic and reproductive outcomes in fibrous dysplasia/McCune-Albright syndrome. Orphanet J Rare Dis. 2019;14(1):90.
  245. Boyce AM, Chong WH, Shawker TH, Pinto PA, Linehan WM, Bhattacharryya N, et al. Characterization and management of testicular pathology in McCune-Albright syndrome. J Clin Endocrinol Metab. 2012;97(9):E1782-90.
  246. Tatsi C, Stratakis CA. Neonatal Cushing Syndrome: A Rare but Potentially Devastating Disease. Clin Perinatol. 2018;45(1):103-18.
  247. Gaujoux S, Salenave S, Ronot M, Rangheard AS, Cros J, Belghiti J, et al. Hepatobiliary and Pancreatic neoplasms in patients with McCune-Albright syndrome. J Clin Endocrinol Metab. 2014;99(1):E97-101.
  248. Zacharin M, Bajpai A, Chow CW, Catto-Smith A, Stratakis C, Wong MW, et al. Gastrointestinal polyps in McCune Albright syndrome. J Med Genet. 2011;48(7):458-61.
  249. Majoor BC, Boyce AM, Bovee JV, Smit VT, Collins MT, Cleton-Jansen AM, et al. Increased Risk of Breast Cancer at a Young Age in Women with Fibrous Dysplasia. J Bone Miner Res. 2018;33(1):84-90.
  250. Lumbroso S, Paris F, Sultan C, European Collaborative S. Activating Gsalpha mutations: analysis of 113 patients with signs of McCune-Albright syndrome--a European Collaborative Study. J Clin Endocrinol Metab. 2004;89(5):2107-13.
  251. Roszko KL, Guthrie L, Li X, Collins MT, de Castro LF, Boyce AM. Identification of GNAS Variants in Circulating Cell-Free DNA from Patients with Fibrous Dysplasia/McCune Albright Syndrome. J Bone Miner Res. 2023;38(3):443-50.
  252. Couch RM, Muller J, Perry YS, Winter JS. Kinetic analysis of inhibition of human adrenal steroidogenesis by ketoconazole. J Clin Endocrinol Metab. 1987;65(3):551-4.
  253. Estrada A, Boyce AM, Brillante BA, Guthrie LC, Gafni RI, Collins MT. Long-term outcomes of letrozole treatment for precocious puberty in girls with McCune-Albright syndrome. Eur J Endocrinol. 2016;175(5):477-83.
  254. Eugster EA, Rubin SD, Reiter EO, Plourde P, Jou HC, Pescovitz OH, et al. Tamoxifen treatment for precocious puberty in McCune-Albright syndrome: a multicenter trial. J Pediatr. 2003;143(1):60-6.
  255. Hu R, Hilakivi-Clarke L, Clarke R. Molecular mechanisms of tamoxifen-associated endometrial cancer (Review). Oncol Lett. 2015;9(4):1495-501.
  256. Sims EK, Garnett S, Guzman F, Paris F, Sultan C, Eugster EA, et al. Fulvestrant treatment of precocious puberty in girls with McCune-Albright syndrome. Int J Pediatr Endocrinol. 2012;2012(1):26.
  257. Nabhan ZM, West KW, Eugster EA. Oophorectomy in McCune-Albright syndrome: a case of mistaken identity. J Pediatr Surg. 2007;42(9):1578-83.
  258. Corica D, Aversa T, Pepe G, De Luca F, Wasniewska M. Peculiarities of Precocious Puberty in Boys and Girls With McCune-Albright Syndrome. Front Endocrinol (Lausanne). 2018;9:337.
  259. Tessaris D, Matarazzo P, Mussa A, Tuli G, Verna F, Fiore L, et al. Combined treatment with bicalutamide and anastrozole in a young boy with peripheral precocious puberty due to McCune-Albright Syndrome. Endocr J. 2012;59(2):111-7.
  260. Partsch CJ, Kreller-Laugwitz G, Sippell WG. [Transitory precocious puberty caused by autonomous ovarian cysts. Clinical, endocrinologic and sonographic course]. Monatsschr Kinderheilkd. 1989;137(4):235-8.
  261. de Sousa G, Wunsch R, Andler W. Precocious pseudopuberty due to autonomous ovarian cysts: a report of ten cases and long-term follow-up. Hormones. 2008;7(2):170-4.
  262. Nayak S, Witchel SF, Sanfilippo JS. Vaginal foreign body: a delayed diagnosis. J Pediatr Adolesc Gynecol. 2014;27(6):e127-9.
  263. Ganer Herman H, Shalev A, Ginat S, Kerner R, Keidar R, Bar J, et al. Clinical characteristics of adnexal torsion in premenarchal patients. Arch Gynecol Obstet. 2016;293(3):603-8.
  264. Adeyemi-Fowode O, Lin EG, Syed F, Sangi-Haghpeykar H, Zhu H, Dietrich JE. Adnexal Torsion in Children and Adolescents: A Retrospective Review of 245 Cases at a Single Institution. J Pediatr Adolesc Gynecol. 2019;32(1):64-9.
  265. Spinelli C, Piscioneri J, Strambi S. Adnexal torsion in adolescents: update and review of the literature. Curr Opin Obstet Gynecol. 2015;27(5):320-5.
  266. Spinelli C, Buti I, Pucci V, Liserre J, Alberti E, Nencini L, et al. Adnexal torsion in children and adolescents: new trends to conservative surgical approach -- our experience and review of literature. Gynecol Endocrinol. 2013;29(1):54-8.
  267. Heo S, Shim YS, Lee HS, Hwang JS. Clinical course of peripheral precocious puberty in girls due to autonomous ovarian cysts. Clin Endocrinol (Oxf). 2023.
  268. Poonai N, Poonai C, Lim R, Lynch T. Pediatric ovarian torsion: case series and review of the literature. Can J Surg. 2013;56(2):103-8.
  269. Gaikwad PM, Goswami S, Sengupta N, Baidya A, Das N. Transformation of Peripheral Sexual Precocity to Central Sexual Precocity Following Treatment of Granulosa Cell Tumor of the Ovary. Cureus. 2022;14(2):e22676.
  270. Bessiere L, Todeschini AL, Auguste A, Sarnacki S, Flatters D, Legois B, et al. A Hot-spot of In-frame Duplications Activates the Oncoprotein AKT1 in Juvenile Granulosa Cell Tumors. EBioMedicine. 2015;2(5):421-31.
  271. Fuller PJ, Leung D, Chu S. Genetics and genomics of ovarian sex cord-stromal tumors. Clin Genet. 2017;91(2):285-91.
  272. Yamamoto H, Sakamoto H, Kumagai H, Abe T, Ishiguro S, Uchida K, et al. Clinical Guidelines for Diagnosis and Management of Peutz-Jeghers Syndrome in Children and Adults. Digestion. 2023:1-13.
  273. De Paolis E, Paragliola RM, Concolino P. Spectrum of DICER1 Germline Pathogenic Variants in Ovarian Sertoli-Leydig Cell Tumor. J Clin Med. 2021;10(9).
  274. Patel SS, Carrick KS, Carr BR. Virilization persists in a woman with an androgen-secreting granulosa cell tumor. Fertil Steril. 2009;91(3):933 e13-5.
  275. Kalfa N, Meduri G, Philibert P, Patte C, Boizet-Bonhoure B, Thibaut E, et al. Unusual virilization in girls with juvenile granulosa cell tumors of the ovary is related to intratumoral aromatase deficiency. Horm Res Paediatr. 2010;74(2):83-91.
  276. Schultz KAP, Williams GM, Kamihara J, Stewart DR, Harris AK, Bauer AJ, et al. DICER1 and Associated Conditions: Identification of At-risk Individuals and Recommended Surveillance Strategies. Clin Cancer Res. 2018;24(10):2251-61.
  277. Trahmono, Wahyudi I, Rodjani A, Situmorang GR, Marzuki NS. Precocious Pseudo-Puberty with Testicular Enlargement: Two Cases of Leydig Cell Tumor with Different Histopathological Results. Res Rep Urol. 2020;12:577-82.
  278. Luckie TM, Danzig M, Zhou S, Wu H, Cost NG, Karaviti L, et al. A Multicenter Retrospective Review of Pediatric Leydig Cell Tumor of the Testis. J Pediatr Hematol Oncol. 2019;41(1):74-6.
  279. De Felici M, Klinger FG, Campolo F, Balistreri CR, Barchi M, Dolci S. To Be or Not to Be a Germ Cell: The Extragonadal Germ Cell Tumor Paradigm. Int J Mol Sci. 2021;22(11).
  280. Cattoni A, Albanese A. Case report: Fluctuating tumor markers in a boy with gonadotropin-releasing hormone-independent precocious puberty induced by a pineal germ cell tumor. Front Pediatr. 2022;10:940656.
  281. . !!! INVALID CITATION !!! (269, 271, 272).
  282. Yhoshu E, Lone YA, Mahajan JK, Singh UB. Hepatoblastoma with Precocious Puberty. J Indian Assoc Pediatr Surg. 2019;24(1):68-71.
  283. Hersmus R, van Bever Y, Wolffenbuttel KP, Biermann K, Cools M, Looijenga LH. The biology of germ cell tumors in disorders of sex development. Clin Genet. 2017;91(2):292-301.
  284. Shenker A, Laue L, Kosugi S, Merendino JJ, Jr., Minegishi T, Cutler GB, Jr. A constitutively activating mutation of the luteinizing hormone receptor in familial male precocious puberty. Nature. 1993;365(6447):652-4.
  285. Themmen APN, Huhtaniemi IT. Mutations of gonadotropins and gonadotropin receptors: elucidating the physiology and pathophysiology of pituitary-gonadal function. Endocr Rev. 2000;21(5):551-83.
  286. Gurnurkar S, DiLillo E, Carakushansky M. A Case of Familial Male-limited Precocious Puberty with a Novel Mutation. J Clin Res Pediatr Endocrinol. 2021;13(2):239-44.
  287. Haddad NG, Eugster EA. Peripheral precocious puberty including congenital adrenal hyperplasia: causes, consequences, management and outcomes. Best Pract Res Clin Endocrinol Metab. 2019;33(3):101273.
  288. Cunha-Silva M, Brito VN, Macedo DB, Bessa DS, Ramos CO, Lima LG, et al. Spontaneous fertility in a male patient with testotoxicosis despite suppression of FSH levels. Hum Reprod. 2018;33(5):914-8.
  289. Kooij CD, Mavinkurve-Groothuis AMC, Kremer Hovinga ICL, Looijenga LHJ, Rinne T, Giltay JC, et al. Familial Male-limited Precocious Puberty (FMPP) and Testicular Germ Cell Tumors. J Clin Endocrinol Metab. 2022;107(11):3035-44.
  290. Reiter EO, Mauras N, McCormick K, Kulshreshtha B, Amrhein J, De Luca F, et al. Bicalutamide plus anastrozole for the treatment of gonadotropin-independent precocious puberty in boys with testotoxicosis: a phase II, open-label pilot study (BATT). J Pediatr Endocrinol Metab. 2010;23(10):999-1009.
  291. Leschek EW, Flor AC, Bryant JC, Jones JV, Barnes KM, Cutler GB, Jr. Effect of Antiandrogen, Aromatase Inhibitor, and Gonadotropin-releasing Hormone Analog on Adult Height in Familial Male Precocious Puberty. J Pediatr. 2017;190:229-35.
  292. Flippo C, Kolli V, Andrew M, Berger S, Bhatti T, Boyce AM, et al. Precocious Puberty in a Boy With Bilateral Leydig Cell Tumors due to a Somatic Gain-of-Function LHCGR Variant. J Endocr Soc. 2022;6(10):bvac127.
  293. Latronico AC, Lins TS, Brito VN, Arnhold IJ, Mendonca BB. The effect of distinct activating mutations of the luteinizing hormone receptor gene on the pituitary-gonadal axis in both sexes. Clin Endocrinol (Oxf). 2000;53(5):609-13.
  294. Vanwyk JJ, Grumbach MM. Syndrome of Precocious Menstruation and Galactorrhea in Juvenile Hypothyroidism - an Example of Hormonal Overlap in Pituitary Feedback. J Pediatr-Us. 1960;57(3):416-35.
  295. Kusuma Boddu S, Ayyavoo A, Hebbal Nagarajappa V, Kalenahalli KV, Muruda S, Palany R. Van Wyk Grumbach Syndrome and Ovarian Hyperstimulation in Juvenile Primary Hypothyroidism: Lessons From a 30-Case Cohort. J Endocr Soc. 2023;7(6):bvad042.
  296. Reddy P, Tiwari K, Kulkarni A, Parikh K, Khubchandani R. Van Wyk Grumbach Syndrome: A Rare Consequence of Hypothyroidism. Indian J Pediatr. 2018;85(11):1028-30.
  297. Zhang H, Geng N, Wang Y, Tian W, Xue F. Van Wyk and Grumbach syndrome: two case reports and review of the published work. J Obstet Gynaecol Res. 2014;40(2):607-10.
  298. Baranowski E, Hogler W. An unusual presentation of acquired hypothyroidism: the Van Wyk-Grumbach syndrome. Eur J Endocrinol. 2012;166(3):537-42.
  299. Anasti JN, Flack MR, Froehlich J, Nelson LM, Nisula BC. A potential novel mechanism for precocious puberty in juvenile hypothyroidism. J Clin Endocrinol Metab. 1995;80(1):276-9.
  300. Polat S, Kulle A, Karaca Z, Akkurt I, Kurtoglu S, Kelestimur F, et al. Characterisation of three novel CYP11B1 mutations in classic and non-classic 11beta-hydroxylase deficiency. Eur J Endocrinol. 2014;170(5):697-706.
  301. Armengaud JB, Charkaluk ML, Trivin C, Tardy V, Breart G, Brauner R, et al. Precocious pubarche: distinguishing late-onset congenital adrenal hyperplasia from premature adrenarche. J Clin Endocrinol Metab. 2009;94(8):2835-40.
  302. Miller WL. Congenital Adrenal Hyperplasia: Time to Replace 17OHP with 21-Deoxycortisol. Horm Res Paediatr. 2019;91(6):416-20.
  303. Sarafoglou K, Merke DP, Reisch N, Claahsen-van der Grinten H, Falhammar H, Auchus RJ. Interpretation of Steroid Biomarkers in 21-Hydroxylase Deficiency and Their Use in Disease Management. J Clin Endocrinol Metab. 2023;108(9):2154-75.
  304. Mallappa A, Merke DP. Management challenges and therapeutic advances in congenital adrenal hyperplasia. Nat Rev Endocrinol. 2022;18(6):337-52.
  305. Witchel SF. Congenital Adrenal Hyperplasia. J Pediatr Adolesc Gynecol. 2017;30(5):520-34.
  306. Wieneke JA, Thompson LD, Heffess CS. Adrenal cortical neoplasms in the pediatric population: a clinicopathologic and immunophenotypic analysis of 83 patients. Am J Surg Pathol. 2003;27(7):867-81.
  307. . !!! INVALID CITATION !!! (297).
  308. Custodio G, Komechen H, Figueiredo FR, Fachin ND, Pianovski MA, Figueiredo BC. Molecular epidemiology of adrenocortical tumors in southern Brazil. Mol Cell Endocrinol. 2012;351(1):44-51.
  309. Lapunzina P. Risk of tumorigenesis in overgrowth syndromes: a comprehensive review. Am J Med Genet C Semin Med Genet. 2005;137C(1):53-71.
  310. Clay MR, Pinto EM, Fishbein L, Else T, Kiseljak-Vassiliades K. Pathological and Genetic Stratification for Management of Adrenocortical Carcinoma. J Clin Endocrinol Metab. 2022;107(4):1159-69.
  311. Kaplowitz PB. For Premature Thelarche and Premature Adrenarche, the Case for Waiting before Testing. Horm Res Paediatr. 2020;93(9-10):573-6.
  312. Freedman SM, Kreitzer PM, Elkowitz SS, Soberman N, Leonidas JC. Ovarian microcysts in girls with isolated premature thelarche. J Pediatr. 1993;122(2):246-9.
  313. Witchel SF, Pinto B, Burghard AC, Oberfield SE. Update on adrenarche. Curr Opin Pediatr. 2020;32(4):574-81.
  314. Kaya G, Abali ZY, Bas F, Poyrazoglu S, Darendeliler F. Body mass index at the presentation of premature adrenarche is associated with components of metabolic syndrome at puberty. European Journal of Pediatrics. 2018;177(11):1593-601.
  315. Ibanez L, Ong K, de Zegher F, Marcos MV, del Rio L, Dunger DB. Fat distribution in non-obese girls with and without precocious pubarche: central adiposity related to insulinaemia and androgenaemia from prepuberty to postmenarche. Clin Endocrinol (Oxf). 2003;58(3):372-9.
  316. Ibanez L, Potau N, Virdis R, Zampolli M, Terzi C, Gussinye M, et al. Postpubertal outcome in girls diagnosed of premature pubarche during childhood: increased frequency of functional ovarian hyperandrogenism. J Clin Endocrinol Metab. 1993;76(6):1599-603.
  317. Tennila J, Jaaskelainen J, Utriainen P, Voutilainen R, Hakkinen M, Auriola S, et al. PCOS Features and Steroid Profiles Among Young Adult Women with a History of Premature Adrenarche. J Clin Endocrinol Metab. 2021;106(9):e3335-e45.
  318. Ramsey JT, Li Y, Arao Y, Naidu A, Coons LA, Diaz A, et al. Lavender Products Associated With Premature Thelarche and Prepubertal Gynecomastia: Case Reports and Endocrine-Disrupting Chemical Activities. J Clin Endocrinol Metab. 2019;104(11):5393-405.
  319. Kim SH, Park MJ. Effects of phytoestrogen on sexual development. Korean J Pediatr. 2012;55(8):265-71.
  320. Cleemann L, Holm K. [Precocious pseudopuberty in a seven year-old girl due to estrogen treatment of labial adhesion]. Ugeskr Laeger. 2011;173(20):1435-6.
  321. Guarneri MP, Brambilla G, Loizzo A, Colombo I, Chiumello G. Estrogen exposure in a child from hair lotion used by her mother: clinical and hair analysis data. Clin Toxicol (Phila). 2008;46(8):762-4.
  322. Martinez-Pajares JD, Diaz-Morales O, Ramos-Diaz JC, Gomez-Fernandez E. Peripheral precocious puberty due to inadvertent exposure to testosterone: case report and review of the literature. J Pediatr Endocrinol Metab. 2012;25(9-10):1007-12.
  323. Castiello F, Freire C. Exposure to non-persistent pesticides and puberty timing: a systematic review of the epidemiological evidence. Eur J Endocrinol. 2021;184(6):733-49.
  324. Watkins DJ, Sanchez BN, Tellez-Rojo MM, Lee JM, Mercado-Garcia A, Blank-Goldenberg C, et al. Phthalate and bisphenol A exposure during in utero windows of susceptibility in relation to reproductive hormones and pubertal development in girls. Environ Res. 2017;159:143-51.
  325. Lee JE, Jung HW, Lee YJ, Lee YA. Early-life exposure to endocrine-disrupting chemicals and pubertal development in girls. Ann Pediatr Endocrinol Metab. 2019;24(2):78-91.
  326. Franssen D, Svingen T, Lopez Rodriguez D, Van Duursen M, Boberg J, Parent AS. A Putative Adverse Outcome Pathway Network for Disrupted Female Pubertal Onset to Improve Testing and Regulation of Endocrine Disrupting Chemicals. Neuroendocrinology. 2022;112(2):101-14.
  327. Taylor KW, Howdeshell KL, Bommarito PA, Sibrizzi CA, Blain RB, Magnuson K, et al. Systematic evidence mapping informs a class-based approach to assessing personal care products and pubertal timing. Environ Int. 2023;181:108307.
  328. Ahn C, Jeung EB. Endocrine-Disrupting Chemicals and Disease Endpoints. Int J Mol Sci. 2023;24(6).
  329. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, et al. EDC-2: The Endocrine Society's Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr Rev. 2015;36(6):E1-E150.
  330. Uldbjerg CS, Koch T, Lim YH, Gregersen LS, Olesen CS, Andersson AM, et al. Prenatal and postnatal exposures to endocrine disrupting chemicals and timing of pubertal onset in girls and boys: a systematic review and meta-analysis. Hum Reprod Update. 2022;28(5):687-716.
  331. De Sanctis V, Rigon F, Bernasconi S, Bianchin L, Bona G, Bozzola M, et al. Age at Menarche and Menstrual Abnormalities in Adolescence: Does it Matter? The Evidence from a Large Survey among Italian Secondary Schoolgirls. Indian J Pediatr. 2019;86(Suppl 1):34-41.
  332. Reindollar RH, Byrd JR, McDonough PG. Delayed sexual development: a study of 252 patients. Am J Obstet Gynecol. 1981;140(4):371-80.
  333. Herman-Giddens ME, Steffes J, Harris D, Slora E, Hussey M, Dowshen SA, et al. Secondary sexual characteristics in boys: data from the Pediatric Research in Office Settings Network. Pediatrics. 2012;130(5):e1058-68.
  334. Jonsdottir-Lewis E, Feld A, Ciarlo R, Denhoff E, Feldman HA, Chan YM. Timing of Pubertal Onset in Girls and Boys With Constitutional Delay. J Clin Endocrinol Metab. 2021;106(9):e3693-e703.
  335. Barroso PS, Jorge AAL, Lerario AM, Montenegro LR, Vasques GA, Lima Amato LG, et al. Clinical and Genetic Characterization of a Constitutional Delay of Growth and Puberty Cohort. Neuroendocrinology. 2020;110(11-12):959-66.
  336. Sukumar SP, Bhansali A, Sachdeva N, Ahuja CK, Gorsi U, Jarial KD, et al. Diagnostic utility of testosterone priming prior to dynamic tests to differentiate constitutional delay in puberty from isolated hypogonadotropic hypogonadism. Clin Endocrinol (Oxf). 2017;86(5):717-24.
  337. Galazzi E, Improda N, Cerbone M, Soranna D, Moro M, Fatti LM, et al. Clinical benefits of sex steroids given as a priming prior to GH provocative test or as a growth-promoting therapy in peripubertal growth delays: Results of a retrospective study among ENDO-ERN centres. Clin Endocrinol (Oxf). 2021;94(2):219-28.
  338. Vezzoli V, Hrvat F, Goggi G, Federici S, Cangiano B, Quinton R, et al. Genetic architecture of self-limited delayed puberty and congenital hypogonadotropic hypogonadism. Front Endocrinol. 2023;13.
  339. Aung Y, Kokotsis V, Yin KN, Banerjee K, Butler G, Dattani MT, et al. Key features of puberty onset and progression can help distinguish self-limited delayed puberty from congenital hypogonadotrophic hypogonadism. Front Endocrinol (Lausanne). 2023;14:1226839.
  340. Gohil A, Eugster EA. Delayed and Precocious Puberty: Genetic Underpinnings and Treatments. Endocrin Metab Clin. 2020;49(4):741-+.
  341. Cassatella D, Howard SR, Acierno JS, Xu C, Papadakis GE, Santoni FA, et al. Congenital hypogonadotropic hypogonadism and constitutional delay of growth and puberty have distinct genetic architectures. European Journal of Endocrinology. 2018;178(4):377-88.
  342. Zhu J, Choa RE, Guo MH, Plummer L, Buck C, Palmert MR, et al. A shared genetic basis for self-limited delayed puberty and idiopathic hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 2015;100(4):E646-54.
  343. Cui X, Cui Y, Shi L, Luan J, Zhou X, Han J. A basic understanding of Turner syndrome: Incidence, complications, diagnosis, and treatment. Intractable Rare Dis Res. 2018;7(4):223-8.
  344. Cameron-Pimblett A, La Rosa C, King TFJ, Davies MC, Conway GS. The Turner syndrome life course project: Karyotype-phenotype analyses across the lifespan. Clin Endocrinol (Oxf). 2017;87(5):532-8.
  345. Gravholt CH, Andersen NH, Conway GS, Dekkers OM, Geffner ME, Klein KO, et al. Clinical practice guidelines for the care of girls and women with Turner syndrome: proceedings from the 2016 Cincinnati International Turner Syndrome Meeting. Eur J Endocrinol. 2017;177(3):G1-G70.
  346. Chan YM, Lippincott MF, Sales Barroso P, Alleyn C, Brodsky J, Granados H, et al. Using Kisspeptin to Predict Pubertal Outcomes for Youth With Pubertal Delay. J Clin Endocrinol Metab. 2020;105(8):e2717-25.
  347. Bojesen A, Juul S, Gravholt CH. Prenatal and postnatal prevalence of Klinefelter syndrome: a national registry study. J Clin Endocrinol Metab. 2003;88(2):622-6.
  348. Tian SY, Li Y, Zhao LM, He HY. [Clinicopathological characteristics of Klinefelter syndrome: a testicular biopsy analysis of 87 cases]. Zhonghua Bing Li Xue Za Zhi. 2023;52(4):341-6.
  349. Hamzik MP, Gropman AL, Brooks MR, Powell S, Sadeghin T, Samango-Sprouse CA. The Effect of Hormonal Therapy on the Behavioral Outcomes in 47,XXY (Klinefelter Syndrome) between 7 and 12 Years of Age. Genes (Basel). 2023;14(7).
  350. Lazos-Ochoa M, Aguirre D, Nieto K, Pena R, Palma I, Kofman S, et al. Extragonadal mediastinal germ cell tumors are often associated with Klinefelter syndrome. Modern Pathol. 2006;19:137-.
  351. Lopez Krabbe HV, Holm Petersen J, Asserhoj LL, Johannsen TH, Christiansen P, Jensen RB, et al. Reproductive hormones, bone mineral content, body composition, and testosterone therapy in boys and adolescents with Klinefelter syndrome. Endocr Connect. 2023;12(7).
  352. Rohayem J, Nieschlag E, Zitzmann M, Kliesch S. Testicular function during puberty and young adulthood in patients with Klinefelter's syndrome with and without spermatozoa in seminal fluid. Andrology. 2016;4(6):1178-86.
  353. Masterson TA, 3rd, Nassau DE, Ramasamy R. A clinical algorithm for management of fertility in adolescents with the Klinefelter syndrome. Curr Opin Urol. 2020;30(3):324-7.
  354. Vena W, Carrone F, Delbarba A, Akpojiyovbi O, Pezzaioli LC, Facondo P, et al. Body composition, trabecular bone score and vertebral fractures in subjects with Klinefelter syndrome. J Endocrinol Invest. 2023;46(2):297-304.
  355. Butler G, Srirangalingam U, Faithfull J, Sangster P, Senniappan S, Mitchell R. Klinefelter syndrome: going beyond the diagnosis. Arch Dis Child. 2023;108(3):166-71.
  356. Tanner M, Miettinen PJ, Hero M, Toppari J, Raivio T. Onset and progression of puberty in Klinefelter syndrome. Clin Endocrinol (Oxf). 2022;96(3):363-70.
  357. Cools M, Nordenstrom A, Robeva R, Hall J, Westerveld P, Fluck C, et al. Caring for individuals with a difference of sex development (DSD): a Consensus Statement. Nat Rev Endocrinol. 2018;14(7):415-29.
  358. Rosario R, Anderson R. The molecular mechanisms that underlie fragile X-associated premature ovarian insufficiency: is it RNA or protein based? Mol Hum Reprod. 2020;26(10):727-37.
  359. Biancalana V, Glaeser D, McQuaid S, Steinbach P. EMQN best practice guidelines for the molecular genetic testing and reporting of fragile X syndrome and other fragile X-associated disorders. Eur J Hum Genet. 2015;23(4):417-25.
  360. Yatsenko SA, Rajkovic A. Genetics of human female infertilitydagger. Biol Reprod. 2019;101(3):549-66.
  361. Flechtner I, Viaud M, Kariyawasam D, Perrissin-Fabert M, Bidet M, Bachelot A, et al. Puberty and fertility in classic galactosemia. Endocr Connect. 2021;10(2):240-7.
  362. Gubbels CS, Land JA, Rubio-Gozalbo ME. Fertility and impact of pregnancies on the mother and child in classic galactosemia. Obstet Gynecol Surv. 2008;63(5):334-43.
  363. Frederick AB, Zinsli AM, Carlock G, Conneely K, Fridovich-Keil JL. Presentation, progression, and predictors of ovarian insufficiency in classic galactosemia. J Inherit Metab Dis. 2018;41(5):785-90.
  364. Derks B, Rivera-Cruz G, Hagen-Lillevik S, Vos EN, Demirbas D, Lai K, et al. The hypergonadotropic hypogonadism conundrum of classic galactosemia. Hum Reprod Update. 2023;29(2):246-58.
  365. Rubio-Gozalbo ME, Gubbels CS, Bakker JA, Menheere PP, Wodzig WK, Land JA. Gonadal function in male and female patients with classic galactosemia. Hum Reprod Update. 2010;16(2):177-88.
  366. Schweitzer S, Shin Y, Jakobs C, Brodehl J. Long-term outcome in 134 patients with galactosaemia. Eur J Pediatr. 1993;152(1):36-43.
  367. Waggoner DD, Buist NR, Donnell GN. Long-term prognosis in galactosaemia: results of a survey of 350 cases. J Inherit Metab Dis. 1990;13(6):802-18.
  368. Waldstreicher J, Seminara SB, Jameson JL, Geyer A, Nachtigall LB, Boepple PA, et al. The genetic and clinical heterogeneity of gonadotropin-releasing hormone deficiency in the human. J Clin Endocrinol Metab. 1996;81(12):4388-95.
  369. Sykiotis GP, Pitteloud N, Seminara SB, Kaiser UB, Crowley WF, Jr. Deciphering genetic disease in the genomic era: the model of GnRH deficiency. Sci Transl Med. 2010;2(32):32rv2.
  370. Sykiotis GP, Plummer L, Hughes VA, Au M, Durrani S, Nayak-Young S, et al. Oligogenic basis of isolated gonadotropin-releasing hormone deficiency. Proc Natl Acad Sci U S A. 2010;107(34):15140-4.
  371. Linscott ML, Chung WCJ. Epigenomic control of gonadotrophin-releasing hormone neurone development and hypogonadotrophic hypogonadism. J Neuroendocrinol. 2020;32(6):e12860.
  372. Harrington J, Palmert MR. An Approach to the Patient With Delayed Puberty. J Clin Endocrinol Metab. 2022;107(6):1739-50.
  373. Molsted K, Kjaer I, Giwercman A, Vesterhauge S, Skakkebaek NE. Craniofacial morphology in patients with Kallmann's syndrome with and without cleft lip and palate. Cleft Palate Craniofac J. 1997;34(5):417-24.
  374. Cassatella D, Howard SR, Acierno JS, Xu C, Papadakis GE, Santoni FA, et al. Congenital hypogonadotropic hypogonadism and constitutional delay of growth and puberty have distinct genetic architectures. Eur J Endocrinol. 2018;178(4):377-88.
  375. Husebye ES, Anderson MS, Kampe O. Autoimmune Polyendocrine Syndromes. N Engl J Med. 2018;378(12):1132-41.
  376. McElreavey K, Jorgensen A, Eozenou C, Merel T, Bignon-Topalovic J, Tan DS, et al. Pathogenic variants in the DEAH-box RNA helicase DHX37 are a frequent cause of 46,XY gonadal dysgenesis and 46,XY testicular regression syndrome. Genet Med. 2020;22(1):150-9.
  377. Foster KL, Lee DJ, Witchel SF, Gordon CM. Ovarian Insufficiency and Fertility Preservation During and After Childhood Cancer Treatment. J Adolesc Young Adult Oncol. 2024.
  378. Matalliotakis M, Koliarakis I, Matalliotaki C, Trivli A, Hatzidaki E. Clinical manifestations, evaluation and management of hyperprolactinemia in adolescent and young girls: a brief review. Acta Biomed. 2019;90(1):149-57.
  379. Lee SL, Lim A, Munns C, Simm PJ, Zacharin M. Effect of Testosterone Treatment for Delayed Puberty in Duchenne Muscular Dystrophy. Horm Res Paediatr. 2020;93(2):108-18.
  380. Ward LM, Weber DR. Growth, pubertal development, and skeletal health in boys with Duchenne Muscular Dystrophy. Curr Opin Endocrinol Diabetes Obes. 2019;26(1):39-48.
  381. Esquivel-Zuniga R, Rogol AD. Functional hypogonadism in adolescence: an overlooked cause of secondary hypogonadism. Endocr Connect. 2023;12(11).
  382. Thavaraputta S, Ungprasert P, Witchel SF, Fazeli PK. Anorexia nervosa and adrenal hormones: a systematic review and meta-analysis. Eur J Endocrinol. 2023;189(3):S64-S73.
  383. Misra M, Golden NH, Katzman DK. State of the art systematic review of bone disease in anorexia nervosa. Int J Eat Disord. 2016;49(3):276-92.
  384. Odle AK, Akhter N, Syed MM, Allensworth-James ML, Benes H, Melgar Castillo AI, et al. Leptin Regulation of Gonadotrope Gonadotropin-Releasing Hormone Receptors As a Metabolic Checkpoint and Gateway to Reproductive Competence. Front Endocrinol (Lausanne). 2017;8:367.
  385. Vazquez MJ, Velasco I, Tena-Sempere M. Novel mechanisms for the metabolic control of puberty: implications for pubertal alterations in early-onset obesity and malnutrition. J Endocrinol. 2019;242(2):R51-R65.
  386. Cano Sokoloff N, Misra M, Ackerman KE. Exercise, Training, and the Hypothalamic-Pituitary-Gonadal Axis in Men and Women. Front Horm Res. 2016;47:27-43.
  387. Gordon CM, Ackerman KE, Berga SL, Kaplan JR, Mastorakos G, Misra M, et al. Functional Hypothalamic Amenorrhea: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2017;102(5):1413-39.
  388. De Lorenzo A, Noce A, Moriconi E, Rampello T, Marrone G, Di Daniele N, et al. MOSH Syndrome (Male Obesity Secondary Hypogonadism): Clinical Assessment and Possible Therapeutic Approaches. Nutrients. 2018;10(4).
  389. Federici S, Goggi G, Quinton R, Giovanelli L, Persani L, Cangiano B, et al. New and Consolidated Therapeutic Options for Pubertal Induction in Hypogonadism: In-depth Review of the Literature. Endocr Rev. 2022;43(5):824-51.
  390. Szeliga A, Podfigurna A, Bala G, Meczekalski B. Kisspeptin and neurokinin B analogs use in gynecological endocrinology: where do we stand? J Endocrinol Invest. 2020;43(5):555-61.
  391. Alexander EC, Faruqi D, Farquhar R, Unadkat A, Ng Yin K, Hoskyns R, et al. Gonadotropins for pubertal induction in males with hypogonadotropic hypogonadism: systematic review and meta-analysis. Eur J Endocrinol. 2024;190(1):S1-S11.
  392. Stancampiano MR, Lucas-Herald AK, Russo G, Rogol AD, Ahmed SF. Testosterone Therapy in Adolescent Boys: The Need for a Structured Approach. Horm Res Paediatr. 2019;92(4):215-28.
  393. Raivio T, Falardeau J, Dwyer A, Quinton R, Hayes FJ, Hughes VA, et al. Reversal of idiopathic hypogonadotropic hypogonadism. N Engl J Med. 2007;357(9):863-73.
  394. Sun T, Xu W, Chen Y, Niu Y, Wang T, Wang S, et al. Reversal of idiopathic hypogonadotropic hypogonadism in a Chinese male cohort. Andrologia. 2022;54(11):e14583.
  395. Klein KO, Phillips SA. Review of Hormone Replacement Therapy in Girls and Adolescents with Hypogonadism. J Pediatr Adolesc Gynecol. 2019;32(5):460-8.
  396. Cheuiche AV, da Silveira LG, de Paula LCP, Lucena IRS, Silveiro SP. Diagnosis and management of precocious sexual maturation: an updated review. Eur J Pediatr. 2021;180(10):3073-87.
  397. Ibanez L, Ferrer A, Marcos MV, Hierro FR, de Zegher F. Early puberty: rapid progression and reduced final height in girls with low birth weight. Pediatrics. 2000;106(5):E72.
  398. Young J, Xu C, Papadakis GE, Acierno JS, Maione L, Hietamaki J, et al. Clinical Management of Congenital Hypogonadotropic Hypogonadism. Endocr Rev. 2019;40(2):669-710.
  399. Varimo T, Hero M, Laitinen EM, Miettinen PJ, Tommiska J, Kansakoski J, et al. Childhood growth in boys with congenital hypogonadotropic hypogonadism. Pediatr Res. 2016;79(5):705-9.
  400. Varimo T, Miettinen PJ, Kansakoski J, Raivio T, Hero M. Congenital hypogonadotropic hypogonadism, functional hypogonadotropism or constitutional delay of growth and puberty? An analysis of a large patient series from a single tertiary center. Hum Reprod. 2017;32(1):147-53.
  401. Neely EK, Hintz RL, Wilson DM, Lee PA, Gautier T, Argente J, et al. Normal ranges for immunochemiluminometric gonadotropin assays. J Pediatr. 1995;127(1):40-6.
  402. Martinez-Aguayo A, Hernández MI, Capurro T, Peña V, Avila A, Salazar T, et al. Leuprolide acetate gonadotrophin response patterns during female puberty. Clin Endocrinol (Oxf). 2010;72(4):489-95.
  403. Cao R, Liu J, Fu P, Zhou Y, Li Z, Liu P. The Diagnostic Utility of the Basal Luteinizing Hormone Level and Single 60-Minute Post GnRH Agonist Stimulation Test for Idiopathic Central Precocious Puberty in Girls. Front Endocrinol (Lausanne). 2021;12:713880.
  404. Howard SR. Interpretation of reproductive hormones before, during and after the pubertal transition-Identifying health and disordered puberty. Clin Endocrinol (Oxf). 2021;95(5):702-15.
  405. Bizzarri C, Spadoni GL, Bottaro G, Montanari G, Giannone G, Cappa M, et al. The response to gonadotropin releasing hormone (GnRH) stimulation test does not predict the progression to true precocious puberty in girls with onset of premature thelarche in the first three years of life. J Clin Endocrinol Metab. 2014;99(2):433-9.
  406. Vestergaard ET, Schjorring ME, Kamperis K, Petersen KK, Rittig S, Juul A, et al. The follicle-stimulating hormone (FSH) and luteinizing hormone (LH) response to a gonadotropin-releasing hormone analogue test in healthy prepubertal girls aged 10 months to 6 years. Eur J Endocrinol. 2017;176(6):747-53.
  407. Harrington J, Palmert MR. Clinical review: Distinguishing constitutional delay of growth and puberty from isolated hypogonadotropic hypogonadism: critical appraisal of available diagnostic tests. J Clin Endocrinol Metab. 2012;97(9):3056-67.
  408. Degros V, Cortet-Rudelli C, Soudan B, Dewailly D. The human chorionic gonadotropin test is more powerful than the gonadotropin-releasing hormone agonist test to discriminate male isolated hypogonadotropic hypogonadism from constitutional delayed puberty. European Journal of Endocrinology. 2003;149(1):23-9.
  409. Chaudhary S, Walia R, Bhansali A, Dayal D, Sachdeva N, Singh T, et al. FSH-stimulated Inhibin B (FSH-iB): A Novel Marker for the Accurate Prediction of Pubertal Outcome in Delayed Puberty. J Clin Endocrinol Metab. 2021;106(9):e3495-e505.
  410. Turcu AF, Mallappa A, Nella AA, Chen X, Zhao L, Nanba AT, et al. 24-Hour Profiles of 11-Oxygenated C(19) Steroids and Delta(5)-Steroid Sulfates during Oral and Continuous Subcutaneous Glucocorticoids in 21-Hydroxylase Deficiency. Front Endocrinol (Lausanne). 2021;12:751191.
  411. Trost LW, Mulhall JP. Challenges in Testosterone Measurement, Data Interpretation, and Methodological Appraisal of Interventional Trials. J Sex Med. 2016;13(7):1029-46.
  412. French D. Clinical utility of laboratory developed mass spectrometry assays for steroid hormone testing. J Mass Spectrom Adv Clin Lab. 2023;28:13-9.
  413. Chin VL, Cai Z, Lam L, Shah B, Zhou P. Evaluation of puberty by verifying spontaneous and stimulated gonadotropin values in girls. J Pediatr Endocrinol Metab. 2015;28(3-4):387-92.
  414. Abbara A, Eng PC, Phylactou M, Clarke SA, Mills E, Chia G, et al. Kisspeptin-54 Accurately Identifies Hypothalamic Gonadotropin-Releasing Hormone Neuronal Dysfunction in Men with Congenital Hypogonadotropic Hypogonadism. Neuroendocrinology. 2021;111(12):1176-86.
  415. Rosenfield RL. Normal and Premature Adrenarche. Endocr Rev. 2021;42(6):783-814.
  416. Chesover AD, Millar H, Sepiashvili L, Adeli K, Palmert MR, Hamilton J. Screening for Nonclassic Congenital Adrenal Hyperplasia in the Era of Liquid Chromatography-Tandem Mass Spectrometry. J Endocr Soc. 2020;4(2):bvz030.
  417. Gao Y, Du Q, Liu L, Liao Z. Serum inhibin B for differentiating between congenital hypogonadotropic hypogonadism and constitutional delay of growth and puberty: a systematic review and meta-analysis. Endocrine. 2021;72(3):633-43.
  418. Binder G, Schweizer R, Blumenstock G, Braun R. Inhibin B plus LH vs GnRH agonist test for distinguishing constitutional delay of growth and puberty from isolated hypogonadotropic hypogonadism in boys. Clin Endocrinol (Oxf). 2015;82(1):100-5.
  419. Mosbah H, Bouvattier C, Maione L, Trabado S, De Filippo G, Cartes A, et al. GnRH stimulation testing and serum inhibin B in males: insufficient specificity for discriminating between congenital hypogonadotropic hypogonadism from constitutional delay of growth and puberty. Hum Reprod. 2020;35(10):2312-22.
  420. Albrethsen J, Ljubicic ML, Juul A. Longitudinal Increases in Serum Insulin-like Factor 3 and Testosterone Determined by LC-MS/MS in Pubertal Danish Boys. J Clin Endocrinol Metab. 2020;105(10).
  421. Segal TY, Mehta A, Anazodo A, Hindmarsh PC, Dattani MT. Role of gonadotropin-releasing hormone and human chorionic gonadotropin stimulation tests in differentiating patients with hypogonadotropic hypogonadism from those with constitutional delay of growth and puberty. J Clin Endocrinol Metab. 2009;94(3):780-5.
  422. Greulich WW PS. Radiographic Atlas of Skeletal Development of the Hand and Wrist. 2nd ed: Stanford University Press; 1999.
  423. Tanner JM WR, Cameron N, et al.,. Assessment of Skeletal Maturity and Prediction of Adult Height (TW2 Method). 2nd ed: Goldstein H (Ed), Academic Press; 1983.
  424. Klein KO, Newfield RS, Hassink SG. Bone maturation along the spectrum from normal weight to obesity: a complex interplay of sex, growth factors and weight gain. J Pediatr Endocrinol Metab. 2016;29(3):311-8.
  425. Oh MS, Kim S, Lee J, Lee MS, Kim YJ, Kang KS. Factors associated with Advanced Bone Age in Overweight and Obese Children. Pediatr Gastroentero. 2020;23(1):89-97.
  426. Bar A, Linder B, Sobel EH, Saenger P, Dimartinonardi J. Bayley-Pinneau Method of Height Prediction in Girls with Central Precocious Puberty - Correlation with Adult Height. J Pediatr-Us. 1995;126(6):955-8.
  427. Eitel KB, Eugster EA. Differences in Bone Age Readings between Pediatric Endocrinologists and Radiologists. Endocrine Practice. 2020;26(3):328-31.
  428. Wang X, Zhou B, Gong P, Zhang T, Mo Y, Tang J, et al. Artificial Intelligence-Assisted Bone Age Assessment to Improve the Accuracy and Consistency of Physicians With Different Levels of Experience. Frontiers in Pediatrics. 2022;10.
  429. Thodberg HH, Thodberg B, Ahlkvist J, Offiah AC. Autonomous artificial intelligence in pediatric radiology: the use and perception of BoneXpert for bone age assessment. Pediatr Radiol. 2022;52(7):1338-46.
  430. de Vries L, Horev G, Schwartz M, Phillip M. Ultrasonographic and clinical parameters for early differentiation between precocious puberty and premature thelarche. Eur J Endocrinol. 2006;154(6):891-8.
  431. Sathasivam A, Rosenberg HK, Shapiro S, Wang H, Rapaport R. Pelvic ultrasonography in the evaluation of central precocious puberty: comparison with leuprolide stimulation test. J Pediatr. 2011;159(3):490-5.
  432. Lee SH, Joo EY, Lee JE, Jun YH, Kim MY. The Diagnostic Value of Pelvic Ultrasound in Girls with Central Precocious Puberty. Chonnam Med J. 2016;52(1):70-4.
  433. Battaglia C, Mancini F, Regnani G, Persico N, Iughetti L, De Aloysio D. Pelvic ultrasound and color Doppler findings in different isosexual precocities. Ultrasound Obstet Gynecol. 2003;22(3):277-83.
  434. Long MG, Boultbee JE, Hanson ME, Begent RH. Doppler time velocity waveform studies of the uterine artery and uterus. Br J Obstet Gynaecol. 1989;96(5):588-93.
  435. Paesano PL, Colantoni C, Mora S, di Lascio A, Ferrario M, Esposito A, et al. Validation of an Accurate and Noninvasive Tool to Exclude Female Precocious Puberty: Pelvic Ultrasound With Uterine Artery Pulsatility Index. Am J Roentgenol. 2019;213(2):451-7.
  436. Cheuiche AV, Moro C, Lucena IRS, de Paula LCP, Silveiro SP. Accuracy of doppler assessment of the uterine arteries for the diagnosis of pubertal onset in girls: a scoping review. Sci Rep. 2023;13(1):5791.
  437. Kafi SE, Alagha E, Shazly MA, Al-Agha A. Pseudo-precocious Puberty Associated with an Adrenocortical Tumor in a Young Child. Cureus. 2019;11(12).
  438. Alagha E, Kafi SE, Shazly MA, Al-Agha A. Precocious Puberty Associated with Testicular Hormone-secreting Leydig Cell Tumor. Cureus. 2019;11(12):e6441.
  439. Eyer de Jesus L, Paz de Oliveira AP, Porto LC, Dekermacher S. Testicular adrenal rest tumors - Epidemiology, diagnosis and treatment. J Pediatr Urol. 2023.
  440. Cantas-Orsdemir S, Garb JL, Allen HF. Prevalence of cranial MRI findings in girls with central precocious puberty: a systematic review and meta-analysis. J Pediatr Endocrinol Metab. 2018;31(7):701-10.
  441. Pedicelli S, Alessio P, Scire G, Cappa M, Cianfarani S. Routine screening by brain magnetic resonance imaging is not indicated in every girl with onset of puberty between the ages of 6 and 8 years. J Clin Endocrinol Metab. 2014;99(12):4455-61.
  442. Kaplowitz PB. Do 6-8 year old girls with central precocious puberty need routine brain imaging? Int J Pediatr Endocrinol. 2016;2016:9.
  443. Mogensen SS, Aksglaede L, Mouritsen A, Sorensen K, Main KM, Gideon P, et al. Pathological and Incidental Findings on Brain MRI in a Single-Center Study of 229 Consecutive Girls with Early or Precocious Puberty. Plos One. 2012;7(1).
  444. Hansen AB, Renault CH, Wojdemann D, Gideon P, Juul A, Jensen RB. Neuroimaging in 205 consecutive Children Diagnosed with Central Precocious Puberty in Denmark. Pediatr Res. 2023;93(1):125-30.
  445. Kim SH, Ahn MB, Cho WK, Cho KS, Jung MH, Suh BK. Findings of Brain Magnetic Resonance Imaging in Girls with Central Precocious Puberty Compared with Girls with Chronic or Recurrent Headache. J Clin Med. 2021;10(10).
  446. Eugster EA. Update on Precocious Puberty in Girls. J Pediatr Adol Gynec. 2019;32(5):455-9.
  447. Yoon JS, So CH, Lee HS, Lim JS, Hwang JS. The prevalence of brain abnormalities in boys with central precocious puberty may be overestimated. Plos One. 2018;13(4).
  448. Kang JY, Kim SH, Kim H, Ki H, Lee MH. Pituitary magnetic resonance imaging abnormalities in young female patients with hypogonadotropic hypogonadism. Obstet Gynecol Sci. 2019;62(4):249-57.
  449. Hacquart T, Ltaief-Boudrigua A, Jeannerod C, Hannoun S, Raverot G, Pugeat M, et al. Reconsidering olfactory bulb magnetic resonance patterns in Kallmann syndrome. Ann Endocrinol (Paris). 2017;78(5):455-61.
  450. Sarfati J, Saveanu A, Young J. Pituitary stalk interruption and olfactory bulbs aplasia/hypoplasia in a man with Kallmann syndrome and reversible gonadotrope and somatotrope deficiencies. Endocrine. 2015;49(3):865-6.
  451. Elnaw EAA, Ibrahim AAB, Abdullah MA. Feminizing adrenocortical adenoma in a girl from a resource-limited setting: a case report. J Med Case Rep. 2021;15(1).
  452. Bulut S, Catli G, Filibeli BE, Manyas H, Ayranci I, Meral R, et al. Virilizing Adrenocortical Carcinoma Oncocytic Variant in a Child with Heterosexual Precocious Puberty and a Literature Review. J Pediatr Res. 2022;9(3):307-13.
  453. Ko JH, Lee HS, Hong J, Hwang JS. Virilizing adrenocortical carcinoma in a child with Turner syndrome and somatic TP53 gene mutation. European Journal of Pediatrics. 2010;169(4):501-4.
  454. Elnaw EAA, Ibrahim AAB, Abdullah MA. Feminizing adrenocortical adenoma in a girl from a resource-limited setting: a case report. J Med Case Rep. 2021;15(1):605.
  455. Gravholt CH, Viuff M, Just J, Sandahl K, Brun S, van der Velden J, et al. The Changing Face of Turner Syndrome. Endocr Rev. 2023;44(1):33-69.
  456. Correa Brito L, Rey RA. Taming Idiopathic Central Precocious Puberty: High Frequency of Imprinting Disorders in Familial Forms. J Clin Endocrinol Metab. 2023;108(8):e636-e7.
  457. Aguirre RS, Eugster EA. Central precocious puberty: From genetics to treatment. Best Pract Res Cl En. 2018;32(4):343-54.
  458. Howard SR, Dunkel L. Delayed Puberty-Phenotypic Diversity, Molecular Genetic Mechanisms, and Recent Discoveries. Endocr Rev. 2019;40(5):1285-317.
  459. Howard SR. Genes underlying delayed puberty. Mol Cell Endocrinol. 2018;476:119-28.
  460. Villanueva C, Jacobson-Dickman E, Xu C, Manouvrier S, Dwyer AA, Sykiotis GP, et al. Congenital hypogonadotropic hypogonadism with split hand/foot malformation: a clinical entity with a high frequency of FGFR1 mutations. Genet Med. 2015;17(8):651-9.
  461. LATRONICO A. Deciphering the genetic basis of central precocious puberty. Rev Esp Endocrinol Pediatr. 2023;14 Suppl(2):17-20
  462. Stecchini MF, Macedo DB, Reis AC, Abreu AP, Moreira AC, Castro M, et al. Time Course of Central Precocious Puberty Development Caused by an MKRN3 Gene Mutation: A Prismatic Case. Horm Res Paediatr. 2016;86(2):126-30.
  463. Teles MG, Bianco SDC, Brito VN, Trarbach EB, Kuohung W, Xu SY, et al. A GPR54-activating mutation in a patient with central precocious puberty. New Engl J Med. 2008;358(7):709-15.
  464. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Shagoury JK, et al. The GPR54 gene as a regulator of puberty. New Engl J Med. 2003;349(17):1614-U8.
  465. Krstevska-Konstantinova M, Jovanovska J, Tasic VB, Montenegro LR, Beneduzzi D, Silveira LF, et al. Mutational analysis of KISS1 and KISS1R in idiopathic central precocious puberty. J Pediatr Endocrinol Metab. 2014;27(1-2):199-201.
  466. Grandone A, Capristo C, Cirillo G, Sasso M, Umano GR, Mariani M, et al. Molecular Screening of MKRN3, DLK1, and KCNK9 Genes in Girls with Idiopathic Central Precocious Puberty. Horm Res Paediatr. 2017;88(3-4):194-200.
  467. Macedo DB, Kaiser UB. DLK1, Notch Signaling and the Timing of Puberty. Semin Reprod Med. 2019;37(4):174-81.
  468. Montenegro L, Labarta JI, Piovesan M, Canton APM, Corripio R, Soriano-Guillen L, et al. Novel Genetic and Biochemical Findings of DLK1 in Children with Central Precocious Puberty: A Brazilian-Spanish Study. J Clin Endocrinol Metab. 2020;105(10).
  469. Soares JM, de Holanda FS, Matsuzaki CN, Sorpreso ICE, Veiga ECD, de Abreu LC, et al. Analysis of the PvuII and XbaI polymorphisms in the estrogen receptor alpha gene in girls with central precocious puberty: a pilot study. Bmc Med Genet. 2018;19.
  470. Lee HS, Park HK, Kim KH, Ko JH, Kim YJ, Yi KH, et al. Estrogen receptor alpha gene analysis in girls with central precocious puberty. J Pediatr Endocr Met. 2013;26(7-8):645-9.
  471. Lee HS, Kim KH, Hwang JS. Association of aromatase (TTTA)n repeat polymorphisms with central precocious puberty in girls. Clin Endocrinol (Oxf). 2014;81(3):395-400.
  472. Tsuji-Hosokawa A, Matsuda N, Kurosawa K, Kashimada K, Morio T. A Case of MECP2 Duplication Syndrome with Gonadotropin-Dependent Precocious Puberty. Horm Res Paediatr. 2017;87(4):271-6.
  473. Canton APM, Krepischi ACV, Montenegro LR, Costa S, Rosenberg C, Steunou V, et al. Insights from the genetic characterization of central precocious puberty associated with multiple anomalies. Hum Reprod. 2021;36(2):506-18.
  474. Czako M, Till A, Zima J, Zsigmond A, Szabo A, Maasz A, et al. Xp11.2 Duplication in Females: Unique Features of a Rare Copy Number Variation. Front Genet. 2021;12:635458.
  475. Ludwig NG, Radaeli RF, da Silva MMX, Romero CM, Carrilho AJF, Bessa D, et al. A boy with Prader-Willi syndrome: unmasking precocious puberty during growth hormone replacement therapy. Arch Endocrin Metab. 2016;60(6):596-600.
  476. Hoffmann K, Heller R. Uniparental disomies 7 and 14. Best Pract Res Clin Endocrinol Metab. 2011;25(1):77-100.
  477. Partsch CJ, Japing I, Siebert R, Gosch A, Wessel A, Sippell WG, et al. Central precocious puberty in girls with Williams syndrome. J Pediatr. 2002;141(3):441-4.
  478. Utine GE, Alikasifoglu A, Alikasifoglu M, Tuncbilek E. Central precocious puberty in a girl with Williams syndrome: the result of treatment with GnRH analogue. Eur J Med Genet. 2006;49(1):79-82.
  479. Kuroki Y, Katsumata N, Eguchi T, Fukushima Y, Suwa S, Kajii T. Precocious puberty in Kabuki makeup syndrome. J Pediatr. 1987;110(5):750-2.
  480. Concolino D, Muzzi G, Pisaturo L, Piccirillo A, Di Natale P, Strisciuglio P. Precocious puberty in Sanfilippo IIIA disease: diagnosis and follow-up of two new cases. Eur J Med Genet. 2008;51(5):466-71.
  481. Ray LA, Eckert GJ, Eugster EA. Long-term experience with the use of a single histrelin implant beyond one year in patients with central precocious puberty. J Pediatr Endocrinol Metab. 2023;36(3):309-12.
  482. Saengkaew T, Howard SR. Genetics of pubertal delay. Clin Endocrinol (Oxf). 2022;97(4):473-82.
  483. Akram M, Raza Rizvi SS, Qayyum M, Handelsman DJ. A classification of genes involved in normal and delayed male puberty. Asian J Androl. 2023;25(2):230-9.
  484. Vilain C, Mortier G, Van Vliet G, Dubourg C, Heinrichs C, de Silva D, et al. Hartsfield Holoprosencephaly-Ectrodactyly Syndrome in Five Male Patients: Further Delineation and Review. American Journal of Medical Genetics Part A. 2009;149a(7):1476-81.
  485. Pitteloud N, Meysing A, Quinton R, Acierno J, Dwyer A, Plummer L, et al. Mutations in fibroblast growth factor receptor 1 cause Kallmann syndrome with a wide spectrum of reproductive phenotypes. Molecular and Cellular Endocrinology. 2006:60-9.
  486. Quennell JH, Mulligan AC, Tups A, Liu X, Phipps SJ, Kemp CJ, et al. Leptin indirectly regulates gonadotropin-releasing hormone neuronal function. Endocrinology. 2009;150(6):2805-12.
  487. Williams KW, Sohn JW, Donato J, Lee CE, Zhao JJ, Elmquist JK, et al. The Acute Effects of Leptin Require PI3K Signaling in the Hypothalamic Ventral Premammillary Nucleus. J Neurosci. 2011;31(37):13147-56.
  488. Donato J, Cravo RM, Frazao R, Elias CF. Hypothalamic Sites of Leptin Action Linking Metabolism and Reproduction. Neuroendocrinology. 2011;93(1):9-18.
  489. Farooqi IS, O'Rahilly S. Mutations in ligands and receptors of the leptin-melanocortin pathway that lead to obesity. Nat Clin Pract Endoc. 2008;4(10):569-77.
  490. Tata B, Huijbregts L, Jacquier S, Csaba Z, Genin E, Meyer V, et al. Haploinsufficiency of Dmxl2, encoding a synaptic protein, causes infertility associated with a loss of GnRH neurons in mouse. PLoS Biol. 2014;12(9):e1001952.
  491. Margolin DH, Kousi M, Chan YM, Lim ET, Schmahmann JD, Hadjivassiliou M, et al. Ataxia, dementia, and hypogonadotropism caused by disordered ubiquitination. N Engl J Med. 2013;368(21):1992-2003.
  492. Synofzik M, Kernstock C, Haack TB, Schols L. Ataxia meets chorioretinal dystrophy and hypogonadism: Boucher-Neuhauser syndrome due to PNPLA6 mutations. J Neurol Neurosurg Psychiatry. 2015;86(5):580-1.
  493. Synofzik M, Gonzalez MA, Lourenco CM, Coutelier M, Haack TB, Rebelo A, et al. PNPLA6 mutations cause Boucher-Neuhauser and Gordon Holmes syndromes as part of a broad neurodegenerative spectrum. Brain. 2014;137(Pt 1):69-77.
  494. Pingault V, Bodereau V, Baral V, Marcos S, Watanabe Y, Chaoui A, et al. Loss-of-function mutations in SOX10 cause Kallmann syndrome with deafness. Am J Hum Genet. 2013;92(5):707-24.
  495. Tziaferi V, Kelberman D, Dattani MT. The role of SOX2 in hypogonadotropic hypogonadism. Sex Dev. 2008;2(4-5):194-9.
  496. Alatzoglou KS, Azriyanti A, Rogers N, Ryan F, Curry N, Noakes C, et al. SOX3 deletion in mouse and human is associated with persistence of the craniopharyngeal canal. J Clin Endocrinol Metab. 2014;99(12):E2702-8.
  497. Boehm U, Bouloux PM, Dattani MT, de Roux N, Dode C, Dunkel L, et al. Expert consensus document: European Consensus Statement on congenital hypogonadotropic hypogonadism--pathogenesis, diagnosis and treatment. Nat Rev Endocrinol. 2015;11(9):547-64.
  498. Joustra SD, Wehkalampi K, Oostdijk W, Biermasz NR, Howard S, Silander TL, et al. IGSF1 variants in boys with familial delayed puberty. Eur J Pediatr. 2015;174(5):687-92.
  499. Niederberger C. Re: Identification of HESX1 mutations in Kallmann syndrome. J Urol. 2014;191(4):1081.
  500. Kim HG, Kurth I, Lan F, Meliciani I, Wenzel W, Eom SH, et al. Mutations in CHD7, encoding a chromatin-remodeling protein, cause idiopathic hypogonadotropic hypogonadism and Kallmann syndrome. Am J Hum Genet. 2008;83(4):511-9.
  501. Marcos S, Sarfati J, Leroy C, Fouveaut C, Parent P, Metz C, et al. The Prevalence of CHD7 Missense Versus Truncating Mutations is Higher in Patients With Kallmann Syndrome than in Typical CHARGE Patients (vol 99, pg E2138, 2014). J Clin Endocr Metab. 2015;100(1):317-.
  502. Balasubramanian R, Choi JH, Francescatto L, Willer J, Horton ER, Asimacopoulos EP, et al. Functionally compromised CHD7 alleles in patients with isolated GnRH deficiency. P Natl Acad Sci USA. 2014;111(50):17953-8.
  503. Timmons M, Tsokos M, Asab MA, Seminara SB, Zirzow GC, Kaneski CR, et al. Peripheral and central hypomyelination with hypogonadotropic hypogonadism and hypodontia. Neurology. 2006;67(11):2066-9.
  504. Sato I, Onuma A, Goto N, Sakai F, Fujiwara I, Uematsu M, et al. A case with central and peripheral hypomyelination with hypogonadotropic hypogonadism and hypodontia (4H syndrome) plus cataract. J Neurol Sci. 2011;300(1-2):179-81.
  505. Pelletier F, Perrier S, Cayami FK, Mirchi A, Saikali S, Tran LT, et al. Endocrine and Growth Abnormalities in 4H Leukodystrophy Caused by Variants in POLR3A, POLR3B, and POLR1C. J Clin Endocrinol Metab. 2021;106(2):e660-e74.
  506. Liu S, Yan L, Zhou X, Chen C, Wang D, Yuan G. Delayed-onset adrenal hypoplasia congenita and hypogonadotropic hypogonadism caused by a novel mutation in DAX1. J Int Med Res. 2020;48(2):300060519882151.
  507. Muscatelli F, Strom TM, Walker AP, Zanaria E, Recan D, Meindl A, et al. Mutations in the Dax-1 Gene Give Rise to Both X-Linked Adrenal Hypoplasia Congenita and Hypogonadotropic Hypogonadism. Nature. 1994;372(6507):672-6.
  508. Jennings JE, Costigan C, Reardon W. Moebius sequence and hypogonadotrophic hypogonadism. Am J Med Genet A. 2003;123A(1):107-10.
  509. Alves C, Franco RR. Prader-Willi syndrome: endocrine manifestations and management. Arch Endocrinol Metab. 2020;64(3):223-34.
  510. Emerick JE, Vogt KS. Endocrine manifestations and management of Prader-Willi syndrome. Int J Pediatr Endocrinol. 2013;2013(1):14.
  511. Forsythe E, Beales PL. Bardet-Biedl syndrome. Eur J Hum Genet. 2013;21(1):8-13.
  512. Desai A, Jha O, Iyer V, Dada R, Kumar R, Tandon N. Reversible hypogonadism in Bardet-Biedl syndrome. Fertil Steril. 2009;92(1):391 e13-5.
  513. Jain V, Foo SH, Chooi S, Moss C, Goodwin R, Berland S, et al. Borjeson-Forssman-Lehmann syndrome: delineating the clinical and allelic spectrum in 14 new families. Eur J Hum Genet. 2023;31(12):1421-9.
  514. Wang T, Ren W, Fu F, Wang H, Li Y, Duan J. Digenic CHD7 and SMCHD1 inheritance Unveils phenotypic variability in a family mainly presenting with hypogonadotropic hypogonadism. Heliyon. 2024;10(1):e23272.
  515. Alavi O, Khamirani HJ, Zoghi S, Feili A, Dastgheib SA, Tabei SMB, et al. Two novel Warburg micro syndrome 1 cases caused by pathogenic variants in RAB3GAP1. Hum Genome Var. 2021;8(1):39.
  516. Kline AD, Moss JF, Selicorni A, Bisgaard AM, Deardorff MA, Gillett PM, et al. Diagnosis and management of Cornelia de Lange syndrome: first international consensus statement. Nat Rev Genet. 2018;19(10):649-66.
  517. Miraoui H, Dwyer AA, Sykiotis GP, Plummer L, Chung W, Feng BH, et al. Mutations in FGF17, IL17RD, DUSP6, SPRY4, and FLRT3 Are Identified in Individuals with Congenital Hypogonadotropic Hypogonadism. American Journal of Human Genetics. 2013;92(5):725-43.
  518. Bianco SDC, Kaiser UB. The genetic and molecular basis of idiopathic hypogonadotropic hypogonadism. Nature Reviews Endocrinology. 2009;5(10):569-76.
  519. Gonzalez-Martinez D, Kim SH, Hu YL, Guimond S, Schofield J, Winyard P, et al. Anosmin-1 modulates fibroblast growth factor receptor 1 signaling in human gonadotropin-releasing hormone olfactory neuroblasts through a heparan sulfate-dependent mechanism. J Neurosci. 2004;24(46):10384-92.
  520. Maione L, Albarel F, Bouchard P, Gallant M, Flanagan CA, Bobe R, et al. R31C GNRH1 mutation and congenital hypogonadotropic hypogonadism. Plos One. 2013;8(7):e69616.
  521. Bouligand J, Ghervan C, Tello JA, Brailly-Tabard S, Salenave S, Chanson P, et al. Isolated Familial Hypogonadotropic Hypogonadism and a GNRH1 Mutation. New Engl J Med. 2009;360(26):2742-8.
  522. Layman LC, Cohen DP, Jin M, Xie J, Li Z, Reindollar RH, et al. Mutations in gonadotropin-releasing hormone receptor gene cause hypogonadotropic hypogonadism. Nature Genetics. 1998;18(1):14-5.
  523. Topaloglu AK, Tello JA, Kotan LD, Ozbek MN, Yilmaz MB, Erdogan S, et al. Inactivating KISS1 mutation and hypogonadotropic hypogonadism. N Engl J Med. 2012;366(7):629-35.
  524. Xu C, Messina A, Somm E, Miraoui H, Kinnunen T, Acierno J, et al. KLB, encoding beta-Klotho, is mutated in patients with congenital hypogonadotropic hypogonadism. Embo Molecular Medicine. 2017;9(10):1379-97.
  525. Fergani C, Navarro VM. Expanding the Role of Tachykinins in the Neuroendocrine Control of Reproduction. Reproduction. 2016;153(1):R1-R14.
  526. Hanchate NK, Giacobini P, Lhuillier P, Parkash J, Espy C, Fouveaut C, et al. SEMA3A, a Gene Involved in Axonal Pathfinding, Is Mutated in Patients with Kallmann Syndrome. Plos Genetics. 2012;8(8).
  527. Abreu AP, Trarbach EB, de Castro M, Frade Costa EM, Versiani B, Matias Baptista MT, et al. Loss-of-function mutations in the genes encoding prokineticin-2 or prokineticin receptor-2 cause autosomal recessive Kallmann syndrome. J Clin Endocrinol Metab. 2008;93(10):4113-8.
  528. Dode C, Teixeira L, Levilliers J, Fouveaut C, Bouchard P, Kottler ML, et al. Kallmann syndrome: mutations in the genes encoding prokineticin-2 and prokineticin receptor-2. PLoS Genet. 2006;2(10):e175.
  529. Kim HG, Ahn JW, Kurth I, Ullmann R, Kim HT, Kulharya A, et al. WDR11, a WD Protein that Interacts with Transcription Factor EMX1, Is Mutated in Idiopathic Hypogonadotropic Hypogonadism and Kallmann Syndrome. American Journal of Human Genetics. 2010;87(4):465-79.
  530. Saengkaew T, Ruiz-Babot G, David A, Mancini A, Mariniello K, Cabrera CP, et al. Whole exome sequencing identifies deleterious rare variants in CCDC141 in familial self-limited delayed puberty. NPJ Genom Med. 2021;6(1):107.
  531. Kotan LD, Hutchins BI, Ozkan Y, Demirel F, Stoner H, Cheng PJ, et al. Mutations in FEZF1 cause Kallmann syndrome. Am J Hum Genet. 2014;95(3):326-31.
  532. Lofrano-Porto A, Barra GB, Giacomini LA, Nascimento PP, Latronico AC, Casulari LA, et al. Luteinizing hormone beta mutation and hypogonadism in men and women. N Engl J Med. 2007;357(9):897-904.
  533. Layman LC, Porto AL, Xie J, da Motta LA, da Motta LD, Weiser W, et al. FSH beta gene mutations in a female with partial breast development and a male sibling with normal puberty and azoospermia. J Clin Endocrinol Metab. 2002;87(8):3702-7.
  534. Berger K, Souza H, Brito VN, d'Alva CB, Mendonca BB, Latronico AC. Clinical and hormonal features of selective follicle-stimulating hormone (FSH) deficiency due to FSH beta-subunit gene mutations in both sexes. Fertil Steril. 2005;83(2):466-70.
  535. Mancini A, Howard SR, Cabrera CP, Barnes MR, David A, Wehkalampi K, et al. EAP1 regulation of GnRH promoter activity is important for human pubertal timing. Human Molecular Genetics. 2019;28(8):1357-68.
  536. Styrkarsdottir U, Thorleifsson G, Sulem P, Gudbjartsson DF, Sigurdsson A, Jonasdottir A, et al. Nonsense mutation in the LGR4 gene is associated with several human diseases and other traits. Nature. 2013;497(7450):517-20.
  537. Zang S, Yin X, Li P. FTO-mediated m(6)A demethylation regulates GnRH expression in the hypothalamus via the PLCbeta3/Ca(2+)/CAMK signalling pathway. Commun Biol. 2023;6(1):1297.

Pediatric implications of normal insulin-GH-IGF-axis physiology

ABSTRACT

 

Understanding the involvement of the insulin-GH-IGF-axis in the different phases of human growth, development, and metabolism is the key to understanding human pathophysiology. The normal physiological actions of the axis optimize human growth and metabolism to impact adult height by approximately one third. IGF binding proteins modulate access of circulating IGF-I to the tissues and modulate IGF-I and -II access to the type 1 IGF receptor (IGF1R) at the cellular level. Complete lack of IGF1R signaling is most likely not compatible with a viable human fetus, while allelic haploinsufficiency impairs brain development and causes severe short stature. Lack of insulin receptor signaling in Leprechaunism may result in the rare event of an alive but severely small for gestational age baby that will only survive if treated with recombinant-IGF-1 to substitute inulin receptor signaling with IGF1R signaling via their common intracellular pathways. IGF-I gene defects result in mental retardation and severe fetal and postnatal growth failure with GH hypersecretion and marked insulin resistance. Likewise, IGF2 gene defects or imprinting defects cause severe fetal growth failure but somewhat less adverse effects on postnatal growth, more variable effects on brain development, and an absence of marked metabolic effects. GH fine-tunes insulin and IGF-I signaling with no impact on IGF-II expression and has a minor impact on fetal development and growth. GH effects on lipolysis are established in the newborn and ensure gluconeogenesis and prevents hypoglycemia after birth. The complete absence of GH expression such as in GHRHR or GH1 gene defects or absence of GH signaling in GHR or STAT5B gene defect leads to an adult height of 120-130cm if untreated, and has severe metabolic consequences. Even excess of insulin, GH, IGF-I and IGF-II signaling are associated with severe metabolic disease and excess growth and/or obesity. Malnutrition or malabsorption causes decreased insulin signaling which reduces GHR expression and blocks the GH signaling pathway leading to IGF-I expression (GHR uncoupling), while GH’s metabolic actions on lipolysis and gluconeogenesis are unaffected. GH signaling attenuate insulin actions on glucose metabolism which causes insulin resistance and hyperinsulinemia or may precipitate diabetes. However, insulin signaling pathways that enhances GHR function or suppress IGFBP-1 or SHBG production are still intact and promote anabolism, optimize growth, enhance androgen actions and play a mechanistic role in premature adrenarche and PCOS. Long-term nutritional deprivation compromises growth, while from a developmental perspective, decreased insulin signaling (leading to GHR uncoupling) prolongs life (at least in some experimental animal models) which ensures that fertile age is reached, and survival of the species is ensured. For the health of the general population, the subtle changes in insulin, GH and IGF-I signaling associated with gene polymorphisms or epigenetic changes programmed during fetal and early postnatal life and affecting gene expression are important. They determine growth and pubertal development in childhood and predispose the individual for developing the metabolic diseases and malignancies in adult life, as predicted by the Barker hypothesis. As the roles of the insulin-GH-IGF-axis in growth and metabolism, often discussed separately, are intimately linked they will be described jointly here.

 

EARLY WORK DEFINING THE INSULIN-GH-IGF-AXIS

 

Daughaday realized that the mitogenic effect of GH in the growth plate was not direct but mediated by Insulin-like Growth Factor-I (IGF-I), at that time named sulphation factor or somatomedin C (1). Another effect of IGF-I was insulin-like and not inhibited by insulin specific antibodies (2,3,4) and therefore it was named non-suppressible Insulin-like activity (NSILA). Hall and Van Wyk purified IGF-I from human muscle extracts (5,6) and realized that these biological activities originated from the same molecule. They also identified significant quantities in blood (7). The primary structure of IGF-I and Insulin-like Growth Factor-II (IGF-II) was discovered by Froesch and coworkers as a result of their persistent work to characterize the metabolic activity of NSILA (8,9). Soon after, the mitogenic activity of the sulphation factor or somatomedin C as well as somatomedin A was shown to be identical to IGF-I (10). Rechler and Nissley demonstrated that IGF-II was identical to multiplication stimulating activity, a factor known to stimulate DNA synthesis in chick embryo fibroblasts (11).

 

The concept that binding proteins existed for peptide hormones like the IGFs, similar to those for steroid and thyroid hormones, were suggested by studies from Zapf and Froesch (12) and by Hintz (13), demonstrating that NSILA was present in high molecular weight complexes in serum. The binding was exclusive to IGFs and did not apply to insulin or proinsulin despite their structural similarities. High molecular weight IGF-I complexes with IGFBPs were GH dependent (14) and formed a ternary complex composed of IGFBP-3 (15), the Acid Labile Subunit (ALS) (16) and IGF-I or IGF-II. Low molecular weight complexes contained IGF-I or IGF-II bound to an insulin regulated liver derived protein IGFBP-1 (17, 18), at first called the 28 kDa binding protein or PP12 (19). The existence of other IGF binding proteins, six in total, became clear when Hossenloop (20) developed Western ligand blotting as a technique to quantify these proteins. The components of the IGF-IGFBP-system are outlined in Figure 1.

 

Figure 1. The primary structures of IGF-I, IGF-II and insulin are similar. IGFs are produced by many differentiated cell types, and their bioactivity in the extracellular fluids or in the circulation are coordinated by six IGF binding proteins (IGFBP-1 through -6). IGFBP-3, the major binding protein in serum is stimulated by GH and it forms a large 150 kDa ternary complex with IGF-I or -II and the GH regulated acid labile subunit (ALS). IGFBP-5, an important supporter of bone tissue formation, also forms ternary complexes with IGF-I or -II and ALS. IGFBP-1, suppressed by insulin, is one of several binding proteins in the smaller 50 kDa binary complexes with IGF-I or –II. IGFBP-2 has inverse association with insulin under many physiological conditions. In contrast, IGFBP-4, -5 and -6 do not appear to be directly regulated by GH or insulin and are important local regulators of IGF activity in bone and the CNS. The type 1 IGF receptor (IGF1R) is the mediator of the mitogenic, anti-apoptotic, differentiating and metabolic effects of both IGF-I and -II. The structural similarity of the IGF1R with the insulin receptor (IR) explains the formation of hybrid receptors in cells that expresses both receptors such as myocytes and pre-adipocytes. Cross reactivity among the ligands and the receptors have been demonstrated, although it has minor importance under physiological conditions but may cause non-islet-cell tumor hypoglycemia due to unprocessed pro-IGFs with markedly decreased binding affinity to IGFBPs. A second receptor, exclusively binding IGF-II, work as a scavenger receptor and is identical to the mannose-6-phosphate receptor, known to direct proteins for degradation in the lysosomes. A second level of control of IGF bioactivity is exerted by IGFBP proteases which release IGF-I activity after fragmentation of IGFBPs. Specific production and regulation of IGFBP proteases at the tissue level controls processes such as ovulation and atherosclerosis. Furthermore, interaction of IGFBPs and IGFBP proteases with the extracellular matrix modify the binding affinity for the IGFs and are involved in prolonging the actions of IGFs at the tissue level. Extracellular matrix also signals though integrin receptors on the cell surface and modifies IGF-1R signaling. This figure also shows the existence of IGFBP-related proteins with markedly lower affinity for the IGFs and with physiological roles not related to their IGF binding.

 

ANIMAL EXPERIMENTS ESSENTIAL FOR THE UNDERSTANDING OF HUMAN INSULIN-GH-IGF AXIS PHYSIOLOGY

 

Insulin, IGF-I, and IGF-II and Their Receptors

 

Efstratiadis’ series of knock-outs of the insulin-GH-IGF-axis in mice in the early 1990s clearly confirmed its importance in fetal and postnatal growth and metabolism (21). It also predicted the phenotype of experiment of nature in humans with gene defects in the axis yet to be discovered. The studies opened new insights, not least the equal importance of IGF-I and IGF-II in fetal growth, reducing birth weight by about 60 % in both Igf1 knock-out (Igf1ko) and Igf2ko animals and demonstrating that the previous perceived concept that there was a fetal (IGF-II) and a postnatal (IGF-I) form of IGF was incorrect. IGF-I and -II had actions through the type 1 IGF receptor (IGF1R) demonstrated by Igf1rko animals with 45% of wild type birth weight and no further effect when crossed with Igf1ko animals. While Igf1ko animals were viable depending on genetic background and were non-fertile, the Igf1rko animals died from respiratory failure but with an absence of apparent malformations. Interestingly, crossing Igf2ko with Igf1rko resulted in further growth retardation indicating that IGF-II had actions through an additional receptor. Another new insight came from knock-out of the ‘mysterious type 2 IGF receptor’, identical to the mannose-6 phosphate receptor (M6P-R), specifically binding IGF-II and involved in internalization of proteins for lysosomal degradation. Knock-out of the Igf2r/M6pr resulted in increased serum and tissue levels of IGF-II and fetal overgrowth (140% of wild-type birth weight) (22). This receptor works to clear IGF-II and its presence in endothelial cells may, at least partly, explain the lack of endocrine actions of IGF-II due to its proteolytic lysosomal degradation (23). Thus, IGF-II effects on fetal growth are paracrine/autocrine actions mediated by the IGF1R. Knock-out of the Igf2r/M6pr gene combined with Igf2ko/Igf1rko could partly rescue growth retardation, a finding that was explained by IGF-II actions via the insulin receptor (INSR). The formation of heterodimers, more commonly named hybrid receptors, between type A or B isoforms of the insulin receptors (INSRA or INSRB) and the IGF1R of which IRA-IGF1R are highly expressed in the fetus (and in malignant cells) and activated by IGF-II, may further point to the importance of IGF-II during the fetal period. INSRB-IGF1R hybrids comprises up to 30 % of INSR and IGF-I receptors in muscle due to high expression of both and this hybrid predominantly responds to IGF-I (less to insulin) and explains the important role of IGF-I in growth and metabolism in skeletal muscle.

 

Postnatally, the Igf1ko mice continued to grow poorly, resulting in an adult weight 30% of wild-type and with poor gonadal function and delayed bone development. Knock-out of GH or its receptor (GHR), both expressed in the mouse fetus, did not affect birth size, indicating that the Igf1 gene is not under GH control during the fetal period. The actions of GH and its receptor on growth in mice were obvious from postnatal day 15 and largely slowed growth resulting in a 50 % reduction of wild-type adult weight. On the other hand, double Ghrko/Igf1ko resulted in further postnatal growth retardation relative to Igf1ko mice completely obstructing further weight gain after postnatal day 15 and supporting previous studies suggesting that progenitor cells in the growth plate require direct GH actions (24).

 

IGFBPs and IGFBP Proteases  

 

Like the above attempts to pinpoint the role of important ligands and receptors in the axis, steps to assess the role of IGF binding proteins involved in modulating IGF-I and IGF-II bioactivity were taken (reviewed by Pintar (25)). In contrast to the pronounced phenotypes caused by mutations in receptors and their ligands, the growth phenotypes of the various IGFBP knock-out animals were far less pronounced as were the metabolic changes observed (26,27,28). It was argued that there is a large degree of redundancy among the functions of the IGFBPs which to some extend contradicts their specialized functions in various tissues (29). However, this idea was to some extent supported by the finding of somewhat more pronounced phenotypes in double and triple knock-out animals (30). This is largely in accordance with the absence of reports of IGFBP gene defects causing growth retardation in humans. The most affected phenotype identified was that of Igfbp4ko mice who were growth retarded at birth and displayed poor postnatal growth (30). No such mutation has been identified in humans. IGFBP-4 is specifically degraded by the metalloproteinase PAPP-A (Pregnancy Associated Plasma Protease -A) produced by the placenta as well as bone and ovary. In Pappa knock-out animals a 20-30% reduction in body weight was reported (31). Interestingly, the growth restriction phenotype of mice null for Pappacould be rescued by disruption of IGF-II imprinting during embryonic development (32).

 

Endocrine Versus Paracrine Autocrine IGF-I

 

One of the controversies of the area has been the relative contribution to linear growth of circulating endocrine IGF-I largely produced by the liver versus peripherally produced IGF-I with major paracrine/autocrine actions on local tissues. The major importance of paracrine/autocrine IGF-I was demonstrated by liver specific Igf1ko mice (Ligf1ko) with largely unaffected longitudinal growth (33). Circulating levels were 20% of wild-type with compensatory elevation of GH, insulin resistance and hyperinsulinemia. With age the animals developed type 2 diabetes, underlining the metabolic consequences of largely elevated GH combined with circulating IGF-I deficiency (34). Somewhat unexpectedly, this animal model closely resembles children and adolescents with type 1 diabetes, as further elaborated on below.

 

Another model to assess the relative importance of endocrine versus paracrine/autocrine IGF-I is the liver-specific Ghrko mouse. It produces a similar phenotype but with more specific hepatic consequences of absent GH signaling (35).

 

INSULIN-GH-IGF AXIS PHYSIOLOGY, A PEDIATRIC PERSPECTIVE

 

Insulin-GH-IGF-Axis and Human Fetal Growth

 

IGF-I controls the pace of the cell cycle from early on in human embryogenesis. INSR and IGF1R is expressed in human pre-implantation blastocysts already from the 8-cell stage, while IGF-II is expressed already in the oocyte (36). After implantation, IGF-I is produced in the human embryo (37), but until then the source of IGF-I is thought to be the female reproductive tract, and it is known that the availability of the IGF-I ligand is important for blastocyst growth in human in vitro fertilization - IVF (38). IGF-I production is controlled by nutritional factors in the early embryo and even later during human fetal development (39). Circulating endocrine IGF-I increases with gestational age (40) and is strongly correlated with fetal growth in the second part of gestation (41,42). However, little has been reported concerning the regulatory control of the IGF1 gene in the human fetus. IGFBPs can be identified in the human fetal circulation (40), and recently the role of IGFBP-5 in regulating fetal growth was suggested by fetal growth retardation in the absence of a specific IGFBP-5 protease, PPAP-A2 (43). Insulin continues to be permissive for IGF-I production even after GH is established as the major pituitary stimuli controlling endocrine as well as paracrine/autocrine IGF-I, as described below.

 

Fetal Growth Restriction and Programming of the Insulin-GH-IGF-Axis Setpoint

 

Insulin resistance has been developmentally advantageous for mankind until very recent decades of excess food and sedentary life style. It was proposed by Barker et al (44) in his ‘fetal and infant origin of adult disease’ hypothesis that intrauterine restriction of growth compensated by excessive postnatal catch-up growth results in an increased risk of developing disease entities of the metabolic syndrome later in life. In his early epidemiological studies, he demonstrated that there is a U-shaped relationship between birth weight and risk of obesity, insulin resistance, type 1 diabetes, hypertension, dyslipidemia and ischemic heart disease, with lower birth weight (within the normal range) imposing a risk. Notably, at higher birth weights this risk rises again which may represent genetic risks of obesity and type 2 diabetes. The concept was that poor fetal nutrition would lower fetal IGF-I and program the child to a low IGF-I setpoint and slower postnatal growth, an epigenetic phenomenon that could be preserved over a few generations (45). At the same time, small for gestational age (SGA) babies becomes insulin resistant (46) and this trait is enforced by a low endocrine IGF-I setpoint (47), creating the best physiological circumstances for the storage of fat during short times of food availability in a world with limited access to food. However, in a world of plenty, this advantage would turn into a disadvantage and give fast increases in body weight, hyperinsulinemia, and the development of metabolic syndrome problems early on (reviewed by Dunger et al (48). New information even suggests that the parental nutritional state can impose epigenetic metabolic changes in the fetus (49).

 

Figure 2. In the fetus (insert) IGF-I increases with gestational age toward birth. Endocrine circulating IGF-I is strongly nutritionally dependent and correlated with birth size. Pituitary GH control of IGF-I production is not fully established during the first year of human life. The ability of serum IGF-I levels to increase during childhood is dependent on the shift from binary complexes of IGF-I with short half-life to a complete dominance of IGF-I bound in a stable ternary complex with the GH dependent proteins IGFBP-3 and ALS. Both these proteins increase, when pituitary GH control of the axis is established. During pubertal development, sex steroids change the set-point of negative IGF-I feedback and allow a peak of IGF-I in mid-puberty. Total IGF-I levels decline to low levels in senescence. Serum IGF-I reference values based on Juul (50).

 

Postnatal Establishment of Pituitary GH Control of IGF-I Production

 

In humans, full GH control of the IGFI gene, as well as the IGFBP3 and Acid Labile Subunit (ALS) genes, is developmentally regulated and established not until the first year of life. IGF-I and IGFBP-3 levels increase slowly from birth until a more rapid increase and peak during puberty, which is followed by a decline toward low levels in senescence (50, 51) (Figure 2). The late establishment of pituitary GH control of the axis is strongly supported by animal data from GHR KO mice reviewed above, and by a new model of Laron syndrome (GHR defect) in pigs (52). In accordance, newborns with mutations in the GH Releasing Hormone Receptor (GHRHR) gene resulting in an isolated GH deficiency (GHD) phenotype was associated with normal birth weight in one cohort (53) and slightly subnormal birth weight in another (54). In a subgroup of 12 children with congenital isolated GHD, birth weight (1·1 ± 0·8 SD) and length (0·5 ± 1·3 SD) was not affected (Mehta A). GH1 mutations appear to be slightly more affected with mean birth weights of −1.0 ± 0.9 (54). Studies of common polymorphisms in GH1 demonstrate dose effects of 150 and 100 grams in term newborns of normal and low birth weight, respectively (55).  Somewhat contradictory to the observations in animals, Savage et al (56) reported 27 prepubertal children with severe GH insensitivity syndrome (GHIS or Severe Primary IGF Deficiency (SPIGFD)) to have a median (range) birth weight SDS of -0.72 (1.75 - (-3.29)) and birth length SDS of -1.59 (0.63 - (-3.63)). SPIGFD in these patients were defined by phenotypic and biochemical characteristics and they were treated with recombinant human (rh)IGF-1 in one of the clinical trials leading to approval of this therapy, as described later. There was no complete genetic characterization of these patients and 7 patients had a normal serum GH Binding Protein (GHBP) suggesting that the extracellular part of GHR was not affected. In a monograph, Professor Zvi Laron (57), who gave his name to this syndrome, reported that birth weight is unaffected while birth length is slightly on the shorter side. In summary, human fetal growth is only marginally affected by GH. GH is detectable and the GHR is expressed in the human fetus and the metabolic effects of GH on lipolysis are essential to maintain normal levels of glucose in the newborn.

 

Given this critical role of GH in adjusting metabolism to the fasting condition, it is plausible that the metabolic effects of GH are required for optimal linear growth of the human fetus and that this explains marginal effects on linear growth of the human fetus. However, strict GH control of IGF-I in the human fetus would predict severe growth retardation of the above-mentioned genetic defects comparable with the birth size observed in defects in the IGF-I gene. And that is not observed: Children with IGF1 defects suffer from far more severe fetal growth retardation with birth weight SDS around -4 and birth length SDS of -5 to -6 (reviewed in (58). In the study by Mehta et al (Mehta A), children with congenital isolated GH deficiency demonstrated growth retardation as already at 6 months of age (−2·6 ± 1·0 SD and −2·2 ± 1·3 SD in weight and length, respectively) prior to starting treatment. This suggests that the developmental GH control of IGF-I production is established early after birth. It is in accordance with data from Jensen RB and Juul A et al (Jensen RB) suggesting that low IGF-I is a marker of GH deficiency early in life. Children with combined pituitary hormone deficiencies were even more growth retarded at 6 months (Metha A).

 

GHR Signaling Pathway to IGF1 Gene Transcription

 

The important cell signaling steps associated with GH stimulated IGF1 gene activation, transcription and IGF-I production are detailed in the Endotext chapter ‘Normal Physiology of Growth Hormone in Adults´. Briefly, GH binding to preformed dimeric GHR - JAK2 complexes introduces structural changes in the receptor complex that separates JAK2 inhibitory and kinase active sites and enables trans-phosphorylation of the two JAK2 molecules (reviewed in (59). The GHR belongs to the class 1 cytokine receptors which uses STAT as one of its principal secondary messengers, and the subsequent phosphorylation of two STAT5b molecules results in a phospo-STAT5b homodimer which translocate to the cell nucleus, binds to STAT5b recognition sites on the IGF1 gene promoter, and initiates transcription (Figure 3).

 

Primary and Secondary IGF-I Deficiency

 

IGF-I deficiency may be the result of low or inadequate production of GH. This condition is known as GH deficiency (GHD) but in analogy with other pituitary deficiencies leading to peripheral hormone deficiencies the term secondary IGF-I deficiency was proposed (Rosenfeld et al). GHD is severe in GH1 or GHRH-R gene defects and in some children with congenital GHD as well as after treatment of brain tumors with radiation therapy. However, these conditions are rare and most children treated with rhGH has less severe GHD or an indication not associated with GHD. Disorders of GH in childhood is outlined in the Endotext chapter Disorders of Growth Hormone in Childhood by Murray and Clayton (132)

 

Primary IGF-I deficiency or GH insensitivity is severe in homozygous genetic defects in genes including the GH receptor (Laron syndrome) gene, STAT-5b gene and IGF-I gene. Less severe growth retardation is reported in children with homozygous genetic defects in the ALS gene. Treatment with rhGH do not improve growth in these cases while rhIGF-1 is efficient in SPIGFD and approved by FDA and EMA. In many patients with severe primary IGF-I deficiency (SPIGFD) defined by low IGF-I (less than – 3 SDS or the 2.5th percentile), severe short stature (Height SDS less than – 3), normal GH secretion and absence of acquired insensitivity to GH (discussed below) genetic defects may be absent. Still treatment with rhIGF-1 may be as efficient as in patients with confirmed homozygous GHR defects (131).

 

Insulin Enhancement of GHR Signaling to IGF1 Gene Transcription

 

Insulin signaling enhances the GH signaling pathway to enable IGF-I production in the fed state and promotes linear growth and other anabolic responses (60, 61, 62). Moreover, GH signaling to elicit IGF1 gene transcription is blocked in the absence of appropriate insulin signaling, a phenomenon also known as un-coupling, and resulting in growth arrest (60, 61, 63, 64) (Figure 3). This is partly a result of insulin’s effects on hepatic GHR expression, and partly a post-receptor signaling effect as unraveled by extensive animal studies (reviewed in (60). In obese individuals with hyperinsulinemia, hepatic GHR expression is enhanced as indicated by elevated GHBP levels reflecting proteolytic cleavage of highly expressed surface GHR and release of the extracellular part to the circulation (65). This allows obese individuals to maintain normal serum IGF-I levels despite markedly diminished GH secretion (66, 67). Consequently, obese individuals have attenuated GH responses to GH secretagogues (68).

 

Figure 3. Multiple, partly identical, pathways have been described to be activated by the GHR, the INSR and many other hormone kinase receptors not shown on the slide. Limiting this cartoon to the GHR and INSR, still the complexity is very high and the potential candidate hubs for crosstalk are numerous. The crosstalk that, following activation of the GHR, leads to resistance to specific signaling events from the INSR (related to glucose metabolism) is more well established and describe in detail in the Endotext chapter ‘Normal Physiology of Growth Hormone in Adults´. In the current review the focus is on the crosstalk that is executed by activation of the INSR and results in enhanced signaling from the GHR leading to gene activation of IGF1 and other GH dependent genes such as IGFBP3 and ALS. There are basically no studies addressing this crosstalk on the cellular level despite the strong evidence for INSR signaling being permissive for IGF1 transcription. Given that mTORC1 and mTORC2, downstream the INSR, are essential hubs for substrate and energy sensing and thus controlling the switch between cell anabolism and catabolism, they appear to be strong candidates to determine whether IGF1 should be on or off. A further argument for their candidate role is the so far limited evidence of mTORC1 and mTORC2 involvement in branched chain amino acid sensing directly enhancing IGF1 transcription. The unique role of the Jak2, STAT5b pathway in connecting the GHR with IGF1 gene activation has not been challenged and is thus the major candidate pathway to be affected by INSR signaling crosstalk. It is less clear which of the signaling pathways from the GHR that results in enhanced lipolysis although the STAT5b pathways has been implicated. This is particularly interesting given that GHR induced lipolysis does not require INSR signaling crosstalk. The reader is encouraged to seek specific information regarding other GHR and INSR pathways depicted in the cartoon but not further discussed in this review.

 

While short-term fasting decreases serum IGF-I but does not affect GHBP (69), the triad of IGF-I deficiency, poor growth and pubertal delay/arrest in long term nutritional deficiency such as in anorexia nervosa is associated with low GHBP that is partly restored with weight gain (63). Also, circulating IGF-I deficiency due to hepatic under-insulinization in type 1 diabetes is associated with low GHBP levels. Normal circulating IGF-I and GHBP are fully restored only after experimental intra-peritoneal (70) or intraportal (71) insulin delivery.

 

Functional/Acquired IGF-I Deficiency - Uncoupling of GHR Signaling to IGF1 Gene Transcription and Maintained GHR Metabolic Signaling Due to Insulin Deficiency

 

Increased GHR signaling in obese children does not generally result in elevated IGF-I, due to negative feedback inhibition of GH. In contrast, impairment of GH signaling due to insulin deficiency cannot generally be compensated by GH hypersecretion. This is true in fasting children (63) and adults (62). The exact mechanisms by which insulin and GH signaling crosstalk on the post-receptor level is not yet understood (Figure 3). More recent data suggesting that FGF21 plays a role in mediating these events needs further confirmation (72). Interestingly, GH signaling leading to activation of lipolysis in adipose tissue and increased hepatic glucose production via both glycogenolysis and gluconeogenesis in the liver, is not affected by the absence of insulin crosstalk (reviewed in (72)). This has important implications in securing substrate mobilization and gluconeogenesis during fasting and explains the cardinal hypoglycemic symptoms in GHD and GHIS newborns in the absence of intrauterine growth retardation (IUGR). GH signaling leading to lipolysis is thought to involve STAT5b. Most information comes from animal models and involves GH signaling in the liver, but in mouse adipose tissue GHR KO downregulates beta-3 adrenergic receptor expression and inhibits lipolysis (73). GH effects are lost if STAT5b signaling is blocked (74), gene transcript profiles of GHR KO and STAT5b KO animals overlap largely, and STAT5b controls key regulator enzymes involved in lipid metabolism (75). However, if STAT5b mediates both metabolic signaling and IGF-I production it still needs to be understood where the two pathways diverge, and why GH metabolic signaling is not blocked in the absence of insulin crosstalk. In humans, recent studies have identified new GH signaling responses involving GH downregulation of fat-specific protein (FSP27), a negative regulator of lipolysis. MEK/ERK activation and inhibition of peroxisome proliferator-activated receptor-γ (PPARγ) are involved, and this offers an alternative signaling pathway from the GHR (76).

 

Interactions Among Endocrine Axes

 

The activity of the insulin-GH-IGF-axis is dependent on the other endocrine axes which have permissive actions on GH stimulated IGF-I expression and affect IGFBPs and proteases (Figure 4). For example, thyroxine is needed to enhance GH effects on endocrine IGF-I expression and a normal GH-IGF-IGFBP-axis is needed for optimal thyroid hormone production (77). Sex steroids further enhance the function of the GH-IGF-axis, most likely by attenuating pituitary and hypothalamic sensitivity to IGF-I negative feedback (78). The pivotal role of sex steroids on the setpoint of the axis is reflected by the peak circulating levels of IGF-I and IGFBP-3 reached in mid-puberty (50,51). On the other hand, GH via its stimulation of local IGF-I is important for testicular production of testosterone and spermatogenesis (79), and the local IGF-IGFBP-axis is involved in selection and growth of the primary follicle in the ovary, estradiol production and ovulation (80, 81). Finally, cortisol has impact on the actions of the GH-IGF-axis on growth by blocking IGF1R signaling leading to apoptosis (82) despite normal endocrine IGF-I levels (83).

 

Figure 4. Hypothalamic GH releasing hormone and somatostatin establish the pulsatile pituitary GH secretion that is established as the main regulator of endocrine and paracrine/autocrine IGF-I production during the first year of life in humans. Insulin is permissive for this regulation by modulating GHR signaling, and normal beta-cell release of insulin is required for normal liver derived endocrine IGF-I levels measured in serum (blue insert) that in most cases is a good marker of the local production and actions of IGF-I. During fasting the GH regulation of IGF-I is uncoupled, resulting in decreased IGF-I (and catabolism) and elevated GH secretion and maintained lipolytic signals securing gluconeogenesis and preventing hypoglycemia. Apart from insulin, the endocrine thyroid axis is important for normal GH induced IGF-I production and during pubertal development sex steroids from the gonads enhance the performance of the GH-IGF-axis presumable by relaxation of the negative IGF-I feedback on GH secretion allowing a higher set-point of the axis. Whether this is an estradiol effect is not fully elucidated but it is suggested by the fact that non-aromatizable androgens such as oxandrolone do not affect IGF-I levels. The actions of the adrenal axis are most likely local and involve actions on IGF1R signaling leading to apoptosis of stem cells in the growth plate and thus irreversible loss of height. Cortisol excess leaves endocrine IGF-I and GH levels largely unaffected.

 

Discordance Between Endocrine and Paracrine/Autocrine IGF-I

 

An important example of metabolic and mitogenic consequences of an unbalanced endocrine versus autocrine/paracrine insulin-GH-IGF-axis comes from observations in children and adolescents with type 1 diabetes (Figure 5). They suffer specifically from insulin deficiency in the hepatic portal circulation as a result of the subcutaneous delivery of insulin (reviewed by Dunger (64)). This attenuates their endocrine production of circulating IGF-I despite excessive GH secretion (84). Circulating IGF-I deficiency and GH hypersecretion induce insulin resistance which is further augmented by insufficient suppression of hepatic glucose output. To overcome this, higher subcutaneous insulin doses are needed to maintain glycemic control, and this results in aggravated systemic hyperinsulinemia. The importance of local tissue hyperinsulinemia and GH hypersecretion in generating high paracrine/autocrine IGF-I production and promoting mitogenic vascular events leading to diabetic long-term complications should not be underestimated. Based on this insight, a promising new drug targeting the alphaVbeta3 integrin affecting IGF-I signaling in smooth muscle cells has been found to inhibit the development of atherosclerotic lesions in diabetic pigs (85). Another consequence of a compensatory increased in local IGF-I activity is the finding of normal childhood and pubertal linear growth despite endocrine IGF-I deficiency in type 1 diabetes (86). It is interesting that the endocrine and paracrine/autocrine changes in the insulin-GH-IGF-axis observed in children with type 1 diabetes closely resembles those observed in liver IGF-I KO mice which eventually leads to diabetes in the KO mice. Given that portal delivery of insulin, which has the potential to completely restore IGF-I levels in type 1 diabetes (70, 71, 87), remains an experimental treatment, rhIGF-1 treatment to restore circulating IGF-I and suppress GH and decrease insulin needs appears to be the most feasible approach to take (88). In a 6-month clinical trial of a single daily injection of rhIGF-1 improved glycemic control in adolescents with type 1 diabetes were found (89). Long-term studies on diabetic vascular complications have yet to be performed.

 

If paracrine/autocrine IGF-I production is lost in addition to liver-derived IGF-I, the metabolic consequences become obvious. This situation was first reported in a boy with a deletion of exon 4 of the IGF-I gene (90) resulting in severe fetal and post-natal growth arrest, poor brain development and extreme insulin resistance with compensatory hyperinsulinemia and acanthosis nigricans. A short trial of treatment with rhIGF-1 resulted in normalization of circulating IGF-I, suppression of GH hypersecretion and a markedly decreased insulin response to a meal tolerance test (91). In this example and in type 1 diabetes, it has been discussed whether the normalization of glucose metabolism following rhIGF-I therapy is most importantly associated with insulin-like effects of IGF-I on glucose uptake in muscle or suppression of GH hypersecretion? Although most studies support the importance of GH suppression, prolonged actions of IGF-I similar to that of long-acting insulin analogs in type 1 diabetic patients are important. IGF-I is equipotent with insulin in stimulating glucose uptake in human muscle but has less effects in fat and liver (92). Reports that newborns with a complete lack of insulin effects due to inactivating defects in the INSR gene can now survive for extended time into adolescence when treated with recombinant IGF-I, that stimulate glucose uptake via the IGF1R sharing common signaling pathways with the INSR, support an important direct role of IGF-1 signaling on metabolism (93).

 

Figure 5. Changes in liver derived endocrine IGF-I measured in the circulation and paracrine/autocrine IGF-I are in most cases concordant. In the absence of practical and validated methods to measure IGF-I at the tissue site of action, paracrine/autocrine IGF-I activity is assessed by determining known physiological actions of IGF-I such as growth or glucose metabolism. Type 1 diabetes is a condition with discordant changes in endocrine vs. paracrine/autocrine changes in IGF-I that in many ways resembles those reported in a mouse model of conditional knock-out of IGF-1 expression in the liver. In type 1 diabetes, insulin deficiency in the liver, caused by a systemic rather than a portal insulin replacement therapy, results in a functional GHR signaling defect to IGF-I transcription (uncoupling). Low endocrine IGF-I production decreases circulating IGF-I and results in decreased negative pituitary feedback and GH hypersecretion. The lack of direct IGF-I effects on glucose uptake in muscle and the diabetogenic effects of GH (including maintained signaling to lipolysis) decreases insulin actions on glucose metabolism (known as insulin resistance). The portal insulin deficiency also fails to suppress hepatic glucose production. In other to maintain glycemic control, the increased insulin requirement can only be met by more subcutaneous insulin leading to systemic hyperinsulinemia. There is no direct information about local paracrine/autocrine IGF-I activity, but there are several indications that tissue hyperinsulinemia and GH hypersecretion results in a compensatory increase of tissue IGF-I activity. Firstly, linear growth is not impaired in children and adolescents with Type 1 Diabetes despite of their low endocrine IGF-I (comparable to levels in short stature children), indicating a compensatory upregulation of local IGF-I activity (IGF-I being the most important stimulator of longitudinal growth). Secondly, it is plausible that increased local IGF-I activity contributes to diabetes complications known to be tightly associated with increased rather than decreased IGF-I activity. While type 1 diabetes is not generally associated with increased risk of cancer, the increase in local production of IGF-I in obesity and type 2 diabetes may contribute.

 

Liver Disease and Endocrine Versus Paracrine/Autocrine IGF-1 Production

 

In children with severe liver disease, there may be similar discrepancies between circulating endocrine levels of IGF-I and IGF-I activity in peripheral levels contributed by paracrine/autocrine secretion of IGF-I (94). However, less is known about peripheral IGF-I activity and it is possible that there are more secondary metabolic and nutritional issues that could lower local IGF-I production and impact on linear growth. Particularly in liver cirrhosis associated with thalassemia major, which concomitantly can impair pituitary GH secretion, there is no secondary upregulation of local IGF-I and linear growth failure is common (95). Recently, increased IGF-I expression was reported in obese children with non-alcoholic fatty liver disease (NAFLD) and it was combined with upregulation of IGF1R (96), not expressed in the normal liver but involved in liver repair, such as after liver resection in a mouse model (97).

 

GH and Cytokine Crosstalk

 

STAT5b phosphorylation is also mediated by activation of other members of the cytokine receptor family and has an impact on immunological function: this is evident from the finding of immune deficient symptoms in children with STAT5b genetic defects (98) but these are not found in GHD and GHIS children.

 

Negative Control of GHR Signaling

 

Control of the GH signaling cascade is also under inhibitory control, principally by two mechanisms. Firstly, tyrosine phosphatases including PTP dephosphorylate GHR associated molecules. In Noonan’s syndrome genetic defects in the PTPN11 gene may affect this pathway and has been implicated in poor growth and poor response to GH therapy, although reports are conflicting (99, 100). In addition, the SOCS gene is activated by GH signaling and works as a short intracellular negative feedback loop which rapidly down-regulate GHR activity by internalization and receptor ubiquitinoylation resulting in lysosomal and proteasomal degradation (101).

 

Other Nutritional Signals to the GH-IGF-Axis

 

Nutritional supplementation increasing dietary protein intake from cow’s milk increases endocrine IGF-I, while an equal intake of animal protein from meat does not (102). It is possible that the amino-acid composition that differs depending on the dietary source may contribute, and there are other constituents in skimmed milk not present in meat. More likely, however, it is explained by a higher carbohydrate intake in the milk group (while fat content was higher with meat supplementation) and the finding that fasting insulin levels doubled (103), in accordance with the essential regulatory role of insulin on GHR signaling discussed above. There is, however, direct evidence for a regulatory role of amino-acids on IGF-I production that is independent of insulin. Branched chained amino-acids (BCAA) are known to stimulate cell growth by the activation of mTORC2, a protein complex that controls protein synthesis in cells by sensing nutrient and energy availability and is also one of the main signaling pathways of IGF-I and insulin (Figure 3). The role of BCAA has been studied in rats given a restricted diet containing high levels of BCAA, compared to a group given low levels of BCAA (104).

 

The availability of nutrients in the circulation might also have a direct effect on the production of IGF-I. Human breast adipocytes cultured in high glucose levels have been found to produce more IGF-I compared to adipocytes cultured in low glucose levels (105).

 

IGF-II

 

IGF-II is a paracrine/autocrine hormone which is as essential for fetal growth as IGF-I (21). IGF-II may also contribute considerable to postnatal growth although the absence of endocrine effects makes it difficult to study in humans. Less is known about the metabolic actions of IGF-II. Growth promoting actions of IGF-II is via the IGF1R and IGF1R-INSR hybrid receptors which has preference for IGF-II actions if the A isoform of the INSR receptor – expressed in the fetus and in malignantly transformed cells - pairs with the IGF1R in the hybrid (106). The human IGF2 gene is an imprinted gene (107) exclusively expressed from the paternal allele in certain tissues (reviewed by Rossignol (108). The imprinted promoter region is found in a complex configuration with the H19 gene on chromosome 11 and shares two important imprinting regions with this gene. Methylation of the imprinting region ICR1 results in expression of the paternal IGF2allele, while H19 gene expression is suppressed. Correct imprinting should lead to expression of the paternal allele only and sufficient expression of IGF-II for normal growth. Loss of methylation of the ICR1 on the paternal allele results in the phenotype of Silver Russell syndrome (SRS) which may also arise from other genetic aberrations that have not yet been linked to the IGF-II production or signaling cascade including maternal uniparental dyssomnia of chromosome 7.

 

SRS is characterized by proportional IUGR with severe SGA at birth, relative sparing of the brain with close to normal head circumference at birth, severe feeding difficulties in infancy and childhood (which in contrast to Prader-Willi syndrome does not rebound into feeding obsession later in life), postnatal growth retardation and body asymmetry. As indicated by the SRS phenotype, IGF2 gene transcription of some organs are not controlled by imprinting. The relative normal development of the brain and the relative macrocephaly of SRS is explained by the lack of imprinting control of IGF-II expression in the brain. Children with genetic mutations in the expressed paternal allele of the IGF2 gene, were reported to have an SRS phenotype. They had somewhat more pronounced psychomotor developmental problems compared with the SRS phenotype, which has increased risk of autism spectrum defects including attention deficit hyperactivity disorder. In SRS, there is a normal postnatal expression of the IGF2 gene in the liver leading to normal levels of circulating IGF-II. Interestingly, the normal endocrine levels of IGF-II do not overcome the postnatal growth restriction. A similar lack of endocrine IGF-II effects on growth and metabolism was reported in the IGF-I deficient child mentioned previously with a loss of exon 4 of the IGF1 gene. He had compensatory increased GH, IGFBP-3, ALS as a result of lack of negative IGF-I feedback and secondary to the increased IGF binding capacity, increased IGF-II (see also the chapter on IGF-binding proteins below). Another observation in favor of this view is the lack of a correlation between newborn cord levels of IGF-II and birth size (41). This contrasts with a strong positive correlation between cord blood IGF-I concentrations and birth size. The role of IGF-II should be viewed in the light of Efstradiadis series of knock-out experiments in mice (21) where the Igf2ko mice had the same degree of fetal growth retardation as the igf1ko which demonstrates that IGF-II is a paracrine/autocrine and not an endocrine hormone.

 

It is possible that circulating IGF-II after release from the ternary complex is cleared from the circulation by binding to the IGF2R – identical to the mannose-6-phosphate receptor – which is associated with lysosomes and results in degradation of IGF-II in endothelial cells (23). In the elegant mouse KO experiments by Efstradiadis et al (21), KO of the IGF2R resulted in fetal overgrowth. However, the largely elevated IGF-II serum levels in that model are more likely a secondary finding, while the lack of clearance of paracrine/autocrine IGF-II is the explanation for the excessive growth.

 

IGF Binding Proteins

 

Six IGFBPs bind IGF-I and IGF-II inside and outside the circulation and has impact on IGF bioactivity (reviewed by Clemmons (109). The IGFBP-related proteins share some structural similarities with the six IGFBPs but have no relevant impact on IGF bioactivity. GF-I passes the endothelium intact primarily via IGF1R mediated transcytosis and this process is essential for endocrine actions of liver derived IGF-I (110). Limited experimental evidence from animal and tissue cultures suggest that IGF-I complexed with IGFBP-1 and -2 may leave the circulation, although the extent and importance is unclear (Bar 1990).  After endothelial passage IGF-I redistributes to soluble IGFBPs in the extravascular fluids or IGFBPs bound on extracellular matrix or cell surfaces (111). The concentration of unbound IGF-I in the circulation is likely to be proportional to unbound IGF-I concentrations in the tissues, but they are not equal and may have different relationship in different target tissues with differentially-expressed IGFBPs.

 

As pointed out earlier in this review, transgenic animal studies disrupting one or more IGFBPs have not suggested that a marked growth phenotype should be expected in children and no IGFBP mutations causing growth retardation in children had been reported to the best of my knowledge. Interestingly, as predicted by the Igfbp4-ko and the pappa-ko animal models described previously, a human PAPP-A2 gene defect with growth phenotype was recently reported as detailed below (43).

 

Free IGF-I Assays

 

Assays claiming to measure free circulating IGF-I have been developed, but it is unclear to what extent different techniques are influenced by redistribution of IGF-I among IGFBPs associated with the assay procedure (112). Anyway, the fact that IGF-I redistribute among extravascular IGFBPs after passing the endothelium is likely to affect the local tissue bioavailability even more. Moreover, the fact that most data in the literature originate from one assay technique established in one single laboratory has resulted in a lack of confirmatory reports. In a few cases, measurements with different free IGF-I assays have been reported from the same study/experiment with large differences in results (113). The bottom line is that measurements of free IGF-I have not been demonstrated to better predict different physiological or pathophysiological conditions in humans and do therefore not provide any clinically important contributions (114, 112). Techniques to assess IGF-I at the tissue site of action pose practical and methodological challenges. Attempts to establish and validate a method to determine local tissue levels by microdialysis have been reported in adolescents with type 1 diabetes, where endocrine levels are a poor marker of local IGF-I activity (115).

 

Ternary Complex Formation

 

The developmental establishment of GH control over the IGF1, IGFBP3 and IGFALS genes in early childhood initiates the dominance of the ternary complex formed by IGF-I or IGF-II and IGFBP-3 (or IGFBP-5) and ALS as the quantitatively most important circulating form of IGF-I and IGF-II (reviewed by Baxter (116). In the human fetus and newborn, serum IGFBP-3 and ALS concentrations are low and ternary complex formation is absent (117). Although IGF2 gene expression is not under GH control, the circulating levels of IGF-II are largely influenced by GH status since IGF-II (as well as IGF-I and IGFBP-3) is rapidly cleared from the circulation if not bound in the ternary complex. This can be observed in children with SPIGFD, who are deficient in IGF-I as well as IGFBP-3 and ALS, and in whom sc injected rhIGF-I displays a very fast serum clearance rate (118). As mentioned, formation of the ternary complex also governs the circulating levels of IGFBP-3 which under physiological conditions is present in a 1:1 molar relationship with IGF-I plus IGF-II. ALS is a large glucoprotein that under physiological conditions are present in a two-fold molar excess (16).

 

Immunometric IGFBP-3 assays have been claimed to be more predictive of GH status in very young children; however, the support for that is weak. It is rather a misconception related to problems of commercial IGF-I assays at the lower end of IGF-I detection. Moreover, IGFBP-3 has been claimed to provide information about IGF-I bioavailability from calculating the molar ratio of total IGF-I to IGFBP-3. Given that both IGF-I and IGFBP-3 are rapidly cleared from the circulation if unbound, using the IGF-I/IGFBP-3 ratio and disregarding IGF-II concentrations (that are 2-3-fold those of IGF-I on a molar basis) does not make any sense. During puberty, for example, IGF-I bioactivity is increased (114). This is dependent on the 3-4-fold increase in total IGF-I (50), which consequently results in an increase in unbound IGF-I, even if the increase is matched with the same absolute molar increase in IGFBP-3 (and complexed with ALS in a ternary complex). A common view is that increased IGF-I bioactivity depends on a higher IGF-I/IGFBP-3 molar ratio during puberty. However, the increase in molar ratio is entirely explained by the fact that IGF-I and IGFBP-3 increase with the same number of moles per liter, but with a larger relative increase in IGF-I than IGFBP-3 and with IGF-II molar concentrations being unchanged (112).

 

IGFBP Proteolysis and Physiological Consequences

 

The fact that proteolytic cleavage of IGFBP-3 is common, and may result in falsely elevated IGFBP-3 immunoactivity, is the most likely reason for observing a low IGF-I/ IGFBP-3 ratio. Under certain physiological conditions first described in pregnancy (119, 120), specific proteases cleave IGFBP-3 into several proteolytic fragments of which each may retain immunoactivity and thus give rise to signals in an immunometric assay (121). This will lead to overestimations of the IGFBP-3 immunoreactivity in pregnancy, which is already truly increased due to increased placental GH tonus. It may also lead to the erroneous conclusion that IGF-I bioactivity is decreased. On the contrary, IGF-I bioactivity is increased in the maternal circulation resulting from increased total serum IGF-I and decreased binding affinity of fragmented IGFBP-3 (122). There is strong experimental evidence that IGFBP proteolysis results in lower IGF binding affinity. The finding that partial IGFBP-3 proteolysis, such as in pregnancy, does not disrupt the ternary complex, has questioned its significance. However, evidence for increased IGF-I bioactivity in a ternary complex with fragmented IGFBP-3 exists (123). IGFBP-3 proteolysis has also been described in insulin resistant states such as fasting, obesity and type 1 and 2 diabetes (62, 124, 125). While several known proteolytic enzymes such as those involved in blood clotting (126) and cancer metastasis (127, 128) have been identified as IGFBP-3 proteases, the identity of the pregnancy protease is still not resolved.

 

Recently, a human gene mutation of PAPP-A2, a circulating and tissue protease with IGFBP-5 (and to some extent IGFBP-3) as its primary substrate (Gaudamauskes et al), was demonstrated to have a marked growth phenotype involving fetal and post-natal growth retardation in children in a consanguineous family (43). Largely elevated levels of circulating IGF-I but as a result of absence of proteolysis of IGFBP-5 and IGFBP-3, necessary for disruption of ternary complex formation, IGF-I bioactivity in serum is low and presumably tissue bioactivity of IGF-I (and IGF-II) is low. Functionally, this is a state of severe primary IGF-I deficiency (despite of elevated total serum IGF-I) and pharmacokinetic studies suggested that sc. Injections of rhIGF-1 resulted in a fraction of unbound IGF-I in serum despite the impaired proteolysis of IGFBP-5 and IGFBP-3 (133). Attempts to improve linear growth by rhIGF-I treatment has been reported to result in some improvements in a few affected children but not all (134, 135)

 

It is beyond the scope of this chapter to review the overwhelming evidence from cell biology experiments demonstrating the important role of IGFBPs in modulating IGF bioactivity and the role of IGFBP proteases and their actions at the cellular level. Furthermore, IGFBPs other than IGFBP-3 may play a role in the access of IGF-I to various tissues (129).

 

SUMMARY

 

In the present review the pivotal role of nutrition and insulin in determining the regulation and actions of the GH-IGF-axis is reviewed. For the pediatrician, caring for patients in a phase of rapid growth and development, it is important to refer to normality and understand the requirements for a normal insulin-GH-IGF-axis in order to succeed in this task. In the complex work-up, treatment and management of growth disorders a thorough understanding of the normal physiology of the axis is essential in taking the right actions (130, 131). From the normal physiology of this axis, it is possible to understand the consequences of various genetic defects and disorders that affect its regulation and function. The most severe conditions associated with defects in the axis may cause a loss of adult height of approximately 1/3 and may cause severe developmental and neurological deficits and compromise pubertal maturation and fertility. Minor changes in the setpoint of the axis caused by programming of the fetus exposed to intra-uterine growth retardation may predispose the individual for poor linear growth and later metabolic disease, insights that the pediatrician should be aware of and consider in order to improve health and prevent later disease.

 

REFERENCES

 

  1. Salmon WD, Daughaday WH. A hormonally controlled serum factor which stimulates sulfate incorporation by cartilage in vitro. J Lab Clin Med. 1957;49:825–826.
  2. Leonards JR. Insulin-like activity of blood, what is it? Federation Proc. 1959;18:272.
  3. Froesch ER, Bürgi WA, Ramseier EB, Bally P, Labhart A. Antibody-suppressible and nonsuppressible insulin-like activities in human serum and their physiological significance. An insulin assay with adipose tissue of increased precision and specificity. J Clin Invest 1963;42:1816– 1834.
  4. Froesch ER, Muller WA, Bürgi H, Waldvogel M, Labhart A. Non-suppressible insulin-like activity of human serum. II. Biological properties of plasma extracts with non-suppressible insulin-like activity. Biochim Biophys Acta. 1966;121:360–374.
  5. Van Wyk JJ, Hall K, Van den Brande JL, Weaver RP. Further purification and characterization of sulfation factor and thymidine factor from acromegalic plasma. J Clin Endocrinol Metab. 1971;32:389–403.
  6. Van Wyk JJ, Hall K, Weaver RP. Partial purification of sulphation factor and thymidine factor from plasma. Biochim Biophys Acta 1969;192:560–562.
  7. Uthne K. Human Somatomedin. Purification and some studies on their biological actions. Thesis, Stockholm, 1973.
  8. Rinderknecht E, Humbel RE. The amino acid sequence of human insulin-like growth factor I and its structural homology with proinsulin. J Biol Chem. 1978;253:2769–2776.
  9. Rinderknecht E, Humbel RE. Primary structure of human insulin-like growth factor II. Febs Letters. 1978;89:283–286.
  10. Klapper DG, Svoboda ME, Van Wyk JJ. Sequence analysis of somatomedin-C: confirmation of identity with insulin-like growth factor I. Endocrinology. 1983;112:2215–2217.
  11. Moses AC, Nissley SP, Short PA, Rechler MM, Podskalny JM. Purification and characterization of multiplication stimulating activity. Insulin- like growth factors purified from rat-liver- cell-conditioned medium. Eur J Biochem. 1980;103:387–400.
  12. Zapf J, Waldvogel M, Froesch ER. Binding of nonsuppressible insulinlike activity to human serum: evidence for a carrier protein. Arch Biochem Biophys. 1975;169:638–645.
  13. Hintz RL, Liu F. Demonstration of specific plasma protein binding sites for somatomedin. J Clin Endocrinol Metab. 1977;45:988– 495.
  14. Hintz RL, Liu F. Demonstration of specific plasma protein binding sites for somatomedin. J Clin Endocrinol Metab. 1977;45:988– 495.
  15. Martin JL, Baxter RC. Insulin-like growth factor-binding protein from human plasma. Purification and characterization. J Biol Chem. 1986;261:8754–8760.
  16. Baxter RC. Characterization of the acid-labile subunit of the growth hormone-dependent insulin-like growth factor binding protein complex. J Clin Endocrinol Metab. 1988;67:265–272.
  17. Póvoa G, Enberg G, Jörnvall H, Hall K. Isolation and characterization of a somatomedin-binding protein from mid-term human amniotic fluid. Eur J Biochem. 1984;144:199-204.
  18. Póvoa G, Roovete A, Hall K. Cross-reaction of serum somatomedin-binding protein in a radioimmunoassay developed for somatomedin-binding protein isolated from human amniotic fluid. Acta Endocrinol (Copenh). 1984;107:563-70.
  19. Suikkari AM, Koivisto VA, Rutanen EM, Yki Jarvinen H, Karonen SL, Seppala M. Insulin regulates the serum levels of low molecular weight insulin-like growth factor-binding protein. J Clin Endocrinol Metab. 1988;66:266–272.
  20. Hossenlopp P, Seurin D, Segovia B, Quinson B, Hardouin S, Binoux M. Analysis of serum insulin-like growth factor binding proteins using Western competitive binding studies. Anal Biochem 1986;154:138–143.
  21. Efstratiadis A. Genetics of mouse growth. Int J Dev Biol. 1998;42:955-976.
  22. Ludwig T, Eggenschwiler J, Fisher P, D'Ercole AJ, Davenport ML, Efstratiadis A. Mouse mutants lacking the type 2 IGF receptor (IGF2R) are rescued from perinatal lethality in Igf2 and Igf1r null backgrounds. Dev Biol. 1996;177:517-535.
  23. Hachiya HL, Carpentier JL, King GL. Comparative studies on insulin-like growth factor II and insulin processing by vascular endothelial cells. Diabetes. 1986;35:1065-1072.
  24. Lindahl A, Isgaard J, Carlsson L, Isaksson OG. Differential effects of growth hormone and insulin-like growth factor I on colony formation of epiphyseal chondrocytes in suspension culture in rats of different ages. Endocrinology. 1987;121:1061-1069.
  25. Pintar JE, Schuller A, Cerro JA, Czick M, Grewal A, Green B. Genetic ablation of IGFBP-2 suggests functional redundancy in the IGFBP family. Prog Growth Factor Res. 1995;6:437-45.
  26. Wood TL, Rogler LE, Czick ME, Schuller AG, Pintar JE. Selective alterations in organ sizes in mice with a targeted disruption of the insulin-like growth factor binding protein-2 gene. Mol Endocrinol. 2000;14:1472-82.
  27. Murali SG, Liu X, Nelson DW, Hull AK, Grahn M, Clayton MK, Pintar JE, Ney DM. Intestinotrophic effects of exogenous IGF-I are not diminished in IGF binding protein-5 knockout mice. Am J Physiol Regul Integr Comp Physiol. 2007;292:R2144-2150.
  28. Ning Y, Hoang B, Schuller AG, Cominski TP, Hsu MS, Wood TL, Pintar JE. Delayed mammary gland involution in mice with mutation of the insulin-like growth factor binding protein 5 gene. Endocrinology. 2007;148:2138-2147.
  29. Pintar JE, Cerro JA, Wood TL. Genetic approaches to the function of insulin-like growth factor-binding proteins during rodent development. Horm Res. 1996;45:172-177.
  30. Ning Y, Schuller AG, Bradshaw S, Rotwein P, Ludwig T, Frystyk J, Pintar JE. Diminished growth and enhanced glucose metabolism in triple knockout mice containing mutations of insulin-like growth factor binding protein-3, -4, and -5. Mol Endocrinol. 2006;20:2173-2186.
  31. Conover CA, Bale LK, Overgaard MT, Johnstone EW, Laursen UH, Füchtbauer EM, Oxvig C, van Deursen J. Metalloproteinase pregnancy-associated plasma protein A is a critical growth regulatory factor during fetal development. Development. 2004;131:1187-1194.
  32. Bale LK, Conover CA. Disruption of insulin-like growth factor-II imprinting during embryonic development rescues the dwarf phenotype of mice null for pregnancy-associated plasma protein-A. J Endocrinol. 2005;186:325-331.
  33. Yakar S, Liu JL, Stannard B, Butler A, Accili D, Sauer B, LeRoith D. Normal growth and development in the absence of hepatic insulin-like growth factor I. Proc Natl Acad Sci U S A. 1999;96:7324-7329.
  34. Yu R, Yakar S, Liu YL, Lu Y, LeRoith D, Miao D, Liu JL. Liver-specific IGF-I gene deficient mice exhibit accelerated diabetes in response to streptozotocin, associated with early onset of insulin resistance. Mol Cell Endocrinol. 2003;204:31-42.
  35. Fan Y, Menon RK, Cohen P, Hwang D, Clemens T, DiGirolamo DJ, Kopchick JJ, Le Roith D, Trucco M, Sperling MA. Liver-specific deletion of the growth hormone receptor reveals essential role of growth hormone signaling in hepatic lipid metabolism. J Biol Chem. 2009;284:19937-19944.
  36. Lighten AD, Hardy K, Winston RM, Moore GE. Expression of mRNA for the insulin-like growth factors and their receptors in human preimplantation embryos. Mol Reprod Dev. 1997;47:134-9.
  37. Han VK, D'Ercole AJ, Lund PK. Cellular localization of somatomedin (insulin-like growth factor) messenger RNA in the human fetus. Science. 1987;236:193-197.
  38. Yu Y, Yan J, Li M, Yan L, Zhao Y, Lian Y, Li R, Liu P, Qiao J. Effects of combined epidermal growth factor, brain-derived neurotrophic factor and insulin-like growth factor-1 on human oocyte maturation and early fertilized and cloned embryo development. Hum Reprod. 2012;27:2146-59.
  39. Rifas-Shiman SL, Fleisch A, Hivert MF, Mantzoros C, Gillman MW, Oken E, Lighten AD. First and second trimester gestational weight gains are most strongly associated with cord blood levels of hormones at delivery important for glycemic control and somatic growth. Metabolism. 2017;69:112-119.
  40. Bang P, Westgren M, Schwander J, Blum WF, Rosenfeld RG, Stangenberg M. Ontogeny of insulin-like growth factor-binding protein-1, -2, and -3: quantitative measurements by radioimmunoassay in human fetal serum. Pediatr Res. 1994;36:528-536.
  41. Lassarre C, Hardouin S, Daffos F, Forestier F, Frankenne F, Binoux M. Serum insulin-like growth factors and insulin-like growth factor binding proteins in the human fetus. Relationships with growth in normal subjects and in subjects with intrauterine growth retardation. Pediatr Res. 1991;29:219-225.
  42. Östlund E, Bang P, Hagenäs L, Fried G. Insulin-like growth factor I in fetal serum obtained by cordocentesis is correlated with intrauterine growth retardation. Hum Reprod. 1997;12:840-844.
  43. Dauber A, Muñoz-Calvo MT, Barrios V, Domené HM, Kloverpris S, Serra-Juhé C, Desikan V, Pozo J, Muzumdar R, Martos-Moreno GÁ, Hawkins F, Jasper HG, Conover CA, Frystyk J, Yakar S, Hwa V, Chowen JA, Oxvig C, Rosenfeld RG, Pérez-Jurado LA, Argente J. Mutations in pregnancy-associated plasma protein A2 cause short stature due to low IGF-I availability. EMBO Mol Med. 2016;8:363-374.
  44. Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986;1:1077-1081.
  45. Tobi EW, Goeman JJ, Monajemi R, Gu H, Putter H, Zhang Y, Slieker RC, Stok AP, Thijssen PE, Müller F, van Zwet EW, Bock C, Meissner A, Lumey LH, Eline Slagboom P, Heijmans BT. DNA methylation signatures link prenatal famine exposure to growth and metabolism. Nat Commun. 2014;5:5592.
  46. Veening MA, Van Weissenbruch MM, Delemarre-Van De Waal HA. Glucose tolerance, insulin sensitivity, and insulin secretion in children born small for gestational age. J Clin Endocrinol Metab. 2002;87:4657-4661.
  47. Sandhu MS, Heald AH, Gibson JM, Cruickshank JK, Dunger DB, Wareham NJ. Circulating concentrations of insulin-like growth factor-I and development of glucose intolerance: a prospective observational study. Lancet. 2002;359:1740-1745.
  48. Dunger D, Yuen K, Ong K. Insulin-like growth factor I and impaired glucose tolerance. Horm Res. 2004;62 Suppl 1:101-107.
  49. Öst A, Lempradl A, Casas E, Weigert M, Tiko T, Deniz M, Pantano L, Boenisch U, Itskov PM, Stoeckius M, Ruf M, Rajewsky N, Reuter G, Iovino N, Ribeiro C, Alenius M, Heyne S, Vavouri T, Pospisilik JA. Paternal diet defines offspring chromatin state and intergenerational obesity. Cell. 2014;159:1352-1364.
  50. Juul A, Bang P, Hertel NT, Main K, Dalgaard P, Jørgensen K, Müller J, Hall K, Skakkebaek NE. Serum insulin-like growth factor-I in 1030 healthy children, adolescents, and adults: relation to age, sex, stage of puberty, testicular size, and body mass index. J Clin Endocrinol Metab. 1994;78:744-752.
  51. Juul A, Dalgaard P, Blum WF, Bang P, Hall K, Michaelsen KF, Müller J, Skakkebaek NE. Serum levels of insulin-like growth factor (IGF)-binding protein-3 (IGFBP-3) in healthy infants, children, and adolescents: the relation to IGF-I, IGF-II, IGFBP-1, IGFBP-2, age, sex, body mass index, and pubertal maturation. J Clin Endocrinol Metab. 1995;80:2534-42.
  52. Hinrichs A, Kessler B, Kurome M, Blutke A, Kemter E, Bernau M, Scholz AM, Rathkolb B, Renner S, Bultmann S, Leonhardt H, de Angelis MH, Nagashima H, Hoeflich A, Blum WF, Bidlingmaier M, Wanke R, Dahlhoff M, Wolf E. Growth hormone receptor-deficient pigs resemble the pathophysiology of human Laron syndrome and reveal altered activation of signaling cascades in the liver. Mol Metab. 2018;11:113-128
  53. Alba M, Salvatori R. Familial Growth Hormone Deficiency and Mutations in the GHRH Receptor Gene. Vitam Horm. 2004;69:209-220.
  54. Alatzoglou KS, Turton JP, Kelberman D, Clayton PE, Mehta A, Buchanan C, Aylwin S, Crowne EC, Christesen HT, Hertel NT, Trainer PJ, Savage MO, Raza J, Banerjee K, Sinha SK, Ten S, Mushtaq T, Brauner R, Cheetham TD, Hindmarsh PC, Mullis PE, Dattani MT. Expanding the spectrum of mutations in GH1 and GHRHR: genetic screening in a large cohort of patients with congenital isolated growth hormone deficiency. J Clin Endocrinol Metab. 2009;94:3191-3199.
  55. The role of growth hormone in determining birth size and early postnatal growth, using congenital growth hormone deficiency (GHD) as a model. Mehta A, Hindmarsh PC, Stanhope RG, Turton JP, Cole TJ, Preece MA, Dattani MT.Clin Endocrinol (Oxf). 2005 Aug;63(2):223-31. doi: 10.1111/j.1365-2265.2005.02330.x.
  56. Savage MO, Blum WF, Ranke MB, Postel-Vinay MC, Cotterill AM, Hall K, Chatelain PG, Preece MA, Rosenfeld RG. Clinical features and endocrine status in patients with growth hormone insensitivity (Laron syndrome). J Clin Endocrinol Metab. 1993;77:1465-1471.
  57. Laron Z. Lessons from 50 years of study of Laron Syndrome. Endocr Pract. 2015;21:1395-1402. Jensen RB, Jeppesen KA, Vielwerth S,  Michaelsen KM,  Main KM,  Skakkebaek NE, Juul Insulin-like growth factor I (IGF-I) and IGF-binding protein 3 as diagnostic markers of growth hormone deficiency in infancy Horm Res 2005;63(1):15-21. doi: 10.1159/000082456.Epub 2004 Nov 30.
  58. Waters MJ, Hoang HN, Fairlie DP, Pelekanos RA, Brown RJ. New insights into growth hormone action. J Mol Endocrinol. 2006;36:1-7.
  59. Maes M, Maiter D, Thissen JP, Underwood LE, Ketelslegers JM. Contributions of growth hormone receptor and postreceptor defects to growth hormone resistance in malnutrition. Trends Endocrinol Metab. 1991;2:92-97.
  60. Argente J, Caballo N, Barrios V, Pozo J, Muñoz MT, Chowen JA, Hernández M. Multiple endocrine abnormalities of the growth hormone and insulin-like growth factor axis in prepubertal children with exogenous obesity: effect of short- and long-term weight reduction. J Clin Endocrinol Metab. 1997;82:2076-2083.
  61. Bang P, Brismar K, Rosenfeld RG, Hall K. Fasting affects serum insulin-like growth factors (IGFs) and IGF-binding proteins differently in patients with noninsulin-dependent diabetes mellitus versus healthy nonobese and obese subjects. J Clin Endocrinol Metab. 1994;78:960-967.
  62. Argente J, Caballo N, Barrios V, Muñoz MT, Pozo J, Chowen JA, Morandé G, Hernández M. Multiple endocrine abnormalities of the growth hormone and insulin-like growth factor axis in patients with anorexia nervosa: effect of short- and long-term weight recuperation. J Clin Endocrinol Metab. 1997;82:2084-2092.
  63. Dunger DB, Acerini CL. IGF-I and diabetes in adolescence. Diabetes Metab. 1998;24:101-107.
  64. Frystyk J, Skjaerbaek C, Vestbo E, Fisker S, Orskov H. Circulating levels of free insulin-like growth factors in obese subjects: the impact of type 2 diabetes. Diabetes Metab Res Rev. 1999;15:314-322.
  65. Van Dam EW, Roelfsema F, Helmerhorst FH, Frölich M, Meinders AE, Veldhuis JD, Pijl H. Low amplitude and disorderly spontaneous growth hormone release in obese women with or without polycystic ovary syndrome. J Clin Endocrinol Metab. 2002;87:4225-4230.
  66. Roemmich JN, Clark PA, Weltman A, Veldhuis JD, Rogol AD. Pubertal alterations in growth and body composition: IX. Altered spontaneous secretion and metabolic clearance of growth hormone in overweight youth. Metabolism. 2005;54:1374-1383.
  67. Stanley TL, Levitsky LL, Grinspoon SK, Misra M. Effect of body mass index on peak growth hormone response to provocative testing in children with short stature. J Clin Endocrinol Metab. 2009;94:4875-4881.
  68. Salgin B, Marcovecchio ML, Hill N, Dunger DB, Frystyk J. The effect of prolonged fasting on levels of growth hormone-binding protein and free growth hormone. Growth Horm IGF Res. 2012;22:76-81.
  69. Hanaire-Broutin H, Sallerin-Caute B, Poncet MF, Tauber M, Bastide R, Chalé JJ, Rosenfeld R, Tauber JP. Effect of intraperitoneal insulin delivery on growth hormone binding protein, insulin-like growth factor (IGF)-I, and IGF-binding protein-3 in IDDM. Diabetologia. 1996;39:1498-1504.
  70. Shishko PI, Dreval AV, Abugova IA, Zajarny IU, Goncharov VC. Insulin-like growth factors and binding proteins in patients with recent-onset type 1 (insulin-dependent) diabetes mellitus: influence of diabetes control and intraportal insulin infusion. Diabetes Res Clin Pract. 1994;25:1-12.
  71. Chia DJ. Mechanisms of growth hormone-mediated gene regulation. Mol Endocrinol. 2014;28:1012-1025.
  72. Heffernan M1, Summers RJ, Thorburn A, Ogru E, Gianello R, Jiang WJ, Ng FM. The effects of human GH and its lipolytic fragment (AOD9604) on lipid metabolism following chronic treatment in obese mice and beta(3)-AR knock-out mice. Endocrinology. 2001;142:5182-5189.
  73. Fain JN, Ihle JH, Bahouth SW. Stimulation of lipolysis but not of leptin release by GH is abolished in adipose tissue from STAT5a and b knockout mice. Biochem Biophys Res Commun. 1999;263:201–205.
  74. Clodfelter KH, Holloway MG, Hodor P, Park SH, Ray WJ, Waxman DJ. Sex-dependent liver gene expression is extensive and largely dependent upon signal transducer and activator of transcription 5b (STAT5b): STAT5b-dependent activation of male genes and repression of female genes revealed by microarray analysis. Mol Endocrinol. 2006;20:1333-1351.
  75. Sharma VM, Vestergaard ET, Jessen N, Kolind-Thomsen P, Nellemann B, Nielsen TS, Vendelbo MH, Møller N, Sharma R, Lee KY, Kopchick JJ, Jørgensen JOL, Puri V. Growth hormone acts along the PPARγ-FSP27 axis to stimulate lipolysis in human adipocytes. Am J Physiol Endocrinol Metab. 2019;316:E34-E42.
  76. Näntö-Salonen K, Muller HL, Hoffman AR, Vu TH, Rosenfeld RG. Mechanisms of thyroid hormone action on the insulin-like growth factor system: all thyroid hormone effects are not growth hormone mediated. Endocrinology. 1993;132:781-788.
  77. Veldhuis JD, Keenan DM, Bailey JN, Adeniji A, Miles JM, Paulo R, Cosma M, Soares-Welch C. Testosterone supplementation in older men restrains insulin-like growth factor's dose-dependent feedback inhibition of pulsatile growth hormone secretion. J Clin Endocrinol Metab. 2009;94:246-254.
  78. Colón E, Svechnikov KV, Carlsson-Skwirut C, Bang P, Soder O. Stimulation of steroidogenesis in immature rat Leydig cells evoked by interleukin-1alpha is potentiated by growth hormone and insulin-like growth factors. Endocrinology. 2005;146:221-30.
  79. Giudice LC. Insulin-like growth factor family in Graafian follicle development and function. J Soc Gynecol Investig. 2001;8(1 Suppl Proceedings):S26-9.
  80. Conover CA, Faessen GF, Ilg KE, Chandrasekher YA, Christiansen M, Overgaard MT, Oxvig C, Giudice LC. Pregnancy-associated plasma protein-a is the insulin-like growth factor binding protein-4 protease secreted by human ovarian granulosa cells and is a marker of dominant follicle selection and the corpus luteum. Endocrinology. 2001;142:2155-2161.
  81. Chrysis D, Zaman F, Chagin AS, Takigawa M, Sävendahl L. Dexamethasone induces apoptosis in proliferative chondrocytes through activation of caspases and suppression of the Akt-phosphatidylinositol 3'-kinase signaling pathway. Endocrinology. 2005;146:1391-1397.
  82. Bang P, Degerblad M, Thorén M, Schwander J, Blum W, Hall K. Insulin-like growth factor (IGF) I and II and IGF binding protein (IGFBP) 1, 2 and 3 in serum from patients with Cushing's syndrome. Acta Endocrinol (Copenh). 1993;128:397-404.
  83. Öberg D, Salemyr J, Örtqvist E, Juul A, Bang P. A longitudinal study of serum insulin-like growth factor-I levels over 6 years in a large cohort of children and adolescents with type 1 diabetes mellitus: A marker reflecting diabetic retinopathy. Pediatr Diabetes. 2018;19:972-978.
  84. Maile LA, Busby WH, Nichols TC, Bellinger DA, Merricks EP, Rowland M, Veluvolu U, Clemmons DR. A monoclonal antibody against alphaVbeta3 integrin inhibits development of atherosclerotic lesions in diabetic pigs. Sci Transl Med. 2010;2:18ra11.
  85. Vella A, Bouatia-Naji N, Heude B, Cooper JD, Lowe CE, Petry C, Ring SM, Dunger DB, Todd JA, Ong KK. Association analysis of the IGF1 gene with childhood growth, IGF-1 concentrations and type 1 diabetes. Diabetologia. 2008;51:811-815.
  86. Hedman CA, Frystyk J, Lindström T, Oskarsson P, Arnqvist HJ. Intraperitoneal insulin delivery to patients with type 1 diabetes results in higher serum IGF-I bioactivity than continuous subcutaneous insulin infusion. Clin Endocrinol (Oxf). 2014;81:58-62.
  87. Saukkonen T, Shojaee-Moradie F, Williams RM, Amin R, Yuen KC, Watts A, Acerini CL, Umpleby AM, Dunger DB. Effects of recombinant human IGF-I/IGF-binding protein-3 complex on glucose and glycerol metabolism in type 1 diabetes. Diabetes. 2006;55:2365-2370.
  88. Acerini CL, Patton CM, Savage MO, Kernell A, Westphal O, Dunger DB. Randomised placebo-controlled trial of human recombinant insulin-like growth factor I plus intensive insulin therapy in adolescents with insulin-dependent diabetes mellitus. Lancet. 1997;350:1199-1204.
  89. Woods KA, Camacho-Hübner C, Savage MO, Clark AJ. Intrauterine growth retardation and postnatal growth failure associated with deletion of the insulin-like growth factor I gene. N Engl J Med. 1996;335:1363-1367.
  90. Woods KA, Camacho-Hübner C, Bergman RN, Barter D, Clark AJ, Savage MO. Effects of insulin-like growth factor I (IGF-I) therapy on body composition and insulin resistance in IGF-I gene deletion. J Clin Endocrinol Metab. 2000;85:1407-1411.
  91. Dohm GL, Elton CW, Raju MS, Mooney ND, DiMarchi R, Pories WJ, Flickinger EG, Atkinson SM Jr, Caro JF. IGF-I--stimulated glucose transport in human skeletal muscle and IGF-I resistance in obesity and NIDDM. Diabetes. 1990;39:1028-1032.
  92. Nakae J, Kato M, Murashita M, Shinohara N, Tajima T, Fujieda K. Long-term effect of recombinant human insulin-like growth factor I on metabolic and growth control in a patient with leprechaunism. J Clin Endocrinol Metab. 1998;83:542-549.
  93. Dehghani SM, Karamifar H, Hamzavi SS, Haghighat M, Malek-Hosseini SA. Serum insulinlike growth factor-1 and its binding protein-3 levels in children with cirrhosis waiting for a liver transplant. Exp Clin Transplant. 2012;10:252-257.
  94. Moayeri H, Oloomi Z. Prevalence of growth and puberty failure with respect to growth hormone and gonadotropins secretion in beta-thalassemia major. Arch Iran Med. 2006;9:329-334.
  95. Alisi A, Pampanini V, De Stefanis C, Panera N, Deodati A, Nobili V, Cianfarani S. Expression of insulin-like growth factor I and its receptor in the liver of children with biopsy-proven NAFLD. PLoS One. 2018;13:e0201566.
  96. E CY, Cai Y, Sun BZ, Guan LY, Jiang T. Hepatic insulin-like growth factor receptor is upregulated by activation of the GSK3B-FOXO3 pathway after partial hepatectomy. J Biol Regul Homeost Agents. 2017;31:549-555.
  97. Kofoed EM, Hwa V, Little B, Woods KA, Buckway CK, Tsubaki J, Pratt KL, Bezrodnik L, Jasper H, Tepper A, Heinrich JJ, Rosenfeld RG. Growth hormone insensitivity associated with a STAT5b mutation. N Engl J Med. 2003;349:1139-1147.
  98. Binder G, Neuer K, Ranke MB, Wittekindt NE. PTPN11 mutations are associated with mild growth hormone resistance in individuals with Noonan syndrome. J Clin Endocrinol Metab. 2005;90:5377-81.
  99. De Rocca Serra-Nédélec A, Edouard T, Tréguer K, Tajan M, Araki T, Dance M, Mus M, Montagner A, Tauber M, Salles JP, Valet P, Neel BG, Raynal P, Yart A. Noonan syndrome-causing SHP2 mutants inhibit insulin-like growth factor 1 release via growth hormone-induced ERK hyperactivation, which contributes to short stature. Proc Natl Acad Sci U S A. 2012;109:4257-4262.
  100. Vesterlund M, Zadjali F, Persson T, Nielsen ML, Kessler BM, Norstedt G, Flores-Morales A. The SOCS2 ubiquitin ligase complex regulates growth hormone receptor levels. PLoS One. 2011;6:e25358.
  101. Hoppe C, Mølgaard C, Juul A, Michaelsen KF. High intakes of skimmed milk, but not meat, increase serum IGF-I and IGFBP-3 in eight-year-old boys. Eur J Clin Nutr. 2004;58):1211-1216.
  102. Hoppe C, Mølgaard C, Vaag A, Barkholt V, Michaelsen KF. High intakes of milk, but not meat, increase s-insulin and insulin resistance in 8-year-old boys. Eur J Clin Nutr. 2005;59:393-398.
  103. Mogami H, Yura S, Itoh H, Kawamura M, Fujii T, Suzuki A, Aoe S, Ogawa Y, Sagawa N, Konishi I, Fujii S. Isocaloric high-protein diet as well as branched-chain amino acids supplemented diet partially alleviates adverse consequences of maternal undernutrition on fetal growth. Growth Horm IGF Res. 2009;19:478-485.
  104. D'Esposito V, Passaretti F, Hammarstedt A, Liguoro D, Terracciano D, Molea G, Canta L, Miele C, Smith U, Beguinot F, Formisano P. Adipocyte-released insulin-like growth factor-1 is regulated by glucose and fatty acids and controls breast cancer cell growth in vitro. Diabetologia. 2012;55:2811-2822.
  105. Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009;30:586-623.
  106. DeChiara TM, Robertson EJ, Efstratiadis A. Parental imprinting of the mouse insulin-like growth factor II gene. Cell. 1991;64:849-859.
  107. Rossignol S, Netchine I, Le Bouc Y, Gicquel C. Epigenetics in Silver-Russell syndrome. Best Pract Res Clin Endocrinol Metab. 2008;22:403-414.
  108. Clemmons DR. Role of IGF-binding proteins in regulating IGF responses to changes in metabolism. J Mol Endocrinol. 2018;61:T139-T169.
  109. Banskota NK, Carpentier JL, King GL. Processing and release of insulin and insulin-like growth factor I by macro- and microvascular endothelial cells. Endocrinology. 1986;119:1904-1913.
  110. Maile LA, Badley-Clarke J, Clemmons DR. The association between integrin-associated protein and SHPS-1 regulates insulin-like growth factor-I receptor signaling in vascular smooth muscle cells. Mol Biol Cell. 2003;14:3519-3528.
  111. Bang P, Ahlsén M, Berg U, Carlsson-Skwirut C. Free insulin-like growth factor I: are we hunting a ghost? Horm Res. 2001;55 Suppl 2:84-93.
  112. Bang P, Thorell A, Carlsson-Skwirut C, Ljungqvist O, Brismar K, Nygren J. Free dissociable IGF-I: Association with changes in IGFBP-3 proteolysis and insulin sensitivity after surgery. Clin Nutr. 2016;35:408-413.
  113. Juul A, Holm K, Kastrup KW, Pedersen SA, Michaelsen KF, Scheike T, Rasmussen S, Müller J, Skakkebaek NE. Free insulin-like growth factor I serum levels in 1430 healthy children and adults, and its diagnostic value in patients suspected of growth hormone deficiency. J Clin Endocrinol Metab. 1997;82:2497-2502.
  114. Ekström K, Pulkkinen MA, Carlsson-Skwirut C, Brorsson AL, Ma Z, Frystyk J, Bang P. Tissue IGF-I Measured by Microdialysis Reflects Body Glucose Utilization After rhIGF-I Injection in Type 1 Diabetes.J Clin Endocrinol Metab. 2015;100:4299-4306.
  115. Baxter RC. Insulin-like growth factor binding proteins in the human circulation: a review. Horm Res. 1994;42:140-144.
  116. Bang P, Stangenberg M, Westgren M, Rosenfeld RG. Decreased ternary complex formation and predominance of a 29 kDa IGFBP-3 fragment in human fetal serum. Growth Regul. 1994;4:68-76.
  117. Ekström K, Carlsson-Skwirut C, Ritzén EM, Bang P. Insulin-like growth factor-I and insulin-like growth factor binding protein-3 cotreatment versus insulin-like growth factor-I alone in two brothers with growth hormone insensitivity syndrome: effects on insulin sensitivity, body composition and linear growth. Horm Res Paediatr. 2011;76:355-366.
  118. Giudice LC, Farrell EM, Pham H, Lamson G, Rosenfeld RG. Insulin-like growth factor binding proteins in maternal serum throughout gestation and in the puerperium: effects of a pregnancy-associated serum protease activity. J Clin Endocrinol Metab. 1990;71:806-816.
  119. Hossenlopp P, Segovia B, Lassarre C, Roghani M, Bredon M, Binoux M. Evidence of enzymatic degradation of insulin-like growth factor-binding proteins in the 150K complex during pregnancy. J Clin Endocrinol Metab. 1990;71:797-805.
  120. Diamandi A, Mistry J, Krishna RG, Khosravi J. Immunoassay of insulin-like growth factor-binding protein-3 (IGFBP-3): new means to quantifying IGFBP-3 proteolysis. J Clin Endocrinol Metab. 2000;85:2327-2333.
  121. Ahlsén M, Carlsson-Skwirut C, Jonsson AP, Cederlund E, Bergman T, Bang P. A 30-kDa fragment of insulin-like growth factor (IGF) binding protein-3 in human pregnancy serum with strongly reduced IGF-I binding. Cell Mol Life Sci. 2007;64:1870-1880.
  122. Yan X, Payet LD, Baxter RC, Firth SM. Activity of human pregnancy insulin-like growth factor binding protein-3: determination by reconstituting recombinant complexes. Endocrinology. 2009;150:4968-4976.
  123. Zachrisson I, Brismar K, Carlsson-Skwirut C, Dahlquist G, Wallensteen M, Bang P. Increased 24 h mean insulin-like growth factor binding protein-3 proteolytic activity in pubertal type 1 diabetic boys. Growth Horm IGF Res. 2000;10:324-331.
  124. Bang P, Brismar K, Rosenfeld RG. Increased proteolysis of insulin-like growth factor-binding protein-3 (IGFBP-3) in noninsulin-dependent diabetes mellitus serum, with elevation of a 29-kilodalton (kDa) glycosylated IGFBP-3 fragment contained in the approximately 130- to 150-kDa ternary complex. J Clin Endocrinol Metab. 1994;78:1119-1127.
  125. Bang P, Fielder PJ. Human pregnancy serum contains at least two distinct proteolytic activities with the ability to degrade insulin-like growth factor binding protein-3. Endocrinology. 1997;138:3912-3917.
  126. Rajah R, Katz L, Nunn S, Solberg P, Beers T, Cohen P. Insulin-like growth factor binding protein (IGFBP) proteases: functional regulators of cell growth. Prog Growth Factor Res. 1995;6:273-84.
  127. Martin DC, Fowlkes JL, Babic B, Khokha R. Insulin-like growth factor II signaling in neoplastic proliferation is blocked by transgenic expression of the metalloproteinase inhibitor TIMP-1. J Cell Biol. 1999;146:881-892.
  128. Gaidamauskas E, Gyrup C, Boldt HB, Schack VR, Overgaard MT, Laursen LS, Oxvig C.IGF dependent modulation of IGF binding protein (IGFBP) proteolysis by pregnancy-associated plasma protein-A (PAPP-A): multiple PAPP-A-IGFBP interaction sites. Biochim Biophys Acta. 2013 Mar;1830(3):2701-9. doi: 10.1016/j.bbagen.2012.11.002.PMID: 23671931
  129. Bang P, Ahmed SF, Argente J, Backeljauw P, Bettendorf M, Bona G, Coutant R, Rosenfeld RG, Walenkamp MJ, Savage MO. Identification and management of poor response to growth-promoting therapy in children with short stature. Clin Endocrinol (Oxf). 2012;77:169-181.
  130. Bang P, Polak M, Woelfle J, Houchard A; EU IGFD Registry Study Group. Effectiveness and Safety of rhIGF-1 Therapy in Children: The European Increlex® Growth Forum Database Experience. Horm Res Paediatr. 2015;83:345-357.
  131. Murray and Clayton. Disorders of growth hormone in childhood. MDText.com 2022
  132. Cabrera-Salcedo C, Mizuno T, Tyzinski L, Andrew M, Vinks AA, Frystyk J, Wasserman H, Gordon CM, Hwa V, Backeljauw P, Dauber A. Pharmacokinetics of IGF-1 in PAPP-A2-Deficient Patients, Growth Response, and Effects on Glucose and Bone Density. J Clin Endocrinol Metab. 2017 Dec 1;102(12):4568-4577. doi: 10.1210/jc.2017-01411.
  133. Muthuvel G, Dauber A, Alexandrou E, Tyzinski L, Andrew M, Hwa V, Backeljauw P. Five-Year Therapy with Recombinant Human Insulin-Like Growth Factor-1 in a Patient with PAPP-A2 Deficiency. Horm Res Paediatr. 2023;96(5):449-457. doi: 10.1159/000529071. Epub 2023 Jan 16.
  134. Muñoz-Calvo MT, Barrios V, Pozo J, Chowen JA, Martos-Moreno GÁ, Hawkins F, Dauber A, Domené HM, Yakar S, Rosenfeld RG, Pérez-Jurado LA, Oxvig C, Frystyk J, Argente J. Treatment With Recombinant Human Insulin-Like Growth Factor-1 Improves Growth in Patients With PAPP-A2 Deficiency. J Clin Endocrinol Metab. 2016 Nov;101(11):3879-3883. doi: 10.1210/jc.2016-2751. Epub 2016 Sep 20.

Evaluation and Treatment of Dyslipidemia in the Elderly

ABSTRACT

 

The definition of elderly is arbitrary. In this chapter we will define elderly as greater than 75 years of age because both the US and European lipids guidelines use this age to differentiate therapy recommendations. Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in the elderly. Age is a key risk factor for ASCVD and with identical risk factors the 10-year risk of an ASCVD event markedly increases with age. In fact, an older individual with excellent risk factors can still have a high risk for having an ASCVD event. ASCVD begins early in life and progresses until it leads to clinical events later in life. The age that one develops clinical manifestations of ASCVD is dependent on the severity of individual risk factors, the number of risk factors, and the duration of exposure to the risk factors. Elderly individuals have a long exposure to risk factors so even when the risk factors are relatively modest the cumulative effects can be sufficient to result in clinical ASCVD events. This explains why age is such a key variable in determining the risk of developing an ASCVD event. Cardiovascular outcome studies have demonstrated that lowering LDL-C levels with statins, ezetimibe, or PCSK 9 monoclonal antibodies will reduce ASCVD events in elderly patients with pre-existing cardiovascular disease (secondary prevention). In elderly patients without cardiovascular disease (primary prevention) the available data does not definitively demonstrate a decrease in ASCVD events with statin or ezetimibe therapy but is suggestive of a benefit (note there are no primary prevention trials with PCSK9 inhibitors). Additional data is required to determine if bempedoic acid and icosapent ethyl reduce ASCVD events in patients ≥ 75 years of age. Studies are currently underway to provide definitive information on whether statin therapy is beneficial as primary prevention in the elderly. In deciding whether to treat an elderly patient with lipid lowering drugs one needs to consider the following factors; the higher the LDL-C level the greater the benefit of lowering LDL-C, the greater the decrease in LDL-C the greater the benefit, the higher the absolute risk of ASCVD the greater the benefit of lowering LDL-C, life expectancy, competing non-cardiovascular disorders, risk of drug side effects, potential for drug interactions, and patient preferences. In elderly patients without pre-existing ASCVD one should estimate the patient’s risk of developing ASCVD events and in conjunction with the general principles described above discuss with the patient a treatment plan. Determining the coronary calcium score can be helpful if there is uncertainty regarding the appropriate decision. If the decision is to treat our goal in primary prevention patients is often an LDL-C < 100mg/dL but in high-risk patients our goal may be an LDL-C < 70mg/dL. Elderly patients with ASCVD should be treated with lipid lowering drugs to reduce ASCVD unless there are contraindications. At a minimum our goal is an LDL-C < 70mg/dL but we would prefer an LDL-C < 55mg/dL if they can be achieved with a statin + ezetimibe. In very high-risk patients our goal is an LDL-C < 55mg/dL and adding a PCSK9 inhibitor may be required to achieve these levels in some patients. Age per se should not be used to withhold therapy with lipid lowering drugs that can reduce the risk of ASCVD events.

 

INTRODUCTION

 

Due to a decreasing birth rate and a longer life expectancy the population is getting older. According to the US census in 2020 there were approximately 50 million people between 65 and 84 years of age (14.9% of the total population) and approximately 6 million between 85 and 99 years of age (1.89% of the total population). The number of Americans ages 65 and older is projected to increase to 82 million by 2050 (23% of the total population). World-wide there are 703 million people aged 65 or older, which is projected to reach 1.5 billion by 2050 (1 in 6 people). It is well recognized that atherosclerotic cardiovascular disease (ASCVD) increases with age and is a major cause of morbidity and mortality in the elderly. In addition to an increased risk of coronary artery disease there is more than a doubling of the prevalence of peripheral arterial disease, cerebrovascular disease, and abdominal aortic aneurism with each decade of life (1). Unfortunately, the elderly (≥ 75 years of age) have not been well represented in lipid lowering cardiovascular outcome trials.

 

The definition of elderly is arbitrary. In this chapter we will define elderly as greater than 75 years of age because both the US and European guidelines use this age to differentiate therapy recommendations. In both the “Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines” and the “2019 ESC/EAS Guidelines for the Management of Dyslipidemias: Lipid Modification to Reduce Cardiovascular Risk” the recommendations for those over 75 years of age differ from recommendations for younger individuals (2,3). Thus, where possible we will focus on studies in individuals greater than 75 years of age.

 

LIPID LEVELS IN THE ELDERLY

 

Lipid levels in US adults from the National Health and Nutrition Examination Survey (NHANES) 2003-2004 are shown in Table 1 (4). Compared to 30–69-year-olds there is a slight decrease in LDL-C, non-HDL-C, and triglycerides with similar HDL-C levels in individuals 70-79 years of age. Other cross-sectional studies have reported similar results (5-11). Prospective studies with longitudinal follow-up have also observed small decreases in total cholesterol, LDL-C, and HDL-C levels in men and women as they become elderly (6,7,9,12-14). It should be noted that the changes in lipid levels reported with aging are relatively small and vary somewhat from study to study. The clinical significance of these small changes is uncertain.

 

Table 1. Lipid Levels in U.S. Adults (NHANES 2003-2004)

Age

LDL-C (mg/dL)

Non-HDL-C (mg/dL)

HDL-C (mg/dL)

Triglycerides (mg/dL)

20-29

104

126

54

105

30-39

120

146

54

118

40-49

124

152

53

144

50-59

123

154

55

141

60-69

126

157

54

145

70-79

119

148

56

133

 

Studies have demonstrated that older individuals have an exaggerated postprandial lipemia compared with younger individuals (15,16). While elevated postprandial triglycerides is associated with an increased risk of ASCVD whether this plays a causal role in increasing ASCVD is uncertain.

 

It is well recognized that with increasing age the likelihood of other medical disorders increases and this can affect lipid levels. For example, inflammation and infections can decrease LDL-C and HDL-C levels (17). Additionally, poor nutrition due to illness or social-economic factors could decrease lipid levels in the elderly. Finally, frailty is a syndrome associated with aging and increases with age. It is usually associated with a lowering of total, LDL-C, and non-HDL cholesterol levels (18-20).

 

AGE IS AN IMPORTANT RISK FOR ATHEROSCLEROTIC CARDIOVASCULAR DISEASE

 

The clearest way to illustrate the importance of age as a key risk factor for atherosclerotic cardiovascular disease (ASCVD) is to compare the 10-year risk at different ages using an updated version of the AHA/ACC pooled cohort equation. Shown in table 2 are four examples of the effect of age on 10-year risk in different clinical situations that demonstrate the marked effect of age on ASCVD risk. Similarly, using the SCORE risk calculator for determining the 10-year risk of fatal cardiovascular disease also demonstrates the very large impact of age on risk (figure 1). It is obvious that age is a major determinant of ASCVD risk.

 

Table 2. Ten Year Risk of Developing ASCVD

 

Age 55

Age 65

Age 75

Male, white, BP 130, TC 200, HDL-C 45, non-smoker, no diabetes

6.3%

13.9%

26.2%

Female, African American, BP120, TC 180, HDL-C 50, non-smoker, no diabetes

2.8%

6.3%

13.2%

Male, African American, BP140, TC 200, HDL-C 50, smoker, no diabetes

10.1%

14.9%

20.5%

Female, white, BP 140, TC 180, HDL-C 50, non-smoker, diabetes

4.4%

12.2%

32.9%

https://tools.acc.org/ascvd-risk-estimator-plus/#!/calculate/estimate/.

BP= systolic BP mm Hg, TC= total cholesterol mg/dL.

 

Figure 1. Systematic Coronary Risk Estimation chart for European populations at high cardiovascular disease risk (from 2019 ESC/EAS Guidelines for the management of dyslipidaemias (3)).

 

In fact, an older individual with excellent risk factors can have a high risk for having an ASCVD event. For example, using the AHA/ACC pooled cohort equation a 75-year-old white male with a total cholesterol of 180mg/dL, an HDL-C of 50mg/dL, a blood pressure of 120/80 mmHg, who is not diabetic, doesn’t smoke, and is on no medications still has a 10-year risk for an ASCVD event of 21.7%. A 75-year-old white female with the same risk factors also has a relatively high 10-year risk (14.1%). Using the SCORE estimator (figure 1) for a fatal CVD event it is also apparent that many older individuals, particularly males, are at high risk even when they are non-smokers with an excellent total cholesterol and blood pressure. For example, a 70-year-old male, non-smoker with a total cholesterol of 160mg/dL and systolic BP of 120 mmHg still has a 13% 10-year risk of death from CVD.

 

WHY ARE OLDER INDIVIDUALS AT HIGHER RISK FOR ASCVD?

 

It is widely recognized that atherosclerosis begins early in life and slowly progresses ultimately resulting in clinical manifestations later in life (21). Numerous studies have demonstrated the presence of atherosclerosis in young individuals (22-27). In the Bogalusa Heart Study autopsies were performed on 204 young people 2 to 39 years of age (22,28). In the coronary arteries fatty streaks were very common (50 percent at 2 to 15 years of age and 85 percent at 21 to 39 years of age). More advanced raised fibrous-plaque lesions in the coronary arteries were present in 8 percent of individuals 2 to 15 years of age and 69 percent of individuals 26 to 39 years of age. The extent of the atherosclerotic lesions correlated positively with BMI, systolic and diastolic BP, total cholesterol, LDL-C, and triglyceride levels and negatively with HDL-C levels. The extent of the atherosclerotic lesions was greatest in individuals who had multiple risk factors. The Pathobiological Determinants of Atherosclerosis in Youth [PDAY] study examined the effect of risk factors for atherosclerosis in 1079 men and 364 women 15 through 34 years of age who died due to accidents, homicide, or suicide (23,29). Atherosclerosis of the aorta and right coronary artery was measured and increased with age, LDL-C levels, glycohemoglobin levels, BMI, and smoking while HDL-C levels were negatively associated with the extent of fatty streaks and raised lesions in the aorta and right coronary artery. Finally, in a study of US service members (mean age 25.9 years; range 18-59 years; 98.3% male) who died of combat or unintentional injuries (n= 3832) the effect of risk factors on coronary atherosclerosis was determined (27). Atherosclerosis prevalence was increased by age, dyslipidemia, hypertension, and obesity. Taken together these studies clearly demonstrate that atherosclerosis begins early in life with the prevalence increasing with age and the extent and onset of lesions is influenced by risk factors, including dyslipidemia.

 

Moreover, the presence of risk factors early in life is associated with an increase in atherosclerosis later in life (30-32). A meta-analysis that included 4380 participants from 4 prospective studies that collected cardiovascular risk factor data during childhood (age 3 to 18 years) and measured carotid intima-media thickness (CIMT) in adulthood (age 20 to 45 years) reported that total cholesterol, triglycerides, BP, and BMI measured in childhood were predictive of elevated CIMT in adults (33). Additionally, increased LDL-C and/or decreased HDL-C during adolescence predict an increase in CIMT later in life (34). Importantly, an increased total cholesterol or BP early in life also predicted an increased risk of developing cardiovascular disease later in life (35-38).

 

Genetic studies have further illustrated the key role of risk factors and duration of exposure to the risk factor as key variables determining the time when clinical manifestations of ASCVD occur. In patients with homozygous familial hypercholesterolemia (FH) LDL-C are markedly elevated and cardiovascular events can occur early in life. Greater than 50% of untreated patients with homozygous FH develop clinically significant ASCVD by the age of 30 and cardiovascular events can occur before age 10 in some patients (39). In patients with heterozygous FH LDL-C levels are elevated but not to the levels seen with homozygous FH and cardiovascular events occur later in life but still at a relatively younger age. Untreated males with heterozygous FH have a 50% risk for a fatal or non-fatal myocardial infarction by 50 years of age whereas untreated females have a 30% chance by age 60 (39). Conversely, individuals with genetic variants in PCSK9, HMG-CoA reductase, LDL receptor, NPC1L1, or ATP citrate lyase that lead to a decrease in LDL-C levels have a reduced risk of developing cardiovascular events (40,41). The relationship between genetic disorders that alter LDL-C levels and the time to develop clinical cardiovascular events is illustrated in figure 2. The figure clearly illustrates that the age when one clinically manifests ASCVD depends on the level of LDL-C. With very high LDL-C levels clinical events occur early in life and with low LDL-C levels events will occur at an older age leading to the concept of LDL years.

 

Figure 2. Relationship between cumulative LDL-C exposure and age of developing cardiovascular disease. (from (41)).

 

The degree and duration of other risk factors also seems to play a role in when the clinical manifestations of ASCVD are expressed. For example, for cigarette smoking, cigarettes/day, smoking duration, and pack-years all increase the risk of cardiovascular disease (42). Interestingly smoking fewer cigarettes/day for a longer duration was more deleterious than smoking more cigarettes/day for a shorter duration (42,43). Additionally, while smoking cessation lowers the risk of ASCVD events an increased risk persists for decades after smoking cessation (44). These observations suggest that the effect of smoking is related to the number of cigarettes smoked and the duration of the smoking (i.e., pack years). Similarly, in patients with diabetes glycemic control and duration of diabetes influences the development of ASCVD complications (45-48). At any given age, a 10-year longer diabetes duration was associated with a 1.1-1.5-fold increased risk of stroke and 1.5-2.0-fold increased risk of MI (45).

 

Thus, ASCVD begins early in life and progresses until it leads to clinical events such as a myocardial infarction or stroke later in life. The age that one develops clinical manifestations of ASCVD is dependent on the severity of individual risk factors, the number of risk factors, and the duration of exposure to the risk factors. Elderly individuals have a long exposure to risk factors so even when the risk factors are relatively modest the cumulative effects can be sufficient to result in clinical ASCVD events. This explains why age is such a key variable in determining the risk of developing an ASCVD event.

 

DOES LIPID LOWERING REDUCE EVENTS IN THE ELDERLY

 

Below we discuss lipid lowering drug studies that report the effect on cardiovascular outcomes that are relevant to clinical decisions in elderly individuals. For additional and more detailed information on lipid lowering cardiovascular outcome studies see the Endotext chapters on “Cholesterol Lowering Drugs” and “Triglyceride Lowering Drugs” (49,50).

 

Statins

 

Few statin studies have focused on lowering LDL-C in elderly patients, which we define as individuals greater than 75 years of age. The Prosper Trial determined the effect of pravastatin 40mg/day (n= 2891) vs. placebo (n= 2913) on cardiovascular events in older subjects (70-82 years of age) with pre-existing vascular disease or who were at high risk for vascular disease (51). The average age in this trial was 75 years of age and approximately 45% had cardiovascular disease. As expected, pravastatin treatment lowered LDL-C by 34% compared to the placebo group. The primary end point was coronary death, non-fatal myocardial infarction, and fatal or non-fatal stroke, which was reduced by 15% (HR 0.85, 95% CI 0.74-0.97, p=0.014). However, in the individuals without pre-existing cardiovascular disease pravastatin did not significantly reduce ASCVD events (HR 0.94; CI 0.77–1.15). In contrast, in individuals with cardiovascular disease pravastatin therapy significantly reduced ASCVD events (HR 0.78, CI 0.66–0.93). Thus, this study demonstrated benefits of statin therapy in the elderly with cardiovascular disease but failed to demonstrate benefit in the elderly without cardiovascular disease.

 

A meta-analysis by the Cholesterol Treatment Trialists of 28 trials with 14,483 of 186,854 found a reduction in LDL-C levels with statin therapy that was similar in the participants ≥75 years of age compared to younger individuals. Moreover, statin therapy resulted in a decrease in cardiovascular events in all age groups including participants ≥75 years of age (Figure 3) (52). In the participants ≥75 there was a 13% reduction in ASCVD events per 39mg/dL decrease in LDL-C (RR 0.87; 95% CI 0.77–0.99). This analysis included four trials done exclusively among people with heart failure or receiving renal dialysis, for whom statin therapy shows little or no benefit (50). A second analysis was performed after elimination of these four trials and there was an 18% reduction in ASCVD events per 39mg/dL decrease in LDL-C (RR 0.82; 95%CI 0.70-0.95). Similar to the Prosper Trial a decrease in ASCVD events was clearly demonstrated in individuals with pre-existing cardiovascular disease (secondary prevention) but in individuals without pre-existing cardiovascular disease (primary prevention) the decrease in ASCVD events was not statistically significant (Figure 4- analysis included all studies). After excluding the trials in patients with heart failure or receiving renal dialysis, statin therapy reduced major ASCVD events by 26% per 39mg/dL decrease in LDL-C (RR 0.74; 95% CI0.60 − 0.91) in patients with pre-existing cardiovascular disease but only by 8% in patients without pre-existing cardiovascular disease (RR 0.92; 95%CI 0.72 − 1.16).

 

Figure 3. Effect of Statin Treatment on Major Vascular Events. Modified from (44).

Figure 4. Effect of Statin Treatment on Major Vascular Events in Individuals With and Without Pre-Existing Cardiovascular Disease. Modified from (44).

 

A statin trial not included in the Cholesterol Treatment Trialists meta-analysis was carried out in patients with an ischemic stroke or a transient ischemic attack who were treated with statins and/or ezetimibe with a target LDL-C level < than 70mg/dL (n= 1430) or an LDL-C 90mg/dL to 110mg/dL (n= 1430) (53). The primary end point was ischemic stroke, myocardial infarction, new symptoms leading to urgent coronary or carotid revascularization, or death from cardiovascular causes. The mean LDL-C level was 65mg/dL in the lower-target group and 96 mg/dL in the higher-target group. After median of 3.5 years the primary end point occurred in 8.5% of the patients in the lower-target group and 10.9% of the patients in the higher target group (HR 0.78; 95% CI 0.61 to 0.98; P=0.04). In patients < 65 years of age only a 7% decrease in the primary end point was observed (HR 0.93; 95%CI 0.63–1.36) whereas more impressive decreases in the primary endpoint were observed in patients 65-75 years of age (37% decrease; HR 0.63 95% CI 0.42–0.95) and > 75 years of age (23% decrease; HR 0.77; 95%CI 0.49–1.22). These results are consistent with the Cholesterol Treatment Trialists meta-analysis demonstrating that elderly patients with pre-existing cardiovascular disease lowering LDL-C levels reduces ASCVD events.

 

There are observational studies demonstrating that statin treatment for the primary prevention of ASCVD is effective in older patients (54-59). For example, in US veterans ≥75 years of age and free of ASCVD at baseline, new statin use was significantly associated with a lower risk of ASCVD events (HR 0.92; 95% CI 0.91-0.94) and cardiovascular mortality (HR 0.80; 95% CI 0.78-0.81) when compared to statin nonusers (55). Similarly, in a Danish nationwide cohort initiation of statin therapy in patients > 70 years of age without pre-existing cardiovascular disease there was a 23% lower risk of major vascular events per 39mg/dL decrease in LDL-C (HR 0.77; 95% CI 0.71-0.83), which was similar to what was observed in younger individuals (54). Finally, in nursing home residents without ASCVD statin use reduced all-cause mortality in individuals with and without dementia (59). These observational studies while suggestive of a benefit of statin therapy for primary prevention in older individuals cannot provide definitive proof as there is always the possibility of residual confounding. Nevertheless, they provide additional support that statin therapy provides benefits in elderly patients without pre-existing cardiovascular disease.

 

Thus, in older patients with cardiovascular disease lowering LDL-C levels with statins clearly reduces cardiovascular events but in older patients without cardiovascular disease the data demonstrating that statins reduce cardiovascular events is less robust but suggests a reduction in ASCVD events.

 

Ezetimibe

 

IMPROVE-IT TRIAL

 

The IMPROVE-IT Trial tested whether the addition of ezetimibe to statin therapy would provide an additional beneficial effect in patients with the acute coronary syndrome (60). The IMPROVE-IT Trial was a large trial with over 18,000 patients randomized to simvastatin 40mg vs. simvastatin 40mg + ezetimibe 10mg per day. On treatment LDL-C levels were 70mg/dL in the statin alone group vs. 54mg/dL in the statin + ezetimibe group. There was a small but significant 6.4% decrease in major cardiovascular events (cardiovascular death, MI, documented unstable angina requiring rehospitalization, coronary revascularization, or stroke) in the statin + ezetimibe group (HR 0.936; 95% CI 0.887-0.988; p=0.016). Cardiovascular death, non-fatal MI, or non-fatal stroke were reduced by 10% (HR 0.90; 95% CI 0.84-0.97; p=0.003). The effect of age on the benefits of statin + ezetimibe therapy is shown in figure 5. In elderly individuals (≥ 75 years of age) the combination of ezetimibe and simvastatin reduced ASCVD events.

 

Figure 5. Primary endpoint in the IMPROVE-IT trial in different age groups. Modified from (60).

 

EWTOPIA 75 TRIAL

 

EWTOPIA 75 was a multicenter, randomized trial in Japan that examined the preventive efficacy of ezetimibe for patients aged ≥75 years (mean age 80.6 years), with elevated LDL-C (≥140 mg/dL) without a history of coronary artery disease (primary prevention) who were not taking lipid lowering drugs (61). Patients were randomized to ezetimibe 10mg (n=1,716) or usual care (n=1,695) and followed for 4.1 years. The primary outcome was a composite of sudden cardiac death, myocardial infarction, coronary revascularization, or stroke. In the ezetimibe group LDL-C was decreased by 25.9% and non-HDL-C by 23.1% while in the usual care group LDL-C was decreased by 18.5% and non-HDL-C by 16.5% (p<0.001 for both lipid parameters). By the end of the trial 9.6% of the patients in the usual care group and 2.1% of the ezetimibe group were taking statins. Ezetimibe reduced the incidence of the primary outcome by 34% (HR 0.66; P=0.002). Additionally, composite cardiac events were reduced by 60% (HR 0.60; P=0.039) and coronary revascularization by 62% (HR 0.38; P=0.007) in the ezetimibe group vs. the control group. There was no difference in the incidence of stroke or all-cause mortality between the groups. It should be noted that the reduction in cardiovascular events was much greater than one would expect based on the absolute difference in LDL-C levels (121mg/dL in ezetimibe group vs. 132mg/dL in usual care group). As stated by the authors “Given the open-label nature of the trial, its premature termination, and issues with follow-up, the magnitude of benefit observed should be interpreted with caution.” Nevertheless, this study suggests that lowering LDL-C in elderly individuals without cardiovascular disease can reduce ASCVD events.

 

RACING TRIAL

 

The RACING trial was a randomized, open-label trial in patients with ASCVD carried out in South Korea (62). Patients were randomly assigned to either rosuvastatin 10 mg with ezetimibe 10 mg (n= 1894) or rosuvastatin 20 mg (n= 1886). The primary endpoint was cardiovascular death, major cardiovascular events, or non-fatal stroke. The median LDL-C level during the study was 58mg/dL in the combination therapy group and 66mg/dL in the statin monotherapy group (p<0.0001). The primary endpoint occurred in 9.1% of the patients in the combination therapy group and 9.9% of the patients in the high-intensity statin monotherapy group (non-inferior). Non-inferiority was observed in patients with baseline LDL-C levels < 100mg/dL and >100mg/dL (63).

 

In the RACING trial 574 participants (15.2%) were aged ≥75 years and there was no difference in the primary endpoint between the combination therapy group and the high-intensity statin monotherapy group in these elderly participants (64). However, in participants ≥75 years of age moderate-intensity statin with ezetimibe combination therapy was associated with lower rates of drug related intolerance with drug discontinuation or dose reduction (2.3% vs 7.2%; P = 0.010).

 

This study demonstrates that moderate intensity statin plus ezetimibe was non-inferior to high-intensity statin therapy with regards to cardiovascular death, major cardiovascular events, or non-fatal stroke. The lower prevalence of discontinuation or dose reduction caused by intolerance to the study drug was seen with combination therapy indicating that using a moderate intensity dose of a statin plus ezetimibe is a useful strategy in patients that do not tolerate high intensity statin therapy or where there are concerns about statin toxicity with high doses.

 

PCSK9 Inhibitors

 

FOURIER TRIAL

 

The FOURIER trial was a randomized, double-blind, placebo-controlled trial of evolocumab vs. placebo in 27,564 patients with ASCVD and an LDL-C level of 70 mg/dL or higher who were on statin therapy (65). The primary end point was cardiovascular death, myocardial infarction, stroke, hospitalization for unstable angina, or coronary revascularization and the key secondary end point was cardiovascular death, myocardial infarction, or stroke. The median duration of follow-up was 2.2 years. Baseline LDL-C levels were 92mg/dL and evolocumab resulted in a 59% decrease in LDL levels (LDL-C level on treatment approximately 30mg/dL). In this trial 6233 of the participants were > 69 years of age and the decrease in LDL was similar in participants > 69 years of age and younger individuals (66). A 14% reduction in the primary endpoint (HR 0.86; 95% CI 0.74–0.99) and a 18% reduction in the secondary endpoint (HR 0.82; 95% CI 0.69–0.98) was observed in the participants > 69 years of age, which was similar to the decreases seen in younger individuals (66). The effect of treatment with evolocumab on the primary and secondary endpoint in specific age groups is shown in table 3 (66). These results demonstrate that lowering LDL-C with a PCSK9 inhibitor decreases ASCVD events in elderly patients.

 

Table 3. Effect of Evolocumab Treatment on Cardiovascular Outcomes in Different Age Groups

 

< 65

65-75

>75

Primary Endpoint

HR 0.86; 95%CI 0.78–0.94

HR 0.86; 95%CI 0.76–0.97

HR 0.78; 95%CI 0.60–1.02

Secondary Endpoint

HR 0.79; 95%CI 0.69–0.90

HR 0.82; 95%CI 0.70–0.95

HR 0.78 95%CI 0.58–1.04

For the primary endpoint the P interaction for the three age groups = 0.84

For the secondary endpoint the P interaction for the three age groups = 0.94.  

 

ODYSSEY TRIAL

 

The ODYSSEY trial was a multicenter, randomized, double-blind, placebo-controlled trial involving 18,924 patients who had an acute coronary syndrome 1 to 12 months earlier, an LDL-C level of at least 70 mg/dL, a non-HDL-C level of at least 100 mg/dL, or an apolipoprotein B level of at least 80 mg/dL while on high intensity statin therapy or the maximum tolerated statin dose (67). Patients were randomly assigned to receive alirocumab 75 mg every 2 weeks or matching placebo. The dose of alirocumab was adjusted to target an LDL-C level of 25 to 50 mg/dL. The primary end point was a composite of death from coronary heart disease, nonfatal myocardial infarction, fatal or nonfatal ischemic stroke, or unstable angina requiring hospitalization. In this trial 5084 (26.9%) individuals were ≥ 65 years of age, 1007 (5.3%) ≥ 75 years of age, and 42 (0.2%) ≥ 85 years of age (68). The baseline and decrease in LDL-C levels were similar in participants ≥65 years of age and those <65 years of age (LDL-C at baseline approximately 94mg/dL and after 4 months of treatment approximately 40mg/dL) (67,68). In the individuals ≥ 65 years of age there was a 22% decrease in the primary endpoint (HR 0.78; 95% CI 0.68–0.91) and in those < 65 years of age a 11% decrease (HR 0.89; 95% CI 0.80–1.00) (68). The secondary endpoint of all-cause death, myocardial infarction, or ischemic stroke was also reduced in the ≥ 65 participants (HR 0.78; 95% CI 0.68–0.90) and < 65 participants (HR 0.91; 0.82–1.02) (68). In participants ≥ 75 years of age the primary endpoint was reduced by 15% (HR 0.85; 95% CI 0.64–1.13) (68). When plotted as a continuous variable the relative benefit of alirocumab over placebo on the primary endpoint was consistent across the entire age range (figure 6).

 

Figure 6. Relative benefit of alirocumab at various ages. Modified from reference (68).

These two studies demonstrate that lowering LDL-C with a PCSK9 inhibitor decreases ASCVD events in elderly patients with pre-existing cardiovascular disease.

 

Bempedoic Acid

 

The CLEAR Outcome trial was a double-blind, randomized, placebo-controlled trial involving patients with cardiovascular disease or at high risk of cardiovascular disease who were unable or unwilling to take statins ("statin-intolerant" patients) (69). The patients were randomized to bempedoic acid 180 mg (n= 6992) or placebo (n= 6978) and the median duration of follow-up was 40.6 months. In this trial 44% of the participants were between ≥65 to < 75 years of age and 15% were ≥ 75 years of age. As expected, LDL-C levels were decreased by 21% in the bempedoic group compared to placebo (29mg/dL difference). The primary endpoint, death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularization, was reduced by 13% in the bempedoic acid group (HR 0.87; 95% CI 0.79- 0.96; P = 0.004). The effect of age on the primary endpoint is shown in table 4.

 

Table 4. Effect of Bempedoic on Cardiovascular Outcomes in Different Age Groups

< 65

HR 0.87; 95% CI 0.74-1.02

≥65-<75

HR 0.83, 95% CI 0.72-0.96

≥ 75

HR 0.95, 95% CI 0.77-1.16

Interaction P value = 0.60

 

Niacin and Fibrates

 

Because of the robust effect of statins in lowering LDL-C levels and cardiovascular events recent trials of both niacin and fibrates have focused on the addition of these lipid lowering drugs to statin therapy. The AIM-HIGH trial was designed to determine if the addition of Niaspan, an extended-release form of niacin, to aggressive statin therapy would result in a further reduction in ASCVD events in patients with pre-existing cardiovascular disease (70) while the HPS 2 Thrive trial determined the effect of adding extended-release niacin (2000mg/day) combined with laropiprant, a prostaglandin D2 receptor antagonist, to statin therapy on ASCVD events in  patients with pre-existing vascular disease (71). Unfortunately, both of these trials failed to demonstrate a decrease in ASCVD events with the addition of niacin to statin therapy. The absence of benefit and increased side effects from niacin therapy has markedly reduced enthusiasm for treating patients with niacin to reduce ASCVD event. For additional details on these two studies and other niacin cardiovascular outcome studies see the Endotext chapter “Triglyceride Lowering Drugs” (49).

 

The ACCORD-LIPID Trial was designed to determine if the addition of fenofibrate to aggressive statin therapy in patients with pre-existing cardiovascular disease or at high risk for developing cardiovascular disease would result in a further reduction in cardiovascular disease in patients with Type 2 diabetes (72). The PROMINENT trial determined whether pemafibrate, a new selective PPAR-alpha activator, in patients on statin therapy with diabetes and pre-existing cardiovascular disease or at high risk for developing cardiovascular disease would reduce cardiovascular events (73). Disappointingly, neither trial demonstrated benefits from adding a fibrate to statin therapy. For additional details on these two studies and other fibrate ASCVD outcome studies see the Endotext chapter “Triglyceride Lowering Drugs” (49).

 

Thus, there is currently little enthusiasm for adding either niacin or a fibrate to statin therapy to reduce ASCVD events. One should recognize that like all studies these trials have limitations, that are discussed in detail in reference (49), and it is possible that future trials could resurrect the use of niacin and/or fibrates for decreasing ASCVD.

 

Omega-3-Fatty Acids (Fish Oil)

 

Numerous studies have determined the effect of low dose fish oil (< 1 gram per day) on ASCVD and found that they do not consistently reduce the risk of cardiovascular disease (49). Described below are ASCVD outcome studies that have used higher doses.

 

JAPAN EPA LIPID INTERVENTION STUDY (JELIS)

 

JELIS was an open label study without a placebo in patients with total cholesterol levels > 254mg/dL with cardiovascular disease (n= 3,664) or without cardiovascular disease (n=14,981) who were randomly assigned to be treated with 1800 mg of EPA (Vascepa) + statin (n=9,326) or statin alone (n= 9,319) with a 5-year follow-up (74). The primary endpoint was any major coronary event, including sudden cardiac death, fatal and non-fatal myocardial infarction, and other non-fatal events including unstable angina pectoris, angioplasty, stenting, or coronary artery bypass grafting. Total cholesterol, LDL-C, and HDL-C levels were similar in the two groups but plasma TGs were modestly decreased in the EPA treated group (5% decrease in EPA group compared to controls; p = 0.0001). In the EPA plus statin group the primary endpoint occurred in 2.8% of the patients vs. 3.5% of the patients in the statin alone group (19% decrease; p = 0.011). In participants < 61 years of age the primary endpoint was reduced by 24% (HR 0.76; 95%CI 0.57–1.00) while in individuals ≥ 61 years of age the primary endpoint was reduced by 16% (HR 0.84; 95% CI 0.68–1.02; p interaction 0.57). Unstable angina and non-fatal coronary events were significantly reduced in the EPA plus statin group but in this study sudden cardiac death and coronary death did not differ between groups. Unstable angina was the main component contributing to the primary endpoint and this is a more subjective endpoint than other endpoints such as a myocardial infarction, stroke, or cardiovascular death. A subjective endpoint has the potential to be an unreliable endpoint in an open label study and is a limitation of the JELIS Study. Unfortunately, we do not have information on elderly patients (≥ 75 years).

 

REDUCE-IT

 

REDUCE-IT was a randomized, double-blind trial of 2 grams twice per day of EPA ethyl ester (icosapent ethyl) (Vascepa) vs. mineral oil placebo in 8,179 patients with hypertriglyceridemia (135mg/dL to 499mg/dL) and established cardiovascular disease or high cardiovascular disease risk (diabetes plus one risk factor) who were on stable statin therapy (75). The primary end point was a composite of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, or unstable angina. At baseline, the median LDL-C level was 75.0 mg/dL, HDL-C level was 40.0 mg/dL, and TG level was 216.0 mg/dL. The median change in TG level from baseline to 1 year was a decrease of 18.3% (−39.0 mg/dL) in the EPA group and an increase of 2.2% (4.5 mg/dL) in the placebo group. After a median of 4.9 years the primary end-point occurred in 17.2% of the patients in the EPA group vs. 22.0% of the patients in the placebo group (HR 0.75; P<0.001), indicating a 25% decrease in events. In participants <65 years of age the primary end point was reduced by 35% (HR 0.65; 95% CI 0.54–0.78) while in participants ≥ 65 years of age the primary end point was reduced by 18% (HR 0.82; 95%CI 0.70–0.97; P Value for Interaction 0.06). The cardiovascular benefits of EPA were similar across baseline levels of TGs (<150, ≥150 to <200, and ≥200 mg per deciliter). Moreover, the cardiovascular benefits of EPA appeared to occur irrespective of the attained TG level at 1 year (≥150 or <150 mg/dL), suggesting that the ASCVD risk reduction was not associated with attainment of a normal TG level. Unfortunately, information on an elderly subgroup (≥ 75 years) is not available. 

 

It should be noted that in this trial mineral oil was used as the placebo. In the placebo group the LDL-C, non-HDL-C, and hsCRP levels were increased compared to the EPA group during the trial (LDL-C 96mg/dL vs 85mg/dL; non-HDL-C 130mg/dL vs. 113mg/dL; hsCRP 2.8mg/L vs. 1.8mg/L). The impact of these adverse changes on clinical outcomes is uncertain and whether they contributed to the apparent beneficial effects observed in the individuals treated with EPA is unknown.

 

STRENGTH TRIAL

 

The STRENGTH Trial was a double-blind, randomized, trial comparing 4 grams per day of a carboxylic acid formulation of omega-3 fatty acids (EPA and DHA; Epanova) (n = 6,539)) vs. corn oil placebo (n = 6539) in statin-treated participants with high cardiovascular risk, hypertriglyceridemia, and low levels of HDL-C (76). Approximately 55% of patients had established cardiovascular disease and approximately 70% had diabetes. Median LDL-C level was 75.0 mg/dL, median TG level was 240 mg/dL, and median HDL-C level was 36 mg/dL. There were minimal differences in the change in LDL-C and HDL-C levels between the treated and placebo groups after treatment for 12 months but as expected there was a greater reduction in TG levels in the group treated with omega-3-fatty acids (−19.0% vs −0.9%). The primary endpoint was a composite of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, or unstable angina requiring hospitalization which occurred in 12.0% of individuals treated with omega-3 CA vs. 12.2% treated with corn oil (HR 0.99; P = .84). There were no significant differences between the treatment groups with regard to the risk of the individual components of the primary end point over the 3-4 years of the study. Thus, in contrast to the JELIS and REDUCE-IT trials the STRENGTH trial did not demonstrate a benefit of treatment with a mixture of omega-3-fatty acids (EPA + DHA).

 

OMEMI TRIAL

 

The OMEMI trial was a randomized trial of 1.8 grams per day of omega-3-fatty acids (930 mg EPA and 660 mg DHA) (n= 505) vs. corn oil placebo (n= 509) in patients aged 70 to 82 years with a recent myocardial infarction (2-8 weeks) (77). Baseline LDL-C was approximately 76mg/dL, HDL-C was 49mg/dL, and TGs 110mg/dL. The primary endpoint was a composite of nonfatal myocardial infarction, unscheduled revascularization, stroke, all-cause death, and heart failure hospitalization after 2 years of follow-up. The primary endpoint occurred in 21.4% of patients on omega-3-fatty acids vs. 20.0% on placebo (HR 1.08; P=0.60). TGs levels decreased 8.1% in the omega-3-fatty acid group and increased 5.1% in the placebo group (between group difference 13.2%; P<0.001) while changes in LDL-C were minimal in both groups. Thus, similar to the STRENGTH trial no benefits on cardiovascular disease were observed with EPA + DHA treatment.

 

SUMMARY

 

  • High dose EPA (JELIS and REDUCE-IT) reduced ASCVD outcomes while high dose EPA+DHA (STENGTH and OMEMI) did not decrease ASCVD outcomes.
  • The decrease in TG levels is not a major contributor to the beneficial effect of high dose EPA as the combination of high dose EPA+DHA lowers TG levels to the same degree as EPA alone without benefit. Additionally, the JELIS trial only lowered TG levels by 5% but nevertheless reduced ASCVD events. It is likely that the beneficial effects of EPA seen in the JELIS and REDUCE-IT trials are multifactorial with TG lowering making only a small contribution to the decrease in cardiovascular disease. Other actions of EPA, such as decreasing platelet function, anti-inflammation, decreasing lipid oxidation, stabilizing membranes, etc. could account for or contribute to the reduction in ASCVD events (78).
  • Whether EPA has special properties that resulted in the reduction in ASCVD events in the REDUCE-IT trial or there were flaws in the trial design (i.e., the use of mineral oil as the placebo) is uncertain and debated. It should be noted that in the REDUCE-IT trial LDL-C and non-HDL-C levels were increased by approximately 10% in the mineral oil placebo group (75). Additionally, Apo B levels were increased by 7% (6mg/dL) by mineral oil (75). Finally, an increase in hsCRP (20-30%) and other biomarkers of atherosclerosis (oxidized LDL-C, IL-6, IL-1 beta, and lipoprotein-associated phospholipase A2) were noted in the mineral oil group (75,79). In the STRENGTH trial there were no differences in LDL-C, Non-HDL-C, HDL-C, Apo B, or hsCRP levels between the treated vs. placebo groups (76). Whether EPA has special properties compared to DHA leading to a reduction in cardiovascular events or the mineral oil placebo resulted in adverse changes increasing ASCVD in the placebo resulting in an artifactual decrease in the EPA group is debated (80,81). Ideally, another large randomized ASCVD trial with EPA ethyl ester (icosapent ethyl) (Vascepa) using a placebo other than mineral oil would help resolve this controversy. In the meantime, clinicians will need to use their clinical judgement on whether to treat patients with modest elevations in TG levels with EPA (icosapent ethyl; Vascepa) balancing the potential benefits of treatment vs. the potential side effects.

 

Summary of Lipid Lowering Drug Studies

 

The above results clearly demonstrate that lowering LDL-C levels with statins, ezetimibe, or PCSK 9 monoclonal antibodies will reduce ASCVD events in elderly patients (≥ 75 years of age) with pre-existing cardiovascular disease. In elderly patients without cardiovascular disease (primary prevention) the available data does not definitively demonstrate a decrease in ASCVD events with statin or ezetimibe therapy but is suggestive of a benefit (note there are no primary prevention trials with PCSK9 inhibitors). Additional data is required to determine if bempedoic acid and icosapent ethyl reduce ASCVD events in patients ≥ 75 years of age.

 

Studies in Progress

 

Studies are currently underway to provide definitive information on whether statin therapy is beneficial as primary prevention in the elderly. STAREE (NCT02099123) is a multicenter randomized trial in Australia of atorvastatin 40mg vs. placebo in adults ≥ 70 years of age without cardiovascular disease and PREVENTABLE (NCT04262206) is a multicenter randomized trial in the USA of atorvastatin vs. placebo in adults ≥ 75 years of age without cardiovascular disease (82,83). Other trials in the elderly that are in progress include SCOPE (NCT03770312) which is a multicenter randomized trial in Korea of low intensity vs. high intensity statin therapy in adults 76-85 years of age without CVD and SITE (Statins In The Elderly) (NCT02547883) which is a trial in France of patients ≥ 75 years of age on statin therapy who will be randomized to continue statin therapy or stop statin therapy.

 

SIDE EFFECTS OF LIPID LOWERING DRUGS

 

In this section we will describe the potential side effects of lipid lowering drugs with an emphasis on side effects likely to be seen in the elderly. Elderly patients may be more susceptible to side effects due to decreased renal function, decreased drug metabolism by the liver, polypharmacy leading to drug interactions, and co-morbidities. For a detailed discussion of the side effects of lipid lowering drugs see the Endotext chapters entitled “Cholesterol Lowering Drugs” and “Triglyceride Lowering Drugs” (49,50).

 

Statins

 

An umbrella review of meta-analyses of observational studies and randomized controlled trials examined 278 unique non-CVD outcomes from 112 meta-analyses of observational studies and 144 meta-analyses of RCTs and found that the only adverse effects associated with statin therapy were the development of diabetes and muscle disorders (84). For a detailed discussion of the side effects of statin therapy a scientific statement from the American Heart Association provides a comprehensive review (85).

 

DIABETES

 

After many years of statin use it was recognized that statins increase the risk of developing diabetes. In a meta-analysis of 13 trials with over 90,000 subjects, there was a 9% increase in the incidence of diabetes during follow-up among subjects receiving statin therapy (86). All statins appear to increase the risk of developing diabetes. In comparisons of intensive vs. moderate statin therapy, Preiss et al observed that patients treated with intensive statin therapy had a 12% greater risk of developing diabetes compared to subjects treated with moderate dose statin therapy (87). Older subjects, obese subjects, and subjects with high glucose levels were at a higher risk of developing diabetes while on statin therapy. In the Prosper trial in elderly subjects (70-82 years of age; average age 75), diabetes developed in 6.6% of patients treated with pravastatin vs. 5.1% of patients in the placebo group (51). Thus, statins may be unmasking and accelerating the development of diabetes that would have occurred naturally in these subjects at some point in time. In patients without risk factors for developing diabetes, treatment with statins does not appear to greatly increase the risk of developing diabetes.

 

In balancing the benefits and risks of statin therapy it is important to recognize that an increase in plasma glucose levels is a surrogate marker for an increased risk of developing micro and macrovascular disease (i.e., an increase in plasma glucose per se is not an event but rather increases the risk of future events). In contrast, statin therapy is preventing actual clinical events that cause morbidity and mortality. Furthermore, it may take many years for an elevated blood glucose to induce diabetic complications while the reduction in cardiovascular events with statin therapy occurs relatively quickly (after one-year benefits are seen). Finally, the number of patients needed to treat with statins to avoid one cardiovascular event is much lower (10-20 depending on the type of patient) than the number of patients needed to treat to cause one patient to develop diabetes (100–200 for one extra case of diabetes) (88). Patients on statin therapy, particularly those with risk factors for the development of diabetes, should be periodically screened for the development of diabetes with measurement of fasting glucose and/or HbA1c levels.

 

COGNITIVE DYSFUNCTION

 

Several randomized clinical trials have examined the effect of statin therapy on cognitive function and have not indicated any increased risk (for review see (89-92)). The Prosper Trial was designed to determine whether statin therapy will reduce cardiovascular disease in older subjects (age 70-82) (51). In this trial cognitive function was assessed repeatedly and no difference in cognitive decline was found in subjects treated with pravastatin compared to placebo (51,93). In the Heart Protection Study over 20,000 patients were randomized to simvastatin 40mg or placebo and again no significant differences in cognitive function was observed between the statin vs. placebo group (94). Additionally, a Cochrane review examined the effect of statin therapy in patients with established dementia and identified 4 studies with 1154 participants (95). In this analysis no benefit or harm of statin therapy on cognitive function could be demonstrated in this high-risk group of patients. Thus, randomized clinical trials do not indicate a significant association between statins and cognitive function.

 

MUSCLE

 

The most common side effect of statin therapy is muscle symptoms and many patients will discontinue the use of statins due to muscle symptoms. These can range from life threatening rhabdomyolysis, which is very rare, to myalgias, which are a common complaint (96). The risk of serious muscle disorders due to statin therapy is very small, particularly if one is aware of the potential drug interactions that increase the risk. In a case-control study with a cohort of 252,460 new users of lipid-lowering medications in U.S. health plans 21 cases of rhabdomyolysis were compared to 200 controls without rhabdomyolysis (97). Statin users >65 years of age had four times the risk of hospitalization for rhabdomyolysis than those under age 65.

The Cholesterol Treatment Trialists analyzed individual participant data on the development of muscle symptoms from 19 double-blind trials of statin versus placebo with 123,940 participants and four double-blind trials of a more intensive vs. a less intensive statin regimen with 30,724 participants (98). After a median follow-up of 4.3 years 27.1% of the individuals taking a statin vs. 26.6% on placebo reported muscle pain or weakness representing a 3% increase greater than placebo (risk ratio- 1.03; 95% CI 1.01-1.06) (Table 5). The specific muscle symptoms caused by statin therapy, myalgia, muscle cramps or spasm, limb pain, other musculoskeletal pain, or muscle fatigue or weakness were similar to those caused by placebo. The slight increase in muscle symptoms in the statin treated individuals was manifest in the first year of therapy but in the later years muscle symptoms were similar in the statin treated and placebo groups. The relative risk of statin induced muscle symptoms was greater in women than men. Intensive statin treatment with 40-80 mg atorvastatin or 20-40 mg rosuvastatin resulted in a higher risk of muscle symptoms than less intensive or moderate-intensity regimens but different statins at equivalent LDL-C lowering doses had similar effects on muscle symptoms. As shown in figure 7 muscle pain or weakness was slightly increased in patients > 65 years of age and similar in patients > 65 and ≤ 75 and those > 75 years of age. It should be noted that in individuals > 75 years of age the occurrence of muscle pain or weakness occurred in 39.6% of the individuals on the placebo, demonstrating the very high occurrence of muscle symptoms in this age group.

 

This study demonstrates that there is a small increase in muscle symptoms that primarily manifests in the first year of therapy. Statin therapy caused approximately 11 additional complaints of muscle pain or weakness per 1000 patients during the first year, but little excess in later years. Of particularly note is that 26.6% of patients taking a placebo had muscle symptoms demonstrating a very high frequency of this clinical complaint (even higher in patients > 75). Given the high prevalence of muscle complaints and the small increase attributed to statins it is very difficult to determine if a muscle complaint is actually due to the statin, which presents great clinical difficulties in patient management.

 

Table 5. Effect of Statin vs. Placebo on Muscle Symptoms

Symptom

Statin Events

Placebo Events

RR (95% CI)

Myalgia

12.0%

11.7%

1·03 (0·99–1·08)

Other musculoskeletal pain

13.3

13.0

1·03 (0·99–1·08)

Any muscle pain

26.9%

26.3%

1·03 (1·01–1·06)

Any muscle pain or weakness

27.1%

26.6%

1·03 (1·01–1·06)

 

Figure 7. Occurrence of muscle pain or weakness in different age groups in the Cholesterol Treatment Trialists meta-analysis.

 

While the results of the randomized trials suggest that muscle symptoms are not frequently induced by statin therapy, in typical clinical settings a significant percentage of patients are unable to tolerate statins due to muscle symptoms (in many studies as high as 5-25% of patients) (99-101). Additionally, when patients know that they are taking a statin they are more likely to have muscle symptoms (i.e. the nocebo effect) (102). Clinically differentiating statin induced myalgias from placebo induced myalgias is difficult, as there are no specific symptoms, signs, or biomarkers that clearly distinguish between the two. Thus, while statin induced myalgias are a real entity careful studies have shown that in the majority of patients with “statin induced muscle symptoms” the symptoms are not actually due to statin therapy. In the clinic it is difficult to be certain whether the muscle symptoms are actually due to true statin intolerance or to other factors.

 

A detailed discussion of statin induced muscle symptoms and a clinical approach to this problem is presented in the Endotext chapter entitled “Cholesterol Lowering Drugs” (50). In the section “Patients with Statin Intolerance” in this chapter we discuss the clinical approach to treating these patients.

 

Ezetimibe

 

Ezetimibe has not demonstrated significant side effects.

 

PCSK9 Monoclonal Antibodies

 

In a subgroup of patients from the FOURIER trial a prospective study of cognitive function (EBBINGHAUS Study) was carried out and no significant differences in cognitive function was observed over a median of 19 months in the PCSK9 treated vs. placebo group (103).

 

An issue of concern is whether lowering LDL-C to very low levels has the potential to cause toxicity. In a number of the PCSK9 studies a significant number of patients had LDL-C levels < 25mg/dL. For example, in the Odyssey long term study 37% of patients on alirocumab had two consecutive LDL-C levels below 25mg/dL and in the Osler long term study in patients treated with evolocumab 13% had values below 25mg/dL (104,105). In these short term PCSK9 studies, toxicity from very low LDL-C levels has not been observed. Additionally, in patients with Familial Hypobetalipoproteinemia LDL levels can be very low and these patients do not have any major disorders other than hepatic steatosis, which is not mechanistically due to low LDL-C levels (106). Similarly, there are rare individuals who are homozygous for loss of function mutations in the PCSK9 gene and they also do not appear to have major medical issues (107). Finally, in a number of statin trials and the IMPROVE-IT trial (statin + ezetimibe) there have been patients with very low LDL-C levels and an increased risk of side effects has not been consistently observed in those patients (108-111). Thus, with the limited data available there does not appear to be a major risk from markedly lowering LDL-C levels.

 

Bempedoic Acid

 

In clinical trials, 26% of bempedoic acid-treated patients with normal baseline uric acid values experienced hyperuricemia one or more times versus 9.5% in the placebo group (package insert). In the CLEAR Outcomes trial elevated uric acid levels occurred in 10.9% of the patients on bempedoic acid compared to 5.6% taking the placebo (69). Elevations in blood uric acid levels may lead to the development of gout. Gout was reported in 1.5% of patients treated with bempedoic acid vs. 0.4% of patients treated with placebo. The risk for gout attacks were higher in patients with a prior history of gout (11.2% for bempedoic acid treatment vs. 1.7% in the placebo group) (package insert). In patients with no prior history of gout only 1% of patients treated with bempedoic acid and 0.3% of the placebo group had a gouty attack (package insert). In the CLEAR Outcomes trial gout was increased in the bempedoic acid group (3.1% vs. 2.1%) (69).

 

In clinical trials tendon rupture occurred in 0.5% of patients treated with bempedoic acid vs. 0% of placebo treated patients and involved the rotator cuff (the shoulder), biceps tendon, or Achilles tendon (package insert). Tendon rupture occurred within weeks to months of starting bempedoic acid and occurred more frequently in patients over 60 years of age, in those taking corticosteroid or fluoroquinolone drugs, in patients with renal failure, and in patients with previous tendon disorders. In the CLEAR Outcomes trial tendon rupture was similar in the bempedoic acid and placebo group (bempedoic acid 1.2% and placebo 0.9%) (69).

 

Bempedoic acid treatment resulted in a mean increase in serum creatinine of 0.05 mg/dL compared to baseline. Approximately 3.8% of patients treated with bempedoic acid had BUN levels that doubled vs. 1.5% in the placebo group and about 2.2% of patients treated with bempedoic acid had creatinine values that increased by 0.5 mg/dL vs. 1.1% in the placebo group (package insert). Renal function returned to baseline when bempedoic acid was discontinued. In the CLEAR Outcomes trial renal impairment was increased in the bempedoic acid group (11.5% vs.8.6%) as was the change from baseline creatinine (0.05±0.2 mg/dL vs. 0.01±0.2 mg/dL) (69).

 

Omega-3-Fatty Acids

 

 At very high doses, omega-3-fatty acids can inhibit platelets and prolong bleeding time. However, at the recommended doses this has not been a major clinical problem but nevertheless when patients are on anti-platelet drugs one should be alert for the possibility of bleeding problems (Package Inserts for Lovaza, Vascepa, and Epanova). In the REDUCE-IT trial serious bleeding events occurred in 2.7% of the patients in the icosapent ethyl group and in 2.1% in the placebo group (P=0.06) (75). There were no fatal bleeding events in either group and the rates of hemorrhagic stroke, serious central nervous system bleeding, and serious gastrointestinal bleeding were not significantly higher in the EPA group than in the placebo group. In the STRENGHT trial any bleeding events and major bleeding events were similar in the omega-3 fatty acid group and placebo group (76).  A recent review found no evidence for discontinuing the use of omega-3 fatty acid treatment before invasive procedures or when given in combination with other agents that affect bleeding (112).

 

An increase in new-onset atrial fibrillation was observed in the REDUCE-IT trial in the patients treated with icosapent ethyl 4 grams/day (5.3% vs. 3.9%) and in the STRENGTH trial in the patients treated with omega-3-fatty acids (2.2% vs 1.3%)

 

CURRENT GUIDELINES AND LDL-C GOALS

 

This section discusses guidelines as they pertain to elderly patients.

 

2018 AHA/ACC/Multi-Society Report

 

The following summarizes the 2018 AHA/ACC guidelines (2).

 

PRIMARY PREVENTION

 

  • For individuals >75 years of age, randomized controlled trials of statin therapy do not provide strong evidence for benefit, so clinical assessment of risk status in a clinician–patient risk discussion is needed for deciding whether to continue or initiate statin treatment.
  • In individuals ≥ 75 years of age with an LDL-C level of 70 to 189mg/dL, initiating a moderate-intensity statin may be reasonable. Goal is to reduce LDL-C by 30-49% (note these guidelines recommend percent reduction rather than absolute LDL-C goals).
  • In individuals ≥ 75 years of age it may be reasonable to stop statin therapy when functional decline (physical or cognitive), multimorbidity, frailty, or reduced life-expectancy limits the potential benefits of statin therapy.
  • A shared decision-making process between clinicians and patients that individualizes decisions is indicated, with regular periodic reassessment.
  • Determining coronary artery calcium (CAC) score will help in determining which patients will benefit the most. For older adults with CAC scores of zero, the likelihood of benefits from statin therapy does not outweigh the risks. Limiting statin therapy to those with CAC scores greater than zero, combined with clinical judgment and patient preference, could provide a valuable awareness with which to inform shared decision-making.

 

SECONDARY PREVENTION

 

  • In patients ≥75 years of age with clinical ASCVD, it is reasonable to initiate moderate- or high-intensity statin therapy after evaluation of the potential for ASCVD risk reduction, adverse effects, and drug–drug interactions, as well as patient frailty and patient preferences. The goal of moderate statin therapy is to reduce LDL-C by 30-49% and the goal of high-intensity statin therapy is to reduce LDL-C by ≥ 50%. In very high-risk patients, a goal of an LDL-C < 70mg/dL and non-HDL-C < 100mg/dL is reasonable.
  • In patients ≥75 years of age who are tolerating high-intensity statin therapy, it is reasonable to continue high-intensity statin therapy after evaluation of the potential for ASCVD risk reduction, adverse effects, and drug-drug interactions, as well as patient frailty and patient preferences.

 

PATIENTS WITH DIABETES

 

  • In patients ≥ 75 years of age with diabetes mellitus and who are already on statin therapy, it is reasonable to continue statin therapy.
  • In patients ≥ 75 years of age with diabetes mellitus without cardiovascular disease it may be reasonable to start moderate statin therapy after a clinician-patient discussion of the potential benefits and risks of therapy. The goal is to decrease LDL-C by 30-49%.

 

2019 ESC/EAS Guidelines

 

The following summarizes the 2019 ESC/EAS guidelines (3).

 

  • Treatment with statins is recommended for older people with ASCVD in the same way as for younger patients.
  • Treatment with statins is recommended for primary prevention, according to the level of risk, in older people aged ≤ 75 years.
  • Initiation of statin treatment for primary prevention in older people aged >75 years may be considered, if at high-risk or above.
  • It is recommended that the statin is started at a low dose if there is significant renal impairment and/or the potential for drug interactions, and then titrated upwards to achieve LDL-C treatment goals.
  • In patients at very-high risk in primary or secondary prevention the goal is a 50% reduction in LDL-C and an LDL-C < 55mg/dL.
  • In patients at high risk in primary or secondary prevention the goal is a 50% reduction in LDL-C and an LDL-C < 70mg/dL.

The ESC/EAS criteria for very high risk and high risk are shown in table 6.

 

Table 6. ESC/EAS Criteria for Very-High Risk and High Risk for ASCVD Events

Very High Risk

Documented ASCVD or unequivocal on imaging

DM with target organ damage or at least three major risk factors, or early onset of T1DM of long duration (>20 years)

Severe CKD (eGFR <30 mL/min/1.73 m2)

A calculated SCORE ≥ 10% for 10-year risk of fatal CVD

Familial Hypercholesterolemia with ASCVD or with another major risk factor

High Risk

Markedly elevated single risk factors, in particular LDL-C >190 mg/dL or BP ≥ 180/110 mmHg

Patients with Familial Hypercholesterolemia without other major risk factors

Patients with DM without target organ damage, with DM duration ≥ 10 years, or another additional risk factor

Moderate CKD (eGFR 30-59 mL/min/1.73 m2)

A calculated SCORE ≥ 5% and <10% for 10-year risk of fatal CVD.

 

Our Approach

 

Our approach is based on concepts taken from both the ACC/AHA and ESC/EAS guidelines (i.e., we try to utilize the best ideas from each guideline). There are several general principles regarding lipid lowering therapy that should be considered in deciding who to treat (113).

 

  • The higher the LDL-C level the greater the benefit of lowering LDL-C.
  • The greater the decrease in LDL-C the greater the benefit.
  • The higher the absolute risk of ASCVD the greater the benefit of lowering LDL-C.

 

Additional factors that need to be considered, particularly in elderly patients, include

 

  • Life expectancy.
  • Competing non-cardiovascular disorders.
  • Risk of drug side effects.
  • Potential for drug interactions.
  • Patient preferences.

 

PRIMARY PREVENTION

 

Given the absence of definitive outcome trials demonstrating the benefit of decreasing LDL-C levels in patients ≥ 75 years of age without cardiovascular disease one must use clinical judgement in deciding who to treat. It should be recognized, as discussed in detail earlier, that the available evidence suggests that decreasing LDL-C will reduce ASCVD events in the elderly. Our approach is to determine ASCVD risk and then balance the risk with competing factors such as life expectancy, non-cardiovascular disorders, potential for drug interactions, and patient preferences. We use the approach described below to determine risk.

 

Step 1- Calculate the 10-year risk of an ASCVD event using the AHA/ACC pooled cohort equation. In Europe one can use the SCORE OP risk prediction algorithms (114). This will provide an estimate of the risk of the patient having an ASCVD event/death.

Step 2- To gain further insight on the risk of ASCVD one can determine if patient has any risk enhancing factors (tables 7 and 8). This can help further stratify the patient’s risk.

Step 3- If after discussion with the patient, you and/or the patient is uncertain on the level of risk and the appropriate treatment plan obtaining a coronary calcium score (CAC) can be helpful. A CAC score of zero indicates low risk for ASCVD and allows one to not start statin therapy (2). Note that a CAC score of zero in cigarette smokers, patients with diabetes mellitus, those with a strong family history of ASCVD, and possibly chronic inflammatory conditions such as HIV, may still be associated with a substantial 10-year risk (2).

 

Following these steps, we can estimate the risk for ASCVD events and in conjunction with the general principles described above discuss with the patient a treatment plan. If the decision is to treat our goal is often an LDL-C < 100mg/dL and non-HDL-C < 130mg/dL but in high-risk patients our goal may be an LDL-C < 70mg/dL and non-HDL-C < 100mg/dL.

 

Table 7. Risk-Enhancing Factors

 Family history of premature ASCVD (males, age <55 y; females, age <65 y)

 Primary hypercholesterolemia (LDL-C ≥160mg/dL; non-HDL-C ≥190mg/dL

 Metabolic syndrome (increased waist circumference, elevated triglycerides [>175 mg/dL], elevated blood pressure, elevated glucose, and low HDL-C [<40 mg/dL in men; <50 in women mg/dL] are factors; tally of 3 makes the diagnosis)

 Chronic kidney disease (eGFR 15–59 mL/min/1.73 m2 with or without albuminuria; not treated with dialysis or kidney transplantation)

 Chronic inflammatory conditions such as psoriasis, RA, or HIV/AIDS

 History of premature menopause (before age 40 y) and history of pregnancy-associated conditions that increase later ASCVD risk such as preeclampsia

 High-risk race/ethnicities (e.g., South Asian ancestry)

 Lipid/biomarkers: Associated with increased ASCVD risk

  Persistently* elevated, primary hypertriglyceridemia (≥175 mg/dL);

  If measured:

  1. Elevated high-sensitivity C-reactive protein (≥2.0 mg/L)

  2. Elevated Lp(a) ≥50 mg/dL or ≥125 nmol/L

  3. Elevated apoB ≥130 mg/dL

  4. ABI <0.9

 ABI= ankle-brachial index, RA= rheumatoid arthritis.

Modified from reference (2).

 

Table 8. Factors Modifying Systematic Coronary Risk Estimation Risks

Social deprivation: the origin of many of the causes of CVD.

Obesity and central obesity as measured by the body mass index and waist circumference, respectively.

Physical inactivity.

Psychosocial stress

Family history of premature CVD (men: <55 years and women: <60 years).

Chronic immune-mediated inflammatory disorder.

Major psychiatric disorders.

Treatment for HIV infection.

Atrial fibrillation.

Left ventricular hypertrophy.

Chronic kidney disease.

Obstructive sleep apnea syndrome.

Metabolic associated fatty liver disease.

Modified from reference (3).

 

PATIENTS WITH DIABETES

 

In patients ≥ 75 with diabetes without pre-existing cardiovascular our approach is very similar to that described for primary prevention. In addition to the risk enhancers listed in tables 7 and 8 there are specific diabetes risk enhancers that clinicians should factor in their decisions (table 9). Also, as noted above, in the presence of diabetes a zero CAC score is not as strong an indicator of low risk for ASCVD as in non-diabetics.

 

In patients with diabetes because they usually have multiple risk factors and are at high risk for ASCVD events our typical LDL-C goal is < 70mg/dL and non-HDL-C < 100mg/dL. In the rare situation where there are minimal other risk factors an LDL-C goal < 100mg/dL and non-HDL-C < 130mg/dL is reasonable. 

 

Table 9. Diabetes-Specific Risk Enhancers That Are Independent of Other Risk Factors

Long duration (≥10 years for type 2 diabetes or ≥20 years for type 1 diabetes)

Albuminuria ≥30 mcg of albumin/mg creatinine

eGFR <60 mL/min/1.73 m

Retinopathy

Neuropathy

ABI <0.9

ABI= ankle-brachial index.

Modified from reference (2).

 

SECONDARY PREVENTION

 

Studies have shown that lowering LDL-C levels with statins, ezetimibe, and PCSK9 monoclonal antibodies reduces ASCVD events in older adults with ASCVD. Thus, unless there are contraindications older patients with ASCVD should be treated with lipid lowering drugs to reduce ASCVD events. In elderly patients we will often employ a modest statin dose (for example atorvastatin 10-20mg or rosuvastatin 5-10mg) in combination with ezetimibe 10mg and then increase the statin dose, if necessary, based on lipid levels and the patient tolerating the treatment regimen. At a minimum our goal is an LDL-C < 70mg/dL and a non-HDL-C level < 100mg/dL but we would prefer lower values (ideally LDL-C < 55mg/dL and non-HDL-C < 85mg/dL) if they can be achieved with a statin + ezetimibe. In very high-risk patients (table 10) our goal is an LDL-C < 55mg/dL and non-HDL-C < 85mg/dL and adding a PCSK9 inhibitor may be required to achieve these levels in some patients.

 

Table 10. Criteria for Very High Risk

Very high-risk includes a history of multiple major ASCVD events or one major ASCVD event and multiple high-risk conditions.

 

Major ASCVD Events

 Recent ACS (within the past 12 months)

 History of MI (other than recent ACS event)

 History of ischemic stroke

 Symptomatic peripheral arterial disease (history of claudication with ABI <0.85, or previous revascularization or amputation)

 

High-Risk Conditions

 Age ≥65 y

 Heterozygous familial hypercholesterolemia

 History of prior coronary artery bypass surgery or percutaneous coronary intervention outside of the major ASCVD event(s)

 Diabetes mellitus

 Hypertension

 CKD (eGFR 15-59 mL/min/1.73 m2)

 Current smoking

 Persistently elevated LDL-C (LDL-C ≥100 mg/dL [≥2.6 mmol/L]) despite maximally tolerated statin therapy and ezetimibe

 History of congestive HF

ABI= ankle-brachial index; ACS= acute coronary syndrome.

Based on reference (2).

 

TREATMENT

 

Lifestyle

 

The lifestyle changes described below are recommended for all adults and are not specific for elderly individuals or for individuals with cardiovascular disease. The lifestyle changes recommended will lower lipid levels and are likely to reduce the risk of ASCVD.

 

EXERCISE

 

There is little debate that exercise is beneficial and that all individuals should be physically active. It is recommended that individuals participate in at least 150 minutes of moderate-intensity aerobic physical activity (for example 30 minutes 5 times per week) or 75 minutes per week of vigorous-intensity physical activity (115,116). Additionally, it is recommended that individuals participate in 2 days per week of muscle-strengthening activity (116). Because of the loss of muscle mass with aging it is very important to incorporate resistance training into the exercise program of elderly individuals.

 

A meta-analysis of exercise in the older individuals (>60 years of age) found that aerobic exercise decreased triglyceride and LDL-C levels and increased HDL-C levels while resistance exercise decreased LDL-C levels (117). Exercise also increases fitness and helps with weight loss. It should be noted that many elderly individuals may have substantial medical and social barriers to participating in exercise programs. Comorbidities, such as osteoarthritis, may limit exercise tolerance and make exercise challenging. Older individuals should be encouraged to be as active as possible.

 

DIET

 

For a detailed discussion of the effect of diet on lipids, lipoproteins and ASCVD see the Endotext chapter entitled “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels (118). There is general agreement on what constitutes a healthy diet and a brief summary of the Guidelines for Americans 2020-2025 is shown in table 11 and the guidelines from the American College of Cardiology/American Heart Association are shown in table 12.

 

Table 11. Guidelines for Americans 2020-2025

Recommend

Limit

Vegetables of all types—dark green; red and orange; beans, peas, and lentils; starchy; and other vegetables

Added sugars—Less than 10 percent of calories per day

Fruits, especially whole fruit

Saturated fat—Less than 10 percent of calories per day

Grains, at least half of which are whole grain

Sodium—Less than 2,300 milligrams per day

Dairy, including fat-free or low-fat milk, yogurt, and cheese, and/or lactose-free versions and fortified soy beverages and yogurt as alternatives

Alcoholic beverages—Adults can

choose not to drink or to drink in moderation by limiting intake to 2 drinks or less in a day for men and 1 drink or less in a day for women

Protein foods, including lean meats, poultry, and eggs; seafood; beans, peas, and lentils; and nuts, seeds, and soy products

 

Oils, including vegetable oils and oils in food, such as seafood and nuts

 

Full guideline is available at DietaryGuidelines.gov

 

Table 12. ACC/AHA Dietary Recommendations to Reduce Risk of ASCVD (115)

1. A diet emphasizing intake of vegetables, fruits, legumes, nuts, whole grains, and fish is recommended

2. Replacement of saturated fat with dietary monounsaturated and polyunsaturated fats can be beneficial

3. A diet containing reduced amounts of cholesterol and sodium can be beneficial

4. As a part of a healthy diet, it is reasonable to minimize the intake of processed meats, refined carbohydrates, and sweetened beverages

5. As a part of a healthy diet, the intake of trans fats should be avoided

 

A summary of the effect of individual dietary constituents on lipid and lipoprotein levels is shown in table 13 (118). This table summarizes the results of numerous randomized trials examining the effect of dietary manipulations on lipid and lipoprotein levels.

 

Table 13. Summary of the Effect of Dietary Constituents on Lipid and Lipoproteins

SFA

Increase LDL-C and modest increase HDL-C

MUFA and PUFA

Decrease LDL-C

TFA

Increase LDL-C and decrease HDL-C

Cholesterol

Increase LDL-C

Carbohydrates

Increase TGs, increase greater with simple sugars particularly fructose

Fiber

Decrease LDL-C

Phytosterols

Decrease LDL-C

SFA= saturated fatty acids, MUFA= monounsaturated fatty acids, PUFA= polyunsaturated fatty acids, TFA= trans fatty acids.

 

There is a huge literature describing the effect of diet on the risk of ASCVD and this literature is often conflicting and controversial (118). Several well recognized investigators have discussed the limitations of the information linking various diets and dietary constituents and the risk of disease (119,120). The major problem is that almost all of the information is based on observational studies and well conducted randomized trials measuring important ASCVD outcomes are very rare. Observational studies can demonstrate associations but do not definitively indicate that there is a cause-and-effect relationship. Unrecognized confounding variables can result in false associations.

 

Some of the more recent randomized dietary trials that have examined the effect of diet on ASCVD events are described below. For a discussion of other studies see reference (118). The PREDIMED trial employing a Mediterranean diet (increased monounsaturated fats) reduced the incidence of major ASCVD events (121). In this multicenter trial, carried out in Spain, over 7,000 patients at high risk for developing ASCVD were randomized to three diets (primary prevention trial). A Mediterranean diet supplemented with extra-virgin olive oil, a Mediterranean diet supplemented with mixed nuts, or a control diet. The average age of participants in this trial was 67. In the patients assigned to the Mediterranean diets there was 29% decrease in the primary end point (MI, stroke, and death from ASCVD). Subgroup analysis demonstrated that the Mediterranean diet was equally beneficial in patients < 70 and ≥ 70 years of age. The Mediterranean diet resulted in only a small but significant increase in HDL-C levels and a small decrease in both LDL-C and TG levels, suggesting that the beneficial effects were not mediated by changes in lipids (122). The CORDIOPREV study and the Lyon Diet Heart Study were randomized trials that demonstrated that a Mediterranean diet reduces ASCVD events in patients with cardiovascular disease (secondary prevention) (123,124). Unfortunately, these studies did not have a sufficient number of patients > 70 years of age for analysis of the effect of the diet in older patients with pre-existing cardiovascular disease.

 

The results of these three randomized trials indicate that following a Mediterranean type diet reduces ASCVD. It is likely that the beneficial effects of the Mediterranean diet on ASCVD is mediated by multiple mechanisms with alterations in lipid levels making only a minor contribution.

 

LIPID LOWERING DRUGS

 

Current guidelines and lipid lowering goals are discussed in the guidelines section above. In this section we will focus on clinical decisions regarding the use of lipid lowering drugs. To maximize benefits of lipid lowering therapy we think it is important to achieve LDL-C goals.

 

Elderly Patients on Lipid Lowering Therapy

 

In elderly patients on lipid lowering therapy, we usually continue therapy if they are tolerating the medications without side effects. We will periodically check a lipid panel to make sure that they are achieving the goals of therapy. If not, we will adjust the lipid lowering medications to achieve the desired LDL-C goals. We will make changes in therapy if circumstances change. For example, if a patient develops metastatic cancer and is transferred to palliative or Hospice care we will stop the lipid lowering therapy. Similarly, if a new drug is required the current lipid lowering drugs may need to be changed to avoid drug interactions. Thus, in most patients continuing lipid lowering therapy is appropriate.

 

Primary Prevention in Elderly Patients

 

In elderly patients we usually initiate therapy using moderate-intensity statin therapy if therapy is indicated as discussed above. We typically use either atorvastatin 10-20mg or rosuvastatin 5-10mg. Our reason for using these statins is that if one needs to lower LDL-C further we can just increase the dose of the statin and not need to start a new statin. In certain circumstances we might use another statin to avoid drug interactions (for example in a patient living with HIV we might use pitavastatin). After 6-12 weeks on statin therapy, we check a lipid panel and if the patient is having any medication side effects. If the LDL-C is not at goal we will either increase the statin dose or if we feel that the patient is at risk for statin toxicity add ezetimibe 10mg instead. In a healthy elderly patient at high risk for ASCVD (for example high LDL-C, diabetes, and hypertension) we do not hesitate to use high-intensity statin therapy (atorvastatin 40-80mg and rosuvastatin 20-40mg) plus ezetimibe 10mg to achieve the LDL-C goal.

 

Secondary Prevention in Elderly Patients

 

Unless than is a contraindication we frequently start these patients on high-intensity statin therapy. After 6-12 weeks on statin therapy, we check a lipid panel and if the patient is having any medication side effects. We will often add ezetimibe as studies have shown that the greater the lowering of LDL-C the greater the reductions in ASCVD events. Additionally, ezetimibe is generic (i.e. inexpensive) and doesn’t typically cause side effects. We will use PCSK9 inhibitors following the principle that the higher the LDL-C and the greater the risk of ASCVD events the greater the cost effectiveness of using expensive PCSK9 inhibitors.

 

Mixed Hyperlipidemia

 

In patients with mixed hyperlipidemia (elevated LDL-C and triglyceride levels) Initial drug therapy should also be a statin unless triglyceride levels are greater than 500-1000mg/dL. If triglycerides are > 500-1000mg/dL initial therapy is directed at lowering triglyceride levels (49,125). In addition to lowering LDL-C levels, statins are also effective in lowering triglyceride levels particularly when the triglycerides are elevated. If LDL-C is not lowered sufficiently ezetimibe is a reasonable next step. The approach to the patient whose LDL-C levels are at goal but the triglycerides and non-HDL-C are still elevated is not clearly defined. As discussed above studies have failed to demonstrate that adding a fibrate or niacin reduces ASCVD events. The REDUCE-IT trial has demonstrated that icosapent ethyl (Vascepa) decreases ASCVD events in this patient population but as discussed in detail above the results of this study are debated because the mineral oil placebo increased LDL-C. non-HDL-C, hsCRP, and other biomarkers associated with an increased risk of ASCVD events. It is debated by various experts whether the beneficial effect seen in this study was due to the positive effects of icosapent ethyl or to negative effects of the placebo. Clinicians will need to use their clinical judgement on whether to treat patients with elevations in TG and non-HDL-C levels with icosapent ethyl balancing the potential benefits of treatment vs. the potential side effects. In making this decision in our elderly patients it is worth noting that in participants <65 years of age the primary end point was reduced by 35% (HR 0.65; 95% CI 0.54–0.78) while in participants ≥ 65 years of age the primary end point was reduced by 18% (HR 0.82; 95%CI 0.70–0.97; P Value for Interaction 0.06). Information on patients ≥ 75 years of age is not available.  

 

Patients with Statin Intolerance

 

Statin intolerance is frequently due to myalgias but on occasion can be due other issues, such as increased liver or muscle enzymes, cognitive dysfunction, or other neurological disorders. The percentage of patients who are “statin intolerant” varies greatly but in clinical practice a significant number of patients have difficulty taking statins.

 

It can be difficult to determine if the muscle symptoms that occur when a patient is taking a statin are actually due to the statin or are unrelated to statin use. The first step in a “statin intolerant patient” is to take a careful history of the nature and location of the muscle symptoms and the timing of onset in relation to statin use to determine whether the presentation fits the typical picture for statin induced myalgias. The characteristic findings with a statin induced myalgia are shown in table 14 and findings that are not typical for statin induced myalgia are shown in table 15. The disappearance of symptoms within a few weeks of stopping statins and the reappearance after restarting statins is very suggestive of the symptoms being due to true statin intolerance. An on-line tool (htpp://tools.acc.org/statinintolerance/#!/) and an app produced by the ACC/AHA are available. This tool characterizes patients based on 8 criteria into possible vs. unlikely to have statin induced muscle symptoms (table 16).

 

Table 14. Characteristic Findings with Statin Induced Myalgia

Symmetric

Proximal muscles

Muscle pain, tenderness, weakness, cramps

Symptom onset < 4 weeks after starting statin or dose increase

Improves within 2-4 weeks of stopping statin

Cramping is unilateral and involves small muscles of hands and feet

Same symptoms occur with re-challenge within 4 weeks

 

Table 15. Symptoms Atypical in Statin Induced Myalgia

Unilateral

Asymmetric

Small muscles

Joint or tendon pain

Shooting pain, muscle twitching or tingling

Symptom onset > 12 weeks

No improvement after discontinuing statin

 

Table 16. Diagnosis of Statin Associated Muscle Symptoms

Symptom timing

Symptom type

Symptom location

Sex

Age

Race/ethnicity

CK elevation > 5 times the upper limit of normal

Known risk factors for statin induced muscle symptoms and non-statin causes of muscle symptoms

 

One should also check a CK level but this is almost always in the normal range. If the CK is not elevated and the symptoms do not suggest a statin induced myalgia one can often reassure the patient and continue statin therapy. This is often successful and studies have shown that many patients that stop taking statins due to “statin induced myalgia” can be successfully treated with a statin (50). If the CK is elevated it should be repeated after instructing the patient to avoid exercise for 48 hours. Also, the CK levels should be compared to CK levels prior to starting therapy if available. If the CK remains elevated (3x upper limit of normal) the statin should be discontinued. Similarly, if the CK is normal but the symptoms are suggestive of a statin induced myalgia the statin should also be discontinued. The next step is to determine if one can identify reversible factors that could be increasing statin toxicity (hypothyroidism, drug interactions).  If none are identified the next step after the myalgias have resolved is to try a low dose of a different statin that is metabolized by a different pathway (for example instead of atorvastatin, which is metabolized by the CYP3A4 pathway, rosuvastatin, which has a different pathway of metabolism). Because statin side effects are dose related, a low dose of a statin may often be tolerated. One can also try several different statins as sometimes a patient may tolerate one statin and not others. A meta-analysis has shown that every other day administration of statins is as effective as daily administration in lowering lipid levels and therefore is a very reasonable strategy (126). In some instances, using a long-acting statin (rosuvastatin or atorvastatin) 1-3 times per week can work (we usually start with once per week and then slowly increase frequency as tolerated) (127). In these circumstances (low doses or 1-3 times per week) the reduction in LDL-C may not be sufficient but one can use combination therapy with other drugs such as ezetimibe, bempedoic acid, or PCSK9 inhibitors to achieve LDL-C target goals.

 

If after trying various approaches a patient still has myalgias and is unable to tolerate statin therapy one needs to utilize other approaches to lower LDL-C levels. We typically use the ezetimibe and bempedoic acid combination pill (Nexlizet), which can lower the LDL-C level by approximately 40%, which is often sufficient (128). If needed one could add a PCSK9 inhibitor to further decrease LDL-C.

 

CONCLUSION

 

ASCVD is a major cause of morbidity and mortality in elderly patients. In elderly patients with pre-existing ASCVD randomized clinical trials have shown that lipid lowering drug therapy with statins, ezetimibe, and PCSK9 inhibitors reduce ASCVD events. Thus, most elderly patients with ASCVD should be treated with lipid lowering drugs unless there are contraindications such as limited life expectancy, competing non-cardiovascular disorders, high risk of drug interactions or drug side effects. In elderly patients without ASCVD if they are already taking lipid lowering drugs and if they are tolerating the medications without side effects continuing therapy is usually reasonable as long as the clinical circumstances have not changed. In elderly patients not on lipid lowering therapy and without cardiovascular disease studies have suggested that lipid lowering therapy is beneficial but further studies are required to definitively demonstrate benefit. In these patients one needs to determine the patient’s risk for ASCVD events and then discuss the potential benefits and side effects with the patient to make a shared decision on whether to initiate therapy. Age per se should not be used to withhold therapy with lipid lowering drugs that can reduce the risk of ASCVD events.

 

ACKNOWLEDGEMENTS

 

This work was supported by grants from the Northern California Institute for Research and Education. The authors would like to thank Dan Streja, the original author of this chapter, who provided the framework for this updated chapter.

 

REFERENCES

 

  1. Savji N, Rockman CB, Skolnick AH, Guo Y, Adelman MA, Riles T, Berger JS. Association between advanced age and vascular disease in different arterial territories: a population database of over 3.6 million subjects. J Am Coll Cardiol 2013; 61:1736-1743
  2. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Jr., Sperling L, Virani SS, Yeboah J. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 139:e1082-e1143
  3. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  4. Ghandehari H, Kamal-Bahl S, Wong ND. Prevalence and extent of dyslipidemia and recommended lipid levels in US adults with and without cardiovascular comorbidities: the National Health and Nutrition Examination Survey 2003-2004. Am Heart J 2008; 156:112-119
  5. Laurenzi M, Mancini M. Plasma lipids in elderly men and women. Eur Heart J 1988; 9 Suppl D:69-74
  6. Weijenberg MP, Feskens EJ, Kromhout D. Age-related changes in total and high-density-lipoprotein cholesterol in elderly Dutch men. Am J Public Health 1996; 86:798-803
  7. Abbott RD, Sharp DS, Burchfiel CM, Curb JD, Rodriguez BL, Hakim AA, Yano K. Cross-sectional and longitudinal changes in total and high-density-lipoprotein cholesterol levels over a 20-year period in elderly men: the Honolulu Heart Program. Ann Epidemiol 1997; 7:417-424
  8. Grunenberger F, Lammi-Keefe CJ, Schlienger JL, Deslypere JP, Hautvast JG. Longitudinal changes in serum lipids of elderly Europeans. SENECA Investigators. Eur J Clin Nutr 1996; 50 Suppl 2:S25-31
  9. Newschaffer CJ, Bush TL, Hale WE. Aging and total cholesterol levels: cohort, period, and survivorship effects. Am J Epidemiol 1992; 136:23-34
  10. Ettinger WH, Wahl PW, Kuller LH, Bush TL, Tracy RP, Manolio TA, Borhani NO, Wong ND, O'Leary DH. Lipoprotein lipids in older people. Results from the Cardiovascular Health Study. The CHS Collaborative Research Group. Circulation 1992; 86:858-869
  11. Curb JD, Reed DM, Yano K, Kautz JA, Albers JJ. Plasma lipids and lipoproteins in elderly Japanese-American men. J Am Geriatr Soc 1986; 34:773-780
  12. Wilson PW, Anderson KM, Harris T, Kannel WB, Castelli WP. Determinants of change in total cholesterol and HDL-C with age: the Framingham Study. J Gerontol 1994; 49:M252-257
  13. Garry PJ, Hunt WC, Koehler KM, VanderJagt DJ, Vellas BJ. Longitudinal study of dietary intakes and plasma lipids in healthy elderly men and women. Am J Clin Nutr 1992; 55:682-688
  14. Ferrara A, Barrett-Connor E, Shan J. Total, LDL, and HDL cholesterol decrease with age in older men and women. The Rancho Bernardo Study 1984-1994. Circulation 1997; 96:37-43
  15. Katsanos CS. Clinical considerations and mechanistic determinants of postprandial lipemia in older adults. Adv Nutr 2014; 5:226-234
  16. Spitler KM, Davies BSJ. Aging and plasma triglyceride metabolism. J Lipid Res 2020; 61:1161-1167
  17. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  18. Zuliani G, Romagnoni F, Bollini C, Leoci V, Soattin L, Fellin R. Low levels of high-density lipoprotein cholesterol are a marker of disability in the elderly. Gerontology 1999; 45:317-322
  19. Ranieri P, Rozzini R, Franzoni S, Barbisoni P, Trabucchi M. Serum cholesterol levels as a measure of frailty in elderly patients. Exp Aging Res 1998; 24:169-179
  20. Pel-Littel RE, Schuurmans MJ, Emmelot-Vonk MH, Verhaar HJ. Frailty: defining and measuring of a concept. J Nutr Health Aging 2009; 13:390-394
  21. Wilson DP. Is Atherosclerosis a Pediatric Disease? In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  22. Berenson GS, Srinivasan SR, Bao W, Newman WP, 3rd, Tracy RE, Wattigney WA. Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. The Bogalusa Heart Study. N Engl J Med 1998; 338:1650-1656
  23. McGill HC, Jr., McMahan CA, Malcom GT, Oalmann MC, Strong JP. Effects of serum lipoproteins and smoking on atherosclerosis in young men and women. The PDAY Research Group. Pathobiological Determinants of Atherosclerosis in Youth. Arterioscler Thromb Vasc Biol 1997; 17:95-106
  24. Tuzcu EM, Kapadia SR, Tutar E, Ziada KM, Hobbs RE, McCarthy PM, Young JB, Nissen SE. High prevalence of coronary atherosclerosis in asymptomatic teenagers and young adults: evidence from intravascular ultrasound. Circulation 2001; 103:2705-2710
  25. Enos WF, Holmes RH, Beyer J. Coronary disease among United States soldiers killed in action in Korea; preliminary report. J Am Med Assoc 1953; 152:1090-1093
  26. McNamara JJ, Molot MA, Stremple JF, Cutting RT. Coronary artery disease in combat casualties in Vietnam. JAMA 1971; 216:1185-1187
  27. Webber BJ, Seguin PG, Burnett DG, Clark LL, Otto JL. Prevalence of and risk factors for autopsy-determined atherosclerosis among US service members, 2001-2011. JAMA 2012; 308:2577-2583
  28. Newman WP, 3rd, Freedman DS, Voors AW, Gard PD, Srinivasan SR, Cresanta JL, Williamson GD, Webber LS, Berenson GS. Relation of serum lipoprotein levels and systolic blood pressure to early atherosclerosis. The Bogalusa Heart Study. N Engl J Med 1986; 314:138-144
  29. McGill HC, Jr., McMahan CA. Determinants of atherosclerosis in the young. Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group. Am J Cardiol 1998; 82:30T-36T
  30. Raitakari OT, Juonala M, Kahonen M, Taittonen L, Laitinen T, Maki-Torkko N, Jarvisalo MJ, Uhari M, Jokinen E, Ronnemaa T, Akerblom HK, Viikari JS. Cardiovascular risk factors in childhood and carotid artery intima-media thickness in adulthood: the Cardiovascular Risk in Young Finns Study. JAMA 2003; 290:2277-2283
  31. Li S, Chen W, Srinivasan SR, Bond MG, Tang R, Urbina EM, Berenson GS. Childhood cardiovascular risk factors and carotid vascular changes in adulthood: the Bogalusa Heart Study. JAMA 2003; 290:2271-2276
  32. Davis PH, Dawson JD, Riley WA, Lauer RM. Carotid intimal-medial thickness is related to cardiovascular risk factors measured from childhood through middle age: The Muscatine Study. Circulation 2001; 104:2815-2819
  33. Juonala M, Magnussen CG, Venn A, Dwyer T, Burns TL, Davis PH, Chen W, Srinivasan SR, Daniels SR, Kahonen M, Laitinen T, Taittonen L, Berenson GS, Viikari JS, Raitakari OT. Influence of age on associations between childhood risk factors and carotid intima-media thickness in adulthood: the Cardiovascular Risk in Young Finns Study, the Childhood Determinants of Adult Health Study, the Bogalusa Heart Study, and the Muscatine Study for the International Childhood Cardiovascular Cohort (i3C) Consortium. Circulation 2010; 122:2514-2520
  34. Magnussen CG, Venn A, Thomson R, Juonala M, Srinivasan SR, Viikari JS, Berenson GS, Dwyer T, Raitakari OT. The association of pediatric low- and high-density lipoprotein cholesterol dyslipidemia classifications and change in dyslipidemia status with carotid intima-media thickness in adulthood evidence from the cardiovascular risk in Young Finns study, the Bogalusa Heart study, and the CDAH (Childhood Determinants of Adult Health) study. J Am Coll Cardiol 2009; 53:860-869
  35. Klag MJ, Ford DE, Mead LA, He J, Whelton PK, Liang KY, Levine DM. Serum cholesterol in young men and subsequent cardiovascular disease. N Engl J Med 1993; 328:313-318
  36. Gray L, Lee IM, Sesso HD, Batty GD. Blood pressure in early adulthood, hypertension in middle age, and future cardiovascular disease mortality: HAHS (Harvard Alumni Health Study). J Am Coll Cardiol 2011; 58:2396-2403
  37. Pletcher MJ, Vittinghoff E, Thanataveerat A, Bibbins-Domingo K, Moran AE. Young Adult Exposure to Cardiovascular Risk Factors and Risk of Events Later in Life: The Framingham Offspring Study. PLoS One 2016; 11:e0154288
  38. Stamler J, Daviglus ML, Garside DB, Dyer AR, Greenland P, Neaton JD. Relationship of baseline serum cholesterol levels in 3 large cohorts of younger men to long-term coronary, cardiovascular, and all-cause mortality and to longevity. JAMA 2000; 284:311-318
  39. Warden BA, Fazio S, Shapiro MD. Familial Hypercholesterolemia: Genes and Beyond. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  40. Ference BA, Ray KK, Catapano AL, Ference TB, Burgess S, Neff DR, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Kastelein JJP, Nicholls SJ. Mendelian Randomization Study of ACLY and Cardiovascular Disease. N Engl J Med 2019; 380:1033-1042
  41. Horton JD, Cohen JC, Hobbs HH. PCSK9: a convertase that coordinates LDL catabolism. J Lipid Res 2009; 50 Suppl:S172-177
  42. Lubin JH, Couper D, Lutsey PL, Woodward M, Yatsuya H, Huxley RR. Risk of Cardiovascular Disease from Cumulative Cigarette Use and the Impact of Smoking Intensity. Epidemiology 2016; 27:395-404
  43. Lubin JH, Albanes D, Hoppin JA, Chen H, Lerro CC, Weinstein SJ, Sandler DP, Beane Freeman LE. Greater Coronary Heart Disease Risk With Lower Intensity and Longer Duration Smoking Compared With Higher Intensity and Shorter Duration Smoking: Congruent Results Across Diverse Cohorts. Nicotine Tob Res 2017; 19:817-825
  44. Ding N, Sang Y, Chen J, Ballew SH, Kalbaugh CA, Salameh MJ, Blaha MJ, Allison M, Heiss G, Selvin E, Coresh J, Matsushita K. Cigarette Smoking, Smoking Cessation, and Long-Term Risk of 3 Major Atherosclerotic Diseases. J Am Coll Cardiol 2019; 74:498-507
  45. Morton JI, Lazzarini PA, Polkinghorne KR, Carstensen B, Magliano DJ, Shaw JE. The association of attained age, age at diagnosis, and duration of type 2 diabetes with the long-term risk for major diabetes-related complications. Diabetes Res Clin Pract 2022; 190:110022
  46. de Jong M, Woodward M, Peters SAE. Duration of diabetes and the risk of major cardiovascular events in women and men: A prospective cohort study of UK Biobank participants. Diabetes Res Clin Pract 2022; 188:109899
  47. Feingold KR. Role of Glucose and Lipids in the Atherosclerotic Cardiovascular Disease in Patients with Diabetes. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  48. Zoungas S, Woodward M, Li Q, Cooper ME, Hamet P, Harrap S, Heller S, Marre M, Patel A, Poulter N, Williams B, Chalmers J. Impact of age, age at diagnosis and duration of diabetes on the risk of macrovascular and microvascular complications and death in type 2 diabetes. Diabetologia 2014; 57:2465-2474
  49. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  50. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  51. Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM, Cobbe SM, Ford I, Gaw A, Hyland M, Jukema JW, Kamper AM, Macfarlane PW, Meinders AE, Norrie J, Packard CJ, Perry IJ, Stott DJ, Sweeney BJ, Twomey C, Westendorp RG. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623-1630
  52. Cholesterol Treatment Trialists Colloboration. Efficacy and safety of statin therapy in older people: a meta-analysis of individual participant data from 28 randomised controlled trials. Lancet 2019; 393:407-415
  53. Amarenco P, Kim JS, Labreuche J, Charles H, Abtan J, Bejot Y, Cabrejo L, Cha JK, Ducrocq G, Giroud M, Guidoux C, Hobeanu C, Kim YJ, Lapergue B, Lavallee PC, Lee BC, Lee KB, Leys D, Mahagne MH, Meseguer E, Nighoghossian N, Pico F, Samson Y, Sibon I, Steg PG, Sung SM, Touboul PJ, Touze E, Varenne O, Vicaut E, Yelles N, Bruckert E. A Comparison of Two LDL Cholesterol Targets after Ischemic Stroke. N Engl J Med 2020; 382:9
  54. Andersson NW, Corn G, Dohlmann TL, Melbye M, Wohlfahrt J, Lund M. LDL-C Reduction With Lipid-Lowering Therapy for Primary Prevention of Major Vascular Events Among Older Individuals. J Am Coll Cardiol 2023; 82:1381-1391
  55. Orkaby AR, Driver JA, Ho YL, Lu B, Costa L, Honerlaw J, Galloway A, Vassy JL, Forman DE, Gaziano JM, Gagnon DR, Wilson PWF, Cho K, Djousse L. Association of Statin Use With All-Cause and Cardiovascular Mortality in US Veterans 75 Years and Older. JAMA 2020; 324:68-78
  56. Kim K, Lee CJ, Shim CY, Kim JS, Kim BK, Park S, Chang HJ, Hong GR, Ko YG, Kang SM, Choi D, Ha JW, Hong MK, Jang Y, Lee SH. Statin and clinical outcomes of primary prevention in individuals aged >75 years: The SCOPE-75 study. Atherosclerosis 2019; 284:31-36
  57. Bergami M, Cenko E, Yoon J, Mendieta G, Kedev S, Zdravkovic M, Vasiljevic Z, Milicic D, Manfrini O, van der Schaar M, Gale CP, Badimon L, Bugiardini R. Statins for primary prevention among elderly men and women. Cardiovasc Res 2022; 118:3000-3009
  58. Lavie G, Hoshen M, Leibowitz M, Benis A, Akriv A, Balicer R, Reges O. Statin Therapy for Primary Prevention in the Elderly and Its Association with New-Onset Diabetes, Cardiovascular Events, and All-Cause Mortality. Am J Med 2021; 134:643-652
  59. O'Sullivan JL, Kohl R, Lech S, Romanescu L, Schuster J, Kuhlmey A, Gellert P, Yasar S. Statin Use and All-Cause Mortality in Nursing Home Residents With and Without Dementia: A Retrospective Cohort Study Using Claims Data. Neurology 2024; 102:e209189
  60. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, Darius H, Lewis BS, Ophuis TO, Jukema JW, De Ferrari GM, Ruzyllo W, De Lucca P, Im K, Bohula EA, Reist C, Wiviott SD, Tershakovec AM, Musliner TA, Braunwald E, Califf RM. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387-2397
  61. Ouchi Y, Sasaki J, Arai H, Yokote K, Harada K, Katayama Y, Urabe T, Uchida Y, Hayashi M, Yokota N, Nishida H, Otonari T, Arai T, Sakuma I, Sakabe K, Yamamoto M, Kobayashi T, Oikawa S, Yamashita S, Rakugi H, Imai T, Tanaka S, Ohashi Y, Kuwabara M, Ito H. Ezetimibe Lipid-Lowering Trial on Prevention of Atherosclerotic Cardiovascular Disease in 75 or Older (EWTOPIA 75): A Randomized, Controlled Trial. Circulation 2019; 140:992-1003
  62. Kim BK, Hong SJ, Lee YJ, Hong SJ, Yun KH, Hong BK, Heo JH, Rha SW, Cho YH, Lee SJ, Ahn CM, Kim JS, Ko YG, Choi D, Jang Y, Hong MK. Long-term efficacy and safety of moderate-intensity statin with ezetimibe combination therapy versus high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease (RACING): a randomised, open-label, non-inferiority trial. Lancet 2022; 400:380-390
  63. Lee B, Hong SJ, Rha SW, Heo JH, Hur SH, Choi HH, Kim KJ, Kim JH, Kim HK, Kim U, Choi YJ, Lee YJ, Lee SJ, Ahn CM, Ko YG, Kim BK, Choi D, Hong MK, Jang Y, Kim JS. Moderate-intensity statin plus ezetimibe vs high-intensity statin according to baseline LDL-C in the treatment of atherosclerotic cardiovascular disease: A post-hoc analysis of the RACING randomized trial. Atherosclerosis 2023; 386:117373
  64. Lee SH, Lee YJ, Heo JH, Hur SH, Choi HH, Kim KJ, Kim JH, Park KH, Lee JH, Choi YJ, Lee SJ, Hong SJ, Ahn CM, Kim BK, Ko YG, Choi D, Hong MK, Jang Y, Kim JS. Combination Moderate-Intensity Statin and Ezetimibe Therapy for Elderly Patients With Atherosclerosis. J Am Coll Cardiol 2023; 81:1339-1349
  65. Sabatine MS, Giugliano RP, Wiviott SD, Raal FJ, Blom DJ, Robinson J, Ballantyne CM, Somaratne R, Legg J, Wasserman SM, Scott R, Koren MJ, Stein EA. Efficacy and safety of evolocumab in reducing lipids and cardiovascular events. N Engl J Med 2015; 372:1500-1509
  66. Sever P, Gouni-Berthold I, Keech A, Giugliano R, Pedersen TR, Im K, Wang H, Knusel B, Sabatine MS, O'Donoghue ML. LDL-cholesterol lowering with evolocumab, and outcomes according to age and sex in patients in the FOURIER Trial. Eur J Prev Cardiol 2021; 28:805-812
  67. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R, Quintero K, Roe MT, Sasiela WJ, Tamby JF, Tricoci P, White HD, Zeiher AM. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med 2018; 379:2097-2107
  68. Sinnaeve PR, Schwartz GG, Wojdyla DM, Alings M, Bhatt DL, Bittner VA, Chiang CE, Correa Flores RM, Diaz R, Dorobantu M, Goodman SG, Jukema JW, Kim YU, Pordy R, Roe MT, Sy RG, Szarek M, White HD, Zeiher AM, Steg PG. Effect of alirocumab on cardiovascular outcomes after acute coronary syndromes according to age: an ODYSSEY OUTCOMES trial analysis. Eur Heart J 2020; 41:2248-2258
  69. Nissen SE, Lincoff AM, Brennan D, Ray KK, Mason D, Kastelein JJP, Thompson PD, Libby P, Cho L, Plutzky J, Bays HE, Moriarty PM, Menon V, Grobbee DE, Louie MJ, Chen CF, Li N, Bloedon L, Robinson P, Horner M, Sasiela WJ, McCluskey J, Davey D, Fajardo-Campos P, Petrovic P, Fedacko J, Zmuda W, Lukyanov Y, Nicholls SJ. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N Engl J Med 2023; 388:1353-1364
  70. AIM-HIGH Investigators, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, McBride R, Teo K, Weintraub W. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011; 365:2255-2267
  71. Hps Thrive Collaborative Group, Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, Tomson J, Wallendszus K, Craig M, Jiang L, Collins R, Armitage J. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 2014; 371:203-212
  72. ACCORD Study Group, Ginsberg HN, Elam MB, Lovato LC, Crouse JR, 3rd, Leiter LA, Linz P, Friedewald WT, Buse JB, Gerstein HC, Probstfield J, Grimm RH, Ismail-Beigi F, Bigger JT, Goff DC, Jr., Cushman WC, Simons-Morton DG, Byington RP. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010; 362:1563-1574
  73. Das Pradhan A, Glynn RJ, Fruchart JC, MacFadyen JG, Zaharris ES, Everett BM, Campbell SE, Oshima R, Amarenco P, Blom DJ, Brinton EA, Eckel RH, Elam MB, Felicio JS, Ginsberg HN, Goudev A, Ishibashi S, Joseph J, Kodama T, Koenig W, Leiter LA, Lorenzatti AJ, Mankovsky B, Marx N, Nordestgaard BG, Pall D, Ray KK, Santos RD, Soran H, Susekov A, Tendera M, Yokote K, Paynter NP, Buring JE, Libby P, Ridker PM. Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N Engl J Med 2022; 387:1923-1934
  74. Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Saito Y, Ishikawa Y, Oikawa S, Sasaki J, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata T, Shimada K, Shirato K. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet 2007; 369:1090-1098
  75. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT, Jr., Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N Engl J Med 2019; 380:11-22
  76. Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Ridker PM, Ray KK, Katona BG, Himmelmann A, Loss LE, Rensfeldt M, Lundstrom T, Agrawal R, Menon V, Wolski K, Nissen SE. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA 2020; 324:2268-2280
  77. Kalstad AA, Myhre PL, Laake K, Tveit SH, Schmidt EB, Smith P, Nilsen DWT, Tveit A, Fagerland MW, Solheim S, Seljeflot I, Arnesen H. Effects of n-3 Fatty Acid Supplements in Elderly Patients After Myocardial Infarction: A Randomized, Controlled Trial. Circulation 2021; 143:528-539
  78. Mason RP, Libby P, Bhatt DL. Emerging Mechanisms of Cardiovascular Protection for the Omega-3 Fatty Acid Eicosapentaenoic Acid. Arterioscler Thromb Vasc Biol 2020; 40:1135-1147
  79. Ridker PM, Rifai N, MacFadyen J, Glynn RJ, Jiao L, Steg PG, Miller M, Brinton EA, Jacobson TA, Tardif JC, Ballantyne CM, Mason RP, Bhatt DL. Effects of Randomized Treatment With Icosapent Ethyl and a Mineral Oil Comparator on Interleukin-1beta, Interleukin-6, C-Reactive Protein, Oxidized Low-Density Lipoprotein Cholesterol, Homocysteine, Lipoprotein(a), and Lipoprotein-Associated Phospholipase A2: A REDUCE-IT Biomarker Substudy. Circulation 2022; 146:372-379
  80. Goff ZD, Nissen SE. N-3 polyunsaturated fatty acids for cardiovascular risk. Curr Opin Cardiol 2022; 37:356-363
  81. Mason RP, Sherratt SCR, Eckel RH. Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab 2023; 37:101681
  82. Joseph J, Pajewski NM, Dolor RJ, Sellers MA, Perdue LH, Peeples SR, Henrie AM, Woolard N, Jones WS, Benziger CP, Orkaby AR, Mixon AS, VanWormer JJ, Shapiro MD, Kistler CE, Polonsky TS, Chatterjee R, Chamberlain AM, Forman DE, Knowlton KU, Gill TM, Newby LK, Hammill BG, Cicek MS, Williams NA, Decker JE, Ou J, Rubinstein J, Choudhary G, Gazmuri RJ, Schmader KE, Roumie CL, Vaughan CP, Effron MB, Cooper-DeHoff RM, Supiano MA, Shah RC, Whittle JC, Hernandez AF, Ambrosius WT, Williamson JD, Alexander KP. Pragmatic evaluation of events and benefits of lipid lowering in older adults (PREVENTABLE): Trial design and rationale. J Am Geriatr Soc 2023; 71:1701-1713
  83. Zoungas S, Curtis A, Spark S, Wolfe R, McNeil JJ, Beilin L, Chong TT, Cloud G, Hopper I, Kost A, Nelson M, Nicholls SJ, Reid CM, Ryan J, Tonkin A, Ward SA, Wierzbicki A. Statins for extension of disability-free survival and primary prevention of cardiovascular events among older people: protocol for a randomised controlled trial in primary care (STAREE trial). BMJ Open 2023; 13:e069915
  84. He Y, Li X, Gasevic D, Brunt E, McLachlan F, Millenson M, Timofeeva M, Ioannidis JPA, Campbell H, Theodoratou E. Statins and Multiple Noncardiovascular Outcomes: Umbrella Review of Meta-analyses of Observational Studies and Randomized Controlled Trials. Ann Intern Med 2018; 169:543-553
  85. Newman CB, Preiss D, Tobert JA, Jacobson TA, Page RL, 2nd, Goldstein LB, Chin C, Tannock LR, Miller M, Raghuveer G, Duell PB, Brinton EA, Pollak A, Braun LT, Welty FK. Statin Safety and Associated Adverse Events: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2019; 39:e38-e81
  86. Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, Seshasai SR, McMurray JJ, Freeman DJ, Jukema JW, Macfarlane PW, Packard CJ, Stott DJ, Westendorp RG, Shepherd J, Davis BR, Pressel SL, Marchioli R, Marfisi RM, Maggioni AP, Tavazzi L, Tognoni G, Kjekshus J, Pedersen TR, Cook TJ, Gotto AM, Clearfield MB, Downs JR, Nakamura H, Ohashi Y, Mizuno K, Ray KK, Ford I. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 2010; 375:735-742
  87. Preiss D, Seshasai SR, Welsh P, Murphy SA, Ho JE, Waters DD, DeMicco DA, Barter P, Cannon CP, Sabatine MS, Braunwald E, Kastelein JJ, de Lemos JA, Blazing MA, Pedersen TR, Tikkanen MJ, Sattar N, Ray KK. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA 2011; 305:2556-2564
  88. Sattar N. Statins and diabetes: What are the connections? Best Pract Res Clin Endocrinol Metab 2023; 37:101749
  89. Rojas-Fernandez C, Hudani Z, Bittner V. Statins and Cognitive Side Effects: What Cardiologists Need to Know. Endocrinol Metab Clin North Am 2016; 45:101-116
  90. Rojas-Fernandez CH, Goldstein LB, Levey AI, Taylor BA, Bittner V. An assessment by the Statin Cognitive Safety Task Force: 2014 update. J Clin Lipidol 2014; 8:S5-16
  91. Richardson K, Schoen M, French B, Umscheid CA, Mitchell MD, Arnold SE, Heidenreich PA, Rader DJ, deGoma EM. Statins and cognitive function: a systematic review. Ann Intern Med 2013; 159:688-697
  92. Olmastroni E, Molari G, De Beni N, Colpani O, Galimberti F, Gazzotti M, Zambon A, Catapano AL, Casula M. Statin use and risk of dementia or Alzheimer's disease: a systematic review and meta-analysis of observational studies. Eur J Prev Cardiol 2022; 29:804-814
  93. Trompet S, van Vliet P, de Craen AJ, Jolles J, Buckley BM, Murphy MB, Ford I, Macfarlane PW, Sattar N, Packard CJ, Stott DJ, Shepherd J, Bollen EL, Blauw GJ, Jukema JW, Westendorp RG. Pravastatin and cognitive function in the elderly. Results of the PROSPER study. J Neurol 2010; 257:85-90
  94. Collins R, Armitage J, Parish S, Sleight P, Peto R, Heart Protection Study Collaborative Group. Effects of cholesterol-lowering with simvastatin on stroke and other major vascular events in 20536 people with cerebrovascular disease or other high-risk conditions. Lancet 2004; 363:757-767
  95. McGuinness B, Craig D, Bullock R, Malouf R, Passmore P. Statins for the treatment of dementia. Cochrane Database Syst Rev 2014:CD007514
  96. Rosenson RS, Baker SK, Jacobson TA, Kopecky SL, Parker BA, The National Lipid Association's Muscle Safety Expert Panel. An assessment by the Statin Muscle Safety Task Force: 2014 update. J Clin Lipidol 2014; 8:S58-71
  97. Schech S, Graham D, Staffa J, Andrade SE, La Grenade L, Burgess M, Blough D, Stergachis A, Chan KA, Platt R, Shatin D. Risk factors for statin-associated rhabdomyolysis. Pharmacoepidemiol Drug Saf 2007; 16:352-358
  98. Cholesterol Treatment Trialists Collaboration. Effect of statin therapy on muscle symptoms: an individual participant data meta-analysis of large-scale, randomised, double-blind trials. Lancet 2022; 400:832-845
  99. Bruckert E, Hayem G, Dejager S, Yau C, Begaud B. Mild to moderate muscular symptoms with high-dosage statin therapy in hyperlipidemic patients--the PRIMO study. Cardiovasc Drugs Ther 2005; 19:403-414
  100. Buettner C, Davis RB, Leveille SG, Mittleman MA, Mukamal KJ. Prevalence of musculoskeletal pain and statin use. J Gen Intern Med 2008; 23:1182-1186
  101. Cohen JD, Brinton EA, Ito MK, Jacobson TA. Understanding Statin Use in America and Gaps in Patient Education (USAGE): an internet-based survey of 10,138 current and former statin users. J Clin Lipidol 2012; 6:208-215
  102. Gupta A, Thompson D, Whitehouse A, Collier T, Dahlof B, Poulter N, Collins R, Sever P. Adverse events associated with unblinded, but not with blinded, statin therapy in the Anglo-Scandinavian Cardiac Outcomes Trial-Lipid-Lowering Arm (ASCOT-LLA): a randomised double-blind placebo-controlled trial and its non-randomised non-blind extension phase. Lancet 2017; 389:2473-2481
  103. Giugliano RP, Mach F, Zavitz K, Kurtz C, Im K, Kanevsky E, Schneider J, Wang H, Keech A, Pedersen TR, Sabatine MS, Sever PS, Robinson JG, Honarpour N, Wasserman SM, Ott BR. Cognitive Function in a Randomized Trial of Evolocumab. N Engl J Med 2017; 377:633-643
  104. Koren MJ, Giugliano RP, Raal FJ, Sullivan D, Bolognese M, Langslet G, Civeira F, Somaratne R, Nelson P, Liu T, Scott R, Wasserman SM, Sabatine MS. Efficacy and safety of longer-term administration of evolocumab (AMG 145) in patients with hypercholesterolemia: 52-week results from the Open-Label Study of Long-Term Evaluation Against LDL-C (OSLER) randomized trial. Circulation 2014; 129:234-243
  105. Robinson JG, Farnier M, Krempf M, Bergeron J, Luc G, Averna M, Stroes ES, Langslet G, Raal FJ, El Shahawy M, Koren MJ, Lepor NE, Lorenzato C, Pordy R, Chaudhari U, Kastelein JJ. Efficacy and safety of alirocumab in reducing lipids and cardiovascular events. N Engl J Med 2015; 372:1489-1499
  106. Shapiro MD, Feingold KR. Monogenic Disorders Causing Hypobetalipoproteinemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  107. McKenney JM. Understanding PCSK9 and anti-PCSK9 therapies. J Clin Lipidol 2015; 9:170-186
  108. LaRosa JC, Grundy SM, Kastelein JJ, Kostis JB, Greten H. Safety and efficacy of Atorvastatin-induced very low-density lipoprotein cholesterol levels in Patients with coronary heart disease (a post hoc analysis of the treating to new targets [TNT] study). Am J Cardiol 2007; 100:747-752
  109. Wiviott SD, Cannon CP, Morrow DA, Ray KK, Pfeffer MA, Braunwald E. Can low-density lipoprotein be too low? The safety and efficacy of achieving very low low-density lipoprotein with intensive statin therapy: a PROVE IT-TIMI 22 substudy. J Am Coll Cardiol 2005; 46:1411-1416
  110. Everett BM, Mora S, Glynn RJ, MacFadyen J, Ridker PM. Safety profile of subjects treated to very low low-density lipoprotein cholesterol levels (<30 mg/dl) with rosuvastatin 20 mg daily (from JUPITER). Am J Cardiol 2014; 114:1682-1689
  111. Giugliano RP, Wiviott SD, Blazing MA, De Ferrari GM, Park JG, Murphy SA, White JA, Tershakovec AM, Cannon CP, Braunwald E. Long-term Safety and Efficacy of Achieving Very Low Levels of Low-Density Lipoprotein Cholesterol : A Prespecified Analysis of the IMPROVE-IT Trial. JAMA Cardiol 2017; 2:547-555
  112. Wachira JK, Larson MK, Harris WS. n-3 Fatty acids affect haemostasis but do not increase the risk of bleeding: clinical observations and mechanistic insights. Br J Nutr 2014; 111:1652-1662
  113. Feingold KR, Chait A. Approach to patients with elevated low-density lipoprotein cholesterol levels. Best Pract Res Clin Endocrinol Metab 2023; 37:101658
  114. Score Op working group E. S. C. Cardiovascular risk collaboration. SCORE2-OP risk prediction algorithms: estimating incident cardiovascular event risk in older persons in four geographical risk regions. Eur Heart J 2021; 42:2455-2467
  115. Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, Himmelfarb CD, Khera A, Lloyd-Jones D, McEvoy JW, Michos ED, Miedema MD, Munoz D, Smith SC, Jr., Virani SS, Williams KA, Sr., Yeboah J, Ziaeian B. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 140:e596-e646
  116. Piercy KL, Troiano RP, Ballard RM, Carlson SA, Fulton JE, Galuska DA, George SM, Olson RD. The Physical Activity Guidelines for Americans. JAMA 2018; 320:2020-2028
  117. Yun H, Su W, Zhao H, Li H, Wang Z, Cui X, Xi C, Gao R, Sun Y, Liu C. Effects of different exercise modalities on lipid profile in the elderly population: A meta-analysis. Medicine (Baltimore) 2023; 102:e33854
  118. Feingold KR. The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  119. Ioannidis JPA. The Challenge of Reforming Nutritional Epidemiologic Research. JAMA 2018; 320:969-970
  120. Nissen SE. U.S. Dietary Guidelines: An Evidence-Free Zone. Ann Intern Med 2016; 164:558-559
  121. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Fito M, Gea A, Hernan MA, Martinez-Gonzalez MA. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N Engl J Med 2018; 378:e34
  122. Estruch R, Martinez-Gonzalez MA, Corella D, Salas-Salvado J, Ruiz-Gutierrez V, Covas MI, Fiol M, Gomez-Gracia E, Lopez-Sabater MC, Vinyoles E, Aros F, Conde M, Lahoz C, Lapetra J, Saez G, Ros E. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145:1-11
  123. de Lorgeril M, Renaud S, Mamelle N, Salen P, Martin JL, Monjaud I, Guidollet J, Touboul P, Delaye J. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet 1994; 343:1454-1459
  124. Delgado-Lista J, Alcala-Diaz JF, Torres-Pena JD, Quintana-Navarro GM, Fuentes F, Garcia-Rios A, Ortiz-Morales AM, Gonzalez-Requero AI, Perez-Caballero AI, Yubero-Serrano EM, Rangel-Zuniga OA, Camargo A, Rodriguez-Cantalejo F, Lopez-Segura F, Badimon L, Ordovas JM, Perez-Jimenez F, Perez-Martinez P, Lopez-Miranda J. Long-term secondary prevention of cardiovascular disease with a Mediterranean diet and a low-fat diet (CORDIOPREV): a randomised controlled trial. Lancet 2022; 399:1876-1885
  125. Chait A, Feingold KR. Approach to patients with hypertriglyceridemia. Best Pract Res Clin Endocrinol Metab 2023; 37:101659
  126. Awad K, Mikhailidis DP, Toth PP, Jones SR, Moriarty P, Lip GYH, Muntner P, Catapano AL, Pencina MJ, Rosenson RS, Rysz J, Banach M. Efficacy and Safety of Alternate-Day Versus Daily Dosing of Statins: a Systematic Review and Meta-Analysis. Cardiovasc Drugs Ther 2017; 31:419-431
  127. Kennedy SP, Barnas GP, Schmidt MJ, Glisczinski MS, Paniagua AC. Efficacy and tolerability of once-weekly rosuvastatin in patients with previous statin intolerance. J Clin Lipidol 2011; 5:308-315
  128. Ballantyne CM, Laufs U, Ray KK, Leiter LA, Bays HE, Goldberg AC, Stroes ES, MacDougall D, Zhao X, Catapano AL. Bempedoic acid plus ezetimibe fixed-dose combination in patients with hypercholesterolemia and high CVD risk treated with maximally tolerated statin therapy. Eur J Prev Cardiol 2020; 27:593-603

 

The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels

ABSTRACT

 

The role of lipids and lipoproteins as causal factors for cardiovascular disease (CVD) is well established. Dietary saturated fatty acids (SFA), which are in milk, butter, cheese, beef, lamb, pork, poultry, palm oil, and coconut oil increase LDL-C and HDL-C. The increase in LDL-C is due to a decrease in hepatic LDL clearance and an increase in LDL production secondary to a decrease in hepatic LDL receptors. Monounsaturated fatty acids (MUFA) are in olive, canola, peanut, safflower, and sesame oil, and avocados, peanut butter, and many nuts and seeds and polyunsaturated fatty acids (PUFA) are in soybean, corn, and sunflower oil, and some nuts and seeds, tofu, and soybeans. Both MUFA and PUFA lower LDL-C by increasing hepatic LDL receptor activity. Dietary cholesterol is found in egg yolks, shrimp, beef, pork, poultry, cheese, and butter and increase LDL-C but the effect is modest and varies with approximately 15-25% of individuals being hyper-responders with more robust increases. Dietary cholesterol reduces hepatic LDL receptor activity, decreasing the clearance and increasing the production of LDL. Trans fatty acids (TFA) occur naturally in meat and dairy products and are formed during the partial hydrogenation of vegetable fat. TFA increase LDL-C and decrease HDL-C. Carbohydrates (CHO) can be divided into high-quality, for example fruits, legumes, vegetables, and whole grains, or low-quality, which include refined grains, starches, and added sugars. CHO increase TG with low quality CHO, particularly added sugars, having a more robust effect. Dietary CHO, particularly fructose, promotes hepatic de novo fatty acid synthesis leading to increased VLDL secretion. Fiber is found mostly in fruits, vegetables, whole and unrefined grains, nuts, seeds, beans, and legumes and phytosterols are naturally occurring constituents of plants and are found in vegetable oils, cereals, nuts, fruit and vegetables. Both dietary fiber and phytosterols decrease LDL-C by decreasing intestinal cholesterol absorption.

Summary of the Effect of Dietary Constituents on Lipid and Lipoproteins

SFA

Increase LDL-C and modest increase HDL-C

MUFA and PUFA

Decrease LDL-C

TFA

Increase LDL-C and decrease HDL-C

Cholesterol

Increase LDL-C

CHO

Increase TGs particularly simple sugars

Fiber

Decrease LDL-C

Phytosterols

Decrease LDL-C

With regards to CVD there are very few well conducted randomized controlled trials and most of the information is derived from observational studies that demonstrate associations. These observational studies have found that fruits, vegetables, beans/legumes, nuts/seeds, whole grains, fish, yogurt, fiber, seafood omega-3 fatty acids, and polyunsaturated fats were associated with a decreased risk of CVD while unprocessed red meats, processed meats, sugar-sweetened beverages, high glycemic load CHO, and trans-fats were associated with an increased risk of CVD. Randomized trials have shown that a Mediterranean diet reduces CVD. Based on this information current guidelines for the general population recommend 1. A diet emphasizing intake of vegetables, fruits, legumes, nuts, whole grains, and fish 2. Replacement of SFA with MUFA and PUFA 3. A reduced amount of dietary cholesterol 4. Minimizing intake of processed meats, refined CHO, and sweetened beverages and 5. Avoidance of TFA. For individuals with a high LDL-C limiting dietary SFA, TFA, and cholesterol and increasing fiber and phytosterols will help lower LDL-C while in individuals with high TG limiting low quality CHO, particularly simple sugars, and ethanol with weight loss, if indicated, will help lower TG.

 

INTRODUCTION

 

There is a huge literature describing the effect of diet on the risk of cardiovascular disease (CVD) and this literature is often conflicting and controversial. Several well recognized investigators have discussed the limitations of the information linking various diets and dietary constituents and the risk of disease (1,2). The major problem is that almost all of the information is based on observational studies and well conducted randomized trials measuring important cardiovascular outcomes are very rare. Observational studies can demonstrate associations but do not necessarily indicate that there is a cause-and-effect relationship. Unrecognized confounding variables can result in false associations. In several instances a robust association was observed in observation trials but randomized trials failed to confirm these observations (3). For example, several observational studies showed that higher vitamin E intake from dietary sources or supplements was associated with a lower risk of CVD (4-8), but randomized controlled trials failed to demonstrate a reduction in cardiovascular events with vitamin E supplementation (9-12). Observational studies have also reported that vitamin B6, B12, or folic acid intake reduced the risk of CVD (13-15), but again randomized controlled trials failed to demonstrate a benefit of increased vitamin intake on CVD (16-19). These results point to potential deficiencies in observational studies and the need to recognize that the associations demonstrated in observational studies may not always be causal. Therefore, in this chapter, where possible, we will focus on randomized controlled trials.

 

Moreover, even the interpretation of the results of observational trials is often debated. For example, a 2019 meta-analysis and systematic review published in the Annals of Internal Medicine reached the conclusion that “the magnitude of association between red and processed meat consumption and all-cause mortality and adverse cardiometabolic outcomes is very small, and the evidence is of low certainty” (20). This conclusion is contrary to the recommendations of almost all dietary guidelines and as would be expected this resulted in a critique challenging this conclusion (21). There are numerous other instances where there are conflicting results and interpretations in the literature linking diet with CVD making it difficult to sort out fact from fiction.

 

The information pertaining to the effect of dietary manipulations on lipid and lipoprotein levels are frequently based on randomized controlled trials rather than observational studies and therefore tend to be more consistent. However, even in these studies the results are sometimes conflicting. There are many factors that could account for this variability including the heterogeneity in study settings, type of individuals studied, study designs, differences in baseline diets, adherence to the study diet, differences in types of diet or dietary composition, methods and accuracy of the methods used to measure lipid and lipoprotein levels, and many other factors.

 

Additionally, the clinician should recognize that the lipid response of an individual patient to dietary manipulations can vary greatly, is very modest on average (in the range of 10% reductions, typically), and in most cases will not prevent the need for lipid lowering medications. The importance of genetic differences on these responses is often under recognized by patients and providers. For example, individuals with an apo E4 allele have a more robust decrease in LDL-C in response to a decrease in dietary fat and cholesterol than subjects carrying the apo E3 or apo E2 alleles (22). Polymorphisms in other genes have also been shown to modulate the lipid and lipoprotein response to dietary manipulations (22,23). Clinical conditions can also affect the response to diet. For example, the expected lipid and lipoprotein response to a low cholesterol, low saturated fatty acids (SFA) diet is blunted in obese individuals (24). Therefore, the effect of a specific diet can vary from individual to individual and the clinician will have to monitor a patient’s response.

 

It should be recognized that when one increases or decreases a particular macronutrient in the diet (lipids, carbohydrates (CHO), or protein) there needs to be a reciprocal change in another macronutrient to maintain caloric balance. It can therefore be difficult to know whether the increase in a particular nutrient or a decrease in another nutrient is accounting for the observed effect (for example decreasing SFA and increasing CHO). Where possible I will try to specify which nutrient was decreased and which was increased in the studies described.

 

Finally, it is important to look at the effect of diet on lipids independent of weight loss. Weight loss per se can affect lipid levels resulting in a decrease in triglycerides and LDL-C and an increase in HDL-C levels (25). For a detailed discussion of the effect of weight loss on lipid levels see the chapter on obesity and dyslipidemia (25).

 

In this chapter we will first discuss the effect of various macronutrients, then specific foods, and finally specific diets on lipids and lipoprotein levels.  

 

DIETARY SATURATED FATTY ACIDS

 

Major sources of saturated fatty acids (SFA) in the diet are milk, butter, cheese, other dairy products, beef, lamb, pork, poultry particularly the skin, palm oil, palm kernel oil, and coconut oil (tables 1 and 3).

 

Table 1. Fatty Acid Composition of Foods High in Saturated Fat

 

Total Fat

grams/100 grams

SFA

grams/100 grams

MUFA

grams/100 grams

PUFA

grams/100 grams

Hamburger

15.0

5.89

6.66

0.49

Pork loin

13.3

5.23

6.19

1.20

Chicken

12.6

3.50

4.93

2.74

Lamb

15.1

6.90

7.00

1.20

Whole milk*

3.9

2.5

1.0

0.1

Gouda cheese**

30.6

20.3

7.4

0.9

Butter***

82.2

52.1

20.9

2.8

*TFA = 0.1g/100g; **TFA = 1.1g/100g; TFA = 2.9g/100g.

TFA= trans fatty acids, MUFA= monounsaturated fatty acids, PUFA= polyunsaturated fatty acids.

 

Effect of Dietary Saturated Fatty Acids on Cardiovascular Disease

 

OBSERVATIONAL STUDIES

 

Dietary guidelines uniformly recommend reducing the intake of SFA. There are a large number of observational trials that have shown an association between dietary SFA intake and CVD (26-31). However, there are meta-analyses that have not found an association between dietary SFA intake and CVD (32-36). A possible explanation for this discordance is whether the SFA in the diet is replaced by polyunsaturated fatty acids (PUFA) vs. replaced by CHO. When SFA is replaced by PUFA there is a reduction in CVD whereas replacement with CHO has no benefit on CVD (27-29,37-39). However, replacement of SFA with high quality CHO may be beneficial (27,37,38). Additionally, in one study SFA from meat was associated with an increased risk of CVD while SFA from dairy products was associated with a decrease in CVD (40). Thus, the source of SFA may be important. 

 

As noted above, observational studies can demonstrate an association but are not able to definitively demonstrate a causal relationship. It is therefore essential to review the results of randomized controlled trials on the effect of decreasing dietary SFA on preventing cardiovascular events.

 

RANDOMIZED CONTROLLED OUTCOME TRIALS

 

This section will review the major randomized trials analyzing the effect of decreasing SFA intake on preventing CVD. Studies with very few participants, few cardiovascular events, or very short-term studies will not be included. It is important to note that many of these studies were carried out in the 1950’s and 1960’s when the diagnosis and treatment of CVD was very primitive compared to current standards. Also, typical diets were much different (higher in SFA) and mean plasma cholesterol levels were higher. Lastly, the methodology of these studies was not up to the current standards by which randomized controlled trials are performed (small number of patients, often not blinded, inadequate statistical power, non-specific endpoints, etc.). Thus, the accuracy of these trials and the relevancy of these older studies to current times is uncertain.

 

In a study from England initiated in 1957, 252 men under the age of sixty-five who recently

had a myocardial infarction were assigned to a low-fat diet or usual diet (41). The low-fat diet was limited to 40 grams per day of fat with decreases in butter and meat. The intake of fat during the trial was approximately 100-120 grams per day in the usual diet group and slightly greater than 40 grams per day in the low-fat diet group. At the time of the study the typical diet was high in SFA so a decrease in total fat would have resulted in a significant decrease in SFA. During the trial serum cholesterol levels were in the 240mg/dL range in the usual diet group and 220mg/dL in the low-fat diet group. There were no differences between the two groups in cardiovascular events during the 5 years of the trial. To see a reduction in cardiovascular events with the modest reduction in serum cholesterol levels this study would have required a much larger number of participants.

 

The Oslo Diet-Heart Study randomized men under 65 years of age with a history of a myocardial infarction to a diet low in SFA and cholesterol, and high in PUFA (n=206) or their usual diet (n=206) (42). Cholesterol levels were approximately 295mg/dL and decreased to approximately 240mg/dL in the patients on the low SFA diet with minimal changes in the control group. After 5 years major cardiovascular events and cardiovascular mortality were reduced in the group on the low SFA diet (Events- 61 low SFA group vs. 81 control group; Mortality- 38 low SFA group vs. 52 control group).

 

The Medical Research Council soya-bean trial randomized men under 60 years of age with a recent myocardial infarction to continue their usual diet (n=194) or a diet low in SFA and containing 85 grams of soya-bean oil daily (PUFA) (n=199) (43). The low SFA diet lowered cholesterol from 272 to 213mg/dL (22% decrease) while in the controls, cholesterol decreased from 273 to 259mg/dL (6% decrease). The primary outcome was first relapse (myocardial infarction, angina, sudden death). After 4 years, 62 of 199 in the soybean oil group had a recurrent coronary event compared with 74 of 194 in the usual diet group; the difference, −18% (95% CI, −38 to 7), was not statistically significant but given the small number of participants was suggestive of benefit.

 

The Los Angeles Veterans Administration Center study randomized 422 men to the conventional control diet and 424 to the experimental diet low in SFA and cholesterol and enriched in PUFA (44,45). 30% of the men had CVD. The baseline plasma cholesterol was 233mg/dL and on treatment there was a 13% decrease in the experimental diet compared to controls. Over 8 years the primary endpoint of myocardial infarction and sudden death from ischemia were reduced in the experimental diet group (control 67 vs experimental diet 45). The difference in the primary end point of the study-sudden death or myocardial infarction was not statistically significant but when these data were pooled with those for cerebral infarction and other secondary end points, the totals were 96 in the control group and 66 in the experimental group; P=0.01. Fatal atherosclerotic events occurred in 70 patients in the control group and 48 in the experimental group (P<0.05). For all primary and secondary end points the incidence rates were 47.7% and 31.3% for the control and experimental groups respectively (P= 0.02).

 

The Finnish Mental Hospital Study was carried out in two mental hospitals. One hospital was switched to a diet low in SFA and cholesterol and relatively high in PUFA, while the other hospital continued the usual hospital diet (46-48). After 6 years the type of diet was reversed in each hospital. The individuals in this study were hospitalized men between 34 to 64 years of age and women age 44 to 64 years. During the study individuals were removed from the study and others added to the study cohort. The serum cholesterol level on the usual diet was 268mg/dL while on the low SFA diet the serum cholesterol level was 226mg/dL. The incidence of CVD was consistently much lower during the low SFA diet periods than during the normal-diet periods but detailed comparisons are difficult due to the lack of randomization of individuals and the adding and removal of individuals during the study leading to only 36% of the men and 20.6% of the women completing both periods of the study. Nevertheless, this study provides evidence of the benefit of a diet low in SFA and cholesterol and enriched in PUFA.

 

The Sydney Diet Heart Study was a randomized controlled trial conducted from 1966 to 1973 that evaluated the effects of increasing linoleic acid from safflower oil (PUFA ~ 15% of calories) in place of SFA (<10% of calories) in men aged 30-59 years with a history of coronary artery disease (49). Participants were randomized to the dietary intervention group (n=221) or a control group with no specific dietary instruction (n=237). Baseline cholesterol levels were ~280mg/dL and decreased to 267mg/dL in the control group and 244mg/dL in the diet intervention group. Compared with the control group, the intervention group had an increased risk of all-cause mortality (17.6% v 11.8%; P=0.051), cardiovascular mortality (17.2% v 11.0%; P=0.037), and mortality from coronary heart disease (16.3% v 10.1%; P=0.036) over the 5 years of the trial. The reason for the increase in mortality is not clear but the safflower oil margarine substitute for animal fats may have contained trans fatty acids, which could have increased CVD.

 

The DART trial was a multicenter trial in men less than 70 years of age with a diagnosis of an acute myocardial infarction (50). There were several different dietary approaches used in this trial but the one of interest reduced fat intake to 30% of total energy and increased the PUFA/SFA ratio to 1.0 (n=1018) vs. no advice (n=1015). The fat advice group reduced SFA from 15% to 11% of total calories, increased PUFA from 7% to 9%, and increased carbohydrate intake from 44% to 46%. Cholesterol levels were reduced by 3.6% (baseline 252mg/dL) in the diet advice group. During the 2-year trial the number of cardiovascular events were similar in the diet group vs. no advice group.

 

The Minnesota Coronary Survey was a 4.5-year, randomized trial that was conducted in six Minnesota state mental hospitals and one nursing home and included 4,393 men and 4,664 women (51). The trial compared the effects of the usual diet (18% SFA, 5% PUFA, 16% monounsaturated fatty acid (MUFA), 446 mg dietary cholesterol per day) versus a low SFA and cholesterol treatment diet (9% SFA, 15% PUFA, 14% MUFA, 166 mg dietary cholesterol per day). The mean duration of time on the diets was 384 days, with 1,568 subjects consuming the diet for over 2 years. The baseline serum cholesterol level was 207 mg/dL, falling to 175 mg/dL in the treatment group and 203 mg/dL in the control group. No differences between the treatment and control groups were observed for cardiovascular events, cardiovascular deaths, or total mortality, perhaps due to the relatively short duration of this study.

 

The Women’s Health Initiative trial randomized 19,541 postmenopausal women 50-79 years of age to the diet intervention group and 29,294 women to usual dietary advice (52). The goal in the diet intervention group was to reduce total fat intake to 20% of calories and increase intake of vegetables/fruits to 5 servings/day and grains to at least 6 servings/day. Fat intake decreased by 8.2% of energy intake in the intervention vs the comparison group, with small decreases in SFA (2.9%), MUFA (3.3%), and PUFA (1.5%) with increased consumption of vegetables, fruits, and grains. LDL-C levels were reduced by 3.55 mg/dL in the intervention group while levels of HDL-C and TGs were not significantly different in the intervention vs comparison groups. The dietary intervention did not significantly decrease CVD. In fact, in the women with pre-existing CVD there was an increase in cardiovascular events with diet therapy.

 

Summary of Dietary Randomized Controlled Trials 

 

In reviewing these randomized controlled trials, it appears that the dietary studies that produce a long-term decrease in plasma cholesterol levels resulted in a reduction in cardiovascular events (Oslo Diet-Heart Study, soya-bean trial, Los Angeles Veterans Administration Center, Finnish Mental Hospital Study) while the dietary studies that did not produce a long-term decrease in plasma cholesterol levels failed to demonstrate a reduction in CVD. The baseline plasma cholesterol levels in the positive studies tended to be high and allowed for a robust cholesterol lowering with dietary manipulation. Additionally, as will presented in the next section the greater the reduction in SFA in the diet the greater the decrease in TC and LDL-C levels and many of the positive studies were carried out in an era when the content of SFA in the diet was high. Additionally, studies in non-human primates have also demonstrated that reducing SFA intake reduces atherosclerosis (53,54).

 

These results correspond very nicely with the large number of trials demonstrating that using a variety of different pharmacologic agents that lower plasma cholesterol levels results in a decrease in cardiovascular events (55). In an analysis comparing cholesterol lowering with diet vs. drug therapy it was observed that a similar decrease in cardiovascular events occurred adjusting for the magnitude of cholesterol lowering (56). Thus, it would appear that diets that decrease dietary SFA and thereby lead to a significant decrease in plasma cholesterol levels for an extended period of time have benefits on CVD with the caveat that there is not an increase in other nutrients that will adversely affect other parameters thereby negating the beneficial effects of decreasing SFA. For example, an increase in dietary simple sugars for SFA could lead to an increase in TG levels with negative effects.

 

REVERSAL OF ATHEROSCLEROSIS TRIALS

 

Two studies have examined the effect of decreasing dietary SFA on atherosclerotic lesions.

 

The St Thomas’ Atherosclerosis Regression Study (STARS) determined the effect of decreasing dietary saturated fat in the diet (n=26) vs. usual diet (n=24) in men less than 66 years of age with a plasma cholesterol greater than 234mg/dL referred for coronary angiography to investigate angina pectoris or other findings suggestive of coronary heart disease (57). In the diet group total fat intake was reduced to 27% of dietary energy, saturated fatty acid content to 8-10% of dietary energy, and dietary cholesterol to 100 mg/1000 kcal; omega-6 and omega-3 polyunsaturated fatty acids were increased to 8% of dietary energy, and plant-derived soluble fiber intake was increased to the equivalent of 3-6 g polygalacturonate/1000 kcal. During the trial LDL-C levels were 163mg/dL in the diet intervention group vs.182mg/dL in the usual diet group. Additionally, TGs decreased in the diet intervention group (206mg/dL to 165mg/dl) with no change in TG levels in the usual diet group. After approximately 3 years coronary angiography revealed that the percentage of patients who showed progression of coronary narrowing was significantly reduced by the dietary intervention (usual diet 46% vs, dietary intervention 15%), whereas the percentage who showed an increase in luminal diameter rose significantly (usual diet 4% vs. dietary intervention 38%). While the number of cardiovascular events was small, they were significantly reduced in the dietary intervention group (usual diet 36% vs dietary intervention 11%; p< 0.05). Finally, the improvement in angiographic appearance correlated with LDL-C levels.

 

The Lifestyle Heart Trial was a one year randomized, controlled trial to determine whether lifestyle changes affect coronary atherosclerosis in patients with angiographically documented coronary artery disease (58). Patients were assigned to the lifestyle group (low-fat vegetarian diet, stopping smoking, stress management training, and moderate exercise) (n= 22) or a usual-care control group (n=19). The lifestyle diet contained approximately 10% of calories as fat PUFA/SFA ratio greater than 1), 15-20% protein, and 70-75% predominantly complex carbohydrates. Cholesterol intake was limited to 5 mg/day or less. In the lifestyle group LDL-C decreased from 153mg/dL to 96mg/dl (37% decrease) whereas in the usual care group LDL-C decreased from 168mg/dL to 159mg/dL. Patients in the lifestyle group reported a 91% decrease in the frequency of angina, a 42% decrease in the duration of angina, and a 28% decrease in the severity of angina. In contrast, patients in the usual care group reported a 165% increase in the frequency of angina, a 95% increase in the duration of angina, and a 39% increase in the severity of angina. In the lifestyle group regression of coronary atherosclerosis occurred in 18 of the 22 patients (82%) whereas in the usual care group progression of coronary atherosclerosis occurred in 10 of 19 patients (53%).

 

These two regression trials provide strong support for the results observed in the randomized cardiovascular outcome studies described above i.e., that lowering LDL-C levels by decreasing dietary SFA can reduce atherosclerosis and cardiovascular events.

 

Effect of Dietary Saturated Fatty Acids on Lipid Levels          

 

It should be recognized that when one increases or decreases a particular macronutrient in the diet there needs to be a reciprocal change in another macronutrient to maintain caloric balance.

The effect of substituting PUFA, MUFA, or carbohydrates (CHO) for SFA is shown in table 1. Note that this table shows the effect of replacing 5% of energy from SFA for the indicated dietary component. Thus, going from a diet where 15% of the calories is from SFA to a diet where 10% of the calories is from SFA is estimated to lower LDL-C levels from 6 to 9mg/dL depending on which dietary component replaces the SFA. To keep this decrease in LDL-C in perspective it is estimated that a 40mg/dL decrease in LDL-C induced by statin therapy will result in an approximate 20% decrease in cardiovascular events over a 5 year period of time but the lifetime benefits of a 10 mg/dL decrease in LDL-C due to genetic variants will result in a 16–18% decrease in cardiovascular events (59). The effect on TGs is dependent on the dietary component replacing SFA with CHO resulting in a large increase in TG levels. One should note that there is also a decrease in HDL-C with replacement of SFA (table 2).

 

Table 2. Effect of Decreasing Dietary Saturated Fatty Acids on Lipid Levels

Dietary Component

LDL-C (mg/dL)

TGs (mg/dL)

HDL-C (mg/dL)

PUFA

−9.0    

-2.0

-1.0

MUFA

-6.5

+1.0

-6.0

CHO

-6.0

+9.5

-2.0

PUFA- polyunsaturated fatty acids; MUFA- monounsaturated fatty acids; CHO- carbohydrates.

Effects on lipoprotein lipids of replacing 5% of energy from SFA with the 5% of energy from the specified dietary component. Table adapted from references (26,60).

 

SFA in the diet predominantly increases LDL-C levels, predominantly larger, cholesterol-enriched LDL, with modest increases in HDL-C (60,61). As expected, Apo B and apo AI levels also increase (60). These effects are observed in both men and women (60). The effect of a decrease or increase in SFA intake on lipids and lipoproteins is linear with a consistent effect on serum lipids and lipoproteins across a wide range of SFA intakes (60). Of note the effects of decreasing SFA intake was observed even when the SFA intake was already less than 10% of the daily energy intake. Most studies have suggested that replacement of SFA with carbohydrate or unsaturated fat modestly increases Lp(a) but the results have varied from study to study with replacement of SFA with unsaturated fat from particular food sources such as nuts showing no increase in Lp(a) (62).

 

Individual SFA have diverse biological and cholesterol-raising effects with chain length of SFA playing an important role in determining the effect on lipid and lipoprotein levels. The most commonly consumed SFA are palmitic acid (16:0; major source: vegetable oil, dairy, and meat), stearic acid (18:0; meat, dairy, and chocolate), myristic acid (14:0; dairy and tropical oil, particularly coconut oil) and lauric acid (12:0; dairy and tropical oil). A meta-analysis of 60 controlled trials by Mensink et al. reported an increase in LDL-C and HDL-C concentrations by isocaloric replacement of carbohydrates with palmitic, myristic, and lauric acids (63). As expected, apolipoprotein B and A-I also increase (60,64). Myristic and palmitic acids increased LDL-C and HDL-C levels to a similar extent, whereas lauric acid had the largest LDL-C- and HDL-C-raising effect (63,65). Stearic acid did not increase LDL-C levels (63,65).The lack of an association between stearic acid and changes in LDL-C levels has been linked to a slower and/or less efficient absorption as well as desaturation of stearic acid to oleic acid (66). Compared with carbohydrates, an increased intake of lauric, myristic, palmitic or stearic acid lowered TG levels (63,65). For a specific individual many factors including lifestyle factors such as overall dietary composition and physical activity, clinical conditions such as obesity, insulin resistance and hypertriglyceridemia, as well as genetic factors may modify these responses.

 

MECHANISM FOR THE INCREASE IN LDL-C

 

Dietary SFA have been shown to decrease hepatic LDL receptor activity, protein, and mRNA levels and this results in a decrease in the clearance of circulating LDL leading to increased LDL-C levels (67,68). Additionally, the decrease in LDL receptors could result in an increase in the conversion of intermediate density lipoproteins to LDL rather than clearance by the liver (i.e., LDL production is enhanced).

 

SFA have been shown to decrease the formation of cholesterol esters, a reaction catalyzed by the enzyme acyl CoA:cholesterol acyltransferase (ACAT) (68). Free cholesterol in the endoplasmic reticulum is the primary regulator of the activation of sterol receptor binding protein (SREBP), which translocates to the nucleus and enhances the transcription of the LDL receptor (69). Elevated levels of cholesterol in the endoplasmic reticulum prevents the activation of SREBP (69). When free cholesterol is esterified into cholesterol esters it no longer prevents the activation of SREBP and the up-regulation LDL receptor expression. Thus, SFA by decreasing the formation of cholesterol esters and increasing free cholesterol may lead to the down-regulation of LDL receptor expression (68).

 

DIETARY MONOUNSATURATED AND POLYUNSATURATED FATTY ACIDS

 

Olive oil, canola oil, peanut oil, safflower oil, sesame oil, avocados, peanut butter, and many nuts and seeds are major sources of MUFA (table 3). Soybean oil, corn oil, sunflower oil, some nuts and seeds such as walnuts and sunflower seeds, tofu, and soybeans are major sources of PUFA (table 3). Omega-3-fatty acids, eicosapentaenoic acid (EPA, 20:5) and docosahexaenoic acid (DHA, 22:6), are mostly found in fish and other seafood, while another omega-3 fatty acid, alpha-linolenic acid (ALA, 18:3) is found mostly in nuts and seeds such as walnuts, flaxseed, and some vegetable oils such as soybean and canola oils. The body is capable of converting ALA into EPA and DHA but the conversion rates are low.

 

Table 3. Fat Composition of Oils, Lard, Butter, and Margarine

Type of Oil

SFA (%)

MUFA (%)

PUFA (%)

Corn oil

13.6

28.97

57.43

Safflower oil (linoleic)

6.51

15.1

78.4

Canola oil

7.46

64.1

28.49

Almond oil

8.59

73.19

18.22

Olive oil

14.19

74.99

10.82

Soybean oil

16.27

23.69

60.0

Sesame oil

14.85

41.53

43.62

Sunflower oil (linoleic)

10.79

20.42

68.8

Avocado oil

12.1

73.8

14.11

Peanut oil

17.77

48.58

33.65

Palm oil

51.57

38.7

9.73

Coconut oil

91.92

6.16

1.91

Lard

41.1

47.23

11.73

Butter

68.1

27.87

4.0

Margarine (soft)

20

47

33

Margarine (hard)

80

14

6

U.S. Department of Agriculture

 

Effect of Dietary Monounsaturated and Polyunsaturated Fatty Acids on Cardiovascular Disease

 

MONOUNSATURATED FATTY ACIDS

 

Many meta-analyses, but not all, have failed to demonstrate that MUFA intake reduces cardiovascular events (29,33,35,70). However, one meta-analysis and the Nurses’ Health Study and Health Professionals Follow-Up Study, two very large observational studies, found that MUFA when delivered from plant sources was protective but MUFA from other sources was not protective from developing cardiovascular events (71,72).

 

The PREDIMED a randomized controlled outcome trial employing a Mediterranean diet (increased MUFA) reduced the incidence of major CVD (73-75). In this multicenter trial, carried out in Spain, over 7,000 individuals at high risk for developing CVD were randomized to three diets (primary prevention trial). A Mediterranean diet supplemented with extra-virgin olive oil, a Mediterranean diet supplemented with mixed nuts, or a control diet. In the patients assigned to the Mediterranean diets there was 29% decrease in the primary composite end point (myocardial infarction, stroke, and death from CVD), which was primarily due to a decrease in strokes. The Mediterranean diet resulted in a small but significant increase in HDL-C levels and a small decrease in both LDL-C and TG levels (76). The changes in lipids were unlikely to account for the beneficial effects of the Mediterranean diet on CVD.

 

The Lyon Diet Heart Study randomized 584 patients who had a myocardial infarction within 6 months to a Mediterranean type diet vs usual diet (77,78). The oils recommended for salads and food preparation were rapeseed and olive oils exclusively. Additionally, they were also supplied with a rapeseed (canola) oil-based margarine. There was a marked reduction in events in the group of patients randomized to the Mediterranean diet (cardiac death and nonfatal myocardial infarction rate was 4.07 per 100 patient years in the control diet vs.1.24 in the Mediterranean diet; p<0.0001). Lipid levels were similar in both groups in this trial (77).

 

The CORDIOPREV study was a single center randomized trial that compared a Mediterranean diet to a low-fat diet in 1,002 patients with cardiovascular disease (79). The Mediterranean diet contained a minimum of 35% of the calories as fat (22% monounsaturated fatty acids, 6% polyunsaturated fatty acids, and <10% saturated fat), 15% proteins, and a maximum of 50% carbohydrates while the low-fat diet contained less than 30% of total fat (<10% saturated fat, 12–14% monounsaturated fatty acids, and 6–8% polyunsaturated fatty acids), 15% protein, and a minimum of 55% carbohydrates. The risk of an ASCVD event was reduced by approximately 25-30% in the Mediterranean diet group. Whether these diets differed in their effects on fasting lipid levels has not been reported.

 

The results of these three randomized trials indicate that a Mediterranean diet enriched in plant MUFA reduce the risk of CVD. It is likely that the beneficial effects of the Mediterranean diet on CVD is mediated by multiple mechanisms with alterations in lipid levels making only a minor contribution. It should be noted that in addition to an increase in MUFA the diet also includes low to moderate red wine consumption, high consumption of whole grains and cereals, low consumption of meat and meat products, increased consumption of fish, and moderate consumption of milk and dairy products. As in many dietary studies it is difficult to change a single variable and therefore the interpretation of which factor or factors account for the benefits is difficult to untangle.

 

POLYUNSATURATED FATTY ACIDS

 

Recent meta-analyses of the effect of PUFA on cardiovascular events in observational studies have demonstrated either no effect or a modestly lower risk of CVD and mortality (80-84). Randomized trials are described in the section on saturated fats and CVD and describe the results of replacing SFA with PUFA. It appears that dietary PUFA has a neutral effect on CVD except in the circumstances where it replaces SFA and results in a sustained decrease in plasma cholesterol levels leading to a decrease in cardiovascular events.

 

OMEGA-3-FATTY ACIDS

 

As discussed in detail in the chapter entitled “Triglyceride  Lowering Drugs” numerous randomized controlled trials of the effect of low dose omega-3-fatty acids (approximately ≤1 gram/day) on CVD have been published and the bulk of the evidence indicates no benefit (85). The effect of pharmacologic doses of omega-3-fatty acids (≥1.8 grams/day) on cardiovascular outcomes is discussed in the chapter entitled “Triglyceride  Lowering Drugs” (85).

 

Effect of Dietary Monounsaturated and Polyunsaturated Fatty Acids on Lipid Levels

 

Table 4 shows the effect of substituting PUFA or MUFA for carbohydrates on LDL-C, HDL-C, and TG levels. Both PUFA and MUFA decrease LDL-C and TGs but PUFA induces a greater decrease (60). Both PUFA and MUFA increase HDL-C levels (60).

 

Table 4. Effect of Decreasing Dietary Carbohydrate on Lipid Levels

Dietary Component

LDL-C (mg/dL)

TGs (mg/dL)

HDL-C (mg/dL)

PUFA

-4.3

-9.2

1.2

MUFA

-1.8

-6.6

1.6

PUFA- polyunsaturated fatty acids; MUFA- monounsaturated fatty acids;

Effects on lipoprotein lipids of replacing 5% of energy from carbohydrates with the 5% of energy from the specified dietary component. Table adapted from reference (60).

 

In a meta-analysis of 14 studies no significant differences in TC, LDL-C, or HDL-C  levels were observed when diets high in MUFA or PUFA were compared directly (86). TG levels were modestly but consistently lower on the diets high in PUFA (P = .05) (86).

 

While high dose omega-3-fatty acids (3-4 grams/day) lower TG levels, lower doses (≤1 gram/day) have minimal effects on lipid levels (85).

 

MECHANISM FOR THE DECREASE IN LDL-C

 

Unsaturated fatty acids increase hepatic LDL receptor activity, protein, and mRNA abundance, which will increase the clearance of LDL from the circulation (67,68). Unsaturated fatty acids are a preferred substrate for ACAT and thereby result in an increase in cholesterol ester formation and a decrease in free cholesterol in the liver (68). A decrease in hepatic free cholesterol will result in the up-regulation of LDL receptor expression leading to a decrease in LDL-C levels. PUFA also increase membrane fluidity leading to an increase in the ability of LDL receptors to bind LDL (67). Additionally, the increase in LDL receptors could result in a decrease in the conversion of intermediate density lipoproteins (IDL) to LDL due to increased uptake of IDL by the liver (i.e., LDL production is decreased).

 

DIETARY TRANS FATTY ACIDS

 

The two major sources of dietary trans fatty acids (TFA) are those that occur naturally in meat and dairy products as a result of anaerobic bacterial fermentation in ruminant animals and those formed during the partial hydrogenation of vegetable fat (the fatty acids in vegetable oils have cis double bonds) (87). Partial hydrogenation and the formation of TFA converts the liquid vegetable oil into a solid form at room temperature allowing for ease of use in food products and increased shelf life (87,88). TFA acids were widely used in baked products, packaged snack foods, margarines, and crackers (88). With the recognition of the adverse effects of TFA the use of partial hydrogenated oils in food products has markedly diminished World-wide and in the US is no longer allowed.

 

Effect of Trans Fatty Acids on Cardiovascular Disease

 

A meta-analysis by de Souza and colleagues of 5 studies with 70,864 participants found that the relative risk of coronary heart disease mortality disease was increased with dietary TFA (1.28; p=0.003) (34). Similarly, the relative risk of coronary heart disease was also increased (1.21; p<0,001) (34). Another meta-analysis by Chowdhury and colleagues of 5 studies with 155,270 participants found that the relative risk of coronary events was increased with higher intake of TFA (RR 1.16; CI 1.06-1.27) (33). It has been estimated that a 2 percent increase in energy intake from TFA was associated with a 23 percent increase in the incidence of coronary heart disease (88). Thus, observational studies have consistently demonstrated that an increase in dietary TFA increase the risk of CVD. Clearly it would not be ethical to carry out randomized trials of the effect of TFA acids on CVD.

 

Effect of Trans Fatty Acids on Lipid Levels

 

The effect of replacing SFA, MUFA or PUFA with TFA acids is shown in table 5. TFA increase LDL-C levels and decrease HDL-C levels. Of note TFA increase LDL-C even when substituting for SFA. There appears to be a nearly linear relationship between TFA intake and LDL-C concentration, but this relationship does not seem to exist between TFA intake and HDL-C (89). HDL-C seems to be lowered significantly by TFA only when intake is >2% to 4% of the total energy intake (89). TFA also increases TG and Lp(a) levels (88). Additionally, dietary TFA increases small dense LDL and the increase correlates with the quantity of TFA in the diet (90). 

 

Table 5. Effect on Lipids of Replacing Various Fatty Acids with Trans Fatty Acids

Dietary Component

LDL-C (mg/dL)

HDL-C (mg/dL)

SFA

2.0

-2.0

PUFA

11.5

-1.3

MUFA

9.5

-1.5

SFA- saturated fatty acids; PUFA- polyunsaturated fatty acids; MUFA- monounsaturated fatty acids. All results are statistically significant (P<0.05) except the increase in LDL-C with SFA replacement. Effects on lipoprotein lipids of replacing 5% of energy from various fatty acids with 5% of the energy from TFA. Table adapted from reference (88).

 

Replacing carbohydrates with TFA results in an increase in LDL-C and apo B and no change in HDL-C, apo AI, or TG levels (63).

 

RUMINANT TRANS FATTY ACID

 

A key question now that TFA derived from partial hydrogenation of vegetable fat in the diet have been markedly reduced is whether ruminant derived TFA which are present in milk, butter, cheese, and beef have harmful effects similar to industrial created TFA. It is important to note that ruminant derived TFA have a different composition with ruminant TFA being enriched in vaccenic acid, which is the predominant TFA, and conjugated linoleic acid (89,91). Also the quantities of ruminant TFA ingested is much lower than the quantities of industrial TFA ingested (89). In an analysis of a large number of studies of the effect of ruminant and industrial TFA on lipid levels it was observed that the effect of ruminant TFA on LDL-C and HDL-C was similar but slightly less than that of industrial TFA (the difference was not significant) (91). Whether the low quantities of ruminant TFA in the diet will influence the risk of CVD is unknown (89) but a meta-analysis of 4 observational trials did not find a link between ruminant-TFA intake (increments ranging from 0.5 to 1.9 g/day) and the risk of CHD (RR=0.92; CI 0.76-1.11; P=0.36) (92). Another meta-analysis also did not find a link between ruminant TFA and CVD (34). 

 

MECHANISM FOR THE LIPID EFFECTS OF TRANS FATTY ACIDS

 

The mechanism for the increase in LDL-C levels by dietary TFA is thought to be due to decreased LDL-Apo B catabolism without a change in LDL-Apo B production (87,93). The decrease in HDL-C induced by TFA has been attributed to an increase in HDL Apo A-I catabolism without a significant change in HDL apoA-1 production rate (87,93). Additionally, TFA increases CETP activity which could increase the transfer of cholesterol esters from HDL to LDL thereby contributing to the decreased HDL-C levels and increased LDL-C levels (94).

 

DIETARY CHOLESTEROL

 

The primary food sources of dietary cholesterol are egg yolks, shrimp, beef, pork, poultry, cheese, and butter with the top five food sources being eggs and mixed egg dishes, chicken, beef, burgers, and cheese (table 6) (95). In the US the typical cholesterol intake varies from 50 to 400mg per day with a mean of 293 mg/day (348 mg/day for men and 242 mg/day for women) (96).

 

Table 6. Cholesterol Content of Food

Food

mg per 100 grams

Egg

373

Butter

215

Shrimp

125

Cheese

108

Beef

90

Chicken

88

Pork

80

Ice Cream

47

 

Effect of Dietary Cholesterol on Cardiovascular Disease

 

In reviews of prospective observational studies an association between dietary cholesterol and CVD has not been clearly demonstrated with some studies reporting an association and others no association (97,98). Most of these studies did not adjust for the amount and types of fatty acids consumed, which could influence the results as foods containing large amounts of cholesterol are also rich in SFA. Dietary cholesterol was not associated with cardiovascular risk among >80,000 nurses and 43,000 male health care professionals after adjusting for energy intake, PUFA, trans fatty acid, and SFA intake (99,100).

 

Most foods that contain cholesterol also contain significant amounts of SFA. An exception are eggs which contain significant amounts of cholesterol and only small amounts of SFA (95). It is therefore of interest to examine the effect of egg consumption on CVD. In an analysis of 7 cohort studies no association between egg intake and coronary heart disease was observed and egg intake may be associated with a reduced risk of stroke (101). A recent meta-analysis of 23 prospective studies with 1,415,839 individuals and a median follow-up of 12.28 years also found that increased consumption of eggs was not associated with increased risk of CVD (102). Other meta-analyses and reviews have also not demonstrated a consistent link between eggs and CVD (98,103-105). However, a recent very large meta-analysis with 3,601,401 participants with 255,479 events showed that the consumption of 1 additional 50-g egg daily was associated with a very small increase in CVD risk (pooled relative risk, 1.04; 95% CI 1.00-1.08) (106). Thus, eggs have either no effect or a very small effect on CVD that can be seen only in very large studies.

 

There appears to be no randomized studies of the effect of decreasing cholesterol intake on CVD. Do recognize that the studies of decreasing dietary SFA intake described earlier also result in a decrease in cholesterol intake. Thus, at this time there is very limited data linking dietary cholesterol intake with an increased risk of CVD. 

 

Effect of Dietary Cholesterol on Lipid Levels   

 

In a meta-analysis of fifty-five studies with 2,652 subjects the predicted change in LDL-C levels for an increase of 100 mg dietary cholesterol per day adjusted for dietary fatty acids ranged from 1.90mg/dL to 4.58 mg/dL depending upon the model employed (107). An increase of 200mg dietary cholesterol per day increased LDL-C levels from 3.80mg/dL to 6.96mg/dL. It should be noted that the effect of dietary cholesterol levels is greater the higher the LDL-C level (107). For a baseline LDL-C level of 100, 125, 150, and 175 mg/dL the predicted increase in LDL-C for a change in dietary cholesterol of 100mg is 2.7, 3.6, 4.6, and 5.5 mg/dL respectively (107). While the absolute increase is greater if the LDL-C level is higher the percentage increase is similar. Moreover, cholesterol feeding does not alter number of LDL particles – instead it increases the cholesterol content of the LDL particles leading to the formation of large buoyant LDL (108).

 

The effect of dietary cholesterol on HDL-C levels differs in males and females. In men an increase of 100mg of dietary cholesterol results in a 0.30 to 1.44mg/dL decrease in HDL-C levels while in women this results in a 0.50 to 1.61 increase in HDL-C levels (107). Dietary cholesterol does not impact TG or VLDL cholesterol levels (97). 

 

Approximately 15-25% of the population have an increased response to dietary cholesterol with greater increases in LDL-C levels (i.e., sensitive or hyper-responders), while the majority respond minimally (i.e., non-sensitive or hypo-responders) (109). An intake of 100 mg/day dietary cholesterol leads to a 3-4-fold difference in LDL-C concentration between hyper- and hypo-responders (an increase of 2.84 mg/dL vs. 0.76 mg/dL (110). The mechanism for the increase in cholesterol absorption in hyper-responders is unknown. On average 50% (typical range 40-60%) of dietary cholesterol is absorbed but this varies from person to person (111). A high-cholesterol diet leads to significant increases in non-HDL-C levels in insulin-sensitive individuals but not in lean or obese insulin-resistant subjects whereas HDL-C levels increased in all 3 groups (112). The above observations demonstrate the variable response of lipid and lipoprotein levels that can occur in response to dietary manipulations and emphasize how the response of an individual can be variable.

 

MECHANISM FOR THE INCREASE IN LDL-C

 

The increase in LDL-C levels by dietary cholesterol is due to a decrease in hepatic LDL receptors (111). Cholesterol absorbed by the small intestine is packaged into chylomicrons which deliver dietary cholesterol to the liver (111). This increases hepatic cholesterol levels which down-regulates the expression of LDL receptors leading to a decrease in the clearance of LDL from the circulation (111). Additionally, the decrease in LDL receptors could result in an increase in the conversion of intermediate density lipoproteins to LDL rather than clearance by the liver (i.e., LDL production is enhanced).

 

DIETARY CARBOHYDRATES

 

Carbohydrates (CHO) can be divided into high-quality CHO, for example fruits, legumes, vegetables, and whole grains, or low-quality CHO, which include refined grains (such as white bread, white rice, cereal, crackers, and bakery desserts), starches (potatoes), and added sugars (sugar-sweetened beverages, candy). The high-quality CHO are typically enriched in fiber and have a low glycemic index/glycemic load (i.e., are slowly absorbed and thus do not rapidly increase plasma glucose levels). The low-quality CHO have a high glycemic index and load and rapidly increase plasma glucose levels.

 

Effect of Dietary Carbohydrates on Cardiovascular Disease

 

OBSERVATIONAL STUDIES

 

When SFA is replaced by CHO there is no reduction in CVD whereas replacement of SFA with high quality CHO may be beneficial (27,37,38). A study by Jakobsen and colleagues found that replacing SFA with CHO with a low-glycemic index value is associated with a lower risk of myocardial infarction whereas replacing SFA with CHO with a high-glycemic index values is associated with a higher risk of myocardial infarction (113). Meta-analyses and reviews of the association of glycemic index with CVD have varied with some showing an association of low glycemic index with CVD and others reporting no link (114,115). Two very large studies found that a diet with a high glycemic index was associated with an increased risk of cardiovascular disease (116,117). It should be noted that in the largest study the relative risk for CVD was relatively modest (RR 1.15; 95% CI 1.11-1.19) (117). An increase in cardiovascular morbidity and mortality was associated with an increase in added sugar intake (118-121). Hazard ratios were 1.30 (95% CI- 1.09-1.55) and 2.75 (95% CI-1.40-5.42), respectively, comparing participants who consumed 10.0% to 24.9% or 25.0% or more calories from added sugar with those who consumed less than 10.0% of calories from added sugar (118). Additionally, in the Health Professionals Follow-up Study participants in the top quartile of sugar-sweetened beverage intake had a 20% higher relative risk of coronary heart disease than those in the bottom quartile (RR=1.20; 95% CI- 1.09-1.33) after adjustment for multiple risk factors (122).

 

RANDOMIZED CONTROLLED TRIALS

 

Three of the randomized trials described above in the SFA and CVD section provide information on the role of CHO on CVD. The British Medical Research Council studied 252 men after a myocardial infarction aiming to reduce total fat from 41% to 22% of calories and maintaining total fat at 41% in the control group (41). The type of fat was similar in the high- and low-fat groups, mainly saturated fat from dairy products and meat. It is likely that the decrease in fat calories was substituted by an increase in CHO calories. The type of CHO that replaced the SFA was not specified but the authors indicated that there was a marked increase in sugar intake in the low- fat diet group. There was no difference between the two groups in cardiovascular events during the 5 years of the trial. The DART study decreased SFA which were substituted with PUFA and CHO (50). During the 2-year trial cardiovascular events were similar in the decreased SFA vs. PUFA and CHO group. Finally, the Women’s Health Initiative trial randomized 19,541 postmenopausal women 50-79 years of age to the diet intervention group and 29,294 women to usual dietary advice (52). The goal in the diet intervention group was to reduce total fat intake to 20% of calories and increase intake of vegetables/fruits to 5 servings/day and grains to at least 6 servings/day (i.e., CHO}. Fat intake decreased by 8.2% of energy intake in the intervention vs the comparison group, with small decreases in SFA (2.9%), MUFA (3.3%), and PUFA (1.5%) fat with increased consumption of CHO. The dietary intervention did not significantly decrease CVD even though the CHO recommended was high quality CHO. These randomized studies do not provide support for a benefit of substituting CHO for fat in reducing CVD. Of particular note is the Women’s Health Initiative which decreased fat intake and increased high quality CHO and observed no cardiovascular benefits in contrast to the results of observational studies.

 

Effect of Dietary Carbohydrates on Lipids

 

Replacing SFA, MUFA, or PUFA with CHO results in an increase in TGs and a decrease in HDL-C levels (60,63). Replacing SFA with CHO results in a decrease in LDL-C while replacing MUFA or PUFA with CHO results in an increase in LDL-C (see tables 2 and 4) (60,63). In addition, dietary CHO increases the quantity of small dense LDL particles (123). The consumption of moderate amounts of fructose or sucrose (40-80 grams/day) in healthy young men was sufficient to increase small dense LDL levels (124). The effect of increasing dietary CHO on Lp(a) levels has been variable (62).

 

Conversely, decreasing CHO in the diet and adding fat results in an increase in LDL-C and HDL-C levels and a decrease in TG levels (125). In a meta-analysis of eleven randomized controlled trials with 1,369 participants comparing low fat/high CHO diet to high fat/low CHO diet it was found that the high fat/low CHO led to an increase in LDL-cholesterol (6.24mg/dL; 95 % CI 0.12- 12.9) and HDL-C (5.46mg/dL; 95% CI 3.51- 7.41) compared with subjects on the low fat/high CHO diets (126). The high fat/low CHO decreased TG levels (-22.9mg/dL; 95 % CI -13.4- -32.6 (126). Another meta-analysis of 23 randomized controlled trials also found that a high fat/low CHO diet increased LDL-C and HDL-C levels and decreased TG levels (127). These studies nicely demonstrates that a high fat diet will increase LDL-C and HDL-C levels while a high CHO diet will increase TG levels and decrease HDL-C levels.

 

COMPARISON OF DIFFERENT CARBOHYDRATES ON LIPIDS

 

A meta-analysis of twenty-eight randomized controlled trials comparing low- with high glycemic index diets (1,272 participants) reported that low glycemic index diets significantly decreased LDL-C levels by 6.2mg/dL; P < 0.0001) with no effect on HDL-C or TGs (128). The decrease in LDL-C was related to the amount of fiber and/or phytosterols in the low glycemic diet (see Fiber and Plant Sterols/Stanols section below).

 

High fructose corn syrup (HFCS) has become a major source of fructose intake (HFCS made for beverages contains 55% fructose and 45% glucose). Because sucrose and HFCS are major contributors to total CHO intake there has been interest in the effect of fructose, glucose, and sucrose on lipid levels. In a comparison of isocalorically substituting starch for glucose, fructose, or sucrose there were no difference in TG levels but there was a decrease in LDL-C (approximately 7.8mg/dL) (129).

 

A meta-analysis by Te Morenga and colleagues examined the effect of the addition of sugar on lipid levels. In studies where energy intake was isocaloric, sugar intake increased TG levels by 11.7mg/dL, LDL-C by 6.6mg/dL, and HDL-C by 0.8mg/dL (130). In a similar meta-analysis by Fattore and colleagues an isocaloric substitution of free sugars for complex CHO increased TGs by 8.3mg/dL, LDL-C by 7.1mg/dL, and HDL-C by 1.3mg/dL (131). The increase in TG and LDL-C levels were larger in the trials where greater amounts of free sugar were employed.

 

In a meta-analysis of adding fructose to the diet there was no significant effect on fasting TG levels at dietary fructose < 100 grams per day but at higher amounts fructose increased fasting TG levels (132). Fructose is more likely to have adverse effects on lipids when intake is high and/or when caloric excess is present. For example, in young healthy individuals, a 2-week intervention with 25% of energy requirements as HFCS or fructose sweetened beverages resulted in significant increases in fasting LDL-C, small dense LDL particles, non-HDL-C, apo B, and HDL-C and postprandial TGs (133). High quantities of glucose did not affect LDL-C, non-HDL-C, Apo B, HDL-C, or postprandial TG levels but did increase fasting TG levels (133).

 

Thus, the effect of CHO on lipids can vary depending upon the particular type of CHO studied (table 7). In the case of glycemic index (complex CHO) and starch vs sugar some of the difference in lipid response could be due to other dietary constituents (i.e., fiber, phytosterols).

 

Table 7. Summary of the Effect of Different Carbohydrates on Lipid and Lipoproteins

Comparisons

Effect on Lipids and Lipoproteins

Low GI vs. High GI

High GI increases LDL-C

Sugar vs. Starch

Sugar increases LDL-C

Sugar vs. Complex CHO

Sugar increases LDL-C and TGs

Fructose vs. Glucose

Fructose increases LDL-C and HDL-C and postprandial TGs

Sugar- sucrose, glucose, or fructose

 

MECHANISM OF THE EFFECTS OF CARBOHYDRATES ON LIPIDS

 

Dietary CHO promote hepatic de novo fatty acid synthesis by providing substrate for fatty acid synthesis (Figure 1). This is particularly the case when there is caloric excess. Additionally, the glucose provided by dietary CHO stimulates insulin secretion which also increases hepatic fatty acid synthesis. The increase in fatty acid synthesis in the liver enhances TG synthesis which promotes VLDL formation and secretion leading to an increase in plasma TG levels. 

 

Figure 1. Carbohydrates stimulate VLDL production by stimulating de novo fatty acid synthesis.

 

Fructose is more potent at increasing de novo fatty acid synthesis than glucose. Small quantities of fructose in the diet are metabolized in the small intestine to glucose and organic acids and do not affect systemic metabolism while high quantities of fructose can escape intestinal metabolism and are delivered to the liver (134). In the liver fructose but not glucose activates SREBP1c and ChREBP leading to the increased expression of the genes that synthesize fatty acids stimulating hepatic lipogenesis (134,135). Additionally, fructose metabolism in the liver is not inhibited providing an unlimited supply of fructose carbons for lipogenesis. In contrast, the first steps in glucose metabolism can be inhibited and thus the utilization of glucose for lipogenesis is regulated (134). In addition, fructose inhibits fatty acid oxidation whereas glucose does not (135). These differences in the metabolism of fructose and glucose in the liver explain the increased ability of fructose to stimulate hepatic lipogenesis and the enhanced formation and secretion of VLDL. In the addition to increased VLDL production fructose does not stimulate the secretion of insulin, which plays a key role in stimulating lipoprotein lipase activity and the clearance of TG rich lipoproteins. The failure of dietary fructose to induce an increase in lipoprotein lipase activity may lead to a decrease in the clearance of TG rich lipoproteins compared to dietary glucose, which stimulates insulin secretion.

 

The elevation in TG rich lipoproteins in turn may have effects on other lipoproteins (25) (Figure 2). Specifically, cholesterol ester transfer protein (CETP) mediates the equimolar exchange of TGs from TG rich VLDL and chylomicrons for cholesterol from LDL and HDL (25). The increase in TG rich lipoproteins per se leads to an increase in CETP mediated exchange, increasing the TG content and decreasing the cholesterol content of both LDL and HDL particles. This CETP-mediated exchange underlies the commonly observed reciprocal relationship of low HDL-C levels when TG levels are high and the increase in HDL-C when TG levels decrease. The TG on LDL and HDL are then hydrolyzed by hepatic lipase and lipoprotein lipase leading to the production of small dense LDL and small HDL particles.

 

Figure 2. The effect of hypertriglyceridemia on LDL and HDL.

 

DIETARY PROTEIN

 

Effect of Dietary Protein on Cardiovascular Disease

 

In a meta-analysis of 10 studies with 425 ,781 participants intake of plant protein was associated with a decrease in cardiovascular mortality (136). Other meta-analyses have also found that intake of plant proteins was associated with a lower risk of cardiovascular mortality (137-139). In some but not all studies animal protein intake increased the risk of cardiovascular mortality (136-139). The differences in outcomes observed between plant and animal proteins could be due to increased intake of SFA with animal proteins and increased fiber and phytosterol intake with plant proteins. 

 

Effect of Dietary Protein on Lipids

 

Because a high protein diet is often associated with an increase in SFA intake it is important to control for this variable in determining the effect of dietary protein on lipid levels. In a meta-analysis of a high vs. low protein diets in individuals on a low-fat diet no difference in LDL-C, HDL-C, or TG levels were observed (140). In another meta-analysis of 24 trials with 1,063 participants that compared isocaloric diets matched for fat intake but with differences in protein and CHO  intakes no differences in LDL-C and HDL-C levels were observed but TG levels were decreased in the high protein diet group (-20.2mg/dL) (141). Greater weight loss and decreased CHO intake in the high protein diet group likely contributed to the decrease in TGs. In a meta-analysis where fat intake was not controlled the high protein diet was associated with an increase in HDL-C levels and a decrease in TG levels (142). It is obviously difficult to determine the effect of dietary protein on lipid levels as other dietary constituents are changing (SFA, CHO) and secondary effects induced by changes in protein intake (weight loss) could influence lipid levels.

 

DIETARY FIBER

 

Dietary fiber are non-digestible carbohydrates including non-starch polysaccharides, cellulose, pectins, hydrocolloids, fructo-oligosaccharides and lignin. Fiber is found mostly in fruits, vegetables, whole grains, nuts, seeds, psyllium seeds, beans, and legumes. There are two main types of dietary fiber; soluble and insoluble. The main sources of soluble fiber are fruits and vegetables and insoluble fiber are cereals and whole-grain products. Most high fiber foods contain both soluble and insoluble fiber. A summary of the fiber content of some foods is shown in tables 8-11.

 

Table 8. Fiber Content of Selected Vegetables

Vegetables

 

Serving

Size

Total Fiber/ Serving (g)

Soluble Fiber/ Serving (g)

Insoluble Fiber/ Serving (g)

Cooked vegetables

Turnip

½ cup

4.8

1.7

3.1

Peas, green, frozen

½ cup

4.3

1.3

3.0

Okra, frozen

½ cup

4.1

1.0

3.1

Potato, sweet, flesh

½ cup

4.0

1.8

2.2

Brussels sprouts

½ cup

3.8

2.0

1.8

Asparagus

½ cup

2.8

1.7

1.1

Kale

½ cup

2.5

0.7

1.8

Broccoli

½ cup

2.4

1.2

1.2

Carrots, sliced

½ cup

2.0

1.1

0.9

Green beans, canned

½ cup

2.0

0.5

1.5

Beets, flesh only

½ cup

1.8

0.8

1.0

Tomato sauce

½ cup

1.7

0.8

0.9

Corn, whole, canned

½ cup

1.6

0.2

1.4

Spinach

½ cup

1.6

0.5

1.1

Cauliflower

½ cup

1.0

0.4

0.6

Turnip

½ cup

4.8

1.7

3.1

Raw vegetables

Carrots, fresh

1, 7 ½ in. long

2.3

1.1

1.2

Celery, fresh

1 cup chopped

1.7

0.7

1.0

Onion, fresh

½ cup chopped

1.7

0.9

0.8

Pepper, green, fresh

1 cup chopped

1.7

0.7

1.0

Cabbage, red

1 cup

1.5

0.6

0.9

Tomato, fresh

1 medium

1.0

0.1

0.9

Mushrooms, fresh

1 cup pieces

0.8

0.1

0.7

Cucumber, fresh

1 cup

0.5

0.2

0.3

Lettuce, iceberg

1 cup

0.5

0.1

0.4

Adapted from Anderson JW. Plant Fiber in Foods. 2nd ed. HCF Nutrition Research Foundation Inc, PO Box 22124, Lexington, KY 40522, 1990.

 

Table 9. Fiber Content of Selected Legumes

Legumes (cooked)

Serving Size

Total Fiber/ Serving (g)

Soluble Fiber/ Serving (g)

Insoluble Fiber/ Serving (g)

Kidney beans, light red

½ cup

7.9

2

5.9

Navy beans

½ cup

6.5

2.2

4.3

Black beans

½ cup

6.1

2.4

3.7

Pinto beans

½ cup

6.1

1.4

4.7

Lentils

½ cup

5.2

0.6

4.6

Black-eyed peas

½ cup

4.7

0.5

4.2

Chick peas, dried

½ cup

4.3

1.3

3

Lima beans

½ cup

4.3

1.1

3.2

Adapted from Anderson JW. Plant Fiber in Foods. 2nd ed. HCF Nutrition Research Foundation Inc, PO Box 22124, Lexington, KY 40522, 1990.

 

Table 10. Fiber Content of Selected Fruits

Fruits

Serving

Size

Total Fiber/ Serving (g)

Soluble Fiber/ Serving (g)

Insoluble Fiber/ Serving (g)

Apricots, fresh w/skin

4

3.5

1.8

1.7

Raspberries, fresh

1 cup

3.3

0.9

2.4

Figs, dried

1 ½

3

1.4

1.6

Mango, fresh

½ small

2.9

1.7

1.2

Orange, fresh

1 small

2.9

1.8

1.1

Pear, fresh, w/skin

½ large

2.9

1.1

1.8

Apple, red, fresh w/skin

1 small

2.8

1

1.8

Strawberries, fresh

1 ¼ cup

2.8

1.1

1.7

Plum, red, fresh

2 medium

2.4

1.1

1.3

Applesauce, canned

½ cup

2

0.7

1.3

Apricots, dried

7 halves

2

1.1

0.9

Peach, fresh, w/skin

1 medium

2

1

1

Kiwifruit, fresh

1 large

1.7

0.7

1

Prunes, dried

3 medium

1.7

1

0.7

Grapefruit, fresh

½ medium

1.6

1.1

0.5

Blueberries, fresh

¾ cup

1.4

0.3

1.1

Cherries, black, fresh

12 large

1.3

0.6

0.7

Banana, fresh

½ small

1.1

0.3

0.8

Melon, cantaloupe

1 cup cubed

1.1

0.3

0.8

Watermelon

1 ¼ cup cubed

0.6

0.4

0.2

Grapes, fresh w/skin

15 small

0.5

0.2

0.3

Raisins, dried

2 tbsp

0.4

0.2

0.2

Adapted from Anderson JW. Plant Fiber in Foods. 2nd ed. HCF Nutrition Research Foundation Inc, PO Box 22124, Lexington, KY 40522, 1990.

 

Table 11. Fiber Content of Grains

Food

Serving

Size

Total Fiber/ Serving (g)

Soluble Fiber/ Serving (g)

Insoluble Fiber/ Serving (g)

Wheat bran

½ cup

12.3

1.0

2.7

Barley, pearled, cooked

½ cup

3.0

0.8

2.2

Oatmeal, dry

⅓ cup

2.7

1.4

11.3

Bread, pumpernickel

1 slice

2.7

1.2

1.5

Wheat flakes

¾ cup

2.3

0.4

1.9

Bread, rye

1 slice

1.8

0.8

1.0

Bread, whole wheat

1 slice

1.5

0.3

1.2

Rice, white, cooked

½ cup

0.8

trace

0.8

Bread, white

1 slice

0.6

0.3

0.3

Adapted from Anderson JW. Plant Fiber in Foods. 2nd ed. HCF Nutrition Research Foundation Inc, PO Box 22124, Lexington, KY 40522, 1990.

 

Effect of Dietary Fiber on Cardiovascular Disease

 

Several meta-analyses have demonstrated that an increase in total fiber, soluble fiber, and insoluble fiber are associated with a decrease in cardiovascular events (143-148). The greater the intake of fiber the greater the reduction in risk of cardiovascular events.  

 

Effect of Dietary Fiber on Lipids

 

In a meta-analysis of randomized controlled trials the effect of fiber on lipid levels was evaluated (149). Increased dietary fiber decreased total cholesterol (TC) (−7.8mg/dL; 95% CI −13.3 to −2.3), LDL-C (−5.5mg/dL; 95% CI −8.6 to −2.3), and HDL-C levels ( −1.17mg/dL; 95% CI −2.34 to −0.39) (149,150), There was no change in TG levels. A meta-analysis of randomized controlled studies of whole-grain foods vs non-whole-grain foods found that the whole-grain diet lowered LDL-C (-3.51mg/dL; P < 0.01) and TC levels (-4.68mg/dL; P < 0.001) compared with the non-whole grain foods (151). HDL-C and TG levels were not significantly altered by the whole grain diet. Moreover, 3.4 g of psyllium (Metamucil), a soluble fiber, decreased LDL-C with no significant effects on HDL-C or TGs (152,153). In a meta-analysis of 28 randomized trials psyllium lowered LDL by 12.9mg/dL (P < 0.00001) (154). A mean reduction in LDL-C concentrations of about 1.1 mg/dL can be expected for each g of water-soluble fiber in the diet (155,156).

 

MECHANISM OF EFFECT OF FIBER ON LDL-C

 

Fiber is thought to decrease cholesterol absorption by the small intestine (157,158). This leads to a decrease in cholesterol content of chylomicrons and a reduction in the delivery of cholesterol to the liver. The decrease in cholesterol in the liver upregulates LDL receptors resulting in a decrease in plasma LDL-C levels. Fiber may also decrease small intestinal absorption of bile acids which will lead to the increased utilization of hepatic cholesterol for the synthesis of bile acids (159). This will also decrease hepatic cholesterol levels inducing an increase in the expression of LDL receptors lowering plasma LDL-C levels. Finally, colonic fermentation of dietary fiber with production of short-chain fatty acids, such as acetate, propionate, and butyrate, is postulated to inhibit hepatic cholesterol synthesis contributing to a decrease in LDL-C levels (159). 

 

PLANT STEROLS AND STANOLS (PHYTOSTEROLS)

 

Plant sterols and plant stanols (phytosterols) are naturally occurring constituents of plants and are found in vegetable oils, such as corn oil, soybean oil, and rapeseed oil and cereals, nuts, fruits, and vegetables. The intake of plant sterols and stanols is about 200–400 mg/day. The most commonly occurring phytosterols in the human diet are β-sitosterol, campesterol, and stigmasterol. Higher intakes can be achieved by consuming a vegetable-based diets such as a vegetarian diet (400-800mg/day) or by consuming food products enriched with plant sterols or stanols (for example margarines or yogurt). If using foods enriched in phytosterols it is best to take them with main meals to enhance their effectiveness. High doses of phytosterols can affect the absorption of fat-soluble vitamins. The plant sterol and stanol content of different foods is shown in table 12.

 

Table 12. Plant Sterol and Stanol Contents in Different Foods

Food item

Plant Sterols

(mg/100 g)

Plant Stanols

(mg/100 g)

Vegetable oils

Corn oil

686-952

23-33

Rapeseed oil (canola oil)

250-767

2-12

Soybean oil

221-328

7

Sunflower oil

263-376

4

Olive oil

144-193

0.3-4

Palm oil

60-78

Traces

Cereals

Corn

66-178

-

Rye

71-113

12-22

Wheat

45-83

17

Barley

80

2

Millet

77

-

Rice

72

3

Oats

35-61

1

Vegetables

Broccoli

39

2

Cauliflower

18-40

Traces

Carrot

12-16

Traces

Lettuce

9-17

0.5

Potato

7

0.6

Tomato

7

1

Fruits and berries

Avocado

75

0.5

Passion fruit

44

Not detected

Raspberry

27

0.2

Orange

24

Not detected

Apple

12-18

0.8

Banana

12-16

Not detected

Adapted from Piironen V and Lampi AM (160)

 

Effect of Phytosterols on Cardiovascular Disease

 

There is minimal data on the effect of phytosterols on cardiovascular events. From the effect on LDL-C levels one would anticipate that phytosterols would reduce CVD.

 

Effect of Phytosterols on Lipids

 

Plant sterols or plant stanols at a dose of 3 grams per day lowers LDL-C by approximately 12% (161). Higher doses do not dramatically further lower LDL-C levels and lower doses have less effect on LDL-C (for example 2 grams/day lowers LDL-C by 8%) (161).  HDL-C levels are not affected by plant sterols or stanols but TG levels decrease modestly (~6%) with a greater absolute reduction in individuals with high TG level (percent change is the same) (162). To achieve these high doses consuming food products enriched is phytosterols is necessary.

 

MECHANISM OF EFFECT OF PHYTOSTEROLS ON LDL-C

 

Plant sterols or plant stanols reduce LDL-C levels by competing with cholesterol for incorporation into micelles in the gastrointestinal tract, resulting in decreased cholesterol absorption (163). This leads to the decreased delivery of cholesterol to the liver and the up-regulation of LDL-receptor expression lowering LDL-C levels.

 

SUMMARY OF THE EFFECT OF DIETARY CONSTITUENTS ON LIPID LEVELS

 

A summary of the major effects of dietary constituents on lipid levels is shown in table 13, typically under isocaloric feeding conditions in short-term feeding studies. Dietary SFA, TFA, and cholesterol increase LDL-C levels whereas CHO increases TG levels. MUFA, PUFA, fiber and phytosterols decrease LDL-C and TFA decrease HDL-C levels.

 

Table 13. Summary of the Effect of Dietary Constituents on Lipid and Lipoproteins

SFA

Increase LDL-C and modest increase HDL-C

MUFA and PUFA

Decrease LDL-C

TFA

Increase LDL-C and decrease HDL-C

Cholesterol

Increase LDL-C

CHO

Increase TGs, increase greater with simple sugars particularly fructose

Fiber

Decrease LDL-C

Phytosterols

Decrease LDL-C

 

EFFECT OF SPECIFIC FOODS ON CARDIOVASCULAR DISEASE

 

There are a large number of observational trials linking various foods with either an increased or decreased risk of CVD. A large meta-analysis by Micha et al reported that fruits, vegetables, beans/legumes, nuts/seeds, whole grains, fish, yogurt, fiber, seafood omega-3 fatty acids, polyunsaturated fats, and potassium were associated with a decreased risk of CVD while unprocessed red meats, processed meats, sugar-sweetened beverages, and sodium were associated with an increased risk of CVD (164). A similar meta-analysis by Bechthold et al found that whole grains, vegetables and fruits, nuts, and fish consumption were associated with a decrease in CVD while red meat, processed meat, and sugar sweetened beverage consumption was associated with an increase in CVD (165). Note, as discussed in the introduction, observational studies have limitations and cannot be assumed to indicate cause and effect. Additional one can find other meta-analyses that reach different conclusions than the results described above. For example, a meta-analysis by Zeraatkar et al and a meta-analysis by Vernooij et al reached the conclusion that meat and processed meat were not associated with a significant increase in CVD (20,166). Thus, one needs recognize that while these studies can suggest beneficial and harmful effects of eating certain foods more definitive studies are required to be certain. For a detailed analysis of the limitations of observational dietary studies see articles by Ioannidis and Nissen (1,2).

 

Only a single randomized trial has examined the effect of specific foods on CVD events. The DART trial randomized men with an acute myocardial infarction to at least two weekly portions (200-400 g) of fatty fish (mackerel, herring, kipper, pilchard, sardine, salmon, or trout) (n=1015) or no dietary advice (n=1018) (50). After approximately 2 years total mortality was significantly lower (RR 0.71; CI 0.54-0.93) in the fish advice group than in the no fish advice group, due to a reduction in ischemic heart disease deaths. There were no significant differences in ischemic heart disease events (RR 0.84; CI 0.66-1.07). In a separate portion of the DART trial there was also a group of men with an acute myocardial infarction randomized to increased intake of cereal fiber (18 grams/day) (n=1017) vs. no dietary advice (n=1016). No reduction in cardiovascular events was seen in the cereal fiber group.

 

Clearly addition randomized trials are required to determine the true benefits of specific foods on cardiovascular events.

 

EFFECT OF SPECIFIC FOODS ON LIPID LEVELS

 

In contrast to the paucity of randomized controlled trials on the effect of specific foods on cardiovascular disease there are an abundance of studies on the effect of specific foods on lipid and lipoprotein levels. Given the large number of studies in many instances I will cite the results of meta-analyses to provide the reader with the typical effects that are observed. It should be noted that the effect of specific foods on lipid and lipoprotein levels tend to be small and therefore the results can be inconsistent from study to study.

 

Nuts and Seeds

 

The most consumed edible tree nuts are almonds, hazelnuts, walnuts, pistachios, pine nuts, cashews, pecans, macadamias, and Brazil nuts. Peanuts are botanically groundnuts or legumes, and are widely considered to be part of the nut food group. Nuts are generally consumed as snacks (fresh or roasted), in spreads (peanut butter, almond paste), or as oils or baked goods. Seeds come in all different sizes, shapes and colors. Popular seeds include flax, pumpkin, sunflower, chia, sesame, and mustard seeds.

 

Nuts and seeds are rich in MUFAs, such as oleic acid and in PUFAs, such as linoleic acid and alpha-linolenic acid (ALA). They also contain small amounts of SFA. Almonds, cashews, hazelnuts, pistachios and macadamian nuts have a high MUFA content (>50%) content when compared with other nuts. For other nuts (e.g., Brazil nuts, pine nuts, and walnuts) the PUFA content is high (>50%), while peanuts and pecans have been found to contain relatively high levels of both MUFA and PUFA (table 14). Nuts are a good source of dietary fiber, ranging from 4-11 g/100 g and phytosterols.

 

Table 14. Nutrient Composition of Nuts

Nuts

 

PUFA

(g/100 g)

MUFA

(g/100 g)

SFA

(g/100 g)

Fiber

(g/100 g)

Walnuts

47.2

8.9

6.1

6.7

Peanuts

15.6

24.4

6.3

8.8

Pistachios

13.7

23.8

5.6

10.3

Almonds

12.3

31.6

3.8

12.5

Hazelnuts

7.9

45.7

4.5

9.7

Cashews

7.8

23.8

7.8

3.3

Pecans

21.6

40.8

6.2

9.6

Macadamias

1.5

58.9

12.1

8.6

 

Consumption of nuts and seeds lower TC and LDL-C levels in healthy subjects or patients with moderate hypercholesterolemia (167-172). Nuts had no significant or minimal effect on increasing HDL-C. The benefits of nuts and seeds vary depending on the type, nutrient composition, and quantity of nuts and seeds consumed. Studies have noted that the estimated cholesterol lowering effect of nuts was greater in individuals with higher initial values of LDL-C and in those with a lower baseline BMI (169).

 

Walnuts: A meta-analysis on the effect of walnuts on lipid levels that included 365 participants showed a decrease in LDL-C (9.2 mg/dL), while HDL-C or TG were not significantly affected (173). In another meta-analysis that analyzed 1,059 participants with a walnut enriched diet LDL-C was lowered by 5.5 mg/dL (174).

 

Almonds: A meta-analysis of 15 studies with 534 participants found that almonds decreased LDL cholesterol (5.8 mg/dL; 95% CI: -9.91, -1.75 mg/dL) and apo B (6.67 mg/dL; 95% CI: -12.63, -0.72 mg/dL) (175). Triglycerides, apo A1, and lipoprotein (a) showed no differences.

 

Pistachio nuts: A meta-analysis of twelve randomized studies reported that pistachio nuts decreased LDL-C -3.82 mg/dL (95% CI, -5.49 to -2.16) and TG -11.19 mg/dL (95% CI, -14.21 to -8.17) levels without effecting HDL-C levels (176).

 

A meta-analysis by Houston et al analyzed the effect of a variety of different nuts on lipid levels (table 15) (177). They found that in general nuts lowered LDL-C and minimally lowered TG levels but had no effect on HDL-C levels. A meta-analysis found that whole flaxseed reduced TC and LDL-C by 6 and 8 mg/dL, respectively (178). Thus, both nuts and seeds lower LDL-C levels.

 

Table 15. Effect of Nuts on Lipid Levels

 

Number of analyses

Number of participants

Effect estimate (mmol/L)

95% CI

LDL Cholesterol

Almond

32

2439

-0.15 [-0.22, -0.08]

Brazil nut

4

307

-0.30 [-0.70, 0.11]

Cashew nut

3

432

 0.02 [-0.12, 0.16]

Hazelnut

6

374

-0.01 [-0.15, 0.12]

Macadamia

6

410

-0.11 [-0.27, 0.04]

Mixed nuts

10

791

 0.04 [-0.06, 0.14]

Peanut

10

1021

 0.08 [-0.04, 0.20]

Pecan

6

295

-0.23 [-0.46, 0.00]

Pistachio

12

736

-0.15 [-0.30, 0.00]

Walnut

35

2582

-0.12 [-0.18, -0.06]

Triglycerides

Almond

32

2439

-0.02 [-0.05, 0.02]

Brazil nut

4

307

 0.04 [-0.54, 0.63]

Cashew nut

3

432

-0.02 [-0.11, 0.07]

Hazelnut

5

313

 0.11 [-0.02, 0.25]

Macadamia

5

342

-0.10 [-0.21, 0.00]

Mixed nuts

11

888

-0.01 [-0.07, 0.06]

Peanut

10

1021

-0.09 [-0.16, -0.02]

Pecan

6

295

-0.11 [-0.24, 0.03]

Pistachio

9

498

-0.12 [-0.21, -0.03]

Walnut

35

3109

-0.09 [-0.12, -0.06]

Table based on data from a meta-analysis by Houston et al (177). To convert mmol/L cholesterol to mg/dL multiply by 39 and to convert mmol/L triglycerides to mg/dL multiply by 88.

 

Whole Grains

 

Whole grains include barley, brown rice, buckwheat, bulgur (cracked wheat), millet, oatmeal, and wild rice. Whole grains contain ~80% more dietary fiber than refined grains, as the latter are milled, a process that removes bran and germ. Refined grains include white flour, white rice, white bread, and corn flower. Health benefits ascribed to whole grains are mainly due to the presence of fiber and bran. A meta-analysis of fifty-five trials with 3900 participants comparing various grains found that oat bran was the most effective intervention strategy for lowering LDL-C (- 12.5mg/dL; 95% CI – 17.2 to – 7.4mg/dL) compared with control (179). Oats also reduced LDC (- 6.6mg/dL; 95% CI – 10.9 to 2.73mg/dL). Barley, brown rice, wheat and wheat bran were not effective in improving blood lipid levels compared with controls. Another meta-analysis also found that whole-grain oats decreased LDL-C levels (–16.7 mg/dL; P < 0.0001) (180).

 

Soy Protein

 

Soybeans and soy products as well as supplements contain soy proteins. In a meta-analysis of 43 randomized studies with 2,607 participants the decrease in LDL-C levels reductions for soy protein ranged between −4.2 and −6.7 mg/dL (P<0.006) (181). Numerous other meta-analyses have reported similar decreases in LDL-C (182-187).  In addition, soy protein also decreases TG levels (~2-10mg/dL) and increases HDL-C levels (~1-2mg/dL). Soy protein does not affect Lp(a) levels (188). The amount of soy protein that is recommended for lipid lowering is 25–50 grams per day (189).

 

The decrease in LDL-C is due to the indirect effect of soy protein decreasing the intake of animal protein (SFA and cholesterol) and the intrinsic effects of bioactive compounds in soy protein (190). The intrinsic effect of soy protein might be mediated by phyto-estrogens that could increase levels of HDL-C and TG and decrease levels of LDL-C (189).   

 

Garlic

 

Garlic supplements are available in several different forms, including garlic powder, allicin, aged garlic extract, and garlic oil. Several meta-analyses have shown that garlic lowers TC levels with variable effects on LDL-C, HDL-C, and TG (191-198). Some studies find a decrease in LDL-C and others a decrease in TG levels. The longer the duration of treatment and the higher the baseline TC the greater the effect. In one meta-analysis TC was reduced by 17 ± 6 mg/dL and low-density lipoprotein cholesterol by 9 ± 6 mg/dL in individuals with elevated TC levels (>200 mg/dL) if treated for longer than 2 months (191). In another meta-analysis garlic powder and aged garlic extract were more effective in reducing TC levels, while garlic oil was more effective in lowering serum TG levels (192). In a meta-analysis of garlic administration to patients with diabetes TC decreased 16.9mg/dL, LDL decreased 9.7mg/dL, TG decreased 12.4mg/dL, and HDL-C increased 3.19mg/dL (all p=0.001) (199). Lp(a) levels are not altered by garlic (198).The mechanism by which garlic alters lipid levels is unknown.

 

Tea

 

Green tea contains many catechins (e.g., epigallocatechin-3-gallate) that influence lipid metabolism in animal models and have been shown to upregulate LDL receptors in liver and suppress PCSK9 production (200,201). Epigallocatechin gallate may also interfere with the intestinal absorption of lipids (202). Most but not all meta-analyses have shown that drinking green tea or black tea decreases TC and LDL-C levels with no significant effect on HDL-C or TG levels (203-214). The reduction in LDL-C is approximately 5-10mg/dL.

 

Coffee

 

Coffee contains cholesterol-increasing compounds; diterpenes such as cafestol and kahweol (215,216). The amount of these cholesterol increasing compounds in coffee depends on how the coffee is prepared (215,216). Boiling coffee beans extracts diterpenes due to the prolonged contact with hot water resulting in high concentrations in the coffee whereas brewed filtered coffee because of the short contact with hot water and retention of diterpenes by the filter paper has lower concentrations of diterpenes. Instant coffee has very low levels of diterpenes (216). The concentration of the cholesterol-raising compound cafestol is negligible in drip-filtered, instant, and percolator coffee but high in unfiltered coffee such as French press, Turkish, or Scandinavian boiled coffee. Levels of cafestol are intermediate in espresso and coffee made in a Moka pot.

 

A meta-analysis of 18 trials found that the consumption of unfiltered, boiled coffee dose-dependently increased TC and LDL-C concentrations (23 mg/dL and 14 mg/dL, respectively), while consumption of filtered coffee resulted in only small changes (TC increased by 3 mg/dL and no effect on LDL-C concentration) (217). Additionally, decaffeinated coffee had a smaller effect and the increase in cholesterol levels was greatest in individuals with hypercholesterolemia. Thus coffee, depending upon how it is prepared, can increase TC and LDL-C levels.

 

Chocolate and Cocoa

 

Cocoa is the non-fat component of finely ground cocoa beans that is used to produce chocolate. Cocoa is rich in flavanols which are low‐molecular‐weight monomeric compounds, such as epicatechin or complex higher‐molecular‐weight oligomeric and polymeric compounds (218). The flavanol content in cocoa products can vary greatly and is dependent on the crop type, post‐harvest handling practices, and manufacturer processing techniques. The flavanol content of milk and white chocolate is low or even absent (218).

 

In a meta-analysis of 21 studies with 986 participants very small effects on LDL-C and HDL-C levels were observed (LDL-C 2.7mg/dL decrease; HDL-C 1.2mg/dL increase) with no change in TG levels with chocolate and/or cocoa intake (219). In another meta-analysis there was a decrease in TG levels (-8.8mg/dL), an increase in HDL-C (2.3mg/dL), and a non-significant decrease in LDL-C (-10.1mg/dL) (220). In studies where the epicatechin dose was greater than 100mg per day the decrease in LDL-C levels was greater (5.5mg/dL) (219). Another meta-analysis of 19 studies found that LDL decreased by 3.3mg/dL and HDL-C increased by 1.8mg/dL with cocoa intake (221). A meta-analysis of 10 clinical trials with 320 participants that focused on dark chocolate found a 6.23mg/dl decrease in LDL-C with no significant changes in HDL-C and TG (222). Thus chocolate/cocoa causes a small decrease in LDL-C levels. 

 

Alcohol

 

It is recommended that females consume no more than 1 drink per day of alcohol (equivalent to 15 grams per day) and that males consume no more than 2 drinks per day (equivalent to 30 grams per day). Alcohol has a relatively high caloric level (7 calories/gram).

 

EFFECT OF ALCOHOL ON LIPID LEVELS

 

In a meta-analysis of 25 studies with an average consumption of 40.9 grams of alcohol per day HDL-C concentrations increased by 5.1 mg/dL (223). HDL-C levels increased by 0.122- 0.133 mg/dL per gram of alcohol per day. Consuming 30 grams of alcohol a day would therefore increase HDL-C concentrations by approximately 3.99 mg/dL compared with an individual who abstains (an 8.3% increase from pretreatment values). The increase in HDL-C was observed regardless of sex, duration of study, median age, or beverage type but the increase was greater in individuals with baseline HDL-C < 40mg/dL and who were sedentary. As expected apo A1 levels also increased. In a meta-analysis of 35 studies TG concentrations increased by 0.19 mg/dL per gram of alcohol consumed a day (P=0.001) and 5.69 mg/dL per 30 g consumed per day (5.9% increase over baseline) (223). The increase in TG levels was seen regardless of beverage type and appeared to be greater in males than females.

 

In a more recent meta-analysis of 33 studies with 796 participants HDL-C levels were increased by 3.67mg/dL by alcohol intake (224). Apo A1 levels were also increased but there were no significant differences in TC, LDL-C, TG, or Lp(a) with alcohol intake. The greater the consumption of alcohol the greater the increase in HDL-C levels. When the consumption of alcohol was greater than 60 grams per day (4 drinks) TG levels were also increased (24.4mg/dL).

 

In a meta-analysis of 14 studies, comparing 548 beer drinkers and 532 controls TC levels were significantly higher in the beer drinkers compared to controls (difference 3.52 mg/dL; p<0.001) (225). In a meta-analysis of 18 studies, comparing 626 beer drinkers and 635 controls HDL-C levels were higher in the beer drinkers compared to controls (difference 3.63 mg/dL: p<0.001) (225). This increase in HDL-C levels in beer drinkers were seen in both males and females. LDL-C and TG levels were not significantly different between beer drinkers and controls (LDL-C difference -2.85 mg/dL; p = 0.070; TG difference 0.40 mg/dL; p = 0.089) (225).

 

Genetic factors play a role in the HDL response to alcohol (226). Individuals with an apoE2 allele have greater HDL-C increase and those with an apoE4 allele have a blunted increase in HDL-C with alcohol intake (226).In addition to an increase in HDL-C levels studies have suggested that the ability of HDL to facilitate the efflux of cholesterol from cells is enhanced by alcohol intake (226,227).

 

One should note in the meta-analyses described above alcohol doesn’t appear to have a major impact on TG levels. However, it must be recognized that the amount of alcohol consumed is a key variable (228,229). At low to moderate amounts alcohol has either no effect or might even decrease TG levels (228). However, at high amounts of alcohol intake increases in TG levels are observed (228,229). As noted above one meta-analysis noted that the consumption of 60 grams per day of alcohol increased TG levels (224). Moreover, alcohol consumed with a meal increases and prolongs the postprandial increase in TG levels (228,229). Additionally, genetic factors and the presence of other abnormalities play a role in the TG response to alcohol intake (229). For example, the increase in TG levels after red wine was -4%, 17%, and 33% in individuals with a BMI 19.60-24.45, 24.46- 26.29, and 26.30-30.44, respectively (P = .001) demonstrating that the increase in TG was strongly influenced by BMI (230). Finally, in patients with pre-existing hypertriglyceridemia moderate alcohol intake increased TG levels (338 ± 71 mg/dL to 498 ± 117 mg/dL; P < 0.05) (231).  

 

MECHANISM FOR THE EFFECT OF ALCOHOL ON HDL

 

 The mechanism for the increase in HDL-C levels is likely due to an increased production of apo A1 and A2 (232). Additionally, alcohol inhibits cholesteryl ester transfer protein (CETP) activity, which will also increase HDL-C levels (229).

 

MECHANISM FOR THE EFFECT OF ALCOHOL ON TRIGLYCERIDES

 

Alcohol increases VLDL secretion by the liver (229). The increased production and secretion of VLDL is due to a number of factors including a) alcohol increases lipolysis in adipose tissue and increases the delivery of fatty acids to the liver b) alcohol increases hepatic fatty acid transporters increasing the uptake of circulating fatty acids c) alcohol increases hepatic de novo fatty acid synthesis d) alcohol decreases the beta oxidation of fatty acids in the liver (229,233,234). Together these effects lead to an increased supply of fatty acids in the liver facilitating TG synthesis and the formation and secretion of VLDL.

 

While moderate alcohol intake increases lipoprotein lipase (LPL) activity acute alcohol intake inhibits LPL activity, which could explain the observation that alcohol consumed with a meal increases postprandial TG levels ((228,229).

 

Summary of the Effect of Specific Foods on Lipid Levels

 

Table 16. Major Effects of Specific Foods on Lipid Levels

Nuts and Seeds

Decrease TC and LDL-C

Whole Grains

Decrease LDL-C

Garlic

Decrease TC, LDL-C, TG

Green and Black Tea

Decrease TC and LDL-C

Coffee (depends on method of preparation)

Increase TC, LDL-C, TG

Cocoa and Chocolate

Decrease LDL-C

 

SPECIFIC DIETS

 

The effect of several dietary strategies on lipid levels is discussed below. Randomized controlled trials on the effect of specific diets on cardiovascular outcomes were discussed in earlier sections (saturated fatty acids section and monounsaturated fatty acids section).  

 

Mediterranean Diet

 

Mediterranean diets have an abundance of plant foods, including vegetables, legumes, nuts, fruits, and grains, fish, and low to moderate red wine consumption. Low consumption of meat and meat products and moderate consumption of milk and dairy products is encouraged. In the PREDIMED trial the Mediterranean diet resulted in a small but significant increase in HDL-C levels and a small decrease in both LDL-C and TG levels (76). In the Lyon Diet Heart Study lipid levels were similar in the Mediterranean and usual diet groups (77). The cardiovascular outcome benefits of both of these randomized outcome trials are discussed in the effect of MUFA on CVD section. In a meta-analysis of the effect of a Mediterranean diet on lipid levels little or no change in LDL-C, HDL-C, and TGs was observed (235). Another meta-analysis reported a 4.6mg/dL decrease in LDL-C and a 0.61mg/dL increase in HDL-C (236).

 

Dietary Approach to Stop Hypertension (DASH) Diet

 

The DASH diet promotes the consumption of fruits, vegetables, low-fat dairy products, whole grains, poultry, fish, and nuts and a decrease in the intake of red meat, sweets, sugar-containing beverages, total fat, saturated fat, and cholesterol. In the initial DASH trial total fat and SFA intake was reduced in the DASH diet group (total fat 27% vs. 39% of calories; SFA 6.2% vs. 15% of calories). MUFA and PUFA intake were similar but cholesterol intake was decreased (194mg/day vs 324mg/day). As expected, CHO and fiber intake were increase (CHO 59% vs. 49% of calories; fiber 35grams/day vs. 17grams/day). The DASH diet lowered TC (15.6 to 19.5mg/dL), LDL-C (11.7 to 15.5mg/dL), and HDL-C (3.12 to 3.90mg/dL) (237). TG levels were not significantly affected. In a meta-analysis of twenty studies of the DASH diet reporting data for 1917 participants TC was decreased (7.8mg/dL; P=0.001) and LDL was decreased (3.9mg/dL; p<0.03) (238). HDL-C and TG levels were not significantly altered (238). Similar results were seen in other meta-analyses (239,240).

 

Portfolio Diet

 

The portfolio dietary pattern is a plant-based dietary pattern that includes four cholesterol-lowering foods; a) tree nuts or peanuts, b) plant protein from soy products, beans, peas, chickpeas, or lentils, c) viscous soluble fiber from oats, barley, psyllium, eggplant, okra, apples, oranges, or berries, and d) plant sterols initially provided in a plant sterol-enriched margarine. In a meta-analysis of 5 studies with 439 participants LDL-C was lowered by 17% (28.5mg/dL; p< 0.001) and TGs by 16% (24.6mg/dL; p< 0.001) with no change in HDL-C or weight (241).

 

Nordic Diet

 

The Nordic diet is based on the consumption of different healthy foods such as whole grains, fruits (such as berries, apples, and pears), vegetables, legumes (such as oats, barley, and almonds), rapeseed oil, fatty fish (such as salmon, herring and mackerel), shellfish, seaweed, low-fat choices of meat (such as poultry and game), low-fat dairy, and decreased intake of salt and sugar-sweetened products. In a meta-analysis of 5 studies LDL-C was decreased by 11.7mg/dL (p= 0.013) with no changes in TG or HDL-C levels (242). In another meta-analysis of 6 studies LDL-C was decreased by 10.1mg/dL with no changes in TG or HDL-C levels (243).

 

Ketogenic Diet

 

Low CHO diets can contain variable amounts of CHO. When the CHO levels are very low, they stimulate the formation of ketones. In a typical ketogenic diet CHO contribute <10% of calories (< 50 grams/day), protein approximately 30% of calories, and fat approximately 60% of calories with no restrictions on the type of fat or cholesterol levels. These diets can be high in beef, poultry, fish, oils, various nuts/seeds, and peanut butter, with moderate amounts of vegetables, salads with low-carbohydrate dressing, cheese, and eggs. Fruits and fruit juices, most dairy products with the exception of hard cheeses and heavy cream, breads, cereals, beans, rice, desserts/sweets, or any other foods containing substantial amounts of CHO are avoided.

 

It is well recognized that a ketogenic diet results in an increase in LDL-C levels, which varies depending upon the type of fat ingested, the degree of carbohydrate restriction, the presence of other medical conditions, weight loss on the diet, and genetic background (244). This increase in LDL-C levels is best illustrated in children treated with a ketogenic diet for epilepsy and in healthy individuals on a ketogenic diet (245-250). In some studies HDL-C is also increased (246-249). A meta-analysis of randomized studies in normal-weight adults found that a ketogenic diet increased LDL-C by 42mg/dL and HDL-C by 13.7mg/dL with no significant changes in TG levels (251). It should be noted that the increase in LDL-C is often not observed or is modest in patients with obesity or the metabolic syndrome (252,253).

 

While the typical increases in LDL-C levels observed with a ketogenic diet are relatively modest, recently a series of reports have described marked elevations in LDL-C levels in some patients on a ketogenic diet (253-255). For example, Goldberg et al reported 5 patients with marked increases in LDL-C levels on a ketogenic diet (256). Three patients had LDL-C levels greater than 500mg/dL. Similarly, Schaffer et al described 3 patients in which a very low carbohydrate diet induced LDL-C levels greater then 400mg/dL (257). Finally, Schmidt et al reported 17 patients with LDL-C levels greater than 200mg/dL on a ketogenic diet (258). In these patients there was an average increase in their LDL-C level of 187 mg/dL (258). The elevations in LDL-C levels decrease towards normal with cessation of the ketogenic diet (256-258). It should be noted that most of the patients with marked elevations in LDL-C in response to a ketogenic diet had normal LDL-C levels prior to the dietary change (255).

 

Many of the individuals who develop marked increases in LDL-C on a very low carbohydrate ketogenic diet have low TG levels, elevated HDL-C levels, and are thin (253,255). This phenotype has been called the lean mass hyper-responder (LMHR) phenotype (253,255). LMHR individuals have been defined as having TG <70mg/dL, HDL-C > 80mg/dL, and LDL-C > 200mg/dL (253,255). The mechanism for the marked increase in LDL-C levels is unknown. It may be due to a genetic predisposition in certain individuals (Apo E2/E2 genotype or high polygenic risk score for hypercholesterolemia) (256). Therefore, it is important for clinicians to monitor lipid levels in patients electing to follow a very low CHO/high fat diet.    

 

Comparison of Low Fat vs. Low Carbohydrate Weight Loss Diets

 

Numerous randomized studies have compared the effect of low fat vs. low CHO weight loss diets on lipid levels. In a study by Foster et al 154 obese individuals were randomized to a low-fat diet and 153 obese individuals to a low CHO diet (259). In the low CHO diet during the first 12 weeks of treatment participants were instructed to limit CHO intake to 20 grams/day in the form of low–glycemic index vegetables after which the diet was gradually liberalized. In the low-fat diet participants were instructed to limit energy intake with approximately 55% of calories from CHO, 30% from fat, and 15% from protein. Participants were instructed to limit calorie intake, with a focus on decreasing fat intake. After 6 months weight loss was similar in both diet groups. The effect on lipid levels at 6 months is shown in table 17. As one would expect the low CHO was very effective at lowering TG levels and increasing HDL-C levels while the low-fat diet was very effective at lowering LDL-C levels. The large weight loss in this trial may have contributed to the large reduction in lipid levels. A review of a large number of meta-analyses comparing a low CHO diet vs. low fat weight loss diet similarly described that the low CHO diet lowered TG levels and increased HDL-C and LDL-C levels compared to the low-fat diet (244). Note the increase in LDL-C with the low-CHO diet was blunted in patients with diabetes or pre-diabetes (244). Also, the increase in LDL-C levels is likely to be greater in low CHO diets that are enriched in SFA (244).

 

Table 17. Comparison of Low Fat vs. Low Carbohydrate Weight Loss Diet on Lipid Levels

 

Low Fat Diet

Low Carbohydrate Diet

 

Weight

-11.3kg

-12.2kg

NS

TGs

-24mg/dL

-49mg/dL

P<0.001

LDL-C

-9.5mg/dL

0.5mg/dL

P<0.001

HDL-C

1.0mg/dL

6.2mg/dL

P<0.001

 

Comparison of Vegetarian and Omnivore Diet on Lipid Levels

 

Vegetarian diets exclude all animal flesh. A meta-analysis of 19 studies comparing a vegetarian vs. omnivore diet found that consumption of vegetarian diets resulted in a 12.2mg/dL decrease in LDL-C (p < 0.001) and 3.4mg/dL decrease in HDL-C (p < 0.001) and a nonsignificant increase in TG levels (5.8 mg/dL; P = 0.090) compared with consumption of an omnivorous diet (260). Vegan diets, which exclude all animal products, were associated with larger LDL-C reductions than lacto-ovo vegetarian diets. A meta-analysis of 11 clinical trials comparing a vegetarian vs. omnivore diet observed similar results (LDL‐C decreased 13.3mg/dL ; P<0.001; HDL decreased 3.9mg/dL; P<0.001) (261). It is likely that a decrease in dietary SFA and cholesterol and an increase in dietary fiber and phytosterols account for the differences in a vegetarian and omnivore diets.

 

Comparison of 14 Different Diets on Lipid Levels

 

In a network meta-analysis of 121 eligible trials with 21, 942 overweight or obese patients Ge and colleagues compared the effect of 14 different diets on LDL-C and HDL-C levels (236). The diets could be grouped into low CHO diets (Atkins, South Beach, Zone), moderate macronutrients diets (Biggest Loser, DASH, Jenny Craig, Mediterranean, Portfolio, Slimming World, Volumetrics, Weight Watchers), and low-fat diets (Ornish, Rosemary Conley). The effect of these different diets on LDL-C and HDL-C levels are shown in table 18. It should be noted that despite considerable weight loss the effect of these diets on LDL-C and HDL-C levels was very modest except for the LDL-C lowering seen with the Portfolio diet. Unfortunately, a comparison of the effect of these diets on TG levels was not reported.

 

Table 18. Effect of Different Diets in Comparison with Usual Diet

Diet vs. Usual Diet

Decrease in Weight (Kg)

Change in LDL-C (mg/dL)

Change in HDL-C (mg/dL)

Atkins

5.46

+2.75

-3.41

Zone

4.07

+2.89

+0.33

Dash

3.63

-3.93

+1.90

Mediterranean

2.87

-4.59

+0.61

Paleolithic

5.31

-7.27

+2.52

Low Fat

4.87

-1.92

+2.13

Jenny Craig

7.77

-0.21

+2.85

Volumetrics

5.95

-7.13

+0.13

Weight Watchers

3.90

-7.13

+0.88

Rosemary Conley

3.76

-7.15

+2.04

Ornish

3.64

-4.71

+4.87

Portfolio

3.64

-21.29

+3.26

Biggest Loser

2.88

-3.90

+0.01

Slimming World

2.15

N/A

N/A

South Beach

9.86

+0.64

-3.60

Dietary Advice

0.31

+2.01

+1.71

 Summary of results of popular named diets network meta-analysis for outcomes at six months

 

In a study carried out in a single center the Atkins, Zone, Weight Watchers, and Ornish diets were compared and the effect on TG levels was also reported (262). Table 19 shows the results of this study at 2 months, a period at which dietary compliance was still high. The magnitude of weight loss was similar but the decrease in LDL-C that occurs with weight loss was blunted with a diet that was high in fat (Atkins diet). In contrast HDL-C levels increased with a high fat diet, particularly SFA (Atkins diet) and decreased with a very low-fat diet (Ornish diet). The weight loss induced decrease in TG levels was blunted by a high CHO intake (Ornish diet). These observations confirm and extend the results described above.

 

Table 19. Effect of Different Diets on Lipid Levels

 

Weight (kg)

LDL-C (mg/dL)

HDL-C (mg/dL)

TG (mg/dL)

Atkins

-3.6

1.3

3.2

-32

Zone

-3.8

-9.7

1.8

-54

Weight Watchers

-3.5

-12.1

-0.2

-9.2

Ornish

-3.6

-16.5

-3.6

-0.4

 

Summary

 

While diets can significantly affect lipid levels it should be recognized that the effect is typically relatively modest compared to drug therapy. Whether these modest effects on lipid levels can reduce the risk of CVD has not been tested in randomized controlled trials and given the difficulty of carrying out such long-term diet studies is likely not to be attempted. However, diet therapy can be initiated early in life and has the potential to result in long-term decreases in lipid levels. Given that studies have shown that long-term modest reductions in LDL-C levels can have major effects on the risk of CVD (a 10mg/dL life-long decrease in LDL-C due to polymorphisms in ATP citrate lyase, HMGCoA reductase, LDL receptor, PCSK9, and NPC1L1 resulted in a 16%-18% decrease in cardiovascular events (263)) it is likely that a similar long-term decrease induced by dietary changes would also be effective in decreasing CVD. A life-long 70mg/dL decrease in TG levels due to polymorphisms in the lipoprotein lipase gene resulted in a 23% decrease in coronary heart disease suggesting that long-term decreases in TG levels due to dietary changes would also be beneficial (264). Thus, long-term reductions in lipid levels induced by diet therapy may reduce the lifetime risk of developing CVD.

 

CURRENT DIETARY GUIDELINES

 

Most dietary guidelines recommended to the general population to prevent disease are very similar so I will only present the recommendations of two organizations. A brief summary of the Guidelines for Americans 2020-2025 is shown in table 20 and the guidelines from the American College of Cardiology/American Heart Association are shown in table 21.

 

Table 20. Guidelines for Americans 2020-2025

Recommend

Limit

Vegetables of all types—dark green; red and orange; beans, peas, and lentils; starchy; and other vegetables

Added sugars—Less than 10 percent of calories per day

Fruits, especially whole fruit

Saturated fat—Less than 10 percent of calories per day

Grains, at least half of which are whole grain

Sodium—Less than 2,300 milligrams per day

Dairy, including fat-free or low-fat milk, yogurt, and cheese, and/or lactose-free versions and fortified soy beverages and yogurt as alternatives

Alcoholic beverages—Adults can

choose not to drink or to drink in moderation by limiting intake to 2 drinks or less in a day for men and 1 drink or less in a day for women

Protein foods, including lean meats, poultry, and eggs; seafood; beans, peas, and lentils; and nuts, seeds, and soy products

 

Oils, including vegetable oils and oils in food, such as seafood and nuts

 

Full guideline is available at DietaryGuidelines.gov

 

Table 21. ACC/AHA Dietary Recommendations to Reduce Risk of ASCVD (265)

1. A diet emphasizing intake of vegetables, fruits, legumes, nuts, whole grains, and fish is recommended

2. Replacement of saturated fat with dietary monounsaturated and polyunsaturated fats can be beneficial

3. A diet containing reduced amounts of cholesterol and sodium can be beneficial

4. As a part of a healthy diet, it is reasonable to minimize the intake of processed meats, refined carbohydrates, and sweetened beverages

5. As a part of a healthy diet, the intake of trans fats should be avoided

ASCVD- Atherosclerotic CVD

 

DIETARY RECOMMENDATIONS FOR PATIENTS WITH LIPID DISORDERS

 

Elevated LDL-C

 

The dietary approach to reduce LDL-C levels is to avoid TFA and decrease SFA and cholesterol intake while increasing intake of fiber and phytosterols (266). Additionally, weight loss if appropriate can be helpful in lowering LDL-C levels (266). Certain foods are effective in lowering LDL-C levels such as tree nuts or peanuts, plant protein from soy products, beans, peas, chickpeas, or lentils, and viscous soluble fiber from oats, barley, psyllium, eggplant, okra, apples, oranges, or berries and if possible, can be added to the individual’s diet (236,241). If one combines multiple nutritional changes one can have significant reductions in LDL-C levels (table 22).

 

Table 22. Effect of Multiple LDL-C Lowering Changes on LDL-C Levels

Nutritional Intervention

Estimated LDL-C Decrease

Replace 5% of energy from SFA with MUFA or PUFA

5% to 10%

7.5 grams/day viscous fiber

6% to 9%

2 grams/day plant sterols/stanols

5% to 8%

Replace 30 grams animal protein or CHO with plant protein

3% to 5%

Loss 5% body weight if excess adiposity

3% to 5%

Total Effect

22% to 37%

Table adapted from (267).

 

While diet alone usually does not reduce LDL-C sufficiently it adds to the beneficial effect of cholesterol lowering drugs. In a comparison of LDL-C lowering a low-fat diet alone lowered LDL-C by 5%, a statin alone by 27%, and the combination of low-fat diet plus statin by 32% demonstrating an independent and additive effect of combining diet and lipid lowering medications (268). 

 

Modestly Elevated Triglycerides

 

The dietary approach to reduce TG levels is to reduce CHO intake particularly simple and refined sugars and to avoid or minimize alcohol intake (266). Weight loss if appropriate can be very helpful in lowering TG levels (25,266).

 

Markedly Elevated Triglycerides

 

In patients with marked elevations in TGs due to the Familial Chylomicronemia Syndrome a diet very low in fat is often necessary to prevent episodes of pancreatitis (<10% of calories from fat) (269). In patients with this disorder medium chain TGs may be helpful. In patients with the Multifactorial Chylomicronemia Syndrome who present with markedly elevated TGs (>1000mg/dL) initial dietary treatment should be a very low-fat diet until the TGs decrease. Once the TGs decrease one can initiate the diet described above for individuals with modestly elevated TGs.

 

Elevated Lipoprotein (a)

 

There is no evidence that healthy dietary changes significantly lower Lp(a) levels (62,270) . In fact, it should be noted that reducing SFA intake while decreasing LDL-C levels increases Lp(a) levels (271). In certain patients with high Lp(a) levels one may need to balance the benefits of decreasing LDL-C levels with the risks of increasing Lp(a) levels (271).

 

Effect of Dietary Advice on Lipid and Lipoprotein Levels

 

In a meta-analysis of 44 randomized studies with 18,175 healthy adult participants comparing dietary advise vs. no or minimal advice found that dietary advice reduced total serum cholesterol by 5.9mg/dL (95% CI 2.3 to 9.0) and LDL-C by 6.2mg/dL (95% CI 3.1 to 9.4) with no change in HDL-C and TG levels (272). In a meta-analysis of 7 studies with 1081 participants that compared consultation with a dietician vs. usual care there was no difference in the absolute change in TC, LDL-C, or HDL-C levels but TG levels were decreased by 19.4mg/dL (95%CI -37.8 to -1.8; p=0.03) (273). Similarly, in a meta-analysis of 5 randomized trials in 912 patients with type 2 diabetes found that dietary advice from a dietician vs. usual care resulted in a small decrease in LDL-C (6.6mg/dL) in the group receiving advice from the dietician (274). Finally, as discussed earlier the Women’s Health Initiative trial randomized 19,541 postmenopausal women 50-79 years of age to the diet intervention group and 29,294 women to usual dietary advice (52). The goal in the diet intervention group was to reduce total fat intake to 20% of calories and increase intake of vegetables/fruits to 5 servings/day and grains to at least 6 servings/day. Fat intake decreased by 8.2% of energy intake in the intervention vs the comparison group, with small decreases in SFA (2.9%), MUFA (3.3%), and PUFA (1.5%) with increased consumption of vegetables, fruits, and grains. LDL-C levels were reduced by 3.55 mg/dL in the intervention group while levels of HDL-C and TGs were not significantly different in the intervention vs comparison groups. Taken together these studies illustrate that diet therapy under many circumstances has only modest effects on lipid and lipoprotein levels. Of course, there are studies and individual patients where major reductions in lipid levels occur. For example, in a life style modification study including a vegetarian diet by Ornish and colleagues a marked decrease in LDL-C was observed (153mg/dL decreasing to 96mg/dL) (58). One is most likely to see dramatic effects the greater the change in diet (for example going from a typical Western diet to a vegetarian low-fat diet) and the higher the baseline lipid levels. Patient ability to follow the dietary advise is crucial for success.

 

ACKNOWLEDGEMENTS

 

This work was supported by grants from the Northern California Institute for Research and Education.

 

REFERENCES

 

  1. Ioannidis JPA. The Challenge of Reforming Nutritional Epidemiologic Research. JAMA 2018; 320:969-970
  2. Nissen SE. U.S. Dietary Guidelines: An Evidence-Free Zone. Ann Intern Med 2016; 164:558-559
  3. Lichtenstein AH. Nutrient supplements and cardiovascular disease: a heartbreaking story. J Lipid Res 2009; 50 Suppl:S429-433
  4. Rimm EB, Stampfer MJ, Ascherio A, Giovannucci E, Colditz GA, Willett WC. Vitamin E consumption and the risk of coronary heart disease in men. N Engl J Med 1993; 328:1450-1456
  5. Kushi LH, Folsom AR, Prineas RJ, Mink PJ, Wu Y, Bostick RM. Dietary antioxidant vitamins and death from coronary heart disease in postmenopausal women. N Engl J Med 1996; 334:1156-1162
  6. Stampfer MJ, Hennekens CH, Manson JE, Colditz GA, Rosner B, Willett WC. Vitamin E consumption and the risk of coronary disease in women. N Engl J Med 1993; 328:1444-1449
  7. Lee CH, Chan RSM, Wan HYL, Woo YC, Cheung CYY, Fong CHY, Cheung BMY, Lam TH, Janus E, Woo J, Lam KSL. Dietary Intake of Anti-Oxidant Vitamins A, C, and E Is Inversely Associated with Adverse Cardiovascular Outcomes in Chinese-A 22-Years Population-Based Prospective Study. Nutrients 2018; 10
  8. Knekt P, Reunanen A, Jarvinen R, Seppanen R, Heliovaara M, Aromaa A. Antioxidant vitamin intake and coronary mortality in a longitudinal population study. Am J Epidemiol 1994; 139:1180-1189
  9. Lee IM, Cook NR, Gaziano JM, Gordon D, Ridker PM, Manson JE, Hennekens CH, Buring JE. Vitamin E in the primary prevention of cardiovascular disease and cancer: the Women's Health Study: a randomized controlled trial. JAMA 2005; 294:56-65
  10. de Gaetano G. Low-dose aspirin and vitamin E in people at cardiovascular risk: a randomised trial in general practice. Collaborative Group of the Primary Prevention Project. Lancet 2001; 357:89-95
  11. Sesso HD, Buring JE, Christen WG, Kurth T, Belanger C, MacFadyen J, Bubes V, Manson JE, Glynn RJ, Gaziano JM. Vitamins E and C in the prevention of cardiovascular disease in men: the Physicians' Health Study II randomized controlled trial. JAMA 2008; 300:2123-2133
  12. Heart Protection Study Collaborative G. MRC/BHF Heart Protection Study of antioxidant vitamin supplementation in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002; 360:23-33
  13. Rimm EB, Willett WC, Hu FB, Sampson L, Colditz GA, Manson JE, Hennekens C, Stampfer MJ. Folate and vitamin B6 from diet and supplements in relation to risk of coronary heart disease among women. JAMA 1998; 279:359-364
  14. Voutilainen S, Rissanen TH, Virtanen J, Lakka TA, Salonen JT. Low dietary folate intake is associated with an excess incidence of acute coronary events: The Kuopio Ischemic Heart Disease Risk Factor Study. Circulation 2001; 103:2674-2680
  15. Jayedi A, Zargar MS. Intake of vitamin B6, folate, and vitamin B12 and risk of coronary heart disease: a systematic review and dose-response meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr 2019; 59:2697-2707
  16. Armitage JM, Bowman L, Clarke RJ, Wallendszus K, Bulbulia R, Rahimi K, Haynes R, Parish S, Sleight P, Peto R, Collins R. Effects of homocysteine-lowering with folic acid plus vitamin B12 vs placebo on mortality and major morbidity in myocardial infarction survivors: a randomized trial. JAMA 2010; 303:2486-2494
  17. Albert CM, Cook NR, Gaziano JM, Zaharris E, MacFadyen J, Danielson E, Buring JE, Manson JE. Effect of folic acid and B vitamins on risk of cardiovascular events and total mortality among women at high risk for cardiovascular disease: a randomized trial. JAMA 2008; 299:2027-2036
  18. Lonn E, Yusuf S, Arnold MJ, Sheridan P, Pogue J, Micks M, McQueen MJ, Probstfield J, Fodor G, Held C, Genest J, Jr. Homocysteine lowering with folic acid and B vitamins in vascular disease. N Engl J Med 2006; 354:1567-1577
  19. Marti-Carvajal AJ, Sola I, Lathyris D, Dayer M. Homocysteine-lowering interventions for preventing cardiovascular events. Cochrane Database Syst Rev 2017; 8:CD006612
  20. Zeraatkar D, Han MA, Guyatt GH, Vernooij RWM, El Dib R, Cheung K, Milio K, Zworth M, Bartoszko JJ, Valli C, Rabassa M, Lee Y, Zajac J, Prokop-Dorner A, Lo C, Bala MM, Alonso-Coello P, Hanna SE, Johnston BC. Red and Processed Meat Consumption and Risk for All-Cause Mortality and Cardiometabolic Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann Intern Med 2019; 171:703-710
  21. Qian F, Riddle MC, Wylie-Rosett J, Hu FB. Red and Processed Meats and Health Risks: How Strong Is the Evidence? Diabetes Care 2020; 43:265-271
  22. Ordovas JM, Lopez-Miranda J, Mata P, Perez-Jimenez F, Lichtenstein AH, Schaefer EJ. Gene-diet interaction in determining plasma lipid response to dietary intervention. Atherosclerosis 1995; 118 Suppl:S11-27
  23. Vazquez-Vidal I, Desmarchelier C, Jones PJH. Nutrigenetics of Blood Cholesterol Concentrations: Towards Personalized Nutrition. Curr Cardiol Rep 2019; 21:38
  24. Flock MR, Green MH, Kris-Etherton PM. Effects of adiposity on plasma lipid response to reductions in dietary saturated fatty acids and cholesterol. Adv Nutr 2011; 2:261-274
  25. Feingold KR. Obesity and Dyslipidemia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  26. Kris-Etherton PM, Petersen K, Van Horn L. Convincing evidence supports reducing saturated fat to decrease cardiovascular disease risk. BMJ Nutr Prev Health 2018; 1:23-26
  27. Li Y, Hruby A, Bernstein AM, Ley SH, Wang DD, Chiuve SE, Sampson L, Rexrode KM, Rimm EB, Willett WC, Hu FB. Saturated Fats Compared With Unsaturated Fats and Sources of Carbohydrates in Relation to Risk of Coronary Heart Disease: A Prospective Cohort Study. J Am Coll Cardiol 2015; 66:1538-1548
  28. Lichtenstein AH. Dietary Fat and Cardiovascular Disease: Ebb and Flow Over the Last Half Century. Adv Nutr 2019; 10:S332-S339
  29. Jakobsen MU, O'Reilly EJ, Heitmann BL, Pereira MA, Balter K, Fraser GE, Goldbourt U, Hallmans G, Knekt P, Liu S, Pietinen P, Spiegelman D, Stevens J, Virtamo J, Willett WC, Ascherio A. Major types of dietary fat and risk of coronary heart disease: a pooled analysis of 11 cohort studies. Am J Clin Nutr 2009; 89:1425-1432
  30. Farvid MS, Ding M, Pan A, Sun Q, Chiuve SE, Steffen LM, Willett WC, Hu FB. Dietary linoleic acid and risk of coronary heart disease: a systematic review and meta-analysis of prospective cohort studies. Circulation 2014; 130:1568-1578
  31. Zheng Y, Fang Y, Xu X, Ye W, Kang S, Yang K, Cao Y, Xu R, Zheng J, Wang H. Dietary saturated fatty acids increased all-cause and cardiovascular disease mortality in an elderly population: The National Health and Nutrition Examination Survey. Nutr Res 2023; 120:99-114
  32. Siri-Tarino PW, Sun Q, Hu FB, Krauss RM. Meta-analysis of prospective cohort studies evaluating the association of saturated fat with cardiovascular disease. Am J Clin Nutr 2010; 91:535-546
  33. Chowdhury R, Warnakula S, Kunutsor S, Crowe F, Ward HA, Johnson L, Franco OH, Butterworth AS, Forouhi NG, Thompson SG, Khaw KT, Mozaffarian D, Danesh J, Di Angelantonio E. Association of dietary, circulating, and supplement fatty acids with coronary risk: a systematic review and meta-analysis. Ann Intern Med 2014; 160:398-406
  34. de Souza RJ, Mente A, Maroleanu A, Cozma AI, Ha V, Kishibe T, Uleryk E, Budylowski P, Schunemann H, Beyene J, Anand SS. Intake of saturated and trans unsaturated fatty acids and risk of all cause mortality, cardiovascular disease, and type 2 diabetes: systematic review and meta-analysis of observational studies. BMJ 2015; 351:h3978
  35. Skeaff CM, Miller J. Dietary fat and coronary heart disease: summary of evidence from prospective cohort and randomised controlled trials. Ann Nutr Metab 2009; 55:173-201
  36. Yamagishi K, Iso H, Yatsuya H, Tanabe N, Date C, Kikuchi S, Yamamoto A, Inaba Y, Tamakoshi A. Dietary intake of saturated fatty acids and mortality from cardiovascular disease in Japanese: the Japan Collaborative Cohort Study for Evaluation of Cancer Risk (JACC) Study. Am J Clin Nutr 2010; 92:759-765
  37. Clifton PM, Keogh JB. A systematic review of the effect of dietary saturated and polyunsaturated fat on heart disease. Nutr Metab Cardiovasc Dis 2017; 27:1060-1080
  38. Sacks FM, Lichtenstein AH, Wu JHY, Appel LJ, Creager MA, Kris-Etherton PM, Miller M, Rimm EB, Rudel LL, Robinson JG, Stone NJ, Van Horn LV. Dietary Fats and Cardiovascular Disease: A Presidential Advisory From the American Heart Association. Circulation 2017; 136:e1-e23
  39. Mozaffarian D, Micha R, Wallace S. Effects on coronary heart disease of increasing polyunsaturated fat in place of saturated fat: a systematic review and meta-analysis of randomized controlled trials. PLoS Med 2010; 7:e1000252
  40. de Oliveira Otto MC, Mozaffarian D, Kromhout D, Bertoni AG, Sibley CT, Jacobs DR, Jr., Nettleton JA. Dietary intake of saturated fat by food source and incident cardiovascular disease: the Multi-Ethnic Study of Atherosclerosis. Am J Clin Nutr 2012; 96:397-404
  41. Low-fat diet in myocardial infarction: A controlled trial. Lancet 1965; 2:501-504
  42. Leren P. The Oslo diet-heart study. Eleven-year report. Circulation 1970; 42:935-942
  43. Controlled trial of soya-bean oil in myocardial infarction. Lancet 1968; 2:693-699
  44. Dayton S, Pearce ML, Goldman H, Harnish A, Plotkin D, Shickman M, Winfield M, Zager A, Dixon W. Controlled trial of a diet high in unsaturated fat for prevention of atherosclerotic complications. Lancet 1968; 2:1060-1062
  45. DAYTON S PM, HASHIMOTO S, DIXON WJ, and TOMIYASU U. A Controlled Clinical Trial of a Diet High in Unsaturated Fat in Preventing Complications of Atherosclerosis. Circulation 1969; 40:II-1–II-63
  46. Turpeinen O, Karvonen MJ, Pekkarinen M, Miettinen M, Elosuo R, Paavilainen E. Dietary prevention of coronary heart disease: the Finnish Mental Hospital Study. Int J Epidemiol 1979; 8:99-118
  47. Miettinen M, Turpeinen O, Karvonen MJ, Pekkarinen M, Paavilainen E, Elosuo R. Dietary prevention of coronary heart disease in women: the Finnish mental hospital study. Int J Epidemiol 1983; 12:17-25
  48. Miettinen M, Turpeinen O, Karvonen MJ, Elosuo R, Paavilainen E. Effect of cholesterol-lowering diet on mortality from coronary heart-disease and other causes. A twelve-year clinical trial in men and women. Lancet 1972; 2:835-838
  49. Ramsden CE, Zamora D, Leelarthaepin B, Majchrzak-Hong SF, Faurot KR, Suchindran CM, Ringel A, Davis JM, Hibbeln JR. Use of dietary linoleic acid for secondary prevention of coronary heart disease and death: evaluation of recovered data from the Sydney Diet Heart Study and updated meta-analysis. BMJ 2013; 346:e8707
  50. Burr ML, Fehily AM, Gilbert JF, Rogers S, Holliday RM, Sweetnam PM, Elwood PC, Deadman NM. Effects of changes in fat, fish, and fibre intakes on death and myocardial reinfarction: diet and reinfarction trial (DART). Lancet 1989; 2:757-761
  51. Frantz ID, Jr., Dawson EA, Ashman PL, Gatewood LC, Bartsch GE, Kuba K, Brewer ER. Test of effect of lipid lowering by diet on cardiovascular risk. The Minnesota Coronary Survey. Arteriosclerosis 1989; 9:129-135
  52. Howard BV, Van Horn L, Hsia J, Manson JE, Stefanick ML, Wassertheil-Smoller S, Kuller LH, LaCroix AZ, Langer RD, Lasser NL, Lewis CE, Limacher MC, Margolis KL, Mysiw WJ, Ockene JK, Parker LM, Perri MG, Phillips L, Prentice RL, Robbins J, Rossouw JE, Sarto GE, Schatz IJ, Snetselaar LG, Stevens VJ, Tinker LF, Trevisan M, Vitolins MZ, Anderson GL, Assaf AR, Bassford T, Beresford SA, Black HR, Brunner RL, Brzyski RG, Caan B, Chlebowski RT, Gass M, Granek I, Greenland P, Hays J, Heber D, Heiss G, Hendrix SL, Hubbell FA, Johnson KC, Kotchen JM. Low-fat dietary pattern and risk of cardiovascular disease: the Women's Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 2006; 295:655-666
  53. Wolfe MS, Sawyer JK, Morgan TM, Bullock BC, Rudel LL. Dietary polyunsaturated fat decreases coronary artery atherosclerosis in a pediatric-aged population of African green monkeys. Arterioscler Thromb 1994; 14:587-597
  54. Rudel LL, Parks JS, Sawyer JK. Compared with dietary monounsaturated and saturated fat, polyunsaturated fat protects African green monkeys from coronary artery atherosclerosis. Arterioscler Thromb Vasc Biol 1995; 15:2101-2110
  55. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  56. Silverman MG, Ference BA, Im K, Wiviott SD, Giugliano RP, Grundy SM, Braunwald E, Sabatine MS. Association Between Lowering LDL-C and Cardiovascular Risk Reduction Among Different Therapeutic Interventions: A Systematic Review and Meta-analysis. JAMA 2016; 316:1289-1297
  57. Watts GF, Lewis B, Brunt JN, Lewis ES, Coltart DJ, Smith LD, Mann JI, Swan AV. Effects on coronary artery disease of lipid-lowering diet, or diet plus cholestyramine, in the St Thomas' Atherosclerosis Regression Study (STARS). Lancet 1992; 339:563-569
  58. Ornish D, Brown SE, Scherwitz LW, Billings JH, Armstrong WT, Ports TA, McLanahan SM, Kirkeeide RL, Brand RJ, Gould KL. Can lifestyle changes reverse coronary heart disease? The Lifestyle Heart Trial. Lancet 1990; 336:129-133
  59. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394
  60. Mensink RP. Effects of saturated fatty acids on serum lipids and lipoproteins: a systematic review and regression analysis. Geneva, Switzerland: Geneva: World Health Organization; 2016.
  61. Dreon DM, Fernstrom HA, Campos H, Blanche P, Williams PT, Krauss RM. Change in dietary saturated fat intake is correlated with change in mass of large low-density-lipoprotein particles in men. Am J Clin Nutr 1998; 67:828-836
  62. Enkhmaa B, Petersen KS, Kris-Etherton PM, Berglund L. Diet and Lp(a): Does Dietary Change Modify Residual Cardiovascular Risk Conferred by Lp(a)? Nutrients 2020; 12
  63. Mensink RP, Zock PL, Kester AD, Katan MB. Effects of dietary fatty acids and carbohydrates on the ratio of serum total to HDL cholesterol and on serum lipids and apolipoproteins: a meta-analysis of 60 controlled trials. Am J Clin Nutr 2003; 77:1146-1155
  64. Wu JHY, Micha R, Mozaffarian D. Dietary fats and cardiometabolic disease: mechanisms and effects on risk factors and outcomes. Nat Rev Cardiol 2019; 16:581-601
  65. Micha R, Mozaffarian D. Saturated fat and cardiometabolic risk factors, coronary heart disease, stroke, and diabetes: a fresh look at the evidence. Lipids 2010; 45:893-905
  66. Flock MR, Kris-Etherton PM. Diverse physiological effects of long-chain saturated fatty acids: implications for cardiovascular disease. Curr Opin Clin Nutr Metab Care 2013; 16:133-140
  67. Fernandez ML, West KL. Mechanisms by which dietary fatty acids modulate plasma lipids. J Nutr 2005; 135:2075-2078
  68. Dietschy JM. Dietary fatty acids and the regulation of plasma low density lipoprotein cholesterol concentrations. J Nutr 1998; 128:444S-448S
  69. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest 2002; 109:1125-1131
  70. Mente A, de Koning L, Shannon HS, Anand SS. A systematic review of the evidence supporting a causal link between dietary factors and coronary heart disease. Arch Intern Med 2009; 169:659-669
  71. Schwingshackl L, Hoffmann G. Monounsaturated fatty acids, olive oil and health status: a systematic review and meta-analysis of cohort studies. Lipids Health Dis 2014; 13:154
  72. Zong G, Li Y, Sampson L, Dougherty LW, Willett WC, Wanders AJ, Alssema M, Zock PL, Hu FB, Sun Q. Monounsaturated fats from plant and animal sources in relation to risk of coronary heart disease among US men and women. Am J Clin Nutr 2018; 107:445-453
  73. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Martinez-Gonzalez MA. Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med 2013; 368:1279-1290
  74. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Fito M, Gea A, Hernan MA, Martinez-Gonzalez MA. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N Engl J Med 2018; 378:e34
  75. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, Gomez-Gracia E, Ruiz-Gutierrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pinto X, Basora J, Munoz MA, Sorli JV, Martinez JA, Martinez-Gonzalez MA. Retraction and Republication: Primary Prevention of Cardiovascular Disease with a Mediterranean Diet. N Engl J Med 2013;368:1279-90. N Engl J Med 2018; 378:2441-2442
  76. Estruch R, Martinez-Gonzalez MA, Corella D, Salas-Salvado J, Ruiz-Gutierrez V, Covas MI, Fiol M, Gomez-Gracia E, Lopez-Sabater MC, Vinyoles E, Aros F, Conde M, Lahoz C, Lapetra J, Saez G, Ros E. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145:1-11
  77. de Lorgeril M, Renaud S, Mamelle N, Salen P, Martin JL, Monjaud I, Guidollet J, Touboul P, Delaye J. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet 1994; 343:1454-1459
  78. de Lorgeril M, Salen P, Martin JL, Monjaud I, Delaye J, Mamelle N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation 1999; 99:779-785
  79. Delgado-Lista J, Alcala-Diaz JF, Torres-Pena JD, Quintana-Navarro GM, Fuentes F, Garcia-Rios A, Ortiz-Morales AM, Gonzalez-Requero AI, Perez-Caballero AI, Yubero-Serrano EM, Rangel-Zuniga OA, Camargo A, Rodriguez-Cantalejo F, Lopez-Segura F, Badimon L, Ordovas JM, Perez-Jimenez F, Perez-Martinez P, Lopez-Miranda J. Long-term secondary prevention of cardiovascular disease with a Mediterranean diet and a low-fat diet (CORDIOPREV): a randomised controlled trial. Lancet 2022; 399:1876-1885
  80. Li J, Guasch-Ferre M, Li Y, Hu FB. Dietary intake and biomarkers of linoleic acid and mortality: systematic review and meta-analysis of prospective cohort studies. Am J Clin Nutr 2020; 112:150-167
  81. Marklund M, Wu JHY, Imamura F, Del Gobbo LC, Fretts A, de Goede J, Shi P, Tintle N, Wennberg M, Aslibekyan S, Chen TA, de Oliveira Otto MC, Hirakawa Y, Eriksen HH, Kroger J, Laguzzi F, Lankinen M, Murphy RA, Prem K, Samieri C, Virtanen J, Wood AC, Wong K, Yang WS, Zhou X, Baylin A, Boer JMA, Brouwer IA, Campos H, Chaves PHM, Chien KL, de Faire U, Djousse L, Eiriksdottir G, El-Abbadi N, Forouhi NG, Michael Gaziano J, Geleijnse JM, Gigante B, Giles G, Guallar E, Gudnason V, Harris T, Harris WS, Helmer C, Hellenius ML, Hodge A, Hu FB, Jacques PF, Jansson JH, Kalsbeek A, Khaw KT, Koh WP, Laakso M, Leander K, Lin HJ, Lind L, Luben R, Luo J, McKnight B, Mursu J, Ninomiya T, Overvad K, Psaty BM, Rimm E, Schulze MB, Siscovick D, Skjelbo Nielsen M, Smith AV, Steffen BT, Steffen L, Sun Q, Sundstrom J, Tsai MY, Tunstall-Pedoe H, Uusitupa MIJ, van Dam RM, Veenstra J, Monique Verschuren WM, Wareham N, Willett W, Woodward M, Yuan JM, Micha R, Lemaitre RN, Mozaffarian D, Riserus U. Biomarkers of Dietary Omega-6 Fatty Acids and Incident Cardiovascular Disease and Mortality. Circulation 2019; 139:2422-2436
  82. Zhu Y, Bo Y, Liu Y. Dietary total fat, fatty acids intake, and risk of cardiovascular disease: a dose-response meta-analysis of cohort studies. Lipids Health Dis 2019; 18:91
  83. Hooper L, Al-Khudairy L, Abdelhamid AS, Rees K, Brainard JS, Brown TJ, Ajabnoor SM, O'Brien AT, Winstanley LE, Donaldson DH, Song F, Deane KH. Omega-6 fats for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11:CD011094
  84. Abdelhamid AS, Martin N, Bridges C, Brainard JS, Wang X, Brown TJ, Hanson S, Jimoh OF, Ajabnoor SM, Deane KH, Song F, Hooper L. Polyunsaturated fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2018; 11:CD012345
  85. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  86. Gardner CD, Kraemer HC. Monounsaturated versus polyunsaturated dietary fat and serum lipids. A meta-analysis. Arterioscler Thromb Vasc Biol 1995; 15:1917-1927
  87. Lichtenstein AH. Dietary trans fatty acids and cardiovascular disease risk: past and present. Curr Atheroscler Rep 2014; 16:433
  88. Mozaffarian D, Katan MB, Ascherio A, Stampfer MJ, Willett WC. Trans fatty acids and cardiovascular disease. N Engl J Med 2006; 354:1601-1613
  89. Nestel P. Trans fatty acids: are its cardiovascular risks fully appreciated? Clin Ther 2014; 36:315-321
  90. Mauger JF, Lichtenstein AH, Ausman LM, Jalbert SM, Jauhiainen M, Ehnholm C, Lamarche B. Effect of different forms of dietary hydrogenated fats on LDL particle size. Am J Clin Nutr 2003; 78:370-375
  91. Brouwer IA, Wanders AJ, Katan MB. Effect of animal and industrial trans fatty acids on HDL and LDL cholesterol levels in humans--a quantitative review. PLoS One 2010; 5:e9434
  92. Bendsen NT, Christensen R, Bartels EM, Astrup A. Consumption of industrial and ruminant trans fatty acids and risk of coronary heart disease: a systematic review and meta-analysis of cohort studies. Eur J Clin Nutr 2011; 65:773-783
  93. Matthan NR, Welty FK, Barrett PH, Harausz C, Dolnikowski GG, Parks JS, Eckel RH, Schaefer EJ, Lichtenstein AH. Dietary hydrogenated fat increases high-density lipoprotein apoA-I catabolism and decreases low-density lipoprotein apoB-100 catabolism in hypercholesterolemic women. Arterioscler Thromb Vasc Biol 2004; 24:1092-1097
  94. van Tol A, Zock PL, van Gent T, Scheek LM, Katan MB. Dietary trans fatty acids increase serum cholesterylester transfer protein activity in man. Atherosclerosis 1995; 115:129-134
  95. Soliman GA. Dietary Cholesterol and the Lack of Evidence in Cardiovascular Disease. Nutrients 2018; 10
  96. Xu Z, McClure ST, Appel LJ. Dietary Cholesterol Intake and Sources among U.S Adults: Results from National Health and Nutrition Examination Surveys (NHANES), 2001(-)2014. Nutrients 2018; 10
  97. Berger S, Raman G, Vishwanathan R, Jacques PF, Johnson EJ. Dietary cholesterol and cardiovascular disease: a systematic review and meta-analysis. Am J Clin Nutr 2015; 102:276-294
  98. Carson JAS, Lichtenstein AH, Anderson CAM, Appel LJ, Kris-Etherton PM, Meyer KA, Petersen K, Polonsky T, Van Horn L. Dietary Cholesterol and Cardiovascular Risk: A Science Advisory From the American Heart Association. Circulation 2020; 141:e39-e53
  99. Hu FB, Stampfer MJ, Manson JE, Rimm E, Colditz GA, Rosner BA, Hennekens CH, Willett WC. Dietary fat intake and the risk of coronary heart disease in women. N Engl J Med 1997; 337:1491-1499
  100. Ascherio A, Rimm EB, Giovannucci EL, Spiegelman D, Stampfer M, Willett WC. Dietary fat and risk of coronary heart disease in men: cohort follow up study in the United States. BMJ 1996; 313:84-90
  101. Alexander DD, Miller PE, Vargas AJ, Weed DL, Cohen SS. Meta-analysis of Egg Consumption and Risk of Coronary Heart Disease and Stroke. J Am Coll Nutr 2016; 35:704-716
  102. Krittanawong C, Narasimhan B, Wang Z, Virk HUH, Farrell AM, Zhang H, Tang WHW. Association Between Egg Consumption and Risk of Cardiovascular Outcomes: A Systematic Review and Meta-Analysis. Am J Med 2021; 134:76-83 e72
  103. Drouin-Chartier JP, Schwab AL, Chen S, Li Y, Sacks FM, Rosner B, Manson JE, Willett WC, Stampfer MJ, Hu FB, Bhupathiraju SN. Egg consumption and risk of type 2 diabetes: findings from 3 large US cohort studies of men and women and a systematic review and meta-analysis of prospective cohort studies. Am J Clin Nutr 2020; 112:619-630
  104. Mazidi M, Katsiki N, Mikhailidis DP, Pencina MJ, Banach M. Egg Consumption and Risk of Total and Cause-Specific Mortality: An Individual-Based Cohort Study and Pooling Prospective Studies on Behalf of the Lipid and Blood Pressure Meta-analysis Collaboration (LBPMC) Group. J Am Coll Nutr 2019; 38:552-563
  105. Xu L, Lam TH, Jiang CQ, Zhang WS, Zhu F, Jin YL, Woo J, Cheng KK, Thomas GN. Egg consumption and the risk of cardiovascular disease and all-cause mortality: Guangzhou Biobank Cohort Study and meta-analyses. Eur J Nutr 2019; 58:785-796
  106. Zhao B, Gan L, Graubard BI, Mannisto S, Albanes D, Huang J. Associations of Dietary Cholesterol, Serum Cholesterol, and Egg Consumption With Overall and Cause-Specific Mortality: Systematic Review and Updated Meta-Analysis. Circulation 2022; 145:1506-1520
  107. Vincent MJ, Allen B, Palacios OM, Haber LT, Maki KC. Meta-regression analysis of the effects of dietary cholesterol intake on LDL and HDL cholesterol. Am J Clin Nutr 2019; 109:7-16
  108. Herron KL, Lofgren IE, Sharman M, Volek JS, Fernandez ML. High intake of cholesterol results in less atherogenic low-density lipoprotein particles in men and women independent of response classification. Metabolism 2004; 53:823-830
  109. Djousse L, Gaziano JM. Dietary cholesterol and coronary artery disease: a systematic review. Curr Atheroscler Rep 2009; 11:418-422
  110. McNamara DJ. The impact of egg limitations on coronary heart disease risk: do the numbers add up? J Am Coll Nutr 2000; 19:540S-548S
  111. Grundy SM. Does Dietary Cholesterol Matter? Curr Atheroscler Rep 2016; 18:68
  112. Tannock LR, O'Brien KD, Knopp RH, Retzlaff B, Fish B, Wener MH, Kahn SE, Chait A. Cholesterol feeding increases C-reactive protein and serum amyloid A levels in lean insulin-sensitive subjects. Circulation 2005; 111:3058-3062
  113. Jakobsen MU, Dethlefsen C, Joensen AM, Stegger J, Tjonneland A, Schmidt EB, Overvad K. Intake of carbohydrates compared with intake of saturated fatty acids and risk of myocardial infarction: importance of the glycemic index. Am J Clin Nutr 2010; 91:1764-1768
  114. Lambadiari V, Korakas E, Tsimihodimos V. The Impact of Dietary Glycemic Index and Glycemic Load on Postprandial Lipid Kinetics, Dyslipidemia and Cardiovascular Risk. Nutrients 2020; 12
  115. Vega-Lopez S, Venn BJ, Slavin JL. Relevance of the Glycemic Index and Glycemic Load for Body Weight, Diabetes, and Cardiovascular Disease. Nutrients 2018; 10
  116. Jenkins DJA, Dehghan M, Mente A, Bangdiwala SI, Rangarajan S, Srichaikul K, Mohan V, Avezum A, Diaz R, Rosengren A, Lanas F, Lopez-Jaramillo P, Li W, Oguz A, Khatib R, Poirier P, Mohammadifard N, Pepe A, Alhabib KF, Chifamba J, Yusufali AH, Iqbal R, Yeates K, Yusoff K, Ismail N, Teo K, Swaminathan S, Liu X, Zatonska K, Yusuf R, Yusuf S. Glycemic Index, Glycemic Load, and Cardiovascular Disease and Mortality. N Engl J Med 2021; 384:1312-1322
  117. Jenkins DJA, Willett WC, Yusuf S, Hu FB, Glenn AJ, Liu S, Mente A, Miller V, Bangdiwala SI, Gerstein HC, Sieri S, Ferrari P, Patel AV, McCullough ML, Le Marchand L, Freedman ND, Loftfield E, Sinha R, Shu XO, Touvier M, Sawada N, Tsugane S, van den Brandt PA, Shuval K, Khan TA, Paquette M, Sahye-Pudaruth S, Patel D, Siu TFY, Srichaikul K, Kendall CWC, Sievenpiper JL. Association of glycaemic index and glycaemic load with type 2 diabetes, cardiovascular disease, cancer, and all-cause mortality: a meta-analysis of mega cohorts of more than 100 000 participants. Lancet Diabetes Endocrinol 2024; 12:107-118
  118. Yang Q, Zhang Z, Gregg EW, Flanders WD, Merritt R, Hu FB. Added sugar intake and cardiovascular diseases mortality among US adults. JAMA Intern Med 2014; 174:516-524
  119. Narain A, Kwok CS, Mamas MA. Soft drinks and sweetened beverages and the risk of cardiovascular disease and mortality: a systematic review and meta-analysis. Int J Clin Pract 2016; 70:791-805
  120. Ho FK, Gray SR, Welsh P, Petermann-Rocha F, Foster H, Waddell H, Anderson J, Lyall D, Sattar N, Gill JMR, Mathers JC, Pell JP, Celis-Morales C. Associations of fat and carbohydrate intake with cardiovascular disease and mortality: prospective cohort study of UK Biobank participants. BMJ 2020; 368:m688
  121. Malik VS, Li Y, Pan A, De Koning L, Schernhammer E, Willett WC, Hu FB. Long-Term Consumption of Sugar-Sweetened and Artificially Sweetened Beverages and Risk of Mortality in US Adults. Circulation 2019; 139:2113-2125
  122. de Koning L, Malik VS, Kellogg MD, Rimm EB, Willett WC, Hu FB. Sweetened beverage consumption, incident coronary heart disease, and biomarkers of risk in men. Circulation 2012; 125:1735-1741, S1731
  123. Gerber PA, Berneis K. Regulation of low-density lipoprotein subfractions by carbohydrates. Curr Opin Clin Nutr Metab Care 2012; 15:381-385
  124. Aeberli I, Gerber PA, Hochuli M, Kohler S, Haile SR, Gouni-Berthold I, Berthold HK, Spinas GA, Berneis K. Low to moderate sugar-sweetened beverage consumption impairs glucose and lipid metabolism and promotes inflammation in healthy young men: a randomized controlled trial. Am J Clin Nutr 2011; 94:479-485
  125. Fechner E, Smeets E, Schrauwen P, Mensink RP. The Effects of Different Degrees of Carbohydrate Restriction and Carbohydrate Replacement on Cardiometabolic Risk Markers in Humans-A Systematic Review and Meta-Analysis. Nutrients 2020; 12
  126. Mansoor N, Vinknes KJ, Veierod MB, Retterstol K. Effects of low-carbohydrate diets v. low-fat diets on body weight and cardiovascular risk factors: a meta-analysis of randomised controlled trials. Br J Nutr 2016; 115:466-479
  127. Hu T, Mills KT, Yao L, Demanelis K, Eloustaz M, Yancy WS, Jr., Kelly TN, He J, Bazzano LA. Effects of low-carbohydrate diets versus low-fat diets on metabolic risk factors: a meta-analysis of randomized controlled clinical trials. Am J Epidemiol 2012; 176 Suppl 7:S44-54
  128. Goff LM, Cowland DE, Hooper L, Frost GS. Low glycaemic index diets and blood lipids: a systematic review and meta-analysis of randomised controlled trials. Nutr Metab Cardiovasc Dis 2013; 23:1-10
  129. Schwingshackl L, Neuenschwander M, Hoffmann G, Buyken AE, Schlesinger S. Dietary sugars and cardiometabolic risk factors: a network meta-analysis on isocaloric substitution interventions. Am J Clin Nutr 2020; 111:187-196
  130. Te Morenga LA, Howatson AJ, Jones RM, Mann J. Dietary sugars and cardiometabolic risk: systematic review and meta-analyses of randomized controlled trials of the effects on blood pressure and lipids. Am J Clin Nutr 2014; 100:65-79
  131. Fattore E, Botta F, Agostoni C, Bosetti C. Effects of free sugars on blood pressure and lipids: a systematic review and meta-analysis of nutritional isoenergetic intervention trials. Am J Clin Nutr 2017; 105:42-56
  132. Livesey G, Taylor R. Fructose consumption and consequences for glycation, plasma triacylglycerol, and body weight: meta-analyses and meta-regression models of intervention studies. Am J Clin Nutr 2008; 88:1419-1437
  133. Stanhope KL, Bremer AA, Medici V, Nakajima K, Ito Y, Nakano T, Chen G, Fong TH, Lee V, Menorca RI, Keim NL, Havel PJ. Consumption of fructose and high fructose corn syrup increase postprandial triglycerides, LDL-cholesterol, and apolipoprotein-B in young men and women. J Clin Endocrinol Metab 2011; 96:E1596-1605
  134. Kroemer G, Lopez-Otin C, Madeo F, de Cabo R. Carbotoxicity-Noxious Effects of Carbohydrates. Cell 2018; 175:605-614
  135. Softic S, Meyer JG, Wang GX, Gupta MK, Batista TM, Lauritzen H, Fujisaka S, Serra D, Herrero L, Willoughby J, Fitzgerald K, Ilkayeva O, Newgard CB, Gibson BW, Schilling B, Cohen DE, Kahn CR. Dietary Sugars Alter Hepatic Fatty Acid Oxidation via Transcriptional and Post-translational Modifications of Mitochondrial Proteins. Cell Metab 2019; 30:735-753 e734
  136. Naghshi S, Sadeghi O, Willett WC, Esmaillzadeh A. Dietary intake of total, animal, and plant proteins and risk of all cause, cardiovascular, and cancer mortality: systematic review and dose-response meta-analysis of prospective cohort studies. BMJ 2020; 370:m2412
  137. Chen Z, Glisic M, Song M, Aliahmad HA, Zhang X, Moumdjian AC, Gonzalez-Jaramillo V, van der Schaft N, Bramer WM, Ikram MA, Voortman T. Dietary protein intake and all-cause and cause-specific mortality: results from the Rotterdam Study and a meta-analysis of prospective cohort studies. Eur J Epidemiol 2020; 35:411-429
  138. Budhathoki S, Sawada N, Iwasaki M, Yamaji T, Goto A, Kotemori A, Ishihara J, Takachi R, Charvat H, Mizoue T, Iso H, Tsugane S. Association of Animal and Plant Protein Intake With All-Cause and Cause-Specific Mortality in a Japanese Cohort. JAMA Intern Med 2019; 179:1509-1518
  139. Qi XX, Shen P. Associations of dietary protein intake with all-cause, cardiovascular disease, and cancer mortality: A systematic review and meta-analysis of cohort studies. Nutr Metab Cardiovasc Dis 2020; 30:1094-1105
  140. Schwingshackl L, Hoffmann G. Long-term effects of low-fat diets either low or high in protein on cardiovascular and metabolic risk factors: a systematic review and meta-analysis. Nutr J 2013; 12:48
  141. Wycherley TP, Moran LJ, Clifton PM, Noakes M, Brinkworth GD. Effects of energy-restricted high-protein, low-fat compared with standard-protein, low-fat diets: a meta-analysis of randomized controlled trials. Am J Clin Nutr 2012; 96:1281-1298
  142. Santesso N, Akl EA, Bianchi M, Mente A, Mustafa R, Heels-Ansdell D, Schunemann HJ. Effects of higher- versus lower-protein diets on health outcomes: a systematic review and meta-analysis. Eur J Clin Nutr 2012; 66:780-788
  143. Threapleton DE, Greenwood DC, Evans CE, Cleghorn CL, Nykjaer C, Woodhead C, Cade JE, Gale CP, Burley VJ. Dietary fibre intake and risk of cardiovascular disease: systematic review and meta-analysis. BMJ 2013; 347:f6879
  144. Pereira MA, O'Reilly E, Augustsson K, Fraser GE, Goldbourt U, Heitmann BL, Hallmans G, Knekt P, Liu S, Pietinen P, Spiegelman D, Stevens J, Virtamo J, Willett WC, Ascherio A. Dietary fiber and risk of coronary heart disease: a pooled analysis of cohort studies. Arch Intern Med 2004; 164:370-376
  145. Kim Y, Je Y. Dietary fibre intake and mortality from cardiovascular disease and all cancers: A meta-analysis of prospective cohort studies. Arch Cardiovasc Dis 2016; 109:39-54
  146. Reynolds A, Mann J, Cummings J, Winter N, Mete E, Te Morenga L. Carbohydrate quality and human health: a series of systematic reviews and meta-analyses. Lancet 2019; 393:434-445
  147. Liu L, Wang S, Liu J. Fiber consumption and all-cause, cardiovascular, and cancer mortalities: a systematic review and meta-analysis of cohort studies. Mol Nutr Food Res 2015; 59:139-146
  148. Wu Y, Qian Y, Pan Y, Li P, Yang J, Ye X, Xu G. Association between dietary fiber intake and risk of coronary heart disease: A meta-analysis. Clin Nutr 2015; 34:603-611
  149. Loveman E, Colquitt J, Rees K. Cochrane corner: does increasing intake of dietary fibre help prevent cardiovascular disease? Heart 2016; 102:1607-1609
  150. Hartley L, May MD, Loveman E, Colquitt JL, Rees K. Dietary fibre for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 2016:CD011472
  151. Hollaender PL, Ross AB, Kristensen M. Whole-grain and blood lipid changes in apparently healthy adults: a systematic review and meta-analysis of randomized controlled studies. Am J Clin Nutr 2015; 102:556-572
  152. Bell LP, Hectorne K, Reynolds H, Balm TK, Hunninghake DB. Cholesterol-lowering effects of psyllium hydrophilic mucilloid. Adjunct therapy to a prudent diet for patients with mild to moderate hypercholesterolemia. JAMA 1989; 261:3419-3423
  153. Anderson JW, Zettwoch N, Feldman T, Tietyen-Clark J, Oeltgen P, Bishop CW. Cholesterol-lowering effects of psyllium hydrophilic mucilloid for hypercholesterolemic men. Arch Intern Med 1988; 148:292-296
  154. Jovanovski E, Yashpal S, Komishon A, Zurbau A, Blanco Mejia S, Ho HVT, Li D, Sievenpiper J, Duvnjak L, Vuksan V. Effect of psyllium (Plantago ovata) fiber on LDL cholesterol and alternative lipid targets, non-HDL cholesterol and apolipoprotein B: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2018; 108:922-932
  155. Brown L, Rosner B, Willett WW, Sacks FM. Cholesterol-lowering effects of dietary fiber: a meta-analysis. Am J Clin Nutr 1999; 69:30-42
  156. Ghavami A, Ziaei R, Talebi S, Barghchi H, Nattagh-Eshtivani E, Moradi S, Rahbarinejad P, Mohammadi H, Ghasemi-Tehrani H, Marx W, Askari G. Soluble Fiber Supplementation and Serum Lipid Profile: A Systematic Review and Dose-Response Meta-Analysis of Randomized Controlled Trials. Adv Nutr 2023; 14:465-474
  157. Cohn JS, Kamili A, Wat E, Chung RW, Tandy S. Reduction in intestinal cholesterol absorption by various food components: mechanisms and implications. Atheroscler Suppl 2010; 11:45-48
  158. Jesch ED, Carr TP. Food Ingredients That Inhibit Cholesterol Absorption. Prev Nutr Food Sci 2017; 22:67-80
  159. Gunness P, Gidley MJ. Mechanisms underlying the cholesterol-lowering properties of soluble dietary fibre polysaccharides. Food Funct 2010; 1:149-155
  160. Piironeen V, Lampi A. Occurrence and levels of phytosterols in foods. New York Marcel Dekker, Inc.
  161. Ras RT, Geleijnse JM, Trautwein EA. LDL-cholesterol-lowering effect of plant sterols and stanols across different dose ranges: a meta-analysis of randomised controlled studies. Br J Nutr 2014; 112:214-219
  162. Demonty I, Ras RT, van der Knaap HC, Meijer L, Zock PL, Geleijnse JM, Trautwein EA. The effect of plant sterols on serum triglyceride concentrations is dependent on baseline concentrations: a pooled analysis of 12 randomised controlled trials. Eur J Nutr 2013; 52:153-160
  163. Calpe-Berdiel L, Escola-Gil JC, Blanco-Vaca F. New insights into the molecular actions of plant sterols and stanols in cholesterol metabolism. Atherosclerosis 2009; 203:18-31
  164. Micha R, Shulkin ML, Penalvo JL, Khatibzadeh S, Singh GM, Rao M, Fahimi S, Powles J, Mozaffarian D. Etiologic effects and optimal intakes of foods and nutrients for risk of cardiovascular diseases and diabetes: Systematic reviews and meta-analyses from the Nutrition and Chronic Diseases Expert Group (NutriCoDE). PLoS One 2017; 12:e0175149
  165. Bechthold A, Boeing H, Schwedhelm C, Hoffmann G, Knuppel S, Iqbal K, De Henauw S, Michels N, Devleesschauwer B, Schlesinger S, Schwingshackl L. Food groups and risk of coronary heart disease, stroke and heart failure: A systematic review and dose-response meta-analysis of prospective studies. Crit Rev Food Sci Nutr 2019; 59:1071-1090
  166. Vernooij RWM, Zeraatkar D, Han MA, El Dib R, Zworth M, Milio K, Sit D, Lee Y, Gomaa H, Valli C, Swierz MJ, Chang Y, Hanna SE, Brauer PM, Sievenpiper J, de Souza R, Alonso-Coello P, Bala MM, Guyatt GH, Johnston BC. Patterns of Red and Processed Meat Consumption and Risk for Cardiometabolic and Cancer Outcomes: A Systematic Review and Meta-analysis of Cohort Studies. Ann Intern Med 2019; 171:732-741
  167. Fraser GE, Sabate J, Beeson WL, Strahan TM. A possible protective effect of nut consumption on risk of coronary heart disease. The Adventist Health Study. Arch Intern Med 1992; 152:1416-1424
  168. Ros E. Nuts and novel biomarkers of cardiovascular disease. Am J Clin Nutr 2009; 89:1649S-1656S
  169. Ros E. Health benefits of nut consumption. Nutrients 2010; 2:652-682
  170. Sabate J, Haddad E, Tanzman JS, Jambazian P, Rajaram S. Serum lipid response to the graduated enrichment of a Step I diet with almonds: a randomized feeding trial. Am J Clin Nutr 2003; 77:1379-1384
  171. Sabate J, Oda K, Ros E. Nut consumption and blood lipid levels: a pooled analysis of 25 intervention trials. Arch Intern Med 2010; 170:821-827
  172. Sabate J, Wien M. Nuts, blood lipids and cardiovascular disease. Asia Pac J Clin Nutr 2010; 19:131-136
  173. Banel DK, Hu FB. Effects of walnut consumption on blood lipids and other cardiovascular risk factors: a meta-analysis and systematic review. Am J Clin Nutr 2009; 90:56-63
  174. Guasch-Ferre M, Li J, Hu FB, Salas-Salvado J, Tobias DK. Effects of walnut consumption on blood lipids and other cardiovascular risk factors: an updated meta-analysis and systematic review of controlled trials. Am J Clin Nutr 2018; 108:174-187
  175. Lee-Bravatti MA, Wang J, Avendano EE, King L, Johnson EJ, Raman G. Almond Consumption and Risk Factors for Cardiovascular Disease: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Adv Nutr 2019; 10:1076-1088
  176. Hadi A, Asbaghi O, Kazemi M, Haghighian HK, Pantovic A, Ghaedi E, Abolhasani Zadeh F. Consumption of pistachio nuts positively affects lipid profiles: A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 2023; 63:5358-5371
  177. Houston L, Probst YC, Chandra Singh M, Neale EP. Tree Nut and Peanut Consumption and Risk of Cardiovascular Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv Nutr 2023; 14:1029-1049
  178. Pan A, Yu D, Demark-Wahnefried W, Franco OH, Lin X. Meta-analysis of the effects of flaxseed interventions on blood lipids. Am J Clin Nutr 2009; 90:288-297
  179. Hui S, Liu K, Lang H, Liu Y, Wang X, Zhu X, Doucette S, Yi L, Mi M. Comparative effects of different whole grains and brans on blood lipid: a network meta-analysis. Eur J Nutr 2019; 58:2779-2787
  180. Marshall S, Petocz P, Duve E, Abbott K, Cassettari T, Blumfield M, Fayet-Moore F. The Effect of Replacing Refined Grains with Whole Grains on Cardiovascular Risk Factors: A Systematic Review and Meta-Analysis of Randomized Controlled Trials with GRADE Clinical Recommendation. J Acad Nutr Diet 2020; 120:1859-1883 e1831
  181. Jenkins DJA, Blanco Mejia S, Chiavaroli L, Viguiliouk E, Li SS, Kendall CWC, Vuksan V, Sievenpiper JL. Cumulative Meta-Analysis of the Soy Effect Over Time. J Am Heart Assoc 2019; 8:e012458
  182. Moradi M, Daneshzad E, Azadbakht L. The effects of isolated soy protein, isolated soy isoflavones and soy protein containing isoflavones on serum lipids in postmenopausal women: A systematic review and meta-analysis. Crit Rev Food Sci Nutr 2020; 60:3414-3428
  183. Tokede OA, Onabanjo TA, Yansane A, Gaziano JM, Djousse L. Soya products and serum lipids: a meta-analysis of randomised controlled trials. Br J Nutr 2015; 114:831-843
  184. Anderson JW, Bush HM. Soy protein effects on serum lipoproteins: a quality assessment and meta-analysis of randomized, controlled studies. J Am Coll Nutr 2011; 30:79-91
  185. Anderson JW, Johnstone BM, Cook-Newell ME. Meta-analysis of the effects of soy protein intake on serum lipids. N Engl J Med 1995; 333:276-282
  186. Reynolds K, Chin A, Lees KA, Nguyen A, Bujnowski D, He J. A meta-analysis of the effect of soy protein supplementation on serum lipids. Am J Cardiol 2006; 98:633-640
  187. Zhan S, Ho SC. Meta-analysis of the effects of soy protein containing isoflavones on the lipid profile. Am J Clin Nutr 2005; 81:397-408
  188. Simental-Mendia LE, Gotto AM, Jr., Atkin SL, Banach M, Pirro M, Sahebkar A. Effect of soy isoflavone supplementation on plasma lipoprotein(a) concentrations: A meta-analysis. J Clin Lipidol 2018; 12:16-24
  189. Hunter PM, Hegele RA. Functional foods and dietary supplements for the management of dyslipidaemia. Nat Rev Endocrinol 2017; 13:278-288
  190. Jenkins DJ, Mirrahimi A, Srichaikul K, Berryman CE, Wang L, Carleton A, Abdulnour S, Sievenpiper JL, Kendall CW, Kris-Etherton PM. Soy protein reduces serum cholesterol by both intrinsic and food displacement mechanisms. J Nutr 2010; 140:2302S-2311S
  191. Ried K, Toben C, Fakler P. Effect of garlic on serum lipids: an updated meta-analysis. Nutr Rev 2013; 71:282-299
  192. Zeng T GF, Zhang C,Song F, Zhao X,  Xi K. A meta‐analysis of randomized, double‐blind, placebo‐controlled trials for the effects of garlic on serum lipid profiles. Journal of the Science of Food and Agricultue 2012; 92:1892-1902
  193. Reinhart KM, Talati R, White CM, Coleman CI. The impact of garlic on lipid parameters: a systematic review and meta-analysis. Nutr Res Rev 2009; 22:39-48
  194. Stevinson C, Pittler MH, Ernst E. Garlic for treating hypercholesterolemia. A meta-analysis of randomized clinical trials. Ann Intern Med 2000; 133:420-429
  195. Kwak JS, Kim JY, Paek JE, Lee YJ, Kim HR, Park DS, Kwon O. Garlic powder intake and cardiovascular risk factors: a meta-analysis of randomized controlled clinical trials. Nutr Res Pract 2014; 8:644-654
  196. Schwingshackl L, Missbach B, Hoffmann G. An umbrella review of garlic intake and risk of cardiovascular disease. Phytomedicine 2016; 23:1127-1133
  197. Sun YE, Wang W, Qin J. Anti-hyperlipidemia of garlic by reducing the level of total cholesterol and low-density lipoprotein: A meta-analysis. Medicine (Baltimore) 2018; 97:e0255
  198. Li S, Guo W, Lau W, Zhang H, Zhan Z, Wang X, Wang H. The association of garlic intake and cardiovascular risk factors: A systematic review and meta-analysis. Crit Rev Food Sci Nutr 2023; 63:8013-8031
  199. Shabani E, Sayemiri K, Mohammadpour M. The effect of garlic on lipid profile and glucose parameters in diabetic patients: A systematic review and meta-analysis. Prim Care Diabetes 2019; 13:28-42
  200. Kitamura K, Okada Y, Okada K, Kawaguchi Y, Nagaoka S. Epigallocatechin gallate induces an up-regulation of LDL receptor accompanied by a reduction of PCSK9 via the annexin A2-independent pathway in HepG2 cells. Mol Nutr Food Res 2017; 61
  201. Cui CJ, Jin JL, Guo LN, Sun J, Wu NQ, Guo YL, Liu G, Dong Q, Li JJ. Beneficial impact of epigallocatechingallate on LDL-C through PCSK9/LDLR pathway by blocking HNF1alpha and activating FoxO3a. J Transl Med 2020; 18:195
  202. Koo SI, Noh SK. Green tea as inhibitor of the intestinal absorption of lipids: potential mechanism for its lipid-lowering effect. J Nutr Biochem 2007; 18:179-183
  203. Onakpoya I, Spencer E, Heneghan C, Thompson M. The effect of green tea on blood pressure and lipid profile: a systematic review and meta-analysis of randomized clinical trials. Nutr Metab Cardiovasc Dis 2014; 24:823-836
  204. Zheng XX, Xu YL, Li SH, Liu XX, Hui R, Huang XH. Green tea intake lowers fasting serum total and LDL cholesterol in adults: a meta-analysis of 14 randomized controlled trials. Am J Clin Nutr 2011; 94:601-610
  205. Hartley L, Flowers N, Holmes J, Clarke A, Stranges S, Hooper L, Rees K. Green and black tea for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 2013:CD009934
  206. Zhao Y, Asimi S, Wu K, Zheng J, Li D. Black tea consumption and serum cholesterol concentration: Systematic review and meta-analysis of randomized controlled trials. Clin Nutr 2015; 34:612-619
  207. Liu W, Wan C, Huang Y, Li M. Effects of tea consumption on metabolic syndrome: A systematic review and meta-analysis of randomized clinical trials. Phytother Res 2020; 34:2857-2866
  208. Yuan F, Dong H, Fang K, Gong J, Lu F. Effects of green tea on lipid metabolism in overweight or obese people: A meta-analysis of randomized controlled trials. Mol Nutr Food Res 2018; 62
  209. Igho-Osagie E, Cara K, Wang D, Yao Q, Penkert LP, Cassidy A, Ferruzzi M, Jacques PF, Johnson EJ, Chung M, Wallace T. Short-Term Tea Consumption Is Not Associated with a Reduction in Blood Lipids or Pressure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Nutr 2020; 150:3269-3279
  210. Momose Y, Maeda-Yamamoto M, Nabetani H. Systematic review of green tea epigallocatechin gallate in reducing low-density lipoprotein cholesterol levels of humans. Int J Food Sci Nutr 2016; 67:606-613
  211. Araya-Quintanilla F, Gutierrez-Espinoza H, Moyano-Galvez V, Munoz-Yanez MJ, Pavez L, Garcia K. Effectiveness of black tea versus placebo in subjects with hypercholesterolemia: A PRISMA systematic review and meta-analysis. Diabetes Metab Syndr 2019; 13:2250-2258
  212. Wang D, Chen C, Wang Y, Liu J, Lin R. Effect of black tea consumption on blood cholesterol: a meta-analysis of 15 randomized controlled trials. PLoS One 2014; 9:e107711
  213. Kim A, Chiu A, Barone MK, Avino D, Wang F, Coleman CI, Phung OJ. Green tea catechins decrease total and low-density lipoprotein cholesterol: a systematic review and meta-analysis. J Am Diet Assoc 2011; 111:1720-1729
  214. Xu R, Yang K, Li S, Dai M, Chen G. Effect of green tea consumption on blood lipids: a systematic review and meta-analysis of randomized controlled trials. Nutr J 2020; 19:48
  215. O'Keefe JH, Bhatti SK, Patil HR, DiNicolantonio JJ, Lucan SC, Lavie CJ. Effects of habitual coffee consumption on cardiometabolic disease, cardiovascular health, and all-cause mortality. J Am Coll Cardiol 2013; 62:1043-1051
  216. Urgert R, Katan MB. The cholesterol-raising factor from coffee beans. Annu Rev Nutr 1997; 17:305-324
  217. Jee SH, He J, Appel LJ, Whelton PK, Suh I, Klag MJ. Coffee consumption and serum lipids: a meta-analysis of randomized controlled clinical trials. Am J Epidemiol 2001; 153:353-362
  218. Latham LS, Hensen ZK, Minor DS. Chocolate--guilty pleasure or healthy supplement? J Clin Hypertens (Greenwich) 2014; 16:101-106
  219. Hooper L, Kay C, Abdelhamid A, Kroon PA, Cohn JS, Rimm EB, Cassidy A. Effects of chocolate, cocoa, and flavan-3-ols on cardiovascular health: a systematic review and meta-analysis of randomized trials. Am J Clin Nutr 2012; 95:740-751
  220. Lin X, Zhang I, Li A, Manson JE, Sesso HD, Wang L, Liu S. Cocoa Flavanol Intake and Biomarkers for Cardiometabolic Health: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Nutr 2016; 146:2325-2333
  221. Shrime MG, Bauer SR, McDonald AC, Chowdhury NH, Coltart CE, Ding EL. Flavonoid-rich cocoa consumption affects multiple cardiovascular risk factors in a meta-analysis of short-term studies. J Nutr 2011; 141:1982-1988
  222. Tokede OA, Gaziano JM, Djousse L. Effects of cocoa products/dark chocolate on serum lipids: a meta-analysis. Eur J Clin Nutr 2011; 65:879-886
  223. Rimm EB, Williams P, Fosher K, Criqui M, Stampfer MJ. Moderate alcohol intake and lower risk of coronary heart disease: meta-analysis of effects on lipids and haemostatic factors. BMJ 1999; 319:1523-1528
  224. Brien SE, Ronksley PE, Turner BJ, Mukamal KJ, Ghali WA. Effect of alcohol consumption on biological markers associated with risk of coronary heart disease: systematic review and meta-analysis of interventional studies. BMJ 2011; 342:d636
  225. Spaggiari G, Cignarelli A, Sansone A, Baldi M, Santi D. To beer or not to beer: A meta-analysis of the effects of beer consumption on cardiovascular health. PLoS One 2020; 15:e0233619
  226. Brinton EA. Effects of ethanol intake on lipoproteins. Curr Atheroscler Rep 2012; 14:108-114
  227. Badia RR, Pradhan RV, Ayers CR, Chandra A, Rohatgi A. The Relationship of Alcohol Consumption and HDL Metabolism in the Multiethnic Dallas Heart Study. J Clin Lipidol 2023; 17:124-130
  228. Kovar J, Zemankova K. Moderate alcohol consumption and triglyceridemia. Physiol Res 2015; 64:S371-375
  229. Klop B, do Rego AT, Cabezas MC. Alcohol and plasma triglycerides. Curr Opin Lipidol 2013; 24:321-326
  230. Cesena FH, Coimbra SR, Andrade AC, da Luz PL. The relationship between body mass index and the variation in plasma levels of triglycerides after short-term red wine consumption. J Clin Lipidol 2011; 5:294-298
  231. Ginsberg H, Olefsky J, Farquhar JW, Reaven GM. Moderate ethanol ingestion and plasma triglyceride levels. A study in normal and hypertriglyceridemic persons. Ann Intern Med 1974; 80:143-149
  232. De Oliveira ESER, Foster D, McGee Harper M, Seidman CE, Smith JD, Breslow JL, Brinton EA. Alcohol consumption raises HDL cholesterol levels by increasing the transport rate of apolipoproteins A-I and A-II. Circulation 2000; 102:2347-2352
  233. You M, Arteel GE. Effect of ethanol on lipid metabolism. J Hepatol 2019; 70:237-248
  234. Sozio M, Crabb DW. Alcohol and lipid metabolism. Am J Physiol Endocrinol Metab 2008; 295:E10-16
  235. Rees K, Takeda A, Martin N, Ellis L, Wijesekara D, Vepa A, Das A, Hartley L, Stranges S. Mediterranean-style diet for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev 2019; 3:CD009825
  236. Ge L, Sadeghirad B, Ball GDC, da Costa BR, Hitchcock CL, Svendrovski A, Kiflen R, Quadri K, Kwon HY, Karamouzian M, Adams-Webber T, Ahmed W, Damanhoury S, Zeraatkar D, Nikolakopoulou A, Tsuyuki RT, Tian J, Yang K, Guyatt GH, Johnston BC. Comparison of dietary macronutrient patterns of 14 popular named dietary programmes for weight and cardiovascular risk factor reduction in adults: systematic review and network meta-analysis of randomised trials. BMJ 2020; 369:m696
  237. Harsha DW, Sacks FM, Obarzanek E, Svetkey LP, Lin PH, Bray GA, Aickin M, Conlin PR, Miller ER, 3rd, Appel LJ. Effect of dietary sodium intake on blood lipids: results from the DASH-sodium trial. Hypertension 2004; 43:393-398
  238. Siervo M, Lara J, Chowdhury S, Ashor A, Oggioni C, Mathers JC. Effects of the Dietary Approach to Stop Hypertension (DASH) diet on cardiovascular risk factors: a systematic review and meta-analysis. Br J Nutr 2015; 113:1-15
  239. Chiavaroli L, Viguiliouk E, Nishi SK, Blanco Mejia S, Rahelic D, Kahleova H, Salas-Salvado J, Kendall CW, Sievenpiper JL. DASH Dietary Pattern and Cardiometabolic Outcomes: An Umbrella Review of Systematic Reviews and Meta-Analyses. Nutrients 2019; 11
  240. Lari A, Sohouli MH, Fatahi S, Cerqueira HS, Santos HO, Pourrajab B, Rezaei M, Saneie S, Rahideh ST. The effects of the Dietary Approaches to Stop Hypertension (DASH) diet on metabolic risk factors in patients with chronic disease: A systematic review and meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 2021; 31:2766-2778
  241. Chiavaroli L, Nishi SK, Khan TA, Braunstein CR, Glenn AJ, Mejia SB, Rahelic D, Kahleova H, Salas-Salvado J, Jenkins DJA, Kendall CWC, Sievenpiper JL. Portfolio Dietary Pattern and Cardiovascular Disease: A Systematic Review and Meta-analysis of Controlled Trials. Prog Cardiovasc Dis 2018; 61:43-53
  242. Ramezani-Jolfaie N, Mohammadi M, Salehi-Abargouei A. The effect of healthy Nordic diet on cardio-metabolic markers: a systematic review and meta-analysis of randomized controlled clinical trials. Eur J Nutr 2019; 58:2159-2174
  243. Massara P, Zurbau A, Glenn AJ, Chiavaroli L, Khan TA, Viguiliouk E, Mejia SB, Comelli EM, Chen V, Schwab U, Riserus U, Uusitupa M, Aas AM, Hermansen K, Thorsdottir I, Rahelic D, Kahleova H, Salas-Salvado J, Kendall CWC, Sievenpiper JL. Nordic dietary patterns and cardiometabolic outcomes: a systematic review and meta-analysis of prospective cohort studies and randomised controlled trials. Diabetologia 2022; 65:2011-2031
  244. Kirkpatrick CF, Bolick JP, Kris-Etherton PM, Sikand G, Aspry KE, Soffer DE, Willard KE, Maki KC. Review of current evidence and clinical recommendations on the effects of low-carbohydrate and very-low-carbohydrate (including ketogenic) diets for the management of body weight and other cardiometabolic risk factors: A scientific statement from the National Lipid Association Nutrition and Lifestyle Task Force. J Clin Lipidol 2019; 13:689-711 e681
  245. Kwiterovich PO, Jr., Vining EP, Pyzik P, Skolasky R, Jr., Freeman JM. Effect of a high-fat ketogenic diet on plasma levels of lipids, lipoproteins, and apolipoproteins in children. JAMA 2003; 290:912-920
  246. Buga A, Welton GL, Scott KE, Atwell AD, Haley SJ, Esbenshade NJ, Abraham J, Buxton JD, Ault DL, Raabe AS, Noakes TD, Hyde PN, Volek JS, Prins PJ. The Effects of Carbohydrate versus Fat Restriction on Lipid Profiles in Highly Trained, Recreational Distance Runners: A Randomized, Cross-Over Trial. Nutrients 2022; 14
  247. Buren J, Ericsson M, Damasceno NRT, Sjodin A. A Ketogenic Low-Carbohydrate High-Fat Diet Increases LDL Cholesterol in Healthy, Young, Normal-Weight Women: A Randomized Controlled Feeding Trial. Nutrients 2021; 13
  248. Creighton BC, Hyde PN, Maresh CM, Kraemer WJ, Phinney SD, Volek JS. Paradox of hypercholesterolaemia in highly trained, keto-adapted athletes. BMJ Open Sport Exerc Med 2018; 4:e000429
  249. Retterstol K, Svendsen M, Narverud I, Holven KB. Effect of low carbohydrate high fat diet on LDL cholesterol and gene expression in normal-weight, young adults: A randomized controlled study. Atherosclerosis 2018; 279:52-61
  250. Lima PA, de Brito Sampaio LP, Damasceno NR. Ketogenic diet in epileptic children: impact on lipoproteins and oxidative stress. Nutr Neurosci 2015; 18:337-344
  251. Joo M, Moon S, Lee YS, Kim MG. Effects of very low-carbohydrate ketogenic diets on lipid profiles in normal-weight (body mass index < 25 kg/m2) adults: a meta-analysis. Nutr Rev 2023; 81:1393-1401
  252. Castellana M, Conte E, Cignarelli A, Perrini S, Giustina A, Giovanella L, Giorgino F, Trimboli P. Efficacy and safety of very low calorie ketogenic diet (VLCKD) in patients with overweight and obesity: A systematic review and meta-analysis. Rev Endocr Metab Disord 2020; 21:5-16
  253. Norwitz NG, Feldman D, Soto-Mota A, Kalayjian T, Ludwig DS. Elevated LDL Cholesterol with a Carbohydrate-Restricted Diet: Evidence for a "Lean Mass Hyper-Responder" Phenotype. Curr Dev Nutr 2022; 6:nzab144
  254. Houttu V, Grefhorst A, Cohn DM, Levels JHM, Roeters van Lennep J, Stroes ESG, Groen AK, Tromp TR. Severe Dyslipidemia Mimicking Familial Hypercholesterolemia Induced by High-Fat, Low-Carbohydrate Diets: A Critical Review. Nutrients 2023; 15
  255. Norwitz NG, Mindrum MR, Giral P, Kontush A, Soto-Mota A, Wood TR, D'Agostino DP, Manubolu VS, Budoff M, Krauss RM. Elevated LDL-cholesterol levels among lean mass hyper-responders on low-carbohydrate ketogenic diets deserve urgent clinical attention and further research. J Clin Lipidol 2022; 16:765-768
  256. Goldberg IJ, Ibrahim N, Bredefeld C, Foo S, Lim V, Gutman D, Huggins LA, Hegele RA. Ketogenic diets, not for everyone. J Clin Lipidol 2021; 15:61-67
  257. Schaffer AE, D'Alessio DA, Guyton JR. Extreme elevations of low-density lipoprotein cholesterol with very low carbohydrate, high fat diets. J Clin Lipidol 2021; 15:525-526
  258. Schmidt T, Harmon DM, Kludtke E, Mickow A, Simha V, Kopecky S. Dramatic elevation of LDL cholesterol from ketogenic-dieting: A Case Series. Am J Prev Cardiol 2023; 14:100495
  259. Foster GD, Wyatt HR, Hill JO, Makris AP, Rosenbaum DL, Brill C, Stein RI, Mohammed BS, Miller B, Rader DJ, Zemel B, Wadden TA, Tenhave T, Newcomb CW, Klein S. Weight and metabolic outcomes after 2 years on a low-carbohydrate versus low-fat diet: a randomized trial. Ann Intern Med 2010; 153:147-157
  260. Yokoyama Y, Levin SM, Barnard ND. Association between plant-based diets and plasma lipids: a systematic review and meta-analysis. Nutr Rev 2017; 75:683-698
  261. Wang F, Zheng J, Yang B, Jiang J, Fu Y, Li D. Effects of Vegetarian Diets on Blood Lipids: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Am Heart Assoc 2015; 4:e002408
  262. Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ. Comparison of the Atkins, Ornish, Weight Watchers, and Zone diets for weight loss and heart disease risk reduction: a randomized trial. JAMA 2005; 293:43-53
  263. Ference BA, Ray KK, Catapano AL, Ference TB, Burgess S, Neff DR, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Kastelein JJP, Nicholls SJ. Mendelian Randomization Study of ACLY and Cardiovascular Disease. N Engl J Med 2019; 380:1033-1042
  264. Ference BA, Kastelein JJP, Ray KK, Ginsberg HN, Chapman MJ, Packard CJ, Laufs U, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Nicholls SJ, Bhatt DL, Sabatine MS, Catapano AL. Association of Triglyceride-Lowering LPL Variants and LDL-C-Lowering LDLR Variants With Risk of Coronary Heart Disease. JAMA 2019; 321:364-373
  265. Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, Himmelfarb CD, Khera A, Lloyd-Jones D, McEvoy JW, Michos ED, Miedema MD, Munoz D, Smith SC, Jr., Virani SS, Williams KA, Sr., Yeboah J, Ziaeian B. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019; 140:e596-e646
  266. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Group ESCSD. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  267. Kirkpatrick CF, Sikand G, Petersen KS, Anderson CAM, Aspry KE, Bolick JP, Kris-Etherton PM, Maki KC. Nutrition interventions for adults with dyslipidemia: A Clinical Perspective from the National Lipid Association. J Clin Lipidol 2023; 17:428-451
  268. Hunninghake DB, Stein EA, Dujovne CA, Harris WS, Feldman EB, Miller VT, Tobert JA, Laskarzewski PM, Quiter E, Held J, et al. The efficacy of intensive dietary therapy alone or combined with lovastatin in outpatients with hypercholesterolemia. N Engl J Med 1993; 328:1213-1219
  269. Subramanian S. Hypertriglyceridemia: Pathophysiology, Role of Genetics, Consequences, and Treatment. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland J, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Purnell J, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  270. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res 2016; 57:1111-1125
  271. Law HG, Meyers FJ, Berglund L, Enkhmaa B. Lipoprotein(a) and diet-a challenge for a role of saturated fat in cardiovascular disease risk reduction? Am J Clin Nutr 2023; 118:23-26
  272. Rees K, Dyakova M, Ward K, Thorogood M, Brunner E. Dietary advice for reducing cardiovascular risk. Cochrane Database Syst Rev 2013:CD002128
  273. Ross LJ, Barnes KA, Ball LE, Mitchell LJ, Sladdin I, Lee P, Williams LT. Effectiveness of dietetic consultation for lowering blood lipid levels in the management of cardiovascular disease risk: A systematic review and meta-analysis of randomised controlled trials. Nutr Diet 2019; 76:199-210
  274. Moller G, Andersen HK, Snorgaard O. A systematic review and meta-analysis of nutrition therapy compared with dietary advice in patients with type 2 diabetes. Am J Clin Nutr 2017; 106:1394-1400

Osteoporosis in Men

ABSTRACT

 

While progress has been made, osteoporosis in men is still under-diagnosed and under-treated.  In general, men fracture about 10 years later than women, with large increases in fracture risk after about age 75, although a small number of men may present with vertebral fractures in middle age. There is overlap between secondary causes of osteoporosis and risk factors for primary osteoporosis, but men with fragility fractures or low bone density require evaluation by history and physical examination as well as a short list of laboratory tests. Bone mineral density by dual-energy x-ray absorptiometry remains the best test for diagnosing osteoporosis in men, although opportunistic bone density measurements from CT scans are promising. Clinicians should recommend a comprehensive program of treatment with fall risk reduction, attention to diet and vitamin D status, and pharmacologic treatment. In general, medications that work in women should lead to fewer fractures in men, although there are few studies in men with fracture risk reduction as the primary outcome. Most men with osteoporosis should be treated with oral or intravenous bisphosphonates, but men at very high fracture risk should be considered for initial anabolic treatment. Compared to women, men are more likely to die after hip fracture. The long-term management of men with osteoporosis is based solely on a few studies in women.

 

INTRODUCTION

 

Despite new information and even some attention in popular publications, osteoporosis in men remains under-appreciated, under-diagnosed, and under-treated. While the evidence base for evaluation and management of male osteoporosis will always be less than that of female osteoporosis, there is enough information available to identify those men at highest risk, evaluate them thoroughly, and treat them with a program that will reduce osteoporotic fractures.  Nonetheless, there are many impediments to quality care at all stages: recognition, diagnosis, assessment, and management (both short- and long-term). In this chapter, the challenges for the primary care and specialty clinician will be addressed with the purpose of providing an approach to reducing osteoporotic fracture in men.

 

DEFINITION, CLASSIFICATION, AND EPIDEMIOLOGY OF MALE OSTEOPOROSIS

 

Definitions of Osteoporosis in Men

 

In an older adult, regardless of gender, a fall from a standing position should not result in a fracture. Hence, one definition of osteoporosis is just such a fracture. By consensus, some fractures are considered osteoporotic; and others may or may not be, even if they occur with minimal trauma. For the most commonly used fracture risk calculator FRAX (see below), low trauma fractures of the spine, hip, forearm (radius and ulna), and humerus are considered osteoporotic. Pelvic, rib, and sternal fractures may also be osteoporotic. Most authorities do not count skull or digital fractures, and ankle fractures are the most controversial. Interestingly, in a study (1) of older men (MrOS, see below), any fracture after age 50 increased the risk of osteoporotic hip fracture, when combined with bone mineral density (BMD) measured by dual energy x-ray absorptiometry (DXA). The above is compatible with the standard definition of osteoporosis as compromised bone strength leading to increased risk of low trauma fracture (2).  A more operational diagnosis relies on DXA measurements, with a BMD T-score of -2.5 or worse in the spine or hip serving as the diagnosis of osteoporosis (3). This means that the patient’s BMD is at least 2.5 standard deviations below the normal young mean. As the BMD decreases, the fracture risk rises markedly. In men there has been great controversy about the normative database that should be used for the calculation of T-scores. Based on the fact that men and women fracture at similar (overlapping but not quite identical) absolute bone density measurements (in g/cm2), several major osteoporosis organizations, including the International Society for Clinical Densitometry (ISCD), recommend use of the young, white female normative database for all T-score calculations (4). The reader is directed to a discussion of this subject (5), and more details about DXA are discussed below. While the man with a T-score of -2.5 or less is clearly at the highest risk for fracture, more fractures occur in men with T-scores between -1 and -2.5, what is called osteopenia or low bone mass. The reason for this is that there are many more men in this category. For example, baseline DXA testing was done in the Rotterdam study (6), a large, long-term observational study. In men, 29% of hip fractures were in those with osteoporosis by DXA, 64% had osteopenia, and 7% had normal bone density. DXA measures bone quantity, and fracture risk is also determined by bone quality, which is impossible to measure definitively with current clinical tools. Thus, fracture risk calculators have been established, based on epidemiological data, to reflect bone quality and add to the predictive power of DXA. The most commonly used fracture risk calculator is FRAX (7), available online as www.sheffield.ac.uk/FRAX/. FRAX calculates the 10-year risk of hip fracture and of major osteoporotic fracture (MOF) based on the femoral neck BMD in g/cm2 plus a series of risk factors: age, sex, previous fracture, parental hip fracture, current smoking, having more than 3 alcoholic drinks daily, rheumatoid arthritis, exposure to systemic glucocorticoid drugs, and secondary osteoporosis. It also can be calculated using the body mass index (BMI) as a surrogate for femoral neck BMD.  While some studies (e.g. 8) suggest that FRAX works better in women than men, the calculator has been adopted internationally. There are other risk calculators, such as the Garvan nomogram (9), which unlike FRAX includes falls as a risk factor for determining fracture risk. It is interesting to note that at age 50, a man has a risk of experiencing an osteoporotic fracture of 13 to 25%, depending on the population studied. A much smaller percentage of men over age 50 have T-scores of -2.5 or worse, although the proportion increases with age. In a study of NHANES data, osteoporosis was defined from FRAX calculations: a 10-year hip fracture risk of > 3% or MOF of > 20% (10,11,12). Using this definition 16% of American men at age 50 and 46% at age 80 met criteria for osteoporosis, much more similar to actual incidences of osteoporotic fracture (12). There is some evidence (e.g. 13) that treating women who meet this fracture risk criterion respond to current osteoporosis treatment. There are, to my knowledge, no studies in men that show that diagnosing osteoporosis in a man by this method and treating him with standard medication leads to fewer fractures. Indeed Ensrud (14) has reported that men with osteoporosis by DXA have the best response to osteoporosis treatment, compared to those with better BMD.  However, as will be described below, studies of osteoporosis medications in men have almost always used the more liberal male normative database for the calculation of the T-score and accepted men with osteopenia plus a history of an osteoporotic fracture for inclusion. In these studies, such men responded to the treatment regimen with improvements in the standard surrogates for fracture. It is also interesting that the Rotterdam study (6) mentioned above also used sex-specific normative databases for the DXA diagnosis of osteoporosis. Had they used the female database for all participants, the group with osteoporosis by DXA at baseline would have accounted for an even smaller percentage of the hip fractures observed. A practical approach to the diagnosis is provided below.

 

There are other potential tools for determining fracture risk. For example, FRAX Plus (15) will be released soon. It will add falls, diabetes mellitus, and other risk factors to the fracture risk prediction. Trabecular Bone Score (16) can be derived from DXA of the spine. It reflects bone architecture and can be added to FRAX calculations. It is thought to be a reflection of bone quality (17). The reader is directed to the chapters on osteoporosis in women, which will include other methods to better quantify fracture risk.

 

Epidemiology of Osteoporosis in Men

 

Fractures in men occur about 10 years later in life than in women (18). Men, with generally bigger bones, have more to lose over time. In addition, men do not undergo the rapid increase of bone turnover that occurs with menopause and the marked drop in estradiol secretion.  Instead, it is well-accepted that the loss of sex steroids in men is a much more gradual process (19), and it is interesting to note that, with aging, BMD is more closely associated with serum bioavailable estradiol levels than with any serum measure of testosterone (20). Nonetheless, in middle-aged men presenting usually with vertebral fractures or low spine BMD by DXA, one of the causes of osteoporosis earlier in life is hypogonadism. This type of osteoporosis is analogous to what Riggs and Melton labelled postmenopausal osteoporosis in a seminal paper (21) many years ago. They described osteoporosis in women soon after menopause as loss of mostly trabecular bone (and thus vertebrae were particularly at risk) and associated with the dramatic drop in ovarian estrogen production. Men with organic causes of hypogonadism (for example, pituitary tumors) may also present with very low serum testosterone levels and osteoporosis. There are other causes of this earlier type of osteoporosis in men, including hypercalciuria (22) and secondary causes, which may not be very apparent clinically.  An example of the latter is celiac disease, which may not bring the patient to clinical attention but can lead to early fracture risk. (See below for other secondary causes of osteoporosis in men).  Finally, there have been reports of genetic disorders leading to so-called idiopathic osteoporosis in men, such as low levels of IGF-I without abnormalities in growth hormone (23) and low serum bioavailable estradiol levels (24). It is much more likely for a man to experience an osteoporotic fracture after age 75 than at middle age, but the clinician needs to know that early osteoporosis occurs and that it should lead to evaluation and treatment.

 

The majority of fractures in men occur later in life. The Rotterdam Study (6) assessed only nonvertebral fractures because the date of vertebral fractures was much more difficult to ascertain. In men the incidence of nonvertebral fracture accelerates after about age 75. The incidence of nonvertebral fractures in men at ages 80 to 84 is about the same as the incidence in women ages 70 to 74. This observation is the basis for stating that fractures occur about 10 years later in men than women, and it may explain why men come to fracture with more co-morbidities than women, a possible explanation for why men do relatively poorly after hip fracture in particular. As used in FRAX, risk factors for fracture, presumably reflecting bone quality, magnify the impact of bone quantity (DXA) on fracture risk. In the FRAX calculation age, prior fracture, and history of parental fracture are the most important variables. Not well known is a report (25) from Leslie and colleagues proposing that the age at which a parent has fractured a hip is important. If the parent has fractured before age 80, this adds greatly to the patient’s risk of fracture, whereas if the parental hip fracture occurred late in life, the impact on fracture risk is much less. The analogy with familial heart disease is striking: early heart disease, particularly in a patient’s mother, makes the patient at much increased risk for cardiac events. 

 

Risk Factors and Secondary Causes of Osteoporosis

 

Table 1 summarizes potential risk factors and secondary causes of osteoporosis, most of which pertain to men as well as to women. Aspects specific to men are discussed below.

 

Table 1. Conditions, Diseases and Medications that Cause or Contribute to Osteoporosis and Fractures

Lifestyle Factors

Low Calcium Intake

Vitamin D Insufficiency

Excess Vitamin A

High Caffeine Intake

High Salt Intake

Aluminum (in antacids)

Inadequate Physical Activity

Immobilization

Smoking

Falling

Thinness

Alcoholism

Genetic Factors

Cystic Fibrosis

Homocystinuria

Osteogenesis Imperfecta

Ehlers-Danlos Syndrome

Hypophosphatasia

Gaucher’s Disease

Idiopathic Hypercalciuria

Porphyria

Glycogen storage diseases

Marfan Syndrome

Riley-Day Syndrome

Hemochromatosis

Menkes Steely Hair Syndrome

Parental History of Hip Fracture

Androgen Insensitivity

Turner’s & Klinefelter’s Syndromes

Endocrine Disorders

Adrenal Insufficiency

Diabetes Mellitus

Hyperthyroidism

Cushing’s Syndrome

Hyperparathyroidism

Hypogonadal States

Panhypopituitarism

Athletic Amenorrhea

Anorexia Nervosa and Bulimia

Hyperprolactinemia

Premature Ovarian Failure

 

Gastrointestinal disorders

Celiac Disease

Inflammatory Bowel Disease

Primary Biliary Cirrhosis

Gastric Bypass

Malabsorption

GI Surgery

Pancreatic Disease

 

Hematologic Disorders

Hemophilia

Multiple Myeloma

Systemic Mastocytosis

Leukemia

Lymphoma

Sickle Cell Disease

Thalassemia

 

Rheumatic and Autoimmune Diseases

Ankylosing Spondylitis

Lupus

Rheumatoid Arthritis

 

Miscellaneous Conditions and Diseases

Chronic Obstructive Pulmonary Disease

Muscular Dystrophy

Amyloidosis

End Stage Renal Disease

Parenteral Nutrition

Chronic Metabolic Acidosis

Epilepsy

Post-Transplant Bone Disease

Congestive Heart Failure

Idiopathic Scoliosis

Prior Fracture as an Adult

Depression

Multiple Sclerosis

Sarcoidosis

HIV/AIDS

 

Medications

Anticoagulants (heparin)

Cancer Chemotherapeutic Drugs

Gonadotropin Releasing Hormone Agonists

Anticonvulsants

Lithium

Aromatase Inhibitors

Depo-medroxyprogesterone

Barbiturates

Glucocorticoids (> 5mg of prednisone or equivalent for > 3 months)

Cyclosporine A

Tacrolimus

 

Table from the Endotext chapter entitled “Osteoporosis: Clinical Evaluation” by E. Michael Lewiecki.

 

The Osteoporotic Fractures in Men Study (MrOS) has provided a great deal of information. This long-term US observational study included about 6000 men for more than 15 years (26). Of the many important findings from the study, one is of particular interest. What are the characteristics of men, in addition to DXA, that predict hip fracture?  In this excellent report (1), several surprising factors were discovered and others were expected. Of the latter group, age >75, current smoking, Parkinson’s disease, hyperthyroidism, hyperparathyroidism, and decreased cognitive function were risk factors that greatly increased the fracture risk prediction, when added to BMD. More interestingly, several other risk factors were found: low dietary protein, any fracture after age 50, divorce, tricyclic anti-depressants or hypoglycemic agents, tall stature, and the inability to do chair stands. Having 4 of these risk factors increased hip fracture risk 5-fold in men with osteoporosis by DXA.

 

Secondary causes of osteoporosis are thought to be particularly important in men, but there is overlap between what might be called a secondary cause of osteoporosis and in another context a risk factor for primary osteoporosis. In addition, while treatment of a secondary cause may be adequate to lower fracture risk, a man will possibly be at risk for primary osteoporosis as he ages – and need osteoporosis specific treatment. Hyperthyroidism, hyperparathyroidism, and hypercalciuria are well-characterized secondary causes of osteoporosis in men.  A particularly important cause is glucocorticoid excess, usually due to treatment of an inflammatory disorder with systemic glucocorticoids. Glucocorticoid-induced osteoporosis (GIOP) is considered the most important medication-related type of osteoporosis and is of particular concern because fracture risk is increased (27) after 3 months of prednisone equivalent doses of 5 to 7.5 mg daily – and maybe earlier (28) and maybe even lower doses.  There is evidence that men are less likely to be evaluated and treated for GIOP (29), perhaps because clinicians again do not think that osteoporosis happens in men. While endogenous Cushing’s syndrome leads to GIOP, most cases are due to exogenous glucocorticoids, and about 1% of the adult population may be taking such medications at any particular time. 

 

Multiple myeloma may present with osteoporosis-like vertebral fractures; hence, this diagnosis must be in the differential diagnosis of the new patient presenting this way. Malabsorption, particularly celiac disease, is another potential secondary cause of osteoporosis. While type 2 diabetes mellitus is clearly associated with increased fracture risk (30), bone density is usually not decreased, whereas in type 1 diabetes mellitus, BMD is variable. Celiac disease is associated with type 1 diabetes mellitus, and thus it should be considered in men with type 1 diabetes mellitus and a fracture. Mastocytosis is associated with osteoporosis, although the mechanism is not fully understood. Hemochromatosis, presumably via some of its consequences is also on the list of secondary causes. Immobilization leads to loss of bone.  Spinal cord injury is much more common in men than women, and bone is lost distal to the cord lesion and may be worse than immobilization per se because of comorbidities (31). The fracture risk is high in men with spinal cord injury, and other types of decreased mobility should be considered when assessing men: stroke, Parkinson’s disease, multiple sclerosis, etc. 

 

Men with HIV now have life expectancies close to those without HIV, but the risk of osteoporosis and fracture is greater. Fractures appear about 10 years earlier than in men without HIV.  In a systematic review of large numbers of people living with HIV (mostly men), the relative risk of a low trauma fracture was 1.51 and the relative risk of a hip fracture was 4.09 (32). In a meta-analysis of bone mineral density testing (33), low bone mass (osteopenia) and osteoporosis by dual energy x-ray absorptiometry (DXA) was more prevalent in persons (again mostly men) with HIV compared to non-infected controls.  Interestingly, initiation of antiretroviral therapy (ART) appeared to be associated with lower bone density, confirmed by a subsequent randomized trial (34). ART with tenofovir alafenamide appears to have better renal safety than tenofovir disoproxil fumarate (35) and may have less impact on bone. Fortunately, men with HIV appear to response well to bisphosphonate therapy for osteoporosis 36).    

 

Osteoporosis and Hypogonadism

 

As mentioned above, men with organic causes of hypogonadism are at risk for fracture and may present at middle age or later with fracture or low BMD. More common and more controversial is the parallel decrease in BMD and serum testosterone levels with aging. While not proven, it is reasonable to assume that as testosterone and muscle mass diminish with age, falls will increase, leading to fractures. The relationship between serum testosterone and BMD is less clear. As previously mentioned, Khosla and colleagues reported (20) that BMD was more strongly related to bioavailable estradiol levels than any measure of testosterone. Of course, in men the major source of estradiol is aromatization of testosterone. The importance of estrogen is illustrated by the impact of iatrogenic hypogonadism. Prostate cancer often responds to androgen withdrawal, and for men with rising PSA levels or other evidence of recurrence or spread, androgen withdrawal may be accomplished by orchiectomy (not done very often today) or by use of analogs of gonadotropin releasing hormone (GnRH).  Some GnRH analogs acutely increase gonadotropins LH and FSH such that an androgen receptor blocker such as bicalutamide or nilutamide is given on a short-term basis until the pituitary is down regulated.  GnRH analog treatment results in serum testosterone levels that are essentially zero and in very low levels of estradiol (the remaining estrogens are presumably from conversion of adrenal androgens). Some men are treated with an androgen blocker alone. In most studies (37) bone loss is much more profound in the men treated with GnRH analogs compared to men treated with only androgen receptor blockers, who have normal estradiol levels. Abiraterone (38) blocks conversion of precursors to androgens and may be used in concert with GnRH analogs.  Prednisone is needed to prevent mineralocorticoid excess due to the enzyme blockage caused by abiraterone. The dose is 5 mg twice daily, a little more than a replacement dose. This potentially adds to the risk that men treated with this combined androgen deprivation therapy (ADT) will have particularly increased fracture risk. However, the most widely cited study (39) of fracture in men on ADT is several years old, done before abiraterone was approved. The important finding from this study was that while ADT given to a man who has a rising PSA level after primary treatment of prostate cancer leads to a 10-year survival rate of 80 to 90%, the 5-year fracture rate was almost 20% in Caucasian men and 2/3 or ¾ of that rate in African-Americans. Thus, the profound hypogonadism of ADT is clearly a major risk for fracture. 

 

This still leaves unanswered whether testosterone given to men with decreased serum testosterone levels associated with aging would benefit from testosterone replacement. There are no studies large enough to show a fracture benefit of such treatment. In a careful study (40) of older men with low serum testosterone levels, testosterone gel or placebo gel was used for one year. At the end of the year, there was a modest increase in BMD by DXA and also by quantitative computed tomography (qCT). More importantly, there was an increase in bone strength by finite element analysis of the qCT data. The Endocrine Society Male Osteoporosis Guideline (41) states that older men at risk for fracture should be treated with osteoporosis-specific medications but those who also have symptomatic hypogonadism can be considered for testosterone replacement. The likely impact of testosterone deficiency on muscle and the bone strength response to testosterone replacement make it plausible that testosterone replacement will lead to fewer fractures. The TRAVERSE study (42) is a large study of testosterone replacement on cardiovascular safety in older hypogonadal men. There was no increase in cardiovascular events in the men treated with testosterone gel (43), nor was there evidence of increased prostate cancer risk or urinary retention (44). Interestingly, there were more fractures in the men receiving testosterone replacement (45). However, the fractures occurred soon after starting replacement, and the majority were ankle and risk fractures (45, 46). This suggested to Grossmann and Anawalt (46) that testosterone-induced changes in behavior may have been the etiology of the fracture increase.

 

SCREENING AND DIAGNOSTIC EVALUATION IN MEN

 

DXA Testing Men

 

From this extensive review of pathogenesis and epidemiology of osteoporosis in men, it is possible to postulate which men should be screened for osteoporosis and how they should be evaluated.  Age is a major risk factor for fracture.  At what age should a man undergo DXA testing and does such testing lead to fewer fractures? The Endocrine Society Guideline (41) suggests DXA testing in most men at age 70 or above. The United States Preventive Services Task Force (47) states that there is insufficient evidence to recommend DXA testing in men, although it supports DXA testing in women by age 65. There are few studies demonstrating that DXA screening in women leads to fewer fractures. The recent SCOOP study (48) from the UK revealed that a two-stage method of choosing women for testing by first calculating FRAX using BMI as a surrogate for femoral neck BMD resulted in fewer hip fractures. In this study, women at low risk for fracture by FRAX were not screened further. Those at high risk were treated, and those in the middle had a DXA. Based on DXA results and recalculation of FRAX with femoral neck DXA results, women at risk were placed on therapy and had fewer fracture than those not screened for osteoporosis. There are no similar prospective studies in men, but Colon-Emeric and colleagues (49) used the Department of Veterans Affairs and Medicare databases to determine the impact of screening men with DXA. Overall, screening did not lead to fewer fractures. However, strategic screening did.  Men aged 80 or older, men on systemic glucocorticoids or ADT, and men with FRAX calculated with BMI (somewhat like the SCOOP study women) had fewer fractures if they were screened by DXA. In addition, men over age 65 with several other risk factors (including rheumatoid arthritis, alcohol or tobacco abuse, chronic obstructive pulmonary disease, chronic liver disease, stroke, Parkinson’s disease, gastrectomy, hyperthyroidism, hyperparathyroidism, or traditional anti-seizure drug use) were also likely to benefit, should they have a DXA done. This study was observational and done with the Department of Veterans Affairs population, which tends to be sicker than the general population and from the population of the prospective study, MrOS. Nonetheless, the findings are compatible with the epidemiology of fractures in men and can serve as a basis for clinical care.  It is unrealistic to expect that a study like SCOOP will be done in men. The SCOOP population was about 12,500 women; a male version would likely need approximately 40,000 participants.  Based on the Colon-Emeric observational study (49) and studies from MrOS (1), Table 2 suggests which older men that should be screened for osteoporosis by DXA.

 

Table 2.  Which Men Should Be Screened (by DXA) for Osteoporosis?

Men > 50 Years Old

After a fragility fracture (usually vertebral in younger group)

On chronic glucocorticoids

Organic causes of hypogonadism

Hypercalciuria

Men > 65 Years Old

All of the above plus:

On androgen deprivation therapy for prostate cancer

High risk for fracture based on FRAX using BMI

Current smoking/COPD

Alcohol abuse/chronic liver disease

Rheumatoid arthritis

Parkinson’s disease or other mobility disorder

Gastrectomy/bariatric surgery

Hyperthyroidism

Hyperparathyroidism

On enzyme-inducing anti-seizure medications for > 2 years

Men > 80 Years Old

If not already screened, all men over 80 should have a DXA (unless there is a contraindication).

    

In the United States, reimbursement for DXA testing is limited. This may be one reason that so few men are assessed for fracture risk. One potential method to identify men at risk for fracture is to assess bone density from CT scans done for other reasons. There are several methods of so-called opportunistic bone density evaluation that have been used (e. g. 50), including a study done in male veterans (51). It is likely that artificial intelligence can be harnessed to make this process even more efficient. Whether finding men at risk this way will lead to more clinical evaluation and treatment and fewer fractures remains to be determined.

 

Beyond DXA: Laboratory Evaluation of Osteoporosis in Men

 

If a man has osteoporosis by DXA or meets other criteria for osteoporosis or has low bone mass (osteopenia) but may be at higher risk for fracture, what other tests should be done? Spine x-rays or vertebral fracture analysis (images of the spine by DXA machines) may reveal vertebral fractures that increase subsequent fracture risk. There are no specific blood tests for osteoporosis, and the evidence base for the tests that follow may be weak. Nonetheless, it makes clinical sense to do a few laboratory tests to look for secondary causes/risk factors for osteoporosis and to ensure the safety of treatment, should it be indicated. Many patients will have had some of these tests as part of their general medical care, so the actual addition to routine testing may be small. For all patients, assessments of serum calcium and phosphate and renal function are necessary to look for hypercalcemia (which might signal hyperparathyroidism) and to determine if some osteoporosis treatments can be safely given.  Avoiding controversies about ideal levels of serum 25-OH vitamin D in the general population, there is consensus that for the patient with osteoporosis, the target level should be 30 ng/ml (52). All of those tests mentioned may help to identify the unusual patient with osteomalacia.  Serum alkaline phosphatase reflects bone formation and turnover, among other things. It is interesting that low serum alkaline phosphatase may be a sign of hypophosphatasia (53), a disorder of variable severity that may present as osteoporosis. Such patients should not be treated with anti-resorptive agents. An automated complete blood count should be done, particularly if there is any suspicion of multiple myeloma because about 75% of such patients will have anemia. All of the above tests, with the exception of 25-OH vitamin D, may be done as routine screening tests in many people visiting primary care clinicians, although measurement of 25-OH vitamin D has become very common as well. Once the 25-OH vitamin D level is at goal, a 24-hour urine for calcium and creatinine (and possibly sodium) may help to signal hypercalciuria, or in in the case of low urinary calcium excretion, may reflect malabsorption. For a patient suspected of hyperparathyroidism or hyperthyroidism, appropriate testing for parathyroid hormone (PTH) or thyroid hormones/TSH should be done. Similarly, for patients in whom there is a suggestion of another secondary cause of osteoporosis, specific tests such as serum protein electrophoresis, celiac antibodies, cortisol, tryptase, etc. can be done.  More controversial is whether serum testosterone should be measured.  Most symptoms of hypogonadism are non-specific, such as fatigue. Decreased libido is considered the most specific symptom, but decreased muscle mass and decreased beard growth might be present.  For the symptomatic man, measurement of early morning testosterone is reasonable. Many experts may suggest measurement of free and bioavailable testosterone as well as gonadotropins. The diagnosis of hypogonadism requires two early morning (preferably fasting) testosterone measurements (54). We would also measure PSA and review the hematocrit and hemoglobin before considering testosterone replacement. In addition, measurement of testosterone should only be done if the clinician would consider testosterone replacement, likely in addition to an osteoporosis-specific treatment. In the Veterans Affairs population, routine laboratory testing was found to reveal new secondary causes and/or osteoporosis risk factors (55). In contrast, in the healthier MrOS cohort, routine testing was found to be less helpful (56).

 

Table 3.  Practical Approach to the Man with Osteoporosis

History and Physical Exam

           Evidence of secondary causes of osteoporosis, risk factors

           Family history

           Height versus maximum attained height

           Gait

           Kyphosis

           General condition of teeth

           Evidence of significant visual abnormalities

           Ability to rise from chair without using hands

           Tenderness to percussion of spine

Standard Laboratory Tests

            Serum Chemistries: Calcium, Phosphate, Alkaline Phosphatase, Albumin

            Measure of Renal Function (e.g. serum creatinine, eGFR)

            Complete blood count

            Serum 25-OH vitamin D

            When 25-OH vitamin D is at goal: 24-hour urine calcium, creatinine, and maybe sodium

Laboratory Tests in Specific Cases (triggered by history and physical exam)

            Thyroid function tests (TSH, Free T4, maybe Total T3)

            Parathyroid hormone (PTH)

            Ionized Calcium

            Total, Free, and Bioavailable Testosterone

            LH, FSH, Prolactin

            CTX or other marker of bone resorption

            Bone Specific Alkaline Phosphatase (or other marker of bone formation)

            Celiac antibodies

            Serum/Urine Protein Electrophoresis

            Magnesium

            Tryptase

            Tests for cortisol excess (e.g. urinary free cortisol, dexamethasone suppression test, midnight salivary cortisol)

Images          

            X-rays of thoracic and lumbar spine

            X-rays of fractured bone

            Pituitary imaging (usually MRI)

 

MANAGEMENT OF OSTEOPOROSIS IN MEN

 

Non-Pharmacologic Management of Osteoporosis

 

One criticism heard about current osteoporosis treatment is that it focuses only on pharmacologic methods. A more comprehensive approach to osteoporosis treatment is preferred. Indeed, there are ways to reduce fracture that do not involve prescription of drugs, and they should be an important part of the therapeutic regimen. While there has been controversy about the role of calcium and vitamin D on fracture risk and on potential side effects, such as cardiovascular events, discussion of these controversies can be found in other chapters. One recent meta-analysis (57) concluded that daily calcium and vitamin D are likely to be salutary for osteoporosis. The widely-cited Institute of Medicine report (58) suggested 1000 to 1200 mg of elemental calcium in the diet and vitamin D intake of 400 to 800 units per day. As stated above, most experts would suggest that a target vitamin D level of 30 ng/ml is reasonable for patients with osteoporosis. From MrOS (1) we learned that the protein content of the diet is also important. A liquid protein supplement might be a good source of calcium and protein for some older men. In my own experience, older men who live alone may have poor diets, and such protein supplements may be an easy way to augment their diet.

 

Fall risk reduction is also very important. In most cases, patients fall first, fracture second.  Thus, attention to eyesight, avoidance of drugs that affect standing blood pressure or cause sleepiness or confusion, and home safety are very important parts of a comprehensive osteoporosis treatment program. Treatment of cataracts, for example, leads to fewer fractures (59). In MrOS (1) use of hypoglycemic agents was associated with increased hip fracture risk.  People with seizure disorders fall; thus, control of epilepsy is important. Avoidance of alcohol, opiates, benzodiazepines, and psychiatric drugs is suggested, but of course some patients may require medications that can cause drowsiness or imbalance. Anti-hypertensive medications need to be titrated such that postural hypotension does not occur. Convincing a man to use a walking aid may be challenging. Night lights, elimination of loose throw rugs and extension cords, and care with pets are also important to avoid falls. Consultation with Occupational Therapy and/or Physical Therapy should be considered in many cases. Exercise prescriptions should aim to improve muscle strength as well as balance. Risk factors for falls are listed in table 4.

 

Table 4. Risk Factors for Falls Adapted From Guidelines of the National Osteoporosis Foundation

Environmental Risk Factors

Lack of assistive devices in bathrooms, loose throw rugs, low level lighting, obstacles in the walking path, slippery outdoor conditions

Medical Risk Factors

Age, anxiety and agitation, arrhythmias, dehydration, depression, female gender, impaired transfer and mobility, malnutrition, orthostatic hypotension, poor vison and use of bifocals, previous fall, reduced mental acuity and diminished cognitive skills, urgent urinary incontinence, Vitamin D insufficiency (serum 25-OH-D < 30ng/ml (75nmol/l)), medications causing over-sedation (narcotic analgesics, anticonvulsants, psychotropics), diabetes

Neurological and Musculoskeletal Risk Factors

Kyphosis, poor balance, reduced proprioception, weak muscles

Other Risk Factors; Fear of falling

The presence of any of these risk factors should trigger consideration of further evaluation and treatment to reduce the risk of falls and fall-related injuries.

Table from the Endotext chapter entitled “Osteoporosis: Clinical Evaluation” by E. Michael Lewiecki.

 

Medications for Osteoporosis in Men

 

The pharmacologic treatment of osteoporosis in men is by and large the same as treatment in women. Alendronate, risedronate, zoledronic acid, denosumab, teriparatide, and abaloparatide are all FDA approved for men with osteoporosis. Most men have been treated with bisphosphonates, similar to women. Alendronate was the first modern bisphosphonate approved by regulatory agencies in the mid-1990’s; it was shown to change surrogates of fracture (BMD and bone turnover markers) in men similarly to women (60). Although fracture risk reduction was not the primary outcome of the study, there were fewer morphometric vertebral fractures in the men randomized to alendronate compared those on placebo.  Similarly, risedronate and zoledronic acid have been shown to increase bone density in men to a similar degree as in women (61, 62). A criticism by some is that current surrogates for fracture may not be adequate, and that raising BMD or suppressing bone turnover markers in men is not enough evidence to conclude that fracture risk will be lowered by bisphosphonates. In a two-year study (63) with morphometric vertebral fractures as the primary outcome, Boonen et al demonstrated that zoledronic acid not only increased BMD in men, compared to placebo infusions, but it also led to fewer vertebral fractures. Specifically, at 2 years there was a 67% relative risk reduction and 3.3% absolute risk reduction in morphometric vertebral fractures. Thus, the clinician can be confident that if the patient is compliant and adherent to bisphosphonate treatment, fracture risk should be decreased.

 

All of the cited studies in men used a male normative database for calculation of the T-score. Men were eligible for the studies if they had osteoporosis by this criterion or had osteopenia (usually a T-score of -2) plus history of a low trauma facture. In women treated with bisphosphonates, vertebral fractures are decreased by about half and hip fractures by a third.  In the zoledronic acid registration trial in women (64), at 2 years the relative risk reduction of morphometric vertebral fractures was 71% and the absolute risk reduction was 5%. Compared to the study in men, at baseline the women were older, were more likely to have had a previous fracture, and had lower BMD.  It is impossible to compare results between the two gender-specific studies in any meaningful way, other than to conclude that zoledronic acid works similarly in men and women. 

 

The usual starting treatment for osteoporosis is oral alendronate 70 mg by mouth once weekly.  As in women, oral alendronate (or most preparations of risedronate) has to be taken on an empty stomach with just a glass of water, and the patient is instructed to take nothing else by mouth for at least 30 minutes. In general, this is not a problem, but for men also taking levothyroxine and/or proton pump inhibitors, timing may be difficult. In patients on levothyroxine, one strategy is to have them take the levothyroxine in the middle of the night, when older men are likely to need to urinate. This does not work for bisphosphonates because lying down after taking the bisphosphonate may lead to esophageal irritation. For the man with gastro-esophageal reflux disease (GERD), avoidance of oral bisphosphonates is indicated if the GERD is not under good control. For such men and for those unable or unwilling to adhere to the correct oral regimen, intravenous zoledronic acid, 5 mg given over 15 minutes or more, is a reasonable choice. The FDA-approved interval for zoledronic acid is one year. In our experience (65, 66), increasing the interval to 1.5 years or so allows all bisphosphonate patients to have a 5-year initial treatment period. Long-term management of osteoporosis in men is discussed below.  An alternative oral treatment is risedronate given as a monthly 150 mg tablet.  For some men, particularly those with a high pill burden, this may be an attractive regimen.

 

As an alternative to bisphosphonates, another anti-resorptive or anti-bone turnover medication is denosumab, an antibody against RANK Ligand. Among the earliest uses of this medication was a study of a high-risk group, men on ADT for prostate cancer. In this important study, Smith and colleagues (67) randomized men receiving GnRH analogs to profoundly suppress testosterone secretion to denosumab or placebo. There were fewer morphometric vertebral fractures in the men who were given denosumab as a subcutaneous injection every 6 months compared to men receiving placebo injections. After a study (68) showing that denosumab altered surrogates for fracture in men similarly to the effect in women, the drug was approved for osteoporosis in men, regardless of etiology. Interestingly, denosumab increases forearm bone density, something not found with bisphosphonate treatment (60). In long term studies of bisphosphonates in women (69, 70) BMD rises and then plateaus after a few years of treatment.  In contrast, studies in women have shown continued increases in BMD for at least 10 years with continued denosumab treatment (71). The consequences of this plus the impact of withdrawal of osteoporosis treatment will be discussed below.

 

As men age, there is thinning of trabeculae, whereas in women there is loss of trabecular number and the spacing between trabeculae increases (72). Thus, while the changes in vertebral fracture risk appear very similar in men and women, the impact on fracture could be different. DXA does not capture all of the changes with time. More recent studies (73) with high resolution peripheral quantitative computed tomography (HR-pQCT) also show sex-specific changes, but the studies are small. To my knowledge, there are no bone biopsy or HR-pQCT studies that demonstrate sex-dependent differences in response to therapy.

 

In women anabolic agents increase trabecular thickness and connectivity and increase cortical bone thickness. Of late, increased use of such agents as the initial treatment has been advocated for those patients at highest risk for fracture based on recent studies in women (74) demonstrating benefits to starting with anabolic treatment. In the United States, only teriparatide and abaloparatide are FDA approved for osteoporosis in men. There is another anabolic agent, romosozumab, that is approved for women, but there is no reason to believe that it would not work in men. There is one published report of improved fracture surrogates in men given romosozumab (75). Abaloparatide works similarly in men and women (76, 77). The use of anabolic agents, regardless of the patient’s sex, is limited by inconvenience of treatment (both teriparatide and abaloparatide are administered as a daily subcutaneous injection) and cost.  While romosozumab is given as a monthly injection in a clinician’s office, its cost in the United States is similar to that of abaloparatide, which is somewhat cheaper than teriparatide. In Japanese women, a higher dose of teriparatide given weekly or semi-weekly has been found to be effective (78), but there have been no studies of similar preparations in Europe or the United States. Based on studies in women, anabolic agents should be considered, including off-label use, for men at the highest risk of fracture. In a 3-year study (79) of men and women with glucocorticoid-induced osteoporosis, teriparatide was shown to result in fewer spine fractures than alendronate. More recently, a study in women (80) showed that anabolic treatment led to fewer fractures than anti-resorptive treatment with risedronate. Until there are better surrogates for fracture, there will never be a study comparing fracture risk in men treated with anabolic agents compared to anti-resorptives. The data from studies in women are convincing, and there is no physiological reason to question whether men would respond differently. A recent systematic review and meta-analysis of randomized controlled trials (81) led to the conclusion that osteoporosis drugs work the same in men and women.

 

In summary, initial treatment of osteoporosis in men should be comprehensive, with attention to diet, exercise, vitamin D, fall risk reduction, and home safety. After vitamin D is satisfactory, and possibly after dental work is completed, most men can be treated with bisphosphonates, usually oral alendronate. For those who cannot take an oral preparation, intravenous zoledronic acid is the drug of choice. For men at the highest risk for fracture, based on fracture history, DXA, risk factors, and risk calculators, a 1 to 2-year course of anabolic treatment should be considered, although teriparatide and abaloparatide can be prescribed for more than 2 years, if needed. For very high-risk patients, the anabolic therapy can be followed by 2 years of denosumab treatment, followed by consolidation with a bisphosphonate. For those men with CKD 4, denosumab is a good choice but must be continued indefinitely. Denosumab is also appealing for men on ADT who receive long-acting GnRH analogs every 6 months because they can receive denosumab at the same visit. However, there are rapid bone loss and potential vertebral fractures in women who have recently withdrawn from denosumab (82, 83). In one observational study in men (84) zoledronic acid prevented the loss of bone after men had discontinuation of denosumab.

 

Long-Term Management of Osteoporosis in Men

 

There are no long-term studies of osteoporosis treatment in men. Hence, all suggestions for management must be made from the few studies in women. The FLEX trial (69) showed that 10 years of alendronate in women led to fewer clinical vertebral fractures than 5 years of alendronate followed by 5 years of placebo tablets. The HORIZON extension trial (70) showed that a plan of 6 annual infusions of zoledronic acid was associated with fewer morphometric vertebral fractures than 3 annual infusions of zoledronic acid followed by 3 placebo infusions.  Based on these studies plus some other information, a task force of the American Society for Bone and Mineral Research recommended an approach to long-term osteoporosis management (85). While the approach was aimed mostly at postmenopausal women, the task force recommended that it be applied to men as well. In this approach, the initial treatment period is 5 years for oral bisphosphonates and 3 years for zoledronic acid. At the end of the initial treatment period, the patient is re-assessed by history, physical examination, and repeat BMD. Those patients remaining at elevated fracture risk should continue treatment and be re-assessed again in 2 to 3 years. Those patients whose fracture risk has been demonstrably decreased by treatment can interrupt therapy and be re-assessed at 2 to 3 years. Beyond 10 years of treatment there are no studies, and so clinical judgement will be necessary to manage such patients. I have proposed, based on studies in women (86, 87) and men (65), that the interval between zoledronic acid infusions can be lengthened such that each patient would receive 3 infusions of zoledronic acid over 5 years. This creates a 5-year initial treatment plan for the majority of people with osteoporosis: all but those at highest risk for fracture. For the latter group, initial therapy should be anabolic for the first 1 to 2 years, and then the patient would be placed on anti-resorptive agents. While this approach to long-term osteoporosis management makes sense, it will likely never be supported by large randomized trials.

 

A summary of a practical approach to the evaluation of osteoporosis in men is shown in Table 5.

 

Table 5.  Approach to Osteoporosis Treatment in Men

For All Men: Conservative Treatment

Fall risk reduction/home safety

Adequate calcium, vitamin D, dietary protein

Weight bearing exercise/balance training

Smoking cessation/minimization of alcohol intake

Treat Secondary Osteoporosis with Specific Therapy

Men with Borderline Fracture Risk

Conservative treatment

Repeat DXA in 2 to 3 years

Use FRAX to demonstrate low risk

Osteoporosis by DXA, Osteopenia + Fracture, High Risk by FRAX

Oral alendronate or risedronate or intravenous zoledronic acid

Clinical reassessment every year

Repeat DXA at 2 to 3 years

Change Rx if response inadequate

Repeat DXA at 5 years to consider drug holiday versus continued Rx

Very High Risk by DXA, FRAX, Clinical Findings

Anabolic Rx for 1 to 2 years

Then denosumab for 2 years

Then 1 year of alendronate or 1 infusion of zoledronic acid

DXA at 2 to 3 and 5 years

Drug holiday versus continued treatment based on fracture risk after treatment

.

CONCLUSIONS

 

Despite the overall paucity of evidence underpinning osteoporosis evaluation and treatment in men, it is important to identify men at risk for fracture, evaluate them efficiently, and treat them.  As more men live long enough to fracture, the burden of male osteoporosis will increase. In addition, because men with hip fracture are more likely to die after fracture (88), compared to women of the same age, improving diagnosis and treatment is likely to save lives, decrease suffering, and lead to lower costs.     

 

REFERENCES

 

  1. Cauley JA, Cawthon PM, Peters KE, Cummings SR, Bauer DC, Taylor BC, Shikany JM, Hoffman AR, Lane NE, Kado DM. Stefanick ML, Orwoll ES. Osteoporotic Fractures in Men (MrOS) Study Group. Risk factors for hip fracture in older men: The Osteoporotic Fractures in Men Study (MrOS). J Bone Miner Res 2016;31:1810-1819.
  2. NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA 2001;285:785-795.
  3. Kanis JA. Diagnosis of osteoporosis and assessment of fracture risk. Lancet 2002;359:1929-1936.
  4. Schousboe JT, Tanner SB, Leslie WD. Definition of osteoporosis by bone density criteria in men: effect of using female instead of male young reference data depends on skeletal site and densitometer manufacturer. J Clin Densitom 2014;17:310-316.
  5. Pasco JA, Lane SE, Brennan SL, Tinney EN, Bucki-Smith G, Dobbins AG, Nicholson GC, Kotowicz MA. Fracture risk among older men: osteopenia and osteoporosis defined by cut-points derived from female versus male reference data. Osteoporos Int 2014;25:857-862.
  6. Trajanoska K, Schoufour JD, de Jonge EA, Kieboom BCT, Mulder M, Stricker BH, Voortman T, Uitterlinden AG, Oei EH, Ikram MA, Zillikens MC, Rivadeneira F, Oei L. Fracture incidence and secular trends between 1989 and 2013 in a population based cohort: the Rotterdam Study. Bone 2018;114:116-124.
  7. Kanis JA, Harvey NC, Johansson H, Oden A, Leslie WD, McCloskey EV. FRAX Update. J Clin Densitom 2017;20:360-367.
  8. Sandhu SK, Nguyen ND, Center JR, Pocock NA, Eisman JA, Nguyen TV. Prognosis of fracture: evaluation of predictive accuracy of the FRAX algorithm and Garvan nomogram. Osteoporos Int 2010;21:863-871.
  9. Ahmed LA, Nguyen ND, Bjornerem A, Joakimsen RM, Jorgensen L, Stormer J, Bliuc D, Center JR, Eisman JA, Nguyen TV, Emaus N. External validation of the Garvan nomogram for predicting absolute fracture risk: the Tromso study. PLoS One 2014;9:e107695.
  10. Looker AC, Sarafrazi Isfahani N, Fan B, Shepherd JA. FRAX-based estimates of 10-year probability of hip and major osteoporotic fracture among adults aged 40 and over: United States, 2013 and 2014. Natl Health Stat Report. 2017;103:1-16.
  11. Toteston AN, Melton LJ 3rd, Dawson-Hughes B, Baim S, Favus MJ, Khosla S, Lindsay RL. Cost-effective osteoporosis treatment thresholds: the United States perspective Osteopors Int 2008;19:437-447.
  12. Wright NC, Saag KG, Dawson-Hughes B, Khosla S, Siris ES. The impact of the new National Bone Health Alliance (NBHA) diagnostic criteria on the prevalence of osteoporosis in the USA. Osteoporos Int 2017;31:1753-1759.
  13. McCloskey EV, Johansson H, Oden A, Vasireddy S, Kayan K, Pande K, JalavaT, Kanis JA. Ten-year fracture probability identifies women who will benefit from clodronate therapy-additional results from a double-blind, placebo controlled randomized study. Ostoeporos Int 2009;20:811-817.
  14. Ensrud KE, Taylor BC, Peters KW, Gourlay ML, Donaldson MG, Leslie WD, Blackwell TL, Fink HA, Orwoll ES, Schousboe J. Implications of expanding indications for drug treatment to prevent fracture in older men in the United States: cross sectional and longitudinal analysis of a prospective cohort study. BMJ 2014;349:g1420.
  15. Schini M, Johansson H, Harvey NC, Lorentzon M, Kanis JA, McCloskey EV. An overview of the use of the fracture risk assessment tool (FRAX) in osteoporosis. J Endocrinol Invest 2023 Oct 24. Doi: 10.1007/s40618-023-02219-9.
  16. Shevroja E, Reginster J-Y, Lamy L, Al-Daghri N, Chandran M, Demoux-Balada A-L, Kohlmeier L, Lecart M-P, Messina D, Camargos BM, Payer J, Tuzun S, Veronese N, Cooper C, McCloskey EV, Harvey NC. Update on the clinical use of the trabecular bone score (TBS) in the management of osteoporosis: results of an expert group meeting organized by the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO), and the International Osteoporosis Foundation (IOF) under the auspices of WHO Collaborating Center for Epidemiology of Musculoskeletal Health and Aging. Osteoporos Int 2023;34:1501-1529.
  17. Schousboe JT, Vo TN, Langsetmo L, Taylor BC, Kats AM, Schwartz AV, Bauer DC, Cauley JA, Ensrud KE, for the Osteoporotic Fractures in Men (MrOS) Study Research Group. Predictors of change in trabecular bone score (TBS) in older men: results from the Osteoporotic Fractures in Men (MrOS) Study. Osteoporos Int 2018;29:49-59.
  18. Schuitt SC, van der Klift M, Weel AE, de Laet CE, Burger H, Seeman E, Hofman A, Uitterlinden AG, ven Leeuwen JP, Pols HA. Fracture incidence and association with bone mineral density in elderly men and women: the Rotterdam Study. Bone 2004;34:195-202.
  19. Harman SM, Metter EJ, Tobin JD, Pearson J, Blackman MR. Longitudinal effects of aging on serum total and free testosterone levels in healthy men. J Clin Endocrinol Metab 2001;86:724-731.
  20. Khosla S, Melton LJ 3rd, Atkinson EJ, O’Fallon WM. Relationship of serum sex steroid levels to longitudinal changes in bone density in young versus elderly men. J Clin Endocrinol Metab 2001;86:3555-3561.
  21. Riggs BL, Melton LJ 3rd. Involutional osteoporosis. N Engl J Med 1986;314:1676-1686.
  22. Ryan LE, Ing SW. Hypercalciuria and bone health. Curr Osteoporos Rep 2012;10:286-295.
  23. Rosen CJ, Kurland ES, Vereault D, Adler RA, Rackoff PJ, Craig WY, Witte S, Rogers J, Bilezikian JP. An association between serum IGF-I and a simple sequence repeat in the IGF-I gene: implications for genetic studies of bone mineral density. J Clin Endocrinol Metab 1998;83:2286-2290.
  24. van Pottelbergh L, Goemaere S, Zmierczak H, Kaufman JM. Perturbed sex steroid status in men with idiopathic osteoporosis and their sons. J Clin Endocrinol Metab 2004;89:4949-4953.
  25. Yang S, Leslie WD, Yan L, Walld R, Roos KKm Morin SN, Majumdar SR, Lix LM. Objectively verified parental hip fracture is an independent risk factor for fracture: a linkage analysis of 478,792 parents and 261,705 offspring. J Bone Miner Res 2016;31:1753-1759.
  26. Cawthon PM, Shahnazari M, Orwoll ES, Lane NE. Osteoporosis in men: findings from the osteoporotic fractures in men study (MrOS). Ther Adv Musculoskel Dis 2016;8:15-27.
  27. van Staa TP, Leufkens HG, Abenhaim L, Zhang B, Cooper C. Oral corticosteroids and fracture risk: relationship to daily and cumulative doses. Rheumatology (Oxford) 2000;39:1383-1389.
  28. Waljee AK, Rogers MA, Lin P, Singal AG, Stein JD, Marks RM, Ayanian JZ, Nallamothu BK. Short term use of oral corticosteroids and related harms among adults in the United States: population based cohort study. BMJ 2017;357:j1415.
  29. Adler RA, Hochberg MC. Glucocorticoid-induced osteoporosis in men. J Endocrinol Investig 2011;34:481-484.
  30. Compston J. Type 2 diabetes mellitus and bone. J Intern Med 2018;283:140-153.
  31. Akhigbe T, Chin AS, Svircev JN, Hoenig H, Burns SP, Weaver FM, Bailey L, Carbone L. A retrospective review of lower extremity fracture care in patients with spinal cord injury. J Spinal Cord Med 2015;38:2-9.
  32. Starup-Linde J, Rosendahl SB, Storgaard M, Langdahl B. Management of osteoporosis in patients living with HIG – A systematic review and meta-analysis. J Acquir Immune Defic Syndr 2020;83:1-8.
  33. Goh SSL. Lai PSM, Tan ATB, Ponnampalavanar S. Reduced bone mineral density in human immunodeficiency virus-infected individuals: a meta-analysis of its prevalence and risk factors. Osteoporos Int 2018;29:595-613.
  34. Hoy JF, Grund B, Roedigner M, Schwartz AV, Shepherd J, Avihingsanon A, Badal-Faesen S, de Wit S, Jacoby S, La Rosa A, Pujari S, Schechter M, White D Engen NW, Ensrud K, Aagaard PD, Carr A, INSIGHT START Bone Mineral Density Substudy Group. Immediate initiation of antiretroviral therapy for HIV infection accelerates bone loss relative to deferring therapy: findings from the START Bone Mineral Density Substudy, a randomized trial. J Bone Miner Res 2017;32:1945-1955.
  35. Gupta SK, Post FA, Arrivas JR, Eron jr JJ, Wohl DA, Clarke AE, Sax PE, Stellbrink H-J, Esser S, Pozniak AL, Podzamczer D, Waters L. Orkin C, Rockstroh JK, Mudrikova T, Negredo E, Elion RA, Guo S, Zhong L, Carter C, Martin H, Brainard D, SenGupta D, Das M. Renal safety of tenofovir alafenamide vs tenofovir disoproxil fumarate: a pooled analysis of 26 clinical trials. AIDS 2019;33:1455-1465.
  36. de Rocha VM, Faria MBB, de Assis dos Reis Jr F, Lima COGX, Fiorelli RKA, Cassiano KM. Use of bisphosphonates, calcium and vitamin D for bone demineralization in patients with human immunodeficiency virus/acquired immune deficiency syndrome: a systematic review and meta-analysis of clinical trials. J Bone Metab 2020;27:175-186.
  37. Smith MR, Goode M, Zietman AL, McGovern FJ, Lee H, Finkelstein JS. Bicalutamide monotherapy versus leuprolide monotherapy for prostate cancer: effects on bone mineral density and body composition. J Clin Oncol 2004;22:2546-2553.
  38. Dalla Volta A, Formenti AM, Berruti A. Higher risk of fragility fractures in prostate cancer patients treated with combined Radium-223 and abiraterone: prednisone may be the culprit. Eur Urol 2019;75:894-896.
  39. Shahinian VB, Kuo YF, Freeman JL, Goodwin JS. Risk of fracture after androgen deprivation for prostate cancer. N Engl J Med 2005;352:154-164.
  40. Snyder PJ, Kopperdahl DL, Stephens-Shields AJ, Ellenberg SS, Cauley JA, Ensrud KE, Lewis CE, Barrett-Connor E, Schwartz AV, Lee DC, Bhasin S, Cunningham GR, Gill TM, Matsumoto AM, Swerdloff RS, Basaria S, Diem SJ, Wang C, Hou X, Cifelli D, Dougar D, Zeldow B, Bauer DC, Keaveny TM. Effect of testosterone treatment on volumetric bone density and strength in older men with low testosterone: a controlled clinical trial. JAMA Intern Med 2017;177:471-479.
  41. Watts NB, Adler RA, Bilezikian JP, Drake MT, Eastell R, Orwoll ES, Finkelstein JS. Osteoporosis in men: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2012;97:1802-1822.
  42. gov Identifier: NCT03518034.
  43. Lincoff AM, Bhasin S, Flevaris P, Mitchell LM, Basaria S. Boden WE, Cunningham GR, Granger CB, Khera M, Thompson IM, Wang Q, Wolski K, Davey D, Kalahasti V, Khan N, Miller MG, Snabes MC, Chan A, Dubenco E, Li X, Yi T, Huang KM, Travison TG, Nissen SE, for the TRAVERSE Study Investigators. Cardiovascular safety of testosterone-replacement therapy. N Engl J Med 2023;389:107-117.
  44. Bhasin S, Travison TG, Pencina KM, O’Leary M, Cunningham GR, Lincoff AM, Nissen SE, Lucia S, Preston MA, Khera M, Khan M, Snabes MC, Li X, Tangen CM, Buhr KA, Thompson Jr, IM. Prostate safety eventst during testosterone replacement therapy in men with hypogonadism; a randomized clinical trial. JAMA Network Open 2023;6:e2348692.
  45. Snyder PJ, Bauer DC, Ellenberg SS, Cauley JA, Buhr KA, Bhasin S, Miller MG, Khan NS, Ki X, Nissen SE. Testosterone treatment and fractures in men with hypogonadism. N Engl J Med 2024;390:203-211.
  46. Grossmann M, Anawalt BD. Breaking news- testosterone treatment and fractures in older men. N Engl J Med 2024;390:268-269.
  47. US Preventive Services Task Force. Screening for osteoporosis to prevent fractures. US Preventive Services Task Force Recommendation Statement. JAMA 2018;319:2521-2531.
  48. Shepstone L, Lenaghan E, Cooper C, Clarke S, Fong-Soe-Khloe R, Fordham R, Gittoes N, Harvey I, Harvey N, Heawood A, Holland R, Howe A, Kanis J, Marshall T, O’Neill T, Peters T, Redmond N, Torgerson D, Turner D, McCloskey E, SCOOP Study Team. Screening in the community to reduce fractures in older women (SCOOP): a randomized controlled trial. Lance 2018;391:741-747.
  49. Colon-Emeric CS, Pieper CF, Van Houtven CH, Grubber JM, Lyles KW, LaFleur J, Adler RA. Limited osteoporosis screening effectiveness due to low treatment rates in a national sample of older men. Mayo Clin Proc 2018;93:1749-1759.
  50. Gruenewald LD, Koch V, Yel I, Eichler K, Gruber-Rouh T, Alizadeh LS, Mahmoudi S, D’Angelo T, Wichmann JL, Wesarg S. Vogl TJ, Booz C. Association of phantomless dual-energy CT-based volumetric bone mineral density with prevalence of acute insufficiency fractures of the spine. Acad Radiol 2023;30:2110-2117.
  51. Teng PF, Chiang JM, Schafer AL, Sukerkar PA, Keaveny TM, Bikle D. Prevalence of osteoporosis in older male veterans receiving hip-containing computed tomography scans: opportunistic use of biomechanical computed tomography analysis (BCT). Osteoporos Int 2023;34:551-561.
  52. El-Hajj Fuleihan G, Bouillon R, Clarke B, Chakhtoura M, Cooper C, McClung M, Singh RJ. Serum 25-hydroxyvitamin D levels: variability, knowledge gaps, and the concept of a desirable range. J Bone Miner Res 2015;30:1119-1133.
  53. Shapiro JR, Lewiecki EM. Hypophosphatasia in adults: clinical assessment and treatment considerations. J Bone Miner Res 2017;32:1977-1980.
  54. Bhasin S, Brito JP, Cunningham GR, Hayes FJ, Hodis HN, Matsumoto AM, Snyder PJ, Swerdloff RS, Wu FC, Yialamas MA. Testosterone therapy in men with hypogonadism: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2018;103:1715-1744.
  55. Ryan CS, Petkov VI, Adler RA. Osteoporosis in men: the value of laboratory testing. Ostoporos Int 2011;22:1845-1853.
  56. Fink HA, Litwack-Harrison S, Tayulor BC, Bauer DC, Orwoll ES, Lee CG, Barrett-Connor E, Schousboe JT, Kado DM, Garimella PS, Ensrud KE, Osteoporosis Fractures in Men (MrOS) Study Group. Clinical utility of routine laboratory testing to identify possible secondary causes in older men with osteoporosis: the Osteoporotic Fractures in Men (MrOS) Study. Osteoporos Int 27:331-338, 2016.
  57. Weaver CM, Alexander DD, Boushy CJ, Dawson-Hughes B, Lappe JM, LeBoff MS, Liu S, Looker AC, Wallace TC, Wang DD. Calcium plus vitamin D supplementation and risk of fractures: an updated meta-analysis from the National Osteoporosis Foundation. Osteoporos Int 2016;27:367-376.
  58. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clifton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Mayne ST, Rosen CJ, Shapses SA. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab 2011;96:53-58.
  59. Tseng VL, Yu F, Lum F, Coleman AL. Risk of fractures following cataract surgery in Medicare beneficiaries. JAMA 2012;308:493-501.
  60. Orwoll E, Ettinger M, Weiss S, Miller P, Kendler D, Graham J, Adami S, Weber K, Lorenc R, Pietschmann P, Vandormael K, Lombardi A. Alendronate for the treatment of osteoporosis in men. N Engl J Med 2000;343:604-610.
  61. Ringe JD, Faber H, Farahmand P, Dorst A. Efficacy of risedronate in men with primary and secondary osteoporosis: results of a 1-year study. Rheumatol Int 2006;26:427-431.
  62. Orwoll ES, Miller PD, Adachi JD, Brown J, Adler RA, Kendler D, Bucci-Rechtweg C, Readie A, Mesenbrink P, Weinstein RS. Efficacy and safety of a once-yearly i.v. infusion of zoledronic acid 5 mg versus a once-weekly 70-mg oral alendronate in the treatment of male osteoporosis: a randomized, multicenter, double-blind, active-controlled trial. J Bone Miner Res 2010;25:2239-2250.
  63. Boonen S, Reginster JY, Kaufman JM, Lippuner K, Zanchetta J, Langdahl B, Rizzoli R, Lipschitz S, Dimai HP, Witvrouw R, Eriksen E, Brixen K, Russo L, Claessens F, Papanastasiou P, Antunez O, Su G, Bucci-Rechtweg C, Hruska J, Incera E, Vanderschueren D, Orwoll E. Fracture risk and zoledronic acid therapy in men with osteoporosis. N Engl J Med 2012;367:1714-1723.
  64. Black DM, Delmas PD, Eastell R, Reid IR, Boonen S, Cauley JA, Cosman F, Lakatos P, Leung PC, Man Z, Mautalen C, Messenbrink P, Hu H, Caminis J, Tong K, Rosario-Jansen T, Krasnow J, Hue TF, Sellmeyer D, Eriksen EF, Cummings SR, HORIZON Pivotal Fracture Trial. Once yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med 2007;356:1809-1822.
  65. Johnson DA, Williams MI, Petkov VI, Adler RA. Zoledronic acid treatment of osteoporosis: effects in men. Endocr Pract 2010;16:960-967.
  66. Adler RA. Duration of anti-resorptive therapy for osteoporosis. Endocrine 2016;51:222-224.
  67. Smith MR, Egerdie B, Hernandez Toriz N, Feldman R, Tammela TTL, Saad F, Heracek J, Szwedowski M, Ke C, Kupiec A, Leder BZ, Goessi C, Denosumab HALT Prostate Cancer Study Group. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med 2009;361:745-755.
  68. Langdahl B, Teglbjaerg CS, Ho PR, Chapurlat R, Czerwinski E, Kendler DL, Reginster JY, Kivitz A, Lewiecki EM, Miller PD, Bolognese MA, McClung MR, Bone HG, Ljinggren O, Abrahmsen B, Gruntmanis U, Yang YC, Wagman RB, Mirza F, Siddhanti S, Orwoll E. A 24-month study evaluating the efficacy and safety of denosumab for the treatment of men with low bone mineral density: results from the ADAMO trial. J Clin Endocrinol Metab 2015;100:1335-1342.
  69. Black DM, Schwartz AW, Ensrud KE, Cauley JA, Levis S, Quandi SA, Satterfield S, Wallace RB, Bauer DC, Palermo L, Wehren LE, Lombardi A, Santora A, Cummings SR, FLEX Research Group. Effects of continuing or stopping alendronate after 5 years of treatment: the Fracture Intervention Trial Long-term Extension (FLEX): a randomized trails. JAMA 2006;296:2927-2938.
  70. Black DM, Reid IR, Boonen S, Bucci-Rechtweg C, Cauley JA, Cosman F, Cummings SR, Hue TF, Lippuner K, Lakatos P, Leung PC, Man Z, Martinez RI, Tan M, Ruzycky ME, Su G, Eastell R. The effect of 3 versus 6 years of zoledronic acid treatment of osteoporosis: a randomized extension to the HORIZON-Pivotal Fracture Trial (PFT). J Bone Miner Res 2012;27:243-254.
  71. Bone HG, Wagman RB, Brandi ML, Brown JP, Chapurlat R, Cummings SR, Czerwinski E, Fahrleitner-Pammer A, Kendler DL, Lippuner K, Reginster JY, Roux C, Malouf J, Bradley MN, Daizadeh NS, Wang A, Dakin P, Pannacciulli N, Dempster DW, Papapoulos S. 10 years of denosumab treatment in postmenopausal women with osteoporosis: results from the phase 3 randomized FREEDOM trial and open-label extension. Lancet Diabetes Endocrinol 2017;5:513-523.
  72. Khosla S, Riggs BL, Atkinson EJ, Oberg AL, McDaniel LJ, Holets M, Peterson JM , Melton LJ 3rd. Effects of sex and age on bone microstructure at the ultradistal radius: a population-based noninvasive in vivo assessment. J Bone Miner Res 2006;21:124-131.
  73. Cheung AM, Adachi JD, Hanley DA, Kendler DL, Davison KS, Josse R, Brown JP, Ste-Marie L-G, Kremer R, Erlandson MC, Dian L, Burghardt AJ, Boyd SK. High-resolution peripheral quantitative computed tomography for the assessment of bone strength and structure: a review by the Canadian Bone Strength Working Group. Curr Osteoporos Rep 2013;11:136-146.
  74. Leder BZ, Tsai JN, Uihlein AV, Wallace PM, Lee II, Neer RM, Burnett-Bowie SA. Denosumab and teriparatide transitions in postmenopausal osteoporosis (the DATA-Switch study): extension of a randomized, controlled trial. Lancet 2015;386:1147-1155.
  75. Lewiecki EM, Blicharski T, Goemaere S, Lippuner K, Meisner PD, Miller PD, Miyauchi A, Maddox J, Chen L, Horlait S. A phase III randomized placebo-controlled trial to evaluate efficacy and safety of romosozumab in men with osteoporosis. J Clin Endocrinol Metab 2018;103:3183-3193.
  76. Czerwinski E, Cardona J, Piebanski R, Recknor C, Vokes T, Saag KG, Binkley N, Lewiecki EM, Adachi J, Knychas D, Kendler D, Orwoll E, Chen Y, Pearman L, Li YH, Mitlak B. The efficacy and safety of abaloparatide-sc in men with osteoporosis: a randomized clinical trial. J Bone Miner Res 2022;37:2435-2442.
  77. Dhaliwal R, Kendler D, Saag K, Ing SW, Singer A, Adler RA, Pearman L, Wang Y, Mitlak B. Response rates for lumbar spine, total hip, and femoral neck bone mineral density in men treated with abaloparatide: results from the ATOM Study. JBMRPlus 2024; doi/10.1093/jbmrpl/ziae009.
  78. Sugimoto T, Shiraki M, Fukunaga M, Kishimoto H, Hagino H, Sone T, Nakano T, Ito M, Yoshikawa H, Minamida T, Tsuruya Y, Nakamura T. Study of twice-weekly injections of teriparatide by comparing efficacy with once-weekly injections in osteoporosis patients: the TWICE Study. Osteoporos Int 2019;30:2321-2331.
  79. Saag KG, Zanchetta JR, Devogelaer JP, Adler RA, Eastell R, See K, Kresge JH, Krohn K, Warner MR. Effects of teriparatide versus alendronate for treating glucocorticoid-induced osteoporosis: thirty-six-month results. Arthritis Rheum 2009:60:3346-3355.
  80. Kendler DL, Marin F, Zerbini CAF, Russo LA, Greenspan SL, Zikan V, Bagur A, Malouf-Sierra J, Lakatos P, Fahrleitner-Panner A, Lespessailles E, Minisola S, Body JJ, Geusens P, Moricke R, Lopez-Romero P. Effects of teriparatide and risedronate on new fractures in post-menopausal women with severe osteoporosis (VERO): a multicenter, double-blind, double-dummy, randomized controlled trial. Lancet 2018;391:230-240.
  81. Beaudart C, Demonceau C, Sabico S, Veronese N, Cooper C, Harvey N, Fuggle N, Bruyere O, Rizzoli R, Reginster J-Y. Efficacy of osteoporosis pharmacologic treatments in men: a systematic review and meta-analysis. Aging Clin Exper Res 2023;35:1789-1806.
  82. Anastasilakis AD, Polyzos SA, Makras P, Aubry-Rozier B, Kaouri S, Lamy O. Clinical features of 24 patients with rebound-associated vertebral fractures after denosumab discontinuation: systematic review and additional cases. J Bone Miner Res 2017;32:1291-1296.
  83. Kendler D, Chines A, Clark P, Ebeling PR, McClung M, Rhee Y, Huang S, Stad RK. Bone mineral density after transitioning from denosumab to alendronate. J Clin Endocrinol Metab 2020;105:e355-264.
  84. Solling AS, Harslof T, Brockstedt HK, Langdahl B. Discontinuation of denosumab in men with prostate cancer. Osteoporos Int 2023;34:291-297.
  85. Adler RA, El-Hajj Fuleihan G, Bauer DC, Camacho PM, Clarke BL, Clines GA, Compston JE, Drake MT, Edwards BJ, Favus MJ, Greenspan SL, McKinney R Jr., Pignolo RJ, Sellmeyer DE. Managing osteoporosis in patients on long-term bisphosphonate treatment: report of a task force of the American Society for Bone and Mineral Research. J Bone Miner Res 2016;31:16-35.
  86. Grey A, Bolland M, Wattie D, Horne A, Gamble G, Reid IR. Prolonged antiresorptive activity of zoledronate: a randomized, controlled trial. J Bone Miner Res 2010;25:2251-2255.
  87. Reid IR, Horne AM, Mihov B, Stewart A, Garratt E, Wong S, Wiessing KR, Bolland MJ, Bastin S, Gamble GD. Fracture prevention with zoledronate in older women with osteopenia. N Engl J Med 2018;379:2407-2416.
  88. Bass E, French DD, Bradham DD, Rubenstein LZ. Risk-adjusted mortality rates of elderly veterans with hip fractures. Ann Epidemiol 2007;17:514-519.

Cushing Syndrome/Disease in Children and Adolescents

ABSTRACT

 

Endogenous Cushing syndrome (CS) is a rare pediatric endocrine condition commonly caused by pituitary corticotroph tumors or less often by adrenal or ectopic sources. The typical presentation of the child with CS includes weight gain with height deceleration, characteristic skin findings, and hormonal and biochemical findings indicative of excessive glucocorticoid production. The diagnostic evaluation of the patient with suspected hypercortisolemia initially involves the confirmation of cortisol excess in blood and/or urine, and then the identification of source. The first line of management usually requires surgical treatment of a pituitary or adrenal lesion. In persistent or recurrent disease, re-operation, medical treatment, or radiation should be considered.

 

INTRODUCTION

 

Cushing syndrome (CS) describes the exposure of the body to supraphysiologic levels of glucocorticoids. Although exogenous (iatrogenic) CS is common, endogenous pediatric CS, is a rare pediatric endocrine condition. Population studies of the incidence of the disease have shown that endogenous CS occurs in about 3-50 cases per million people per year, depending on the population studied; pediatric patients in these studies represent 6-7% of all cases (1-3).  

 

ETIOLOGY

 

Endogenous CS can be classified as ACTH-dependent (pituitary or ectopic) or ACTH-independent CS (adrenal-related, Table 1) (4). The etiology of pediatric CS differs based on the age group of the patient (5). In patients younger than 5 years of age, ACTH-independent CS is more common compared to older children and adolescents who usually present with ACTH-dependent CS. Ectopic CS (ECS) is rare at any age group (5).

 

Table 1. Causes of Cushing Syndrome

Type

Source

 Mechanism

Exogenous

 

Iatrogenic

 Exogenous administration of supraphysiologic doses of glucocorticoids or ACTH

Endogenous

 

ACTH-dependent

Pituitary

Corticotroph pituitary neuroendocrine tumor

Pituitary blastoma

Ectopic

 Neuroendocrine tumors secreting ACTH and/or CRH

 

 

 ACTH-independent

Unilateral adrenal (except in metastatic disease)

 Cortisol-secreting adrenocortical adenomas and carcinomas

Bilateral adrenals

Bilateral micronodular adrenocortical disease

-       Primary pigmented nodular adrenocortical disease (PPNAD), isolated or in the context of Carney complex

-       Isolated micronodular adrenocortical disease (iMAD)

Bilateral macronodular adrenocortical disease

 

ACTH-Dependent Cushing Syndrome

 

ACTH-dependent CS is most commonly due to a corticotroph pituitary neuroendocrine tumor (PitNET, also called pituitary adenoma or Cushing disease, (CD). These are monoclonal lesions that continue to express some of the characteristics of the normal corticotroph cell which can be useful in the diagnostic evaluation of patients (6, 7). Corticotroph-secreting PitNETs are usually microadenomas with median diameter of 5mm and do not often show signs of invasion to the cavernous sinus or other parasellar structures (8). Rare cases of aggressive PitNETs have been reported in the pediatric population with either resistance to treatment or distant metastasis (metastatic PitNETs) (9). These are associated with specific histologic subtypes, such as Crooke cell adenomas (9).

 

Infantile onset of ACTH-dependent CS with a pituitary lesion is often due to a pituitary blastoma. In 2014 de Kock et al, collected tissues from several infants who had been diagnosed with very young onset CD and reported that the tumors were consistent with pituitary blastomas as they had histologic findings of undifferentiated epithelium Rathke-like cells, mixed with hormone producing cells (10). They were able to identify germline and/or somatic DICER1 gene defects in these patients, suggesting that pituitary blastoma is a rare but almost pathognomonic presentation of DICER1 syndrome (10).

 

ECS is due to neuroendocrine tumors secreting ACTH and/or CRH outside the hypothalamic-pituitary axis. In older children and adolescents, the most common source of ECS are bronchial carcinoids, thymic carcinoids, and gastro-entero-pancreatic NETs (11-13). By contrast, in children younger than 5-10 years of age, ECS often presents in the context of pediatric specific tumors such as Wilm’s tumors, neuroblastomas, and others (13, 14).

 

ACTH-Independent Cushing Syndrome

 

ACTH-independent CS is commonly caused by unilateral adrenocortical tumors, cortisol-producing adenomas or carcinomas (5). Cortisol-producing adenomas are benign lesions with isolated cortisol secretion, while adrenocortical carcinomas are aggressive tumors and may commonly co-secrete cortisol and androgens in up to 80% of all cases (15, 16).

 

Bilateral adrenocortical disorders account for <2% of all cases of CS but some subtypes may be more prevalent in children compared to adults given their association with germline genetic defects (17). Micronodular adrenocortical disease is the most common type of bilateral adrenocortical disorder in pediatric patients. This category may be further divided in primary pigmented micronodular adrenocortical disease (PPNAD) where the adrenals present with multiple dark brown pigmented micronodules (due to lipofuscin deposition with most with diameter of <1cm) with internodular cortical atrophy, or the absence of these findings referred to as isolated micronodular adrenocortical disease (i-MAD) (18). PPNAD may be identified in the context of Carney complex (CNC) and less often as isolated PPNAD (19). Bilateral macronodular adrenocortical disease presenting with bilateral macronodules (most with diameter of ≥1cm) is rare in the pediatric population.

 

GENETICS

 

Genetic causes are found in less than half of the patients with pediatric CD and more commonly in adrenal-related CS. For patients presenting with pediatric onset CS, it is recommended to obtain genetic testing directed to the source of hypercortisolemia, i.e. adrenal vs. pituitary causes.  Although the yield in CD may be low, in cases of pituitary blastomas or bilateral micronodular disease genetic testing has higher chance of identifying the genetic cause and lead to screening for other related manifestations that may be important, such as cardiac myxomas in patients with CNC.

 

ACTH-Dependent Cushing Syndrome

 

Germline mutations are identified in less than 10% of patients with pediatric CD (8). Of the most common causes are MEN1 (causing multiple endocrine neoplasia type 1 syndrome, MEN1), CDKN1B (causing MEN4), and CABLES1 gene defects (20). Genes associated with familiar isolated pituitary adenoma (FIPA) syndrome, such as AIP, SDHx, and MAX, or syndromes associated with pituitary tumors amongst other manifestations, such as CNC due to PRKAR1A gene defects, do not commonly cause corticotropinomas and have only been reported in few case reports (21).

 

As mentioned above, young children (<2 years old) presenting with pituitary blastomas should be screened for DICER1 gene defects (10). DICER1 codes for an endoribonuclease that processes miRNAs (22). Patients with DICER1 or pleuropulmonary syndrome present with multiple tumors in lungs, kidneys, multinodular goiter, and other manifestations. Pituitary blastomas are present in less than 10% of all patients and always within the first years of life (23).

 

Somatic mutations are more likely to be identified in corticotropinomas. USP8 mutations in the 14-3-3 binding motif hotspot region of the gene have been reported as the cause of 40-60% of adults with CD (24, 25). Pediatric data suggest that USP8 mutations are less common and identified in up to 30% of cases (26). USP8 is a deubiquitinase involved in recycling of the epidermal growth factor receptor (EGFR) and mutations in the hotspot region led to increased catalytic activity, activation of the EGF pathway, and increased POMC expression. In children, USP8 mutant tumors presented with larger size and higher risk for persistent disease after surgery or recurrence after initial remission (26). Data in adult patients did not confirm this finding, and the prognostic value of identifying a USP8 mutation is still unclear (27). Other somatic mutations identified in corticotropinomas include USP48, TP53, and BRAF, but the incidence in pediatric patients is unknown (28). Finally, in a subset of patients with pediatric corticotropinomas large genomic chromosomal deletions/gains are identified and are associated with larger tumor and higher risk of invasion of the cavernous sinus (29).

 

ECS may present in various neuroendocrine tumors and the genetic background is associated with the primary tumor. MEN1, MEN2 (RET gene mutations), and some gene fusions have been described according to the tissue involved in ectopic ACTH secretion (30, 31).

 

ACTH-Independent Cushing Syndrome

 

Pediatric cortisol producing adrenocortical carcinomas may present in the context of TP53 mutations (32). In the Brazilian South and Southeast population, high prevalence of a germline founder TP53 mutation (p.R337H) is associated with high incidence of pediatric adrenocortical carcinomas (33, 34). Germline TP53 mutations may also present as Li-Fraumeni syndrome where patients have high risk for breast, central nervous system, bone, and other tumors (35). Cortisol-producing adrenocortical adenomas may be associated with gene defects leading to activation of the cyclic AMP (cAMP) protein kinase A (PKA) pathway, such as somatic mutations in PRKACA, PRKAR1A, and PRKACB genes (36, 37). Finally, somatic gene defects in the Wnt signaling pathway have also been identified in adrenocortical tumors (38).

 

ACTH-independent CS due to PPNAD presents commonly in the context of CNC (39). CNC is an autosomal dominant multiple neoplasia syndrome caused by inactivating mutations of the gene PRKAR1A, coding for the regulatory subunit 1 alpha of PKA, or less often linked to a second locus at chromosome 2p16 (40-42). Inactivating mutations in PRKAR1Alead to constitutive activation of PKA and downstream pathways (18). Patients with CNC present with several manifestations including PPNAD, pituitary abnormalities most often presenting as growth hormone dysregulation or acromegaly, thyroid nodules or carcinomas, testicular tumors, cardiac and skin myxomas, characteristic skin lesions, breast myxomatosis or adenomas, osteo-chondro-myxomas and psammomatous melanotic schwannomas (40). PPNAD in CNC is often diagnosed in the third decade of life but patients as young as in the first decade of life have been reported (43). Additional information about CNC can be found in the chapter entitled “Carney Complex” of Endotext (40).

 

Additional genetic defects associated with bilateral adrenocortical disease include PRKACA genomic gains, PDE11A, and PDE8A gene defects identified in patients with macronodular adrenocortical disease or isolated micronodular disease (44-46). PRKACA codes for the catalytic subunit of PKA, and chromosomal gains lead to increased PKA signaling (47). PDE11A and PDE8A codes for phosphor-diesterases that catalyze and decrease cAMP levels. Inactivating mutations in these genes lead to increased circulation of cAMP and increased PKA activity (44, 48). Macronodular adrenocortical disease due to ARMC5 gene defects often seen in adults is rare in the pediatric population (49).

 

Neonatal ACTH-independent CS may be seen in the context of McCune-Albright syndrome (MAS) (50). In these cases, CS presents within the first year of life and may have detrimental and rapidly developing symptoms which may even lead to death. However, if managed medically, neonatal CS in MAS may resolve on its own (51). Rare cases of neonatal onset adrenocortical disease have also been reported in the context of Beckwith-Wiedemann syndrome (52, 53).

 

PRESENTATION

 

The presentation of pediatric CS has similarities and differences from that in adults (Table 2).

 

Table 2. Presenting Findings in Pediatric Patients with Cushing Syndrome

Clinical findings

Anthropometric

Height deceleration

Weight gain

Cardiovascular

Hypertension

Musculoskeletal

Fractures

Proximal muscle weakness

Skin

Striae

Facial Plethora

Easy bruising

Acne

Hirsutism

Acanthosis nigricans

Abnormal fat deposition

Neuropsychiatric

Behavioral changes (compulsive behavior, overachievement tendency, irritability)

Psychiatric disorders (depression, anxiety)

Changes in cognitive function

Sleep disturbance (difficulty falling asleep)

Memory problems

Reproductive system

Delayed puberty

Irregular menses

Immunologic

Increased risk for infections

Laboratory and imaging findings

Complete blood count

Elevated total white blood cell, neutrophil and monocyte counts

Decreased lymphocyte and eosinophil count

Elevated neutrophil-to-lymphocyte ratio

Biochemistry

Hypokalemia

Hypercalciuria

Elevated ALT

Hyperlipidemia

Hyperglycemia with elevated insulin levels

Coagulation factors

Increased coagulation factors

Decreased aPTT

Echocardiogram

Cardiac hypertrophy

DXA

Decreased bone mineral density

 

The hallmark of pediatric CS is weight gain with concomitant height deceleration (Figure 1) (54). This finding can help discriminate patients with CS from with simple obesity who often have preserved height percentile (55). Fat deposition in pediatric patients may not be as prominently centripetal as noted in adults, and may present often as generalized obesity similar to other causes (56). Although height deceleration is seen in most cases of growing children, patients may not be short at presentation, may have completed growth by the time hypercortisolemia occurred, or may be exposed to episodic hypercortisolemia which may have more limited effect on their height (5, 8, 57). Bone age is often within the expected range for the chronologic age or advanced in pediatric patients with endogenous CS, and is correlated with the levels of adrenal androgens which are often increased in ACTH-dependent CS (58).

 

Figure 1. Typical growth chart of a pediatric patient with Cushing syndrome (A) compared to a child with obesity (B).

 

Dermatologic findings present in CS include striae (which are present in 60-80% of patients and may not have the characteristic appearance of deep purple color and thickness as in adults), facial plethora, acne (more common in ACTH-dependent CS possibly due to stimulation of adrenal zona reticularis by ACTH), hirsutism in women, hypertrichosis, acanthosis nigricans, and easy bruising (8, 56, 59).

 

Patients with CS often present with delayed puberty in males and females and/or irregular menses and secondary amenorrhea in females (8).

 

As in patients with iatrogenic CS, pediatric patients with endogenous CS present with decreased bone mineral density, with lower scores in the spinal measurements (60-62).  Proximal muscle weakness although reported is less frequent than adult patients (54).

 

Pediatric patients with endogenous CS, especially younger in age, often present with behavioral and neurocognitive changes. They may report behavioral changes including compulsive behaviors with overachievement goals, described as excellent students, along with increased anxiety and irritability (63). They may also report mood changes, depressed mood, sleep problems, and memory issues similar to adults. Headaches are common in pediatric patients and can be noted in up to 80% of them (8).

 

Hypercortisolemia and its related obesity lead to metabolic syndrome (64). Patients often present with insulin resistance and up to 30% of patients may have impaired glucose metabolism (56). Hyperlipidemia and elevated ALT as a surrogate marker of metabolic associated fatty liver disease (MAFLD) are also present in almost half of the patients (8, 56). Hypertension is present in almost 50% of patients with endogenous CS and cases of posterior reversible encephalopathy syndrome (PRES) due to hypertensive emergency have been reported as the initial manifestation of CS in pediatric patients (8, 65).

 

Similar to adults, pediatric patients with CS present with a hypercoagulable state associated with abnormal levels of procoagulants, antifibrinolytics, and anticoagulant factors, such as factor VIII, antithrombin III, protein C and S, and prolonged partial thromboplastin time (PTT) (66). Although in adult patients with CS the risk of venous thromboembolism is more studied, the exact incidence, risk, and thromboprophylaxis protocols in children are not as well delineated and depend on clinical judgement (67).

 

Additional findings in pediatric CS include characteristic abnormalities in CBC due to glucocorticoids effects including increased WBC count, neutrophil count, low normal lymphocyte count, and increased neutrophil-to-lymphocyte ratio (NLR) (68). Although immunosuppression may lead to severe infections in patients with significantly elevated cortisol levels, in most pediatric cases infections are limited to less clinically significant areas such as skin infections etc. (69). However, in very young patients, especially in neonatal CS, or patients with severe hypercortisolemia, such as in ECS, opportunistic infections may lead to significant morbidity and even death and prophylaxis should be initiated (14, 70).

 

Electrolyte abnormalities seen in endogenous CS include hypokalemia, uncommon overall but seen more frequently in ECS, and hypercalciuria which may lead to nephrolithiasis (8, 71).

 

Patients with hypercortisolemia also present with other hormonal defects including abnormal thyroid function test with a pattern of central hypothyroidism, abnormal GH secretion with IGF-1 levels usually preserved within the reference range, and suppressed gonadotropins (72-75). Tumor stalk compression effects may lead to hyperprolactinemia, although this is uncommon due to the small size of most corticotropinomas. Androgen levels are commonly elevated in ACTH-dependent CS due to adrenal zona reticularis stimulation from ACTH, or in adrenocortical carcinomas where co-secretion of cortisol and DHEAS may be seen.

 

DIAGNOSIS

 

The diagnostic evaluation of pediatric CS follows the guidelines of the endocrine and pituitary society adjusted for the pediatric population (7, 76, 77). Screening for hypercortisolemia is preferably done with at least two of the following tests: 24-hour urinary free cortisol (UFC, measured on 2-3 days), midnight (or late night) cortisol measured on 2-3 days, and suppression of cortisol to low dose dexamethasone (76). Specific details on these tests can be found in the chapter entitled “Endocrine Testing Protocols: Hypothalamic Pituitary Adrenal Axis” of Endotext (78).

 

Confirming the Diagnosis of Cushing Syndrome

 

The loss of the diurnal rhythm of ACTH/cortisol secretion is the first abnormality noted in patients with CS (79, 80). In clinical practice, salivary cortisol has been used to measure midnight or late night cortisol levels as it is convenient and can be collected at home (78, 81). If this is not available, then serum midnight cortisol is an alternative accurate screening test (77). A serum cortisol level of ≥4.4mcg/dL was able to distinguish almost all pediatric patients with CS with a sensitivity of 99% and a specificity of 100% (7). Serum cortisol needs to be measured from an indwelling catheter that has been placed at least 2 hours prior to sampling. We instruct patients to turn off all screens by 10pm and blood should be collected without awakening the patient (82).

 

The 24-hour urine collection should be performed on two or three days, to ensure optimal urine collection and account for the known day-to-day variability in urinary cortisol in patients with CS (83, 84). It is generally recommended to collect urine on days of routine physical activities and avoid days when increased stressful activities are expected, like competitive sports games etc. (85). Additionally, patients are advised to consume normal amount of fluids as excessive fluid intake and urine output may lead to false positive results (86). The urine samples should be measured for urine creatinine to ensure normal kidney function, but we do not routinely correct UFC levels for the urine creatinine level as this may lead to inaccurate results (87).

 

The low dose or 1mg overnight dexamethasone suppression test is performed similar to adults (78). Dose adjustment has been used in several studies, though no study has been done to specifically investigate the appropriate dose in children with CS. Various protocols recommend the use of 15mcg/kg, 25mcg/kg, or 0.3mg/m2 (max 1mg) once at 11pm for the overnight test or 1200mcg/kg/day (max 2mg/day) divided Q6 hours for two days (88, 89). Measurement of a serum dexamethasone level at the same time as cortisol is important to ensure the desired dexamethasone level has been reached.

 

If screening labs suggest cortisol excess, it is important to rule out physiologic/non-neoplastic hypercortisolism (previously known as pseudo-Cushing syndrome) (90). If suspicion is high, additional testing should be considered, including dexamethasone-CRH test (if available) or DDAVP stimulation test. If results remain inconsistent, close monitoring with repeat physical examination and labs within 3 months should be offered to monitor clinical and biochemical findings while at the same treating causes that may contribute to activation of the hypothalamic-pituitary-adrenal axis (90).

 

Identifying the Source of Hypercortisolemia

 

Once endogenous CS is confirmed, the next step is to identify the source of hypercortisolemia. ACTH levels are used to guide next steps. Elevated ACTH levels of >20-29pg/mL suggest ACTH-dependent CS while suppressed ACTH is consistent with ACTH-independent CS (7). Intermediate values may need further evaluation for both ACTH-dependent and ACTH-independent causes, but most often a non-suppressed ACTH level suggests ACTH-dependent CS, except in the case of mild subclinical hypercortisolemia or cyclical CS.

 

In cases of ACTH-dependent CS, additional biochemical and imaging studies include pituitary MRI (with and without contrast, pituitary protocol), CRH stimulation test (if available), DDAVP stimulation test and/or high dose dexamethasone suppression test. Corticotroph PitNETs are often shown as hypo-enhancing microadenomas in pituitary MRI (Figure 2), but a normal/negative MRI may be seen in up to 30% of patients (91). In cases of normal MRI or biochemical testing inconsistent with pituitary source, bilateral inferior petrosal sinus sampling (BIPSS) is the gold standard in diagnosing or ruling out CD. Non-invasive strategies are described if BIPSS is not feasible and/or not available (92). In our pediatric patients, all patients who showed suppression to high dose dexamethasone administration and stimulation to CRH/DDAVP consistent with a pituitary source, had CD irrespective of imaging findings (8).

 

For patients suspected to have ECS, further evaluation should include imaging of the neck, chest, abdomen, and pelvis with thin cuts as carcinoids can be small in diameter. Chest imaging is preferably done with CT due to higher accuracy in the lung parenchyma, but some centers use MRI for abdominal/pelvic imaging to reduce radiation. Nuclear imaging, preferably with Ga-68 DOTATATE or, if not available or negative, with 18F-FDG PET, may identify some of these ectopic sources (11, 13, 93). If a lesion found on imaging studies is suspicious but not convincing, one may attempt venous sampling close to the possible lesion for measurement of CRH and/or ACTH and compare the levels to a peripheral source (11). If a gradient is reported then this may further support the diagnosis of ectopic tumor (11). Other markers of potential interest in these cases include chromogranin A and CRH, which may be helpful in the follow-up of patients. Patients with ECS may present with pituitary hyperplasia if CRH is co-secreted, which should be considered when interpreting the imaging and biochemical results.

 

When ACTH-independent CS is suspected, imaging of the adrenals is the best next step. Imaging can be preferably with CT since it has good accuracy for lesions <1cm and less artifacts due to motion, but MRI may be an alternative to avoid radiation exposure. Ultrasound however is not accurate in identifying adrenal lesions other than large adrenocortical tumors (14). In ACTH-independent CS, it is important to review the anatomy of both adrenals; noting a unilateral lesion with atrophy of the contralateral adrenal supports the diagnosis of unilateral disease, whereas bilateral symmetrical adrenal enlargement or bilateral normal appearing adrenals suggests bilateral disease (Figure 2). In case of bilateral micronodular adrenocortical disease, adrenal anatomy is often read as normal or sometimes asymmetric appearance of the contour of the adrenals described as “beads on a string” may be apparent (94).

 

When bilateral adrenocortical disease is suspected, confirmation of the diagnosis prior to proceeding with surgical intervention involves the performance of Liddle’s test (95). The paradoxical increase of urinary free cortisol or 17-hydroxy steroids with increasing doses of dexamethasone is pathognomonic for PPNAD (95).

 

Figure 2. Typical imaging findings in a patient with Cushing disease (A-B), a cortisol-producing adrenal adenoma (C) and bilateral micronodular adrenocortical disease (E). Postcontrast sagittal (A) and coronal (B) MRI images of the pituitary showing a microadenoma (tip of arrows) as hypoechoic lesion. (C) Axial adrenal CT of a patient with a left adrenal adenoma (yellow asterisk) and atrophic contralateral adrenal (blue outline). (E) Axial adrenal CT of a patient with bilateral micronodular adrenocortical disease showing normal appearing adrenals (blue outline) with bilaterally symmetric thickness of the limbs of the adrenals.

 

TREATMENT

 

Surgical intervention is the first line of treatment in all types of CS whenever the source is identified. In patients with CD, transsphenoidal resection of the pituitary tumor is the preferred approach. Endoscopic or microscopic approaches have been attempted. A recent meta-analysis has not showed significant differences in the remission rate between the two approaches overall, but endoscopic approach may be preferrable in macroadenomas (96, 97). In very young patients, pneumatization of the sphenoid sinus may be incomplete and the surgical approach more be more difficult but transsphenoidal access is still possible (98). In rare cases of very young children with pituitary lesions or in giant complex pituitary tumors, the transcranial approach may be considered (99). Remission is defined as postoperative nadir cortisol levels of <2-5mcg/dl and early postoperative hypocortisolemia is a sensitive marker of durable remission (76, 100). In cases of non-remission patients may be managed with immediate reoperation and partial hypophysectomy (101). In the pediatric cohorts the remission rate after surgery ranges from 62 to 98% depending on the cohort and the criteria used (8, 102-104).

 

In cases of ACTH-independent CS, bilateral or unilateral adrenalectomy is recommended depending on the underlying cause (105). Although unilateral adrenalectomy has been suggested in cases of bilateral macronodular adrenocortical disease, data on unilateral adrenalectomy in micronodular adrenocortical disease are not clear (105).

 

ECS should primarily be managed with surgical resection.

 

In cases of persistent CD after surgery, medical therapy or radiation should be considered. At this time, no medical therapy for CS in the pediatric population has been approved by the FDA in the US and all treatments are considered as off-label use, but ketoconazole is approved by the European Medical Association for children >12 years of age. Medical therapies are divided in those directed to adrenal steroidogenesis, to pituitary tumor function, or to peripheral glucocorticoid action. Most commonly, steroidogenesis inhibitors are considered first line as they are more potent and faster acting. Of these, ketoconazole, metyrapone, osilodrostat, levo-ketoconazole and others have been used (106). Radiation therapy could be considered as an alternative second-line treatment but requires medical management and close monitoring until the radiation effect is apparent (107, 108). Finally, bilateral adrenalectomy is reserved for cases of severe CS persistent despite surgical or medical intervention. This is followed by lifelong adrenal insufficiency and patients should be monitored for the risk of Nelson syndrome (109).

 

POSTOPERATIVE MANAGEMENT

 

After successful surgical management, patients experience adrenal insufficiency. In CD the median duration of adrenal insufficiency is almost 12 months (110). Additionally, management of patients after remission of CS should also target symptoms of glucocorticoid withdrawal which may require supraphysiologic doses of glucocorticoids for a period of time and slow tapering to physiologic levels (111).

 

After recovery of the axis, regular screening for possible recurrence should be offered. Long term recurrence has been reported in 8-20% of pediatric patients with CD after initial postoperative remission (8). Screening for recurrence should be done preferably as in adults with two midnight or late-night salivary cortisol levels or with overnight dexamethasone suppression test annually (76).

 

REFERENCES

 

  1. Broder MS, Neary MP, Chang E, Cherepanov D, Ludlam WH. Incidence of Cushing's syndrome and Cushing's disease in commercially-insured patients <65 years old in the United States. Pituitary 2015, 18(3):283-289.
  2. Ragnarsson O, Olsson DS, Chantzichristos D, Papakokkinou E, Dahlqvist P, Segerstedt E, Olsson T, Petersson M, Berinder K, Bensing S, Hoybye C, Eden Engstrom B, Burman P, Bonelli L, Follin C, Petranek D, Erfurth EM, Wahlberg J, Ekman B, Akerman AK, Schwarcz E, Bryngelsson IL, Johannsson G. The incidence of Cushing's disease: a nationwide Swedish study. Pituitary 2019, 22(2):179-186.
  3. Holst JM, Horvath-Puho E, Jensen RB, Rix M, Kristensen K, Hertel NT, Dekkers OM, Sorensen HT, Juul A, Jorgensen JOL. Cushing's syndrome in children and adolescents: a Danish nationwide population-based cohort study. Eur J Endocrinol 2017, 176(5):567-574.
  4. Tatsi C, Stratakis CA. Pediatric Cushing Syndrome; an Overview. Pediatr Endocrinol Rev 2019, 17(2):100-109.
  5. Storr HL, Chan LF, Grossman AB, Savage MO. Paediatric Cushing's syndrome: epidemiology, investigation and therapeutic advances. Trends Endocrinol Metab 2007, 18(4):167-174.
  6. Herman V, Fagin J, Gonsky R, Kovacs K, Melmed S. Clonal origin of pituitary adenomas. J Clin Endocrinol Metab 1990, 71(6):1427-1433.
  7. Batista DL, Riar J, Keil M, Stratakis CA. Diagnostic tests for children who are referred for the investigation of Cushing syndrome. Pediatrics 2007, 120(3):e575-586.
  8. Tatsi C, Kamilaris C, Keil M, Saidkhodjaeva L, Faucz FR, Chittiboina P, Stratakis CA. Paediatric Cushing syndrome: a prospective, multisite, observational cohort study. Lancet Child Adolesc Health 2024, 8(1):51-62.
  9. Tatsi C, Stratakis CA. Aggressive pituitary tumors in the young and elderly. Rev Endocr Metab Disord 2020, 21(2):213-223.
  10. de Kock L, Sabbaghian N, Plourde F, Srivastava A, Weber E, Bouron-Dal Soglio D, Hamel N, Choi JH, Park SH, Deal CL, Kelsey MM, Dishop MK, Esbenshade A, Kuttesch JF, Jacques TS, Perry A, Leichter H, Maeder P, Brundler MA, Warner J, Neal J, Zacharin M, Korbonits M, Cole T, Traunecker H, McLean TW, Rotondo F, Lepage P, Albrecht S, Horvath E, Kovacs K, Priest JR, Foulkes WD. Pituitary blastoma: a pathognomonic feature of germ-line DICER1 mutations. Acta Neuropathol 2014, 128(1):111-122.
  11. Karageorgiadis AS, Papadakis GZ, Biro J, Keil MF, Lyssikatos C, Quezado MM, Merino M, Schrump DS, Kebebew E, Patronas NJ, Hunter MK, Alwazeer MR, Karaviti LP, Balazs AE, Lodish MB, Stratakis CA. Ectopic adrenocorticotropic hormone and corticotropin-releasing hormone co-secreting tumors in children and adolescents causing cushing syndrome: a diagnostic dilemma and how to solve it. J Clin Endocrinol Metab 2015, 100(1):141-148.
  12. More J, Young J, Reznik Y, Raverot G, Borson-Chazot F, Rohmer V, Baudin E, Coutant R, Tabarin A, Groupe Francais des Tumeurs E. Ectopic ACTH syndrome in children and adolescents. J Clin Endocrinol Metab 2011, 96(5):1213-1222.
  13. Yami Channaiah C, Karlekar M, Sarathi V, Lila AR, Ravindra S, Badhe PV, Malhotra G, Memon SS, Patil VA, Pramesh CS, Bandgar T. Paediatric and adolescent ectopic Cushing's syndrome: systematic review. Eur J Endocrinol 2023, 189(4):S75-S87.
  14. Tatsi C, Stratakis CA. Neonatal Cushing Syndrome: A Rare but Potentially Devastating Disease. Clin Perinatol 2018, 45(1):103-118.
  15. Ribeiro RC, Pinto EM, Zambetti GP, Rodriguez-Galindo C. The International Pediatric Adrenocortical Tumor Registry initiative: contributions to clinical, biological, and treatment advances in pediatric adrenocortical tumors. Mol Cell Endocrinol 2012, 351(1):37-43.
  16. Mete O, Erickson LA, Juhlin CC, de Krijger RR, Sasano H, Volante M, Papotti MG. Overview of the 2022 WHO Classification of Adrenal Cortical Tumors. Endocr Pathol 2022, 33(1):155-196.
  17. Kamilaris CDC, Stratakis CA, Hannah-Shmouni F. Molecular Genetic and Genomic Alterations in Cushing's Syndrome and Primary Aldosteronism. Front Endocrinol (Lausanne) 2021, 12:632543.
  18. Tirosh A, Valdes N, Stratakis CA. Genetics of micronodular adrenal hyperplasia and Carney complex. Presse Med 2018, 47(7-8 Pt 2):e127-e137.
  19. Stratakis CA. Adrenocortical tumors, primary pigmented adrenocortical disease (PPNAD)/Carney complex, and other bilateral hyperplasias: the NIH studies. Horm Metab Res 2007, 39(6):467-473.
  20. Makri A, Bonella MB, Keil MF, Hernandez-Ramirez L, Paluch G, Tirosh A, Saldarriaga C, Chittiboina P, Marx SJ, Stratakis CA, Lodish M. Children with MEN1 gene mutations may present first (and at a young age) with Cushing disease. Clin Endocrinol (Oxf) 2018, 89(4):437-443.
  21. Tatsi C, Flippo C, Stratakis CA. Cushing syndrome: Old and new genes. Best Pract Res Clin Endocrinol Metab 2020, 34(2):101418.
  22. Foulkes WD, Priest JR, Duchaine TF. DICER1: mutations, microRNAs and mechanisms. Nat Rev Cancer 2014, 14(10):662-672.
  23. Faure A, Atkinson J, Bouty A, O'Brien M, Levard G, Hutson J, Heloury Y. DICER1 pleuropulmonary blastoma familial tumour predisposition syndrome: What the paediatric urologist needs to know. J Pediatr Urol 2016, 12(1):5-10.
  24. Reincke M, Sbiera S, Hayakawa A, Theodoropoulou M, Osswald A, Beuschlein F, Meitinger T, Mizuno-Yamasaki E, Kawaguchi K, Saeki Y, Tanaka K, Wieland T, Graf E, Saeger W, Ronchi CL, Allolio B, Buchfelder M, Strom TM, Fassnacht M, Komada M. Mutations in the deubiquitinase gene USP8 cause Cushing's disease. Nat Genet 2015, 47(1):31-38.
  25. Ma ZY, Song ZJ, Chen JH, Wang YF, Li SQ, Zhou LF, Mao Y, Li YM, Hu RG, Zhang ZY, Ye HY, Shen M, Shou XF, Li ZQ, Peng H, Wang QZ, Zhou DZ, Qin XL, Ji J, Zheng J, Chen H, Wang Y, Geng DY, Tang WJ, Fu CW, Shi ZF, Zhang YC, Ye Z, He WQ, Zhang QL, Tang QS, Xie R, Shen JW, Wen ZJ, Zhou J, Wang T, Huang S, Qiu HJ, Qiao ND, Zhang Y, Pan L, Bao WM, Liu YC, Huang CX, Shi YY, Zhao Y. Recurrent gain-of-function USP8 mutations in Cushing's disease. Cell Res 2015, 25(3):306-317.
  26. Faucz FR, Tirosh A, Tatsi C, Berthon A, Hernandez-Ramirez LC, Settas N, Angelousi A, Correa R, Papadakis GZ, Chittiboina P, Quezado M, Pankratz N, Lane J, Dimopoulos A, Mills JL, Lodish M, Stratakis CA. Somatic USP8 Gene Mutations Are a Common Cause of Pediatric Cushing Disease. J Clin Endocrinol Metab 2017, 102(8):2836-2843.
  27. Albani A, Perez-Rivas LG, Dimopoulou C, Zopp S, Colon-Bolea P, Roeber S, Honegger J, Flitsch J, Rachinger W, Buchfelder M, Stalla GK, Herms J, Reincke M, Theodoropoulou M. The USP8 mutational status may predict long-term remission in patients with Cushing's disease. Clin Endocrinol (Oxf) 2018.
  28. Chen J, Jian X, Deng S, Ma Z, Shou X, Shen Y, Zhang Q, Song Z, Li Z, Peng H, Peng C, Chen M, Luo C, Zhao D, Ye Z, Shen M, Zhang Y, Zhou J, Fahira A, Wang Y, Li S, Zhang Z, Ye H, Li Y, Shen J, Chen H, Tang F, Yao Z, Shi Z, Chen C, Xie L, Wang Y, Fu C, Mao Y, Zhou L, Gao D, Yan H, Zhao Y, Huang C, Shi Y. Identification of recurrent USP48 and BRAF mutations in Cushing's disease. Nat Commun 2018, 9(1):3171.
  29. Tatsi C, Pankratz N, Lane J, Faucz FR, Hernandez-Ramirez LC, Keil M, Trivellin G, Chittiboina P, Mills JL, Stratakis CA, Lodish MB. Large Genomic Aberrations in Corticotropinomas Are Associated With Greater Aggressiveness. J Clin Endocrinol Metab 2019, 104(5):1792-1801.
  30. Corsello A, Ramunno V, Locantore P, Pacini G, Rossi ED, Torino F, Pontecorvi A, De Crea C, Paragliola RM, Raffaelli M, Corsello SM. Medullary Thyroid Cancer with Ectopic Cushing's Syndrome: A Case Report and Systematic Review of Detailed Cases from the Literature. Thyroid 2022, 32(11):1281-1298.
  31. Agaimy A, Kasajima A, Stoehr R, Haller F, Schubart C, Togel L, Pfarr N, von Werder A, Pavel ME, Sessa F, Uccella S, La Rosa S, Kloppel G. Gene fusions are frequent in ACTH-secreting neuroendocrine neoplasms of the pancreas, but not in their non-pancreatic counterparts. Virchows Arch 2023, 482(3):507-516.
  32. Wagner J, Portwine C, Rabin K, Leclerc JM, Narod SA, Malkin D. High frequency of germline p53 mutations in childhood adrenocortical cancer. J Natl Cancer Inst 1994, 86(22):1707-1710.
  33. Latronico AC, Pinto EM, Domenice S, Fragoso MC, Martin RM, Zerbini MC, Lucon AM, Mendonca BB. An inherited mutation outside the highly conserved DNA-binding domain of the p53 tumor suppressor protein in children and adults with sporadic adrenocortical tumors. J Clin Endocrinol Metab 2001, 86(10):4970-4973.
  34. Pinto EM, Billerbeck AE, Villares MC, Domenice S, Mendonca BB, Latronico AC. Founder effect for the highly prevalent R337H mutation of tumor suppressor p53 in Brazilian patients with adrenocortical tumors. Arq Bras Endocrinol Metabol 2004, 48(5):647-650.
  35. Schneider K, Zelley K, Nichols KE, Garber J: Li-Fraumeni Syndrome. In: GeneReviews((R)). edn. Edited by Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A. Seattle (WA); 1993.
  36. Goh G, Scholl UI, Healy JM, Choi M, Prasad ML, Nelson-Williams C, Kunstman JW, Korah R, Suttorp AC, Dietrich D, Haase M, Willenberg HS, Stalberg P, Hellman P, Akerstrom G, Bjorklund P, Carling T, Lifton RP. Recurrent activating mutation in PRKACA in cortisol-producing adrenal tumors. Nat Genet 2014, 46(6):613-617.
  37. Drougat L, Settas N, Ronchi CL, Bathon K, Calebiro D, Maria AG, Haydar S, Voutetakis A, London E, Faucz FR, Stratakis CA. Genomic and sequence variants of protein kinase A regulatory subunit type 1beta (PRKAR1B) in patients with adrenocortical disease and Cushing syndrome. Genet Med 2021, 23(1):174-182.
  38. Bonnet S, Gaujoux S, Launay P, Baudry C, Chokri I, Ragazzon B, Libe R, Rene-Corail F, Audebourg A, Vacher-Lavenu MC, Groussin L, Bertagna X, Dousset B, Bertherat J, Tissier F. Wnt/beta-catenin pathway activation in adrenocortical adenomas is frequently due to somatic CTNNB1-activating mutations, which are associated with larger and nonsecreting tumors: a study in cortisol-secreting and -nonsecreting tumors. J Clin Endocrinol Metab 2011, 96(2):E419-426.
  39. Stratakis CA, Carney JA, Lin JP, Papanicolaou DA, Karl M, Kastner DL, Pras E, Chrousos GP. Carney complex, a familial multiple neoplasia and lentiginosis syndrome. Analysis of 11 kindreds and linkage to the short arm of chromosome 2. J Clin Invest 1996, 97(3):699-705.
  40. Kaltsas G, Kanakis G, Chrousos G: Carney Complex. In: Endotext. edn. Edited by Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J et al. South Dartmouth (MA); 2000.
  41. Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS, Stratakis CA. Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients with the Carney complex. Nat Genet 2000, 26(1):89-92.
  42. Stratakis CA, Jenkins RB, Pras E, Mitsiadis CS, Raff SB, Stalboerger PG, Tsigos C, Carney JA, Chrousos GP. Cytogenetic and microsatellite alterations in tumors from patients with the syndrome of myxomas, spotty skin pigmentation, and endocrine overactivity (Carney complex). J Clin Endocrinol Metab 1996, 81(10):3607-3614.
  43. Espiard S, Vantyghem MC, Assie G, Cardot-Bauters C, Raverot G, Brucker-Davis F, Archambeaud-Mouveroux F, Lefebvre H, Nunes ML, Tabarin A, Lienhardt A, Chabre O, Houang M, Bottineau M, Stroer S, Groussin L, Guignat L, Cabanes L, Feydy A, Bonnet F, North MO, Dupin N, Grabar S, Duboc D, Bertherat J. Frequency and Incidence of Carney Complex Manifestations: A Prospective Multicenter Study With a Three-Year Follow-Up. J Clin Endocrinol Metab 2020, 105(3).
  44. Horvath A, Mericq V, Stratakis CA. Mutation in PDE8B, a cyclic AMP-specific phosphodiesterase in adrenal hyperplasia. N Engl J Med 2008, 358(7):750-752.
  45. Libe R, Fratticci A, Coste J, Tissier F, Horvath A, Ragazzon B, Rene-Corail F, Groussin L, Bertagna X, Raffin-Sanson ML, Stratakis CA, Bertherat J. Phosphodiesterase 11A (PDE11A) and genetic predisposition to adrenocortical tumors. Clin Cancer Res 2008, 14(12):4016-4024.
  46. Lodish MB, Yuan B, Levy I, Braunstein GD, Lyssikatos C, Salpea P, Szarek E, Karageorgiadis AS, Belyavskaya E, Raygada M, Faucz FR, Izzat L, Brain C, Gardner J, Quezado M, Carney JA, Lupski JR, Stratakis CA. Germline PRKACA amplification causes variable phenotypes that may depend on the extent of the genomic defect: molecular mechanisms and clinical presentations. Eur J Endocrinol 2015, 172(6):803-811.
  47. Carney JA, Lyssikatos C, Lodish MB, Stratakis CA. Germline PRKACA amplification leads to Cushing syndrome caused by 3 adrenocortical pathologic phenotypes. Hum Pathol 2015, 46(1):40-49.
  48. Horvath A, Boikos S, Giatzakis C, Robinson-White A, Groussin L, Griffin KJ, Stein E, Levine E, Delimpasi G, Hsiao HP, Keil M, Heyerdahl S, Matyakhina L, Libe R, Fratticci A, Kirschner LS, Cramer K, Gaillard RC, Bertagna X, Carney JA, Bertherat J, Bossis I, Stratakis CA. A genome-wide scan identifies mutations in the gene encoding phosphodiesterase 11A4 (PDE11A) in individuals with adrenocortical hyperplasia. Nat Genet 2006, 38(7):794-800.
  49. Espiard S, Drougat L, Libe R, Assie G, Perlemoine K, Guignat L, Barrande G, Brucker-Davis F, Doullay F, Lopez S, Sonnet E, Torremocha F, Pinsard D, Chabbert-Buffet N, Raffin-Sanson ML, Groussin L, Borson-Chazot F, Coste J, Bertagna X, Stratakis CA, Beuschlein F, Ragazzon B, Bertherat J. ARMC5 Mutations in a Large Cohort of Primary Macronodular Adrenal Hyperplasia: Clinical and Functional Consequences. J Clin Endocrinol Metab 2015, 100(6):E926-935.
  50. Brown RJ, Kelly MH, Collins MT. Cushing syndrome in the McCune-Albright syndrome. J Clin Endocrinol Metab 2010, 95(4):1508-1515.
  51. Dumitrescu CE, Collins MT. McCune-Albright syndrome. Orphanet J Rare Dis 2008, 3:12.
  52. Carney JA, Ho J, Kitsuda K, Young WF, Jr., Stratakis CA. Massive neonatal adrenal enlargement due to cytomegaly, persistence of the transient cortex, and hyperplasia of the permanent cortex: findings in Cushing syndrome associated with hemihypertrophy. Am J Surg Pathol 2012, 36(10):1452-1463.
  53. Schweiger BM, Esakhan CL, Frishberg D, Grand K, Garg R, Sanchez-Lara PA. Pediatric Cushing syndrome: An early sign of an underling cancer predisposition syndrome. Am J Med Genet A 2021, 185(9):2824-2828.
  54. Magiakou MA, Mastorakos G, Oldfield EH, Gomez MT, Doppman JL, Cutler GB, Jr., Nieman LK, Chrousos GP. Cushing's syndrome in children and adolescents. Presentation, diagnosis, and therapy. N Engl J Med 1994, 331(10):629-636.
  55. Greening JE, Storr HL, McKenzie SA, Davies KM, Martin L, Grossman AB, Savage MO. Linear growth and body mass index in pediatric patients with Cushing's disease or simple obesity. J Endocrinol Invest 2006, 29(10):885-887.
  56. Keil MF, Graf J, Gokarn N, Stratakis CA. Anthropometric measures and fasting insulin levels in children before and after cure of Cushing syndrome. Clin Nutr 2012, 31(3):359-363.
  57. Gunther DF, Bourdeau I, Matyakhina L, Cassarino D, Kleiner DE, Griffin K, Courkoutsakis N, Abu-Asab M, Tsokos M, Keil M, Carney JA, Stratakis CA. Cyclical Cushing syndrome presenting in infancy: an early form of primary pigmented nodular adrenocortical disease, or a new entity? J Clin Endocrinol Metab 2004, 89(7):3173-3182.
  58. Lodish MB, Gourgari E, Sinaii N, Hill S, Libuit L, Mastroyannis S, Keil M, Batista DL, Stratakis CA. Skeletal maturation in children with Cushing syndrome is not consistently delayed: the role of corticotropin, obesity, and steroid hormones, and the effect of surgical cure. J Pediatr 2014, 164(4):801-806.
  59. Stratakis CA, Mastorakos G, Mitsiades NS, Mitsiades CS, Chrousos GP. Skin manifestations of Cushing disease in children and adolescents before and after the resolution of hypercortisolemia. Pediatr Dermatol 1998, 15(4):253-258.
  60. Leong GM, Abad V, Charmandari E, Reynolds JC, Hill S, Chrousos GP, Nieman LK. Effects of child- and adolescent-onset endogenous Cushing syndrome on bone mass, body composition, and growth: a 7-year prospective study into young adulthood. J Bone Miner Res 2007, 22(1):110-118.
  61. Lodish MB, Hsiao HP, Serbis A, Sinaii N, Rothenbuhler A, Keil MF, Boikos SA, Reynolds JC, Stratakis CA. Effects of Cushing disease on bone mineral density in a pediatric population. J Pediatr 2010, 156(6):1001-1005.
  62. Ward LM. Glucocorticoid-Induced Osteoporosis: Why Kids Are Different. Front Endocrinol (Lausanne) 2020, 11:576.
  63. Merke DP, Giedd JN, Keil MF, Mehlinger SL, Wiggs EA, Holzer S, Rawson E, Vaituzis AC, Stratakis CA, Chrousos GP. Children experience cognitive decline despite reversal of brain atrophy one year after resolution of Cushing syndrome. J Clin Endocrinol Metab 2005, 90(5):2531-2536.
  64. Ferrau F, Korbonits M. Metabolic comorbidities in Cushing's syndrome. Eur J Endocrinol 2015, 173(4):M133-157.
  65. Lodish M, Patronas NJ, Stratakis CA. Reversible posterior encephalopathy syndrome associated with micronodular adrenocortical disease and Cushing syndrome. Eur J Pediatr 2010, 169(1):125-126.
  66. Birdwell L, Lodish M, Tirosh A, Chittiboina P, Keil M, Lyssikatos C, Belyavskaya E, Feelders RA, Stratakis CA. Coagulation Profile Dynamics in Pediatric Patients with Cushing Syndrome: A Prospective, Observational Comparative Study. J Pediatr 2016, 177:227-231.
  67. Wagner J, Langlois F, Lim DST, McCartney S, Fleseriu M. Hypercoagulability and Risk of Venous Thromboembolic Events in Endogenous Cushing's Syndrome: A Systematic Meta-Analysis. Front Endocrinol (Lausanne) 2018, 9:805.
  68. Wurth R, Rescigno M, Flippo C, Stratakis CA, Tatsi C. Inflammatory biomarkers in the evaluation of pediatric endogenous Cushing syndrome. Eur J Endocrinol 2022, 186(4):503-510.
  69. Tatsi C, Boden R, Sinaii N, Keil M, Lyssikatos C, Belyavskaya E, Rosenzweig SD, Stratakis CA, Lodish MB. Decreased lymphocytes and increased risk for infection are common in endogenous pediatric Cushing syndrome. Pediatr Res 2018, 83(2):431-437.
  70. Gkourogianni A, Lodish MB, Zilbermint M, Lyssikatos C, Belyavskaya E, Keil MF, Stratakis CA. Death in pediatric Cushing syndrome is uncommon but still occurs. Eur J Pediatr 2015, 174(4):501-507.
  71. Rahman SH, Papadakis GZ, Keil MF, Faucz FR, Lodish MB, Stratakis CA. Kidney Stones as an Underrecognized Clinical Sign in Pediatric Cushing Disease. J Pediatr 2016, 170:273-277 e271.
  72. Weinberg JR, Voudouri M, Keil M, Stratakis CA, Tatsi C. The utility of IGF1 in the evaluation of pediatric patients with endogenous hypercortisolemia. Pediatr Res 2023.
  73. Stratakis CA, Mastorakos G, Magiakou MA, Papavasiliou E, Oldfield EH, Chrousos GP. Thyroid function in children with Cushing's disease before and after transsphenoidal surgery. J Pediatr 1997, 131(6):905-909.
  74. Unuane D, Tournaye H, Velkeniers B, Poppe K. Endocrine disorders & female infertility. Best Pract Res Clin Endocrinol Metab 2011, 25(6):861-873.
  75. Magiakou MA, Mastorakos G, Gomez MT, Rose SR, Chrousos GP. Suppressed spontaneous and stimulated growth hormone secretion in patients with Cushing's disease before and after surgical cure. J Clin Endocrinol Metab 1994, 78(1):131-137.
  76. Fleseriu M, Auchus R, Bancos I, Ben-Shlomo A, Bertherat J, Biermasz NR, Boguszewski CL, Bronstein MD, Buchfelder M, Carmichael JD, Casanueva FF, Castinetti F, Chanson P, Findling J, Gadelha M, Geer EB, Giustina A, Grossman A, Gurnell M, Ho K, Ioachimescu AG, Kaiser UB, Karavitaki N, Katznelson L, Kelly DF, Lacroix A, McCormack A, Melmed S, Molitch M, Mortini P, Newell-Price J, Nieman L, Pereira AM, Petersenn S, Pivonello R, Raff H, Reincke M, Salvatori R, Scaroni C, Shimon I, Stratakis CA, Swearingen B, Tabarin A, Takahashi Y, Theodoropoulou M, Tsagarakis S, Valassi E, Varlamov EV, Vila G, Wass J, Webb SM, Zatelli MC, Biller BMK. Consensus on diagnosis and management of Cushing's disease: a guideline update. Lancet Diabetes Endocrinol 2021.
  77. Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM, Montori VM. The diagnosis of Cushing's syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2008, 93(5):1526-1540.
  78. Babic N, Yeo KTJ, Hannoush ZC, Weiss RE: Endocrine Testing Protocols: Hypothalamic Pituitary Adrenal Axis. In: Endotext. edn. Edited by Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J et al. South Dartmouth (MA); 2000.
  79. Newell-Price J, Trainer P, Perry L, Wass J, Grossman A, Besser M. A single sleeping midnight cortisol has 100% sensitivity for the diagnosis of Cushing's syndrome. Clin Endocrinol (Oxf) 1995, 43(5):545-550.
  80. Tatsi C, Stratakis C: Cushing Disease: Diagnosis and Treatment. In: Pituitary Disorders of Childhood Diagnosis and Clinical Management. edn. Edited by Kohn B: Springer Nature Switzerland AG; 2019: 89-114.
  81. Carroll T, Raff H, Findling JW. Late-night salivary cortisol for the diagnosis of Cushing syndrome: a meta-analysis. Endocr Pract 2009, 15(4):335-342.
  82. Newell-Price J, Trainer P, Besser M, Grossman A. The diagnosis and differential diagnosis of Cushing's syndrome and pseudo-Cushing's states. Endocr Rev 1998, 19(5):647-672.
  83. Stratakis CA. An update on Cushing syndrome in pediatrics. Ann Endocrinol (Paris) 2018, 79(3):125-131.
  84. Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, Sen K, Salgado LR, Colao A, Biller BM, Pasireotide BSG. High variability in baseline urinary free cortisol values in patients with Cushing's disease. Clin Endocrinol (Oxf) 2014, 80(2):261-269.
  85. Luger A, Deuster PA, Kyle SB, Gallucci WT, Montgomery LC, Gold PW, Loriaux DL, Chrousos GP. Acute hypothalamic-pituitary-adrenal responses to the stress of treadmill exercise. Physiologic adaptations to physical training. N Engl J Med 1987, 316(21):1309-1315.
  86. Mericq MV, Cutler GB, Jr. High fluid intake increases urine free cortisol excretion in normal subjects. J Clin Endocrinol Metab 1998, 83(2):682-684.
  87. Bertrand PV, Rudd BT, Weller PH, Day AJ. Free cortisol and creatinine in urine of healthy children. Clin Chem 1987, 33(11):2047-2051.
  88. Hindmarsh PC, Brook CG. Single dose dexamethasone suppression test in children: dose relationship to body size. Clin Endocrinol (Oxf) 1985, 23(1):67-70.
  89. Ferrigno R, Hasenmajer V, Caiulo S, Minnetti M, Mazzotta P, Storr HL, Isidori AM, Grossman AB, De Martino MC, Savage MO. Paediatric Cushing's disease: Epidemiology, pathogenesis, clinical management and outcome. Rev Endocr Metab Disord 2021, 22(4):817-835.
  90. Findling JW, Raff H. DIAGNOSIS OF ENDOCRINE DISEASE: Differentiation of pathologic/neoplastic hypercortisolism (Cushing's syndrome) from physiologic/non-neoplastic hypercortisolism (formerly known as pseudo-Cushing's syndrome). Eur J Endocrinol 2017, 176(5):R205-R216.
  91. Tatsi C, Bompou ME, Flippo C, Keil M, Chittiboina P, Stratakis CA. Paediatric patients with Cushing disease and negative pituitary MRI have a higher risk of nonremission after transsphenoidal surgery. Clin Endocrinol (Oxf) 2021, 95(6):856-862.
  92. Frete C, Corcuff JB, Kuhn E, Salenave S, Gaye D, Young J, Chanson P, Tabarin A. Non-invasive Diagnostic Strategy in ACTH-dependent Cushing's Syndrome. J Clin Endocrinol Metab 2020, 105(10).
  93. Varlamov E, Hinojosa-Amaya JM, Stack M, Fleseriu M. Diagnostic utility of Gallium-68-somatostatin receptor PET/CT in ectopic ACTH-secreting tumors: a systematic literature review and single-center clinical experience. Pituitary 2019, 22(5):445-455.
  94. Courcoutsakis NA, Tatsi C, Patronas NJ, Lee CC, Prassopoulos PK, Stratakis CA. The complex of myxomas, spotty skin pigmentation and endocrine overactivity (Carney complex): imaging findings with clinical and pathological correlation. Insights Imaging 2013, 4(1):119-133.
  95. Stratakis CA, Sarlis N, Kirschner LS, Carney JA, Doppman JL, Nieman LK, Chrousos GP, Papanicolaou DA. Paradoxical response to dexamethasone in the diagnosis of primary pigmented nodular adrenocortical disease. Ann Intern Med 1999, 131(8):585-591.
  96. Chen J, Liu H, Man S, Liu G, Li Q, Zuo Q, Huo L, Li W, Deng W. Endoscopic vs. Microscopic Transsphenoidal Surgery for the Treatment of Pituitary Adenoma: A Meta-Analysis. Front Surg 2021, 8:806855.
  97. Broersen LHA, Biermasz NR, van Furth WR, de Vries F, Verstegen MJT, Dekkers OM, Pereira AM. Endoscopic vs. microscopic transsphenoidal surgery for Cushing's disease: a systematic review and meta-analysis. Pituitary 2018, 21(5):524-534.
  98. Marino AC, Taylor DG, Desai B, Jane JA, Jr. Surgery for Pediatric Pituitary Adenomas. Neurosurg Clin N Am 2019, 30(4):465-471.
  99. Luzzi S, Giotta Lucifero A, Rabski J, Kadri PAS, Al-Mefty O. The Party Wall: Redefining the Indications of Transcranial Approaches for Giant Pituitary Adenomas in Endoscopic Era. Cancers (Basel) 2023, 15(8).
  100. Ironside N, Chatain G, Asuzu D, Benzo S, Lodish M, Sharma S, Nieman L, Stratakis CA, Lonser RR, Chittiboina P. Earlier post-operative hypocortisolemia may predict durable remission from Cushing's disease. Eur J Endocrinol 2018, 178(3):255-263.
  101. Ram Z, Nieman LK, Cutler GB, Jr., Chrousos GP, Doppman JL, Oldfield EH. Early repeat surgery for persistent Cushing's disease. J Neurosurg 1994, 80(1):37-45.
  102. Yordanova G, Martin L, Afshar F, Sabin I, Alusi G, Plowman NP, Riddoch F, Evanson J, Matson M, Grossman AB, Akker SA, Monson JP, Drake WM, Savage MO, Storr HL. Long-term outcomes of children treated for Cushing's disease: a single center experience. Pituitary 2016, 19(6):612-624.
  103. Batista DL, Oldfield EH, Keil MF, Stratakis CA. Postoperative testing to predict recurrent Cushing disease in children. J Clin Endocrinol Metab 2009, 94(8):2757-2765.
  104. Savage MO, Chan LF, Grossman AB, Storr HL. Work-up and management of paediatric Cushing's syndrome. Curr Opin Endocrinol Diabetes Obes 2008, 15(4):346-351.
  105. Meloche-Dumas L, Mercier F, Lacroix A. Role of unilateral adrenalectomy in bilateral adrenal hyperplasias with Cushing's syndrome. Best Pract Res Clin Endocrinol Metab 2021, 35(2):101486.
  106. Castinetti F. Cushing's disease: role of preoperative and primary medical therapy. Pituitary 2022, 25(5):737-739.
  107. Castinetti F, Brue T, Ragnarsson O. Radiotherapy as a tool for the treatment of Cushing's disease. Eur J Endocrinol 2019, 180(5):D9-D18.
  108. Geer EB, Ayala A, Bonert V, Carmichael JD, Gordon MB, Katznelson L, Manuylova E, Shafiq I, Surampudi V, Swerdloff RS, Broder MS, Cherepanov D, Eagan M, Lee J, Said Q, Neary MP, Biller BMK. Follow-up intervals in patients with Cushing's disease: recommendations from a panel of experienced pituitary clinicians. Pituitary 2017, 20(4):422-429.
  109. Reincke M, Fleseriu M. Cushing Syndrome: A Review. JAMA 2023, 330(2):170-181.
  110. Tatsi C, Neely M, Flippo C, Bompou ME, Keil M, Stratakis CA. Recovery of hypothalamic-pituitary-adrenal axis in paediatric Cushing disease. Clin Endocrinol (Oxf) 2021, 94(1):40-47.
  111. Zhang CD, Li D, Singh S, Suresh M, Thangamuthu K, Nathani R, Achenbach SJ, Atkinson EJ, Van Gompel JJ, Young WF, Bancos I. Glucocorticoid withdrawal syndrome following surgical remission of endogenous hypercortisolism: a longitudinal observational study. Eur J Endocrinol 2023, 188(7):592-602.

 

Familial Isolated Pituitary Adenoma

ABSTRACT

 

Familial Isolated Pituitary Adenoma (FIPA) is a term used to identify a genetic condition with pituitary tumors without other endocrine or other associated abnormalities. FIPA families contribute around 2% to the overall incidence of pituitary tumors. FIPA is a heterogeneous disease both in terms of the clinical phenotype as well as from the genetic background point of view. Some FIPA families have been identified to have germline mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene leading to incomplete penetrance of young-onset, mostly growth hormone, mixed growth hormone/prolactin-secreting, or prolactin-secreting pituitary adenomas. Due to the low penetrance, almost half of the AIPmutation-positive patients do not have a positive family history. Duplication of the orphan G protein coupled receptor GPR101 gene, located on Xq26.3, leads to high penetrance pituitary hyperplasia or adenoma resulting in infant-onset GH excess, usually with concomitant hyperprolactinemia, named X-linked acrogigantism (XLAG). The majority of the FIPA families, however, have no known genetic mutation. Their clinical picture includes various types of pituitary adenomas, either homogeneous (all affected family members have the same adenoma type) or heterogeneous (different adenoma types within the same family), presenting with low penetrance and an age of onset not significantly different from patients with sporadic pituitary adenomas. Here we review the clinical features, genetics and screening aspects of FIPA.

 

INTRODUCTION

 

Familial Isolated Pituitary Adenoma (FIPA) is a relatively new term. Introduced by Professor Beckers in 1999, FIPA describes families with pituitary adenoma and no other associated symptoms (1, 2). As opposed to occurring in isolation, familial pituitary adenomas have been recognized in several syndromic diseases, such as the classical MEN1 syndrome or Carney complex or the most recently described, such as hereditary paraganglioma syndromes (3-5), MEN4, and DICER1 syndrome (6) (Figure 1).  For additional information we refer the reader to other chapters within ENDOTEXT on syndromic familial pituitary adenomas.

 

Figure 1. Germline or Mosaic Mutations Causing Pituitary Tumors. Details for the syndromic forms can be found, among others, in the following sections https://www.endotext.org/chapter/multiple-endocrine-neoplasia-type-i/, https://www.endotext.org/chapter/carney-complex/, https://www.endotext.org/chapter/pituitary-adenomas-in-childhood/ and in these references (6-10).

 

Descriptions of familial pituitary adenoma families have been around for several hundreds of years, but only over the last decade has the clinical phenotype and, in some cases, the genetic abnormality been described. Interestingly, some of the patients with germline mutations present as simplex patients without any known family history, either due to low penetrance or due to de novo mutations.

 

Figure 2. Family Trees Demonstrating Examples of the Various Types of FIPA Families. In some AIP mutation-negative FIPA families unaffected obligate carriers can be identified by their position in the family tree, while in other family’s possible carriers of the unidentified gene cannot be identified. AIP mutation-positive kindreds can be ‘families’ or simplex cases. Most XLAG kindreds are simplex cases with females having de novo germline mutations while males have somatic mosaic mutations.

 

Previous data suggest that FIPA families contribute around 2% of the overall incidence of pituitary tumors, but this number may increase with increasing recognition of this clinical entity.

 

Around 10-20% of all FIPA families and 50% of familial isolated GH-producing Tumor families (11, 12) have been identified to have mutations within the aryl hydrocarbon receptor interacting protein (AIP) gene, located at 11q13. Germline mutations in AIP have also been identified in patients with young-onset pituitary adenomas, mostly GH-secreting or prolactin-secreting or silent GH/prolactin-producing adenomas with no apparent family history. These are called ‘simplex’ cases. Until recently, no somatic mutations had been described in the AIP gene in pituitary or other tumors (1). Duplication of the orphan G protein-coupled receptor GPR101 causes X-linked acrogigantism (XLAG) (13).While most of the XLAG cases are due to de novo mutations (germline or somatic mosaicism (14, 15)), to date three families have also been described. The causative gene for the rest and therefore the vast majority (90% only considering kindreds with 2 or more affected subjects) of FIPA families is currently unknown (16). Recently, a microdeletion upstream the GHRH gene, on chromosome 20, has been identified as another possible cause of severe infant-onset gigantism (17). New candidate genes are under active investigation in somatic and familial cases of pituitary adenomas (18), but some need further validation. Representative examples of FIPA family trees are shown in Figure 2.

 

CLINICAL FEATURES OF FIPA

 

Families with AIP mutations usually have a characteristic phenotype, which is usually substantially different from that ofAIP mutation-negative phenotype. In this section, we compare characteristics of AIP-mutated and non-AIP-mutated FIPA. Germline chromosomal defects leading to gigantism, including XLAG and a recently described microdeletion in chromosome 20 that leads to GHRH overexpression, have a drastically different phenotype and are discussed separately below.

 

Tumor Types

 

FIPA families can be homologous (i.e. all affected family members have the same type of tumor) or heterologous (i.e. family members can have different type of tumor) (Figure 2). Therefore, pure acromegaly, pure prolactinoma, and pure non-functioning pituitary adenoma (NFPA) families have been identified, while also mixed families such as acromegaly-prolactinoma, acromegaly-NFPA, prolactinoma-NFPA, prolactin-corticotrophinoma or even acromegaly-prolactinoma-NFPA families have been described. Somato-mammotrophinomas occur commonly, but are not consistently reported, probably as a result of variations in the reporting of tumor histology type. Figure 3a, b and c demonstrate the distribution of histological tumor types in FIPA families.

 

Figure 3a. Proportion of histological tumor types in the AIP positive FIPA population in the International FIPA Consortium cohort (n=911) (19).

Figure 3b. Proportion of tumor types in AIP mutation-positive FIPA families (12).

Figure 3c. Proportion of tumor types in AIP mutation-negative FIPA families (12).

 

In a study including familial as well as simplex (apparently sporadic) patients with germline AIP mutations, 78% of 96 patients developed GH-secreting adenomas (20) (half of the GH-secreting adenomas were somato-mammotrophinomas), 13.5% of patients developed prolactinomas, 7% developed non-functioning pituitary adenomas (NFPAs), and 1 patient developed a TSH-secreting adenoma. In another study, comprising 171 patients carrying AIPmutations, based on clinical diagnosis 70% had somatotrophinomas, 11% mixed GH/PRLomas, 12% had prolactinomas, and 8% had clinically non-functioning tumors (12). On histological testing some tumors show plurihormonal profile (Figure 3b). It is important to note that some non-functioning tumors are found to be somatotroph/lactotroph upon histological examination (21) – these are therefore ‘silent adenomas’. The distribution of tumors amongst 318 non-AIPmutated FIPA families (1310 patients) is represented in Figure 3c (12). Somatotrophinomas are the most common tumor type in both AIP mutation-positive and negative FIPA families (12, 19).

 

Gender Distribution

 

While higher numbers of males are identified with AIP mutations both in familial and simplex setting (12, 20), ascertainment bias due to physiological later puberty of boys and their normally taller stature cannot be ruled out (19), as in a carefully-studied large AIP mutation family equal number of affected males and females are present (22). There is a greater prevalence of females within AIP mutation-negative families, probably due to a higher number of prolactinomas (19).

 

Age of Onset

 

AIP gene mutation-positive FIPA patients have an earlier age of onset of diagnosis compared to those with AIP mutation-negative familial (23) or sporadic (20) pituitary adenomas. The age of onset of pituitary adenoma symptoms is 8 years earlier in the AIP mutation-positive group (mean age 19 years, SD ± 9.5, p<0.001), with diagnosis being made 6 years earlier (mean age of diagnosis 24.3, SD ± 11.9 vs 30, SD ± 13.5, p<0.001) than in the AIP mutation-negative population (12). In our international FIPA cohort, the familial cohort with AIP mutation-positive tumors had a peak age of onset during the 2nd and 3rd decades of life, with 65% of these patients’ developing symptoms aged ≤18 years (28.8% in the AIP mutation-negative group) and 87% by the age of 30 years (12). Previous work has shown that those families with AIP mutation-negative tumors demonstrate a more even spread of occurrence between the ages of 20 and 50, with a peak incidence around the age of 30 years old (19); the latest data suggests that the modal age group (42%) is 20-29 years (12). 

 

Young (<30 years) onset simplex patients, the AIP mutation-positive group, also developed tumors at a younger age than the mutation-negative group, with median ages of 16 years (IQR 14.8-22.3) and 22 years (IQR 16-26) respectively (19).

 

In the Bart’s international cohort, over 80% of the families with AIP mutations have at least one affected patient with gigantism or disease onset before the age of 18 years, while only 3 out of 46 AIP mutation-negative families have an onset of pituitary adenoma before the age of 18 years (23). Interestingly, probably due to earlier recognition of symptoms in affected FIPA families, the age of tumor onset appeared to be earlier in the second generation than in the first (mean age 29 ±10.2 years vs. 50.5± 14.2 years p<0.0001) (24).

 

Disease Penetrance

 

Disease penetrance in FIPA is incomplete. As there is a clear natural bias of affected patient referral and the clinical and genetic data in the individual families are incomplete, the calculation of disease penetrance is difficult. Additionally, it is important that penetrance always be considered in the context of the subject’s age.

 

In AIP positive mutation families, current data suggests 12.5-30% penetrance, but ranges between 10-90%, also depending on available data (19, 20, 23). It seems that the nature of the AIP mutation (truncating or non-truncating) does not have any effect on penetrance (19).

In AIP mutation-negative families, penetrance calculations are even more difficult as carrier unaffected family members (other than obligate carriers) cannot be distinguished from non-carrier unaffected subjects. The current calculation based on affected subjects, obligate carriers and 50% of potential carriers suggest 38±16% (23), but this is obviously a very significant overestimate.

 

Another way to compare penetrance between AIP positive and negative families is to count the known affected subjects within families. Penetrance in AIP mutation-negative families is probably lower than in AIP mutation-positive families, as the mean number of patients with disease in AIP mutation-positive families is 3.2±1.8 and in AIP mutation-negative families 2.2±0.5, P<0.001 (23).

 

De novo AIP mutation has been described in two cases so far: in a child with prolactinoma (c.721A>T; p.Lys241*) where the AIP mutation was not found in the parents (paternity confirmed) or his sister (19, 25). A second case was with identical twin girls, where both of them carry a mutation in the leukocyte derived DNA (p.R304*), while their parents (paternity confirmed) were negative (26).

 

Phenocopies (patients who show manifestations of a disease that are usually associated with mutations of a particular gene but instead are, in this case, due to another etiology) (27) have been described in families with AIP mutations (16, 23) and are probably present in AIP negative families as well, therefore careful and cautious genetic studies and counselling need to be conducted in every family.

 

Tumor Behavior

 

SIZE

 

FIPA patients in general have larger, more aggressive tumors and earlier onset of disease compared to sporadic pituitary adenomas (11, 20, 23, 28).

 

Macroadenomas predominate amongst AIP mutation-negative and positive FIPA groups. However, when compared to sporadic pituitary adenomas, AIP gene mutation-positive FIPA patients were more likely to have larger tumors (1, 11, 19, 28) and macroadenomas (19), and these tumors were more likely to invade the extrasellar region (19, 20).

 

There was no statistical difference between the AIP mutation-positive and negative groups in the occurrence of giant (>40mm) adenomas (19), nor in the incidence of macroadenomas (mutation-positive 83.2% vs 79.2% p=0.259) or cavernous sinus invasion (mutation-positive 36.7% vs 28.3%, p=0.122) (12). Suprasellar extension was more frequent in the pituitary adenomas of AIP mutation-positive FIPA patients (mutation-positive 54.3% vs 42.4%, p=0.043).

 

No correlation was observed between the presence of truncating and non-truncating AIP mutations and the size of the pituitary adenoma, the incidence of macroadenoma or the propensity to invade extrasellar structures (19).

 

APOPLEXY PROPENSITY   

 

Pituitary apoplexy is a relatively rare event; incidence is variously estimated to be as high as 6.8% (in 560 adenoma cases) (29) to as low as 0.6% (in 664 adenoma cases) (30). In a previous study, it was shown that apoplexy occurred more commonly in individuals with AIP mutation-positive tumors than those with mutation-negative tumors (7.6% vs 1.3% of cases respectively) (19). No size difference was observed between tumors that did and those that did not undergo apoplexy in the AIP mutation-positive tumor group (19). Excluding simplex cases from these analyses (i.e. just considering patients with a family history of pituitary adenomas) demonstrated an even bigger disparity in apoplexy incidence with AIP mutation-positive tumors having an apoplexy rate of 10.6% vs 2.3% in mutation-negative families (19). The latest data from the international FIPA consortium has shown similar rates of apoplexy (8.2% vs 3.6% respectively, p=0.009) (12). Familial apoplexy has also been described in AIP mutation-positive families (19, 31). It was previously observed that GH-secreting tumors with AIP mutations were significantly more likely than their mutation-negative counterparts to undergo apoplexy (19) and this has been demonstrated once again (8.3% vs 2.8% p=0.005) (12). The mechanism for this observation is unclear.

 

Treatment Resistance

 

Many of the somatotrophinomas described in FIPA families have been described as sparsely granulated adenomas (1), a subtype which has been previously suggested to respond less well to somatostatin analogues and to be more aggressive (32, 33). Sparsely granulated adenomas occur more commonly in AIP mutation-positive GH-secreting adenomas than in their mutation-negative GH secreting counterparts (19). In one study (12), all of the AIP mutation-positive somatotrophinomas were sparsely granulated, compared to 68% in the AIP mutation-negative group (p<0.001).

 

There is speculation that somatostatin analogues mediate their anti-proliferative effects through AIP up-regulation, which in turn increases the expression of ZAC1, a tumor suppressor gene known to be upregulated by somatostatin analogues  (34, 35), therefore, dysfunction at the AIP step would reduce the expression of ZAC1 and so the usefulness of this class of drug (36).  Another potential mechanism for this treatment resistance involving defective Gαi signaling has been postulated and is discussed in detail below.

 

It has previously been observed that AIP mutation-positive tumors are more difficult to treat - mutation-positive somatotrophinomas undergo less shrinkage and there is a smaller reduction in GH production with first generation somatostatin analogues than in the mutation-negative sporadic patients (1, 20, 28, 37). This may be accounted for by a relative paucity of expression of SSTR2 in the former (38); however, in human samples rather, a higher level of SSTR2 was found (36), and this is also seen in a pituitary Aip-knockout mouse model (39, 40). A greater need for re-operation after initial surgery and a greater use of multiple therapies and >2 types of therapy, including radiotherapy (12) and the failure of pegvisomant to control IGF-1 (20) have also been described. However, some studies (19) failed to demonstrate any difference in the number of therapeutic interventions between AIP positive and negative mutation tumors. Where primary surgery has failed to control the tumor’s GH production, there is some evidence that pegvisomant (37, 41), or pasireotide in patients whose tumor expresses the type 5 somatostatin receptor (38, 42), may reduce the IGF-1 burden.  In some cases, drastic treatment is necessary: for example, in the youngest known case, who presented at the age of 4 years-old, surgery followed by first generation somatostatin analogue, temozolomide, bevacizumab, radiotherapy, pegvisomant, gamma knife therapy and somatostatin analogue combined with increasing dose of pegvisomant, was necessary (43).

 

No correlation was observed between the presence of truncating and non-truncating AIP mutation tumors and the number of treatment modalities required by these patients (19).

 

In addition to sparsely granulated histopathology, other well-known predictive factors of resistance to first generation somatostatin analogues are younger age at diagnosis, hyperintense T2 image on MRI, and low tumor expression of somatostatin receptor subtype 2 (44). Recently, a machine-learning based model accounting for age at diagnosis, sex, pretreatment GH and IGF-1 levels, tumor granulation pattern and expression of somatostatin receptor subtypes 2 and 5 was shown to predict therapeutic response to first generation somatostatin analogues with high negative and positive predictive values (45).

 

Currently, some experts already suggest that the first-line medical treatment for patients that show one or more of these features could be pegvisomant or pasireotide; and that pegvisomant could be preferred in patients with diabetes or low somatostatin receptor subtype 5, whilst pasireotide could be preferred in the presence of significant tumor volume (44). Therefore, in select cases, these two drugs could be considered early in postsurgical medical therapy in patients with persistent disease, especially in younger patients with ongoing uncontrolled height gain, as seen in patients with AIPmutations.

 

Hormone Secretion

 

When matched with acromegaly mutation-negative controls, AIP mutation-positive somatotrophinomas produce more growth hormone (GH) (20) but there was no difference in the levels of IGF-1 (12, 20). Prolactin co-secretion was more common in AIP mutation-positive GH secreting tumors than their non-AIP mutated counterparts (19).

 

Gigantism was observed to be more common among AIP mutation-positive patients (55.9% vs 18.2%, p=0.005) and was the most common clinical diagnosis (12) – which is predicted by their earlier onset of disease, with cases in males predominating in both AIP positive and negative patients (19): 60% of FIPA families in one study had at least one case of gigantism and instances of two cases of gigantism within the same family only occurred in AIP mutation-positive families (19).

 

No correlation was observed between the presence of truncating and non-truncating AIP mutations and the incidence of GH secreting tumors (19); however, there was a significantly greater prevalence of gigantism amongst the GH secreting tumor patients in those with truncating as opposed to non-truncating AIP mutations (54.7% vs 30%). There is also a suggestion that patients with GH-secreting adenomas and the truncating R304* mutation present more commonly at a very young age then rest of the described AIP mutation-positive population with GH secreting adenomas.

 

A previous case report described the co-existence of pituitary hyperplasia and pituitary adenoma in two AIP mutation-positive adenomas from a family. Loss of heterozygosity was seen in the adenoma tissue but not in the surrounding hyperplastic tissue and loss of AIP protein expression was seen in the adenoma tissue with preservation of AIPexpression in the hyperplastic tissue (46). Villa and colleagues hypothesize that this may demonstrate that tumorigenesis is a multi-stage event starting with hyperplasia in haploinsufficient tissue and then the development of further genetic events (including loss of the one remaining wild-type AIP allele) leading to true adenoma formation. They suggest that this could explain the incomplete penetrance seen in pituitary disease in AIP mutation-positive subjects (46).

 

In GH-secreting non-AIP mutated sporadic pituitary tumors, an association was noted between the levels of AIP staining on histology and the aggressiveness of the adenoma. Low levels of AIP staining were associated with a more aggressive phenotype (higher Ki-67 index and a greater likelihood of suprasellar tumor extension) when compared to tumors with higher levels of AIP staining. In the same tumors, none of those with low AIP staining showed significant shrinkage despite pre-operative treatment with a somatostatin analogue. Tumors treated pre-operatively with somatostatin analogues that did shrink showed a higher level of AIP on immunohistochemistry (47).

 

No difference in rates of hypopituitarism was seen between AIP mutation-positive and negative patients with pituitary adenomas at diagnosis (12).

 

Other Tumors in Individuals with an AIP Mutation

 

In one study (19) involving 290 AIP mutation-positive individuals (some with pituitary adenomas), there were 10 cases of tumors occurring outside of the pituitary gland in 9 individuals. These included a gastrointestinal stromal tumor, glioma, meningioma, non-Hodgkin’s lymphoma, and spinal ependymoma. Parathyroid adenomas were excluded from this analysis due to the rare finding of AIP mutations in parathyroid adenomas (48), as were colonic polyps and thyroid nodules due to their frequent occurrence in patients with acromegaly (19). Four of the 9 individuals with extra-pituitary tumors had GH-secreting pituitary tumors, the other 5 were AIP mutation carriers without pituitary tumors.

 

While AIP acts as a tumor suppressor gene in the pituitary gland, and patients with pituitary tumors show heterozygous loss-of-function mutations of AIP, a possible role for AIP as an oncogene has been described in other tumor types. To date, increased expression of AIP was found in association with increased tumorigenic and metastatic properties of colorectal cancer cells (49), with increased survival of primary diffuse large B cell lymphoma (DLBCL) cells (50), and with a bad prognosis in cholangiocarcinoma (51). In colorectal cancer, increased AIP expression was associated with increased cell migration and epithelial-to-mesenchymal transition, possibly by the facilitation of N-cadherin expression and suppression of functional E-cadherin on the cell surface (49). On the other hand, for DLBCL, AIP promoted tumor survival by reducing ubiquitin-mediated proteasomal degradation of BCL6, a protein that reduces the transcription of pro-apoptotic genes such as TP53 and that is frequently overexpressed in DLBCL (50).

 

Therefore, AIP behaves as a double agent, either as a tumor suppressor or as an oncogene, and further studies on AIP regulation mechanisms will be essential for a better understanding of AIP derived tumorigenesis and for unravelling new possible therapeutic targets (52).

 

THE GENETICS OF FIPA

 

The currently known genes causing FIPA are AIP and GPR101 and we will discuss the diseases associated with these genes in detail. Furthermore, there are some pituitary adenoma cases described with other germline mutations, that will be more briefly addressed, as they are still under investigation and require additional validation.

 

AIP

 

There are over 100 heterozygous mutations identified in AIP, showing an autosomal dominant inheritance pattern with incomplete penetrance (53). Mutations that affect the AIP gene commonly lead to truncated or missing protein due to nonsense mutations, small deletions or large deletions, insertions, splicing or promoter mutations, while 21% result in full length mutated protein due to missense mutations or in-frame deletions or insertions (Figure 4). Large deletions cannot be identified with Sanger sequencing and other technologies, such as MLPA, or next generation sequencing methods are required to identify them.

 

Figure 4. Distribution of mutation types found within the AIP gene in the International FIPA consortium (12).

Figure 5. The three-dimensional structure of the AIP protein. Three characteristic tetratricopeptide (TPR) domains, the A and B helices of the first TPR domain, orange, TPR2 blue. TPR3 green and the 7th C-terminal alpha helix with light blue (54, 55).

 

The AIP protein is a well-conserved molecular chaperone, with multiple binding partners. It has three tetratricopeptide (TPR) repeats, conserved anti-parallel pair of alpha helices and a final 7th alpha helix at its carboxyl terminal end (Figure 5). This C-terminal section is known to be important for interaction with other proteins and therefore, it is postulated, that in the case of FIPA it loses its ability to bind its binding partners, such as the aryl hydrocarbon receptor (AHR) or phosphodiesterase (PDE) subtype 4A5, and therefore loses its activity as a tumor suppressor (56).

 

There are a few mutational hotspots, the majority affecting CpG sites, where a mutation has been identified in several independent patients or families (Table 1).

 

Table 1. A Few Examples of AIP Mutation ‘Hotspots’

Variant

References (examples)

c.910C>T; p.R304*

 

 

Cazabat et al. 2007 (57)

Daly et al. 2007 (11)

Georgitsi et al. 2007 (58)

Igreja et al. 2010 (23)

Leontinou et al. 2008 (28)

Variglou et al. 2009 (59)

Vierimaa et al. 2006 (16)

Chahal et al. 2011 (60)

Hernandez-Ramirez et al. 2015 (19)

Ramirez Rentaria et al. 2016 (26)

Marques et al. 2020 (12)

c.811C>T; p.R271W

Daly et al. 2007 (11)

Jennings et al. 2009 (61)

Hernandez-Ramirez et al. 2015 (19)

c.721A>T; p.R81*

Leontiou et al. 2008 (28)

Toledo et al. 2010 (62)

Hernandez-Ramirez et al. 2015 (19)

Marques et al. 2020 (12)

 

AIP Mouse Models

 

AIP knockout in mice is lethal in utero and is associated with ventricular septal defects, double outlet right ventricle and pericardial edema (63). The embryonic mice are also unable to undergo a crucial step in initiating adult erythropoiesis at E11-14, a step which is vital for embryonic survival beyond E13.5 (64). This suggests that AIP may have an important role to play in fetal growth signaling in utero.

 

Heterozygote AIP knockout mice invariably develop mostly GH-secreting pituitary tumors, with 100% penetrance by the age of 18 months, compared to wild-type mice where around 1/3 of mice spontaneously developed prolactin-secreting adenomas, but no GH adenomas are observed (65). AIP expression was lost in these GH-secreting tumors and this corresponded to higher tumor proliferation rates (65), compared to spontaneous pituitary adenomas in the wild-type littermates, with normal AIP expression. These data mirror the increased aggressiveness of tumors seen in mutation-positive FIPA families (11, 20, 23, 28). ARNT expression was also lost in the mouse tumors (65), reflecting a pattern observed in human mutation-positive tumors (66) and therefore suggesting a possible role for loss of ARNT in the development of pituitary tumors (65). Somatotroph-specific AIP deficient mice (sAipKO) have also been created, using Cre/Lox and Flp/Frt technology (67). In keeping with the heterozygote AIP knockout mice described above, >80% of the sAipKO mice developed GH secreting adenomas by 40 weeks of age, by 18 weeks they also displayed elevated IGF-1 and GH levels, increased body and organ size (compared to control animals) and glucose intolerance. Pituitary hyperplasia was consistently observed in the sAipKO mice (on histology and on MRI imaging), suggesting (but not absolutely proving) a progression from hyperplasia to adenoma. The investigators point out that 40 weeks of age for a mouse represents ‘middle adulthood’ and so hypothesize that, in common with other tumors, additional somatic mutations are required on top of the AIP loss of function for somatotroph tumors to occur (67). A pituitary-specific Aipknockout using the Hesx1/Cre model has also developed gigantism with elevated IGF-1 levels (40).

 

ARNT knockout mice die in utero in early gestation (68, 69): the reasons for this are disputed, in one study it appeared that there was faulty angiogenesis in the yolk sac (69), whilst in another the embryos survived slightly longer and had a normally developed yolk sac vasculature but the placental vasculature failed to develop correctly. The embryos in the latter study also displayed a range or anomalies, including neural tube closure defects, brain hypoplasia and placental hemorrhage (68). It has been hypothesized, therefore, that ARNT plays a role in angiogenesis in response to hypoxia secondary to the increasing tissue mass in embryonic development (69).

 

Ahr knockout mice are viable, though they too suffer physiologic dysfunction, including cardiac hypertrophy (with cardiac myocyte enlargement but without the molecular signatures that would indicate cardiac overload) and subsequent cardiomyopathy (70). These mice also have hypertension (71), reduced body weight, reduced reproductive capabilities, smaller livers as a result of a patent ductus venosus, persistence of fetal vascular and liver parenchymal structures and aberrant vasculature in the kidneys. This underlines the importance of AhR signaling mechanisms in the development of a normal, mature vasculature (72).  AhR protein-protein interactions were further characterized, with one of the most interesting interactions being with the mitochondrial protein MRPL40 (73), which codes for a mitochondrial ribosomal 39S subunit. Deletions in this gene have been associated with the 22q11.2 deletion syndromes Velo-cardial facial syndrome and Di George syndrome (OMIM #188400), both of which involve congenital cardiac malformations, further suggesting the importance of AhR in normal cardiac development.

 

It has been suggested that interplay between AhR and ARNT/HIF1α may govern normal vascular development (72).

 

MECHANISM OF TUMORIGENESIS IN PITUITARY ADENOMAS WITH AIP MUTATIONS

 

In the pituitary, AIP is a tumor suppressor, and truncating mutations presumably lead to loss of function mutations. However, for missense mutations change in protein folding or loss of partner protein binding sites could explain the lack of function. Based on data from half-life studies, (74) it seems that a significant proportion of the missense mutations lead to unstable proteins and rapid degradation explaining the loss of function. Furthermore, in vitro measured half-life of missense proteins correlated well with age of onset of disease. (74)

AIP interacts with numerous other molecules (see Table 2), full details of each of these interactions has recently been summarized (56).

 

Table 2. A List of Factors that Have Been Demonstrated to Interact with the AIP Protein (56)

Viral Proteins

Hepatitis B Virus X protein (HBV X)

Epstein Barr Virus Nuclear Antigen 3 (EBNA3)

AIP-AHR-Hsp90 Complex

Aryl Hydrocarbon Receptor (AHR)

Heat Shock Protein 90 (Hsp90)

Heat Shock Cognate 70 (Hsc70)

Aryl Hydrocarbon Receptor Nuclear Translocator (ARNT)

p23

AIP self-association

Cytoskeletal Proteins

Possible interaction with actin

Tubulin (75)

Phosphodiesterases

PDE4A5

PDE2A3

Nuclear Receptors

Estrogen Receptor α (ERα)

Glucocorticoid Receptor (GR)

Peroxisome Proliferator-Activated Receptor α (PPARα)

Thyroid Hormone Receptor β1 (TRβ1)

Transmembrane Receptors

RET

EGFR

G Proteins

Translocase of the Outer Membrane of Mitochondria (TOMM20) Proteins (64)

Survivin (64)

Cardiac Troponin Interacting Kinase 3 (TNNI3K)

Protein Kinase A (76)

 

The exact mechanism by which AIP mutations lead to pituitary tumor formation is unclear; however, several theories have been put forward. AHR is widely expressed in the body and binds numerous compounds, both endogenous and exogenous (77, 78). It is a nuclear transcription factor and prior to ligand binding it is found in the cellular cytoplasm, bound to AIP (77, 78). It is known that AHR is a receptor for environmental pollutants, such as dioxin – a known carcinogen. The binding of dioxin leads to increased AHR nuclear translocation, with activation of detoxification mechanisms (79), including increased expression of the enzyme CYP1A1, which has also been shown to bio-activate polycyclic aromatic hydrocarbon carcinogens (80, 81). Interestingly, an increase of acromegaly incidence (82) has been described in a heavily polluted industrial area. Pituitary adenoma incidence was also studied in an area heavily polluted with dioxin after a chemical factory accident, but data were not sufficient to draw appropriate conclusions (83). A recent follow-up study (84) examined links between the characteristics of patients with GH-secreting pituitary adenomas, residing in an area of high pollution and AHR/AIP variants. It was found that pituitary tumors were significantly larger and IGF-1 burden significantly greater in patients with AHR/AIP gene variants who lived in polluted areas compared to either those who had no gene variants and lived in the same highly polluted areas or those who had gene variants but lived in cleaner areas. Further, the use of somatostatin analogues in patients with GH-secreting pituitary adenomas, who also had AHR/AIP gene variants and lived in highly polluted areas, seemed to be less effective (IGF-1 only normalized in 14%). Overall, the reduction in GH/IGF-1 levels did not reach statistical significance. GH secreting pituitary patients with no AHR/AIP variants had a statistically significant reduction in GH/IGF-1, as did those without gene variants living in polluted areas (IGF-1 normalized in 54-56% of cases).  These data need confirmation.

 

Fibroblasts with heterozygous AIP mutations taken from patients have lower AIP protein levels (probably through nonsense-mediated decay of truncated proteins (74)) compared to wild-type fibroblast controls, but AHR expression is unaffected. However, AIP mutation did result in altered regulation of the AHR transcriptional target CYP1B1, both with and without AHR ligand stimulation (85). The mechanism by which this happens and therefore the role of AHR in signaling in pituitary tumorigenesis is still to be elucidated.

 

It has been noted that the loss of function of the AIP gene allows dysregulated ERα mediated gene transcription by its disinhibition (86). Cumulatively, high levels of estrogen and therefore estrogen mediated gene transcription products have been associated with an increased risk of developing various tumors, including pituitary tumors (86, 87) and so this work provides a novel avenue for investigation into pituitary tumorigenesis.

 

In the previous years, the role of cAMP elevation in pituitary tumors has been further investigated - It had previously been noted that cAMP levels were elevated in a subset of pituitary tumors (88). cAMP is a mitogenic factor in somatotroph cells, this therefore suggests a link between its dysregulation and tumor growth (89, 90). AIP is known to be a binding partner of some of the phosphodiesterases. AIP binding to PDE4A appears to inhibit its phosphodiesterase activity; however, this did not appear to prevent the cell’s in vitro ability to reduce forskolin-induced cAMP driven transcription. Therefore, it was felt unlikely that AIP-phosphodiesterase was the mechanism for cAMP elevation in pituitary tumors (91).  The same study also hypothesized that  AIP’s interactions with other binding partners is vital in its role of reducing cAMP, as R304* mutant AIP transfected cells (which produces a truncated AIP protein, losing its protein-interacting C-terminal) were not able to reduce  cAMP signaling in the same way that  wild-type AIP transfected cells could (91).  This correlated with reduced GH secretion after forskolin stimulation in the wild-type AIP cells, but not in the AIP mutant cells (91).

 

Disordered cAMP regulation is also seen in McCune Albright syndrome – where there is a mutation of the GNAS1 gene which results in a constitutionally active Gαs and raised cAMP (92), and Carney complex (93) – where there is an inactivating mutation in the PRKAR1A gene, a subunit of Protein Kinase A (PKA), a cAMP dependent kinase (94).There is evidence that AIP interacts with some of the subfamily protein of Gα (95), providing a possible way through which AIP can influence intracellular cAMP levels. To investigate this further, Tuominen et al. (96) developed an immortalized fibroblast cell line from the embryos of an AIP knockout mouse. AIP knockout in the mouse embryonic fibroblasts (MEFs) cell line resulted in higher cAMP level with a 2-3 times increase the AIP knockout cells. This result was concordant with AIP knockdown in a rat pituitary tumor cell line, with an observed 20-30% rise in cAMP levels.

 

Figure 6. Role of G alpha Inhibitory Protein. (A) - cells with normally functioning G alpha inhibitory protein (Gai-2) respond to stimulation of the somatostatin receptor (SSTR) by somatostatin (SST) by inhibiting the action of adenylyl cyclase, reducing the cell's secretory and proliferative capabilities. The role of AIP in this process is unknown, but cells with defective/absent AIP (B) also have a reduction in Gai-2 and so a lack of response from SST binding to SSTR with resulting disinhibition of adenylyl cyclase and increased GH secretion and cell proliferation.

 

Sequential knockdown of the Ga subfamily of proteins (Ga12, Ga13, Ga11, Gaq, Ga14, Ga15, and Gas,) produced only a significant reduction in cAMP levels in AIP knockout mouse cells when Gas and Ga13 were knocked down, although this effect was not sufficient to explain the observed difference in cAMP levels between AIP knockout and wild-type cells (96). Sequential knockdowns of the Ga inhibitory subfamilies (Gai-1, Gai-2, and Gai-3) was also performed. Gai-2 and Gai-3 knockdown caused a rise in cAMP levels by 77% and 115% respectively in wild-type MEFs, but minimal changes in the cAMP levels in AIP knockout cells. This was interpreted as evidence of a pre-existing defect in the Gai system of the AIPknockout cells (96) (Figure 6).

 

Immunohistochemical staining was subsequently performed on human somatotrophinomas which showed a reduction in the Gai-2 expression in AIP mutation-positive tumors compared to mutation-negative tumors (96, 97). No difference was observed in the expression of Gai-3 between the two types of tumors (96).

 

These findings may also explain the observed phenomenon whereby AIP mutation-positive tumors appear to respond poorly to somatostatin analogue treatment, as somatostatin receptors mediate reduction in cAMP levels through the Gaisystem (98), particularly through Gai-2 and therefore defective Gai signaling in AIP mutation-positive tumors maybe abrogate the effect of these drugs (96).

 

There is also in vitro evidence that AIP may play a role in reducing PKA activity through binding to its subunits (catalytic Cα and regulatory R1α). It was shown that AIP is able to interact with these two subunits, either as part of the PKA complex or separately. Cα stabilizes AIP and also R1α.  Overexpression of AIP lowered PKA activity, perhaps through inhibition of Cα or through the stabilization of the inactivating Cα-R1α complex. AIP overexpression also led to lower levels of Cα in the nucleus. Conversely, AIP silencing led to an increase in PKA activity. AIP’s interaction with these subunits is partly mediated by its c-terminal and so this may explain why common AIP truncation mutations (such as R304*), which affect this region, have a shorter protein half-life. It is hypothesized that this would then lead to lower intracellular AIP levels and may contribute to tumorigenesis through increased PKA activity (76).

 

The most recent and plausible mechanism relates to an interaction between AIP and the tyrosine kinase receptor RET. Although the first report on this interaction was over a decade ago (59), only recently there has been new insight about how this interaction affects tumorigenesis in the pituitary gland (99).  RET is a dependent receptor in somatotroph cells: in the absence of its ligand GDNF, the monomeric RET receptor is processed intracellularly by caspase-3, leading to PIT1 accumulation and upregulation of the RET/PIT1/ARF/p53-apoptotic pathway (99).  AIP was shown to be a key factor in the initial steps of this pathway, by forming a complex with RET/caspase-3/PKCδ, that allows for the intracellular processing of RET. In the absence of AIP or in the presence of pathological mutations in AIP, there is an inhibition of RET-induced apoptosis, that may be a key feature in somatotroph hyperplasia and adenoma formation (99).  However, PIT1 is a transcription factor that is present in somatotroph, lactotroph and thyrotroph cells; therefore, despite previous studies focusing mostly in somatotroph tumors, the same pathway is probably involved in other tumor types, such as prolactinomas (99), and this seems to be the explanation for the tissue specificity of AIP mutations. In line with this finding, the reported pituitary tumors in patients with AIP mutations are mostly GH and/or prolactin secreting tumors, but also clinically non-functioning adenomas with positive GH and/or prolactin immunostaining and, in one case, thyrotropinoma (12, 20, 100). There have been no unequivocal cases of corticotrophinomas or gonadotroph adenomas in patients with pathological AIP mutations. This extraordinary finding may pave the way for new therapeutic options in sporadic and familial cases of pituitary tumors with AIP mutations.

 

The increased tendency of AIP mutation-positive tumors to invade locally may be a result of an altered tumor microenvironment. One study (40) observed markedly more infiltration of tumors by macrophages in human AIP mutation-positive adenomas compared to sporadic somatotroph tumors. There was also an upregulation in the tumor-derived cytokine, CCL5, which is chemotactic for leukocytes. The macrophages themselves may play an important role in breaching local structures with their secretion of matrix metalloproteinases (MMP2 & 9) (101). Gene expression profiling experiments comparing AIP mutation-positive human pituitary adenomas to sporadic human pituitary adenomas showed a partial epithelial to mesenchymal transition pattern in keeping with a tumor that invades locally but exceedingly rarely metastasizes (40). In recent years, intensive research on pituitary tumor microenvironment has expanded our knowledge on pituitary tumor behavior and tumorigenesis mechanisms and raised the possibility for immunotherapy in aggressive and refractory pituitary tumors (102).

 

In contrast, few studies have focused on the mechanisms of AIP regulation. miR-34, a microRNA that binds to the 3-UTR region of AIP, was shown to be overexpressed and to downregulate AIP at the protein level in sporadic somatotrophinomas with low AIP expression (103) and in somatotrophinomas due to germline AIP mutations (104). Additionally, the high expression of miR-34 is one of the mechanisms driving the increased intracellular cAMP levels seen in AIP mutation-positive tumors (104). Thus, overexpression of miR-34 promotes cell proliferation and migration and may be responsible for the invasive phenotype and typical resistance to first generation somatostatin analogues seen in these tumors (103, 104). Recently, a regulation of AIP at the transcription level was also proposed. GTF2B, a transcription factor that binds the 5-UTR region of AIP, was shown to promote AIP expression and inhibit somatotroph cell proliferation and invasion (105).

 

AIP Mutations and Associations with Other Tumors

 

Germline AIP variants (R304Q, this variant is controversial, likely to be benign) were noted in sporadic parathyroid adenomas in 2 (unrelated) out of 136 patients in one study. One of these patients had a co-existent MEN1 mutation; both had reduced AIP staining in their tumors at histology (48). Concomitant AIP and MEN1 deletions through chromosomal translocations with a variety of partners are also associated with hibernomas (benign brown fat tumors). AIP transcription is down-regulated in these tumors (106) and its loss results in the upregulation of the brown fat marker UCP1 (107). Two patients from different FIPA kindreds, carriers of germline pathogenic mutations in AIP (Leu115Trpfs*41 and p.Q285*) with unaffected pituitary, were described to have follicular thyroid carcinomas showing loss of heterozygosity in the AIPlocus in the tumor tissue (42, 108), raising the possibility for a role of AIP mutation as an initiating event in both pituitary and thyroid. However, differentiated thyroid carcinoma (DTC) is rare in acromegaly, and the most frequent tumor mutations found in patients with known pathogenic AIP mutations are very similar to the ones found in sporadic cases, mostly comprising mutations of BRAF and NRAS (108). Therefore, the potential role of AIP mutations as a possible rare initiating event on the pathogenesis of DTC, although unlikely, requires further investigation.

 

OTHER POSSIBLE CANDIDATE GENES

 

Currently, only two well-characterized genes have been implicated in the pathogenesis of FIPA: AIP, the most common one, and GPR101. However, they only account for a minority of patients with FIPA, while other genes remain largely unknown.

 

At present, the genetics of familial and apparently sporadic pituitary tumors is under active investigation and some new candidate genes have been identified, but additional data is required to convincingly support them as a possible cause of FIPA.

 

Recently, germline loss of function mutations in the peptidylglycine α-amidating monooxygenase (PAM) gene were described in one family with pituitary gigantism and in multiple sporadic cases of several types of pituitary adenomas (18). PAM plays an important role in post-translational processing and secretion of hormones and is highly expressed in all pituitary cells, but the mechanisms linking its altered function with hormone hypersecretion still require clarification. Also, the fact that some of the identified PAM variants were relatively common, and that no deleterious variants were identified in other familial cases from 17 FIPA kindreds in the validation cohort raises some reasonable doubts. Therefore, additional studies in FIPA kindreds are required to further explore and validate this new candidate gene.

 

Another gene, described in sporadic corticotrophinomas, is CABLES1. Heterozygous germline mutations in CABLES1appear to decrease the negative feedback response from glucocorticoids, resulting in increased corticotroph cell growth. They were identified in two young adults, two children with Cushing’s Disease, and in one unaffected parent (109); but, to date, there have been no reports of possible familial cases with this mutation. Cushing disease is only rarely described in FIPA families, mostly in kindreds with heterogeneous tumor types (19). In homogenous corticotroph adenoma families no CABLES1 mutation has been identified (Korbonits unpublished observation). Corticotrophinomas have not been reported in kindreds with AIP mutation (19), and this is also in line with the recently described RET-derived AIP tissue specificity for PIT1 expressing cells (99).

 

A gain of function mutation in PRLR has been described in association with sporadic and familial prolactinomas (110), but additional data is needed to convincingly reinforce that association. Other germline mutations have also been associated with familial pituitary tumors (RXRG, TH, CDH23)(53, 111, 112), but lack functional validation studies as well as independent confirmation to support them as possible candidates involved in the pathogenesis of FIPA (113).

 

Additional conditions with excess GH in the absence of pituitary tumors have been described, and include germline mutations in genes such as IGSF1 and NF1.

 

IGSF1 is a transmembrane glycoprotein that is highly expressed in the anterior pituitary and hypothalamus, and that is considered essential for normal hormone production (114-116). Loss-of-function mutations in IGSF1 have been associated with an X-linked syndrome of central hypothyroidism and a variable prevalence of other endocrinopathies, including disharmonious pubertal development with delayed testosterone rise but normal or advanced testicular growth and postpubertal macroorchidism, hyperprolactinemia and GH dysregulation (114, 117). A minority of male children with such mutations show partial and transient GH deficiency, while adults more often show high IGF-1 levels, a 2- to 3-fold increase in GH pulsatile and basal secretion and mild acromegaloid features (117, 118). Similar features of GH excess were observed in mice (117). A potentially pathogenic variant in IGSF1 was described in three individuals from the same family showing somato-mammotroph hyperplasia or tumor and gigantism (115), but, to date, most case series of patients with IGSF1 mutations have consistently showed normal height and no evidence of pituitary tumors (116, 117, 119). It has been proposed that IGSF1 acts as a regulator of pituitary hormone synthesis, but the mechanism behind this is still poorly understood (114, 117).

 

Pathogenic mutations in the NF1 gene lead to neurofibromin deficiency and neurofibromatosis type 1 (NF-1). NF-1 is an autosomal dominant condition with increased risk of several benign and malignant tumors, including optic pathway gliomas (OPG), that are frequently diagnosed at a young age. An association between NF-1 and increased growth velocity or tall stature due to GH excess has been described in several case series, with a prevalence ranging from 4.5% (120) to 46% (in large deletions of NF1) (121). Excess GH is diagnosed in children with NF-1 and OPG, with a prevalence of 10.9% in this patient group according to the largest series published (122). The most plausible and widely accepted mechanism to explain this association is an induced hypothalamic dysfunction from infiltrative OPG, with reduced somatostatinergic inhibition of GH secretion, corresponding to the fact that there is absence of other pituitary abnormalities in the majority of cases (123). Another suggestion is that GPR101 dysregulation may occur. However, there are some case reports of NF-1 with concomitant pituitary hyperplasia or tumor, with or without OPG, which leads to the hypothesis that GHRH overexpression may be another possible mechanism leading to excess GH (123). Nevertheless, the pathophysiology of GH excess in NF-1 remains to be clarified.

 

GERMLINE CHROMOSOMAL DEFECTS PRESENTING WITH PITUITARY HYPERSECRETION/GIGANTISM- XLAG

 

This is a unique condition described in 2014 caused by a microduplication at Xq26.3 area containing the GPR101 gene, resulting in the overexpression of the orphan G protein coupled receptor GPR101 (13). It may be familial or sporadic, and can be due to a germline or a mosaic somatic mutation (14, 15). It shows an X-linked dominant inheritance with complete penetrance. Most cases are de novo germline (female) or mosaic (males) cases, with, to date, only three kindreds described where affected mothers passed on the mutation to male offspring (124-126). It constitutes 8-10% of the cases with gigantism (125, 127), and practically all the non-syndromic infant-onset gigantism.

 

XLAG Characteristics

 

In addition to the most prominent symptom of very early-onset gigantism with significantly elevated growth velocity, acral enlargement and coarse facial features are also observed (37). Fasting hyperinsulinemia was noted in 1/3 of patients and around 20% had acanthosis nigricans (125). Elevated BMI is often observed, and up to 1/3 of patient with XLAG have increased appetite, something not noted previously in gigantism. Hyperprolactinemia accompanies the GH excess in over 80% of the cases. Three quarters of the patients are females. GHRH levels can be normal or slightly elevated, and in some patients a paradoxical response was seen to the TRH test (127).

 

Tumor Types

 

All GPR101 duplication-related pituitary tumors described so far are GH producing, with the majority also secreting prolactin. There are a few cases of pure GH excess patients, some of these with hyperplasia rather than tumor (128). A rare GPR101 germline variant (p.E308D) does not play a role in somatotrophinoma tumorigenesis based on human (127, 129, 130) and in vitro data (131).

 

Age of Onset

 

Accelerated growth has been reported as early as 2-3 months of age (125), and abnormal hormone levels started to develop soon after birth in a prenatally diagnosed case (126). The median age of onset of rapid growth is at 1 year (range 0.5-2) with a median age at diagnosis being 3 years old (range 1-22) (13, 132).

 

Somatic Mosaicism

 

It seems that male patients, except the few familial cases, in which a germline duplication is inherited from an affected mother (124-126), have mosaic GPR101 duplication with pituitary tissue (and other tissues) showing the microduplication, while blood-derived DNA is negative or has a low level of mutation burden (14, 15, 127). The phenotype of somatic and germline GPR101 duplication patients is the same (132).

 

Tumor Behavior

 

SIZE

 

The size of the pituitary is variable in XLAG cases ranges from large tumors (133) to pituitary hyperplasia (13, 14, 127). It is currently unclear why some patients develop tumors while others have hyperplasia, both have been described in males and females. While Ki-67 is low in the tumor samples in most cases and such tumors do not show any tendency to invasion or apoplexy (127), invasive growth and a high Ki-67 has also been described (126, 133).

 

HORMONE SECRETION

 

Xq26.3 microduplication tumors invariably secrete GH and frequently also prolactin (13, 125). Random levels of GH were markedly raised in one study of 18 XLAG patients with a median of 52.5 times the upper limit of normal (range 6-300 times upper limit of normal) (125).

 

TREATMENT

 

Treatment of XLAG is complex and the tumors may grow rapidly, producing not only local effects due to their size but also causing worsening systemic manifestations of gigantism through their hormone production if not treated promptly (133). Despite widespread expression of type 2 somatostatin receptors, it has proved difficult to control GH levels in XLAG with somatostatin analogues or prolactin with dopamine agonists, even at relatively high doses. Extensive neurosurgery is often needed and effective, but the rates of post-operative hypopituitarism are high (125). In contrast, radiation therapy typically does not lead to disease control (125, 133). First generation somatostatin analogues are also usually ineffective in controlling GH hypersecretion, even in the presence of high tumor expression of somatostatin receptor 2 (125). In patients not controlled by surgery, the GH antagonist pegvisomant has proven effective in controlling IGF-1 levels (14, 41, 125, 128), but radiotherapy may be used as an alternative for tumor control if radical surgery is not possible. Patients with pituitary hyperplasia have previously been treated with hypophysectomy (134), while now combined treatment with somatostatin analogue, cabergoline and pegvisomant provides appropriate control (14). If lesion control and prolactin is not an issue, then patients can be treated just with pegvisomant (135).

 

Mechanism of Tumorigenesis in XLAG

 

It is unclear what role the hypothalamus plays and what is the role of the pituitary tissue in this disease. As some patients do not have a tumor, but produce very high level of GH, abnormal hypothalamic regulation could play a key role. Indeed, some patients have elevated circulating GHRH levels and mutated cells respond strongly to GHRH (136). GPR101 is strongly expressed in the normal pituitary during fetal development, from 19 weeks of gestation onwards, with levels declining through to ‘very low’ in adult life, suggesting a role in pituitary maturation (137). It is strongly over-expressed (both mRNA and protein) in the pituitary lesions of XLAG patients (131, 138). A recent paper has identified the mechanism for this. The duplication disrupts the regulatory region borders around the GPR101 gene (the so-called topologically associated domain or TAD) and this leads to overexpression of GPR101 by regulatory elements that normally do not regulate the expression of this gene (139). Therefore, XLAG is the first endocrine TADopathy. GPR101 has been shown to strongly activate the cAMP pathway. This therefore suggests a mechanism by which its overexpression may lead to tumorigenesis. The transient overexpression of GPR101 in GH3 rat pituitary tumor cells produced increased cellular proliferation and an increase in GH secretion, supporting this hypothesis (13).

 

MICRODELETION CAUSING GHRH OVEREXPRESSION

 

This novel condition, described for the first time in 2023 (17), is another genetic cause of severe non-syndromic infant-onset gigantism. It is caused by a heterozygous microdeletion upstream of the GHRH gene, in chromosome 20, that leads to aberrant splicing and produces a chimeric mRNA consisting of exon 1 of the TTI1 gene followed by all the coding exons of the GHRH gene. Since TTI1 is ubiquitously expressed and exon 1 has features of an active promotor, this fusion gene leads to constitutive GHRH overexpression and ectopic production of GHRH. There is only one case described so far, in a Japanese woman, that unfortunately already passed away. Her clinical phenotype was very similar to X-LAG, with significant weight gain starting a few months after birth and rapid growth diagnosed in the first years of life. She had marked GH elevation, prolactin elevation and no evidence of pituitary tumor in the MRI. She had no familial history of tall stature. Treatment with radiotherapy and bromocriptine did not ensure a complete biochemical response and the patient reached an adult height of 197.4 cm. Genome-edited mice with this mutation exhibited the same phenotype of prominent growth starting in the first weeks of life, pituitary hyperplasia and GHRH expression in several tissues besides the hypothalamus, validating the hypothesis that pituitary gigantism was driven by constitutive GHRH overexpression due to an acquired promoter.

 

CLINICAL MANAGEMENT IN FIPA

 

Pituitary adenoma patients with family members also with pituitary adenoma need to be studied for signs and symptoms of MEN1 and Carney complex (Figure 7). If MEN1 and Carney complex are ruled out by the family history and biochemical and clinical assessment of the index patient and family members, the diagnosis of FIPA needs to be considered. These patients would benefit from referral to genetic counselling. Currently, patients can be offered screening for AIP mutations. Childhood-onset pituitary adenoma cases, even without family history, should also been offered genetic counselling and screening for AIP mutation, as a high percentage of young-onset GH-secreting adenomas show mutations in the AIP gene (20, 60, 140, 141). Around 12% of patients diagnosed with a pituitary tumor before the age of 30 years (and 20% of pediatric patients) were found to have a germline AIP mutation in one study (142) and so it has been recommended that AIP mutation screening be conducted in anyone diagnosed with a somatotroph or lactotroph adenoma or a macroadenoma (diameter >10mm) before the age of 30 years (143), and also in any cases of gigantism. One study which examined the incidence in apparently sporadic young-onset pituitary adenoma patients found 6.8% to have an AIP mutation, with a slightly lower incidence of 10.5% in those sporadic patients with somatotrophinomas. Reassuringly, the incidence of mutation in sporadic prolactinoma was only 1.5% (12).

 

Those diagnosed with a pituitary tumor after the age of 40 years are unlikely to have a germline mutation (none were found in a sample of 443 patient with pituitary adenomas of all histiotypes) (57) and so screening in this latter population is likely to be unrewarding.

The phenomenon of phenocopy needs to be kept in mind both in AIP mutation-positive and AIP mutation-negative families (16, 23).

 

Figure 7. Proposed strategy for evaluating the patient with pituitary adenoma with (A) – negative family history and (B) – positive family history (*rare case report).

It is suggested that family members of an AIP mutation-positive proband should undergo genetic testing (Figure 8 suggests a strategy for this process), though this testing may involve significant numbers of people from the affected family and is probably best carried out in genetic centers that are able to arrange testing and counselling of many people, have experience of discussing results of screening, and can maintain family registers (22). Salivary DNA testing is available for those that are needle-phobic.

 

Figure 8. A proposed strategy for family screening in a family with an AIP mutation-positive proband. *Family member are first degree relatives of those with AIP mutations, or of obligate carriers. Further screening targets are then identified through genetic testing.

 

AIP mutation carriers should be referred to an endocrine service (pediatric or adult) for baseline assessment (clinical examination, biochemical testing, and MRI) (141). MRI can be delayed for young children if clinical and biochemical results are normal (143).  Children aged 4 years and older should be evaluated annually, with height and weight measurements, height velocity, and pituitary function testing (143). The frequency of imaging surveillance if biochemical and clinical findings are normal is difficult to judge with the available data: every 5 years was suggested until the age of 30 (143), with annual clinical assessment and basal hormone profiling (19). More recently, the emergence of an inverted-U shape pattern to the age of onset has led to the suggestion that if there is no evidence of disease at the age of 20 years, then surveillance protocols can be relaxed slightly (12).

 

The youngest case identified of AIP mutation-positive patient with a large macroadenoma with apoplexy was 4 years old with significant symptoms and rapid growth velocity already from age 3 years (43). Although only 15% of the AIP cases present symptoms before the age of 10 years (19), and the above mentioned patient is the single case known presenting before the age of 5 years, these data need to be taken into account when counselling AIP mutation-positive families for the timing of genetic screening and starting clinical follow-up (141, 143).

 

If AIP screening, which includes exons, exon-intron junction and promoter area sequencing as well as MLPA is negative, then currently no further genetic screening is possible. In AIP mutation-negative family’s potential carriers with a 50% chance inheriting the disease-causing mutation should be offered clinical assessment. The age of first clinical assessment of family members in AIP-negative families should be around early teenage years as the current youngest case was found at the age of 12 years (143).

 

We have already prospectively diagnosed several pituitary adenomas (both functioning macroadenomas and non-functioning microadenomas) in our cohort in both AIP-positive and AIP mutation-negative families (12, 60).  Screening allows the early detection and treatment of those with adenomas, perhaps before the endocrine effects become apparent or before the local effects of tumor bulk are problematic. It is important to draw the attention of the family to the possible symptoms of pituitary disease, as awareness of symptoms results in earlier diagnosis of the disease in subsequent generations (1, 11). Data on long-term follow-up of asymptomatic carriers is currently being collected. In our clinic, we see asymptomatic young (<30 years old) carriers once a year and after a normal baseline MRI we will consider a repeat MRI in 5 years. We consider relaxing follow-up at 30 years and stopping follow-up at 50 years for AIP mutation-positive family members if no tumor has been detected by this time.

 

The relatively high frequency of pituitary incidentalomas in the general population (144) also needs to be carefully considered both in AIP positive and negative cases. One paper (22) has suggested repeating an MRI pituitary and hormone testing at 6 months after the discovery of a pituitary incidentaloma in AIP mutation-positive individual with normal biochemistry, with annual hormone testing thereafter if the MRI was unchanged.

 

Those with apparently cured AIP mutation-positive tumors (but without external beam radiotherapy) should be followed up carefully as any residual pituitary tissue will be heterozygous for the AIP mutation and so there is a risk of the occurrence of further pituitary adenomas (22).

 

SUMMARY

 

FIPA is a condition where there is an inherited propensity to the development of pituitary adenomas. The causative gene for the vast majority (76%) of kindreds is unknown: 21% of these have a mutation in the AIP gene, 3% have a duplication on the X chromosome (X-linked acrogigantism, XLAG).

 

There are significant phenotypic differences between these groups, with XLAG presenting with infant-onset gigantism (range 0.5-2 years) most often with prolactin co-secretion, AIP cases presenting with childhood-onset GH or prolactin-secreting tumors, while the spectrum of AIP-negative FIPA kindred represent the full spectrum of pituitary adenoma subtypes with age of onset between the ages of 20 and 50 years with a peak incidence around the age of 30 years.

 

FIPA patients are more likely to have larger (macroadenomas), more aggressive tumors, and an earlier onset of disease compared to sporadic pituitary adenomas. AIP mutation-positive tumors are more likely to be larger and invade the extrasellar region than sporadic adenomas. It has also been observed that the AIP mutated adenomas are more prone to undergoing apoplexy than AIP mutation-negative adenomas. All XLAG tumors described so far are GH producing, with a majority also secreting prolactin. XLAG can result in a spectrum of pituitary gland appearances, ranging from large adenomas to pituitary hyperplasia. The tumors tend not to invade or undergo apoplexy.

 

AIP mutated adenomas are more difficult to treat than their non-mutated counterparts, they are more likely to be resistant to somatostatin analogue therapy, more likely to require radiotherapy, and have higher rates of failure to gain control of IGF-1 with pegvisomant treatment.

 

Treatment of XLAG is also challenging. Tumors can grow rapidly and are difficult to control even with high doses of somatostatin analogue or dopamine agonists. Pegvisomant is effective in normalizing IGF-1, while tumor control may need radical surgery or radiotherapy.

 

FIPA Diagnosis and Screening

 

The first step in trying to establish a diagnosis in patients with pituitary adenomas and with a family history of pituitary adenoma should be to exclude MEN1 and Carney complex. This can be achieved through the taking of a thorough family history and through the clinical and biochemical assessment of the index patient, and if possible other affected family members. If these conditions are excluded then the diagnosis of FIPA should be considered, and these patients should be referred for genetic counselling. Additionally, any childhood onset pituitary adenoma case (irrespective of family history), any somatotroph or lactotroph adenoma, or any macroadenoma diagnosed before the age of 30 and any cases of gigantism should all be referred for genetic counselling. No cases of AIP germline mutation were found in a large study of patients diagnosed with a pituitary tumor after the age of 40 years – and for this reason, genetic screening in this population is unlikely to be rewarding.

 

AIP mutation carriers should be referred to an endocrine service (pediatric or adult) for baseline assessment (clinical examination, biochemical testing, and MRI). MRI can be delayed for young children if clinical and biochemical results are normal. Children aged 4 years and older should be evaluated annually, with height and weight measurements, height velocity, and pituitary function testing. If biochemical and clinical findings are normal then 5-yearly MRIs until the age of 30, with annual clinical assessment and basal hormone profiling, is the suggested follow-up protocol.

 

For AIP positive families we suggest starting genetic screening as soon as the family agrees as the youngest case identified was at the age of 4 years with 1-year history of symptoms, presenting with a large macroadenoma.

 

If AIP screening, which includes exons, exon-intron junction and promoter area sequencing as well as multiple ligation probe amplification (MLPA), is negative, then currently no further genetic screening is possible. In AIP mutation-negative families, potential carriers with a 50% chance of inheriting the disease-causing mutation should be offered clinical assessment. The age of first clinical assessment of family members in AIP negative families should be around early teenage years as the current youngest case was found at the age of 12 years.

 

Prospectively-diagnosed pituitary adenomas have been shown to have a better outcome. Screening allows the early detection and treatment of those with adenomas, perhaps before the endocrine effects become apparent or before the local effects of tumor bulk become problematic. It is important to draw the attention of the family to the possible symptoms of pituitary disease, as awareness of symptoms results in earlier diagnosis of the disease in subsequent generations. In unaffected AIP mutation carriers, follow-up can be relaxed at the age of 30 years if no tumor has been detected by this time, and follow-up can cease at 50 years, based on the available data. The relatively high frequency of pituitary incidentalomas in the general population also needs to be carefully considered both in AIP positive and negative family members. One strategy involves repeating an MRI pituitary and hormone testing at 6-12 months after the discovery of a pituitary incidentaloma in AIP mutation-positive individuals with normal biochemistry, with annual hormone testing thereafter if the MRI is unchanged.

 

ACKNOWLEDGEMENT

 

We are grateful for Dr Craig Stiles (Barts Health NHS Trust, London), who contributed to the previous version of this Endotext chapter.

 

REFERENCES

 

  1. Chahal HS, Chapple JP, Frohman LA, Grossman AB, Korbonits M. Clinical, genetic and molecular characterization of patients with familial isolated pituitary adenomas (FIPA). Trends Endocrinol Metab. 2010;21:419-27.
  2. Verloes A, Stevenaert A, Teh BT, Petrossians P, Beckers A. Familial acromegaly: case report and review of the literature. Pituitary. 1999;1(3-4):273-7.
  3. Xekouki P, Pacak K, Almeida M, Wassif CA, Rustin P, Nesterova M, et al. Succinate dehydrogenase (SDH) D subunit (SDHD) inactivation in a growth-hormone-producing pituitary tumor: a new association for SDH? J Clin Endocrinol Metab. 2012;97(3):E357-66.
  4. Dénes J, Swords F, Rattenberry E, Stals K, Owens M, Cranston T, et al. Heterogeneous genetic background of the association of pheochromocytoma/paraganglioma and pituitary adenoma - results from a large patient cohort. J Clin Endocrinol Metab. 2015;100(3):E531-E41.
  5. O'Toole SM, Denes J, Robledo M, Stratakis CA, Korbonits M. 15 YEARS OF PARAGANGLIOMA: The association of pituitary adenomas and phaeochromocytomas or paragangliomas. Endocr Relat Cancer. 2015;22(4):T105-22.
  6. de Kock L, Sabbaghian N, Plourde F, Srivastava A, Weber E, Bouron-Dal SD, et al. Pituitary blastoma: a pathognomonic feature of germ-line DICER1 mutations. Acta Neuropathol. 2014;128:111-22.
  7. Uraki S, Ariyasu H, Doi A, Furuta H, Nishi M, Sugano K, et al. Atypical pituitary adenoma with MEN1 somatic mutation associated with abnormalities of DNA mismatch repair genes; MLH1 germline mutation and MSH6 somatic mutation. Endocr J. 2017;64(9):895-906.
  8. Voisin MR, Almeida JP, Perez-Ordonez B, Zadeh G. Recurrent Undifferentiated Carcinoma of the Sella in a Patient with Lynch Syndrome. World Neurosurg. 2019;132:219-22.
  9. Bengtsson D, Joost P, Aravidis C, Askmalm Stenmark M, Backman AS, Melin B, et al. Corticotroph Pituitary Carcinoma in a Patient With Lynch Syndrome (LS) and Pituitary Tumors in a Nationwide LS Cohort. J Clin Endocrinol Metab. 2017;102(11):3928-32.
  10. Nachtigall LB, Guarda FJ, Lines KE, Ghajar A, Dichtel L, Mumbach G, et al. Clinical MEN-1 Among a Large Cohort of Patients With Acromegaly. J Clin Endocrinol Metab. 2020;105(6).
  11. Daly AF, Vanbellinghen JF, Khoo SK, Jaffrain-Rea ML, Naves LA, Guitelman MA, et al. Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. J Clin Endocrinol Metab. 2007;92(5):1891-6.
  12. Marques P, Caimari F, Hernandez-Ramirez LC, Collier D, Iacovazzo D, Ronaldson A, et al. Significant Benefits of AIP Testing and Clinical Screening in Familial Isolated and Young-onset Pituitary Tumors. J Clin Endocrinol Metab. 2020;105(6).
  13. Trivellin G, Daly AF, Faucz FR, Yuan B, Rostomyan L, Larco DO, et al. Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. N Engl J Med. 2014;371:2363-74.
  14. Rodd C, Millette M, Iacovazzo D, Stiles CE, Barry S, Evanson J, et al. Somatic GPR101 duplication causing X-linked acrogigantism (XLAG)-diagnosis and management. J Clin Endocrinol Metab. 2016;101(5):1927-30.
  15. Daly AF, Yuan B, Fina F, Caberg JH, Trivellin G, Rostomyan L, et al. Somatic mosaicism underlies X-linked acrogigantism syndrome in sporadic male subjects. Endocr Relat Cancer. 2016;23(4):221-33.
  16. Vierimaa O, Georgitsi M, Lehtonen R, Vahteristo P, Kokko A, Raitila A, et al. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science. 2006;312(5777):1228-30.
  17. Katoh-Fukui Y, Hattori A, Zhang R, Terao M, Takada S, Nakabayashi K, et al. Chromosomal microdeletion leading to pituitary gigantism through hormone-gene overexpression. Hum Mol Genet. 2023;32(14):2318-25.
  18. Trivellin G, Daly AF, Hernández-Ramírez LC, Araldi E, Tatsi C, Dale RK, et al. Germline loss-of-function PAM variants are enriched in subjects with pituitary hypersecretion. medRxiv. 2023.
  19. Hernandez-Ramirez LC, Gabrovska P, Denes J, Stals K, Trivellin G, Tilley D, et al. Landscape of Familial Isolated and Young-Onset Pituitary Adenomas: Prospective Diagnosis in AIP Mutation Carriers. J Clin Endocrinol Metab. 2015;100(9):E1242-54.
  20. Daly AF, Tichomirowa MA, Petrossians P, Heliovaara E, Jaffrain-Rea ML, Barlier A, et al. Clinical characteristics and therapeutic responses in patients with germ-line AIP mutations and pituitary adenomas: an international collaborative study. J Clin Endocrinol Metab. 2010;95(11):E373-83.
  21. Drummond J, Roncaroli F, Grossman AB, Korbonits M. Clinical and Pathological Aspects of Silent Pituitary Adenomas. J Clin Endocrinol Metab. 2019;104(7):2473-89.
  22. Williams F, Hunter S, Bradley L, Chahal HS, Storr H, Akker SA, et al. Clinical experience in the screening and management of a large kindred with familial isolated pituitary adenoma due to an aryl hydrocarbon receptor interacting protein (AIP) mutation. J Clin Endocrinol Metab. 2014;99(4):1122-31.
  23. Igreja S, Chahal HS, King P, Bolger GB, Srirangalingam U, Guasti L, et al. Characterization of aryl hydrocarbon receptor interacting protein (AIP) mutations in familial isolated pituitary adenoma families. Hum Mutat. 2010;31(8):950-60.
  24. Daly AF, Jaffrain-Rea ML, Ciccarelli A, Valdes-Socin H, Rohmer V, Tamburrano G, et al. Clinical characterization of familial isolated pituitary adenomas. J Clin Endocrinol Metab. 2006;91(9):3316-23.
  25. Stratakis CA, Tichomirowa MA, Boikos S, Azevedo MF, Lodish M, Martari M, et al. The role of germline AIP, MEN1, PRKAR1A, CDKN1B and CDKN2C mutations in causing pituitary adenomas in a large cohort of children, adolescents, and patients with genetic syndromes. Clin Genet. 2010;78(5):457-63.
  26. Ramirez-Renteria C, Hernandez-Ramirez LC, Portocarrero-Ortiz L, Vargas G, Melgar V, Espinosa E, et al. AIP mutations in young patients with acromegaly and the Tampico Giant: the Mexican experience. Endocrine. 2016;53(2):402-11.
  27. Turner JJ, Christie PT, Pearce SH, Turnpenny PD, Thakker RV. Diagnostic challenges due to phenocopies: lessons from Multiple Endocrine Neoplasia type1 (MEN1). Hum Mutat. 2010;31(1):E1089-E101.
  28. Leontiou CA, Gueorguiev M, van der Spuy J, Quinton R, Lolli F, Hassan S, et al. The role of the aryl hydrocarbon receptor-interacting protein gene in familial and sporadic pituitary adenomas. J Clin Endocrinol Metab. 2008;93(6):2390-401.
  29. Wakai S, Fukushima T, Teramoto A, Sano K. Pituitary apoplexy: its incidence and clinical significance. J Neurosurg. 1981;55(2):187-93.
  30. Mohr G, Hardy J. Hemorrhage, necrosis, and apoplexy in pituitary adenomas. Surg Neurol. 1982;18(3):181-9.
  31. Xekouki P, Mastroyiannis SA, Avgeropoulos D, de la Luz Sierra M, Trivellin G, Gourgari EA, et al. Familial pituitary apoplexy as the only presentation of a novel AIP mutation. Endocr Relat Cancer. 2013;20(5):L11-4.
  32. Bhayana S, Booth GL, Asa SL, Kovacs K, Ezzat S. The implication of somatotroph adenoma phenotype to somatostatin analog responsiveness in acromegaly. J Clin Endocrinol Metab. 2005;90(11):6290-5.
  33. Stefaneanu L, Kovacs K, Thapar K, Horvath E, Melmed S, Greenman Y. Octreotide effect on growth hormone and somatostatin subtype 2 receptor mRNAs of the human pituitary somatotroph adenomas. Endocr Pathol. 2000;11(1):41-8.
  34. Theodoropoulou M, Zhang J, Laupheimer S, Paez-Pereda M, Erneux C, Florio T, et al. Octreotide, a somatostatin analogue, mediates its antiproliferative action in pituitary tumor cells by altering phosphatidylinositol 3-kinase signaling and inducing Zac1 expression. Cancer Res. 2006;66(3):1576-82.
  35. Theodoropoulou M, Tichomirowa MA, Sievers C, Yassouridis A, Arzberger T, Hougrand O, et al. Tumor ZAC1 expression is associated with the response to somatostatin analog therapy in patients with acromegaly. Int J Cancer. 2009;125(9):2122-6.
  36. Chahal HS, Trivellin G, Leontiou CA, Alband N, Fowkes RC, Tahir A, et al. Somatostatin analogs modulate AIP in somatotroph adenomas: the role of the ZAC1 pathway. J Clin Endocrinol Metab. 2012;97(8):E1411-20.
  37. Korbonits M, Blair JC, Boguslawska A, Ayuk J, Davies JH, Druce MR, et al. Consensus guideline for the diagnosis and management of pituitary adenomas in childhood and adolescence: Part 2, specific diseases. Nat Rev Endocrinol. 2024.
  38. Iacovazzo D, Carlsen E, Lugli F, Chiloiro S, Piacentini S, Bianchi A, et al. Factors predicting pasireotide responsiveness in somatotroph pituitary adenomas resistant to first-generation somatostatin analogues: an immunohistochemical study. Eur J Endocrinol. 2016;174(2):241-50.
  39. Solomou A, Herincs M, Roncaroli F, Vignola ML, Gaston-Massuet C, Korbonits M, editors. Investigating the role of AIP in mouse pituitary adenoma formation. Endocrine Abstracts; 2017; Harrogate BES2017.
  40. Barry S, Carlsen E, Marques P, Stiles CE, Gadaleta E, Berney DM, et al. Tumor microenvironment defines the invasive phenotype of AIP-mutation-positive pituitary tumors. Oncogene. 2019;38(27):5381-95.
  41. Joshi K, Daly AF, Beckers A, Zacharin M. Resistant Paediatric Somatotropinomas due to AIP Mutations: Role of Pegvisomant. Horm Res Paediatr. 2018;90(3):196-202.
  42. Daly AF, Rostomyan L, Betea D, Bonneville JF, Villa C, Pellegata NS, et al. AIP-mutated acromegaly resistant to first-generation somatostatin analogs: long-term control with pasireotide LAR in two patients. Endocr Connect. 2019;8(4):367-77.
  43. Dutta P, Reddy KS, Rai A, Madugundu AK, Solanki HS, Bhansali A, et al. Surgery, octreotide, temozolomide, bevacizumab, radiotherapy, and pegvisomant treatment of an AIP mutation positive child. J Clin Endocrinol Metab. 2019;104(8):3539-44.
  44. Lim DST, Fleseriu M. Personalized Medical Treatment of Patients With Acromegaly: A Review. Endocr Pract. 2022;28(3):321-32.
  45. Wildemberg LE, da Silva Camacho AH, Miranda RL, Elias PCL, de Castro Musolino NR, Nazato D, et al. Machine Learning-based Prediction Model for Treatment of Acromegaly With First-generation Somatostatin Receptor Ligands. J Clin Endocrinol Metab. 2021;106(7):2047-56.
  46. Villa C, Lagonigro MS, Magri F, Koziak M, Jaffrain-Rea ML, Brauner R, et al. Hyperplasia-adenoma sequence in pituitary tumorigenesis related to aryl hydrocarbon receptor interacting protein (AIP) gene mutation. Endocr Relat Cancer. 2011;18(3):347-56.
  47. Jaffrain-Rea ML, Rotondi S, Turchi A, Occhi G, Barlier A, Peverelli E, et al. Somatostatin analogues increase AIP expression in somatotropinomas, irrespective of Gsp mutations. Endocr Relat Cancer. 2013;20(5):753-66.
  48. Pardi E, Marcocci C, Borsari S, Saponaro F, Torregrossa L, Tancredi M, et al. Aryl hydrocarbon receptor interacting protein (AIP) mutations occur rarely in sporadic parathyroid adenomas. J Clin Endocrinol Metab. 2013;98(7):2800-10.
  49. Solís-Fernández G, Montero-Calle A, Sánchez-Martínez M, Peláez-García A, Fernández-Aceñero MJ, Pallarés P, et al. Aryl-hydrocarbon receptor-interacting protein regulates tumorigenic and metastatic properties of colorectal cancer cells driving liver metastasis. Br J Cancer. 2022;126(11):1604-15.
  50. Sun D, Stopka-Farooqui U, Barry S, Aksoy E, Parsonage G, Vossenkämper A, et al. Aryl Hydrocarbon Receptor Interacting Protein Maintains Germinal Center B Cells through Suppression of BCL6 Degradation. Cell Rep. 2019;27(5):1461-71.e4.
  51. Zhu H, Zhao H, Wang J, Zhao S, Ma C, Wang D, et al. Potential prognosis index for m(6)A-related mRNA in cholangiocarcinoma. BMC Cancer. 2022;22(1):620.
  52. Haworth O, Korbonits M. AIP: A double agent? The tissue-specific role of AIP as a tumour suppressor or as an oncogene.Br J Cancer. 127. England: © 2022. The Author(s), under exclusive licence to Springer Nature Limited.; 2022. p. 1175-6.
  53. Loughrey PB, Korbonits M. Genetics of Pituitary Tumours. In: Igaz P, Patocs A, editors. Genetics of Endocrine Diseases and Syndromes. Exp Suppl. 111. 2019/10/08 ed2019. p. 171-211.
  54. Morgan RM, Hernández-Ramírez LC, Trivellin G, Zhou L, Roe SM, Korbonits M, et al. Structure of the TPR domain of AIP: lack of client protein interaction with the C-terminal alpha-7 helix of the TPR domain of AIP is sufficient for pituitary adenoma predisposition. PLoS One. 2012;7(12):e53339.
  55. Linnert M, Haupt K, Lin YJ, Kissing S, Paschke AK, Fischer G, et al. NMR assignments of the FKBP-type PPIase domain of the human aryl-hydrocarbon receptor-interacting protein (AIP). Biomol NMR Assign. 2012;6(2):209-12.
  56. Trivellin G, Korbonits M. AIP and its interacting partners. J Endocrinol. 2011;210(2):137-55.
  57. Cazabat L, Bouligand J, Salenave S, Bernier M, Gaillard S, Parker F, et al. Germline AIP mutations in apparently sporadic pituitary adenomas: prevalence in a prospective single-center cohort of 443 patients. J Clin Endocrinol Metab. 2012;97(4):E663-E70.
  58. Georgitsi M, Raitila A, Karhu A, Tuppurainen K, Makinen MJ, Vierimaa O, et al. Molecular diagnosis of pituitary adenoma predisposition caused by aryl hydrocarbon receptor-interacting protein gene mutations. Proc Natl Acad Sci USA. 2007;104(10):4101-5.
  59. Vargiolu M, Fusco D, Kurelac I, Dirnberger D, Baumeister R, Morra I, et al. The tyrosine kinase receptor RET interacts in vivo with aryl hydrocarbon receptor-interacting protein to alter survivin availability. J Clin Endocrinol Metab. 2009;94(7):2571-8.
  60. Chahal HS, Stals K, Unterlander M, Balding DJ, Thomas MG, Kumar AV, et al. AIP mutation in pituitary adenomas in the 18th century and today. N Engl J Med. 2011;364(1):43-50.
  61. Jennings JE, Georgitsi M, Holdaway I, Daly A, Tichomirowa M, Beckers A, et al. Aggressive pituitary adenomas occurring in young patients in a large Polynesian kindred with a germline R271W mutation in the AIP gene. Eur J Endocrinol. 2009;161(5):799-804.
  62. Toledo RA, Mendonca BB, Fragoso MC, Soares IC, Almeida MQ, Moraes MB, et al. Isolated familial somatotropinoma: 11q13-loh and gene/protein expression analysis suggests a possible involvement of AIP also in non-pituitary tumorigenesis. Clinics (Sao Paulo). 2010;65(4):407-15.
  63. Lin BC, Sullivan R, Lee Y, Moran S, Glover E, Bradfield CA. Deletion of the aryl hydrocarbon receptor-associated protein 9 leads to cardiac malformation and embryonic lethality. J Biol Chem. 2007;282(49):35924-32.
  64. Kang BH, Xia F, Pop R, Dohi T, Socolovsky M, Altieri DC. Developmental control of apoptosis by the immunophilin aryl hydrocarbon receptor-interacting protein (AIP) involves mitochondrial import of the survivin protein. J Biol Chem. 2011;286(19):16758-67.
  65. Raitila A, Lehtonen HJ, Arola J, Heliovaara E, Ahlsten M, Georgitsi M, et al. Mice with inactivation of aryl hydrocarbon receptor-interacting protein (Aip) display complete penetrance of pituitary adenomas with aberrant ARNT expression. Am J Pathol. 2010;177(4):1969-76.
  66. Heliovaara E, Raitila A, Launonen V, Paetau A, Arola J, Lehtonen H, et al. The expression of AIP-related molecules in elucidation of cellular pathways in pituitary adenomas. Am J Pathol. 2009;175(6):2501-7.
  67. Gillam MP, Ku CR, Lee YJ, Kim J, Kim SH, Lee SJ, et al. Somatotroph-Specific Aip-Deficient Mice Display Pretumorigenic Alterations in Cell-Cycle Signaling. J Endocr Soc. 2017;1(2):78-95.
  68. Maltepe E, Schmidt JV, Baunoch D, Bradfield CA, Simon MC. Abnormal angiogenesis and responses to glucose and oxygen deprivation in mice lacking the protein ARNT. Nature. 1997;386(6623):403-7.
  69. Kozak KR, Abbott B, Hankinson O. ARNT-deficient mice and placental differentiation. Dev Biol. 1997;191(2):297-305.
  70. Vasquez A, Atallah-Yunes N, Smith FC, You X, Chase SE, Silverstone AE, et al. A role for the aryl hydrocarbon receptor in cardiac physiology and function as demonstrated by AhR knockout mice. Cardiovasc Toxicol. 2003;3(2):153-63.
  71. Lund AK, Goens MB, Kanagy NL, Walker MK. Cardiac hypertrophy in aryl hydrocarbon receptor null mice is correlated with elevated angiotensin II, endothelin-1, and mean arterial blood pressure. Toxicol Appl Pharmacol. 2003;193(2):177-87.
  72. Lahvis GP, Lindell SL, Thomas RS, McCuskey RS, Murphy C, Glover E, et al. Portosystemic shunting and persistent fetal vascular structures in aryl hydrocarbon receptor-deficient mice. Proc Natl Acad Sci USA. 2000;97(19):10442-7.
  73. Tappenden DM, Hwang HJ, Yang L, Thomas RS, Lapres JJ. The Aryl-Hydrocarbon Receptor Protein Interaction Network (AHR-PIN) as Identified by Tandem Affinity Purification (TAP) and Mass Spectrometry. J Toxicol. 2013;2013:279829.
  74. Hernandez-Ramirez LC, Martucci F, Morgan RM, Trivellin G, Tilley D, Ramos-Guajardo N, et al. Rapid Proteasomal Degradation of Mutant Proteins Is the Primary Mechanism Leading to Tumorigenesis in Patients With Missense AIP Mutations. J Clin Endocrinol Metab. 2016;101(8):3144-54.
  75. Hernandez-Ramirez LC, Morgan RML, Barry S, D'Acquisto F, Prodromou C, Korbonits M. Multi-chaperone function modulation and association with cytoskeletal proteins are key features of the function of AIP in the pituitary gland. Oncotarget. 2018;9(10):9177-98.
  76. Schernthaner-Reiter MH, Trivellin G, Stratakis CA. Interaction of AIP with protein kinase A (cAMP-dependent protein kinase). Hum Mol Genet. 2018.
  77. Hillegass JM, Murphy KA, Villano CM, White LA. The impact of aryl hydrocarbon receptor signaling on matrix metabolism: implications for development and disease. Biol Chem. 2006;387(9):1159-73.
  78. de Oliveira SK, Smolenski A. Phosphodiesterases link the aryl hydrocarbon receptor complex to cyclic nucleotide signaling. Biochem Pharmacol. 2008.
  79. Kang BH, Altieri DC. Regulation of survivin stability by the aryl hydrocarbon receptor-interacting protein. J Biol Chem. 2006;281(34):24721-7.
  80. Monteiro P, Gilot D, Le FE, Rauch C, Lagadic-Gossmann D, Fardel O. Dioxin-mediated up-regulation of aryl hydrocarbon receptor target genes is dependent on the calcium/calmodulin/CaMKIalpha pathway. Mol Pharmacol. 2008;73(3):769-77.
  81. Barouki R, Coumoul X, Fernandez-Salguero PM. The aryl hydrocarbon receptor, more than a xenobiotic-interacting protein. FEBS Lett. 2007;581(19):3608-15.
  82. Cannavo S, Ferraù F, Ragonese M, Curto L, Torre ML, Magistri M, et al. Increased prevalence of acromegaly in a highly polluted area. Eur J Endocrinol. 2010;163(4):509-13.
  83. Pesatori AC, Baccarelli A, Consonni D, Lania A, Beck-Peccoz P, Bertazzi P, et al. Aryl hydrocarbon receptor interacting protein and pituitary adenomas: a population-based study on subjects exposed to dioxin after the Seveso, Italy, accident. Eur J Endocrinol. 2008;159(6):699-703.
  84. Cannavo S, Ragonese M, Puglisi S, Romeo PD, Torre ML, Alibrandi A, et al. Acromegaly Is More Severe in Patients With AHR or AIP Gene Variants Living in Highly Polluted Areas. J Clin Endocrinol Metab. 2016;101(4):1872-9.
  85. Lecoq AL, Viengchareun S, Hage M, Bouligand J, Young J, Boutron A, et al. AIP mutations impair AhR signaling in pituitary adenoma patients fibroblasts and in GH3 cells. Endocr Relat Cancer. 2016;23(5):433-43.
  86. Cai W, Kramarova TV, Berg P, Korbonits M, Pongratz I. The immunophilin-like protein XAP2 is a negative regulator of estrogen signaling through interaction with estrogen receptor alpha. PLoS One. 2011;6(10):e25201.
  87. Heaney AP, Horwitz GA, Wang Z, Singson R, Melmed S. Early involvement of estrogen-induced pituitary tumor transforming gene and fibroblast growth factor expression in prolactinoma pathogenesis. Nat Med. 1999;5(11):1317-21.
  88. Landis CA, Masters SB, Spada A, Pace AM, Bourne HR, Vallar L. GTPase inhibiting mutations activate the a chain of Gs and stimulate adenylyl cyclase in human pituitary tumors. Nature. 1989;340:692-6.
  89. Bertherat J, Chanson P, Montminy M. The cyclic adenosine 3',5'-monophosphate-responsive factor CREB is constitutively activated in human somatotroph adenomas. Mol Endocrinol. 1995;9(7):777-83.
  90. Vallar L, Spada A, Giannattasio G. Altered Gs and adenylate cyclase activity in human GH-secreting pituitary adenomas. Nature. 1987;330(6148):566-8.
  91. Formosa R, Xuereb-Anastasi A, Vassallo J. Aip regulates cAMP signalling and GH secretion in GH3 cells. Endocr Relat Cancer. 2013;20(4):495-505.
  92. Vasilev V, Daly AF, Thiry A, Petrossians P, Fina F, Rostomyan L, et al. McCune-Albright syndrome: a detailed pathological and genetic analysis of disease effects in an adult patient. J Clin Endocrinol Metab. 2014;99(10):E2029-E38.
  93. Tsang KM, Starost MF, Nesterova M, Boikos SA, Watkins T, Almeida MQ, et al. Alternate protein kinase A activity identifies a unique population of stromal cells in adult bone. Proc Natl Acad Sci USA. 2010;107(19):8683-8.
  94. Robinson-White A, Hundley TR, Shiferaw M, Bertherat J, Sandrini F, Stratakis CA. Protein kinase-A activity in PRKAR1A-mutant cells, and regulation of mitogen-activated protein kinases ERK1/2. Hum Mol Genet. 2003;12(13):1475-84.
  95. Nakata A, Urano D, Fujii-Kuriyama Y, Mizuno N, Tago K, Itoh H. G-protein signalling negatively regulates the stability of aryl hydrocarbon receptor. EMBO Rep. 2009;10(6):622-8.
  96. Tuominen I, Heliovaara E, Raitila A, Rautiainen MR, Mehine M, Katainen R, et al. AIP inactivation leads to pituitary tumorigenesis through defective Galphai-cAMP signaling. Oncogene. 2015;34(9):1174-84.
  97. Ritvonen E, Pitkanen E, Karppinen A, Vehkavaara S, Demir H, Paetau A, et al. Impact of AIP and inhibitory G protein alpha 2 proteins on clinical features of sporadic GH-secreting pituitary adenomas. Eur J Endocrinol. 2017;176(2):243-52.
  98. Liu YF, Jakobs KH, Rasenick MM, Albert PR. G protein specificity in receptor-effector coupling. Analysis of the roles of G0 and Gi2 in GH4C1 pituitary cells. J Biol Chem. 1994;269(19):13880-6.
  99. Garcia-Rendueles AR, Chenlo M, Oroz-Gonjar F, Solomou A, Mistry A, Barry S, et al. RET signalling provides tumorigenic mechanism and tissue specificity for AIP-related somatotrophinomas. Oncogene. 2021;40(45):6354-68.
  100. Caimari F, Hernández-Ramírez LC, Dang MN, Gabrovska P, Iacovazzo D, Stals K, et al. Risk category system to identify pituitary adenoma patients with AIP mutations. J Med Genet. 2018;55(4):254-60.
  101. Liu W, Matsumoto Y, Okada M, Miyake K, Kunishio K, Kawai N, et al. Matrix metalloproteinase 2 and 9 expression correlated with cavernous sinus invasion of pituitary adenomas. J Med Invest. 2005;52(3-4):151-8.
  102. Marques P, Korbonits M. Tumour microenvironment and pituitary tumour behaviour. J Endocrinol Invest. 2023;46(6):1047-63.
  103. Dénes J, Kasuki L, Trivellin G, Colli LM, Takiya CM, Stiles CE, et al. Regulation of aryl hydrocarbon receptor interacting protein (AIP) protein expression by MiR-34a in sporadic somatotropinomas. PLoS One. 2015;10(2):e0117107.
  104. Bogner EM, Daly AF, Gulde S, Karhu A, Irmler M, Beckers J, et al. miR-34a is upregulated in AIP-mutated somatotropinomas and promotes octreotide resistance. Int J Cancer. 2020;147(12):3523-38.
  105. Cai F, Chen S, Yu X, Zhang J, Liang W, Zhang Y, et al. Transcription factor GTF2B regulates AIP protein expression in growth hormone-secreting pituitary adenomas and influences tumor phenotypes. Neuro Oncol. 2022;24(6):925-35.
  106. Nord KH, Magnusson L, Isaksson M, Nilsson J, Lilljebjorn H, Domanski HA, et al. Concomitant deletions of tumor suppressor genes MEN1 and AIP are essential for the pathogenesis of the brown fat tumor hibernoma. Proc Natl Acad Sci U S A. 2010;107(49):21122-7.
  107. Magnusson L, Hansen N, Saba KH, Nilsson J, Fioretos T, Rissler P, et al. Loss of the tumour suppressor gene AIP mediates the browning of human brown fat tumours. J Pathol. 2017;243(2):160-4.
  108. Coopmans EC, Muhammad A, Daly AF, de Herder WW, van Kemenade FJ, Beckers A, et al. The role of AIP variants in pituitary adenomas and concomitant thyroid carcinomas in the Netherlands: a nationwide pathology registry (PALGA) study. Endocrine. 2020;68(3):640-9.
  109. Hernandez-Ramirez LC, Gam R, Valdes N, Lodish MB, Pankratz N, Balsalobre A, et al. Loss-of-function mutations in the CABLES1 gene are a novel cause of Cushing's disease. Endocr Relat Cancer. 2017;24(8):379-92.
  110. Gorvin CM, Newey PJ, Rogers A, Stokes V, Neville MJ, Lines KE, et al. Association of prolactin receptor (PRLR) variants with prolactinomas. Hum Mol Genet. 2019;28(6):1023-37.
  111. Zhang Q, Peng C, Song J, Zhang Y, Chen J, Song Z, et al. Germline Mutations in CDH23, Encoding Cadherin-Related 23, Are Associated with Both Familial and Sporadic Pituitary Adenomas. Am J Hum Genet. 2017;100(5):817-23.
  112. Melo FM, Couto PP, Bale AE, Bastos-Rodrigues L, Passos FM, Lisboa RG, et al. Whole-exome identifies RXRG and TH germline variants in familial isolated prolactinoma. Cancer Genet. 2016;209(6):251-7.
  113. Srirangam Nadhamuni V, Korbonits M. Novel Insights into Pituitary Tumorigenesis: Genetic and Epigenetic Mechanisms. Endocr Rev. 2020;41(6):821-46.
  114. Bernard DJ, Brûlé E, Smith CL, Joustra SD, Wit JM. From Consternation to Revelation: Discovery of a Role for IGSF1 in Pituitary Control of Thyroid Function. J Endocr Soc. 2018;2(3):220-31.
  115. Faucz FR, Horvath AD, Azevedo MF, Levy I, Bak B, Wang Y, et al. Is IGSF1 involved in human pituitary tumor formation? Endocr Relat Cancer. 2015;22(1):47-54.
  116. Sun Y, Bak B, Schoenmakers N, van Trotsenburg AS, Oostdijk W, Voshol P, et al. Loss-of-function mutations in IGSF1 cause an X-linked syndrome of central hypothyroidism and testicular enlargement. Nat Genet. 2012;44(12):1375-81.
  117. Joustra SD, Roelfsema F, van Trotsenburg ASP, Schneider HJ, Kosilek RP, Kroon HM, et al. IGSF1 Deficiency Results in Human and Murine Somatotrope Neurosecretory Hyperfunction. J Clin Endocrinol Metab. 2020;105(3):e70-84.
  118. Kardelen AD, Karakılıç Özturan E, Poyrazoğlu Ş, Baş F, Ceylaner S, Joustra SD, et al. A Novel Pathogenic IGSF1 Variant in a Patient with GH and TSH Deficiency Diagnosed by High IGF-I Values at Transition to Adult Care. J Clin Res Pediatr Endocrinol. 2023;15(4):431-7.
  119. Joustra SD, Heinen CA, Schoenmakers N, Bonomi M, Ballieux BE, Turgeon MO, et al. IGSF1 Deficiency: Lessons From an Extensive Case Series and Recommendations for Clinical Management. J Clin Endocrinol Metab. 2016;101(4):1627-36.
  120. Carmi D, Shohat M, Metzker A, Dickerman Z. Growth, puberty, and endocrine functions in patients with sporadic or familial neurofibromatosis type 1: a longitudinal study. Pediatrics. 1999;103(6 Pt 1):1257-62.
  121. Mautner VF, Kluwe L, Friedrich RE, Roehl AC, Bammert S, Högel J, et al. Clinical characterisation of 29 neurofibromatosis type-1 patients with molecularly ascertained 1.4 Mb type-1 NF1 deletions. J Med Genet. 2010;47(9):623-30.
  122. Cambiaso P, Galassi S, Palmiero M, Mastronuzzi A, Del Bufalo F, Capolino R, et al. Growth hormone excess in children with neurofibromatosis type-1 and optic glioma. Am J Med Genet A. 2017;173(9):2353-8.
  123. Hannah-Shmouni F, Trivellin G, Beckers P, Karaviti LP, Lodish M, Tatsi C, et al. Neurofibromatosis Type 1 Has a Wide Spectrum of Growth Hormone Excess. J Clin Med. 2022;11(8).
  124. Glasker S, Vortmeyer AO, Lafferty AR, Hofman PL, Li J, Weil RJ, et al. Hereditary pituitary hyperplasia with infantile gigantism. J Clin Endocrinol Metab. 2011;96(12):E2078-E87.
  125. Beckers A, Lodish MB, Trivellin G, Rostomyan L, Lee M, Faucz FR, et al. X-linked acrogigantism syndrome: clinical profile and therapeutic responses. Endocr Relat Cancer. 2015;22(3):353-67.
  126. Wise-Oringer BK, Zanazzi GJ, Gordon RJ, Wardlaw SL, William C, Anyane-Yeboa K, et al. Familial X-Linked Acrogigantism: Postnatal Outcomes and Tumor Pathology in a Prenatally Diagnosed Infant and His Mother. J Clin Endocrinol Metab. 2019;104(10):4667-75.
  127. Iacovazzo D, Caswell R, Bunce B, Jose S, Yuan B, Hernandez-Ramirez LC, et al. Germline or somatic GPR101 duplication leads to X-linked acrogigantism: a clinico-pathological and genetic study. Acta Neuropathol Commun. 2016;4(1):56.
  128. Burren CP, Williams G, Coxson E, Korbonits M. Effective Long-term Pediatric Pegvisomant Monotherapy to Final Height in X-linked Acrogigantism. JCEM Case Reports. 2023;1(3).
  129. Lecoq AL, Bouligand J, Hage M, Cazabat L, Salenave S, Linglart A, et al. Very low frequency of germline GPR101 genetic variation and no biallelic defects with AIP in a large cohort of patients with sporadic pituitary adenomas. Eur J Endocrinol. 2016;174(4):523-30.
  130. Ferrau F, Romeo PD, Puglisi S, Ragonese M, Torre ML, Scaroni C, et al. Analysis of GPR101 and AIP genes mutations in acromegaly: a multicentric study. Endocrine. 2016;54(3):762-7.
  131. Trivellin G, Bjelobaba I, Daly AF, Larco DO, Palmeira L, Faucz FR, et al. Characterization of GPR101 transcript structure and expression patterns. J Mol Endocrinol. 2016;57(2):97-111.
  132. Iacovazzo D, Korbonits M. Gigantism: X-linked acrogigantism and GPR101 mutations.Growth Horm IGF Res2016.
  133. Naves LA, Daly AF, Dias LA, Yuan B, Zakir JC, Barra GB, et al. Aggressive tumor growth and clinical evolution in a patient with X-linked acro-gigantism syndrome. Endocrine. 2016;51(2):236-44.
  134. Moran A, Asa SL, Kovacs K, Horvath E, Singer W, Sagman U, et al. Gigantism due to pituitary mammosomatotroph hyperplasia. N Engl J Med. 1990;323(5):322-7.
  135. Coxson E, Iacovazzo D, Bunce B, Jose S, Ellard S, Sampson J, et al., editors. Pegvisomant treatment for X-linked acrogigantism syndrome. Endocrine Abstracts; 2015.
  136. Daly AF, Lysy PA, Desfilles C, Rostomyan L, Mohamed A, Caberg JH, et al. GHRH excess and blockade in X-LAG syndrome. Endocr Relat Cancer. 2016;23(3):161-70.
  137. Trivellin G, Faucz FR, Daly AF, Beckers A, Stratakis CA. GPR101, an orphan GPCR with roles in growth and pituitary tumorigenesis. Endocr Relat Cancer. 2020.
  138. Trivellin G, Hernandez-Ramirez LC, Swan J, Stratakis CA. An orphan G-protein-coupled receptor causes human gigantism and/or acromegaly: Molecular biology and clinical correlations. Best Pract Res Clin Endocrinol Metab. 2018;32(2):125-40.
  139. Franke M, Daly AF, Palmeira L, Tirosh A, Stigliano A, Trifan E, et al. Duplications disrupt chromatin architecture and rewire GPR101-enhancer communication in X-linked acrogigantism. Am J Hum Genet. 2022;109(4):553-70.
  140. Cazabat L, Bouligand J, Chanson P. AIP mutation in pituitary adenomas. N Engl J Med. 2011;364(20):1973-4.
  141. Korbonits M, Blair JC, Boguslawska A, Ayuk J, Davies JH, Druce MR, et al. Consensus guideline for the diagnosis and management of pituitary adenomas in childhood and adolescence: Part 1, general recommendations. Nat Rev Endocrinol. 2024.
  142. Tichomirowa MA, Barlier A, Daly AF, Jaffrain-Rea ML, Ronchi CL, Yaneva M, et al. High prevalence of AIP gene mutations following focused screening in young patients with sporadic pituitary macroadenomas. Eur J Endocrinol. 2011;165(4):509-15.
  143. Korbonits M, Storr H, Kumar AV. Familial pituitary adenomas - Who should be tested for AIP mutations? Clin Endocrinol (Oxf). 2012;77(3):351-6.
  144. Ezzat S, Asa SL, Couldwell WT, Barr CE, Dodge WE, Vance ML, et al. The prevalence of pituitary adenomas: a systematic review. Cancer. 2004;101(3):613-9.

Gastrointestinal Disorders in Diabetes

ABSTRACT

 

Gastrointestinal manifestations of type 1 and 2 diabetes are common and represent a substantial cause of morbidity and health care costs, as well as a diagnostic and therapeutic challenge. Predominant among them, and most extensively studied, is abnormally delayed gastric emptying or diabetic gastroparesis. Abnormally increased retention of gastric contents may be associated with symptoms, including nausea, vomiting, postprandial fullness, bloating, and early satiety, which may be debilitating. However, the relationship of upper gastrointestinal symptoms with the rate of gastric emptying is relatively weak. Moreover, gastrointestinal symptoms also occur frequently in people without diabetes, which may compromise the capacity to discriminate gastrointestinal dysfunction resulting from diabetes from common gastrointestinal disorders such as functional dyspepsia. A definitive diagnosis of gastroparesis thus necessitates measurement of gastric emptying by a sensitive technique, such as scintigraphy or a stable-isotope breath test. There is an inter-dependent relationship of gastric emptying with postprandial glycemia. Elevated blood glucose (hyperglycemia) slows gastric emptying while, conversely, the rate of emptying is a major determinant of the glycemic response to a meal. The latter recognition has stimulated the development of dietary and pharmacological (e.g. short-acting GLP-1 receptor agonists) approaches to improve postprandial glycemic control in type 2 diabetes by slowing gastric emptying. The outcome of current management of symptomatic diabetic gastroparesis is often sub-optimal - optimizing glycemic control, the correction of nutritional deficiencies, and use of pharmacotherapy, are important. A number of promising and novel pharmacotherapeutic agents are in development. This chapter focusses on gastric motor function, but also provides an overview of the manifestations of esophageal, gall bladder, small and large intestinal function, in diabetes.

 

INTRODUCTION

 

The gastrointestinal tract extends from the mouth to the anus and performs functions vital to sustaining life including ingestion, breakdown and digestion of nutrients, facilitating nutrient absorption and preparation and expulsion of the waste product. Gastrointestinal symptoms occur commonly in people with diabetes, and include gastro-esophageal reflux, bloating, nausea, constipation, diarrhea, and fecal incontinence. It has been suggested that more than 50% of individuals attending outpatient diabetic clinics will at some stage experience a distressing gastrointestinal symptom. Gastrointestinal motor dysfunction is also common in diabetes and may have an impact on glycemic control. Of the motor dysfunctions, gastroparesis, or delayed gastric emptying, is the most important and will be discussed in relatively greater detail. This chapter is limited to the gastrointestinal manifestations of type 1 and 2 diabetes and does not address other causes of diabetes, such as that related to cystic fibrosis. 

 

GASTROINTESTINAL SYMPTOMS

 

Gastrointestinal symptoms are exhibited frequently in type 1 and 2 diabetes and most, but not all, studies suggest that they are significantly more common in diabetes than in controls without diabetes (1); reported inconsistencies likely reflect discrepancies in the methodology used and the patient populations studied. It should be appreciated that gastrointestinal symptoms are often not volunteered, particularly those considered embarrassing (such as fecal incontinence) and it would not be surprising if current estimates are less than is really the case. Symptoms, unfortunately, continue to be evaluated in clinical trials solely using participant ‘self-report’ despite its appreciated unreliability, rather than simple, validated measures, which are used widely in the assessment of ‘functional’ gastrointestinal disorders (e.g.  irritable bowel syndrome and functional depression) (2)  (1)  Symptoms appear to be more common in women with diabetes, as is the case with functional gastrointestinal disorders (3). While it is unclear whether symptom prevalence varies between type 1 and type 2 diabetes there is no doubt that gastrointestinal symptoms have a substantial negative impact on quality of life in people with diabetes (4). There is, however, a poor correlation between gastrointestinal symptoms and measures of function, such as the rate of gastric emptying. The natural history of gastrointestinal symptoms remains poorly defined, although it is known that onset and disappearance of symptoms is common i.e. there is considerable ‘symptom turnover’ - approximately 15-25 % over a 2-year period has been observed in type 2 patients (1). This symptom turnover has been reported to be associated with the onset of depression, but not with autonomic neuropathy or glycemic control (5).  

 

GASTROINTESTINAL MANIFESTATIONS IN DIABETES

 

Esophagus

 

The esophagus, a muscular tube connecting the pharynx to the stomach, enables propulsion of swallowed food, with a sphincter at either end (the upper and lower esophageal sphincters) to prevent esophago-pharyngeal and gastro-esophageal reflux, respectively.

 

Two common esophageal symptoms are heartburn (as part of gastro-esophageal reflux disease) and dysphagia (potentially indicating esophageal motor dysfunction). Techniques to evaluate esophageal motility include conventional and high-resolution manometry (HRM). Scintigraphy can measure esophageal transit but has not been standardized and is not commonly employed in clinical settings.

 

The relationship between esophageal transit and gastric emptying in diabetes is poor (6). Acute hyperglycemia inhibits esophageal motility (7), and reduces the basal lower esophageal sphincter pressure (8). While the esophagus has been less well studied than the stomach, it is clear that disordered esophageal function occurs frequently and that disordered motility in both the esophagus and stomach may share a similar pathogenesis. It has been postulated that the major mechanism underlying esophageal dysmotility is a reduction of cholinergic activity and vagal parasympathetic dysfunction (9). The pathological abnormalities associated with gastroparesis, such as a reduction in interstitial cells of Cajal and inhibitory intrinsic neurons, have also been postulated to be relevant to esophageal dysmotility (10). Diffuse esophageal muscular hypertrophy was reported in two-thirds of people with diabetes in one case series (11).

 

There are limited evidence-based options for the management of esophageal disorders in diabetes. General measures include lifestyle modifications (improved glycemic control, weight loss, dietary modifications, and physical exercise). Prokinetic agents have been used, albeit with limited evidence to support efficacy. The latter include dopaminergic agents (metoclopramide, domperidone), serotonin receptor agonists (cisapride), and motilin agonists (erythromycin). Botulinum toxin was trialed in a pilot study in patients with achalasia (including those with diabetes) and peripheral neuropathy and improvements in effective peristalsis induction and contraction amplitude were reported (12).

 

Gastro-esophageal reflux disease (GERD) is extremely common in the general population and also frequently seen in diabetes. In non-erosive GERD, treatment involves lifestyle measures (bed elevation of 30 degrees at head- end) and use of proton-pump inhibitors. In a community study, a reduced rate of heartburn was found in type 1 patients when compared with a control population (13), although this observation remains to be confirmed and the implications are unclear.

 

Disordered esophageal motility, especially the elderly, increases the risk of ‘pill-induced esophagitis’, with mucosal injury due to prolonged exposure to impacted medications (14). Diabetes is an independent risk factor (14), and the condition usually presents as chest pain with or without odynophagia. Treatment involves withdrawal of the offending agent and use of proton pump inhibitors (15).

 

Stomach - Diabetic Gastroparesis

 

INTRODUCTION

 

Delayed gastric emptying in diabetes was first reported almost a century ago, but it was Kassander who, in 1958, documented asymptomatic increased gastric retention of barium in diabetes and coined the descriptive term ‘gastroparesis diabeticorum’ (16). Interestingly, Kassander also suggested in their paper that gastroparesis could adversely impact glycemic control. Some sixty years on, diabetic gastroparesis, traditionally defined as abnormally delayed gastric emptying of solid food in the absence of mechanical obstruction, remains a diagnostic and management challenge (17). Gastroparesis occurs in both type 1 and 2 diabetes and may not, necessarily, be indicative of a poor prognosis (18,19).

 

The rate of gastric emptying is now appreciated as a major determinant of postprandial glycemia in both health and diabetes (20), and novel anti-diabetic medications, such as short acting GLP-1 receptor agonists, diminish postprandial glycemic excursions predominantly by slowing gastric emptying, are used widely.

 

EPIDEMIOLOGY OF DIABETIC GASTROPARESIS

 

The ‘true’ incidence and prevalence of diabetic gastroparesis globally remain uncertain largely due to inconsistencies in the definition of gastroparesis, study populations, and methodology. It is, however, clear that diabetes is a leading cause of gastroparesis, accounting for about 30% of cases in tertiary referral studies (17). A recent analysis of data from the follow-up arm of the landmark prospective study in type 1 diabetes, called DCCT-EDIC (Diabetes Control and Complications –Epidemiology of Diabetes Interventions and Complications) found that delayed gastric emptying of a solid meal occurred in 47% of this population, consistent with the prevalence reported in other cross-sectional studies (21). Previously believed to be essentially a complication of advanced type 1 diabetes (T1D), it is now apparent that gastroparesis also occurs frequently in type 2 diabetes (T2D) (16,22). Risk factors for gastroparesis include a long duration of diabetes, the presence of other microvascular complications, female gender, obesity. and smoking (17).  In a recent report from the NIH Gastroparesis Consortium, the proportion of T1D and T2D was comparable (although many more people with T2D have gastroparesis as its prevalence is much higher), although a US-based community study based on symptomatic cases, reported an incidence of approximately 5% in T1D and 1% in T2D (compared with 0.01% in controls) (23). Data from the US indicate that hospitalizations due to diabetic gastroparesis rose 158% between 1995-2004, which may reflect a true increase in incidence and / or greater clinical awareness of the condition (24). Not surprisingly, health care costs related to diabetic gastroparesis have also increased substantially in recent years. It should, however, also be noted that the awareness of the central importance of glycemic control to the development and progression of microvascular complications, and the consequent increased priority in management to improve it, may have led to a reduction in the incidence of gastroparesis. Consistent with this, it has recently been shown that in well-controlled T2D, even when longstanding, the prevalence of gastroparesis is low and, not infrequently, gastric emptying is modestly accelerated (19,25).

 

DIAGNOSIS OF DIABETIC GASTROPARESIS

 

As alluded to, the presence of gastrointestinal symptoms is poorly predictive of delayed gastric emptying. It is well established that patients with debilitating upper GI symptoms may have normal, or even rapid emptying, while others with unequivocally markedly delayed emptying may report few, or no symptoms. Measurement of gastric emptying, after exclusion of mechanical obstruction at the gastric outlet or proximal small intestine is, accordingly, mandatory for a formal diagnosis of gastroparesis, for which scintigraphy, developed in the 1970s, remains the ‘gold standard’ technique. An attempt has been made to standardize the methodology, with the American Neurogastroenterology and Motility Society and the Society of Nuclear Medicine defining gastroparesis by the intra-gastric retention of >60% of a standardized meal at 2 hours and/or >10% at 4 hours (26). The test meal advocated in the consensus statement comprises two egg-whites, two slices of bread and jam (30 g) with water (120 ml), providing 255 kcal with little fat (72% carbohydrate, 24% protein, 2% fat and 2% fiber) (26). While a useful exercise, the probability of universal adoption of a specific meal, especially outside Western cultures, is intuitively low. The advantages of scintigraphy are its capacity for precise, concurrent measurement of both solid and liquid meal components (the ‘consensus’ test meal only labels the solid component); however, it involves radiation exposure and requires sophisticated equipment and technical expertise. Acceptable alternatives include 13C based breath tests and ultrasonography, neither of which involve radiation exposure, although the latter is operator-dependent (22). Newer techniques, such as the wireless motility capsule, MRI, and SPECT imaging have emerged, but at present these should be considered less accurate than scintigraphy and / or only relevant to a research setting (17).

 

PATHOGENESIS OF DIABETIC GASTROPARESIS

 

Gastric emptying is a complex, coordinated process by which chyme is delivered to the small intestine at a tightly regulated rate and involves the gastro-intestinal musculature, nervous systems (intrinsic and extrinsic), gastric ‘pacemaker’ (so-called ‘Interstitial cells of Cajal or ICC), immune cells, and fibroblast-like cells that stain positive for platelet derived growth factor receptor alpha.  In the fasting state, a cyclical pattern of contractile activity known as the ‘migrating motor complex’ (MMC) sweeps from the stomach through to the small intestine, which serves a “housekeeping’ role i.e. facilitating the movement of ingestible food particles and bacteria from the stomach through the intestine (27). There are distinct phases of the MMC: phase I consists of motor quiescence lasting approximately 40 min, phase II, approximately 50 min, is comprised of irregular contractions, and phase III is characterized by regular contractions (at approximately 3 per min in the stomach and about 10-12 per small intestine) for 10 min during which the bulk of indigestible solids are emptied (28). Following meal ingestion, the MMC is replaced by a ‘postprandial’ motor pattern. Solids are then mixed with gastric acid and ground into small particles (usually < 1-2 mm) in the distal stomach. Gastric accommodation is mediated by vagal and nitrergic mechanisms, antral contractions by vagal and intrinsic cholinergic mediation, and pyloric relaxation by nitrergic mechanisms (17). The resultant chyme is delivered through the pylorus to the proximal duodenum predominantly in a pulsatile manner (22,27). It is now appreciated that the rate of emptying is regulated primarily by nutrient-induced inhibitory feedback arising from the small intestine, rather than by ‘intragastric’ mechanisms (29). Digestible solids and high nutrient liquids empty from the stomach in an overall linear fashion as a result of this feedback (6); solid emptying is preceded by an initial so-called ‘lag-phase’ of 20-40 min during which solids are ground into small particles. In contrast to solids, low or non-nutrient liquids empty in an overall, volume-dependent, monoexponential pattern because small intestinal feedback is less (27). A number of gut peptides play a key role in providing intestinal feedback, including GLP-1, CCK, and peptide YY. In contrast, ghrelin and motilin, which accelerate gastric emptying, are suppressed following food intake (22,27). Both the length and region of small intestine exposed to nutrients modulate feedback to slow gastric emptying (30).

 

Disordered gastric emptying represents the outcome of impairments of variable combinations of these diverse components. Advances in understanding the underlying pathophysiology have been made over the past decade, particularly through the efforts of the NIH-funded Gastroparesis Clinical Research Consortium. Histological studies from this group and others have shown a reduction in the number of interstitial cells of Cajal in diabetic gastroparesis, which correlates with the magnitude of delay in emptying (31). Interstitial cells of Cajal loss appears to be driven by an immune infiltrate involving a shift from protective M2, to classically activated, M1 macrophages, with defective regulation of heme oxygenase-1 and resultant oxidative stress.  Altered expression of the Ano-1 gene which influences conduction in the Interstitial cells of Cajal has also been reported (32). A reduction in inhibitory neurons expressing nitric oxide synthase also appears to contribute (31).

 

GASTRIC EMPTYING AND GLYCEMIA (FIGURE 1)

 

Figure 1. Bidirectional relationship between gastric emptying and glycemia. Abbreviations: CCK, cholecystokinin; GIP, gastric inhibitory polypeptide; GLP-1, glucagon-like peptide 1; PYY, peptide YY. Reproduced with permission from Philips et al (22)

 

Gastric emptying exhibits a wide inter-individual variation (ranging between 1-4kcal /min in health and even wider in diabetes because of the high prevalence of gastroparesis and, less often, abnormally rapid emptying) (Figure 2).  Gastric emptying is a major determinant of postprandial glycemia across glucose-tolerant states and these relationships are time- dependent. In individuals with normal glucose tolerance, following a 75g oral glucose drink, the early (approximately 30 min) rise in glucose is directly proportional to the rate of emptying, while the 120 min value (the standard endpoint in an OGTT) is inversely related. This relationship shifts to the right as glucose tolerance worsens, such that both 30- and 120-min glucose values are directly proportional to the gastric emptying rate in type 2 diabetes(33-35).Epidemiological studies indicate that about 50% of people with impaired glucose tolerance or IGT will develop frank type 2 diabetes and, hence, factors affecting progression are of considerable interest. We have shown that the disposition index – a predictor of progression to type 2 diabetes – is inversely related to the gastric emptying rate (36), suggesting that the rate of emptying may influence the progression. There is evidence that the 1-hour plasma glucose level in a 75g oral glucose tolerance test is strongly associated with the risk of future type 2 diabetes (37)  and this is known to be dependent on the rate of gastric emptying (34,35).

 

In type 2 diabetes, slowing of gastric emptying (such as by morphine) reduces the postprandial glycemic profile, while accelerating emptying by pro-kinetics (such as erythromycin) increases it (38). Bypassing the stomach and delivering glucose directly into the small intestine at specified rates (within the physiological span of gastric emptying) via naso- duodenal catheters has been used as a model to characterize the impact of gastric emptying on glycemia. These 'surrogate' studies indicate that gastric emptying is a major determinant of postprandial insulin secretion and the magnitude of the so-called 'incretin' effect (the augmented insulin secretory response to oral or enteral, compared with intravenous, glucose). Moreover, the relative contribution of the two 'incretin' hormones (GIP and GLP-1) to the incretin effect in health varies such that GIP is the predominant contributor when glucose enters the small intestine at 2kcal/ min or less, with GLP-1 contributing only at higher rates of duodenal glucose delivery (3 or 4 kcal per min) (39). It is, therefore, likely that the relative contributions of GIP and GLP-1 to the postprandial insulin response and glycemia depend on an individual’s intrinsic rate of emptying. Variations in blood glucose also affect gastric emptying. Through ‘glucose clamp’ studies, we have shown that abrupt elevations in blood glucose slows gastric emptying in a ‘dose- dependent’ manner, i.e. the slowing is dependent on the magnitude of the elevation in blood glucose (7).  Moreover, when blood glucose is ‘clamped’ at about 8 mmol/L or 144 mg/dL (i.e. physiological hyperglycemia), gastric emptying is modestly slower in both health and well-controlled type 1 diabetes (40). This may, however, not apply to spontaneous elevations in blood glucose (41)  and further clarification is required. On the other hand, acute hypoglycemia (blood glucose about 2.6 mmol/L or 46.8 mg/dL) accelerates gastric emptying markedly in both groups (42), and is likely to represent an important counter-regulatory mechanism. It follows that the acute glycemic environment, by altering gastric emptying, is likely to influence intestinal absorption of nutrients, as well as oral medications, which has hitherto been poorly appreciated in clinical practice. The impact of chronic glycemic control on gastric emptying remains uncertain.

 

Figure 2. Gastric emptying of solids (minced beef) (A), shown as the retention at 100 min (percent); and the gastric emptying of liquids (10% dextrose) (B), shown as the 50% emptying time (minutes) in 101 outpatients with diabetes. The normal range is indicated by the shaded area. Reproduced with permission from Jones et al (43).

 

In people with insulin-treated diabetes (type 1 or type 2), it is important to match exogenous insulin delivery with the availability of carbohydrate to minimize the risk of postprandial hypoglycemia. It is, therefore, intuitively likely that delayed gastric emptying predisposes to lower blood glucose concentrations in the early postprandial period (so-called ‘gastric hypoglycemia’) (44) and subsequent hyperglycemia. A study in type 1 patients reported that insulin requirements were lower in those with gastroparesis during the first 120 min post-meal, but greater during 180-240 min, compared to patients with normal gastric emptying (45). It is increasingly appreciated that greater glycemic variability is associated with worse outcomes (46). Knowledge of the rate of emptying may potentially assist the clinician in developing strategies to reduce postprandial glycemic variability in individual patients, although this needs to be evaluated formally.

 

MANAGEMENT OF SYMPTOMATIC GASTROPARESIS (FIGURE 3)

 

Figure 3. Treatment Algorithm for Diabetic Gastroparesis. PRN, as needed. Reproduced with permission from Du et al (1)

 

General Measures

 

Management of gastroparesis should be individualized. In clinical practice, patients are generally advised to consume small, frequent meals that are low in fat and fiber, with more calories as liquids than solids; ingested solids should be those that fragment readily into small particles (47). It should, however, be noted that this advice has not been rigorously evaluated and may be difficult to adhere to, so that the involvement of a dietitian is recommended (48). While optimizing glycemic control is intuitively important, given the inhibitory effect of acute hyperglycemia on gastric emptying, this has not been clearly established to be the case in the chronic setting, although the use of continuous subcutaneous glucose infusion and continuous glucose monitoring has recently been advocated (49).

 

Concurrent medications should be reviewed and, if possible, those which may slow gastric emptying (e.g. opiates, anticholinergics) ceased. In this regard, it should be appreciated that short-acting GLP-1 receptor agonists (e.g. exenatide BD and lixisenatide) and the amylin analogue, pramlintide (50), improve chronic glycemic control primarily by slowing gastric emptying.

 

Medications

 

Although studies involving pro-kinetic medications for treatment of gastroparesis have nearly all been of short duration and involved a modest number of participants, these drugs are used widely and form the mainstay of therapy. Major limitations are their adverse effect profile and tachyphylaxis i.e. diminution in pharmacological effect over time. Tachyphylaxis is thought to particularly affect motilin agonists, although this has not been well studied. Cisapride (a 5HT4 agonist) was used widely for symptomatic management, but shown subsequently to be associated with cardiac adverse effects (prolonged QT interval and ‘torsades de pointes’) and taken off the market. The most commonly used medications are discussed below. Some prokinetic drugs also have antiemetic properties.

 

Metoclopramide, a dopamine D2 receptor antagonist, improves gastric emptying (48), and can be administered via oral, intranasal, and subcutaneous routes, but is associated with central nervous system adverse events (including tardive dyskinesia), which may be irreversible. Accordingly, the US Food and Drug Administration (FDA) recommends short duration (12 weeks) use only. An intranasal formulation of metoclopramide under development was reported to be efficacious in women, but not men, implying the potential importance of gender in selecting the route of delivery (51). Metoclopramide can also be injected subcutaneously in an attempt to abort attacks of vomiting. It is the only medication that is approved currently by the FDA for the management of gastroparesis.

 

Domperidone is another D2 receptor antagonist, but unlike metoclopramide, does not cross the blood-brain barrier and is associated with fewer adverse events, with apparently comparable improvements in gastric emptying and upper gastrointestinal symptoms (48,52). Domperidone may prolong the QT interval and affect metabolism of other medications through the CYP2D6 pathway (48).

 

The antibiotic, erythromycin, is a motilin receptor agonist and is effective acutely, and inexpensive, but needs to be administered frequently and may also prolong the QT interval and interact with other medications, in this case through the CYP3A4 pathway (48). Acute, intravenous, administration of erythromycin markedly accelerates delayed gastric emptying (53) and may assist in the placement of neuroenteric tubes (54). The gastrokinetic effect of erythromycin is, however, subject to tachyphylaxis (55).

 

A number of novel agents are in Phase 2-3 trials, including ghrelin and 5HT4 receptor agonists. Ghrelin (sometimes referred to as the 'hunger' hormone) is secreted from the fundus of the stomach and has important roles in nutrient sensing and appetite regulation. Administration of ghrelin accelerates gastric emptying in both animals and humans (56). The outcome of phase 2 trials of the ghrelin agonist, relamorelin, have been promising, with a reduction in upper gastrointestinal symptoms in type 1- and 2 patients with gastroparesis as well as an acceleration of gastric emptying (57). An international phase 3 trial is in progress. Similarly, the oral highly selective 5 HT4 agonists, velusetrag (which was marketed for constipation) and prucalopride, accelerate gastric emptying (58,59). A recent study reported that 4 weeks’ of treatment with prucalopride in 32 people with gastroparesis (including 6 with diabetes) improved both symptoms and accelerated gastric emptying, although sub-group analysis of the diabetic cohort was not performed due to small numbers (59).

 

Treatment-Refractory Gastroparesis

 

Gastroparesis refractory to dietary and pharmacological intervention is debilitating for the patient and management represents a substantial challenge. Bypassing the stomach using jejunal or parenteral feeding, may be required to sustain nutrition. Gastric electrical stimulation (GES) using the ‘Enterra’ device) appeared to be a promising therapeutic option when initial unblinded studies were indicative of symptom improvement (22,48) and is currently approved by the FDA for ‘humanitarian exemption’; however, a subsequent blinded study failed to show a difference between periods where the stimulator was switched ‘on’ or ‘off’ (22,48,60,61).  A recently reported randomized cross-over trial reported a reduction in frequency of refractory vomiting following GES for a 4-month period in gastroparesis with or without diabetes but improvement in symptom control did not accelerate gastric emptying or benefit quality of life (62). Similarly, pyloric botulinum toxin injections have fared much better in uncontrolled, than in sham-controlled trials (22).  Surgical and endoscopic interventions, such as pyloroplasty and pyloromyotomy, and acupuncture have been described in literature, but lack controlled outcome data (22,48).

 

Gall Bladder

 

Gall stones are encountered more frequently in people with diabetes, which is not surprising given that risk factors for the development of stones, such as intestinal dysmotility, obesity, and hypertriglyceridemia, are more common in this group (particularly type 2 diabetes) (63). In addition, impairment of gall bladder motility and autonomic neuropathy, as well as factors such as cholesterol supersaturation and crystal nucleation promoting factors, are considered important. Common techniques used to measure gall bladder motor function include ultrasound and scintigraphy. Some studies have found increased fasting gall bladder volume, while in others, there was no difference, or even a reduction, in people with diabetes. It is possible that differences in the techniques employed (ultrasound or scintigraphy), and the presence of autonomic neuropathy may account for these discrepancies. Many studies, however, have reported impairment in postprandial gall bladder emptying in diabetes, sometimes termed ‘diabetic cholecystoparesis’ (63). It is also possible that delayed gastric emptying contributes to delayed emptying from the gall bladder. In health, acute hyperglycemia inhibits gall bladder motility in a dose-dependent manner (64). An increased prevalence of gall bladder-related disorders (including cholecystitis and cholelithiasis) is associated with the use of GLP-1 receptor agonists (65) and may potentially relate to a drug-induced prolongation of gall bladder refilling time (66). Similarly, an increase in gall-bladder disease has been reported post-bariatric surgery in obese individuals (including those with diabetes) with the implication that dramatic weight loss may predispose (67).

 

Small Intestine

 

While diabetic enteropathy is common, it has been studied much less comprehensively than diabetic gastroparesis (68). Symptoms of constipation and diarrhea are discussed in the section on large intestinal disorders in diabetes, which follows.

 

Traditionally, vagal dysfunction has been regarded as the major impairment in diabetic enteropathy. However, as is the case with gastroparesis, recent evidence has suggested a critical role for both interstitial cells of Cajal and nNOS (31). Acute hyperglycemia also has a major effect on postprandial small intestinal motility in health (and, presumably, diabetes) by reducing the amplitude of duodenal and jejunal pressure waves, as well as retarding duodenal-cecal transit (69).

 

Small intestinal bacterial overgrowth (SIBO), probably secondary to altered small intestinal motility, is commonly encountered in diabetes; estimates range between 15-40% in type 1 diabetic cohorts. A major limitation of these studies is lack of a ‘gold standard’ method for diagnosis.

 

There is limited information about small intestinal glucose absorptive function in diabetes but, based on animal models, it has been suggested that carbohydrate digestion is disordered. For example, streptozotocin-induced diabetes in rats, is associated with an increase in mucosal absorption of glucose (70). We have demonstrated that small intestinal glucose absorption is comparable in uncomplicated type 1 patients and healthy controls, but probably affected by both duodenal motility and the prevailing glycemic environment (71) - when blood glucose was elevated, intestinal glucose absorption was increased, while absorption was comparable to that in healthy controls during euglycemia. A fundamental limitation in interpreting the outcome of the numerous studies which have reported the potent modulatory effect of the rate of gastric emptying on postprandial glycemia is their failure to discriminate between effects mediated by changes in gastric emptying from those potentially secondary to changes in small intestinal transit  (72).

 

DIAGNOSIS OF ENTEROPATHY

 

Diabetic enteropathy is often a diagnosis of exclusion. It is essential to exclude underlying non-diabetes related etiologies where relevant – for example, testing for celiac disease in type 1 patients is recommended. It should be appreciated that gastrointestinal adverse effects occur frequently with commonly used anti-diabetic medications. Metformin, GLP-1RAs, SGLT2 inhibitors, and particularly alpha-glucosidase inhibitors (e.g. acarbose), which are used widely, are commonly associated with intestinal symptoms.

 

Small intestinal manometry (measurement of contractile activity) may provide mechanistic insights, but its use is limited to specialized centers. Scintigraphy can quantify small intestinal transit, but the diagnostic significance is uncertain. More recently, technologies, including ingestible wireless capsules (such as the SmartPill) and continuous tracking of capsules (3D-Transit system), have been employed; these are promising, but require further validation before clinical exploitation. Small intestinal bacterial overgrowth can be diagnosed by aspiration and culture of intestinal fluid or breath tests, but both have substantial limitations and neither technique can be regarded as a “gold standard”.

 

MANAGEMENT OF ENTEROPATHY

 

Symptom management with medications is common. Prokinetic agents used for gastroparesis are commonly employed for management of disordered intestinal motility, but much less well evaluated. Small intestinal bacterial overgrowth can be treated with antibiotics, such as rifamixin (most common but expensive), amoxicillin-clavulanic acid, or metronidazole. Not surprisingly, small intestinal bacterial overgrowth frequently relapses.

 

Large Intestine

 

The major function of the colon is to re-absorb water and electrolytes from the intraluminal contents, to concentrate and solidify the waste product, and prepare for its elimination. The most common lower gastrointestinal symptoms are constipation, diarrhea, abdominal pain, and distention. It is difficult to estimate a ‘true’ incidence and prevalence. Cohort studies have reported the presence of chronic constipation in up to 25% of people with type 1 and 2 diabetes, while that of chronic diarrhea is up to 5% (73).  Bytzer et al reported a higher prevalence of constipation and diarrhea in people with type 2 diabetes (15.6% compared with 10% in those without diabetes) (3). A recent report analyzing data from the large-scale US public survey, NHANES, found that chronic diarrhea was more common in people with type 1 and 2 diabetes compared with non-diabetic controls (~ 11% vs 6%) (74).

 

CONSTIPATION

 

The etiology of constipation in diabetes is likely to be multifactorial. A study involving only 10 patients found prolonged colonic transit time in those with constipation (13). Autonomic neuropathy is thought to be important; constipation is more common in those with diabetes and autonomic impairment (75). Validated techniques for evaluation include colonic transit scintigraphy and the use of radio-opaque markers and wireless motility capsules (76), but their utility in routine clinical practice has not been fully established.

 

Management of diabetic constipation must include a medication history review and those that may cause constipation should be ceased, if feasible (figure 4). For mild constipation, the American Diabetes Association recommends lifestyle modification such as increased physical exercise and dietary fiber. Over-the-counter laxatives (bulk, osmotic or stimulatory) such as Senna, Bisacodyl and water- soluble fiber supplements are commonly prescribed. Other medications like lactulose, linaclotide, and lubiprostone (the latter two available by prescription in the United States) have been used. There are no head-to-head trials to determine which agent is superior. However, it has been suggested that lactulose may potentiate glucose- lowering (77). Lubiprostone, which acts by direct activation of CIC-2 chloride channels on enterocytes, has been reported to improve both spontaneous bowel movements and accelerate colon transit in a randomized controlled trial in a cohort with diabetes (78). In a randomized trial cholinesterase inhibition with pyridostigmine in 30 people with diabetes (12 T1D, 18 T2D) and chronic constipation, there were superior improvements in both bowel function and colonic transit compared with placebo (79).

 

Figure 4. Algorithm for Management of Chronic Constipation in Patients with Diabetes.

 

CHRONIC DIARRHEA

 

“Diabetic diarrhea” has been traditionally considered a manifestation of autonomic neuropathy (80). The typical symptom is large volume, painless, nocturnal, diarrhea with or without fecal incontinence. Again, the diagnosis essentially represents one of exclusion and it is important to distinguish diarrhea from fecal incontinence. It should be remembered that widely used glucose-lowering therapies, including metformin (malabsorptive), acarbose (osmotic), and GLP-1 receptor agonists, not infrequently cause diarrhea. It is likely that optimizing glycemic control is important in the management of diabetic diarrhea (81), but again, this has not been rigorously evaluated. Dietary strategies include a low FODMAP (Fermentable Oligosaccharides, Disaccharides, Monosaccharides and Polyols) diet under guidance of a qualified dietitian, although this has not been evaluated specifically for the diabetes population in clinical trials. Loperamide, an over-the-counter mu opioid receptor agonist, is used widely. Bile acid sequestrants, such as cholestyramine and colesevelam, are used when bile salt malabsorption is suspected, and have the added advantage of reducing LDL cholesterol and glycated hemoglobin. Other agents include clonidine, diphenoxylate, octreotide, and ondansetron (figure 5).

 

It has been reported that people with diabetes, especially type 1 diabetes, are more likely to have inflammatory bowel disease (IBD) such as ulcerative colitis. Diabetes also appears to be an independent risk factor for Clostridium difficile infection where metformin appears to be protective, probably via its action on the gut microbiota (82). It has also been suggested that there is a link between diabetes and colorectal malignancy (83), and diabetes is associated with worse outcomes and response to colorectal surgery. Interestingly, some observational studies suggest that metformin may have chemo-preventative properties against colorectal malignancy (84).

 

Figure 5. Algorithm for Management of Chronic Diarrhea in Patients with Diabetes.

 

Rectum and Anus

 

Fecal incontinence occurs more frequently in people with diabetes and is associated with the duration of disease, and the presence of microvascular complications, including autonomic and peripheral neuropathy (85). Both internal anal sphincter tone and anal squeeze pressures are reduced in diabetes compared with healthy controls (86,87). A key step in management is to exclude important differential diagnoses, such as colorectal malignancy and irritable bowel disease (88). No single test can be regarded as ‘gold standard’, but anorectal manometry (conventional, 3D or high resolution) is very useful in clinical practice to estimate ano-rectal motor abnormalities, while barium defecography is useful to detect rectal motory, sensory and structural abnormalities (89).

 

Treatment of fecal incontinence is rarely curative, and the focus of management is to improve symptoms and quality of life. Fecal impaction with overflow can be managed by initial manual removal of stool from the rectum and enemas (promoting evacuation) and the subsequent prescription of bulk laxatives, increasing fiber intake, and toilet training. Operant reconditioning of rectosphincteric responses, called ‘biofeedback’ training, was first described by Engel et al in 1974 (90) and can be useful in treating fecal, as well as urinary, incontinence. The technique involves visual demonstration of voluntary contraction of external anal sphincter (EAS) contraction to the patient and training to improve the quality of the response (both strength and duration). Biofeedback training is effective in the longer term in only about 60% of patients in clinical trials; those with a low bowel satisfaction score and having digital evacuations fare better (91).

 

GASTROINTESTINAL EFFECTS OF ANTI-DIABETIC MEDICATIONS AND THEIR IMPLICATIONS FOR CLINICAL PRACTICE

 

Gastrointestinal adverse effects are extremely common in people treated with glucose-lowering medications for type 2 diabetes. In the case of alpha glucosidase inhibitors such as acarbose and miglitol, these effects (e.g., diarrhea and abdominal distention) are predictable sequelae of the malabsorption of carbohydrate (92) . There is new information in relation to two classes of medications (biguanides and GLP-1 receptor agonists).

 

Metformin, a biguanide of herbal origin, remains a first line pharmacological agent of choice for type 2 diabetes. The precise mechanisms of action remain uncertain, although it clearly has multiple effects, including in the liver (block gluconeogenesis), as an insulin sensitizer, and direct actions through the gut, including slowing of gastric emptying (93) . Up to 25% of people using metformin report gastrointestinal adverse events, particularly diarrhea and nausea. Common outpatient clinic strategies to minimize these include initiating treatment at a low dose (i.e., 500mg/day) and gradually up-titrating to usually ~2000 mg/day, use of extended-release formulations and avoiding ingestion on an empty stomach, although evidence to support these approaches is not robust (94).

 

Similarly, GLP-1 receptor agonists (but not DPP-IV inhibitors which lead to only a modest rise in plasma GLP-1 levels), commonly cause gastrointestinal adverse effects. As mentioned, GLP-1 is a gut-based peptide with a profound, but variable, action to slow gastric emptying. This slowing is more marked when baseline gastric emptying is relatively more rapid and is predictive of the reduction in blood glucose following a meal (95) . GLP-1 plays a physiological role to slow gastric emptying - gastric emptying is accelerated by the specific GLP-1 antagonist, exendin 9-39 (96)  and delayed by exogenous administration of GLP-1 in modestly supra-physiological plasma levels (97). Upper gastrointestinal events induced by GLP-1 are, likely to reflect, in part, delayed emptying. As effects are also observed in the fasting state, a direct action on CNS GLP-1 receptors (most notably, area postrema in the brain stem) has also been postulated. A direct effect on the gut is likely to contribute to lower gastrointestinal adverse events such as diarrhea. GLP-1 secreting cells (specialized entero-endocrine ‘L’ cells) are found throughout the gastrointestinal tract, and GLP-1 may exert a local excitatory action in smooth muscle or through the intramural autonomic plexus to increase motility and induce diarrhea (98,99) . A fundamental limitation of the vast majority of clinical trials involving GLP-1 receptor agonists is that gastrointestinal adverse effects have been assessed using participant recall and not validated questionnaires. Nevertheless, results from large cardiovascular outcome trials relating to the use of GLP-1 agonists indicate that the proportion of participants discontinuing GLP-1 receptor agonists due to adverse gastrointestinal events ranges between 4.5 to 13% (100) . Nausea appears to be the most common symptom (up to 25%), with vomiting and diarrhea reported by about 10% (101). A retrospective analysis of 32 phase-3 trials involving ‘long’ and ‘short’ acting GLP-1 receptor agonists reported that gastrointestinal adverse effects are also dose-dependent, and that ‘long’ acting GLP-1 receptor agonists are associated with less nausea and vomiting, but more diarrhea when compared to short-acting GLP-1 receptor agonists (101). Symptoms are reported most frequently at the time of initiation of a GLP-1 receptor agent and may persist for several hours or days probably dependent on the Tmax of the drug (100). Gradual titration of dose is recommended, although evidence to support this approach is uncontrolled.

 

We, and others, have demonstrated employing the gold standard technique of scintigraphy to quantify gastric emptying and both ‘long’ and ‘short’ acting GLP-1 receptor agonists delay gastric emptying, although the magnitude of this deceleration appears to be greater with ‘short’ acting GLP-1 receptor agonists (95,102-104) . Moreover, it is appreciated that GLP-1 receptor agonists may slow gastric emptying profoundly in doses much lower than those used in the management of type 2 diabetes (2). It had been suggested, incorrectly, that long acting GLP-1 receptor agonists, which are now the most widely used form, have no effect on gastric emptying with sustained use (2). A further limitation of clinical trials of GLP-1 receptor agonists is that gastric emptying has either not been measured or a sub-optimal technique used (105).

 

Instances of apparently GLP-1 receptor agonist-induced gastroparesis are increasingly appearing in the medical literature as case reports (106). The prevalence of marked delay in gastric emptying induced by GLP-1 receptor agonists remains uncertain but has stimulated guidelines in relation to their use prior to elective surgery or endoscopy. For example, the American Society of Anesthesiologists (ASA), has recently published consensus guidelines on pre-operative management of people using GLP-1 agonists and have advised withholding a long-acting agent for at least one week prior to the procedure/surgery (107). Such recommendations lack a strong evidence base. It is uncertain whether these recommendations from the ASA will be universally adopted but it appears intuitively unlikely. Recently a UK-based expert group comprising endocrinologists, anesthetists, and pharmacists have recommended against this generic advice  (107) primarily on the lack of robust data demonstrating an increased risk of aspiration under anesthesia, while being on a GLP-1 receptor agonist, that the recommended duration of avoidance may be inadequate (for example, in people taking 1mg semaglutide, avoidance of one week is likely to reduce the plasma drug concentration by about half, which is still likely to slow gastric emptying), at least in some people and reintroduction of GLP-1 receptor agonists once normal food intake has been established has not clearly defined and there is intuitively the high potential for a deterioration in glycemic control, postoperatively including an increase in glycemic variability. They instead recommend that preoperative assessment for risk of aspiration be individualized.

 

In people co-prescribed with insulin and GLP-1 receptor agonists, there is likely to be an increased risk of a mismatch between insulin delivery and availability and intestinal glucose absorption due to prolonged gastric retention to predispose to hypoglycemia. Clinicians should be circumspect in prescribing a GLP-1 agonist and insulin combination in those who have impaired awareness of hypoglycemia or suspicion of delayed gastric emptying.

 

PANCREATIC EXOCRINE SUFFICIENCY IN DIABETES

 

There is an intricate anatomical association of endocrine and exocrine components of the pancreas which appears to translate to a reciprocal relationship between endocrine and exocrine dysfunction (108). However, a wide variation in the prevalence of pancreatic exocrine insufficiency in diabetes has been reported, with evidence that it is greater in type 1 (approx. 25-75%). compared with type 2 (approx. 25-50%) diabetes (109). The majority of these studies are in hospitalized populations; it is likely that prevalence in the community is lower. Our recent study of community type 2 patients reported a lower prevalence of 9% (110). The etiology in type 1 is thought to be a combination of lack of insulin (+/- glucagon and somatostatin), autoimmunity, autonomic neuropathy, and microvascular damage while the latter two contribute to pancreatic exocrine insufficiency in type 2 diabetes – it has been suggested that this may explain why pancreatic exocrine insufficiency is more common in patients with type 1 diabetes (109). Common symptoms are variable and include diarrhea (steatorrhea), abdominal pain, and failure to thrive in children. It is important to discriminate pancreatic and non-pancreatic causes of malabsorption. The relatively deep-seated location of the pancreas hinders easy assessment of its exocrine function. Diagnostic tests can be direct or indirect (111). Direct tests involve stimulation with exogenous hormones or nutrients while simultaneously collecting pancreatic secretions via duodenal intubation. This technique has many logistical issues (high costs, requirement of expertise, invasive nature) which limits its clinical utility despite being the most sensitive and specific. Examples of indirect tests include the 3-day fecal fat, fecal elastase-1 measurement, and breath tests (14C-triolein). Of these, the most common indirect and non-invasive (as well as relatively inexpensive) test in clinical practice is measurement of fecal elastase 1. It has been suggested that a fecal elastase-1 level less than 200 ug/g stool is indicative of mild pancreatic exocrine insufficiency, and a level of 100 ug/g stool of severe pancreatic exocrine insufficiency (108). It should be appreciated that sensitivity (55%) and specificity (60%) of fecal elastase-1 in diagnosing pancreatic exocrine insufficiency are modest. Measurement of fat-soluble vitamins may be indicated.

 

The principles of general management of pancreatic exocrine insufficiency include consumption of smaller, frequent meals, abstinence from alcohol, and involvement of an experienced dietitian. Pancreatic enzyme replacement therapy is regarded as the cornerstone of treatment (108). It is uncertain whether supplementation with pancreatic enzyme replacement therapy in those with type 2 diabetes and pancreatic exocrine insufficiency reduces postprandial glycemic excursions (110). Adjunctive therapies such as acid-suppressing agents are reserved for those with symptoms despite high-dose pancreatic enzyme replacement therapy.

 

CONCLUSIONS

 

Both gastrointestinal symptoms and dysmotility are common in diabetes and represent an important component of management. Gastric emptying is also a major determinant of postprandial glycemic control and may be modulated therapeutically to improve it. Current management of disordered gastrointestinal function, particularly gastroparesis, is primarily empirical, although a number of novel agents are in development; results of these clinical trials are eagerly anticipated.

 

REFERENCES

 

  1. Du YT, Rayner CK, Jones KL, Talley NJ, Horowitz M. Gastrointestinal Symptoms in Diabetes: Prevalence, Assessment, Pathogenesis, and Management. Diabetes Care 2018; 41:627-637
  2. Jalleh RJ, Jones KL, Nauck M, Horowitz M. Accurate Measurements of Gastric Emptying and Gastrointestinal Symptoms in the Evaluation of Glucagon-like Peptide-1 Receptor Agonists. Ann Intern Med 2023; 176:1542-1543
  3. Bytzer P, Talley NJ, Leemon M, Young LJ, Jones MP, Horowitz M. Prevalence of gastrointestinal symptoms associated with diabetes mellitus: a population-based survey of 15,000 adults. Archives of internal medicine 2001; 161:1989-1996
  4. Talley NJ, Young L, Bytzer P, Hammer J, Leemon M, Jones M, Horowitz M. Impact of chronic gastrointestinal symptoms in diabetes mellitus on health-related quality of life. The American journal of gastroenterology 2001; 96:71-76
  5. Quan C, Talley NJ, Jones MP, Spies J, Horowitz M. Gain and loss of gastrointestinal symptoms in diabetes mellitus: associations with psychiatric disease, glycemic control, and autonomic neuropathy over 2 years of follow-up. The American journal of gastroenterology 2008; 103:2023-2030
  6. Horowitz M, Maddox AF, Wishart JM, Harding PE, Chatterton BE, Shearman DJ. Relationships between oesophageal transit and solid and liquid gastric emptying in diabetes mellitus. European journal of nuclear medicine 1991; 18:229-234
  7. Fraser RJ, Horowitz M, Maddox AF, Harding PE, Chatterton BE, Dent J. Hyperglycaemia slows gastric emptying in type 1 (insulin-dependent) diabetes mellitus. Diabetologia 1990; 33:675-680
  8. De Boer SY, Masclee AA, Lam WF, Lamers CB. Effect of acute hyperglycemia on esophageal motility and lower esophageal sphincter pressure in humans. Gastroenterology 1992; 103:775-780
  9. Lam WF, Masclee AA, de Boer SY, Lamers CB. Hyperglycemia reduces gastric secretory and plasma pancreatic polypeptide responses to modified sham feeding in humans. Digestion 1993; 54:48-53
  10. Monreal-Robles R, Remes-Troche JM. Diabetes and the Esophagus. Curr Treat Options Gastroenterol 2017; 15:475-489
  11. Iyer SK, Chandrasekhara KL, Sutton A. Diffuse muscular hypertrophy of esophagus. The American journal of medicine 1986; 80:849-852
  12. Tack J, Zaninotto G. Therapeutic options in oesophageal dysphagia. Nature reviews Gastroenterology & hepatology 2015; 12:332-341
  13. Maleki D, Locke GR, 3rd, Camilleri M, Zinsmeister AR, Yawn BP, Leibson C, Melton LJ, 3rd. Gastrointestinal tract symptoms among persons with diabetes mellitus in the community. Archives of internal medicine 2000; 160:2808-2816
  14. Kikendall JW. Pill-induced esophagitis. Gastroenterol Hepatol (N Y) 2007; 3:275-276
  15. Dumic I, Nordin T, Jecmenica M, Stojkovic Lalosevic M, Milosavljevic T, Milovanovic T. Gastrointestinal Tract Disorders in Older Age. Can J Gastroenterol Hepatol 2019; 2019:6757524
  16. Kassander P. Asymptomatic gastric retention in diabetics (gastroparesis diabeticorum). Annals of internal medicine 1958; 48:797-812
  17. Camilleri M, Chedid V, Ford AC, Haruma K, Horowitz M, Jones KL, Low PA, Park SY, Parkman HP, Stanghellini V. Gastroparesis. Nat Rev Dis Primers 2018; 4:41
  18. Chang J, Russo A, Bound M, Rayner CK, Jones KL, Horowitz M. A 25-year longitudinal evaluation of gastric emptying in diabetes. Diabetes care 2012; 35:2594-2596
  19. Watson LE, Phillips LK, Wu T, Bound MJ, Jones KL, Horowitz M, Rayner CK. Longitudinal evaluation of gastric emptying in type 2 diabetes. Diabetes research and clinical practice 2019; 154:27-34
  20. Marathe CS, Rayner CK, Jones KL, Horowitz M. Relationships between gastric emptying, postprandial glycemia, and incretin hormones. Diabetes care 2013; 36:1396-1405
  21. Bharucha AE, Batey-Schaefer B, Cleary PA, Murray JA, Cowie C, Lorenzi G, Driscoll M, Harth J, Larkin M, Christofi M, Bayless M, Wimmergren N, Herman W, Whitehouse F, Jones K, Kruger D, Martin C, Ziegler G, Zinsmeister AR, Nathan DM, Diabetes C, Complications Trial-Epidemiology of Diabetes I, Complications Research G. Delayed Gastric Emptying Is Associated With Early and Long-term Hyperglycemia in Type 1 Diabetes Mellitus. Gastroenterology 2015; 149:330-339
  22. Phillips LK, Deane AM, Jones KL, Rayner CK, Horowitz M. Gastric emptying and glycaemia in health and diabetes mellitus. Nature reviews Endocrinology 2015; 11:112-128
  23. Jung HK, Choung RS, Locke GR, 3rd, Schleck CD, Zinsmeister AR, Szarka LA, Mullan B, Talley NJ. The incidence, prevalence, and outcomes of patients with gastroparesis in Olmsted County, Minnesota, from 1996 to 2006. Gastroenterology 2009; 136:1225-1233
  24. Wang YR, Fisher RS, Parkman HP. Gastroparesis-related hospitalizations in the United States: trends, characteristics, and outcomes, 1995-2004. The American journal of gastroenterology 2008; 103:313-322
  25. Boronikolos GC, Menge BA, Schenker N, Breuer TG, Otte JM, Heckermann S, Schliess F, Meier JJ. Upper gastrointestinal motility and symptoms in individuals with diabetes, prediabetes and normal glucose tolerance. Diabetologia 2015; 58:1175-1182
  26. Camilleri M, Parkman HP, Shafi MA, Abell TL, Gerson L, American College of G. Clinical guideline: management of gastroparesis. The American journal of gastroenterology 2013; 108:18-37; quiz 38
  27. Marathe CS, Rayner CK, Jones KL, Horowitz M. Effects of GLP-1 and incretin-based therapies on gastrointestinal motor function. Experimental diabetes research 2011; 2011:279530
  28. Sarna SK. Cyclic motor activity; migrating motor complex: 1985. Gastroenterology 1985; 89:894-913
  29. Lin HC, Kim BH, Elashoff JD, Doty JE, Gu YG, Meyer JH. Gastric emptying of solid food is most potently inhibited by carbohydrate in the canine distal ileum. Gastroenterology 1992; 102:793-801
  30. Rigda RS, Trahair LG, Little TJ, Wu T, Standfield S, Feinle-Bisset C, Rayner CK, Horowitz M, Jones KL. Regional specificity of the gut-incretin response to small intestinal glucose infusion in healthy older subjects. Peptides 2016; 86:126-132
  31. Kashyap P, Farrugia G. Diabetic gastroparesis: what we have learned and had to unlearn in the past 5 years. Gut 2010; 59:1716-1726
  32. Mazzone A, Bernard CE, Strege PR, Beyder A, Galietta LJ, Pasricha PJ, Rae JL, Parkman HP, Linden DR, Szurszewski JH, Ordog T, Gibbons SJ, Farrugia G. Altered expression of Ano1 variants in human diabetic gastroparesis. The Journal of biological chemistry 2011; 286:13393-13403
  33. Jalleh RJ, Jones KL, Rayner CK, Marathe CS, Wu T, Horowitz M. Normal and disordered gastric emptying in diabetes: recent insights into (patho)physiology, management and impact on glycaemic control. Diabetologia 2022; 65:1981-1993
  34. Marathe CS, Horowitz M, Trahair LG, Wishart JM, Bound M, Lange K, Rayner CK, Jones KL. Relationships of Early And Late Glycemic Responses With Gastric Emptying During An Oral Glucose Tolerance Test. J Clin Endocrinol Metab 2015; 100:3565-3571
  35. Jalleh RJ, Wu T, Jones KL, Rayner CK, Horowitz M, Marathe CS. Relationships of Glucose, GLP-1, and Insulin Secretion With Gastric Emptying After a 75-g Glucose Load in Type 2 Diabetes. J Clin Endocrinol Metab 2022; 107:e3850-e3856
  36. Marathe CS, Rayner CK, Lange K, Bound M, Wishart J, Jones KL, Kahn SE, Horowitz M. Relationships of the early insulin secretory response and oral disposition index with gastric emptying in subjects with normal glucose tolerance. Physiological reports 2017; 5
  37. Abdul-Ghani MA, DeFronzo RA. Plasma glucose concentration and prediction of future risk of type 2 diabetes. Diabetes Care 2009; 32 Suppl 2:S194-198
  38. Gonlachanvit S, Hsu CW, Boden GH, Knight LC, Maurer AH, Fisher RS, Parkman HP. Effect of altering gastric emptying on postprandial plasma glucose concentrations following a physiologic meal in type-II diabetic patients. Digestive diseases and sciences 2003; 48:488-497
  39. Marathe CS, Rayner CK, Bound M, Checklin H, Standfield S, Wishart J, Lange K, Jones KL, Horowitz M. Small intestinal glucose exposure determines the magnitude of the incretin effect in health and type 2 diabetes. Diabetes 2014; 63:2668-2675
  40. Schvarcz E, Palmer M, Aman J, Horowitz M, Stridsberg M, Berne C. Physiological hyperglycemia slows gastric emptying in normal subjects and patients with insulin-dependent diabetes mellitus. Gastroenterology 1997; 113:60-66
  41. Aigner L, Becker B, Gerken S, Quast DR, Meier JJ, Nauck MA. Day-to-Day Variations in Fasting Plasma Glucose Do Not Influence Gastric Emptying in Subjects With Type 1 Diabetes. Diabetes Care 2021; 44:479-488
  42. Russo A, Stevens JE, Chen R, Gentilcore D, Burnet R, Horowitz M, Jones KL. Insulin-induced hypoglycemia accelerates gastric emptying of solids and liquids in long-standing type 1 diabetes. The Journal of clinical endocrinology and metabolism 2005; 90:4489-4495
  43. Jones KL, Russo A, Stevens JE, Wishart JM, Berry MK, Horowitz M. Predictors of delayed gastric emptying in diabetes. Diabetes care 2001; 24:1264-1269
  44. Horowitz M, Jones KL, Rayner CK, Read NW. 'Gastric' hypoglycaemia--an important concept in diabetes management. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society 2006; 18:405-407
  45. Ishii M, Nakamura T, Kasai F, Onuma T, Baba T, Takebe K. Altered postprandial insulin requirement in IDDM patients with gastroparesis. Diabetes care 1994; 17:901-903
  46. Ceriello A, Monnier L, Owens D. Glycaemic variability in diabetes: clinical and therapeutic implications. The lancet Diabetes & endocrinology 2019; 7:221-230
  47. Olausson EA, Storsrud S, Grundin H, Isaksson M, Attvall S, Simren M. A small particle size diet reduces upper gastrointestinal symptoms in patients with diabetic gastroparesis: a randomized controlled trial. The American journal of gastroenterology 2014; 109:375-385
  48. Tornblom H. Treatment of gastrointestinal autonomic neuropathy. Diabetologia 2016; 59:409-413
  49. Calles-Escandon J, Koch KL, Hasler WL, Van Natta ML, Pasricha PJ, Tonascia J, Parkman HP, Hamilton F, Herman WH, Basina M, Buckingham B, Earle K, Kirkeby K, Hairston K, Bright T, Rothberg AE, Kraftson AT, Siraj ES, Subauste A, Lee LA, Abell TL, McCallum RW, Sarosiek I, Nguyen L, Fass R, Snape WJ, Vaughn IA, Miriel LA, Farrugia G, Consortium NGCR. Glucose sensor-augmented continuous subcutaneous insulin infusion in patients with diabetic gastroparesis: An open-label pilot prospective study. PLoS One 2018; 13:e0194759
  50. Samsom M, Szarka LA, Camilleri M, Vella A, Zinsmeister AR, Rizza RA. Pramlintide, an amylin analog, selectively delays gastric emptying: potential role of vagal inhibition. American journal of physiology Gastrointestinal and liver physiology 2000; 278:G946-951
  51. Parkman HP, Carlson MR, Gonyer D. Metoclopramide nasal spray is effective in symptoms of gastroparesis in diabetics compared to conventional oral tablet. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society 2014; 26:521-528
  52. Patterson D, Abell T, Rothstein R, Koch K, Barnett J. A double-blind multicenter comparison of domperidone and metoclopramide in the treatment of diabetic patients with symptoms of gastroparesis. The American journal of gastroenterology 1999; 94:1230-1234
  53. Urbain JL, Vantrappen G, Janssens J, Van Cutsem E, Peeters T, De Roo M. Intravenous erythromycin dramatically accelerates gastric emptying in gastroparesis diabeticorum and normals and abolishes the emptying discrimination between solids and liquids. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 1990; 31:1490-1493
  54. Griffith DP, McNally AT, Battey CH, Forte SS, Cacciatore AM, Szeszycki EE, Bergman GF, Furr CE, Murphy FB, Galloway JR, Ziegler TR. Intravenous erythromycin facilitates bedside placement of postpyloric feeding tubes in critically ill adults: a double-blind, randomized, placebo-controlled study. Critical care medicine 2003; 31:39-44
  55. Jones KL, Berry M, Kong MF, Kwiatek MA, Samsom M, Horowitz M. Hyperglycemia attenuates the gastrokinetic effect of erythromycin and affects the perception of postprandial hunger in normal subjects. Diabetes care 1999; 22:339-344
  56. Camilleri M, Acosta A. Emerging treatments in Neurogastroenterology: relamorelin: a novel gastrocolokinetic synthetic ghrelin agonist. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society 2015; 27:324-332
  57. Lembo A, Camilleri M, McCallum R, Sastre R, Breton C, Spence S, White J, Currie M, Gottesdiener K, Stoner E, Group RMT. Relamorelin Reduces Vomiting Frequency and Severity and Accelerates Gastric Emptying in Adults With Diabetic Gastroparesis. Gastroenterology 2016; 151:87-96 e86
  58. Manini ML, Camilleri M, Goldberg M, Sweetser S, McKinzie S, Burton D, Wong S, Kitt MM, Li YP, Zinsmeister AR. Effects of Velusetrag (TD-5108) on gastrointestinal transit and bowel function in health and pharmacokinetics in health and constipation. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society 2010; 22:42-49, e47-48
  59. Carbone F, Van den Houte K, Clevers E, Andrews CN, Papathanasopoulos A, Holvoet L, Van Oudenhove L, Caenepeel P, Arts J, Vanuytsel T, Tack J. Prucalopride in Gastroparesis: A Randomized Placebo-Controlled Crossover Study. The American journal of gastroenterology 2019; 114:1265-1274
  60. Abell T, McCallum R, Hocking M, Koch K, Abrahamsson H, Leblanc I, Lindberg G, Konturek J, Nowak T, Quigley EM, Tougas G, Starkebaum W. Gastric electrical stimulation for medically refractory gastroparesis. Gastroenterology 2003; 125:421-428
  61. McCallum RW, Snape W, Brody F, Wo J, Parkman HP, Nowak T. Gastric electrical stimulation with Enterra therapy improves symptoms from diabetic gastroparesis in a prospective study. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association 2010; 8:947-954; quiz e116
  62. Ducrotte P, Coffin B, Bonaz B, Fontaine P, Des Varannes SB, Zerbib F, Caiazzo R, Charles Grimaud J, Mion F, Hadjadj S, Elie Valensi P, Vuitton L, Charpentier G, Ropert A, Altwegg R, Pouderoux P, Dorval E, Dapoigny M, Duboc H, Yves Benhamou P, Schmidt A, Donnadieu N, Gourcerol G, Guerci B, Group Er. Gastric Electrical Stimulation Reduces Refractory Vomiting in a Randomized Cross-Over Trial. Gastroenterology 2019;
  63. Pazzi P, Scagliarini R, Gamberini S, Pezzoli A. Review article: gall-bladder motor function in diabetes mellitus. Alimentary pharmacology & therapeutics 2000; 14 Suppl 2:62-65
  64. Gielkens HA, van Oostayen JA, Frolich M, Biemond I, Lamers CB, Masclee AA. Dose-dependent inhibition of postprandial gallbladder motility and plasma hormone secretion during acute hyperglycemia. Scandinavian journal of gastroenterology 1998; 33:1074-1079
  65. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB, Committee LS, Investigators LT. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. The New England journal of medicine 2016; 375:311-322
  66. Gether IM, Nexoe-Larsen C, Knop FK. New Avenues in the Regulation of Gallbladder Motility-Implications for the Use of Glucagon-Like Peptide-Derived Drugs. The Journal of clinical endocrinology and metabolism 2019; 104:2463-2472
  67. Pineda O, Maydon HG, Amado M, Sepulveda EM, Guilbert L, Espinosa O, Zerrweck C. A Prospective Study of the Conservative Management of Asymptomatic Preoperative and Postoperative Gallbladder Disease in Bariatric Surgery. Obes Surg 2017; 27:148-153
  68. Cogliandro RF, Rizzoli G, Bellacosa L, De Giorgio R, Cremon C, Barbara G, Stanghellini V. Is gastroparesis a gastric disease? Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society 2019; 31:e13562
  69. Russo A, Fraser R, Horowitz M. The effect of acute hyperglycaemia on small intestinal motility in normal subjects. Diabetologia 1996; 39:984-989
  70. Adachi T, Mori C, Sakurai K, Shihara N, Tsuda K, Yasuda K. Morphological changes and increased sucrase and isomaltase activity in small intestines of insulin-deficient and type 2 diabetic rats. Endocrine journal 2003; 50:271-279
  71. Rayner CK, Schwartz MP, van Dam PS, Renooij W, de Smet M, Horowitz M, Smout AJ, Samsom M. Small intestinal glucose absorption and duodenal motility in type 1 diabetes mellitus. The American journal of gastroenterology 2002; 97:3123-3130
  72. Chaikomin R, Wu KL, Doran S, Jones KL, Smout AJ, Renooij W, Holloway RH, Meyer JH, Horowitz M, Rayner CK. Concurrent duodenal manometric and impedance recording to evaluate the effects of hyoscine on motility and flow events, glucose absorption, and incretin release. Am J Physiol Gastrointest Liver Physiol 2007; 292:G1099-1104
  73. Lysy J, Israeli E, Goldin E. The prevalence of chronic diarrhea among diabetic patients. The American journal of gastroenterology 1999; 94:2165-2170
  74. Sommers T, Mitsuhashi S, Singh P, Hirsch W, Katon J, Ballou S, Rangan V, Cheng V, Friedlander D, Iturrino J, Lembo A, Nee J. Prevalence of Chronic Constipation and Chronic Diarrhea in Diabetic Individuals in the United States. The American journal of gastroenterology 2019; 114:135-142
  75. Prasad VG, Abraham P. Management of chronic constipation in patients with diabetes mellitus. Indian J Gastroenterol 2017; 36:11-22
  76. Rao SS, Camilleri M, Hasler WL, Maurer AH, Parkman HP, Saad R, Scott MS, Simren M, Soffer E, Szarka L. Evaluation of gastrointestinal transit in clinical practice: position paper of the American and European Neurogastroenterology and Motility Societies. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society 2011; 23:8-23
  77. Panesar PS, Kumari S. Lactulose: production, purification and potential applications. Biotechnol Adv 2011; 29:940-948
  78. Christie J, Shroff S, Shahnavaz N, Carter LA, Harrison MS, Dietz-Lindo KA, Hanfelt J, Srinivasan S. A Randomized, Double-Blind, Placebo-Controlled Trial to Examine the Effectiveness of Lubiprostone on Constipation Symptoms and Colon Transit Time in Diabetic Patients. The American journal of gastroenterology 2017; 112:356-364
  79. Bharucha AE, Low P, Camilleri M, Veil E, Burton D, Kudva Y, Shah P, Gehrking T, Zinsmeister AR. A randomised controlled study of the effect of cholinesterase inhibition on colon function in patients with diabetes mellitus and constipation. Gut 2013; 62:708-715
  80. Celik AF, Osar Z, Damci T, Pamuk ON, Pamuk GE, Ilkova H. How important are the disturbances of lower gastrointestinal bowel habits in diabetic outpatients? The American journal of gastroenterology 2001; 96:1314-1316
  81. Vinik AI, Erbas T. Diabetic autonomic neuropathy. Handb Clin Neurol 2013; 117:279-294
  82. Eliakim-Raz N, Fishman G, Yahav D, Goldberg E, Stein GY, Zvi HB, Barsheshet A, Bishara J. Predicting Clostridium difficile infection in diabetic patients and the effect of metformin therapy: a retrospective, case-control study. Eur J Clin Microbiol Infect Dis 2015; 34:1201-1205
  83. Yuhara H, Steinmaus C, Cohen SE, Corley DA, Tei Y, Buffler PA. Is diabetes mellitus an independent risk factor for colon cancer and rectal cancer? The American journal of gastroenterology 2011; 106:1911-1921; quiz 1922
  84. Franciosi M, Lucisano G, Lapice E, Strippoli GF, Pellegrini F, Nicolucci A. Metformin therapy and risk of cancer in patients with type 2 diabetes: systematic review. PLoS One 2013; 8:e71583
  85. Epanomeritakis E, Koutsoumbi P, Tsiaoussis I, Ganotakis E, Vlata M, Vassilakis JS, Xynos E. Impairment of anorectal function in diabetes mellitus parallels duration of disease. Dis Colon Rectum 1999; 42:1394-1400
  86. Sun WM, Katsinelos P, Horowitz M, Read NW. Disturbances in anorectal function in patients with diabetes mellitus and faecal incontinence. Eur J Gastroenterol Hepatol 1996; 8:1007-1012
  87. Schiller LR, Santa Ana CA, Schmulen AC, Hendler RS, Harford WV, Fordtran JS. Pathogenesis of fecal incontinence in diabetes mellitus: evidence for internal-anal-sphincter dysfunction. The New England journal of medicine 1982; 307:1666-1671
  88. Norton C, Thomas L, Hill J, Guideline Development G. Management of faecal incontinence in adults: summary of NICE guidance. BMJ 2007; 334:1370-1371
  89. Carrington EV, Scott SM, Bharucha A, Mion F, Remes-Troche JM, Malcolm A, Heinrich H, Fox M, Rao SS, International Anorectal Physiology Working G, the International Working Group for Disorders of Gastrointestinal M, Function. Expert consensus document: Advances in the evaluation of anorectal function. Nature reviews Gastroenterology & hepatology 2018; 15:309-323
  90. Engel BT, Nikoomanesh P, Schuster MM. Operant conditioning of rectosphincteric responses in the treatment of fecal incontinence. The New England journal of medicine 1974; 290:646-649
  91. Narayanan SP, Bharucha AE. A Practical Guide to Biofeedback Therapy for Pelvic Floor Disorders. Curr Gastroenterol Rep 2019; 21:21
  92. Hanefeld M. The role of acarbose in the treatment of non-insulin-dependent diabetes mellitus. J Diabetes Complications 1998; 12:228-237
  93. Sansome DJ, Xie C, Veedfald S, Horowitz M, Rayner CK, Wu T. Mechanism of glucose-lowering by metformin in type 2 diabetes: Role of bile acids. Diabetes Obes Metab 2020; 22:141-148
  94. Bonnet F, Scheen A. Understanding and overcoming metformin gastrointestinal intolerance. Diabetes Obes Metab 2017; 19:473-481
  95. Linnebjerg H, Park S, Kothare PA, Trautmann ME, Mace K, Fineman M, Wilding I, Nauck M, Horowitz M. Effect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetes. Regul Pept 2008; 151:123-129
  96. Deane AM, Nguyen NQ, Stevens JE, Fraser RJ, Holloway RH, Besanko LK, Burgstad C, Jones KL, Chapman MJ, Rayner CK, Horowitz M. Endogenous glucagon-like peptide-1 slows gastric emptying in healthy subjects, attenuating postprandial glycemia. J Clin Endocrinol Metab 2010; 95:215-221
  97. Little TJ, Pilichiewicz AN, Russo A, Phillips L, Jones KL, Nauck MA, Wishart J, Horowitz M, Feinle-Bisset C. Effects of intravenous glucagon-like peptide-1 on gastric emptying and intragastric distribution in healthy subjects: relationships with postprandial glycemic and insulinemic responses. J Clin Endocrinol Metab 2006; 91:1916-1923
  98. Hellström PM, Näslund E, Edholm T, Schmidt PT, Kristensen J, Theodorsson E, Holst JJ, Efendic S. GLP-1 suppresses gastrointestinal motility and inhibits the migrating motor complex in healthy subjects and patients with irritable bowel syndrome. Neurogastroenterol Motil 2008; 20:649-659
  99. Nakamori H, Iida K, Hashitani H. Mechanisms underlying the prokinetic effects of endogenous glucagon-like peptide-1 in the rat proximal colon. Am J Physiol Gastrointest Liver Physiol 2021; 321:G617-g627
  100. Nauck MA, Quast DR, Wefers J, Meier JJ. GLP-1 receptor agonists in the treatment of type 2 diabetes - state-of-the-art. Mol Metab 2021; 46:101102
  101. Bettge K, Kahle M, Abd El Aziz MS, Meier JJ, Nauck MA. Occurrence of nausea, vomiting and diarrhoea reported as adverse events in clinical trials studying glucagon-like peptide-1 receptor agonists: A systematic analysis of published clinical trials. Diabetes Obes Metab 2017; 19:336-347
  102. Maselli D, Atieh J, Clark MM, Eckert D, Taylor A, Carlson P, Burton DD, Busciglio I, Harmsen WS, Vella A, Acosta A, Camilleri M. Effects of liraglutide on gastrointestinal functions and weight in obesity: A randomized clinical and pharmacogenomic trial. Obesity (Silver Spring) 2022; 30:1608-1620
  103. Jones KL, Huynh LQ, Hatzinikolas S, Rigda RS, Phillips LK, Pham HT, Marathe CS, Wu T, Malbert CH, Stevens JE, Lange K, Rayner CK, Horowitz M. Exenatide once weekly slows gastric emptying of solids and liquids in healthy, overweight people at steady-state concentrations. Diabetes Obes Metab 2020; 22:788-797
  104. Jensterle M, Ferjan S, Ležaič L, Sočan A, Goričar K, Zaletel K, Janez A. Semaglutide delays 4-hour gastric emptying in women with polycystic ovary syndrome and obesity. Diabetes Obes Metab 2023; 25:975-984
  105. Horowitz M, Rayner CK, Marathe CS, Wu T, Jones KL. Glucagon-like peptide-1 receptor agonists and the appropriate measurement of gastric emptying. Diabetes Obes Metab 2020; 22:2504-2506
  106. Kalas MA, Galura GM, McCallum RW. Medication-Induced Gastroparesis: A Case Report. J Investig Med High Impact Case Rep 2021; 9:23247096211051919
  107. Dhatariya K, Levy N, Russon K, Patel A, Frank C, Mustafa O, Newland-Jones P, Rayman G, Tinsley S, Dhesi J. Perioperative use of glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors for diabetes mellitus. Br J Anaesth 2024;
  108. Toouli J, Biankin AV, Oliver MR, Pearce CB, Wilson JS, Wray NH, Australasian Pancreatic C. Management of pancreatic exocrine insufficiency: Australasian Pancreatic Club recommendations. The Medical journal of Australia 2010; 193:461-467
  109. Piciucchi M, Capurso G, Archibugi L, Delle Fave MM, Capasso M, Delle Fave G. Exocrine pancreatic insufficiency in diabetic patients: prevalence, mechanisms, and treatment. Int J Endocrinol 2015; 2015:595649
  110. Riceman MD, Bound M, Grivell J, Hatzinikolas S, Piotto S, Nguyen NQ, Jones KL, Horowitz M, Rayner CK, Phillips LK. The prevalence and impact of low faecal elastase-1 in community-based patients with type 2 diabetes. Diabetes research and clinical practice 2019; 156:107822
  111. Altay M. Which factors determine exocrine pancreatic dysfunction in diabetes mellitus? World J Gastroenterol 2019; 25:2699-2705

Understanding Ethical Dilemmas in Pediatric Lipidology- Genetic Testing in Youth

ABSTRACT

 

Over the past 25 years there has been an increasing focus on early identification of individuals at-risk of premature cardiovascular disease (CVD), with the goal of improving outcomes and reducing premature CVD-related events such as myocardial infarction and stroke. In 2011, a National Heart, Lung and Blood Institute (NHLBI) Expert Panel recommended universal cholesterol screening of all children, irrespective of health status and family history, beginning at 10 years-of-age (range 9-11) and, if normal, repeated once between 17 and 20 years-of-age (1). Children found to have significant hypercholesterolemia are encouraged to adopt a heart-healthy lifestyle and, when appropriate, offered treatment with lipid-lowering medication, starting at 8 years-of-age and older. Research studies have convincingly demonstrated the safety and effectiveness of lipid-lowering medications in reducing risk and improving outcomes in adults, providing indirect support for universally cholesterol screening of children. Data from individuals with familial hypercholesterolemia (FH), treated for 20 years with pravastatin starting at a young age, have shown no adverse effects of growth, development, or reproductive function during adulthood. Shared decision-making in this population, however, is complex. Unlike most adults who are capable of making informed healthcare decisions, children have a wide range of developmentally-related intellectual and cognitive function, creating unique challenges in their ability to 1) understand long-term risk and benefit; and 2) make informed decisions regarding testing and medical management. In addition, some children have mental health and developmental disabilities that limit their cognitive abilities and judgement. Furthermore, legal guardians have the moral responsibility and legal right to make decisions on behalf of a minor.  In this article, we will discuss 1) privacy, discrimination, and the legal rights of children; 2) ethical considerations and concerns and 3) recommendations for clinicians when providing medical care of children with disorders of lipid and lipoprotein metabolism.

 

OVERVIEW OF LIPID AND LIPOPROTEIN DISORDERS IN YOUTH

 

Children with abnormal levels of lipids and lipoproteins are generally identified as result of targeted, universal or occasionally, coincidental testing.  Current recommendations for lipid screening of children are listed below.

 

  1. Targeted screening in all children ≥2 years of age in whom:
    1. One or both biologic parents are known to have hypercholesterolemia or are receiving lipid-lowering medications
    2. Who have a family history of premature cardiovascular disease (men <55 years of age and women <65 years of age)
    3. Whose family history is unknown (e.g., children who were adopted)
  2. Universal screening of all children 10 years of age (range 9-11), regardless of general health or the presence/absence of CVD risk factors. If normal, repeat screening is recommended at 17-20 years-of-age.

 

Since hypercholesterolemia is often caused by an underlying genetic mutation, such as in FH, cascade screening of biologic relatives is also recommended.  Cascade screening involves systematic testing of all first-degree relatives (parents and siblings) of a child with FH, followed by testing of second- and third-degree relatives if any of the first-degree relatives are affected. The most practical approach to cascade screening is biochemical testing of cholesterol, which is inexpensive, readily available and can be performed without the need for fasting. However, up to 25% of family members may be misdiagnosed as being either affected or unaffected when screening is based on cholesterol levels alone. Testing for a known genetic mutation in the family combined with fasting or non-fasting LDL-C levels will yield the most definitive information. While helpful if known, the child’s family history is often unknown, incomplete, or inaccurate. Reliance upon family history alone fails to identify as many as 30-60% of children with significant hypercholesterolemia. For additional information see the Endotext chapters entitled “Guidelines for Screening, Prevention, Diagnosis, and Treatment of Dyslipidemia in Children and Adolescents” and “Principles of Genetic Testing for Dyslipidemia in Children”.

 

Abnormalities of lipids and lipoproteins in youth may be caused by genetic mutations, acquired conditions, or both.  Those with acquired conditions, such as obesity and insulin resistance, are encouraged to adopt a heart-healthy lifestyle, which includes a low-fat,   calorically appropriate carbohydrate diet, weight loss if overweight or obese, participation in 30-60 minutes of moderate-to-vigorous physical activity per day and smoking avoidance or cessation.  Those suspected of having a genetic mutation are generally diagnosed based upon clinical criteria with or without genetic testing.

 

Genetic mutations that cause lipid and lipoprotein abnormalities vary depending upon the mode of inheritance (autosomal co-dominant vs autosomal recessive), the type of mutation present (slice vs missense), the number of genes involved (monogenic vs polygenic) and their phenotypic expression. When a genetic mutation is present, its expression may potentially be modified by other gene abnormalities (often small effect mutations) and environmental factors (e.g., obesity, insulin resistance, medications). For additional information see the Endotext chapter entitled “Genetics and Dyslipidemia”.

 

Early identification and treatment of children with clinically suspected or genetically confirmed FH has become increasingly common.  However long-term outcome studies demonstrating the safety and efficacy of this approach are lacking. Since lifestyles and therapeutic options are likely to change over the extended period of time that would be necessary to reach “hard” end points in children with FH, such as myocardial infarction and stroke, outcome studies are unlikely to be forthcoming.  Given the significant benefit statins have shown in reducing CVD-related mortality in adults, it has been suggested that withholding effective treatment in moderate-to-high risk children would be unethical (2). For additional information see the Endotext chapter entitled “Familial Hypercholesterolemia”.

 

A novel approach has been suggested to potentially lower costs and avoid prolonged exposure of at-risk children to lipid-lowering medication, while offering timely and presumably effective intervention.  Rather than continuous treatment implemented at an early age, Robinson and Gidding proposed intermittent lipid-lowering medication guided by noninvasive measures of atherosclerosis, such as carotid intima-media thickness (3). As with conventional approaches, the goal of such therapy would be regression of atherosclerotic lesions, with retreatment periodically throughout adulthood as needed.  While intriguing, the benefits of this recommendation have not been proven.

 

To date recommendations for early identification and treatment of children with hypercholesterolemia have focused primary on the potential benefits.  Fortunately, no significant physical or psychological harms have been shown in children who have undergone early screening and treatment. However, healthcare providers who advocate screening, genetic testing and treatment of children should carefully consider potential ethical issues, including the rights of the child to participate in clinical decision-making, the presumed benefits to the child and the family, as well as potential harms.

 

 

Over the last 50 years, in the U.S. Congress has passed a variety of laws to assure the privacy of an individual’s health information and eliminate discrimination based upon an individual’s health status. While most clinicians have an awareness of these laws, it is unclear how clinicians use this information in clinical decision-making, particularly as it relates to the current or future interests of the child.

 

Privacy

 

In 1996, Congress passed the Health Insurance Portability and Accountability Act or HIPAA. This law mandates the protection and confidential handling of protected health information, including genetic information. Furthermore, HIPAA states that genetic information in the absence of a diagnosis (e.g., predictive genetic test results) cannot be considered a pre-existing condition. Since children with heterozygous FH are rarely affected by their hypercholesterolemia during childhood, genetic testing would be considered “predictive” of adult-onset disease.  Children found to have a pathogenic or presumed pathogenic mutation, therefore, are afforded privacy under HIPPA and are not consider to have a pre-existing condition.

 

The Genetic Information Nondiscrimination Act (GINA), passed in 2008, adds to HIPPA by prohibiting health insurers and employers from asking or requiring a person to take a genetic test and using genetic information in 1) setting insurance rates and 2) making employment decisions.

 

Discrimination and Pre-existing Medical Conditions

 

Prior to 2014, insurance companies based eligibility for and the cost of health insurance on the presence or absence of pre-existing medical conditions.  A pre-existing condition is typically one for which an individual has received treatment or a diagnosis before being enrolled in a health plan.  Because they were determined by insurance providers, criteria defining pre-existing conditions varied widely. This meant that when applying for health insurance individuals, including children, previously diagnosed with and/or treated for hypercholesterolemia were considered to have a pre-existing condition.

 

Since 2014, with the passage of the Affordable Care Act, insurance companies can no longer deny coverage or discriminate against individuals due to a pre-existing condition. Nor can individuals be charged significantly higher premiums, subjected to an extended waiting period, or have their benefits curtailed or coverage withdrawn because of a pre-existing condition.  However, this protection does not extend to an individual’s ability to obtain nor the rates charged for life, disability, and long-term care insurance.

 

Despite these reassurances, in some cases exemptions may apply, particularly for members of the United States military, veterans obtaining healthcare through the Veterans Administration (VA), and individuals who receive services through the Indian Health Service.

 

Children’s Rights

 

A child’s rights can be considered in two parts 1) nurturance rights, i.e., the right to care and protection and 2) self-determination rights, i.e., the right to have some measure of control over their own lives.  Historically, society has focused on the former. Increasingly there is a growing emphasis on shared decision-making in medicine that recognizes children have the right to take an active part in many of the decisions regarding their own lives. While such efforts are commendable, the ability of children to become actively and willfully involved in the decision process is complicated by normal, and sometimes abnormal, growth and development. This raises an important question about a child’s ability to understand their rights in a reasonable and meaningful way (4). It also assumes that healthcare providers are trained, capable of and willing to provide developmentally-appropriate information to children in a comprehendible and non-threatening way.

 

In the 1980s, Melton (5, 6) suggested that children progress through three distinct stage-like levels of reasoning about rights: Level 1, children exhibit an egocentric orientation where they perceive rights in terms of privileges that are bestowed or withdrawn on the whims of an authority figure. Level 2 children see rights as being based on fairness, maintaining social order and obeying rules. Finally, in Level 3 rights are seen in terms of abstract universal principles. Subsequent models favored the gradual acquisition of context specific knowledge (7-9).  When and how well a child progresses from limited to abstract reasoning presents challenges for physicians who strive to involve children in decisions regarding early screening and intervention for CVD risk prevention.

 

MIGHT EARLY DIAGNOSIS AND TREATMENT OF HYPERCHOLESTEROLEMIA COMPROMISE A CHILD’S FUTURE RIGHTS?

 

Laws such as HIPPA, the Affordable Care Act, and GINA protect privacy and prohibit health insurance companies from denying coverage or discriminating against individuals due to a pre-existing condition, including hypercholesterolemia. Nonetheless, current laws do not preclude an individual being denied other forms of coverage, such as life, disability, or long-term care insurance. Furthermore, laws governing privacy, healthcare, and insurance coverage are subject to change over the course of the child's lifetime. This potential vulnerability needs to be considered by clinicians who provide care to children and fully disclosed to the family prior to diagnostic evaluation and treatment of children with hypercholesterolemia. To the extent that they can participate in such conversations, children should be included in the clinical decision-making. The accelerated risk of atherosclerosis beginning in young adults notwithstanding, the urgency of screening and early treatment of children needs to be considered in the context of the child’s overall best interest and, ideally, with their approval.  

 

ETHICAL CONSIDERATIONS AND CONCERNS

 

Since 1953, there has been an impressive increase in new technology and expanded uses of genetic testing and screening. Application of these diagnostic tools in minors has increasingly become commonplace, raising concerns about ethical issues. While pediatric screening and genetic testing are much less common outside of newborn screening, universal screening and increased use of genetic testing has been advocated by many national professional organizations and societies. Justification for such recommendations cite early identification of a child with an underlying genetic abnormality as an opportunity to initiate treatment that may prevent or reduce morbidity or mortality.

 

Over the past 50 years, genetic testing has increasingly played an important role in helping to understand the basis of many disorders of lipid and lipoprotein metabolism, identifying those who are affected and aiding our understanding of an individual’s risk. While only a minority of individuals with hypercholesterolemia who undergo genetic testing are found to have a pathogenic mutation, epidemiologic and Medallion randomization studies suggest these individuals are at significantly higher risk of premature ASCVD-related morbidity and mortality than the general population. 

 

Genetic testing of an asymptomatic child based upon an abnormal blood test and/or positive family history for a specific genetic condition, such as FH, has also been proposed, particularly if early treatment may affect future morbidity or mortality.  Some genetic tests can reasonability predict disease which only manifest in adults. 

 

Ultimately, decisions about whether to offer genetic testing and screening should be driven by the best interest of the child. This, perhaps, is best determined by a thoughtful discussion between the child’s healthcare provider, the parents, and, when appropriate, the child.  Current recommendations and guidelines suggest early intervention to achieve the best outcomes. Yet, there is no clear definition as to the optimum age at which intervention should be recommended, nor clear understanding about a child’s ability to understand and make a rational decision regarding testing and/or treatment.

 

The genetic testing of children raises specific considerations. Because of the need to respect a children's rights, caution has been advised in performing genetic tests during childhood. Newborn genetic testing is now ubiquitous, yet it is not always seen as routine for older children despite specific indications. Testing for drug responsiveness or disease susceptibility is supported by the ethical principle of beneficence when the benefit/risk ratio is in favor of discovering these results during childhood. Possible harms are seen when such knowledge may impact a child negatively, or foreclose future autonomy about the decision to accept the consequences of such testing. Therefore, there is a difference between genetic confirmation in symptomatic children, and that of pre-symptomatic children in which the benefit may accrue later, but the risks may occur in childhood. Such immediate risks potentially include stigmatization by the disease, depression, or decreased self-esteem. Conversely, altered family dynamics may result in parental favoritism, and survivor's guilt in siblings who test negative. This limitation on future autonomy is not confined to just refusing or allowing an adult decision for testing, but also dealing with the impact on future employment, education, and social relationships when the diagnosis is made at an early age.

 

Tests which help diagnose an ongoing, treatable condition that could affect current and future manifestations and complications clearly can be in the child's best interest. However, when a child is asymptomatic and the disorder is late-onset, it is no longer obvious that such a diagnosis during childhood is in the child's best interest. Therefore, it is advised the children only undergo genetic testing when there is immediate medical benefit in childhood, either through diagnosis and treatment of a disease manifesting in the pediatric age range, or a disease whose prevention is possible and should not be delayed. Under these circumstances, informed decision-making is essential, with parental permission being linked to the child's assent whenever possible.

 

CHOLESTEROL SCREENING AND TREATMENT

 

Currently, universally cholesterol testing is recommended for all children in the U.S., starting at 10 years-of-age (range 9-11). The primary purpose of cholesterol screening is to identify individuals with familial hypercholesterolemia.  For those found to have a significant elevation of cholesterol a low-fat diet is recommended. Lipid-lowering medications, such as a statin, are recommended for children with a persistently elevated LDL-C, starting at approximately 8-10 years-of-age. 

 

Genetic Testing

 

Genetic testing of all children suspected of having FH has been recommended (10). The purported benefits of genetic testing are 1) to assist in clinical decision-making regarding the need for lipid-lowering medication, 2) to help determine the appropriate on-treatment goal of LCL cholesterol; and 3) facilitate cascade screening of biologic relatives.

 

To help better understand the complexities of genetic testing and provide guidance, in 2013 both the American Academy Pediatrics (AAP) and the American College of Medical Genetics (ACMG) published recommendations for genetic testing of children. These guidelines are particularly relevant for those providing care for children with lipid and lipoprotein disorders since, with the exception of homozygous disease, children with heterozygous FH are asymptomatic. Hence, genetic testing in this unique population would be considered “predictive” of adult disease.

 

However, although there is much emphasis on early screening and genetic testing of children for FH, children have a variety of genetic conditions that affect other lipids and lipoproteins as well, such as triglycerides. The infantile form of lysosomal acid lipase deficiency, for example, is generally fatal in the absence of early diagnosis and enzyme replacement therapy. Thus, biochemical screening and genetic testing in this condition becomes imperative in order to reduce early morbidity and prevent premature mortality. Examples of other conditions in which there is a sense of urgency include familial chylomicronemia syndrome (FCS), cerebrotendinous xanthomatosis (CTX), and homozygous mutations of MTTP (abetalipoproteinemia), APOB (familial hypobetalipoproteinemia), and SAR1 (chylomicron retention disease).  When considering screening and genetic testing of children with lipid and lipoprotein disorders, therefore, “one size” clearly does not fit all circumstances. Clinicians must consider each child and condition as unique, carefully weighing the presumed benefits and potential harms individually, before making diagnostic and therapeutic recommendations.  

 

In deciding whether a child should undergo predictive genetic testing, the AAP and ACMG emphasize that the focus must be on the child’s medical best interest. Both organizations concluded that unless ameliorative interventions are available during childhood, children should not undergo testing for predispositions to adult-onset conditions and clinicians should generally decline to order testing. With the exception of those with homozygous FH, this suggests that children with heterozygous disease could defer treatment until adulthood. There is convincing evidence using noninvasive techniques, however, that early initiation of lipid-lowering medication can significantly reduce subclinical atherosclerosis. It is presumed that as a consequence of early and persistent LDL-cholesterol lowering that ASCVD-related events will be prevented or delayed. Yet proof of improved outcomes is currently limited and generally inferred from adult data.  

 

The AAP and ACMG did recognize that the potential psychosocial benefits and harms to the child and the extended family also need to be carefully considered. Extending consideration beyond the child’s medical best interest not only acknowledges the traditional deference given to parents about how they raise their children, but also recognizes that the interest of a child is embedded in and dependent on the interests of the family unit.

 

Predictive genetic testing for adult-onset conditions generally should be deferred unless an intervention initiated in childhood may reduce morbidity or mortality. In some families, the psychosocial burden of ambiguity may be so great as to justify testing during childhood, particularly when parents and mature adolescents jointly express interest in doing so.

 

AAP AND ACMG RECOMMENDATIONS

 

Genetic testing performed in children can be considered either as diagnostic or predictive (11).

 

  1. Diagnostic Genetic Testing - Is performed on a child with physical, developmental, or behavioral features consistent with a potential genetic syndrome or for pharmacogenetic drug selection and dosing decisions. Medical benefits include the possibility of preventive or therapeutic interventions, decisions about surveillance, the clarification of diagnosis and prognosis, and recurrence risks. If the medical benefits of a test are uncertain, will not be realized until a later time, or do not clearly outweigh the medical risks, the justification for testing is less compelling.

 

  1. Predictive Genetic Testing - Is performed on an asymptomatic child with a positive family history for a specific genetic condition, particularly if early surveillance or treatment may affect morbidity or mortality. When there is uncertainty that the presence of a genetic mutation will give rise to clinical manifestations, testing is referred to as “pre-dispositional” testing. Most predictive genetic testing for adult-onset conditions is pre-dispositional.

 

Recommendations for Genetic Testing of Children

 

  1. General
    1. Decisions about whether to offer genetic testing and screening should be driven by the best interest of the child.
    2. Genetic testing is best offered in the context of genetic counseling.
  2. Diagnostic Testing
    1. In a child with symptoms of a genetic condition:
      1. Parents or guardians should be informed about the risks and benefits of testing, and their permission should be obtained.
      2. Ideally and when appropriate, the assent of the child should be obtained.
    2. When performed for therapeutic purposes:
      1. Pharmacogenetic testing of children is acceptable, with permission of parents or guardians and, when appropriate, the child’s assent.
      2. If a pharmacogenetic test result carries implications beyond drug targeting or dose-responsiveness, the broader implications should be discussed before testing.
    3. Newborn Screening
      1. The AAP and ACMG support the mandatory offering of newborn screening for all children. Parents should have the option of refusing the procedure, and an informed refusal should be respected.
    4. Carrier Testing
      1. The AAP and ACMG do not support routine carrier testing in minors when such testing does not provide health benefits in childhood. This recommendation accords with previous statements supporting the future decisional autonomy of the minor, who will be able to make an informed choice about testing once he or she reaches the age of majority.
      2. For pregnant adolescents or for adolescents considering reproduction, genetic testing and screening should be offered as clinically indicated, and the risks and benefits should be clearly explained.
    5. Predictive Genetic Testing
      1. Parents or guardians may authorize predictive genetic testing for asymptomatic children at risk of childhood onset conditions.
      2. Ideally, the assent of the child should be obtained.
      3. Predictive genetic testing for adult-onset conditions generally should be deferred unless an intervention initiated in childhood may reduce morbidity or mortality.
      4. An exception might be made for families in whom diagnostic uncertainty poses a significant psychosocial burden, particularly when an adolescent and his or her parents concur in their interest in predictive testing.
      5. For ethical and legal reasons, health care providers should be cautious about providing predictive genetic testing to minors without the involvement of their parents or guardians, even if a minor is mature. Results of such tests may have significant medical, psychological, and social implications, not only for the minor, but also for other family members.

 

Potential Benefits and Harms of Predictive Genetic Testing of Children. Adapted from (11)

Medical

 

Benefits

Possibility of evolving therapeutic interventions, targeted surveillance, refinement of prognosis and clarification of diagnosis

Harms

Misdiagnosis to the extent that genotype does not correlate with phenotype, ambiguous results in which a specific phenotype cannot be predicted and use of ineffective or harmful preventive or therapeutic interventions.

Psychosocial

 

Benefits

Reduction of uncertainty and anxiety, the opportunity for psychological adjustment, the ability to make realistic life plans and sharing the information with family members.

Harms

Alteration of self-image, distortion of parental perception of the child, increased anxiety and guilt, altered expectation by self and others, familial stress related to identification of other at-risk family members, difficulty obtaining life and/or disability insurance, and the detection of misattributed parentage.

Reproductive

 

Benefits

Avoiding the birth of a child with genetic disease or having time to prepare for the birth of a child with genetic disease.

Harms

Changing family-planning decisions on the basis of social pressures.

 

It is essential that parents, guardians and maturing minors receive genetic counseling before undergoing predictive testing, which includes a discussion of the limits of genetic knowledge and treatment capabilities as well as the potential for psychological harm, stigmatization, and discrimination (12).

 

If an adolescent declines genetic testing, and the benefits of knowing will not be relevant for years to decades, the adolescent’s decision should be final. If a minor is young or immature, genetic testing should be delayed until the minor can actively participate. 

 

If predictive testing of conditions for which childhood interventions will ameliorate future harm, this may favor early testing. In such cases, parental authority to determine medical treatment supersedes the minor’s preferences with regard to liberty and privacy.

 

CONCLUSION

 

Although recommended for all individuals, including children, with clinically suspected familial hypercholesterolemia, genetic testing should be approached with caution. Parents and, when appropriate, children should be provided with a comprehensive discussion of the pros and cons of genetic testing, and informed about out-of-pocket costs prior to testing.

 

REFERENCES

 

  1. Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents, National Heart, Lung, and Blood Institute. Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: summary report. Pediatrics. 2011;128(Suppl 5):S213-S256. doi:10.1542/peds.2009-2107C
  2. Wilson DP, Gidding SS. Atherosclerosis: Is a cure in sight? J Clin Lipidol. 2015;9(5 Suppl):1. doi:S1933-2874(15)00266-4
  3. Robinson JG, Gidding SS. Curing atherosclerosis should be the next major cardiovascular prevention goal. J Am Coll Cardiol. 2014;63(25 Pt A):2779-2785. doi:S0735-1097(14)02138-X
  4. Ruck MD, Abramovitch R, Keating DP. Children's and adolescents' understanding of rights: balancing nurturance and self-determination. Child Dev. 1998;69(2):404-417.
  5. Melton GB. Children's concepts of their rights. J Clin Child Psychol. 1980;9(3):186-190. doi:10.1080/15374418009532985
  6. Melton GB. Child advocacy: Psychological issues and interventions. Plenum; 1983.
  7. Peterson-Badali M, Abramovitch R. Grade related changes in young people's reasoning about plea decisions. Law Hum Behav. 1993;17(5):537-552.
  8. Saywitz KJ. Children’s conceptions of the legal system: “Court is a place to play basketball”. Ceci J, Ross DF, Toglia MP, eds. Perspectives on children’s testimony. Springer-Verlag; 1989:131-157.
  9. Scott ES, Reppucci D, Woolard JL. Evaluating adolescent decision making in legal contexts. Law Hum Behav. 1995;19(3):221-244.
  10. Sturm AC, Knowles JW, Gidding SS, Ahmad ZS, Ahmed CD, Ballantyne CM, Baum SJ, Bourbon M, Carrié A, Cuchel M, de Ferranti SD, Defesche JC, Freiberger T, Hershberger RE, Hovingh GK, Karayan L, Kastelein JJP, Kindt I, Lane SR, Leigh SE, Linton MF, Mata P, Neal WA, Nordestgaard BG, Santos RD, Harada-Shiba M, Sijbrands EJ, Stitziel NO, Yamashita S, Wilemon KA, Ledbetter DH, Rader DJ; the Familial Hypercholesterolemia Foundation. Clinical genetic testing for familial hypercholesterolemia: JACC Scientific Expert Panel. J Am Coll Cardiol. 2018;72(6):662-680. doi:S0735-1097(18)35065-4
  11. American Academy of Pediatrics Committee on Bioethics, Committee on Genetics, and American College of Medical Genetics and Genomics Social, Ethical, Legal Issues Committee. Ethical and policy issues in genetic testing and screening of children. Pediatrics. 2013;131(3):620-622. doi:10.1542/peds.2012-3680
  12. American Academy of Pediatrics Committee on Genetics. Molecular genetic testing in pediatric practice: A subject review. Pediatrics. 2000;106(6):1494-1497. doi:10.1542/peds.106.6.1494

Cholesterol Lowering Drugs

ABSTRACT

 

There are currently several different classes of drugs available for lowering cholesterol levels. There are currently seven HMG-CoA reductase inhibitors (statins) approved for lowering cholesterol levels and they are the first line drugs for treating cholesterol disorders and can lower LDL-C levels by as much as 60%. Statins also are effective in reducing triglyceride levels in patients with hypertriglyceridemia. Statins lower LDL levels by inhibiting HMG-CoA reductase activity leading to decreases in hepatic cholesterol content resulting in an up-regulation of hepatic LDL receptors, which increases the clearance of LDL. The major side effects are muscle complications and an increased risk of diabetes. The different statins have varying drug interactions. Ezetimibe lowers LDL-C levels by approximately 20% by inhibiting cholesterol absorption by the intestines leading to the decreased delivery of cholesterol to the liver, a decrease in hepatic cholesterol content, and an up-regulation of hepatic LDL receptors. Ezetimibe is very useful as add on therapy when statin therapy is not sufficient or in statin intolerant patients. Ezetimibe has few side effects. Bile acid sequestrants lower LDL-C by10-30% by decreasing the absorption of bile acids in the intestine which decreases the bile acid pool consequently stimulating the synthesis of bile acids from cholesterol leading to a decrease in hepatic cholesterol content and an up-regulation of hepatic LDL receptors. Bile acid sequestrants can be difficult to use as they decrease the absorption of multiple drugs, may increase triglyceride levels, and cause constipation and other GI side effects. They do improve glycemic control in patients with diabetes, which is an additional benefit. PCSK9 inhibitors, either monoclonal antibodies or small interfering RNA, lower LDL-C by 50-60% by decreasing PCSK9, which decreases the degradation of LDL receptors. PCSK9 inhibitors also decrease Lp(a) levels. PCSK9 inhibitors are very useful when maximally tolerated statin therapy do not reduce LDL sufficiently and in statin intolerant patients. PCSK9 inhibitors have very few side effects. Bempedoic acid lowers LDL-C by 15-25% by inhibiting hepatic ATP citrate lyase activity resulting in a decrease in cholesterol synthesis in the liver, a decrease in hepatic cholesterol content, and an up-regulation of LDL receptors. Bempedoic acid is employed in patients who do not reach their LDL-C goals on maximally tolerated statin therapy or in patients who do not tolerate statins. Bempedoic acid is associated with elevations in uric acid levels and gouty attacks. Lomitapide and evinacumab are approved for lowering LDL levels in patients with homozygous familiar hypercholesterolemia, as they are not dependent on LDL receptors for decreasing LDL levels. Lomitapide inhibits microsomal triglyceride transfer protein decreasing the formation of chylomicrons in the intestine and VLDL in the liver. Lomitapide has the potential to cause liver toxicity and therefore they were approved with a risk evaluation and mitigation strategy (REMS) to reduce risk. Evinacumab is a monoclonal antibody that inhibits the activity of angiopoietin-like protein 3 resulting in the increased activity of lipoprotein lipase and endothelial cell lipase resulting in a decrease in LDL-C, HDL-C, and triglyceride levels. Mipomersen, which is no longer available, is a second-generation apolipoprotein anti-sense oligonucleotide that decreases apolipoprotein B synthesis resulting in a reduction in the formation and synthesis of VLDL and was approved for the treatment of homozygous familial hypercholesterolemia.

 

INTRODUCTION

 

This chapter will discuss the currently available drugs for lowering total cholesterol levels, especially LDL-C: statins, ezetimibe, bile acid sequestrants, PCSK9 inhibitors, bempedoic acid, lomitapide, mipomersen, and evinacumab. We will not discuss the effect of lifestyle changes or food additives, such as phytosterols, on LDL-C as this is addressed in the chapter entitled “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” (1). Additionally, we will not discuss guidelines for determining who to treat, how aggressively to treat, or targets of treatment as these issues are discussed in detail in the chapters entitled “Guidelines for the Management of High Blood Cholesterol” and “Approach to the Patient with Dyslipidemia” (2,3).

 

STATINS

 

Introduction

 

In the 1970s Dr. Akira Endo, working at Sankyo, discovered that compounds isolated from fungi inhibited the activity of HMG-CoA reductase, a key enzyme in the synthesis of cholesterol (4). Further studies at Merck led to the development of the first HMG-CoA reductase inhibitor, lovastatin, approved in 1987 for the treatment of hypercholesterolemia (5). There are currently seven HMG-CoA reductase inhibitors (statins) approved in the United States for lowering cholesterol levels. Three statins are derived from fungi (lovastatin, simvastatin, and pravastatin) and four statins are synthesized (atorvastatin, rosuvastatin, fluvastatin, and pitavastatin). Most of these statins are now generic drugs and therefore they are relatively inexpensive. Which particular statin one elects to use may depend on the degree of cholesterol lowering needed and the potential of drug-drug interactions. Statins are the first line drugs for treating elevated cholesterol levels and therefore one of the most widely utilized class of drugs. Statins have revolutionized the field of preventive cardiology and made an important contribution to the reduction in atherosclerotic cardiovascular events.

 

Effect on Statins on Lipid and Lipoprotein Levels

 

The major effect of statins is lowering LDL-C levels. The effect of the various statins at different doses on LDL-C levels is shown in Table 1. As can be seen in Table 1 different statins have varying abilities to lower LDL-C with maximal reductions of approximately 60% seen with rosuvastatin 40mg. Doubling the dose of a statin results in an approximate 6% further decrease in LDL-C levels. The percent reduction in LDL-C levels is similar in patients with high and low starting LDL-C levels but the absolute decrease is greater if the starting LDL-C is high. Because of this profound ability of statins to lower LDL-C levels, treatment with these drugs as monotherapy is often sufficient to lower LDL-C below target levels.

 

Table 1. Approximate Effect of Different Doses of Statins on LDL-C Levels

% LDL Reduction

Simvastatin (Zocor)

Atorvastatin (Lipitor)

Lovastatin (Mevacor)

Pravastatin (Pravachol)

Fluvastatin (Lescol)

Rosuvastatin (Crestor)

Pitavastatin (Livalo)

27

10mg

-

20mg

20mg

40mg

-

-

34

20mg

10mg

40mg

40mg

80mg

-

1mg

41

40mg

20mg

80mg

80mg

-

-

2mg

48

80mg

40mg

-

-

-

10mg

4mg

54

-

80mg

-

-

-

20mg

-

60

-

-

-

-

-

40mg

-

Data modified from package inserts

 

As would be predicted from the effect of statins on LDL-C levels, statins are also very effective in lowering non-HDL-C levels (LDL-C is the major contributor to non-HDL-C levels) (6,7). In addition, statins also lower plasma triglyceride levels (8,9). The ability of statins to lower triglyceride levels correlates with the reduction in LDL-C (9). Statins that are most efficacious in lowering LDL-C are also most efficacious in lowering plasma triglyceride and VLDL-C levels. Notably the percent reduction in plasma triglyceride levels is dependent on the baseline triglyceride levels (9). For example, in patients with normal triglyceride levels (<150mg/dL), simvastatin 80mg per day lowered plasma triglyceride levels by 11%. In contrast, if plasma triglyceride levels were elevated (> 250mg/dL), simvastatin 80mg per day lowered triglyceride levels by 40% (9). In patients with both elevated LDL-C and triglyceride levels statin therapy can be very effective in improving the lipid profile and are therefore the first line class of drugs to treat patients with mixed hyperlipidemia unless the triglyceride levels are markedly elevated (>500-1000mg/dL). As expected, given the ability of statins to lower LDL-C and triglyceride/VLDL levels, statin therapy is very effective in lowering apolipoprotein B levels (6,7).

 

Of note despite the ability of statins to lower LDL-C, non-HDL-C, and apolipoprotein B levels, statins do not lower Lp(a) levels and may even increase levels (10,11). Finally, statins modestly increase HDL-C levels (8,12,13). In most studies HDL-C levels increase between 5-10% with statin therapy. Interestingly, while low dose atorvastatin increases HDL levels similar to other statins at high doses the effect of atorvastatin is blunted with either very modest increases or no change observed (12).

 

Table 2. Effect of Statins on Lipid/Lipoprotein Levels

LDL-C

Decrease

Non-HDL-C

Decrease

Apolipoprotein B

Decrease

Triglycerides

Variable. If TG levels increased will decrease

HDL-C

Small Increase

Lp(a)

No change or small increase

 

Non-Lipid Effects of Statins

 

In addition to effects on lipid metabolism statins also have pleiotropic effects that may not be directly related to alterations in lipid metabolism (14). For example, statins are anti-inflammatory and consistently decrease CRP levels (15). Other pleiotropic effects of statins include anti-proliferative effects, antioxidant properties, anti-thrombosis, improving endothelial dysfunction, and attenuating vascular remodeling (14). Whether these pleiotropic effects contribute to the beneficial effects of statins in preventing cardiovascular disease is uncertain and much of the beneficial effect of statins on cardiovascular disease can be attributed to reductions in lipid levels.

 

Mechanism Accounting for the Statin Induced Lipid Effects

 

Statins are competitive inhibitors of HMG-CoA reductase, which leads to a decrease in cholesterol synthesis in the liver. This inhibition of hepatic cholesterol synthesis results in a decrease in cholesterol in the endoplasmic reticulum resulting in the movement of sterol regulatory element binding proteins (SREBPs) from the endoplasmic reticulum to the golgi where they are cleaved by proteases into active transcription factors (16). The SREBPs then translocate to the nucleus where they increase the expression of a number of genes including HMG-CoA reductase and, most importantly, the LDL receptor (16). The increased expression of HMG-CoA reductase restores hepatic cholesterol synthesis towards normal while the increased expression of the LDL receptor results in an increase in the number of LDL receptors on the plasma membrane of hepatocytes leading to the accelerated clearance of LDL (Figure 1) (16). The increased clearance of LDL accounts for the reduction in plasma LDL-C levels. In patients with a total absence of LDL receptors (Homozygous Familiar Hypercholesterolemia) statin therapy is not very effective in lowering LDL-C levels.

 

Figure 1. Mechanism for the Decrease in LDL Levels

 

In addition to lowering LDL and VLDL levels by accelerating the clearance of lipoproteins some studies have also shown that statins reduce the production and secretion of VLDL particles by the liver (17). This could contribute to the decrease in triglyceride levels. The mechanism by which statins increase HDL-C levels is not clear. The small increase in Lp(a) may be due to increased production as studies have shown that incubating HepG2 hepatocytes with a statin increased the expression of LPA mRNA and apolipoprotein(a) protein (18).

 

Pharmacokinetics and Drug Interactions

 

Statins have different pharmacokinetic properties which can explain clinically important differences in safety and drug interactions (19-22). Most statins are lipophilic except for pravastatin and rosuvastatin, which are hydrophilic. Lipophilic statins can enter cells more easily but the clinical significance of this difference is not clear. Most of the clearance of statins is via the liver and GI tract (19-21). Renal clearance of statins in general is low with atorvastatin having a very low renal clearance making this particular drug the statin of choice in patients with significant renal disease. The half-life of statins varies greatly with lovastatin, pravastatin, simvastatin, and fluvastatin having a short half-life (1-3 hours) while atorvastatin, rosuvastatin, and pitavastatin having a long half-life (19-22). In patient’s intolerant of statins, the use of a long-acting statin every other day or 2 times per week has been employed. Short acting statins are most effective when administered in the evening when HMG-CoA reductase activity is maximal while the efficacy of long-acting statins is equivalent whether given in the AM or PM (23). In patients who prefer to take their statin in the morning one should use a long-acting statin.

 

A key difference between statins is their pathway of metabolism. Simvastatin, lovastatin, and atorvastatin are metabolized by the CYP3A4 enzymes and drugs that affect the CYP3A4 pathway can alter the metabolism of these statins (19-22,24). Fluvastatin is metabolized mainly by CYP2C9 with a small contribution by CYP2C8 (19-21,24). Pitavastatin and rosuvastatin are minimally metabolized by the CYP2C9 pathway (19-21,24). Pravastatin is not metabolized at all via the CYP enzyme system (19-21).

 

Drugs that inhibit CYP3A4 can impede the metabolism of simvastatin, lovastatin, and to a smaller extent atorvastatin resulting in high serum levels of these drugs (19-22,24). These higher levels are associated with adverse effects particularly muscle toxicity. Drugs that inhibit CYP3A4 include intraconazole, ketoconazole, erythromycin, clarithromycin, HIV protease inhibitors (amprenavir, darunavir, fosamprenavir, indinavir, nelfinavir, ritonavir, and saquinavir), amiodarone, diltiazem, verapamil, and cyclosporine (19-22,24). It should be noted that grapefruit juice contains compounds that inhibit CYP3A4 and the consumption of grapefruit juice can significantly increase statin blood levels (25). The inhibition of CYP3A4 by grapefruit juice is dose dependent and increases with the concentration and volume of grapefruit juice ingested. One glass of grapefruit juice everyday can influence the metabolism of statins that are metabolized by the CYP3A4 pathway (25). If a patient requires treatment with a drug that inhibits CYP3A4 the clinician has a number of options to avoid potential drug interactions. One could use a statin that is not metabolized via the CYP3A4 system such as pravastatin or rosuvastatin, one could use an alternative drug to the CYP3A4 inhibitor (for example instead of using erythromycin use azithromycin), or one could temporarily suspend for a short period of time the use of the statin that is metabolized by the CYP3A4 pathway (this is particularly useful when a short course of treatment with an antifungal, antiviral, or antibiotic is required). Drugs that inhibit CYP2C9 do not seem to increase the toxicity of fluvastatin, pitavastatin, or rosuvastatin probably because metabolism via the CYP2C9 pathway is not a dominant pathway.

 

Most statins are transported into the liver and other tissues by organic anion transporting polypeptides, particularly OATP1B1 (19-21,24). Drugs, such as clarithromycin, ritonavir, indinavir, saquinavir, and cyclosporine that inhibit OATP1B1 can increase serum statin levels thereby increasing the risk of statin muscle toxicity (19-21,24). Fluvastatin is the statin that is least affected by OATP1B1 inhibitors. In fact, fluvastatin 40mg per day has been studied in patients receiving renal transplants concomitantly treated with cyclosporine and over a five year study period the risk of myopathy or rhabdomyolysis was not increased in the fluvastatin treated patients compared to those treated with placebo (26).

 

Gemfibrozil inhibits the glucuronidation of statins, which accounts for a significant portion of the metabolism of most statins (24). This can lead to the reduced clearance of statins and elevated blood levels increasing the risk of muscle toxicity (24). The only statin whose metabolism is not altered by gemfibrozil is fluvastatin (24). Notably, fenofibrate, another fibrate that has very similar effects on lipid and lipoprotein levels as gemfibrozil, does not inhibit statin glucuronidation (24). Therefore, in patients on statin therapy who also need treatment with a fibrate one should use fenofibrate and not gemfibrozil in combination with statin therapy. Studies have shown that fenofibrate combined with statins does not significantly increase toxicity (27).

 

There are other drug interactions with statins whose mechanisms are unknown. For example, the lopinavir/ritonavir combination used to treat HIV increases rosuvastatin levels by 2-5-fold and atazanavir/ritonavir increases rosuvastatin levels by 2-6-fold (28-32). Similarly, the tipranavir/ritonavir combination increases rosuvastatin levels 2-fold and atorvastatin levels 8-fold (31). When HIV patients are on these drugs other statins should be used to lower LDL-C levels. The use of statins in patients with HIV is discussed in detail in the Endotext chapter entitled “Lipid Disorders in People with HIV” (33).

 

Thus, despite the excellent safety record of statins, careful attention must be paid to the potential drug-drug interactions. For additional information see Kellick et al (22,24).

 

Effect of Statin Therapy on Clinical Outcomes

 

A large number of studies using a variety of statins in diverse patient populations have shown that statin therapy reduces atherosclerotic cardiovascular disease. The Cholesterol Treatment Trialists have published meta-analyses derived from individual subject data. Their first publication included data from 14 trials with over 90,000 subjects (34). There was a 12% reduction in all-cause mortality in the statin treated subjects, which was mainly due to a 19% reduction in coronary heart disease deaths. Non-vascular causes of death were similar in the statin and placebo groups indicating that statin therapy and lowering LDL-C did not increase the risk of death from other causes such as cancer, respiratory disease, etc. Of particular note there was a 23% decrease in major coronary events per 1 mmol/L (39mg/dL) reduction in LDL-C. Decreases in other vascular outcomes including non-fatal MI, coronary heart disease death, vascular surgery, and stroke were also reduced by 20-25% per 1 mmol/L (39mg/dL) reduction in LDL-C. Additionally, analysis of these studies demonstrated that the greater the reduction in absolute LDL-C levels the greater the decrease in cardiovascular events.  For example, while a 40mg/dL decrease in LDL-C will reduce coronary events by approximately 20%, an 80mg/dL decrease in LDL-C will reduce events by approximately 40%. These results support aggressive lipid lowering with statin therapy.

 

Of note the decrease in the number of events begins to be seen in the first year of therapy indicating that the ability of statins to reduce events occurs relatively quickly and increases over time. The ability of statins to reduce cardiovascular events was seen in a wide diversity of patients including those with and without a history of prior cardiovascular disease, patients over age 65 and younger than age 65, males and females, and patients with and without a history of diabetes or hypertension. Additionally, the beneficial effects of statins were seen regardless of the baseline lipid levels. Subjects with elevated or low LDL-C, HDL-C, or triglyceride levels all had similar decreases in the relative risk of cardiovascular events.

 

A subsequent publication by the Cholesterol Treatment Trialists has focused on five studies with over 39,000 subjects that have compared usual vs. intensive statin therapy (35). It was noted that there was a 0.51mmol/L (20mg/dL) further reduction in LDL-C in the intensively treated subjects. This further decrease in LDL-C resulted in a15% reduction in cardiovascular events. The strong numerical relationship between decreases in LDL-C levels and the reduction in cardiovascular events provides evidence indicating that much of the beneficial effect of statins is accounted for by lipid lowering.

 

In addition, the authors added 7 additional trials to their original comparison of statin treatment vs. placebo for a total of 21 trials with over 129,000 subjects. In this larger cohort a 1mmol/L (39mg/dL) decrease in LDL was associated with a 21% reduction in major cardiovascular events. As seen previously the benefits of statin therapy were seen in a wide variety of subjects including patients older than age 75, obese patients, cigarette smokers, patients with decreased renal function, and patients with low and high HDL-C levels. Additionally, a reduction of cardiovascular events with statin therapy was seen regardless of baseline LDL-C levels.

 

A more recent meta-analysis by the Cholesterol Treatment Trialists examined the effect of statins in 27 trials that included 46,675 women and 127,474 men (36). They found that statin therapy was similarly effective in reducing cardiovascular events in both men and women. Thus, there is an overwhelming database of randomized clinical outcome trials showing the benefits of statin therapy in reducing cardiovascular disease, which, coupled with their excellent safety profile, has resulted in statins becoming a very widely used class of drugs and first line therapy for the prevention of cardiovascular disease.  

 

Effect of Statins Therapy on Clinical Outcomes in Specific Patient Groups

 

PRIMARY PREVENTION

 

While there is no doubt that individuals with pre-existing cardiovascular disease require statin therapy, the use of statins for primary prevention was initially debated. There are now a large number of statin primary prevention studies. The Cholesterol Treatment Trialists reported that statin therapy was very effective in reducing cardiovascular events in subjects without a history of vascular disease and the relative risk reduction was similar to subjects with a history of cardiovascular events (35). Additionally, vascular deaths were reduced by statin treatment even in subjects without a history of vascular disease. As expected, non-vascular deaths were not altered in these subjects without a history of pre-existing vascular disease. Additionally, the Cholesterol Treatment Trialists compared the benefits of statin therapy based on baseline risk of developing cardiovascular disease (<5%, ≥5% to <10%, ≥10% to <20%, ≥20% to <30%, ≥30%) (37). The proportional reduction in major vascular events was at least as big in the two lowest risk categories as in the higher risk categories indicating that subjects at low-risk benefit from statin therapy. Similar to the Cholesterol Treatment Trialists analysis, a Cochrane review published in 2013 on the effect of statins in primary prevention patients reached the following conclusion: “Reductions in all-cause mortality, major vascular events, and revascularizations were found with no excess adverse events among people without evidence of CVD treated with statins” (38). An additional study (HOPE-3 trial), not included in the above analyses, has been completed that focused on intermediate risk patients without cardiovascular disease. In this trial 12,705 men and women who had at least one risk factor for cardiovascular disease were randomized to 10mg rosuvastatin vs. placebo (39). Rosuvastatin treatment resulted in a 27% reduction in LDL-C levels and a 24% decrease in cardiovascular events providing additional evidence that statin treatment can reduce events in primary prevention patients. It is therefore clear that statins are effective in safely reducing events in primary prevention patients.

 

The key issue is “which primary prevention patients should be treated” and this is still controversial. It should be noted that the higher the baseline risk the greater the absolute reduction in events with statin therapy. For example, in a high-risk patient with a 20% risk of developing a vascular event, a 25% risk reduction will result in a 15% risk of an event (absolute decrease of 5%). In contrast in a low-risk patient with a 4% risk of developing a vascular event, a 25% risk reduction will result in a 3% risk (absolute decrease of only 1%). Thus, the absolute benefit of statin therapy over the short term will depend on the risk of developing cardiovascular disease.

 

Additionally, based on the Cholesterol Treatment Trialists results the reduction in cardiovascular events is dependent on the absolute decrease in LDL-C levels. Thus, the effect of statin treatment will be influenced by baseline LDL-C levels. For example, a 50% decrease in LDL-C is 80mg/dL if the starting LDL is 160mg/dL and only 40mg/dL if the starting LDL-C is 80mg/dL. Based on studies showing that a decrease in LDL-C of 1 mmol/L (40mg/dL) reduces cardiovascular events by ~20% the relative benefit of statin therapy will be greater in the patient with the starting LDL-C of 160mg/dL (40% decrease in events) than in the patient with the starting LDL-C of 80mg/dL (20% decrease in events). Thus, decisions on treatment need to factor in both relative risk and baseline LDL levels.

 

Finally, it should be recognized that clinical trials represent short term reductions in LDL-C levels (typically 2-5 years) in a disorder that begins early in life and progresses over decades. Life-long decreases in LDL-C levels due to genetic polymorphisms are associated with a much greater reduction in cardiovascular events than would be expected based on the clinical trial results (40). These results suggest that earlier and longer lasting therapy that decreases LDL-C levels will result in a greater reduction in cardiovascular events (41). An in depth discussion of the benefits of early therapy is discussed in the following reference (42).

 

ELDERLY

 

Few studies have focused on lowering LDL-C in elderly patients, which we define as individuals greater than 75 years of age (this is based on the ACC/AHA guidelines using age 75 in their decision algorithms) (3). The Prosper Trial determined the effect of pravastatin 40mg/day (n= 2891) vs. placebo (n= 2913) on cardiovascular events in older subjects (70-82) with pre-existing vascular disease or who were at high risk for vascular disease (43). The average age in this trial was 75 years of age and approximately 45% had cardiovascular disease. As expected, pravastatin treatment lowered LDL-C by 34% compared to the placebo group. The primary end point was coronary death, non-fatal myocardial infarction, and fatal or non-fatal stroke which was reduced by 15% (HR 0.85, 95% CI 0.74-0.97, p=0.014). However, in the individuals without pre-existing cardiovascular disease pravastatin did not significantly reduce cardiovascular events (HR- 0.94; CI- 0.77–1.15). In contrast, in individuals with cardiovascular disease pravastatin therapy reduced cardiovascular events (HR- 0.78, CI- 0.66–0.93). Thus, this study demonstrated benefits of statin therapy in the elderly with cardiovascular disease but failed to demonstrate benefit in the elderly without cardiovascular disease.

 

A meta-analysis by the Cholesterol Treatment Trialists of 28 trials with 14,483 of 186,854 participants older than 75 years of age found a decrease in cardiovascular events in all age groups including participants older than 75 years of age (Figure 2) (44). Similar to the Prosper Trial a decrease in cardiovascular events was clearly demonstrated in individuals with pre-existing cardiovascular disease (secondary prevention) but in individuals without pre-existing cardiovascular disease (primary prevention) the decrease in cardiovascular events was not statistically significant (Figure 3). Thus, in older patients with cardiovascular disease lowering LDL-C levels with statins clearly reduces cardiovascular events but in older patients without cardiovascular disease the data demonstrating that statins reduce cardiovascular events is less robust but suggests a reduction in cardiovascular events.

 

Figure 2. Effect of Statin Treatment on Major Vascular Events. Modified from (44).

Figure 3. Effect of Statin Treatment on Major Vascular Events in Individuals With and Without Pre-Existing Cardiovascular Disease. Modified from (44).

 

Studies are currently underway to provide definitive information on whether statin therapy is beneficial as primary prevention in the elderly. STAREE (NCT02099123) is a multicenter randomized trial in Australia of atorvastatin 40mg vs. placebo in adults ≥ 70 years of age without cardiovascular disease and PREVENTABLE (NCT04262206) is a multicenter randomized trial in the USA of atorvastatin vs. placebo in adults ≥ 75 years of age without cardiovascular disease (45,46).

 

WOMEN

 

As noted above a meta-analysis by the Cholesterol Treatment Trialists examined the effect of statins in 27 trials that included 46,675 women and 127,474 men (36). They found that statin therapy was similarly effective in reducing cardiovascular events in both men and women.

 

ASIANS

 

Pharmacokinetic data have shown that the serum levels of statins are higher in Asians than in Caucasians (47). Moreover, Asians achieve similar LDL lowering at lower statin doses than Caucasians (47). Therefore, the statin dose used should be lower in Asians. For example, the starting dose of rosuvastatin is 5mg in Asians as compared to 10mg in Caucasians. Additionally, the maximum recommended dose of statin is lower in Japan vs. the United States (Table 3). In contrast, studies suggest that South Asian patients may be treated with atorvastatin and simvastatin at doses typically applied to white patients (48). Studies have demonstrated that statins reduce cardiovascular events in Asians (49,50)

 

Table 3. Maximum Statin Dose in Japan and United States

Statin

Japan

United States

Atorvastatin

40

80

Fluvastatin

60

80

Pravastatin

20

80

Rosuvastatin

20

40

Simvastatin

20

40

 

DIABETES

 

Statin trials, including both primary and secondary prevention trials, have consistently shown the beneficial effect of statins on cardiovascular disease in patients with diabetes (51). The Cholesterol Treatment Trialists analyzed data from 18,686 subjects with diabetes (mostly type 2 diabetes) from 14 randomized trials (52). In the statin treated group there was a 9% decrease in all-cause mortality, a 13% decrease in vascular mortality, and a 21% decrease in major vascular events per 1mmol/L (39mg/dL) reduction in LDL-C. The beneficial effect of statin therapy was seen in both primary and secondary prevention patients. The effect of statin treatment on cardiovascular events in patients with diabetes was similar to that seen in non-diabetic subjects. It should be noted that while the data for patients with type 2 diabetes is robust, the number of patients with type 1 diabetes in these trials is relatively small and the results less definitive. Also, of note is that information on young patients with diabetes (< age 40) is very limited. Thus, these studies indicate that statins are beneficial in reducing cardiovascular disease in patients with diabetes. For addition details on the treatment of dyslipidemia in patients with diabetes see the chapter entitled “Dyslipidemia in Patients with Diabetes” (51).

 

RENAL DISEASE

 

The Cholesterol Treatment Trialists examined the effect of renal function on statin effectiveness. They reported that the relative risk reduction for cardiovascular events was similar if the eGFR was < 60ml/min as compared to > 90 or 60-90 (35). In a follow-up analysis it was reported that the relative risk reduction per 1mMol/l (~39mg/dL) decrease in LDL-C levels with statin therapy was 0·78 for an eGFR ≥60 mL/min, 0·76 for an eGFR 45 to <60 mL/min, 0·85 for an eGFR 30 to <45 mL/min, and 0·85 for an eGFR <30 mL/min in patients not on dialysis (53). In patients on dialysis the relative risk reduction was 0·94 (99% CI 0·79-1·11). Similarly, a meta-analysis of 57 studies with >143,000 participants with renal disease not on dialysis reported a 31% reduction in major cardiovascular events in statin treated subjects compared to placebo groups (54). Thus, in patients with renal disease not on dialysis, treatment with statins is beneficial and should be utilized in this population at high risk for vascular disease.

 

In contrast to the above results, studies examining the role of statins in dialysis patients have not found a benefit from statin therapy. The Deutsche Diabetes Dialyse Studie (4D) randomized 1,255 type 2 diabetic subjects on hemodialysis to either 20 mg atorvastatin or placebo (55). The LDL-cholesterol reduction was similar to that seen in non-dialysis patients but there was no significant reduction in cardiovascular death, nonfatal myocardial infarction, or stroke in the atorvastatin treated compared to the placebo group. Similarly, A Study to Evaluate the Use of Rosuvastatin in Subjects on Regular Hemodialysis (AURORA) randomized 2,776 subjects on hemodialysis to rosuvastatin 10 mg or placebo (56). Again, the LDL-cholesterol lowering in dialysis patients was similar to that seen in other studies but there was no significant effect on the primary endpoint of cardiovascular death, nonfatal myocardial infarction, or stroke. A meta-analysis of 25 studies involving 8,289 dialysis patients found no benefit of statin therapy on major cardiovascular events, cardiovascular mortality, all-cause mortality, or myocardial infarction, despite efficacious lipid lowering. The reason for the failure of statins in patients on maintenance dialysis is unclear but could be due to a number of factors including the possibility that the marked severity of atherosclerosis in end stage renal disease may limit reversal, that different mechanisms of atherosclerosis progression occur in dialysis patients (for example an increased role for inflammation, oxidation, or thrombosis), or that cardiovascular events in this patient population may not be due to atherosclerosis. We would recommend continuing statin therapy in patients on dialysis who have been previously treated with statins but not initiating therapy in the rare statin naïve patient beginning dialysis.

 

Statins are primarily metabolized in the liver and therefore the need to adjust the statin dose is not usually needed in patients with renal disease until the eGFR is < 30ml/min. The effect of renal dysfunction on statin clearance varies from statin to statin (57). For some statins such as atorvastatin, there is no need to adjust the dose in renal disease because there is limited renal clearance (57). However, for other statins it is recommended to adjust the dose in patients when the eGFR is < 30ml/min. In patients with an eGFR < 30ml/min the maximum dose of rosuvastatin is 10mg, simvastatin 40mg, pitavastatin 2mg, pravastatin 20mg, lovastatin 20mg, and fluvastatin 40mg per day (57).

 

For additional information on the treatment of dyslipidemia in patients with renal disease see the chapter entitled “Dyslipidemia in Chronic Kidney Disease” (57).

 

CONGESTIVE HEART FAILURE

 

In the Corona study 5,011 patients with New York Heart Association class II, III, or IV ischemic, systolic heart failure (most were class III) were randomly assigned to receive 10 mg of rosuvastatin or placebo per day (58). While rosuvastatin treatment reduced LDL-C levels by 45% compared to placebo, rosuvastatin did not decrease death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke. Similarly, the GISSI-HF trial randomized 4,574 patients with class II, III, of IV congestive heart failure (most were class II) to 10mg of rosuvastatin or placebo (59). The primary endpoints were time to death, and time to death or admission to hospital for cardiovascular reasons and these were similar in the statin and placebo groups. Why statin treatment was not beneficial in patients with congestive heart failure is unknown.

 

LIVER DISEASE

 

Many patients with liver disease, particularly those with nonalcoholic fatty liver disease (NAFLD), are at high risk for cardiovascular disease and therefore require statin therapy (60). There have been concerns that these patients would not tolerate statin therapy and that statin therapy would worsen their underlying liver disease. Fortunately, there are now studies of statin therapy in patients with abnormal liver function tests and underlying liver disease at baseline (60-62). With a variety of statins, studies have demonstrated no significant worsening of liver disease and in fact several studies have suggested improvement in liver function tests with statin therapy (62). This is true for patients with hepatitis C, NAFLD/NASH, and primary biliary cirrhosis. Additionally, in the GREACE trial, statin treatment reduced cardiovascular events in patients with moderately abnormal liver function tests (transaminases < 3x the upper limit of normal) (63). Thus, in patients with mild liver disease without elevations in bilirubin or abnormalities in synthetic function, statins are safe and reduce the risk of cardiovascular disease. 

 

For additional information on the treatment of dyslipidemia in patients with liver disease see the chapter entitled “Lipid and Lipoprotein Metabolism in Liver Disease” (64).

 

HIV

 

Patients living with HIV have an increased risk of cardiovascular disease (33). A trial randomized 7,769 participants with HIV infection with a low-to-moderate risk of cardiovascular disease to either pitavastatin 4 mg or placebo (65). The primary outcome was the occurrence of cardiovascular death, myocardial infarction, hospitalization for unstable angina, stroke, transient ischemic attack, peripheral arterial ischemia, revascularization, or death from an undetermined cause. In the pitavastatin group cardiovascular events were decreased by 35% (HR, 0.65; 95% CI, 0.48 to 0.90; P=0.002). For additional information on the use of statins in HIV patients see the Endotext chapter “Lipid Disorders in People with HIV” (33).  

 

Statin Side Effects

 

An umbrella review of meta-analyses of observational studies and randomized controlled trials examined 278 unique non-CVD outcomes from 112 meta-analyses of observational studies and 144 meta-analyses of RCTs and found that the only adverse effects associated with statin therapy were the development of diabetes and muscle disorders (66). For a detailed discussion of the side effects of statin therapy a scientific statement from the American Heart Association provides a comprehensive review (67).

 

DIABETES

 

After many years of statin use it was recognized that statins increase the risk of developing diabetes. In a meta-analysis of 13 trials with over 90,000 subjects, there was a 9% increase in the incidence of diabetes during follow-up among subjects receiving statin therapy (68). All statins appear to increase the risk of developing diabetes. In comparisons of intensive vs. moderate statin therapy, Preiss et al observed that patients treated with intensive statin therapy had a 12% greater risk of developing diabetes compared to subjects treated with moderate dose statin therapy (69). Older subjects, obese subjects, and subjects with high glucose levels were at a higher risk of developing diabetes while on statin therapy (70). Thus, statins may be unmasking and accelerating the development of diabetes that would have occurred naturally in these subjects at some point in time. In patients without risk factors for developing diabetes, treatment with statins does not appear to increase the risk of developing diabetes.

 

In patients with diabetes, an analysis of 9 studies with over 9,000 patients with diabetes reported that the patients randomized to statin therapy had a 0.12% higher A1c than the placebo group indicating that statin therapy is associated with only a very small increase in A1c levels in patients with diabetes that is unlikely to be clinically significant (71). Individual studies, such as CARDS and the Heart Protection Study, have also shown only a very modest effect of statins on A1c levels in patients with diabetes (72,73).

 

The mechanism by which statins increase the risk of developing diabetes is unknown (74). A study has demonstrated that a polymorphism in the gene for HMG-CoA reductase that results in a decrease in HMG-CoA reductase activity and a small decrease in LDL levels is also associated with an increase in body weight and plasma glucose and insulin levels (75). Additionally, a cross sectional study that compared the change in BMI in individuals on statins to individuals not on statins observed an increased BMI in the subjects taking statins (+1.3 in stain users vs. + 0.4 in non-users over a 10 year period; p=0.02) (76). These observations suggest that the inhibition of HMG-CoA reductase per se may be contributing to the statin induced increased risk of diabetes via weight gain. However, studies have now shown that polymorphisms in different genes (NPC1L1 and PCSK9) that lead to a decrease in LDL-C levels are also associated with an increase in diabetes suggesting that decreases in LDL-C levels per se alter glucose metabolism and increase the risk of diabetes (74,77). How a decrease in LDL-C levels might affect glucose metabolism is unknown. Clearly further studies are required to understand the mechanisms by which statins increase the risk of developing diabetes.

 

In balancing the benefits and risks of statin therapy it is important to recognize that an increase in plasma glucose levels is a surrogate marker for an increased risk of developing micro and macrovascular disease (i.e., an increase in plasma glucose per se is not an event but rather increases the risk of future events). In contrast, statin therapy is preventing actual clinical events that cause morbidity and mortality. Furthermore, it may take many years for an elevated blood glucose to induce diabetic complications while the reduction in cardiovascular events with statin therapy occurs relatively quickly. Finally, the number of patients needed to treat with statins to avoid one cardiovascular event is much lower (10-20 depending on the type of patient) than the number of patients needed to treat to cause one patient to develop diabetes (100–200 for one extra case of diabetes) (74). Patients on statin therapy, particularly those with risk factors for the development of diabetes, should be periodically screened for the development of diabetes with measurement of fasting glucose or A1c levels.

 

CANCER

 

Analysis of 14 trials with over 90,000 subjects by the Cholesterol Treatment Trialists did not demonstrate an increased risk of cancer or any specific cancer with statin therapy (34). An update with an analysis of 27 trials with over 174,000 participants also did not observe an increase in cancer incidence or death (36). Additionally, no differences in cancer rates were observed with any particular statin.

 

COGNITIVE DYSFUNCTION

 

Several randomized clinical trials have examined the effect of statin therapy on cognitive function and have not indicated any increased risk (78-80). The Prosper Trial was designed to determine whether statin therapy will reduce cardiovascular disease in older subjects (age 70-82) (43). In this trial cognitive function was assessed repeatedly and no difference in cognitive decline was found in subjects treated with pravastatin compared to placebo (43,81). In the Heart Protection Study over 20,000 patients were randomized to simvastatin 40mg or placebo and again no significant differences in cognitive function was observed between the statin vs. placebo group (82). Additionally, a Cochrane review examined the effect of statin therapy in patients with established dementia and identified 4 studies with 1154 participants (83). In this analysis no benefit or harm of statin therapy on cognitive function could be demonstrated in this high-risk group of patients. Thus, randomized clinical trials do not indicate a significant association.

 

HEMORRAGIC STROKE

 

In a scientific statement from the American Heart Association on statin safety reached the following conclusions; “The available data in aggregate show no increased risk of brain hemorrhage with statin use in primary stroke prevention populations. An increased risk in secondary stroke prevention populations is possible, but the absolute risk is very small, and the benefit in reducing overall stroke and other vascular events generally outweighs that risk” (67).

 

LIVER DISEASE

 

It was in initially thought that statins induced liver dysfunction and it was recommended that liver function tests be routinely obtained while patients were taking statins. However, studies have now shown that the risk of liver function test abnormalities in patients taking statins is very small (61). For example, in a survey of 35 randomized studies involving > 74,000 subjects, elevations in transaminases were seen in 1.4% of statin treated subjects and 1.1% of controls (84). Similarly, in a meta-analysis of > 49,000 patients from 13 placebo controlled studies, the incidence of transaminase elevations greater than three times the upper limit of normal was 1.14% in the statin group and 1.05% in the placebo group (85). Moreover, even when the transaminase levels are elevated, repeat testing often demonstrates a return towards normal levels (86). The increases in transaminase levels with statin therapy are dose related with high doses of statins leading to more frequent elevations (87). At this time, routine monitoring of liver function tests in patients taking statins is no longer recommended. However, obtaining baseline liver function tests prior to starting statin therapy is indicated (61). If liver function tests are obtained during statin treatment, one should not be overly concerned with modestly elevated transaminase levels (less than 3x the upper limit of normal) (61). If the transaminase is greater than 3x the upper limit of normal the test should be repeated and if it remains > 3x the upper limit of normal, statin therapy should be stopped and the patient evaluated (61).

 

A more clinically important issue is whether statins lead to an increased risk of liver failure. Studies have suggested that the incidence of liver failure in patients taking statins is very similar to the rate observed in the general population (approx. 1 case per 1 million patient years) (88,89). Thus, statin therapy causing serious liver injury is a very rare event.

 

Non-alcoholic fatty liver disease (NAFLD) is very common and is associates with obesity, metabolic syndrome, diabetes, and cardiovascular disease. In patients with NAFLD studies have shown that statins decrease liver enzymes and reduce steatosis (90).

 

MUSCLE

 

The most common side effect of statin therapy is muscle symptoms. These can range from life threatening rhabdomyolysis to myalgias (Table 4) (91).

 

Table 4. Spectrum of Statin Induced Muscle Disorders (Adapted from J. Clinical Lipidology 8: S58-71, 2014)

Myalgia- aches, soreness, stiffness, tenderness, cramps with normal CK levels

Myopathy- muscle weakness with or without increased CK

Myositis- muscle inflammation

Myonecrosis- mild (CK >3x ULN); moderate (CK> 10x ULN); severe (CK> 50x ULN)

Rhabdomyolysis- myonecrosis with myoglobinuria or acute renal failure

 

Many patients will discontinue the use of statins due to muscle symptoms. Risk factors associated with an increased incidence of statin associated muscle symptoms are listed in Table 5 (92,93).

 

Table 5. Risk Factors for Statin Myopathy

Medications that alter statin metabolism

Older age

Female

Hypothyroidism

Excess alcohol

Vitamin D deficiency

History of muscle disorders

Renal disease

Liver disease

Personal or family history of statin intolerance

Low BMI

Polymorphism in SLCO1B1 gene

High dose statin

Drug-drug interactions

 

 The Cholesterol Treatment Trialists analyzed individual participant data on the development of muscle symptoms from 19 double-blind trials of statin versus placebo with 123,940 participants and four double-blind trials of a more intensive vs. a less intensive statin regimen with 30,724 participants (94). After a median follow-up of 4.3 years 27.1% of the individuals taking a statin vs. 26.6% on placebo reported muscle pain or weakness representing a 3% increase greater than placebo (risk ratio- 1.03; 95% CI 1.01-1.06) (Table 6). The specific muscle symptoms caused by statin therapy, myalgia, muscle cramps or spasm, limb pain, other musculoskeletal pain, or muscle fatigue or weakness were similar to those caused by placebo. The increase in muscle symptoms in the statin treated individuals was manifest in the first year of therapy but in the later years muscle symptoms were similar in the statin treated and placebo groups. The relative risk of statin induced muscle symptoms was greater in women than men. Intensive statin treatment with 40-80 mg atorvastatin or 20-40 mg rosuvastatin resulted in a higher risk of muscle symptoms than less intensive or moderate-intensity regimens but different statins at equivalent LDL-C lowering doses had similar effects on muscle symptoms. This study demonstrates that there is a small increase in muscle symptoms that primarily manifests in the first year of therapy. Statin therapy caused approximately 11 additional complaints of muscle pain or weakness per 1000 patients during the first year, but little excess in later years. Of particularly note is that 26.6% of patients taking a placebo had muscle symptoms demonstrating a very high frequency of this clinical complaint. Given the high prevalence of muscle complaints and the small increase attributed to statins it is very difficult to determine if a muscle complaint is actually due to the statin, which presents great clinical difficulties in patient management.                                                       

 

Table 6. Effect of Statin vs. Placebo on Muscle Symptoms

Symptom

Statin Events

Placebo Events

RR (95% CI)

Myalgia

12.0%

11.7%

1·03 (0·99–1·08)

Other musculoskeletal pain

13.3

13.0

1·03 (0·99–1·08)

Any muscle pain

26.9%

26.3%

1·03 (1·01–1·06)

Any muscle pain or weakness

27.1%

26.6%

1·03 (1·01–1·06)

Modified from (94).

 

While the results of the randomized trials suggest that muscle symptoms are not frequently induced by statin therapy, in typical clinical settings a significant percentage of patients are unable to tolerate statins due to muscle symptoms (in many studies as high as 5-25% of patients) (95-97). Recently there was a randomized trial that explored the issue of myopathy with statin therapy in great detail (98). In this trial the effect of atorvastatin 80mg a day vs. placebo for 6 months on creatine kinase (CK), exercise capacity, and muscle strength was studied in 420 healthy, statin-naive subjects. Atorvastatin treatment led to a modest increase in CK levels (20.8U/L) with no change observed in the placebo group. None of the subjects had an elevation of CK > 10x the upper limits of normal. There were no changes in muscle strength or exercise capacity with atorvastatin treatment. However, myalgia was reported in 19 subjects (9.4%) in the atorvastatin group compared to 10 subjects (4.6%) in the placebo group (p=0.05).  In this study “myalgia” was considered to be present if all of the following occurred: (1) subjects reported new or increased muscle pain, cramps, or aching not associated with ex­ercise; (2) symptoms persisted for at least 2 weeks; (3) symptoms resolved within 2 weeks of stopping the study drug; and (4) symp­toms reoccurred within 4 weeks of restarting the study medication. Notably these myalgias were not associated with elevated CK levels. In the atorvastatin group the myalgias tended to occur soon after therapy (average 35 days) whereas in the placebo group myalgias occur later (average 61 days). In the atorvastatin group the symptoms were predominantly localized to the legs and included aches, cramps, and fatigue, whereas in the placebo group they were more diverse including whole body fatigue, foot cramps, worsening of pain in previous injuries, and groin pain. A number of conclusions can be reached from this study. First, statin treatment does in fact increase the incidence of myalgias. Second, a substantial number of patients treated with placebo will also develop myalgias. Third, clinically differentiating statin induced myalgias from placebo induced myalgias is difficult, as there are no specific symptoms, signs, or biomarkers that clearly distinguish between the two. It should be recognized that the patient population typically treated with statins (patients 50-80 years of age) often have muscle symptoms in the absence of statin therapy and it is therefore difficult to be certain that the muscle symptoms described by the patient are actually due to statin therapy.

 

Additionally, when patients know that they are taking a statin they are more likely to have muscle symptoms (i.e. the nocebo effect). This was nicely demonstrated in the ASCOT-LLA extension trial (99). In the initial phase of the study the patients were randomly assigned to atorvastatin 10 mg (n= 5101) or matching placebo (n= 5079) in a double-blind fashion. During the 3.3 years of the double blinded phase adverse muscle symptoms were very similar in the atorvastatin and placebo groups (HR 1.03; p=0.72). This double-blind phase was followed by a non-blinded non-randomized extension where 6409 patients were treated with atorvastatin 10mg and 3490 were untreated. During the 2.3 years of this extension study muscle symptoms were significantly increased in the atorvastatin group (HR 1·41; p=0.006).    

 

In a very small study in the Annals of Internal Medicine eight patients with “statin related myalgia” were re-challenged with statin or placebo and there were no statistically significant differences in the recurrence of myalgias on the statin or placebo (100). This approach has been expanded upon in other studies. In 120 patients with “statin induced myalgia” patients were randomized in a double blinded crossover trial to either simvastatin 20mg per day or placebo and the occurrence of muscle symptoms was determined (101). Only 36% of these patients were confirmed to actually have statin induced myalgia (presence of symptoms on simvastatin without symptoms on placebo). In a similar study, Nissen and colleagues studied 491 patients with “statin induced myalgia” treating with either atorvastatin 20mg per day or placebo in a double-blind crossover trial (102). In this trial 42.6% of patients were confirmed to have statin induced muscle symptoms. In a trial of 156 patients with prior statin induced muscle symptoms patients were treated with alternating periods of atorvastatin 20mg or placebo (103). In this trial no difference in muscle symptoms was found between the statin and placebo treatment periods. A smaller crossover trial in 49 patients who had stopped statin therapy also found no difference in muscle symptoms when patients were taking atorvastatin 20mg or placebo (104)

 

Thus, while statin induced myalgias are a real entity careful studies have shown that in the majority of patients with “statin induced muscle symptoms” the symptoms are not actually due to statin therapy. In the clinic it is difficult to be certain whether the muscle symptoms are actually due to true statin intolerance or to other factors. The approach to treating these patients will be discussed later in this chapter (Treatment of Stain Intolerant Patients). While some patients will not tolerate statin therapy due to myalgias, this side effect does not appear to result in serious morbidity or long-term consequences. In contrast, studies have found that discontinuing statins increases the risk of myocardial infarctions and death from cardiovascular disease (105,106).

 

Fortunately, the more serious muscle related side effects of statin therapy are rare. In a meta-analysis of 21 statin vs. placebo trials there was an excess risk of rhabdomyolysis of 1.6 patients per 100,000 patient years or a standardized rate of 0.016/patient years (86). Other studies report a rate of rhabdomyolysis between 0.03- 0.16 per 1,000 patient years (107). Similarly, the risk of statin induced myositis (muscle symptoms with an increase in CK 10 times the upper limits of normal) is also very low. In an analysis of 21 randomized trials myositis occurred in only 5 patients per 100,000 person years or 0.05/1000 patient years (86). The higher the dose of statin used the greater the risk of myositis and rhabdomyolysis. In a comparison of five trials that compared high dose statin vs. low dose statin there was an excess risk of rhabdomyolysis of 4 per 10,000 people treated (35). The likely basis for an increased risk of myositis or rhabdomyolysis is elevated statin blood levels, which are more likely to occur with high doses of statins. In the development of statins, manufacturers have studied higher doses that are not approved for clinical use. For example, simvastatin and pravastatin at 160mg per day were studied but discontinued due to an increased incidence of muscle side effects (108,109). The use of simvastatin 80mg per day, a previously approved dose, was discontinued due to an increased risk of muscle side effects. Similarly, pitavastatin at doses greater than 4mg per day was investigated, but development was abandoned when an increased risk of rhabdomyolysis was observed. Along similar lines, in many of the patients that develop rhabdomyolysis, the etiology can be linked to the use of other drugs that alter statin metabolism thereby increasing statin blood levels (93). For example, prior to drug interactions being recognized the use of cyclosporine, gemfibrozil, HIV protease inhibitors, and erythromycin in conjunction with certain statins was linked with the development of rhabdomyolysis (93). Finally, common variants in SLCO1B1, which encodes the organic anion-transporting polypeptide OATP1B1, are strongly associated with an increased risk of statin-induced myopathy (110). OATP1B1 facilitates the transport of statins into the liver and certain polymorphisms are associated with an increased risk of developing statin induced muscle disorders, due to the decreased transport of statins into the liver resulting in increased blood levels (111). The exact mechanism by which elevated blood levels induce muscle toxicity remains to be elucidated.

 

Recently it has been recognized that a very small number of patients taking statins develop a progressive autoimmune necrotizing myopathy, which is characterized by progressive symmetric proximal muscle weakness, elevated CK levels (typically >10x the ULN), and antibodies against HMG-CoA reductase (112). It is estimated that this occurs in 2 or 3 per 100,000 patients treated with a statin (112). This myopathy may begin soon after initiating statin therapy or develop after a patient has been on statins for many years (112). Muscle biopsy reveals necrotizing myopathy without severe inflammation (112). In contrast to the typical muscle disorders induced by statin therapy, the autoimmune myopathy progresses despite discontinuing therapy. Spontaneous improvement is not typical and most patients will need to be treated with immunosuppressive therapy (glucocorticoids plus methotrexate, azathioprine, or mycophenolate mofetil) (112). It should be recognized that this disorder can occur in individuals that have not been exposed to statin therapy (113). Statins likely potentiate the development of this disorder in susceptible individuals, perhaps by increasing HMG-CoA reductase levels.

 

From the above certain conclusions can be reached. First, the risk of serious muscle disorders due to statin therapy is very small, particularly if one is aware of the potential drug interactions that increase the risk. Second, the muscle toxicity is usually linked to elevated statin blood levels and the higher the dose of the statin the more likely the chance of developing toxicity. Third, myalgias in patients taking statins are very common and can be due to statin treatment. However, in the individual patient, it is very difficult to know if the myalgia is actually secondary to statin therapy and in many, if not most patients, the myalgias are not due to statin therapy. Fourth, the muscle symptoms that occur in association with statin treatment are a major reason why patients discontinue statin use and therefore better diagnostic algorithms and treatments are required to allow patients to better comply with these highly effective treatments to reduce cardiovascular disease. 

 

Contraindications

 

Previously statins were contraindicated in pregnant women or lactating women. However, in July 2021 the FDA requested the removal of the strongest recommendation against using statins during pregnancy. They continue to advise against the use of statins in pregnancy given the limited data and quality of information available. The decision of whether to continue a statin during pregnancy requires shared decision-making between the patient and clinician, and healthcare professionals need to discuss the risks versus the benefits in high-risk women, such as those with homozygous FH or prior ASCVD events, that may benefit from statin therapy. For a detailed discussion of the use of statins during pregnancy see the Endotext chapter entitled “Effect of Pregnancy on Lipid Metabolism and Lipoprotein Levels” (114).

 

In addition, liver function tests should be obtained prior to initiating statin treatment and moderate to severe liver disease is a contraindication to statin therapy (61). 

 

Summary

 

An enormous data base has accumulated which demonstrates that statins are very effective at reducing the risk of cardiovascular disease and that statins have an excellent safety profile. The risk benefit ratio of treating patients with statins is very favorable and has resulted in this class of drugs being widely utilized to lower serum lipid levels and to reduce the risk of cardiovascular disease and death.

 

EZETIMBE (ZETIA)

 

Introduction

 

Ezetimibe (Zetia) inhibits the absorption of cholesterol by the intestine thereby resulting in modest decreases in LDL-C levels (115). Ezetimibe is primarily used in combination with statin therapy when statin treatment alone does not lower LDL-C levels sufficiently or when patients only tolerate a low statin dose. It may also be used as monotherapy or in combination with other lipid lowering drugs to lower LDL-C levels in patients with statin intolerance. Finally, it is the drug of choice in patients with the rare genetic disorder sitosterolemia, which is discussed in detail in the chapter “Sitosterolemia” (116). Ezetimibe is relatively inexpensive as it is now a generic drug.

 

Effect of Ezetimibe on Lipid and Lipoprotein Levels

 

Pandor and colleagues have published a meta-analysis of ezetimibe monotherapy that included 8 studies with 2,722 patients (117). They reported that ezetimibe decreased LDL-C levels by 18.6%, decreased triglyceride levels by 8.1%, and increased HDL-C levels by 3% compared to placebo. In a pooled analysis by Morrone and colleagues of 27 studies with 11, 714 subjects treated with ezetimibe in combination with statin therapy similar results were observed (118). Specifically, LDL-C levels were decreased by 15.1%, non-HDL-C levels by 13.5%, triglycerides by 4.7%, apolipoprotein B levels by 10.8%, and HDL-C levels were increased by 1.6%. The combination of a high dose potent statin plus ezetimibe can lower LDL-C levels by 70% (119). A meta-analysis of the effect of ezetimibe on Lp(a) revealed that with either monotherapy or combination with statin there was no change in Lp(a) levels (120). The effect of ezetimibe on lipid parameters occurs quickly and can be seen after 2 weeks of treatment. In patients with Heterozygous Familial Hypercholesterolemia who have marked elevations in LDL-C levels, the addition of ezetimibe to statin therapy resulted in a further 16.5% decrease in LDL-C levels (121). Thus, in comparison with statins, ezetimibe treatment produces modest decreases in LDL-C levels (15-20%). In addition to these changes in lipid parameters, ezetimibe in combination with a statin decreased hs-CRP by 10-19% compared to statin monotherapy (122,123). However, ezetimibe alone does not decrease hs-CRP levels (123).

 

Table 7. Effect of Ezetimibe on Lipid/Lipoprotein Levels

LDL-C

Decrease

Non-HDL-C

Decrease

Apolipoprotein B

Decrease

Triglycerides

Small decrease

HDL-C

Small increase

Lp(a)

No change

 

Mechanisms Accounting for the Ezetimibe Induced Lipid Effects

 

NPC1L1 (Niemann-Pick C1-like 1 protein) is highly expressed in the intestine with the greatest expression in the proximal jejunum, which is the major site of intestinal cholesterol absorption (124,125). Knock out animals deficient in NPC1L1 have been shown to have a decrease in intestinal cholesterol absorption (124). Ezetimibe binds to NPC1L1 and inhibits cholesterol absorption (115,124,125). In animals lacking NPC1L1, ezetimibe has no effect on intestinal cholesterol absorption, demonstrating that ezetimibe’s effect on cholesterol absorption is mediated via NPC1L1 (115,125). Thus, a major site of action of ezetimibe is to block the absorption of cholesterol by the intestine (115,125). Cholesterol in the intestinal lumen is derived from both dietary cholesterol (approximately 25%) and biliary cholesterol (approximately 75%); thus the majority is derived from the bile (125). As a consequence, even in patients that have very little cholesterol in their diet, ezetimibe will decrease cholesterol absorption. While ezetimibe is very effective in blocking intestinal cholesterol absorption it does not interfere with the absorption of triglycerides, fatty acids, bile acids, or fat-soluble vitamins including vitamin D and K.

 

When intestinal cholesterol absorption is decreased the chylomicrons formed by the intestine contain less cholesterol and thus the delivery of cholesterol from the intestine to the liver is diminished (126). This results in a decrease in the cholesterol content of the liver, leading to the activation of SREBPs, which enhance the expression of LDL receptors resulting in an increase in LDL receptors on the plasma membrane of hepatocytes (Figure 1) (126). Thus, similar to statins the major mechanism of action of ezetimibe is to decrease the levels of cholesterol in the liver resulting in an increase in the number of LDL receptors leading to the increased clearance of circulating LDL (126). In addition, the decreased cholesterol delivery to the liver may also decrease the formation and secretion of VLDL (126).

 

In addition to NPC1L1 expression in the intestine this protein is also expressed in the liver where it mediates the transport of cholesterol from the bile back into the liver (127). The inhibition of NPC1L1 in the liver will result in the increased secretion of cholesterol in bile and thereby could also contribute to a decrease in the cholesterol content of the liver and an increase in LDL receptor expression and a decrease in VLDL production.

 

Pharmacokinetics and Drug Interactions

 

Following absorption by intestinal cells ezetimibe is rapidly glucuronidated. The glucuronidated ezetimibe is then secreted into the portal circulation and rapidly taken up by the liver where it is secreted into the bile and transported back to the intestine (115). This enterohepatic circulation repeatedly returns ezetimibe to its site of action (note glucuronidated ezetimibe is a very effective inhibitor of NPC1L1) (115). Additionally, this enterohepatic circulation accounts for the long duration of action of ezetimibe and limits peripheral tissue exposure (115). Ezetimibe is not significantly excreted by the kidneys and thus the dose does not need to be adjusted in patients with renal disease.

 

Ezetimibe is not metabolized by the P450 system and does not have many drug interactions (115). It should be noted that cyclosporine does increase ezetimibe levels.

 

Effect of Ezetimibe Therapy on Clinical Outcomes

 

There have been a limited number of ezetimibe clinical outcome trials. Two have studied the effect of ezetimibe in combination with a statin vs. placebo making it virtually impossible to determine if ezetimibe per se has beneficial effects. However, one study has compared ezetimibe plus a statin vs. a statin alone and one study compared ezetimibe vs. placebo. Finally, a study compared moderate-intensity statin with ezetimibe vs. high-intensity statin monotherapy.

 

SEAS TRIAL

 

The SEAS Trial was a randomized trial of 1,873 patients with mild-to-moderate, asymptomatic aortic stenosis (128). The patients received either simvastatin 40mg per day in combination with ezetimibe 10mg per day vs. placebo daily. The primary outcome was a composite of major cardiovascular events, including death from cardiovascular causes, aortic-valve replacement, non-fatal myocardial infarction, hospitalization for unstable angina pectoris, heart failure, coronary-artery bypass grafting, percutaneous coronary intervention, and non-hemorrhagic stroke. Secondary outcomes were events related to aortic-valve stenosis and ischemic cardiovascular events. Simvastatin plus ezetimibe lowered LDL-C levels by 61% compared to placebo. There were no significant differences in the primary outcome between the treated vs. placebo groups. Similarly, the need for aortic valve replacement was also not different between the treated and placebo groups. However, fewer patients had ischemic cardiovascular events in the simvastatin plus ezetimibe treated group than in the placebo group (hazard ratio, 0.78; 95% CI, 0.63 to 0.97; P=0.02), which was primarily accounted for by a decrease in the number of patients who underwent coronary-artery bypass grafting. The design of this study does not allow for one to determine if the beneficial effect on ischemic cardiovascular events typically produced by statin therapy was enhanced by the addition of ezetimibe.

 

SHARP TRIAL

 

The SHARP Trial was a randomized trial of 9,270 patients with chronic kidney disease (3,023 on dialysis and 6,247 not on dialysis) with no known history of myocardial infarction or coronary revascularization (129). Patients were randomly assigned to simvastatin 20 mg plus ezetimibe 10 mg daily vs. placebo. The primary outcome was first major atherosclerotic event (non-fatal myocardial infarction or coronary death, non-hemorrhagic stroke, or any arterial revascularization procedure). Treatment with simvastatin plus ezetimibe resulted in a decrease in LDL-C of 0.85 mmol/L (~34mg/dL). This decrease in LDL-C was associated with a 17% reduction in major atherosclerotic events. In patients on hemodialysis there was a 5% decrease in cardiovascular events that was not statistically significant. Unfortunately, similar to the SEAS Trial, it is impossible to determine whether the addition of ezetimibe improved outcomes above and beyond what would have occurred with statin treatment alone.

 

IMPROVE-IT TRIAL

 

The IMPROVE-IT Trial tested whether the addition of ezetimibe to statin therapy would provide an additional beneficial effect in patients with the acute coronary syndrome (130). The IMPROVE-IT Trial was a large trial with over 18,000 patients randomized to simvastatin 40mg vs. simvastatin 40mg + ezetimibe 10mg per day. On treatment LDL-C levels were 70mg/dL in the statin alone group vs. 54mg/dL in the statin + ezetimibe group. There was a small but significant 6.4% decrease in major cardiovascular events (cardiovascular death, MI, documented unstable angina requiring rehospitalization, coronary revascularization, or stroke) in the statin + ezetimibe group (HR 0.936 CI (0.887, 0.988) p=0.016). Cardiovascular death, non-fatal MI, or non-fatal stroke were reduced by 10% (HR 0.90 CI (0.84, 0.97) p=0.003). There was a significant 21% reduction in ischemic stroke (HR, 0.79; 95% CI, 0.67-0.94; P=0.008) and a nonsignificant increase in hemorrhagic stroke (HR, 1.38; 95% CI, 0.93-2.04; P=0.11) (131). Patients with a prior stroke were at a higher risk of stroke recurrence and the risk of a subsequent stroke was reduced by 40% (HR, 0.60; 95% CI, 0.38-0.95; P=0.030) with ezetimibe added to simvastatin therapy (131). In patients with diabetes or other high risk factors the benefits of adding ezetimibe to statin therapy was enhanced (132). In fact, patients without DM and at low or moderate risk demonstrated no benefit with the addition of ezetimibe to simvastatin (132). Similarly, patients who also had peripheral arterial disease or a history of cerebral vascular disease also had the greatest absolute benefits from the addition of ezetimibe (133). Thus, the addition of ezetimibe to statin therapy is of greatest benefit in patients at high risk (for example patients with diabetes, peripheral vascular disease, cerebrovascular disease, etc.).

 

The results of this study have a number of important implications. First, it demonstrates that combination therapy has benefits above and beyond statin therapy alone. Second, it provides further support for the hypothesis that lowering LDL per se will reduce cardiovascular events. The reduction in cardiovascular events was similar to what one would predict based on the Cholesterol Treatment Trialists results. Third, it suggests that lowering LDL levels into the 50s will have benefits above and beyond lowering LDL levels to the 70mg/dL range in patients with diabetes or other factors that result in a high risk for cardiovascular events. These results have implications for determining goals of therapy and provide support for combination therapy.

 

EWTOPIA 75

 

This was a multicenter, randomized trial in Japan that examined the preventive efficacy of ezetimibe for patients aged ≥75 years (mean age 80.6 years), with elevated LDL-C (≥140 mg/dL) without a history of coronary artery disease who were not taking lipid lowering drugs (134). Patients were randomized to ezetimibe 10mg (n=1,716) or usual care (n=1,695) and followed for 4.1 years. The primary outcome was a composite of sudden cardiac death, myocardial infarction, coronary revascularization, or stroke. In the ezetimibe group LDL-C was decreased by 25.9% and non-HDL-C by 23.1% while in the usual care group LDL-C was decreased by 18.5% and non-HDL-C by 16.5% (p<0.001 for both lipid parameters). By the end of the trial 9.6% of the patients in the usual care group and 2.1% of the ezetimibe group were taking statins. Ezetimibe reduced the incidence of the primary outcome by 34% (HR 0.66; P=0.002). Additionally, composite cardiac events were reduced by 60% (HR 0.60; P=0.039) and coronary revascularization by 62% (HR 0.38; P=0.007) in the ezetimibe group vs. the control group. There was no difference in the incidence of stroke or all-cause mortality between the groups. It should be noted that the reduction in cardiovascular events was much greater than one would expect based on the absolute difference in LDL-C levels (121mg/dL in ezetimibe group vs. 132mg/dL). As stated by the authors “Given the open-label nature of the trial, its premature termination, and issues with follow-up, the magnitude of benefit observed should be interpreted with caution.” Nevertheless, this study provides additional support that ezetimibe can reduce cardiovascular events.

 

RACING TRIAL

 

The RACING trial was a randomized, open-label trial in patients with atherosclerotic cardiovascular disease carried out in South Korea (135). Patients were randomly assigned to either rosuvastatin 10 mg with ezetimibe 10 mg (n= 1894) or rosuvastatin 20 mg (n= 1886). The primary endpoint was cardiovascular death, major cardiovascular events, or non-fatal stroke. The median LDL-C level during the study was 58mg/dL in the combination therapy group and 66mg/dL in the statin monotherapy group (p<0·0001). The primary endpoint occurred in 9.1% of the patients in the combination therapy group and 9·9% of the patients in the high-intensity statin monotherapy group (non-inferior). Non-inferiority was observed in patients with LDL-C levels < 100mg/dL and >100mg/dL and in patients greater than 75 years of age (136,137).

 

This study demonstrates that moderate intensity statin with ezetimibe was non-inferior to high-intensity statin therapy with regards to cardiovascular death, major cardiovascular events, or non-fatal stroke. Interestingly a lower prevalence of discontinuation or dose reduction caused by intolerance to the study drug was seen with combination therapy. This indicates that using a moderate intensity dose of a statin with ezetimibe is a useful strategy in patients that do not tolerate high intensity statin therapy.

 

Side Effects

 

Ezetimibe has not demonstrated significant side effects. In monotherapy trials, the effect on liver function tests was similar to placebo. In a meta-analysis by Toth et al. of 27 randomized trials in > 20,000 participants evaluating statin plus ezetimibe vs. statin alone the incidence of liver function test abnormalities was slightly greater in the combination therapy group (statin alone- 0.35% vs. statin plus ezetimibe 0.56%) (138). In contrast, Luo and colleagues in a meta-analysis of 20 randomized with > 14,000 subjects did not observe a difference in liver function tests in the ezetimibe plus statin vs. statin alone group (139). With regards to muscle side effects, a meta-analysis of seven randomized trials by Kashani and colleagues found that monotherapy with ezetimibe or ezetimibe in combination with a statin did not increase the risk of myositis compared to placebo or monotherapy with a statin (140). Similarly, Luo et al also did not observe that combination therapy with ezetimibe and a statin increased the risk of myositis (139). In a meta-analysis by Savarese et al. of 7 randomized long-term studies including SEAS, SHARP, and IMPROVE-IT, the incidence of cancer was similar in patients treated with ezetimibe vs. patients not treated with ezetimibe (141). This confirms a previous study that also did not demonstrate an increased cancer risk in the three largest ezetimibe trials (142). Ezetimibe does not appear to have adverse effects on fasting glucose levels or A1c levels (143).

 

Thus, over many years of use ezetimibe has been shown to be a very safe drug without major side effects.

 

Contraindications

 

Ezetimibe is contraindicated in patients with active liver disease. The use of ezetimibe during pregnancy and lactation has not been studied.  

 

Summary

 

Ezetimibe has a modest ability to lower LDL-C levels and can be a very useful adjunct to statin therapy. When added to statin therapy it will lower the LDL-C by an additional 15-20% which is equivalent to three titrations of the statin dose (for example adding ezetimibe is equivalent to increasing atorvastatin from 10mg to 80mg per day). Additionally, the combination of a high dose of a potent statin (rosuvastatin 40mg per day) with ezetimibe was able to lower the LDL by approximately 70%, which will allow many patients to reach their LDL goal (123). In patient’s intolerant of statins who either cannot take a statin or can only take low doses of a statin, ezetimibe is extremely useful in further lowering LDL-C. The ease of taking ezetimibe, the lack of serious side effects, and that it is inexpensive as it is now a generic drug make it an obvious second choice drug after statins to lower LDL-C levels.   

 

BILE ACID SEQUESTRANTS 

 

Introduction

 

There are three bile acid sequestrants approved for use in the United States. The first bile acid sequestrant, cholestyramine (Questran), was developed in the 1950s and was the second drug available to lower cholesterol levels (niacin was the first drug). Colestipol (Colestid) was developed in the 1970s and is very similar to cholestyramine. In 2000, Colesevelam (Welchol) was approved. Colesevelam has enhanced binding and affinity for bile acids compared to cholestyramine and colestipol and therefore can be given in much lower doses reducing some side effects (144).

 

​Cholestyramine is available as a powder and the dose ranges from 8-24 grams per day given with meals. Colestipol is available as a tablet and the dose ranges from 2-16 grams per day given with meals or granules and the dose ranges from 5-30 grams per day given with meals. The dose of colesevelam is 3.75 grams per day and can be given as tablets (​take 6 tablets once daily or 3 tablets twice daily), oral suspension (​take one packet once daily), or chewable bars (take one bar once daily). Because bile acid sequestrants mechanism of action starts with the binding of bile acids in the intestine (see below) these drugs are most effective when administered with meals.

 

Effect of Bile Acid Sequestrants on Lipid and Lipoprotein Levels

 

The major effect of bile acid sequestrants is to lower LDL-C levels in a dose dependent fashion. Depending upon the specific drug and dose the decrease in LDL-C ranges from approximately 5 to 30% (144-146). The effect of monotherapy with bile acid sequestrants on LDL-C levels observed in various studies is shown in table 8.

 

Table 8. Effect of Bile Acid Sequestrants on LDL-C

Drug

LDL lowering

Cholestyramine 4g/day

7% decrease

Cholestyramine 24g/day

28% decrease

Colestipol 4g/day

12% decrease

Colestipol 16g/day

24% decrease

Colesevelam 3.8g/day

15% decrease

Colesevelam 4.3g/day

18% decrease

 

Bile acid sequestrants are typically used in combination with statins and the addition of bile acid sequestrants to statin therapy will result in a further 10% to 25% decrease in LDL-C levels (144-146). Combination therapy can result in a 60% reduction in LDL-C levels when high doses of potent statins are combined with high doses of bile acid sequestrants. Bile acid sequestrants will also further lower LDL-C levels by as much as 18% when added to statins and ezetimibe (147). This is particularly useful in patients with Heterozygous Familial Hypercholesterolemia who can have very high LDL-C levels at baseline. Additionally, in patients who are statin intolerant, the combination of a bile acid sequestrant and ezetimibe resulted in an additional 10-20% decrease in LDL-C compared to either drug alone  (148,149). Thus, both in monotherapy and in combination with other drugs that lower LDL-C levels, bile acid sequestrants are effective in lowering LDL-C levels

 

Bile acid sequestrants have a very modest effect on HDL-C levels, typically resulting in a 3-9% increase (144-146). The effect of bile acid sequestrants on triglyceride levels varies (144-146). In patients with normal triglyceride levels, bile acid sequestrants increase triglyceride levels by a small amount. However, as baseline triglyceride levels increase, the effect of bile acid sequestrants on plasma triglyceride levels becomes greater, and can result in substantial increases in triglyceride levels. In patients with triglycerides > 400mg/dL the use of bile acid sequestrants is contraindicated.

 

Table 9. Effect of Bile Acid Sequestrants on Lipid/Lipoprotein Levels

LDL-C

Decrease

Non-HDL-C

Decrease

Apolipoprotein B

Decrease

Triglycerides

Variable. If TG levels elevated will increase significantly

HDL-C

Small Increase

Lp(a)

No change

 

Non-Lipid Effects of Bile Acid Sequestrants

 

Bile acid sequestrants have been shown to reduce fasting glucose and hemoglobin A1c levels (150). Colesevelam has been most intensively studied and in a number of different studies colesevelam has decreased A1c levels by approximately 0.5-1.0% in patients also treated with a variety of glucose lowering drugs including metformin, sulfonylureas, and insulin. The Food and Drug Administration (FDA) has approved colesevelam for improving glycemic control in patients with type 2 diabetes.

 

Bile acid sequestrants decrease CRP. For example, Devaraj et al have shown that colesevelam decreases hs-CRP by 18% compared to placebo (151). In combination with a statin, colesevelam reduced hs-CRP levels by 23% compared to statin alone (152). 

 

Mechanisms Accounting for Bile Acid Sequestrants Induced Lipid Effects

 

Bile acid sequestrants bind bile acids in the intestine, preventing their reabsorption in the terminal ileum leading to the increased fecal excretion of bile acids (153). This decrease in bile acid reabsorption reduces the size of the bile acid pool, which stimulates the conversion of cholesterol into bile acids in the liver (153). This increase in bile acid synthesis decreases hepatic cholesterol levels leading to the activation of SREBPs that up-regulate the expression of the enzymes required for the synthesis of cholesterol and the expression of LDL receptors (153). The increase in hepatic LDL receptors results in the increased clearance of LDL from the circulation leading to a decrease in serum LDL-C levels (Figure 1). Thus, similar to statins and ezetimibe, bile acids lower plasma LDL-C levels by decreasing hepatic cholesterol levels, which stimulates LDL receptor production and thereby accelerates the clearance of LDL from the blood.

 

The key regulator of bile acid synthesis is FXR (farnesoid X receptor), a nuclear hormone receptor that forms a heterodimer with RXR to regulate gene transcription (154,155). Bile acids down-regulate cholesterol 7α hydroxylase, the first enzyme in the bile acid synthetic pathway by several FXR mediated mechanisms. In the ileum, bile acids via FXR stimulate the production of FGF19, which is secreted into the portal vein and inhibits cholesterol 7α hydroxylase expression in the liver (154). Additionally, in the liver, bile acids activate FXR leading to the increased expression of SHP (small heterodimer partner), which inhibits the transcription of cholesterol 7α hydroxylase (155). Thus, a decrease in bile acids will lead to the decreased activation of FXR in the liver and intestines and thereby result in an increase in cholesterol 7α hydroxylase expression and the increased conversion of cholesterol to bile acids resulting in a decrease in hepatic cholesterol content.

 

Decreased activation of FXR can also explain the adverse effects of bile acid sequestrants on triglyceride levels (156,157). Activation of FXR increases the expression of apolipoprotein C-II, apolipoprotein A-V, and the VLDL receptor, proteins that decrease plasma triglyceride levels while decreasing the expression of apolipoprotein C-III, a protein that is associated with increases in plasma triglycerides (156,157). Thus, activation of FXR would be expected to decrease triglyceride levels as increases in apolipoprotein C-II, apolipoprotein A-V, and the VLDL receptor and decreases in apolipoprotein C-III would reduce plasma triglyceride levels. With bile acid sequestrants the activation of FXR would be reduced and decreases in the expression of apolipoprotein C-II, apolipoprotein A-V, and the VLDL receptor and increased expression of apolipoprotein C-III would increase plasma triglyceride levels.

 

The mechanism by which treatment with bile acid sequestrants improves glycemic control is unclear (158). 

 

Pharmacokinetics and Drug Interactions

 

Bile acid sequestrants are not absorbed and not altered by digestive enzymes and thus their primary effects are localized to the intestine (144-146). It should be noted that bile acid sequestrants can indirectly have systemic effects by decreasing the reabsorption of bile acids and thereby reducing the exposure of cells to bile acids, which are biologically active compounds.

 

Unfortunately, in the intestine bile acid sequestrants can impede the absorption of many other drugs (144-146). This is particularly true for cholestyramine and colestipol which are used in large quantities (maximum doses- cholestyramine 24 grams per day; colestipol 30 grams per day). In contrast, colesevelam, which requires a much lower quantity of drug because of its high affinity and binding capacity for bile salts, has less of an effect on the absorption of other drugs (recommended dose of colesevelam 3.75 grams/day). Of particular note colesevelam does not interfere with absorption of statins, fenofibrate, or ezetimibe. A list of some of the drugs whose absorption is affected by cholestyramine or colestipol is shown in table 10 and a list of drugs whose absorption is affected by colesevelam is shown in table 11.

 

Table 10.  Some of the Drugs Affected by Cholestyramine/Colestipol

Statins

Ezetimibe

Gemfibrozil

Fenofibrate

Thiazides

Furosemide

Spironolactone

Digoxin

Warfarin

L-thyroxine

Corticosteroids

Vitamin K

Cyclosporine

Raloxifine

NSAIDs

Sulfonylureas

Aspirin

Beta blockers

Tricyclic

 

 

Table 11. Some of the Drugs Affected by Colesevelam

L-thyroxine

Cyclosporine

Glimepiride

Glipizide

Glyburide

Phenytoin

Olmesartan

Warfarin

Oral contraceptives

 

 

 

 

It is currently recommended that medications should be taken either 4 hours before or 4 hours after taking bile acid sequestrants. This is particularly important with drugs that have a narrow toxic/therapeutic window, such as thyroid hormone, digoxin, or warfarin. It can be very difficult for many patients, particularly those on multiple medications, to take bile acid sequestrants given the need to separate pill ingestion.

 

Cholestyramine and colestipol may also interfere with the absorption of fat-soluble vitamins. Taking a multivitamin 4 hours before or after these drugs can reduce the likelihood of a vitamin deficiency.

 

Effect of Bile Acid Sequestrants on Clinical Outcomes

 

The Lipid Research Clinics Coronary Primary Prevention Trial (LRC-CPPT) of cholestyramine vs. placebo was the first large drug study to explore the effect of specifically lowering LDL-C on cardiovascular outcomes (159). LRC-CPPT was a multicenter, randomized, double-blind study in 3,806 asymptomatic middle-aged men with primary hypercholesterolemia. The treatment group received cholestyramine 24 grams per day and the control group received a placebo for an average of 7.4 years. In the cholestyramine group total and LDL-C was decreased by 8.5% and 12.6% as compared to the placebo group. In the cholestyramine group there was a 19% reduction in risk (p < 0.05) of the primary end point accounted for by a 24% reduction in definite CHD death and a 19% reduction in nonfatal myocardial infarction. In addition, the incidence rates for new positive exercise tests, angina, and coronary bypass surgery were reduced by 25%, 20%, and 21%, respectively, in the cholestyramine group. The reduction in events correlated with the decrease in LDL-C levels (160). Of note, compliance with cholestyramine 24 grams per day was limited with many patients taking much less than the prescribed doses. These results indicate that lowering LDL-C with bile acid sequestrant monotherapy reduces cardiovascular disease.

 

In addition to the LRC-CPPT clinical outcome study, two studies have examined the effect of cholestyramine monotherapy on angiographic changes in the coronary arteries. The National Heart, Lung, and Blood Institute Type II Coronary Intervention Study and the St Thomas Atherosclerosis Regression Study reported that cholestyramine decreased the progression of atherosclerosis (161,162). There are a number of studies that have employed bile acid sequestrants in combination with other drugs and have shown a reduction in the progression of atherosclerosis or an increase in the regression of atherosclerosis but given the use of multiple drugs it is difficult to attribute the beneficial effects to the bile acid sequestrants (163-165). Unfortunately, there are no clinical outcome studies comparing statins alone vs. statins plus bile acid sequestrants.

 

Side Effects

 

Bile acid sequestrants do not have major systemic side effects as they are not absorbed and remain in the intestinal tract. However, they do cause gastrointestinal (GI) side effects (144-146). Constipation is a very common side effect and can be severe. In addition, patients will often complain of bloating, abdominal discomfort, and aggravation of hemorrhoids. Because of GI distress, a significant number of patients will discontinue therapy with bile acid sequestrants. These GI side effects are much more common with cholestyramine and colestipol compared to colesevelam, which is much better tolerated. One can reduce or ameliorate these GI side effects by increasing hydration, adding fiber to the diet (psyllium), and using stool softeners. Notably, bile acid sequestrants do not cause liver or muscle problems.

 

One should also be aware that bile acid sequestrants can be difficult for many patients to take. Colestipol and colesevelam pills are large and can be difficult for some patients to swallow. Additionally, patients need to take a large number of these pills (colesevelam- 6 pills per day; colestipol- as many as 16 pills per day). The granular forms of cholestyramine and colestipol do not dissolve and are ingested as a suspension in liquid. Many patients find mixing with water leads to an unpalatable mixture that is difficult to take. Sometimes mixing with fruit juice, apple sauce, mash potatoes, etc. make the mixture more palatable. The suspension form of colesevelam with either 1.875 or 3.75 grams is preferred by many patients.

 

As noted, earlier bile acid sequestrants can increase triglyceride levels, particularly in patients with elevated baseline triglyceride levels.

 

Contraindications

 

Bile acid sequestrants usually should be avoided in patients with pre-existing GI disorders. Bile acid sequestrants are contraindicated in patients with recent or repeated intestinal obstruction and patients with plasma triglyceride levels > 400mg/dL. In contradistinction from other lipid lowering drugs, bile acid sequestrants are not contraindicated during pregnancy or lactation (category B) (166). In women of child bearing age who are planning to become pregnant bile acid sequestrants can be a good choice to lower LDL levels.

 

Summary

 

Bile acid sequestrants are useful secondary drugs for the treatment of elevated LDL-C levels. They are typically used in combination with statin therapy as a second line drug or as an addition to statin plus ezetimibe therapy as a third line drug. In statin intolerant patients the combination of ezetimibe and a bile acid sequestrant is frequently employed. Bile acid sequestrants can be difficult drugs for patients to take due to GI side effects, difficulty taking the medication, and the need to avoid taking these drugs with other medications. To improve compliance with these drugs the clinician needs to spend time educating the patient on how to take these drugs and how to avoid side effects. Because of these difficulties other cholesterol lowering drugs are used more commonly than bile acid sequestrants. In patients with type 2 diabetes who need an improvement in glycemic control and LDL-C lowering colesevelam can be used to target both abnormalities.

 

PCSK9 MONOCLONAL ANTIBODIES

 

Introduction

 

In 2015 two monoclonal antibodies that inhibit PCSK9 (proprotein convertase subtilisin kexin type 9) were approved for the lowering of LDL-C levels. Alirocumab (Praluent) is produced by Regeneron/Sanofi and evolocumab (Repatha) is produced by Amgen (167,168). Alirocumab is administered as either 75mg or 150mg subcutaneously every 2 weeks or 300mg once a month while evolocumab is administered as either 70mg subcutaneously every 2 weeks or 420mg subcutaneously once a month.

 

Effect of PCSK inhibitors on Lipid and Lipoprotein Levels

 

There are a large number of studies that have examined the effect of PCSK9 inhibitors on lipid and lipoprotein levels. A meta-analysis of 24 studies comprising 10,159 patients reported a reduction in LDL-C levels of approximately 50% and in an increase in HDL of 5-8% (169). Notably, in 12 RCTs with 6,566 patients, Lp(a) levels were reduced by 25-30% (169). The higher the baseline Lp(a) the greater the reduction with treatment (170). It should be recognized that most LDL-C lowering drugs (statins, ezetimibe, bempedoic acid, and bile acid sequestrants) do not lower Lp(a) levels. PCSK9 inhibitors have not been shown to decrease hs-CRP levels (171).

 

MONOTHERAPY

 

Both alirocumab and evolocumab have been studied as monotherapy vs. ezetimibe. In the Mendel-2 study patients were randomly assigned to evolocumab, placebo, or ezetimibe (172). In the evolocumab group, LDL-C levels decreased by 57% while in the ezetimibe group LDL-C levels decreased by 18% compared to placebo. Additionally, non-HDL-C was decreased by 49%, apolipoprotein B by 47%, triglycerides by 5.3% (NS), and Lp(a) by 18.5% while HDL levels increased by 5.5% in the evolocumab treated subjects. In a study of alirocumab vs. ezetimibe, LDL-C levels were reduced by 47% in the alirocumab group and 16% in the ezetimibe group (173). In addition, alirocumab decreased non-HDL-C by 41%, apolipoprotein B by 37%, triglycerides by 12%, and Lp(a) by 17% and increased HDL by 6%. Thus, PCSK9 monoclonal antibodies are very effective in lowering pro-atherogenic lipoproteins when used in monotherapy and have a more robust effect than ezetimibe.

 

IN COMBINATION WITH STATINS

 

In the Odyssey Combo I study, patients on maximally tolerated statin therapy were randomized to alirocumab or placebo (174). Similar to monotherapy results, when alirocumab was added to statin therapy there was a further decrease in LDL-C levels by 46%, non-HDL-C by 38%, apolipoprotein B by 36%, and Lp(a) by 15% with an increase in HDL of 7% and no change in triglyceride levels. In the Odyssey Combo II study, patients on maximally tolerated statin therapy were randomized to alirocumab vs. ezetimibe (175). Alirocumab reduced LDL levels by 51% while ezetimibe reduced LDL by 21%, demonstrating that even when added to statin therapy, alirocumab has a significantly greater ability to reduce LDL-C levels than ezetimibe. In Odyssey Combo II, non-HDL-C levels were decreased by 42%, apolipoprotein B by 41%, triglycerides by 13%, and Lp(a) by 28% while HDL increased by 9% in the alirocumab treated group. In the Laplace-2 study, evolocumab was added to various statins used at different doses (176). It didn’t make any difference which statin was being used (atorvastatin, rosuvastatin, or simvastatin) or what dose (atorvastatin 10mg or 80mg; rosuvastatin 5mg or 40mg); the addition of evolocumab resulted in an approximately 60% further decrease in LDL-C levels beyond statin alone. Additionally, the Laplace-2 trial also showed that evolocumab was much more potent than ezetimibe when added to statin therapy (evolocumab resulted in an approximately 60% decrease in LDL vs. while ezetimibe resulted in an approximately 20-25% reduction).

 

IN COMBINATION WITH STATINS AND EZETIMIBE  

 

When evolocumab was added to patients receiving atorvastatin 80mg and ezetimibe 10mg there was 48% further reduction in LDL-C levels indicating that even in patients on very aggressive lipid lowering therapy the addition of a PCSK9 inhibitor can still result in a marked reduction in LDL-C (177). In addition to decreasing LDL-C there was also a 41% decrease in non-HDL-C, a 38% decrease in apolipoprotein B, and a 19% decrease in Lp(a) when evolocumab was added to statin plus ezetimibe therapy.

 

PATIENTS WITH HETEROZYGOUS FAMILIAL HYPERCHOLESTEROLEMIA  

 

Both alirocumab and evolocumab have been tested in patients with Heterozygous Familial Hypercholesterolemia (178,179). In the Rutherford-2 trial, evolocumab lowered LDL-C by 60%, non-HDL-C by 56%, apolipoprotein B by 49%, Lp(a) by 31%, and triglycerides by 22% while increasing HDL by 8% (178). In the Odyssey FH I and FH II studies, alirocumab lowered LDL-C by approximately 55%, non-HDL-C by ~50%, apolipoprotein B by ~43%, Lp(a) by ~19% and triglycerides by ~14% while increasing HDL by ~7% (179). Thus, in these difficult to treat patients PCSK9 monoclonal antibodies were still very effective at lowering pro-atherogenic lipoproteins.

 

PATIENTS WITH HOMOZYGOUS FAMILIAL HYPERCHOLESTEROLEMIA  

 

Evolocumab resulted in a 21-31% decrease in LDL-C levels compared to placebo in patients with Homozygous Familial Hypercholesterolemia (180,181). The response to therapy appears to be dependent on the underlying genetic cause. Patients with mutations in the LDL receptor leading to the expression of defective receptors respond to therapy whereas patients with mutations leading to negative receptors (null variants) have a poor response (180-182). Given the mechanism by which PCSK9 inhibitors lower LDL-C levels it is not surprising that patients that do not have any functional LDL receptors will not respond to therapy (see section on Mechanism of Lipid Lowering). Alirocumab decreased LDL-C by 35.6%, non-HDL-C by 32.9%, apolipoprotein B by 29.8%, and lipoprotein (a) by 28.4% (183). Given that PCSK9 monoclonal antibodies decrease LDL-C levels in some patients with Familial Hypercholesterolemia these drugs can be useful in this very difficult to treat patient population.

 

STATIN INTOLERANT PATIENTS  

 

A number of studies have examined the effect of PCSK9 monoclonal antibodies in statin intolerant patients (myalgias) and compared the response to ezetimibe treatment (102,184,185). As expected, treatment with a PCSK9 inhibitor was more effective in lowering LDL-C levels than ezetimibe. Importantly, muscle symptoms were less frequent in the PCSK9 treated patients than those treated with ezetimibe, indicating that PCSK9 monoclonal antibodies will be an effective treatment choice in statin intolerant patients with myalgias.

 

PATIENTS WITH DIABETES  

 

A meta-analysis of three trials with 413 patients with type 2 diabetes found that in patients with type 2 diabetes evolocumab caused a 60% decrease in LDL-C compared to placebo and a 39% decrease in LDL-C compared to ezetimibe treatment (186). In addition, in patients with type 2 diabetes, evolocumab decreased non-HDL-C 55% vs. placebo and 34% vs. ezetimibe) and Lp(a) (31% vs. placebo and 26% vs. ezetimibe). These beneficial effects were not affected by glycemic control, insulin use, renal function, and cardiovascular disease status. Thus, PCSK9 inhibitors are effective therapy in patients with type 2 diabetes and the beneficial effects on pro-atherogenic lipoproteins is similar to what is observed in non-diabetic patients.

 

PATIENTS WITH HYPERTRIGLYCERIDEMIA  

 

There are no studies that have examined the effect of PCSK9 monoclonal antibodies in patients with marked elevations in triglyceride levels (>400mg/dL).

 

Table 12. Effect of PCSK9 Inhibitors on Lipid/Lipoprotein Levels

LDL-C

Decrease

Non-HDL-C

Decrease

Apolipoprotein B

Decrease

Triglycerides

No change or small decrease

HDL-C

Small Increase

Lp(a)

Decrease

 

Mechanism Accounting for the PCSK9 Inhibitor Induced Lipid Effects

 

The linkage of PCSK9 with lipoprotein metabolism was first identified by Abifadel and colleagues in 2003, when they demonstrated that certain mutations in PCSK9 could result in the phenotypic appearance of Familiar Hypercholesterolemia (187). Subsequent studies demonstrated that gain of function mutations in PCSK9 are an uncommon cause of Familiar Hypercholesterolemia (167,168,188). In 2005 it was shown that loss of function mutations in PCSK9 resulted in lower LDL-C levels and this decrease in LDL-C levels was associated with a reduction in the risk of cardiovascular events (189,190).

 

The main route of clearance of clearance of plasma LDL is via LDL receptors in the liver (191). When the LDL particle binds to the LDL receptor the LDL particle- LDL receptor complex is taken into the liver by endocytosis (191). The LDL particle and the LDL receptor then disassociate and the LDL lipoprotein particle is delivered to lysosomes where it is degraded and the LDL receptor returns to the plasma membrane (Figure 2) (191). After endocytosis LDL receptors recirculate back to the plasma membrane over 100 times.

 

PCSK9 is predominantly expressed in the liver and secreted into the circulation. Once extracellular, PCSK9 can bind to the LDL receptor and alter the metabolism of the LDL receptor (192,193). Instead of the LDL receptor recycling to the plasma membrane the LDL receptor bound to PCSK9 remains associated with the LDL particle and is delivered to the lysosomes where it is also degraded (Figure 4) (192,193). This results in a decrease in the number of plasma membrane LDL receptors resulting in the decreased clearance of circulating LDL leading to elevations in plasma LDL-C levels.

 

The PCSK9 monoclonal antibodies bind PCSK9 preventing the PCSK9 from interacting with LDL receptors and thereby preventing PCSK9 from inducing LDL receptor degradation (192,193). The decreased LDL receptor degradation results in an increase in hepatic LDL receptors on the plasma membrane leading to the increased clearance of LDL and decreases in plasma LDL-C levels (194,195). Thus, similar to statins, ezetimibe, bempedoic acid, and bile acid sequestrants, PCSK9 inhibitors are reducing plasma LDL-C levels by up-regulating hepatic LDL receptors. The difference is that PCSK9 inhibitors are decreasing the degradation of LDL receptors while statins, ezetimibe, bempedoic acid, and bile acid sequestrants stimulate the production of LDL receptors.

 

Figure 4. PCSK9 Directs LDL Receptor to Degradation in Lysosome.

 

The expression of PCSK9 is stimulated by SREBP-2 (192,193). Statins and other drugs that lower hepatic cholesterol levels lead to the activation of SREBP-2 and thereby increase plasma PCSK9 levels (192,193). Inhibition of PCSK9 with monoclonal antibodies is more effective in lowering plasma LDL-C levels in patients on statin therapy due to the higher levels of plasma PCSK9 in these individuals.

 

The mechanism by which PCSK9 inhibitors reduce Lp(a) levels is unclear. Studies have shown that PCSK9 inhibitors increase the catabolism of lipoprotein(a) particles (196,197). In some circumstances PCSK9 inhibitors may also decrease the production rate (197). It has been postulated that increasing hepatic LDL receptor levels in the setting of marked reductions in circulating LDL levels will result in the clearance of Lp(a) by liver LDL receptors (198).

 

Pharmacokinetics and Drug Interactions

 

PCSK9 monoclonal antibodies are eliminated primarily by cellular endocytosis, phagocytosis, and target-mediated clearance. They are not metabolized or cleared by the liver or kidneys and therefore there is no need to adjust the dose in patients with either liver or kidney disease. There are no interactions with the cytochrome P450 system or transport proteins and thus the risk of drug-drug interactions is minimal. Currently there are no reported drug-drug interactions with PCSK9 monoclonal antibodies.

 

Effect of PCSK9 Inhibitors on Clinical Outcomes

 

FOURIER TRIAL

 

The FOURIER trial was a randomized, double-blind, placebo-controlled trial of evolocumab vs. placebo in 27,564 patients with atherosclerotic cardiovascular disease and an LDL-C level of 70 mg/dL or higher who were on statin therapy (199). The primary end point was cardiovascular death, myocardial infarction, stroke, hospitalization for unstable angina, or coronary revascularization and the key secondary end point was cardiovascular death, myocardial infarction, or stroke. The median duration of follow-up was 2.2 years. Baseline LDL-C levels were 92mg/dL and evolocumab resulted in a 59% decrease in LDL levels (LDL-C level on treatment approximately 30mg/dL). Evolocumab treatment significantly reduced the risk of the primary end point (hazard ratio, 0.85; 95% confidence interval (CI), 0.79 to 0.92; P<0.001) and the key secondary end point (hazard ratio, 0.80; 95% CI, 0.73 to 0.88; P<0.001). The results were consistent across key subgroups, including the subgroup of patients in the lowest quartile for baseline LDL-C levels (median, 74 mg/dL). Of note, a similar decrease in cardiovascular events occurred in patients with diabetes treated with evolocumab and glycemic control was not altered (200). Additionally, in patients with peripheral arterial disease evolocumab also reduced cardiovascular events (201). Further analysis has shown that in the small number of patients with a baseline LDL-C level less than 70mg/dL, evolocumab reduced cardiovascular events to a similar degree as in the patients with an LDL-C greater than 70mg/dL (202). The lower the on-treatment LDL-C levels (down to levels below 20mg/dL), the lower the cardiovascular event rate, suggesting that greater reductions in LDL-C levels will result in greater reductions in cardiovascular disease (203). Finally, the relative risk reductions with evolocumab for the cardiovascular events tended to be greater in high-risk subgroups (20% for those with a more recent MI, 18% with multiple prior MI, and 21% with residual multivessel coronary artery disease), whereas the relative risk reduction was 5% to 8% in patients without these risk factors (204). This observation suggests that certain groups of patients will derive greater benefit from the addition of a PCSK9 inhibitor.

 

It should be noted that that the duration of the FOURIER trial was very short and it is well recognized from previous statin trials that the beneficial effects of lowering LDL-C levels take time with only modest effects observed during the first year of treatment. In the FOURIER trial the reduction of cardiovascular death, myocardial infarction, or stroke was 16% during the first year but was 25% beyond 12 months.

 

ODYSSEY TRIAL

 

The ODYSSEY trial was a multicenter, randomized, double-blind, placebo-controlled trial involving 18,924 patients who had an acute coronary syndrome 1 to 12 months earlier, an LDL-C level of at least 70 mg/dL, a non-HDL-C level of at least 100 mg/dL, or an apolipoprotein B level of at least 80 mg/dL while on high intensity statin therapy or the maximum tolerated statin dose (205). Patients were randomly assigned to receive alirocumab 75 mg every 2 weeks or matching placebo. The dose of alirocumab was adjusted to target an LDL-C level of 25 to 50 mg/dL. The primary end point was a composite of death from coronary heart disease, nonfatal myocardial infarction, fatal or nonfatal ischemic stroke, or unstable angina requiring hospitalization. During the trial LDL-C levels in the placebo group was 93-103mg/dL while in the alirocumab group LDL-C levels were 40mg/dL at 4 months, 48mg/dL at 12 months, and 66mg/dL at 48 months (the increase with time was due to discontinuation of alirocumab or a decrease in dose). The primary endpoint was reduced by 15% in the alirocumab group (HR 0.85; 95% CI 0.78 to 0.93; P<0.001). In addition, total mortality was reduced by 15% in the alirocumab group (HR 0.85; 95% CI 0.73 to 0.98). The absolute benefit of alirocumab was greatest in patients with a baseline LDL-C level greater than 100mg/dL. In patients with an LDL-C level > than 100mg/dL the number needed to treat with alirocumab to prevent an event was only 16. It should be noted that the duration of this trial was very short (median follow-up 2.8 years) which may have minimized the beneficial effects. Additionally, because alirocumab 75mg every 2 weeks was stopped if the LDL-C level was < 15mg/dL on two consecutive measurements the beneficial effects may have been blunted (7.7% of patients randomized to alirocumab were switched to placebo).

 

SUMMARY OF OUTCOME TRIALS

 

It should be noted that that the duration of the PCSK9 outcome trials were relatively short and it is well recognized from previous statin trials that the beneficial effects of lowering LDL-C levels take time with only modest effects observed during the first year of treatment. In the FOURIER trial the reduction of cardiovascular death, myocardial infarction, or stroke was 16% during the first year but was 25% beyond 12 months. In the ODYSSEY trial the occurrence of cardiovascular events was similar in the alirocumab and placebo group during the first year of the study with benefits of alirocumab appearing after year one. Thus, the long-term benefits of treatment with a PCSK9 inhibitor may be greater than that observed during these relatively short-term studies.

 

GLAGOV TRIAL

 

While not an outcome trial the GLAGOV trial provides further support for the benefits of further lowering of LDL-C levels with a PCSK9 inhibitor added to statin therapy (206). This trial was a double-blind, placebo-controlled, randomized trial of evolocumab vs. placebo in 968 patients presenting for coronary angiography. The primary efficacy measure was the change in percent atheroma volume (PAV) from baseline to week 78, measured by serial intravascular ultrasonography (IVUS) imaging. Secondary efficacy measures included change in normalized total atheroma volume (TAV) and percentage of patients demonstrating plaque regression. As expected, there was a marked decrease in LDL-C levels in the evolocumab group (Placebo 93mg/dL vs. evolocumab 37mg/dL; p<0.001). PAV increased 0.05% with placebo and decreased 0.95% with evolocumab (P < .001) while TAV decreased 0.9 mm3 with placebo and 5.8 mm3 with evolocumab (P < .001). There was a linear relationship between achieved LDL-C and change in PAV (i.e., the lower the LDL-C the greater the regression in atheroma volume down to an LDL-C of 20mg/dL). Additionally, evolocumab induced plaque regression in a greater percentage of patients than placebo (64.3% vs 47.3%; P < .001 for PAV and 61.5% vs 48.9%; P < .001 for TAV). These results demonstrate the anti-atherogenic effects of PCSK9 inhibitors. Other trials in different patient populations have also shown that treatment with PCSK9 inhibitors are anti-atherogenic (207,208). 

 

VENOUS THROMBOEMBOLISM

 

In the FOURIER trial treatment with evolocumab resulted in a reduction in venous thromboembolism (VTE) (HR 0.71; 95% CI, 0.50-1.00; P=0.05) (209). Interestingly no effect was observed in the 1st year (HR, 0.96; 95% CI, 0.57-1.62) but a 46% reduction in VTE (HR, 0.54; 95% CI, 0.33-0.88; P=0.014) beyond 1 year occurred. In patients with low baseline Lp(a) levels, evolocumab reduced Lp(a) by only 7 nmol/L and had no effect on VTE risk but in patients with high baseline Lp(a) levels, evolocumab reduced Lp(a) by 33 nmol/L and risk of VTE by 48% (HR, 0.52; 95% CI, 0.30-0.89; P=0.017). In the ODYSSEY OUTCOMES trial, the risk of VTE was reduced but just missed being statistically significant (HR, 0.67; 95% CI, 0.44-1.01; P=0.06) (210). A meta-analysis of FOURIER and ODYSSEY OUTCOMES demonstrated a 31% relative risk reduction in VTE with PCSK9 inhibition (HR, 0.69; 95% CI, 0.53-0.90; P=0.007) (209).

 

Side Effects

 

The major side effect of PCSK9 monoclonal antibodies has been injection site reactions including erythema, itching, swelling, pain, and tenderness. Allergic reactions have been reported and as with any protein there is potential immunogenicity. In general side effects have been minimal, which is not surprising, as monoclonal antibodies do not typically have off target side effects. Since PCSK9 does not appear to have important functions other than regulating LDL receptor degradation, it is not surprising that inhibiting PCSK9 function has not resulted in major side effects.

 

A meta-analysis of 20 randomized controlled trials with 68,123 subjects found a very modest effect on fasting glucose (mean difference 1.88 mg/dL) and A1c levels (mean difference 0.032%) and did not observe an increased risk of developing diabetes (211). It should be recognized that the duration of these trials was relatively short (median follow-up 78 weeks) and therefore further long-term studies are required.

 

In the large outcome trials (ODYSSEY and FOURIER) there was no significant difference between the PCSK9 treated group vs. the placebo group with regard to adverse events (including new-onset diabetes and neurocognitive events). The only exception was the expected increase in injection-site reactions in the patients treated with a PCSK9 inhibitor. Additionally, in a subgroup of patients from the FOURIER trial a prospective study of cognitive function (EBBINGHAUS Study) was carried out and no significant differences in cognitive function was observed over a median of 19 months in the PCSK9 treated vs. placebo group (212). It should be recognized that while short-term treatment with PCSK9 inhibitors have not demonstrated any significant side effects it is possible that long-term use could lead to unexpected side-effects.

 

An issue of concern is whether lowering LDL-C to very low levels has the potential to cause toxicity. In a number of the PCSK9 studies a significant number of patients had LDL-C levels < 25mg/dL. For example, in the Odyssey long term study 37% of patients on alirocumab had two consecutive LDL-C levels below 25mg/dL and in the Osler long term study in patients treated with evolocumab 13% had values below 25mg/dL (213,214). In these short term PCSK9 studies, toxicity from very low LDL-C levels has not been observed. Additionally, in patients with Familial Hypobetalipoproteinemia LDL levels can be very low and these patients do not have any major disorders other than hepatic steatosis, which is not mechanistically due to low LDL-C levels (215). Similarly, there are rare individuals who are homozygous for loss of function mutations in the PCSK9 gene and they also do not appear to have major medical issues (168). Finally, in a number of statin trials there have been patients with very low LDL-C levels and an increased risk of side effects has not been consistently observed in those patients (216-218). Thus, with the limited data available there does not appear to be a major risk of markedly lowering LDL-C levels.   

 

Contraindications

 

Other than a history of a hypersensitivity to these drugs there are currently no contraindications. There are no studies during pregnancy or lactation.

 

Summary

 

PCSK9 monoclonal antibodies robustly reduce LDL-C levels when used as monotherapy, in combination with statins, or when added to the combination of statins + ezetimibe. In distinction to most other cholesterol lowering drugs the PCSK9 inhibitors also decrease Lp(a) levels. Outcome studies have clearly demonstrated that decreasing LDL-C levels with PCSK9 inhibitors reduces cardiovascular events. The side effect profile appears to be very favorable and there are no drug-drug interactions. The major limitation is the high expense of these drugs, which has limited their widespread use.

 

INCLISIRAN (LEQVIO)

 

Introduction

 

Inclisiran (Leqvio) is a double-stranded, siRNA (small interfering RNA) conjugated on the sense strand with triantennary N-acetylgalactosamine (GalNAc) to facilitate uptake into hepatocytes (219). In hepatocytes, inclisiran stimulates the catalytic breakdown of PCSK9 mRNA thereby reducing the hepatic synthesis of PCSK9 and markedly decreasing plasma PCSK9 levels (219,220). The recommended dose of inclisiran is 284 mg by subcutaneous injection, followed with a repeat injection at 3 months, and then every 6 months (package insert). If a dose is missed by more than 3 months it is recommended to repeat the dosage schedule described above (package insert). It is recommended that inclisiran be administered by a healthcare professional.

 

Effect on Inclisiran on Lipid and Lipoprotein Levels

 

There have been several large trials examining the efficacy of inclisiran. The ORION-10 trial was conducted in the United States and included adults with atherosclerotic cardiovascular disease on a maximally tolerated statin with an LDL-C > 70mg/dL (220). Patients were randomized to inclisiran 284mg (n=781) at initial visit, 3 months, 9 months, and 15 months or placebo (n=780) and followed for 540 days. After 3 months the LDL-C was reduced by approximately 50% and this reduction was sustained throughout the duration of the trial (at 540 days the LDL-c was reduced by 52.3% (P<0.001)). As expected, total cholesterol (-33%), non-HDL-C (-47%), and apolipoprotein B (-43%) were also decreased. Additionally, triglyceride (-13%) and Lp(a) (-26%) levels were decreased while HDL-C levels (+5.1%) and hsCRP (+8.8%) were slightly increased. ORION-11 was a very similar trial with an identical protocol conducted in Europe and South Africa and included adults with ASCVD or an ASCVD risk equivalent on maximally tolerated statin therapy (inclisiran n=810 and placebo n=807) (220). At 540 days LDL-C was reduced by 49.9% (P<0.001). Changes in other lipid parameters were similar to those observed in ORION 10. Subgroup analysis revealed that in both the ORION 10 and 11 trials that all subgroups had a similar reduction in LDL-C levels with inclisiran therapy including subjects with diabetes, moderate renal impairment, and greater than 75 years of age (220). Statin therapy and whether statin therapy was moderate intensity or high intensity also did not affect the reduction in LDL-C (220). Additionally, in patients with renal disease, including individuals with an estimated creatinine clearance between 15-29 mL/min, the reduction in LDL-C levels with inclisiran administration were similar to individuals with normal renal function (221). The decrease in LDL-C with inclisiran treatment has been shown to persist for 4 years (222).

 

HETEROZYGOUS FAMILIAL HYPERCHOLESTEROLEMIA

 

The effect of inclisiran on LDL-C levels was determined in patients with heterozygous familial hypercholesterolemia who were randomized to receive subcutaneous injections of inclisiran 284mg (n= 242) or placebo (n=240) on days 1, 90, 270, and 450 (223). The mean baseline LDL-C level was 153±54mg/dL and 90% of the patients were receiving statins with most on high intensity statins (75%). At day 510 LDL-C levels were reduced by 47.9% compared to placebo (P<0.001). The reduction in LDL-C was similar in all genotypes of familial hypercholesterolemia. Total cholesterol was reduced by 33%, non-HDL-C by 44%, Lp(a) by 17.2%, and triglycerides by 12%. HDL-C and hsCRP were not markedly altered.

 

HOMOZYGOUS FAMILIAL HYPERCHOLESTEROLEMIA

 

A small study reported that inclisiran treatment lowered LDL-C levels in 3 of 4 patients with homozygous familiar hypercholesterolemia  (17.5% to 37% decrease) but less than that seen in individuals with fully functioning LDL receptors (224). A larger more recent trial failed to demonstrate a decrease in LDL-C levels with inclisiran treatment (225). Of note there was considerable variation in the LDL-C response, which could be due to differences in genetic variants. Individuals with null-null LDL receptor variants (i.e. no functioning LDL receptors) are unlikely to respond to inclisiran due to the absence of LDL receptors and the group treated with inclisiran in this study was enriched in patients with this genotype, which could explain the absence of a significant reduction in LDL-C.     

 

Mechanisms Accounting for Inclisiran Induced Lipid Effects

 

The mechanism of action of inclisiran is the same as for PCSK9 monoclonal antibodies (219). Briefly, decreasing the production of PCSK9 in the liver, the primary source of circulating PCSK9, leads to a decrease in plasma PCSK9 levels resulting in a decrease in LDL receptor degradation (219). An increase in the number of hepatic LDL receptors increases the clearance of LDL leading to a decrease in LDL-C levels (219).  

 

Pharmacokinetics and Drug Interactions

 

There are no drug interactions. The reduction in LDL-C occurs within 14 days after drug administration and persists for an extended period of time allowing for administration every 6 months.

 

Effect of Inclisiran on Clinical Outcomes

 

No outcome studies are currently available. A cardiovascular outcome study (ORION-4) is ongoing and includes 15,000 patients with established ASCVD. The trial duration is five years and completion is expected in 2024 (NCT03705234) (ClinicalTrials.gov, 2020a).

 

Side Effects

 

The only adverse reactions associated with inclisiran were injection site reactions including rash, pain, and erythema (220). In an analysis of 7 studies with 3,576 patients treated with inclisiran for up to 6 years and 1,968 patients treated with placebo for up to 1.5 years, hepatic, muscle, and kidney events; incident diabetes; and elevations of creatine kinase or creatinine were not increased in patients treated with inclisiran (226).

 

Contraindications

 

In patients with severe hepatic or renal impairment inclisiran should be used with caution as there is limited data and experience in these patients. There are no studies during pregnancy or lactation.

 

Summary

 

Inclisiran very effectively lowers LDL-C levels. The major advantage of this drug compared to PCSK9 monoclonal antibodies is the ability to administer inclisiran every 6 months, which may improve compliance.

 

BEMPEDOIC ACID (NEXLETOL)

 

Introduction

 

Bempedoic acid was approved in the US in February 2020 and is an adenosine triphosphate-citrate lyase (ACL) inhibitor. It is administered orally once daily with or without food at a dose of 180mg (Nexletol). It is also available as a combination tablet containing 180 mg of bempedoic acid and 10 mg of ezetimibe (Nexlizet).

 

Effect on Bempedoic on Lipid and Lipoprotein Levels

 

EFFECT WITHOUT STATINS

 

In a study that randomized 345 patients with hypercholesterolemia (LDL-C 158mg/dL) and a history of intolerance to statin to either bempedoic acid or placebo (2:1), bempedoic acid decreased LDL-C by 21.4%, non-HDL-C by 17.9%, and apolipoprotein B by 15% (227). One third of patients were on background non-statin therapy most commonly ezetimibe and fish oil. Triglyceride levels were not altered but there was a small decrease in HDL-C levels that was statistically significant (-4.5%).

 

IN COMBINATION WITH STATINS

 

There have been two large trials that determined the effect of adding bempedoic acid to statin therapy. In a study that randomized 779 patients on maximally tolerated statin therapy +/- ezetimibe (only a small number on ezetimibe) with an LDL-C level greater than 70mg/dL (baseline LDL-C 120mg/dL) to either bempedoic acid or placebo it was observed that bempedoic acid decreased LDL-C levels by 17.4% compared to placebo (p<0.001) (228). In addition, non-HDL-C and apolipoprotein B levels were decreased by 13% compared to placebo while there was no significant change in triglyceride levels. Bempedoic acid decreased HDL-C levels by approximately 6%. In a similar study, patients with atherosclerotic cardiovascular disease, heterozygous familial hypercholesterolemia, or both with an LDL-C level greater than 70 mg/dL (baseline LDL-C 103mg/dL) while on maximally tolerated statin therapy with or without additional lipid-lowering therapy (only a small number on ezetimibe) were randomized to bempedoic acid (n= 1,488) or placebo (n= 742) (229). Compared to placebo, treatment with bempedoic acid decreased LDL-C by 18.1%, non-HDL-C by 13.5%, and apolipoprotein B by 11.9%. Triglyceride levels were unchanged but HDL-C decreased by 5.92%. Of note in both of the above studies the decrease in LDL-C was maintained over 52 weeks.

 

Notably, the addition of bempedoic acid to atorvastatin 80mg per day was still capable of significantly decreasing LDL-C (22%), non-HDL-C (13%), and apolipoprotein B (-15%) compared to placebo (230). The addition of bempedoic acid to high dose atorvastatin therapy did not cause meaningful changes in atorvastatin pharmacokinetics.   

 

IN COMBINATION WITH EZETIMIBE

 

Patients on maximally tolerated statin therapy with LDL-C levels greater 100 mg/dL if they had cardiovascular disease and/or Familiar Hypercholesterolemia or greater than 130 mg/dL if they had multiple CVD risk factors were randomized to bempedoic acid + ezetimibe, bempedoic acid alone, ezetimibe alone, or placebo (231). The key results of this study are shown in Table 14. Changes from baseline in HDL-C and triglyceride level were modest (<10%) in all treatment groups. In another study patients with a history of statin intolerance on ezetimibe therapy were randomized to bempedoic acid (n=181) or placebo (n= 88) (232). Compared to placebo, bempedoic acid decreased LDL-C by 28.5%, non-HDL-C by -23.6%, and apolipoprotein B by -19.3%. As seen in other studies bempedoic acid did not alter triglyceride levels but slightly decreased HDL-C levels (approximately 6% decrease compared to placebo).

 

Table 14. Effect of Bempedoic Acid and Ezetimibe on Lipid Parameters (231)

 

LDL-C

Non-HDL-C

Apo B

hsCRP

Bempedoic acid + ezetimibe

-38%

-33.7%

-30.1

-35.1

Bempedoic acid

-19%

-15.9%

-17.3

-31.9

Ezetimibe

-25%

-21.7

-20.8

-8.2

 Results are percent decrease compared to the placebo group.

 

Summary

 

Bempedoic acid typically lowers LDL-C by 15-25%, non-HDL-C by 10-20%, and apolipoprotein B levels by 10-20% with no significant effects on triglyceride levels. HDL-C levels decrease by 5-8% and Lp(a) are unchanged (233).

 

Table 15. Effect of Bempedoic Acid on Lipid/Lipoprotein Levels

LDL-C

Decrease

Non-HDL-C

Decrease

Apolipoprotein B

Decrease

Triglycerides

No change

HDL-C

Small decrease

Lp(a)

No change

 

Non-Lipid Effects of Bempedoic Acid

 

Bempedoic acid decreases hsCRP levels (see table 14, 16).

 

Table 16. Effect of Bempedoic Acid on hsCRP Levels

Reference

Percent decrease in hsCRP

(227)

-24.3

(228)

-8.7

(229)

-21.5

(230)

-44

(232)

-31

 

In the CLEAR Outcome study with a median follow-up of 3.4 years there was no difference in the development of new onset diabetes in the bempedoic acid and placebo groups (429 of 3848, -11·1% with bempedoic acid vs 433 of 3749, 11·5% with placebo; HR 0.95; 95% CI 0.83-1.09) (234). Additionally, during the study HbA1c concentrations and fasting glucose levels were similar between the bempedoic acid and placebo groups in patients who had either prediabetes or normoglycemia. In the CLEAR Outcome study in patients with diabetes the prevalence of worsening diabetes was similar in the bempedoic acid and placebo group (235,236)   

 

Mechanisms Accounting for Bempedoic Acid Induced Lipid Effects

 

Bempedoic acid is a potent inhibitor of ATP-citrate lyase, which catalyzes the formation of acetyl-CoA in the cytoplasm (237). Acetyl-CoA is a precursor for the synthesis of cholesterol (figure 5). The inhibition of ATP-citrate lyase by bempedoic acid decreases cholesterol synthesis in liver reducing hepatic intracellular cholesterol levels (237). Of note, bempedoic acid is a pro-drug and conversion to its CoA-derivative by very-long-chain acyl-CoA synthetase-1 is required for inhibition of cholesterol synthesis (237). Very-long-chain acyl-CoA synthetase-1 is highly expressed in the liver but is not expressed in adipose tissue, kidney, intestine or skeletal muscle (237). The inability of bempedoic acid to be activated in muscle and inhibit cholesterol synthesis suggests that bempedoic acid is unlikely result in muscle toxicity.

 

Figure 5. Inhibition of Cholesterol Synthesis by Bempedoic Acid.

 

The decrease in plasma LDL-C levels in patients treated with bempedoic acid is primarily due to an increase in hepatic LDL receptors secondary to the inhibition of cholesterol synthesis resulting in a reduction in hepatic cholesterol levels (237). It should be noted that bempedoic acid also decreases circulating LDL-C levels in LDL receptor deficient mice and LDL receptor deficient miniature pigs indicating that mechanisms in addition to up-regulation of hepatic LDL receptors may contribute to the decrease in LDL-C levels (237). The inhibition of hepatic cholesterol synthesis may decrease the production and secretion of VLDL, which could contribute to a decrease in LDL-C.

 

Pharmacokinetics and Drug Interactions

 

No dose adjustments are required in patients with mild or moderate renal or hepatic impairment or in the elderly (package insert). Concomitant use of bempedoic acid with simvastatin or pravastatin causes an increase in the concentrations of these drugs and therefore may increase the risk of myopathy (package insert). This drug interaction may be secondary to bempedoic acid inhibiting organic anion-transporting polypeptide OATP1B1. It is recommended to avoid concomitant use of bempedoic acid with simvastatin greater than 20 mg/day or pravastatin 40mg/day. While concomitant administration of bempedoic acid with atorvastatin or rosuvastatin elevated the area under the curve by 1.7-fold these elevations were generally within the individual statin exposures and do not impact dosing recommendations (package insert).

 

Effect of Bempedoic Acid on Clinical Outcomes

 

In animal models of atherosclerosis, treatment with bempedoic acid had favorable effects on atherosclerosis (237). Moreover, genetic variants of ATP citrate lyase that lower LDL-C levels are associated with a decrease in cardiovascular disease suggesting that bempedoic acid will have favorable effects on reducing the risk of cardiovascular disease (238).

 

The CLEAR Outcome trial was a double-blind, randomized, placebo-controlled trial involving patients with cardiovascular disease or at high risk of cardiovascular disease who were unable or unwilling to take statins ("statin-intolerant" patients) (239). The patients were randomized to bempedoic acid 180 mg (n= 6992) or placebo (n= 6978) and the median duration of follow-up was 40.6 months. As expected, LDL-C levels were decreased by 21% in the bempedoic group compared to placebo (29mg/dL difference). The primary endpoint, death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularization, was reduced by 13% in the bempedoic acid group (HR 0.87; 95% CI 0.79 to 0.96; P = 0.004). Bempedoic acid also decreased fatal and non-fatal myocardial infarctions and coronary revascularization but had no significant effects on fatal or nonfatal stroke, death from cardiovascular causes, and death from any cause. In the patients who were at high risk for cardiovascular disease (primary prevention), 66% had diabetes, and the primary endpoint was reduced by 30% in the bempedoic acid group (HR 0.70; 95% CI, 0.55-0.89; P = .002) (235). In patients with diabetes with or without cardiovascular disease the primary endpoint was reduced by 17% in the bempedoic acid group (HR 0.83; 95% CI 0.72-0.95) (234). This study clearly demonstrates that treatment with bempedoic acid reduces the risk cardiovascular events.     

 

Side Effects

 

HYPERURICEMIA

 

In clinical trials, 26% of bempedoic acid-treated patients with normal baseline uric acid values experienced hyperuricemia one or more times versus 9.5% in the placebo group (package insert). In the CLEAR Outcomes trial elevated uric acid levels occurred in 10.9% of the patients on bempedoic acid compared to 5.6% taking the placebo (239). The increase in uric acid is due to bempedoic acid inhibiting renal tubular OAT2. The Increase in uric acid levels typically occurred within the first 4 weeks of treatment and persisted throughout treatment. After 12 weeks of treatment, the mean placebo-adjusted increase in uric acid compared to baseline was 0.8 mg/dL for patients treated with bempedoic acid (package insert). Elevations in blood uric acid levels may lead to the development of gout. Gout was reported in 1.5% of patients treated with bempedoic acid vs. 0.4% of patients treated with placebo. The risk for gout attacks were higher in patients with a prior history of gout (11.2% for bempedoic acid treatment vs. 1.7% in the placebo group) (package insert). In patients with no prior history of gout only 1% of patients treated with bempedoic acid and 0.3% of the placebo group had a gouty attack (package insert). In the CLEAR Outcomes trial gout was increased in the bempedoic acid group (3.1% vs. 2.1%) (239).

 

TENDON RUPTURE

 

In clinical trials tendon rupture occurred in 0.5% of patients treated with bempedoic acid vs. 0% of placebo treated patients and involved the rotator cuff (the shoulder), biceps tendon, or Achilles tendon (package insert). Tendon rupture occurred within weeks to months of starting bempedoic acid and occurred more frequently in patients over 60 years of age, in those taking corticosteroid or fluoroquinolone drugs, in patients with renal failure, and in patients with previous tendon disorders. In the CLEAR Outcomes trial tendon rupture was similar in the bempedoic acid and placebo group (bempedoic acid 1.2% and placebo 0.9%) (239).

 

RENAL FUNCTION

 

Bempedoic acid treatment resulted in a mean increase in serum creatinine of 0.05 mg/dL compared to baseline. Approximately 3.8% of patients treated with bempedoic acid had BUN levels that doubled vs. 1.5% in the placebo group and about 2.2% of patients treated with bempedoic acid had creatinine values that increased by 0.5 mg/dL vs. 1.1% in the placebo group (package insert). Renal function returned to baseline when bempedoic acid was discontinued. In the CLEAR Outcomes trial renal impairment was increased in the bempedoic acid group (11.5% vs.8.6%) as was the change from baseline creatinine (0.05±0.2 mg/dL vs. 0.01±0.2 mg/dL)  (239).

 

CHOLELITHIASIS

 

In the CLEAR Outcomes trial cholelithiasis was increased in the bempedoic acid group (2.2 vs 1.2) (239).

 

BENIGN PROSTATIC HYPERPLASIA

 

Bempedoic acid was associated with an increased risk of benign prostatic hyperplasia (BPH) in men with no reported history of BPH, occurring in 1.3% of NEXLETOL-treated patients versus 0.1% of placebo-treated patients (package insert).

 

MISCELLANEOUS LABORATORY ABNORMALITIES

 

Approximately 5.1% of patients on bempedoic acid vs. 2.3% on placebo had decreases in hemoglobin levels of 2 or more g/dL and below the lower limit of normal on one or more occasion. Anemia was reported in 2.8% of patients treated with bempedoic acid and 1.9% of patients treated with placebo. Hemoglobin decrease was generally asymptomatic and did not require medical intervention (package insert).

 

Approximately 9.0% of bempedoic acid treated patients with a normal baseline leukocyte count decreased leukocyte count to less than the lower limit of normal on one or more occasions vs. 6.7% in the placebo group. The leukocyte decrease was generally asymptomatic and did not require medical intervention (package insert).

 

Approximately 10.1% of bempedoic acid treated patients vs. 4.7% in the placebo group had

increases in platelet counts of 100× 109/L or more on one or more occasion. The platelet count increase was asymptomatic, did not result in an increased risk for thromboembolic events, and did not require medical intervention (package insert).

 

Increases to more than 3× the upper limit of normal (ULN) in AST occurred in 1.4% of patients treated with bempedoic acid vs. 0.4% of placebo patients, and increases to more than 5× ULN occurred in 0.4% of bempedoic acid treated patients vs. 0.2% of placebo-treated patients. Increases in ALT were similar in bempedoic acid treated patients and placebo-treated patients. Elevations in transaminases were generally asymptomatic and not associated with elevations ≥2× ULN in bilirubin or with cholestasis. In most cases, the elevations were transient and resolved or improved with continued therapy or after discontinuation of therapy (package insert).

 

Contraindications

 

The use of bempedoic acid during pregnancy and lactation has not been studied (package insert).

 

Summary

 

In patients on statins and ezetimibe with an LDL-C that is not at goal the addition of bempedoic acid is a reasonable third drug. In addition, in patients that cannot tolerate statin therapy the combination of ezetimibe and bempedoic acid may allow for the lowering of LDL-C to goal. One can expect a reduction in LDL-C of approximately 15-25% with bempedoic acid monotherapy therapy or when used in combination with other LDL-C lowering drugs.

 

LOMITAPID (JUXTAPID)

 

Introduction

 

Lomitapide (Juxtapid), a selective microsomal triglyceride transfer protein inhibitor, was approved in December 2012 for lowering LDL-C levels in adults with Homozygous Familial Hypercholesterolemia (240-242). As will be discussed below it lowers LDL-C levels by an LDL receptor independent mechanism.

 

Effect on Lomitapide on Lipid and Lipoprotein Levels

 

The effect of lomitapide on lipid and lipoprotein levels has been studied in patients with Homozygous Familial Hypercholesterolemia. The pivotal study was a 78-week single arm open label study in 29 patients receiving treatment for Homozygous Familial Hypercholesterolemia (243). Lomitapide was initiated at 5mg per day and was up-titrated to 60mg per day based on tolerability and liver function tests. On an intention to treat basis, LDL-C was decreased by 40% and apolipoprotein B by 39%. In patients who were actually taking lomitapide, LDL-C levels were reduced by 50%. In addition to decreasing LDL-C levels, non-HDL-C levels were decreased by 50%, Lp(a) by 15%, and triglycerides by 45%. Interestingly HDL and apolipoprotein A-I levels were decreased by 12% and 14% respectively in this study. Follow-up revealed that the decrease in LDL-C could be sustained for a prolonged period of time (294 weeks) (244).

 

The effect of lomitapide has also been studied in patients without Homozygous Familial Hypercholesterolemia. A study by Samaha and colleagues compared the effect of ezetimibe and lomitapide in patients with elevated cholesterol levels(245). Patients were treated with ezetimibe alone, lomitapide alone, or the combination of ezetimibe and lomitapide. Ezetimibe monotherapy led to a 20–22% decrease in LDL-C levels, lomitapide monotherapy led to a dose dependent decrease in LDL-cholesterol levels (19% at 5.0 mg, 26% at 7.5 mg and 30% at 10 mg). Combined therapy produced a larger dose-dependent decrease in LDL-C levels (35%, 38% and 46%, respectively).  Additionally, lomitapide decreased triglycerides by 10%, non-HDL-C by 27%, apolipoprotein B by 24%, and Lp(a) by 17%.

 

The above studies demonstrate that lomitapide decreases LDL-C, non-HDL-C, triglycerides, and Lp(a) levels.

 

Mechanism Accounting for the Lomitapide Induced Lipid Effects

 

Lomitapide is a selective inhibitor of microsomal triglyceride transfer protein (MTTP) (240-242). MTTP is located in the endoplasmic reticulum of hepatocytes and enterocytes where it plays a key role in transferring triglycerides onto newly synthesized apolipoprotein B leading to the formation of VLDL and chylomicrons (246). Loss of function mutations in both alleles of MTTP results in abetalipoproteinemia, which is characterized by the virtual absence of apolipoprotein B, VLDL, chylomicrons, and LDL in the plasma due to the failure of the liver and intestine to produce VLDL and chylomicrons (215). Lomitapide by inhibiting MTTP activity reduces the secretion of chylomicrons by the intestine and VLDL by the liver leading to a decrease in LDL, apolipoprotein B, triglycerides, non-HDL-C, and Lp(a) (240-242). 

 

Pharmacokinetics and Drug Interactions

 

Lomitapide is extensively metabolized in the liver by the CYP3A4 pathway (240,241). Therefore, lomitapide is contraindicated in patients on strong CYP3A4 inhibitors and lower doses should be used in patients on weak inhibitors. Of particular note, in patients on atorvastatin the maximal dose of lomitapide is 30mg per day and lomitapide should not be used in patients taking more than 20mg of simvastatin (240,241). Lomitapide can increase warfarin levels and therefore close monitoring is required. Finally, given the risk of liver abnormalities (see side effect section) the avoidance of alcohol or a reduction in alcohol intake is prudent.

 

Effect of Lomitapide on Clinical Outcomes

 

There are no clinical outcome trials but it is presumed that lowering LDL-C levels in patients with Homozygous Familial Hypercholesterolemia will reduce cardiovascular events. After initiating lomitapide therapy 1.7 cardiovascular events per 1000 patient months on treatment was observed vs. 26.1 cardiovascular events per 1000 patient months in a comparison cohort (247).

 

Side Effects

 

As expected from its mechanism of action lomitapide causes side effects in the GI tract and liver. In the GI tract diarrhea, nausea, vomiting, and dyspepsia occur very commonly (240-242). In the pivotal study in patients with Homozygous Familial Hypercholesterolemia, 90% of the patients developed GI symptoms during drug titration (243). GI side effects are potentiated by high fat meals and it is therefore recommended that dietary fat be limited. Approximately 10% of patients will discontinue lomitapide, mostly from diarrhea. Lomitapide also reduces the absorption of fat soluble vitamins and therefore patients need to take vitamin supplements (240,241). Additionally, it may also block the absorption of essential fatty acids and it is therefore recommended that supplements of essential fatty acids also be provided (at least 200 mg linoleic acid, 210 mg alpha-linolenic acid (ALA), 110 mg eicosapentaenoic acid (EPA), and 80 mg docosahexaenoic acid (DHA) (240,241).

 

Blocking the formation of VLDL in the liver can lead to fatty liver with elevated liver enzymes (240-242). Approximately 30% of patients will develop increased transaminase levels but in the small number of patients studied this has not resulted in liver failure. After stopping the drug, the transaminases have returned to normal. Whether long term treatment with lomitapide will lead to an increase in liver disease is unknown. There is a single case of a patient with lipoprotein lipase deficiency who was treated for 13 years with lomitapide who developed steatohepatitis and fibrosis (248). In an observational study of a small number of patients on lomitapide for > 5 years liver failure or cirrhosis was not noted (249). In another study in Italy, 34 patients were treated with lomitapide for more than 9 years and elevations in hepatic fat were mild-to-moderate, hepatic stiffness remained normal, and the mean FIB-4 score remained below the fibrosis threshold (250). The studies suggest that in most patients’ severe liver disease will not develop. To reduce the risk of liver dysfunction it is important that patients avoid or limit alcohol intake and avoid drugs that inhibit Cyp3A4 activity.

 

Because of the high potential risk of serious complications the FDA has mandated several measures to ensure that patients are closely followed and monitored for liver toxicity ((Risk Evaluation and Mitigation Strategy (REMS) Program) (240,241). ALT, AST, alkaline phosphatase, and total bilirubin should be measured before initiating treatment. During the first year, liver function tests should be measured prior to each increase in dose or monthly, whichever occurs first. After the first year, liver function tests should be measured at least every 3 months and before any increase in dose.

 

Contraindications

 

Lomitapide should not be used during pregnancy and in patients with moderate or severe liver disease. In addition, it should not be used in patients on strong CYP3A4 inhibitors.

 

Summary

 

Lomitapide is approved only for the treatment of lipid disorders in patients with Homozygous Familiar Hypercholesterolemia. The frequent GI side effects and the potential risk of serious liver disease greatly limit the use of this drug and it should be reserved for the patients in which more benign therapies are not sufficient in lowering LDL-C into a reasonable range. It is used as an adjunct to other lipid lowering therapies and lipoprotein apheresis in patients with Homozygous Familiar Hypercholesterolemia.

 

MIPOMERSEN (KYNAMRO)

 

Introduction

 

Mipomersen (Kynamro) is a second generation apolipoprotein B antisense oligonucleotide that was approved in January 2013 for the treatment of patients with Homozygous Familiar Hypercholesterolemia (241,242,251). It is administered as a 200mg subcutaneous injection once a week (241,242,251). As will be discussed below, it lowers LDL-C levels by an LDL receptor independent mechanism. In May 2018 sales were discontinued due to safety concerns related to increased liver transaminases and fatty liver.

 

Effect on Mipomersen on Lipid and Lipoprotein Levels

 

In the pivotal trial, 51 patients with Homozygote Familial Hypercholesterolemia on treatment were randomized to additional treatment with mipomersen (n= 34) or placebo (n=17) and followed for 26 weeks (252). Mipomersen lowered LDL-C levels by 21% and apolipoprotein B levels by 24% compared to placebo. In addition, non-HDL-C was decreased by 21.6%, triglycerides by 17%, and Lp(a) by 23% while HDL and apolipoprotein A-I were increased by 11.2% and 3.9% respectively.

 

Mipomersen has also been studied in patients with Heterozygous Familial Hypercholesterolemia. In a double-blind, placebo-controlled, randomized trial, patients on maximally tolerated statin therapy were treated weekly with subcutaneous mipomersen 200 mg or placebo for 26 weeks (253). LDL-C levels decreased by 33% in the mipomersen group compared to placebo. Additionally, mipomersen significantly reduced apolipoprotein B by 26%, triglycerides by 14%, and Lp(a) by 21% compared to placebo with no significant changes in HDL-C levels. In an extension follow-up study the beneficial effects of mipomersen were maintained for at least 2 years (254). 

 

In a meta-analysis of 8 randomized studies with 462 subjects with either non-specified hypercholesterolemia or Heterozygous Familial Hypercholesterolemia, Panta and colleagues reported that mipomersen decreased LDL-C levels by 32% compared to placebo (255). Additionally, non-HDL-C was decreased by 31%, apolipoprotein B by 33%, triglycerides by 36%, and Lp(a) by 26% with no effect on HDL-C levels.

 

Mechanism Accounting for the Mipomersen Induced Lipid Effects

 

Apolipoprotein B 100 is the main structural protein of VLDL and LDL and is required for the formation of VLDL and LDL (191). Familiar Hypobetalipoproteinemia is a genetic disorder due to a mutation of one apolipoprotein B allele that is characterized by very low concentrations of LDL and apolipoprotein B due to the decreased production of lipoproteins by the liver (215). Mipomersen, an apolipoprotein B antisense oligonucleotide, mimics Familiar Hypobetalipoproteinemia by inhibiting apolipoprotein B 100 production in the liver by pairing with apolipoprotein B mRNA preventing its translation (241,242,251). This decrease in apolipoprotein B synthesis results in a decrease in hepatic VLDL production leading to a decrease in LDL levels.

 

Pharmacokinetics and Drug Interactions

 

No significant drug interactions have been reported. Given the risk of liver abnormalities (see side effect section) the avoidance of alcohol or a reduction in alcohol intake would be prudent.

 

Effect of Mipomersen on Clinical Outcomes

 

There are no clinical outcome trials but it is presumed that lowering LDL-C levels in patients with Homozygous Familial Hypercholesterolemia will reduce cardiovascular events. In a study comparing cardiovascular events in patients with Homozygous Familial Hypercholesterolemia in the 24 months prior to initiating mipomersen therapy and after initiating mipomersen revealed a decrease in events (prior to treatment 61.5% of patients had an event vs. 9.6% after initiating mipomersen; P < .0001) (256). In this trial mipomersen resulted in a mean absolute reduction in LDL-C of 70 mg/dL (-28%), non-HDL cholesterol of 74 mg/dL (-26%), and Lp(a) of 11 mg/dL (-17%).

 

Side Effects

 

The most common side effect is injection site reactions, which occur in 75-98% of patients and typically consist of one or more of the following: erythema, pain, tenderness, pruritus, and local swelling (241,242,251).  Additional, influenza like symptoms, which typically occur within 2 days after an injection, occur in 30-50% of patients and include one or more of the following: influenza-like illness, pyrexia, chills, myalgia, arthralgia, malaise or fatigue which result in a substantial percentage of patients discontinuing therapy (241,242,251).

 

A major safety concern is liver toxicity (241,242,251). By inhibiting VLDL formation and secretion the risk of fatty liver is increased. Fatty liver has been observed in 5-20% of patients treated with mipomersen (241,242,251). In 10-15% of patients treated with mipomersen increases in transaminases occur (241,242,251). Additionally, liver biopsies from 7 patients after a minimum of 6 months of mipomersen therapy have demonstrated the presence of fatty liver although there was no inflammation despite elevations in liver enzymes (257). Liver function should be measured prior to initiating therapy and monthly during the first year and every 3 months after the first year. Fortunately, when treatment is discontinued liver function tests and fatty liver return to normal.

 

Because of the potential for liver toxicity this drug is no longer available.

 

Contraindications

 

Mipomersen is contraindicated in patients in patients with liver disease or severe renal disease. Mipomersen is not recommended for use during pregnancy or lactation. In animal studies mipomersen has not resulted in fetal abnormalities.

 

Summary

 

Mipomersen was approved only for the treatment of lipid disorders in patients with Homozygous Familiar Hypercholesterolemia. The potential risk of serious liver disease greatly limits the use of this drug and therefore it was reserved for patients in which more benign therapies were not sufficient in lowering LDL-C into a reasonable range. It was used as an adjunct to other lipid lowering therapies in patients with Homozygous Familiar Hypercholesterolemia but because of safety concerns is no longer available.

 

EVINACUMAB (EVKEEZA)

 

Introduction

 

Evinacumab is a human monoclonal antibody against angiopoietin-like protein 3 (ANGPTL3). It is approved for the treatment of Homozygous Familial Hypercholesterolemia. Evinacumab decreases LDL-C levels by mechanisms independent of LDL receptor activity. The recommended dose of evinacumab is 15 mg/kg administered by intravenous infusion over 60 minutes every 4 weeks.

 

Effect on Evinacumab on Lipid and Lipoprotein Levels

 

HOMOZYGOUS FAMILIAL HYPERCHOLESTEROLEMIA

 

A double-blind, placebo-controlled trial randomly treated patients with Homozygous Familial Hypercholesterolemia with an intravenous infusion of evinacumab 15 mg/Kg every 4 weeks (n= 43) or placebo (n= 22) (258). The individuals in this trial were on lipid lowering therapy (94% were on a statin with 77% on a high-intensity statin, 77% on a PCSK9 inhibitor, 75% on ezetimibe, 25% on lomitapide, and 34% undergoing apheresis) and the mean baseline LDL-C level was approximately 250-260mg/dL. After 24 weeks of treatment patients in the evinacumab group had a 47% reduction in LDL-C levels vs. a 1.9% increase in the placebo group (table 17). This decrease in LDL-C levels was observed after 2 weeks of therapy and was observed regardless of concomitant use of other lipid lowering drugs or apheresis. Notably, in individuals with null-null LDL receptor variants evinacumab resulted in a 43% decrease in LDL-C levels indicating that evinacumab therapy was effective in the absence of functional LDL receptors. As expected, total cholesterol, non-HDL-C cholesterol, and apo B levels were also decreased. Moreover, triglyceride levels decreased 55% and HDL-C levels decreased 30% with evinacumab administration while Lp(a) levels were unchanged.

 

Table 17. Effect of Evinacumab on Lipid Levels in Homozygous Familial Hypercholesterolemia

 

LDL-C

Apo B

Non-HDL-C

TG

HDL-C

Baseline mg/dL

255

171

278

124

44

Evinacumab % Change

−47%

−41%

−50%

−55%

−30%

Placebo  % Change

+2%

−5%

+2%

−5%

+1%

 

REFRACTORY HYPERCHOLESTEROLEMIA

 

In a double-blind, placebo-controlled trial, patients with refractory hypercholesterolemia with a screening LDL-C level > 70 mg/dL with atherosclerosis or LDL-C > 100 mg/dL without atherosclerosis were randomized to receive subcutaneous or intravenous evinacumab or placebo (259). The hypercholesterolemia was refractory to treatment with a PCSK9 inhibitor and a statin at a maximum tolerated dose, with or without ezetimibe. In this trial a number of different treatment regimens of evinacumab were employed (intravenous or subcutaneous; different doses) and in this summary only the results of intravenous evinacumab 15 mg/kg every 4 weeks (39 patients) vs. placebo (34 patients) will be presented. Baseline LDL-C levels were approximately 145mg/dL. After 16 weeks of treatment the LDL-C level was decreased by 50% with evinacumab administration vs. a 0.6% decrease with placebo. An extension of this trial for 72 weeks found that the reduction in LDL-C were sustained (260). The decrease in LDL-C was observed after 2 weeks of treatment. As expected, total cholesterol, non-HDL-C, and apo B levels also decreased in the evinacumab group. Evinacumab administration decreased triglyceride levels by 53% and HDL-C levels by 31%. In contrast to the results in the homozygous familiar hypercholesterolemia study described above in this study evinacumab decreased Lp(a) levels by 16%. The effect of the subcutaneous administration of evinacumab on lipid levels was similar to that observed with intravenous administration.

 

The effect of evinacumab on triglyceride levels in patients with marked hypertriglyceridemia is described in the Endotext chapter “Triglyceride Lowering Drugs” (261).

 

Mechanism Accounting for the Evinacumab Induced Lipid Effects

 

ANGPTL3 inhibits lipoprotein lipase (LPL) activity thereby slowing the clearance of VLDL and chylomicrons resulting in an increase in plasma triglyceride levels (262,263). Mice deficient in ANGPTL3 have lower plasma triglyceride levels while mice overexpressing ANGPTL3 have elevated plasma triglyceride levels (263). Evinacumab by inhibiting the ability of ANGPTL3 to inhibit LPL activity will accelerate the clearance of TG rich lipoproteins decreasing plasma triglyceride levels (263). Furthermore, ANGPTL3 has also been shown to reduce endothelial lipase activity (263). Endothelial lipase is a phospholipase that catabolizes phospholipids on HDL and accelerates HDL clearance (264,265). Evinacumab by inhibiting the ability of ANGPTL3 to inhibit endothelial lipase activity will lead to a decrease in HDL levels (266).

 

The mechanism(s) that explain the decrease in LDL-C levels with evinacumab administration is not completely understood. A study has demonstrated that the increase in endothelial lipase activity induced by evinacumab leads to VLDL remodeling and lipid depletion that increases VLDL clearance when the LDL receptor is absent (267). This decrease in VLDL, the precursor of LDL, limits LDL particle production resulting in a reduction in plasma LDL-C levels (267). Kinetic studies in four patients with homozygous familial hypercholesterolemia observed that evinacumab markedly increased the fractional catabolic rate of IDL (intermediate-density lipoprotein) and LDL apoB (268). Whether decreases in VLDL production also plays a role in the decrease in LDL-C levels with evinacumab treatment requires additional studies. It should be noted that inhibition of ANGPTL3 decreases LDL-C levels independent of LDL receptor activity (269).

 

Pharmacokinetics and Drug Interactions

 

There are no significant drug interactions.

 

Effect of Evinacumab on Clinical Outcomes

 

There are no cardiovascular outcome studies. In two patients with homozygous Familial Hypercholesterolemia evinacumab therapy markedly reduced LDL-C levels with a concomitant decrease in plaque volume determined by coronary computed tomography angiography (268).  

 

Homozygosity for loss-of-function mutations in ANGPTL3 is associated with significantly lower plasma levels of LDL-C, HDL-C, and triglycerides (familial combined hypolipidemia) (215,263,270). Heterozygous carriers of loss-of-function mutations in ANGPTL3, which occur at a frequency of about 1:300, have significantly lower total cholesterol, LDL-C, and triglyceride levels than noncarriers (263). Moreover, patients carrying loss-of-function variants in ANGPTL3 have a significantly lower risk of coronary artery disease (271,272). Additionally, in an animal model of atherosclerosis treatment with evinacumab decreased atherosclerotic lesion area and necrotic content (271). Taken together these observations suggest that inhibiting ANGPTL3 with evinacumab will reduce cardiovascular disease.

 

Side Effects

 

Serious hypersensitivity reactions have occurred with evinacumab. In clinical trials, 1 (1%) of evinacumab treated patients experienced anaphylaxis vs. 0% of patients who received placebo (package insert).

 

Contraindications

 

Based on animal studies, evinacumab may cause fetal harm when administered to pregnant patients (package insert). Patients should be advised of the potential risks to the fetus of pregnancy. Patients who may become pregnant should be advised to use effective contraception during treatment with evinacumab and for at least 5 months following the last dose.

 

Summary

 

In patients with Homozygous Familiar Hypercholesterolemia the ability of evinacumab to lower LDL-C levels independent of LDL receptor activity makes this agent very useful in these patients. Most patients with Homozygous Familial Hypercholesterolemia do not achieve goal LDL-C levels with triple drug therapy with maximally tolerated statin therapy, ezetimibe, and a PCSK9 inhibitor and therefore the addition of evinacumab will be needed in many of these patients. Evinacumab is also effective in patients with refractory hypercholesterolemia but the drug is not yet FDA approved in this situation. Nevertheless, one can foresee in patients with refractory hypercholesterolemia at high risk for cardiovascular events the use of evinacumab. In addition to lowering LDL-C levels evinacumab also lowers triglyceride levels and could be useful in selected patients with very severe hypertriglyceridemia (261,273).  

 

APPROACH TO TREATING PATIENTS WITH HYPERCHOLESTEROLEMIA

 

Introduction

 

The issues of deciding who to treat, how aggressive to treat, and the goals of therapy are discussed in detail in the chapter “Guidelines for the Management of High Blood Cholesterol” and therefore will not be addressed in this chapter (3). Additionally, the role of life style changes to lower LDL-C is discussed in great depth in chapter “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” and therefore will also not be addressed here (1). Rather we will focus on how to use the drugs discussed in this chapter to treat various categories of patients. The factors to consider when deciding which drugs are appropriate to use for lowering plasma LDL-C levels are; the efficacy in lowering LDL-C levels, the effect on other lipid and lipoprotein levels, the ability to reduce cardiovascular events, the side effects of drug therapy, the ease of complying with the drug regimen, and the cost of the drugs. Many statins and ezetimibe are generic drugs and therefore they are relatively inexpensive.

 

Isolated Hypercholesterolemia with Cardiovascular Disease

 

In patients with isolated hypercholesterolemia and cardiovascular disease, initial drug therapy should be high intensity statin therapy (atorvastatin 40-80mg or rosuvastatin 20-40mg). In patients with cardiovascular disease, one should aim to lower the LDL-C to below 70mg/dL. Many experts, based on studies comparing statin alone vs. statin + ezetimibe or statin + a PCSK9 inhibitor, would recommend a more aggressive LDL-C goal in high-risk patients (LDL-C <55mg/dL). If statin therapy alone is not sufficient adding ezetimibe, is a reasonable next step. Because a considerable amount of data indicates that the lower the LDL-C the greater the reduction in cardiovascular events many experts would use a combination of high intensity statin therapy plus ezetimibe in all high-risk patients to maximize LDL-C reduction. Ezetimibe is inexpensive, easy to take, has few side effects, will modestly lower LDL-C, and has been shown in combination with statins to further reduce cardiovascular events. High dose statin and ezetimibe will lower LDL-C by as much as 70%, which will lower LDL-C to goal in a large number of patients who do not have a genetic basis for their elevated LDL-C levels. If the combination of statin plus ezetimibe does not lower the LDL to goal one can add a third drug. If the LDL is close to goal, one could add a bile acid sequestrant such as colesevelam or bempedoic acid. If the LDL is not very close to goal one could instead use a statin +/- ezetimibe plus a PCSK9 inhibitor, which will result in marked reductions in LDL-C levels. If the patient has diabetes with a moderately elevated A1c level using a bile acid sequestrant such as colesevelam instead of ezetimibe or in combination with ezetimibe could improve both glycemic control and further lower LDL levels. If the cost of PCSK9 inhibitors decrease the earlier use of these drugs will become feasible.

 

Isolated Hypercholesterolemia in Primary Prevention

 

In patients with isolated hypercholesterolemia (LDL-C < 190mg/dL) without cardiovascular disease initial drug therapy is with a statin. The statin dose should be chosen based on the percent reduction in LDL-C required to lower the LDL-C level to below the target goal (typically < 100mg/dL but if multiple risk factors with a high risk for cardiovascular events is present many experts would aim for <70mg/dL). As discussed earlier, the side effects of statin therapy increase with higher doses so one should not automatically start with high doses, but instead should choose a dose balancing the benefits and risks. Generic statins are inexpensive drugs and are very effective in both lowering LDL-C levels and reducing cardiovascular events. Additionally, they have an excellent safety profile. If the initial statin dose does not lower LCL-C sufficiently, one can then increase the dose or add ezetimibe. If the maximal statin dose does not lower LDL-C sufficiently adding ezetimibe is a reasonable next step if the LDL-C level is in a reasonable range and an additional 20-25% reduction in LDL will be sufficient. High dose statin and ezetimibe will lower LDL-C by as much as 70%, which will lower LDL-C to goal in the majority of patients who do not have a genetic basis for their elevated LDL-C levels. If the combination of statin plus ezetimibe does not lower the LDL-C to goal one can add a third drug, such as bempedoic acid or colesevelam. If the patient has diabetes with a moderately elevated A1c level using colesevelam instead of ezetimibe or in combination with ezetimibe could improve both glycemic control and further lower LDL-C levels.

 

Mixed Hyperlipidemia

 

In patients with mixed hyperlipidemia (elevated LDL-C and triglyceride levels) Initial drug therapy should also be a generic statin unless triglyceride levels are greater than 500-1000mg/dL. If triglycerides are > 500-1000mg/dL initial therapy is directed at lowering triglyceride levels (261). In addition to lowering LDL-C levels, statins are also effective in lowering triglyceride levels particularly when the triglycerides are elevated. If LDL-C is not lowered sufficiently ezetimibe is a reasonable next step. Bile acid sequestrants are not appropriate drugs in patients with hypertriglyceridemia. The approach to the patient whose LDL-C levels are at goal but the triglycerides and non-HDL-C are still elevated is discussed in the chapter on triglyceride lowering drugs (261).

 

Heterozygous Familial Hypercholesterolemia

 

In patients with Heterozygous Familial Hypercholesterolemia or other disorders with very elevated LDL-C levels (>190mg/dL), high doses of a potent statin such as atorvastatin 40-80mg or rosuvastatin 20-40mg are the first step to lower LDL-C levels. In many patients this will not be sufficient. If the LDL-C levels are above goal then adding ezetimibe is a reasonable next step. If after ezetimibe the LDL-C is still slightly above goal triple drug therapy with bempedoic acid or a bile acid sequestrant can be employed. If on statin alone or with the combination of statin and ezetimibe the LDL-C still needs to be markedly reduced a PCSK9 inhibitor may be a better choice as these drugs can markedly lower LDL-C levels.

 

Homozygous Familiar Hypercholesterolemia

 

In patients with Homozygous Familiar Hypercholesterolemia initial therapy with a maximally tolerated statin and ezetimibe can be instituted. This will likely not result in an acceptable LDL-C level and then one can add a PCSK9 inhibitor. Because these therapies depend on LDL receptor activity to lower LDL-C a high percentage of patients will not reach goal and then one can add lomitapide and/or evinacumab, drugs that lower LDL-C levels independent of LDL receptor activity. Because side effects are fewer with evinacumab this is the preferred initial drug in most patients. Studies have shown that with the addition of evinacumab many patients will reach acceptable LDL-C levels. If LDL-C levels are still not acceptable one could then initiate lipoprotein apheresis (274).  

 

Statin Intolerance

 

Statin intolerance is frequently due to myalgias but on occasion can be due other issues, such as increased liver or muscle enzymes, cognitive dysfunction, or other neurological disorders. The percentage of patients who are “statin intolerant” varies greatly but in clinical practice a significant number of patients have difficulty taking statins.

 

As discussed earlier it can be difficult to determine if the muscle symptoms that occur when a patient is taking a statin are actually due to the statin or are unrelated to statin use. The first step in a “statin intolerant patient” is to take a careful history of the nature and location of the muscle symptoms and the timing of onset in relation to statin use to determine whether the presentation fits the typical picture for statin induced myalgias. The characteristic findings with a statin induced myalgia are shown in table 18 and findings that are not typical for statin induced myalgia are shown in table 19. The disappearance of symptoms within a few weeks of stopping statins and the reappearance after restarting statins is very suggestive of the symptoms being due to true statin intolerance. An on-line tool (htpp://tools.acc.org/statinintolerance/#!/) and an app produced by the ACC/AHA are available. This tool characterizes patients based on 8 criteria into possible vs. unlikely to have statin induced muscle symptoms (table 20)

 

Table 18. Characteristic Findings with Statin Induced Myalgia

Symmetric

Proximal muscles

Muscle pain, tenderness, weakness, cramps

Symptom onset < 4 weeks after starting statin or dose increase

Improves within 2-4 weeks of stopping statin

Cramping is unilateral and involves small muscles of hands and feet

Same symptoms occur with re-challenge within 4 weeks

 

Table 19. Symptoms Atypical in Statin Induced Myalgia

Unilateral

Asymmetric

Small muscles

Joint or tendon pain

Shooting pain, muscle twitching or tingling

Symptom onset > 12 weeks

No improvement after discontinuing statin

 

Table 20. Diagnosis of Statin Associated Muscle Symptoms

Symptom timing

Symptom type

Symptom location

Sex

Age

Race/ethnicity

CK elevation > 5 times the upper limit of normal

Known risk factors for statin induced muscle symptoms and non-statin causes of muscle symptoms

 

One should also check a CK level but this is almost always in the normal range. If the CK is not elevated and the symptoms do not suggest a statin induced myalgia one can often reassure the patient and continue statin therapy. This is often successful and studies have shown that many patients that stop taking statins due to “statin induced myalgia” can be successfully treated with a statin. If the CK is elevated it should be repeated after instructing the patient to avoid exercise for 48 hours. Also, the CK levels should be compared to CK levels prior to starting therapy. If the CK remains elevated (3x upper limit of normal) the statin should be discontinued. Similarly, if the CK is normal but the symptoms are suggestive of a statin induced myalgia the statin should also be discontinued. The next step is to determine if one can identify reversible factors that could be increasing statin toxicity (hypothyroidism, drug interactions).  If none are identified the next step after the myalgias have resolved is to try a low dose of a different statin that is metabolized by a different pathway (for example instead of atorvastatin, which is metabolized by the CYP3A4 pathway, rosuvastatin, which has a different pathway of metabolism). Because statin side effects are dose related, a low dose of a statin may often be tolerated. One can also try several different statins as sometimes a patient may tolerate one statin and not others. A meta-analysis has shown that every other day administration of statins is as effective as daily administration in lowering lipid levels and therefore is a very reasonable strategy (275). In some instances, using a long-acting statin (rosuvastatin or atorvastatin) 1-3 times per week can work (we usually start with once per week and then slowly increase frequency as tolerated) (276). In these circumstances (low doses or 1-3 times per week) the reduction in LDL-C may not be sufficient but one can use combination therapy with other drugs such as ezetimibe, bempedoic acid, bile acid sequestrants, or PCSK9 inhibitors to achieve LDL target goals.

 

Many providers have combined Coenzyme Q10 with statins to prevent statin induced myalgias. However, randomized trials with Coenzyme Q10 supplementation have not consistently shown benefit (277-282). A trial, which carefully screened patients to make sure they actually had statin induced myalgias, failed to show a benefit from Coenzyme Q10 supplementation (101). It has also been recommended that vitamin D supplementation be used to prevent statin induced myalgias but a large randomized trial failed to show a reduction in muscle symptoms with vitamin D therapy (283).

 

If after trying various approaches a patient still has myalgias and is unable to tolerate statin therapy one needs to utilize other approaches to lower LDL levels. Similarly, if there are other reasons why a patient cannot take a statin, such as developing muscle pathology, one will also need to utilize other approaches to lower LDL levels. These patients can be treated with ezetimibe, bempedoic acid, bile acid sequestrants, or PCSK 9 inhibitors either as monotherapy or in combination to achieve LDL goals.

 

There are patients who will refuse statins and other drug therapy because they do not believe in taking pharmaceuticals but will take natural products. In these patients we have employed red yeast rice, which decreases LDL-C because it contains a form of lovastatin (284,285). It is effective but one should recognize that the quality control is not similar to the standards of pharmaceutical products and that there can be batch to batch variations. Furthermore, there is a risk of drug-drug interactions if used with inhibitors of CYP3A4. However, in this particular patient population, who refuses to take statins or other drugs, this can be a reasonable alternative. If a patient just refuses statins (usually based on a belief that statins are toxic) we will employ other cholesterol lowering drugs.

 

CONCLUSIONS

 

With currently available drugs to lower LDL-C levels we are now able to markedly reduce LDL-C levels and achieve our LDL-C goals in the vast majority of patients and thereby reduce the risk of cardiovascular disease. Patients with Homozygous Familial Hypercholesterolemia and some patients with Heterozygous Familial Hypercholesterolemia still present major clinical challenges and it can be very difficult in these patients to achieve LDL-C goals.

 

ACKNOWLEDGEMENTS

 

This work was supported by grants from the Northern California Institute for Research and Education.

 

REFERENCES

 

  1. Feingold KR. The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  2. Feingold KR. Approach to the Patient with Dyslipidemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  3. Grundy SM, Feingold KR. Guidelines for the Management of High Blood Cholesterol. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  4. Endo A. A gift from nature: the birth of the statins. Nat Med 2008; 14:1050-1052
  5. Alberts AW. Discovery, biochemistry and biology of lovastatin. Am J Cardiol 1988; 62:10J-15J
  6. Ballantyne CM, Andrews TC, Hsia JA, Kramer JH, Shear C, Efficacy ASGACC, Safety S. Correlation of non-high-density lipoprotein cholesterol with apolipoprotein B: effect of 5 hydroxymethylglutaryl coenzyme A reductase inhibitors on non-high-density lipoprotein cholesterol levels. Am J Cardiol 2001; 88:265-269
  7. Jones PH, Hunninghake DB, Ferdinand KC, Stein EA, Gold A, Caplan RJ, Blasetto JW. Effects of rosuvastatin versus atorvastatin, simvastatin, and pravastatin on non-high-density lipoprotein cholesterol, apolipoproteins, and lipid ratios in patients with hypercholesterolemia: additional results from the STELLAR trial. Clin Ther 2004; 26:1388-1399
  8. Jones PH, Davidson MH, Stein EA, Bays HE, McKenney JM, Miller E, Cain VA, Blasetto JW. Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* Trial). Am J Cardiol 2003; 92:152-160
  9. Stein EA, Lane M, Laskarzewski P. Comparison of statins in hypertriglyceridemia. Am J Cardiol 1998; 81:66B-69B
  10. Bos S, Yayha R, van Lennep JE. Latest developments in the treatment of lipoprotein (a). Curr Opin Lipidol 2014; 25:452-460
  11. van Capelleveen JC, van der Valk FM, Stroes ES. Current therapies for lowering lipoprotein (a). J Lipid Res 2016; 57:1612-1618
  12. Adams SP, Tsang M, Wright JM. Lipid-lowering efficacy of atorvastatin. Cochrane Database Syst Rev 2015; 3:CD008226
  13. Adams SP, Sekhon SS, Wright JM. Lipid-lowering efficacy of rosuvastatin. Cochrane Database Syst Rev 2014; 11:CD010254
  14. Liao JK. Clinical implications for statin pleiotropy. Curr Opin Lipidol 2005; 16:624-629
  15. Joshi PH, Jacobson TA. Therapeutic options to further lower C-reactive protein for patients on statin treatment. Curr Atheroscler Rep 2010; 12:34-42
  16. Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell 2015; 161:161-172
  17. Huff MW, Burnett JR. 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors and hepatic apolipoprotein B secretion. Curr Opin Lipidol 1997; 8:138-145
  18. Tsimikas S, Gordts P, Nora C, Yeang C, Witztum JL. Statin therapy increases lipoprotein(a) levels. Eur Heart J 2020; 41:2275-2284
  19. Causevic-Ramosevac A, Semiz S. Drug interactions with statins. Acta Pharm 2013; 63:277-293
  20. Hu M, Tomlinson B. Evaluation of the pharmacokinetics and drug interactions of the two recently developed statins, rosuvastatin and pitavastatin. Expert Opin Drug Metab Toxicol 2014; 10:51-65
  21. Sirtori CR. The pharmacology of statins. Pharmacol Res 2014; 88:3-11
  22. Bellosta S, Corsini A. Statin drug interactions and related adverse reactions: an update. Expert Opin Drug Saf 2018; 17:25-37
  23. Awad K, Serban MC, Penson P, Mikhailidis DP, Toth PP, Jones SR, Rizzo M, Howard G, Lip GYH, Banach M. Effects of morning vs evening statin administration on lipid profile: A systematic review and meta-analysis. J Clin Lipidol 2017; 11:972-985 e979
  24. Kellick KA, Bottorff M, Toth PP, The National Lipid Association's Safety Task Force. A clinician's guide to statin drug-drug interactions. J Clin Lipidol 2014; 8:S30-46
  25. Lee JW, Morris JK, Wald NJ. Grapefruit Juice and Statins. Am J Med 2016; 129:26-29
  26. Holdaas H, Fellstrom B, Jardine AG, Holme I, Nyberg G, Fauchald P, Gronhagen-Riska C, Madsen S, Neumayer HH, Cole E, Maes B, Ambuhl P, Olsson AG, Hartmann A, Solbu DO, Pedersen TR. Effect of fluvastatin on cardiac outcomes in renal transplant recipients: a multicentre, randomised, placebo-controlled trial. Lancet 2003; 361:2024-2031
  27. ACCORD Study Group, Ginsberg HN, Elam MB, Lovato LC, Crouse JR, 3rd, Leiter LA, Linz P, Friedewald WT, Buse JB, Gerstein HC, Probstfield J, Grimm RH, Ismail-Beigi F, Bigger JT, Goff DC, Jr., Cushman WC, Simons-Morton DG, Byington RP. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010; 362:1563-1574
  28. Busti AJ, Bain AM, Hall RG, 2nd, Bedimo RG, Leff RD, Meek C, Mehvar R. Effects of atazanavir/ritonavir or fosamprenavir/ritonavir on the pharmacokinetics of rosuvastatin. J Cardiovasc Pharmacol 2008; 51:605-610
  29. Gervasoni C, Riva A, Rizzardini G, Clementi E, Galli M, Cattaneo D. Potential association between rosuvastatin use and high atazanavir trough concentrations in ritonavir-treated HIV-infected patients. Antivir Ther 2015; 20:449-451
  30. Kiser JJ, Gerber JG, Predhomme JA, Wolfe P, Flynn DM, Hoody DW. Drug/Drug interaction between lopinavir/ritonavir and rosuvastatin in healthy volunteers. J Acquir Immune Defic Syndr 2008; 47:570-578
  31. Pham PA, la Porte CJ, Lee LS, van Heeswijk R, Sabo JP, Elgadi MM, Piliero PJ, Barditch-Crovo P, Fuchs E, Flexner C, Cameron DW. Differential effects of tipranavir plus ritonavir on atorvastatin or rosuvastatin pharmacokinetics in healthy volunteers. Antimicrob Agents Chemother 2009; 53:4385-4392
  32. van der Lee M, Sankatsing R, Schippers E, Vogel M, Fatkenheuer G, van der Ven A, Kroon F, Rockstroh J, Wyen C, Baumer A, de Groot E, Koopmans P, Stroes E, Reiss P, Burger D. Pharmacokinetics and pharmacodynamics of combined use of lopinavir/ritonavir and rosuvastatin in HIV-infected patients. Antivir Ther 2007; 12:1127-1132
  33. Sarkar S, Brown TT. Lipid Disorders in People with HIV. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  34. Baigent C, Keech A, Kearney PM, Blackwell L, Buck G, Pollicino C, Kirby A, Sourjina T, Peto R, Collins R, Simes R, Cholesterol Treatment Trialists Collaborators. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet 2005; 366:1267-1278
  35. Cholesterol Treatment Trialists Collaboration, Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, Bhala N, Peto R, Barnes EH, Keech A, Simes J, Collins R. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 2010; 376:1670-1681
  36. Cholesterol Treatment Trialists Collaboration, Fulcher J, O'Connell R, Voysey M, Emberson J, Blackwell L, Mihaylova B, Simes J, Collins R, Kirby A, Colhoun H, Braunwald E, La Rosa J, Pedersen TR, Tonkin A, Davis B, Sleight P, Franzosi MG, Baigent C, Keech A. Efficacy and safety of LDL-lowering therapy among men and women: meta-analysis of individual data from 174,000 participants in 27 randomised trials. Lancet 2015; 385:1397-1405
  37. Cholesterol Treatment Trialists Collaboration, Mihaylova B, Emberson J, Blackwell L, Keech A, Simes J, Barnes EH, Voysey M, Gray A, Collins R, Baigent C. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet 2012; 380:581-590
  38. Taylor F, Huffman MD, Macedo AF, Moore TH, Burke M, Davey Smith G, Ward K, Ebrahim S. Statins for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 2013; 1:CD004816
  39. Yusuf S, Bosch J, Dagenais G, Zhu J, Xavier D, Liu L, Pais P, Lopez-Jaramillo P, Leiter LA, Dans A, Avezum A, Piegas LS, Parkhomenko A, Keltai K, Keltai M, Sliwa K, Peters RJ, Held C, Chazova I, Yusoff K, Lewis BS, Jansky P, Khunti K, Toff WD, Reid CM, Varigos J, Sanchez-Vallejo G, McKelvie R, Pogue J, Jung H, Gao P, Diaz R, Lonn E. Cholesterol Lowering in Intermediate-Risk Persons without Cardiovascular Disease. N Engl J Med 2016;
  40. Ference BA. Mendelian randomization studies: using naturally randomized genetic data to fill evidence gaps. Curr Opin Lipidol 2015; 26:566-571
  41. Brown MS, Goldstein JL. Biomedicine. Lowering LDL--not only how low, but how long? Science 2006; 311:1721-1723
  42. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394
  43. Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM, Cobbe SM, Ford I, Gaw A, Hyland M, Jukema JW, Kamper AM, Macfarlane PW, Meinders AE, Norrie J, Packard CJ, Perry IJ, Stott DJ, Sweeney BJ, Twomey C, Westendorp RG. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623-1630
  44. Cholesterol Treatment Trialists Collaboration. Efficacy and safety of statin therapy in older people: a meta-analysis of individual participant data from 28 randomised controlled trials. Lancet 2019; 393:407-415
  45. Joseph J, Pajewski NM, Dolor RJ, Sellers MA, Perdue LH, Peeples SR, Henrie AM, Woolard N, Jones WS, Benziger CP, Orkaby AR, Mixon AS, VanWormer JJ, Shapiro MD, Kistler CE, Polonsky TS, Chatterjee R, Chamberlain AM, Forman DE, Knowlton KU, Gill TM, Newby LK, Hammill BG, Cicek MS, Williams NA, Decker JE, Ou J, Rubinstein J, Choudhary G, Gazmuri RJ, Schmader KE, Roumie CL, Vaughan CP, Effron MB, Cooper-DeHoff RM, Supiano MA, Shah RC, Whittle JC, Hernandez AF, Ambrosius WT, Williamson JD, Alexander KP. Pragmatic evaluation of events and benefits of lipid lowering in older adults (PREVENTABLE): Trial design and rationale. J Am Geriatr Soc 2023; 71:1701-1713
  46. Zoungas S, Curtis A, Spark S, Wolfe R, McNeil JJ, Beilin L, Chong TT, Cloud G, Hopper I, Kost A, Nelson M, Nicholls SJ, Reid CM, Ryan J, Tonkin A, Ward SA, Wierzbicki A. Statins for extension of disability-free survival and primary prevention of cardiovascular events among older people: protocol for a randomised controlled trial in primary care (STAREE trial). BMJ Open 2023; 13:e069915
  47. Liao JK. Safety and efficacy of statins in Asians. Am J Cardiol 2007; 99:410-414
  48. Gupta M, Braga MF, Teoh H, Tsigoulis M, Verma S. Statin effects on LDL and HDL cholesterol in South Asian and white populations. J Clin Pharmacol 2009; 49:831-837
  49. Sakamoto T, Kojima S, Ogawa H, Shimomura H, Kimura K, Ogata Y, Sakaino N, Kitagawa A. Effects of early statin treatment on symptomatic heart failure and ischemic events after acute myocardial infarction in Japanese. Am J Cardiol 2006; 97:1165-1171
  50. Pais P, Jung H, Dans A, Zhu J, Liu L, Kamath D, Bosch J, Lonn E, Yusuf S. Impact of blood pressure lowering, cholesterol lowering and their combination in Asians and non-Asians in those without cardiovascular disease: an analysis of the HOPE 3 study. Eur J Prev Cardiol 2019; 26:681-697
  51. Feingold KR. Dyslipidemia in Patients with Diabetes. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  52. Cholesterol Treatment Trialists Collaboration, Kearney PM, Blackwell L, Collins R, Keech A, Simes J, Peto R, Armitage J, Baigent C. Efficacy of cholesterol-lowering therapy in 18,686 people with diabetes in 14 randomised trials of statins: a meta-analysis. Lancet 2008; 371:117-125
  53. Cholesterol Treatment Trialists Collaboration , Herrington WG, Emberson J, Mihaylova B, Blackwell L, Reith C, Solbu MD, Mark PB, Fellstrom B, Jardine AG, Wanner C, Holdaas H, Fulcher J, Haynes R, Landray MJ, Keech A, Simes J, Collins R, Baigent C. Impact of renal function on the effects of LDL cholesterol lowering with statin-based regimens: a meta-analysis of individual participant data from 28 randomised trials. Lancet Diabetes Endocrinol 2016; 4:829-839
  54. Su X, Zhang L, Lv J, Wang J, Hou W, Xie X, Zhang H. Effect of Statins on Kidney Disease Outcomes: A Systematic Review and Meta-analysis. Am J Kidney Dis 2016;
  55. Wanner C, Krane V, Marz W, Olschewski M, Mann JF, Ruf G, Ritz E. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 2005; 353:238-248
  56. Fellstrom BC, Jardine AG, Schmieder RE, Holdaas H, Bannister K, Beutler J, Chae DW, Chevaile A, Cobbe SM, Gronhagen-Riska C, De Lima JJ, Lins R, Mayer G, McMahon AW, Parving HH, Remuzzi G, Samuelsson O, Sonkodi S, Sci D, Suleymanlar G, Tsakiris D, Tesar V, Todorov V, Wiecek A, Wuthrich RP, Gottlow M, Johnsson E, Zannad F. Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med 2009; 360:1395-1407
  57. Rosenstein K, Tannock LR. Dyslipidemia in Chronic Kidney Disease. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2022.
  58. Kjekshus J, Apetrei E, Barrios V, Bohm M, Cleland JG, Cornel JH, Dunselman P, Fonseca C, Goudev A, Grande P, Gullestad L, Hjalmarson A, Hradec J, Janosi A, Kamensky G, Komajda M, Korewicki J, Kuusi T, Mach F, Mareev V, McMurray JJ, Ranjith N, Schaufelberger M, Vanhaecke J, van Veldhuisen DJ, Waagstein F, Wedel H, Wikstrand J. Rosuvastatin in older patients with systolic heart failure. N Engl J Med 2007; 357:2248-2261
  59. Tavazzi L, Maggioni AP, Marchioli R, Barlera S, Franzosi MG, Latini R, Lucci D, Nicolosi GL, Porcu M, Tognoni G. Effect of rosuvastatin in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial. Lancet 2008; 372:1231-1239
  60. Arvind A, Osganian SA, Cohen DE, Corey KE. Lipid and Lipoprotein Metabolism in Liver Disease. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA) 2019.
  61. Bays H, Cohen DE, Chalasani N, Harrison SA, The National Lipid Association's Statin Safety Task Force. An assessment by the Statin Liver Safety Task Force: 2014 update. J Clin Lipidol 2014; 8:S47-57
  62. Herrick C, Bahrainy S, Gill EA. Statins and the Liver. Endocrinol Metab Clin North Am 2016; 45:117-128
  63. Athyros VG, Tziomalos K, Gossios TD, Griva T, Anagnostis P, Kargiotis K, Pagourelias ED, Theocharidou E, Karagiannis A, Mikhailidis DP. Safety and efficacy of long-term statin treatment for cardiovascular events in patients with coronary heart disease and abnormal liver tests in the Greek Atorvastatin and Coronary Heart Disease Evaluation (GREACE) Study: a post-hoc analysis. Lancet 2010; 376:1916-1922
  64. Cohen DE, Corey KE. Lipid and Lipoprotein Metabolism in Liver Disease. In: De Groot LJ, Beck-Peccoz P, Chrousos G, Dungan K, Grossman A, Hershman JM, Koch C, McLachlan R, New M, Rebar R, Singer F, Vinik A, Weickert MO, eds. Endotext. South Dartmouth (MA) 2019.
  65. Grinspoon SK, Fitch KV, Zanni MV, Fichtenbaum CJ, Umbleja T, Aberg JA, Overton ET, Malvestutto CD, Bloomfield GS, Currier JS, Martinez E, Roa JC, Diggs MR, Fulda ES, Paradis K, Wiviott SD, Foldyna B, Looby SE, Desvigne-Nickens P, Alston-Smith B, Leon-Cruz J, McCallum S, Hoffmann U, Lu MT, Ribaudo HJ, Douglas PS. Pitavastatin to Prevent Cardiovascular Disease in HIV Infection. N Engl J Med 2023; 389:687-699
  66. He Y, Li X, Gasevic D, Brunt E, McLachlan F, Millenson M, Timofeeva M, Ioannidis JPA, Campbell H, Theodoratou E. Statins and Multiple Noncardiovascular Outcomes: Umbrella Review of Meta-analyses of Observational Studies and Randomized Controlled Trials. Ann Intern Med 2018; 169:543-553
  67. Newman CB, Preiss D, Tobert JA, Jacobson TA, Page RL, 2nd, Goldstein LB, Chin C, Tannock LR, Miller M, Raghuveer G, Duell PB, Brinton EA, Pollak A, Braun LT, Welty FK. Statin Safety and Associated Adverse Events: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2019; 39:e38-e81
  68. Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, Seshasai SR, McMurray JJ, Freeman DJ, Jukema JW, Macfarlane PW, Packard CJ, Stott DJ, Westendorp RG, Shepherd J, Davis BR, Pressel SL, Marchioli R, Marfisi RM, Maggioni AP, Tavazzi L, Tognoni G, Kjekshus J, Pedersen TR, Cook TJ, Gotto AM, Clearfield MB, Downs JR, Nakamura H, Ohashi Y, Mizuno K, Ray KK, Ford I. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 2010; 375:735-742
  69. Preiss D, Seshasai SR, Welsh P, Murphy SA, Ho JE, Waters DD, DeMicco DA, Barter P, Cannon CP, Sabatine MS, Braunwald E, Kastelein JJ, de Lemos JA, Blazing MA, Pedersen TR, Tikkanen MJ, Sattar N, Ray KK. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA 2011; 305:2556-2564
  70. Feingold KR. Dyslipidemia in Diabetes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA) 2020.
  71. Erqou S, Lee CC, Adler AI. Statins and glycaemic control in individuals with diabetes: a systematic review and meta-analysis. Diabetologia 2014; 57:2444-2452
  72. Colhoun HM, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Livingstone SJ, Thomason MJ, Mackness MI, Charlton-Menys V, Fuller JH. Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the Collaborative Atorvastatin Diabetes Study (CARDS): multicentre randomised placebo-controlled trial. Lancet 2004; 364:685-696
  73. Collins R, Armitage J, Parish S, Sleigh P, Peto R. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial. Lancet 2003; 361:2005-2016
  74. Sattar N. Statins and diabetes: What are the connections? Best Pract Res Clin Endocrinol Metab 2023; 37:101749
  75. Swerdlow DI, Preiss D, Kuchenbaecker KB, Holmes MV, Engmann JE, Shah T, Sofat R, Stender S, Johnson PC, Scott RA, Leusink M, Verweij N, Sharp SJ, Guo Y, Giambartolomei C, Chung C, Peasey A, Amuzu A, Li K, Palmen J, Howard P, Cooper JA, Drenos F, Li YR, Lowe G, Gallacher J, Stewart MC, Tzoulaki I, Buxbaum SG, van der AD, Forouhi NG, Onland-Moret NC, van der Schouw YT, Schnabel RB, Hubacek JA, Kubinova R, Baceviciene M, Tamosiunas A, Pajak A, Topor-Madry R, Stepaniak U, Malyutina S, Baldassarre D, Sennblad B, Tremoli E, de Faire U, Veglia F, Ford I, Jukema JW, Westendorp RG, de Borst GJ, de Jong PA, Algra A, Spiering W, Maitland-van der Zee AH, Klungel OH, de Boer A, Doevendans PA, Eaton CB, Robinson JG, Duggan D, Consortium D, Consortium M, InterAct C, Kjekshus J, Downs JR, Gotto AM, Keech AC, Marchioli R, Tognoni G, Sever PS, Poulter NR, Waters DD, Pedersen TR, Amarenco P, Nakamura H, McMurray JJ, Lewsey JD, Chasman DI, Ridker PM, Maggioni AP, Tavazzi L, Ray KK, Seshasai SR, Manson JE, Price JF, Whincup PH, Morris RW, Lawlor DA, Smith GD, Ben-Shlomo Y, Schreiner PJ, Fornage M, Siscovick DS, Cushman M, Kumari M, Wareham NJ, Verschuren WM, Redline S, Patel SR, Whittaker JC, Hamsten A, Delaney JA, Dale C, Gaunt TR, Wong A, Kuh D, Hardy R, Kathiresan S, Castillo BA, van der Harst P, Brunner EJ, Tybjaerg-Hansen A, Marmot MG, Krauss RM, Tsai M, Coresh J, Hoogeveen RC, Psaty BM, Lange LA, Hakonarson H, Dudbridge F, Humphries SE, Talmud PJ, Kivimaki M, Timpson NJ, Langenberg C, Asselbergs FW, Voevoda M, Bobak M, Pikhart H, Wilson JG, Reiner AP, Keating BJ, Hingorani AD, Sattar N. HMG-coenzyme A reductase inhibition, type 2 diabetes, and bodyweight: evidence from genetic analysis and randomised trials. Lancet 2015; 385:351-361
  76. Sugiyama T, Tsugawa Y, Tseng CH, Kobayashi Y, Shapiro MF. Different time trends of caloric and fat intake between statin users and nonusers among US adults: gluttony in the time of statins? JAMA Intern Med 2014; 174:1038-1045
  77. Higuchi S, Izquierdo MC, Haeusler RA. Unexplained reciprocal regulation of diabetes and lipoproteins. Curr Opin Lipidol 2018; 29:186-193
  78. Rojas-Fernandez C, Hudani Z, Bittner V. Statins and cognitive side effects: what cardiologists need to know. Cardiol Clin 2015; 33:245-256
  79. Rojas-Fernandez CH, Goldstein LB, Levey AI, Taylor BA, Bittner V, The National Lipid Association's Safety Task Force. An assessment by the Statin Cognitive Safety Task Force: 2014 update. J Clin Lipidol 2014; 8:S5-16
  80. Richardson K, Schoen M, French B, Umscheid CA, Mitchell MD, Arnold SE, Heidenreich PA, Rader DJ, deGoma EM. Statins and cognitive function: a systematic review. Ann Intern Med 2013; 159:688-697
  81. Trompet S, van Vliet P, de Craen AJ, Jolles J, Buckley BM, Murphy MB, Ford I, Macfarlane PW, Sattar N, Packard CJ, Stott DJ, Shepherd J, Bollen EL, Blauw GJ, Jukema JW, Westendorp RG. Pravastatin and cognitive function in the elderly. Results of the PROSPER study. J Neurol 2010; 257:85-90
  82. Collins R, Armitage J, Parish S, Sleight P, Peto R. Effects of cholesterol-lowering with simvastatin on stroke and other major vascular events in 20536 people with cerebrovascular disease or other high-risk conditions. Lancet 2004; 363:757-767
  83. McGuinness B, Craig D, Bullock R, Malouf R, Passmore P. Statins for the treatment of dementia. Cochrane Database Syst Rev 2014; 7:CD007514
  84. Kashani A, Phillips CO, Foody JM, Wang Y, Mangalmurti S, Ko DT, Krumholz HM. Risks associated with statin therapy: a systematic overview of randomized clinical trials. Circulation 2006; 114:2788-2797
  85. de Denus S, Spinler SA, Miller K, Peterson AM. Statins and liver toxicity: a meta-analysis. Pharmacotherapy 2004; 24:584-591
  86. Law M, Rudnicka AR. Statin safety: a systematic review. Am J Cardiol 2006; 97:52C-60C
  87. Alsheikh-Ali AA, Maddukuri PV, Han H, Karas RH. Effect of the magnitude of lipid lowering on risk of elevated liver enzymes, rhabdomyolysis, and cancer: insights from large randomized statin trials. J Am Coll Cardiol 2007; 50:409-418
  88. Russo MW, Scobey M, Bonkovsky HL. Drug-induced liver injury associated with statins. Semin Liver Dis 2009; 29:412-422
  89. Tolman KG. Defining patient risks from expanded preventive therapies. Am J Cardiol 2000; 85:15E-19E
  90. Boutari C, Pappas PD, Anastasilakis D, Mantzoros CS. Statins' efficacy in non-alcoholic fatty liver disease: A systematic review and meta-analysis. Clin Nutr 2022; 41:2195-2206
  91. Rosenson RS, Baker SK, Jacobson TA, Kopecky SL, Parker BA, The National Lipid Association's Muscle Safety Expert Panel. An assessment by the Statin Muscle Safety Task Force: 2014 update. J Clin Lipidol 2014; 8:S58-71
  92. Stroes ES, Thompson PD, Corsini A, Vladutiu GD, Raal FJ, Ray KK, Roden M, Stein E, Tokgozoglu L, Nordestgaard BG, Bruckert E, De Backer G, Krauss RM, Laufs U, Santos RD, Hegele RA, Hovingh GK, Leiter LA, Mach F, Marz W, Newman CB, Wiklund O, Jacobson TA, Catapano AL, Chapman MJ, Ginsberg HN, European Atherosclerosis Society Consensus Panel. Statin-associated muscle symptoms: impact on statin therapy-European Atherosclerosis Society Consensus Panel Statement on Assessment, Aetiology and Management. Eur Heart J 2015; 36:1012-1022
  93. Thompson PD, Clarkson P, Karas RH. Statin-associated myopathy. JAMA 2003; 289:1681-1690
  94. Cholesterol Treatment Trialists Collaboration. Effect of statin therapy on muscle symptoms: an individual participant data meta-analysis of large-scale, randomised, double-blind trials. Lancet 2022; 400:832-845
  95. Bruckert E, Hayem G, Dejager S, Yau C, Begaud B. Mild to moderate muscular symptoms with high-dosage statin therapy in hyperlipidemic patients--the PRIMO study. Cardiovasc Drugs Ther 2005; 19:403-414
  96. Buettner C, Davis RB, Leveille SG, Mittleman MA, Mukamal KJ. Prevalence of musculoskeletal pain and statin use. J Gen Intern Med 2008; 23:1182-1186
  97. Cohen JD, Brinton EA, Ito MK, Jacobson TA. Understanding Statin Use in America and Gaps in Patient Education (USAGE): an internet-based survey of 10,138 current and former statin users. J Clin Lipidol 2012; 6:208-215
  98. Parker BA, Capizzi JA, Grimaldi AS, Clarkson PM, Cole SM, Keadle J, Chipkin S, Pescatello LS, Simpson K, White CM, Thompson PD. Effect of statins on skeletal muscle function. Circulation 2013; 127:96-103
  99. Gupta A, Thompson D, Whitehouse A, Collier T, Dahlof B, Poulter N, Collins R, Sever P. Adverse events associated with unblinded, but not with blinded, statin therapy in the Anglo-Scandinavian Cardiac Outcomes Trial-Lipid-Lowering Arm (ASCOT-LLA): a randomised double-blind placebo-controlled trial and its non-randomised non-blind extension phase. Lancet 2017; 389:2473-2481
  100. Joy TR, Monjed A, Zou GY, Hegele RA, McDonald CG, Mahon JL. N-of-1 (single-patient) trials for statin-related myalgia. Ann Intern Med 2014; 160:301-310
  101. Taylor BA, Lorson L, White CM, Thompson PD. A randomized trial of coenzyme Q10 in patients with confirmed statin myopathy. Atherosclerosis 2015; 238:329-335
  102. Nissen SE, Stroes E, Dent-Acosta RE, Rosenson RS, Lehman SJ, Sattar N, Preiss D, Bruckert E, Ceska R, Lepor N, Ballantyne CM, Gouni-Berthold I, Elliott M, Brennan DM, Wasserman SM, Somaratne R, Scott R, Stein EA. Efficacy and Tolerability of Evolocumab vs Ezetimibe in Patients With Muscle-Related Statin Intolerance: The GAUSS-3 Randomized Clinical Trial. JAMA 2016; 315:1580-1590
  103. Herrett E, Williamson E, Brack K, Beaumont D, Perkins A, Thayne A, Shakur-Still H, Roberts I, Prowse D, Goldacre B, van Staa T, MacDonald TM, Armitage J, Wimborne J, Melrose P, Singh J, Brooks L, Moore M, Hoffman M, Smeeth L. Statin treatment and muscle symptoms: series of randomised, placebo controlled n-of-1 trials. BMJ 2021; 372:n135
  104. Howard JP, Wood FA, Finegold JA, Nowbar AN, Thompson DM, Arnold AD, Rajkumar CA, Connolly S, Cegla J, Stride C, Sever P, Norton C, Thom SAM, Shun-Shin MJ, Francis DP. Side Effect Patterns in a Crossover Trial of Statin, Placebo, and No Treatment. J Am Coll Cardiol 2021; 78:1210-1222
  105. Nielsen SF, Nordestgaard BG. Negative statin-related news stories decrease statin persistence and increase myocardial infarction and cardiovascular mortality: a nationwide prospective cohort study. Eur Heart J 2016; 37:908-916
  106. De Vera MA, Bhole V, Burns LC, Lacaille D. Impact of statin adherence on cardiovascular disease and mortality outcomes: a systematic review. Br J Clin Pharmacol 2014; 78:684-698
  107. Cziraky MJ, Willey VJ, McKenney JM, Kamat SA, Fisher MD, Guyton JR, Jacobson TA, Davidson MH. Risk of hospitalized rhabdomyolysis associated with lipid-lowering drugs in a real-world clinical setting. J Clin Lipidol 2013; 7:102-108
  108. Davidson MH, Stein EA, Dujovne CA, Hunninghake DB, Weiss SR, Knopp RH, Illingworth DR, Mitchel YB, Melino MR, Zupkis RV, Dobrinska MR, Amin RD, Tobert JA. The efficacy and six-week tolerability of simvastatin 80 and 160 mg/day. Am J Cardiol 1997; 79:38-42
  109. Rosenson RS, Bays HE. Results of two clinical trials on the safety and efficacy of pravastatin 80 and 160 mg per day. Am J Cardiol 2003; 91:878-881
  110. Search Collaborative Group, Link E, Parish S, Armitage J, Bowman L, Heath S, Matsuda F, Gut I, Lathrop M, Collins R. SLCO1B1 variants and statin-induced myopathy--a genomewide study. N Engl J Med 2008; 359:789-799
  111. Hopewell JC, Reith C, Armitage J. Pharmacogenomics of statin therapy: any new insights in efficacy or safety? Curr Opin Lipidol 2014; 25:438-445
  112. Mammen AL. Statin-Associated Autoimmune Myopathy. N Engl J Med 2016; 374:664-669
  113. Mohassel P, Mammen AL. Anti-HMGCR Myopathy. J Neuromuscul Dis 2018; 5:11-20
  114. Wild R, Feingold KR. Effect of Pregnancy on Lipid Metabolism and Lipoprotein Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  115. Bruckert E, Giral P, Tellier P. Perspectives in cholesterol-lowering therapy: the role of ezetimibe, a new selective inhibitor of intestinal cholesterol absorption. Circulation 2003; 107:3124-3128
  116. Liebeskind A, Peterson AL, Wilson D. Sitosterolemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  117. Pandor A, Ara RM, Tumur I, Wilkinson AJ, Paisley S, Duenas A, Durrington PN, Chilcott J. Ezetimibe monotherapy for cholesterol lowering in 2,722 people: systematic review and meta-analysis of randomized controlled trials. J Intern Med 2009; 265:568-580
  118. Morrone D, Weintraub WS, Toth PP, Hanson ME, Lowe RS, Lin J, Shah AK, Tershakovec AM. Lipid-altering efficacy of ezetimibe plus statin and statin monotherapy and identification of factors associated with treatment response: a pooled analysis of over 21,000 subjects from 27 clinical trials. Atherosclerosis 2012; 223:251-261
  119. Ballantyne CM, Weiss R, Moccetti T, Vogt A, Eber B, Sosef F, Duffield E. Efficacy and safety of rosuvastatin 40 mg alone or in combination with ezetimibe in patients at high risk of cardiovascular disease (results from the EXPLORER study). Am J Cardiol 2007; 99:673-680
  120. Sahebkar A, Simental-Mendia LE, Pirro M, Banach M, Watts GF, Sirtori C, Al-Rasadi K, Atkin SL. Impact of ezetimibe on plasma lipoprotein(a) concentrations as monotherapy or in combination with statins: a systematic review and meta-analysis of randomized controlled trials. Sci Rep 2018; 8:17887
  121. Kastelein JJ, Akdim F, Stroes ES, Zwinderman AH, Bots ML, Stalenhoef AF, Visseren FL, Sijbrands EJ, Trip MD, Stein EA, Gaudet D, Duivenvoorden R, Veltri EP, Marais AD, de Groot E. Simvastatin with or without ezetimibe in familial hypercholesterolemia. N Engl J Med 2008; 358:1431-1443
  122. Sager PT, Capece R, Lipka L, Strony J, Yang B, Suresh R, Mitchel Y, Veltri E. Effects of ezetimibe coadministered with simvastatin on C-reactive protein in a large cohort of hypercholesterolemic patients. Atherosclerosis 2005; 179:361-367
  123. Ballantyne CM, Houri J, Notarbartolo A, Melani L, Lipka LJ, Suresh R, Sun S, LeBeaut AP, Sager PT, Veltri EP. Effect of ezetimibe coadministered with atorvastatin in 628 patients with primary hypercholesterolemia: a prospective, randomized, double-blind trial. Circulation 2003; 107:2409-2415
  124. Yu L. The structure and function of Niemann-Pick C1-like 1 protein. Curr Opin Lipidol 2008; 19:263-269
  125. Turley SD, Dietschy JM. Sterol absorption by the small intestine. Curr Opin Lipidol 2003; 14:233-240
  126. Telford DE, Sutherland BG, Edwards JY, Andrews JD, Barrett PH, Huff MW. The molecular mechanisms underlying the reduction of LDL apoB-100 by ezetimibe plus simvastatin. J Lipid Res 2007; 48:699-708
  127. Pramfalk C, Jiang ZY, Parini P. Hepatic Niemann-Pick C1-like 1. Curr Opin Lipidol 2011; 22:225-230
  128. Rossebo AB, Pedersen TR, Boman K, Brudi P, Chambers JB, Egstrup K, Gerdts E, Gohlke-Barwolf C, Holme I, Kesaniemi YA, Malbecq W, Nienaber CA, Ray S, Skjaerpe T, Wachtell K, Willenheimer R. Intensive lipid lowering with simvastatin and ezetimibe in aortic stenosis. N Engl J Med 2008; 359:1343-1356
  129. Baigent C, Landray MJ, Reith C, Emberson J, Wheeler DC, Tomson C, Wanner C, Krane V, Cass A, Craig J, Neal B, Jiang L, Hooi LS, Levin A, Agodoa L, Gaziano M, Kasiske B, Walker R, Massy ZA, Feldt-Rasmussen B, Krairittichai U, Ophascharoensuk V, Fellstrom B, Holdaas H, Tesar V, Wiecek A, Grobbee D, de Zeeuw D, Gronhagen-Riska C, Dasgupta T, Lewis D, Herrington W, Mafham M, Majoni W, Wallendszus K, Grimm R, Pedersen T, Tobert J, Armitage J, Baxter A, Bray C, Chen Y, Chen Z, Hill M, Knott C, Parish S, Simpson D, Sleight P, Young A, Collins R. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet 2011; 377:2181-2192
  130. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, Darius H, Lewis BS, Ophuis TO, Jukema JW, De Ferrari GM, Ruzyllo W, De Lucca P, Im K, Bohula EA, Reist C, Wiviott SD, Tershakovec AM, Musliner TA, Braunwald E, Califf RM. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387-2397
  131. Bohula EA, Wiviott SD, Giugliano RP, Blazing MA, Park JG, Murphy SA, White JA, Mach F, Van de Werf F, Dalby AJ, White HD, Tershakovec AM, Cannon CP, Braunwald E. Prevention of Stroke with the Addition of Ezetimibe to Statin Therapy in Patients With Acute Coronary Syndrome in IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial). Circulation 2017; 136:2440-2450
  132. Giugliano RP, Cannon CP, Blazing MA, Nicolau JC, Corbalan R, Spinar J, Park JG, White JA, Bohula EA, Braunwald E. Benefit of Adding Ezetimibe to Statin Therapy on Cardiovascular Outcomes and Safety in Patients With Versus Without Diabetes Mellitus: Results From IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial). Circulation 2018; 137:1571-1582
  133. Bonaca MP, Gutierrez JA, Cannon C, Giugliano R, Blazing M, Park JG, White J, Tershakovec A, Braunwald E. Polyvascular disease, type 2 diabetes, and long-term vascular risk: a secondary analysis of the IMPROVE-IT trial. Lancet Diabetes Endocrinol 2018; 6:934-943
  134. Ouchi Y, Sasaki J, Arai H, Yokote K, Harada K, Katayama Y, Urabe T, Uchida Y, Hayashi M, Yokota N, Nishida H, Otonari T, Arai T, Sakuma I, Sakabe K, Yamamoto M, Kobayashi T, Oikawa S, Yamashita S, Rakugi H, Imai T, Tanaka S, Ohashi Y, Kuwabara M, Ito H. Ezetimibe Lipid-Lowering Trial on Prevention of Atherosclerotic Cardiovascular Disease in 75 or Older (EWTOPIA 75): A Randomized, Controlled Trial. Circulation 2019; 140:992-1003
  135. Kim BK, Hong SJ, Lee YJ, Hong SJ, Yun KH, Hong BK, Heo JH, Rha SW, Cho YH, Lee SJ, Ahn CM, Kim JS, Ko YG, Choi D, Jang Y, Hong MK. Long-term efficacy and safety of moderate-intensity statin with ezetimibe combination therapy versus high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease (RACING): a randomised, open-label, non-inferiority trial. Lancet 2022; 400:380-390
  136. Lee B, Hong SJ, Rha SW, Heo JH, Hur SH, Choi HH, Kim KJ, Kim JH, Kim HK, Kim U, Choi YJ, Lee YJ, Lee SJ, Ahn CM, Ko YG, Kim BK, Choi D, Hong MK, Jang Y, Kim JS. Moderate-intensity statin plus ezetimibe vs high-intensity statin according to baseline LDL-C in the treatment of atherosclerotic cardiovascular disease: A post-hoc analysis of the RACING randomized trial. Atherosclerosis 2023; 386:117373
  137. Lee SH, Lee YJ, Heo JH, Hur SH, Choi HH, Kim KJ, Kim JH, Park KH, Lee JH, Choi YJ, Lee SJ, Hong SJ, Ahn CM, Kim BK, Ko YG, Choi D, Hong MK, Jang Y, Kim JS. Combination Moderate-Intensity Statin and Ezetimibe Therapy for Elderly Patients With Atherosclerosis. J Am Coll Cardiol 2023; 81:1339-1349
  138. Toth PP, Morrone D, Weintraub WS, Hanson ME, Lowe RS, Lin J, Shah AK, Tershakovec AM. Safety profile of statins alone or combined with ezetimibe: a pooled analysis of 27 studies including over 22,000 patients treated for 6-24 weeks. Int J Clin Pract 2012; 66:800-812
  139. Luo L, Yuan X, Huang W, Ren F, Zhu H, Zheng Y, Tang L. Safety of coadministration of ezetimibe and statins in patients with hypercholesterolaemia: a meta-analysis. Intern Med J 2015; 45:546-557
  140. Kashani A, Sallam T, Bheemreddy S, Mann DL, Wang Y, Foody JM. Review of side-effect profile of combination ezetimibe and statin therapy in randomized clinical trials. Am J Cardiol 2008; 101:1606-1613
  141. Savarese G, De Ferrari GM, Rosano GM, Perrone-Filardi P. Safety and efficacy of ezetimibe: A meta-analysis. Int J Cardiol 2015; 201:247-252
  142. Peto R, Emberson J, Landray M, Baigent C, Collins R, Clare R, Califf R. Analyses of cancer data from three ezetimibe trials. N Engl J Med 2008; 359:1357-1366
  143. Wu H, Shang H, Wu J. Effect of ezetimibe on glycemic control: a systematic review and meta-analysis of randomized controlled trials. Endocrine 2018; 60:229-239
  144. Aldridge MA, Ito MK. Colesevelam hydrochloride: a novel bile acid-binding resin. Ann Pharmacother 2001; 35:898-907
  145. Heel RC, Brogden RN, Pakes GE, Speight TM, Avery GS. Colestipol: a review of its pharmacological properties and therapeutic efficacy in patients with hypercholesterolaemia. Drugs 1980; 19:161-180
  146. Insull W, Jr. Clinical utility of bile acid sequestrants in the treatment of dyslipidemia: a scientific review. South Med J 2006; 99:257-273
  147. Huijgen R, Abbink EJ, Bruckert E, Stalenhoef AF, Imholz BP, Durrington PN, Trip MD, Eriksson M, Visseren FL, Schaefer JR, Kastelein JJ. Colesevelam added to combination therapy with a statin and ezetimibe in patients with familial hypercholesterolemia: a 12-week, multicenter, randomized, double-blind, controlled trial. Clin Ther 2010; 32:615-625
  148. Zema MJ. Colesevelam HCl and ezetimibe combination therapy provides effective lipid-lowering in difficult-to-treat patients with hypercholesterolemia. Am J Ther 2005; 12:306-310
  149. Bays H, Rhyne J, Abby S, Lai YL, Jones M. Lipid-lowering effects of colesevelam HCl in combination with ezetimibe. Curr Med Res Opin 2006; 22:2191-2200
  150. Fonseca VA, Handelsman Y, Staels B. Colesevelam lowers glucose and lipid levels in type 2 diabetes: the clinical evidence. Diabetes Obes Metab 2010; 12:384-392
  151. Devaraj S, Autret B, Jialal I. Effects of colesevelam hydrochloride (WelChol) on biomarkers of inflammation in patients with mild hypercholesterolemia. Am J Cardiol 2006; 98:641-643
  152. Bays HE, Davidson M, Jones MR, Abby SL. Effects of colesevelam hydrochloride on low-density lipoprotein cholesterol and high-sensitivity C-reactive protein when added to statins in patients with hypercholesterolemia. Am J Cardiol 2006; 97:1198-1205
  153. Einarsson K, Ericsson S, Ewerth S, Reihner E, Rudling M, Stahlberg D, Angelin B. Bile acid sequestrants: mechanisms of action on bile acid and cholesterol metabolism. Eur J Clin Pharmacol 1991; 40 Suppl 1:S53-58
  154. Kliewer SA, Mangelsdorf DJ. Bile Acids as Hormones: The FXR-FGF15/19 Pathway. Dig Dis 2015; 33:327-331
  155. Chiang JY. Bile acids: regulation of synthesis. J Lipid Res 2009; 50:1955-1966
  156. Porez G, Prawitt J, Gross B, Staels B. Bile acid receptors as targets for the treatment of dyslipidemia and cardiovascular disease. J Lipid Res 2012; 53:1723-1737
  157. Edwards PA, Kast HR, Anisfeld AM. BAREing it all: the adoption of LXR and FXR and their roles in lipid homeostasis. J Lipid Res 2002; 43:2-12
  158. Bronden A, Knop FK. Gluco-Metabolic Effects of Pharmacotherapy-Induced Modulation of Bile Acid Physiology. J Clin Endocrinol Metab 2020; 105
  159. The Lipid Research Clinics Coronary Primary Prevention Trial results. I. Reduction in incidence of coronary heart disease. JAMA 1984; 251:351-364
  160. The Lipid Research Clinics Coronary Primary Prevention Trial results. II. The relationship of reduction in incidence of coronary heart disease to cholesterol lowering. JAMA 1984; 251:365-374
  161. Levy RI, Brensike JF, Epstein SE, Kelsey SF, Passamani ER, Richardson JM, Loh IK, Stone NJ, Aldrich RF, Battaglini JW, et al. The influence of changes in lipid values induced by cholestyramine and diet on progression of coronary artery disease: results of NHLBI Type II Coronary Intervention Study. Circulation 1984; 69:325-337
  162. Watts GF, Lewis B, Brunt JN, Lewis ES, Coltart DJ, Smith LD, Mann JI, Swan AV. Effects on coronary artery disease of lipid-lowering diet, or diet plus cholestyramine, in the St Thomas' Atherosclerosis Regression Study (STARS). Lancet 1992; 339:563-569
  163. Blankenhorn DH, Nessim SA, Johnson RL, Sanmarco ME, Azen SP, Cashin-Hemphill L. Beneficial effects of combined colestipol-niacin therapy on coronary atherosclerosis and coronary venous bypass grafts. JAMA 1987; 257:3233-3240
  164. Brown G, Albers JJ, Fisher LD, Schaefer SM, Lin JT, Kaplan C, Zhao XQ, Bisson BD, Fitzpatrick VF, Dodge HT. Regression of coronary artery disease as a result of intensive lipid-lowering therapy in men with high levels of apolipoprotein B. N Engl J Med 1990; 323:1289-1298
  165. Kane JP, Malloy MJ, Ports TA, Phillips NR, Diehl JC, Havel RJ. Regression of coronary atherosclerosis during treatment of familial hypercholesterolemia with combined drug regimens. JAMA 1990; 264:3007-3012
  166. Ito MK, McGowan MP, Moriarty PM, National Lipid Association Expert Panel on Familial Hypercholesterolemia. Management of familial hypercholesterolemias in adult patients: recommendations from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol 2011; 5:S38-45
  167. Giugliano RP, Sabatine MS. Are PCSK9 Inhibitors the Next Breakthrough in the Cardiovascular Field? J Am Coll Cardiol 2015; 65:2638-2651
  168. McKenney JM. Understanding PCSK9 and anti-PCSK9 therapies. J Clin Lipidol 2015; 9:170-186
  169. Navarese EP, Kolodziejczak M, Schulze V, Gurbel PA, Tantry U, Lin Y, Brockmeyer M, Kandzari DE, Kubica JM, D'Agostino RB, Sr., Kubica J, Volpe M, Agewall S, Kereiakes DJ, Kelm M. Effects of Proprotein Convertase Subtilisin/Kexin Type 9 Antibodies in Adults With Hypercholesterolemia: A Systematic Review and Meta-analysis. Ann Intern Med 2015; 163:40-51
  170. O'Donoghue ML, Fazio S, Giugliano RP, Stroes ESG, Kanevsky E, Gouni-Berthold I, Im K, Lira Pineda A, Wasserman SM, Ceska R, Ezhov MV, Jukema JW, Jensen HK, Tokgozoglu SL, Mach F, Huber K, Sever PS, Keech AC, Pedersen TR, Sabatine MS. Lipoprotein(a), PCSK9 Inhibition, and Cardiovascular Risk. Circulation 2019; 139:1483-1492
  171. Sahebkar A, Di Giosia P, Stamerra CA, Grassi D, Pedone C, Ferretti G, Bacchetti T, Ferri C, Giorgini P. Effect of monoclonal antibodies to PCSK9 on high-sensitivity C-reactive protein levels: a meta-analysis of 16 randomized controlled treatment arms. Br J Clin Pharmacol 2016; 81:1175-1190
  172. Koren MJ, Lundqvist P, Bolognese M, Neutel JM, Monsalvo ML, Yang J, Kim JB, Scott R, Wasserman SM, Bays H. Anti-PCSK9 monotherapy for hypercholesterolemia: the MENDEL-2 randomized, controlled phase III clinical trial of evolocumab. J Am Coll Cardiol 2014; 63:2531-2540
  173. Roth EM, Taskinen MR, Ginsberg HN, Kastelein JJ, Colhoun HM, Robinson JG, Merlet L, Pordy R, Baccara-Dinet MT. Monotherapy with the PCSK9 inhibitor alirocumab versus ezetimibe in patients with hypercholesterolemia: results of a 24 week, double-blind, randomized Phase 3 trial. Int J Cardiol 2014; 176:55-61
  174. Kereiakes DJ, Robinson JG, Cannon CP, Lorenzato C, Pordy R, Chaudhari U, Colhoun HM. Efficacy and safety of the proprotein convertase subtilisin/kexin type 9 inhibitor alirocumab among high cardiovascular risk patients on maximally tolerated statin therapy: The ODYSSEY COMBO I study. Am Heart J 2015; 169:906-915 e913
  175. Cannon CP, Cariou B, Blom D, McKenney JM, Lorenzato C, Pordy R, Chaudhari U, Colhoun HM. Efficacy and safety of alirocumab in high cardiovascular risk patients with inadequately controlled hypercholesterolaemia on maximally tolerated doses of statins: the ODYSSEY COMBO II randomized controlled trial. Eur Heart J 2015; 36:1186-1194
  176. Robinson JG, Nedergaard BS, Rogers WJ, Fialkow J, Neutel JM, Ramstad D, Somaratne R, Legg JC, Nelson P, Scott R, Wasserman SM, Weiss R. Effect of evolocumab or ezetimibe added to moderate- or high-intensity statin therapy on LDL-C lowering in patients with hypercholesterolemia: the LAPLACE-2 randomized clinical trial. JAMA 2014; 311:1870-1882
  177. Blom DJ, Hala T, Bolognese M, Lillestol MJ, Toth PD, Burgess L, Ceska R, Roth E, Koren MJ, Ballantyne CM, Monsalvo ML, Tsirtsonis K, Kim JB, Scott R, Wasserman SM, Stein EA. A 52-week placebo-controlled trial of evolocumab in hyperlipidemia. N Engl J Med 2014; 370:1809-1819
  178. Raal FJ, Stein EA, Dufour R, Turner T, Civeira F, Burgess L, Langslet G, Scott R, Olsson AG, Sullivan D, Hovingh GK, Cariou B, Gouni-Berthold I, Somaratne R, Bridges I, Scott R, Wasserman SM, Gaudet D. PCSK9 inhibition with evolocumab (AMG 145) in heterozygous familial hypercholesterolaemia (RUTHERFORD-2): a randomised, double-blind, placebo-controlled trial. Lancet 2015; 385:331-340
  179. Kastelein JJ, Ginsberg HN, Langslet G, Hovingh GK, Ceska R, Dufour R, Blom D, Civeira F, Krempf M, Lorenzato C, Zhao J, Pordy R, Baccara-Dinet MT, Gipe DA, Geiger MJ, Farnier M. ODYSSEY FH I and FH II: 78 week results with alirocumab treatment in 735 patients with heterozygous familial hypercholesterolaemia. Eur Heart J 2015; 36:2996-3003
  180. Raal FJ, Honarpour N, Blom DJ, Hovingh GK, Xu F, Scott R, Wasserman SM, Stein EA. Inhibition of PCSK9 with evolocumab in homozygous familial hypercholesterolaemia (TESLA Part B): a randomised, double-blind, placebo-controlled trial. Lancet 2015; 385:341-350
  181. Raal FJ, Hovingh GK, Blom D, Santos RD, Harada-Shiba M, Bruckert E, Couture P, Soran H, Watts GF, Kurtz C, Honarpour N, Tang L, Kasichayanula S, Wasserman SM, Stein EA. Long-term treatment with evolocumab added to conventional drug therapy, with or without apheresis, in patients with homozygous familial hypercholesterolaemia: an interim subset analysis of the open-label TAUSSIG study. Lancet Diabetes Endocrinol 2017; 5:280-290
  182. Stein EA, Honarpour N, Wasserman SM, Xu F, Scott R, Raal FJ. Effect of the proprotein convertase subtilisin/kexin 9 monoclonal antibody, AMG 145, in homozygous familial hypercholesterolemia. Circulation 2013; 128:2113-2120
  183. Blom DJ, Harada-Shiba M, Rubba P, Gaudet D, Kastelein JJP, Charng MJ, Pordy R, Donahue S, Ali S, Dong Y, Khilla N, Banerjee P, Baccara-Dinet M, Rosenson RS. Efficacy and Safety of Alirocumab in Adults With Homozygous Familial Hypercholesterolemia: The ODYSSEY HoFH Trial. J Am Coll Cardiol 2020; 76:131-142
  184. Stroes E, Colquhoun D, Sullivan D, Civeira F, Rosenson RS, Watts GF, Bruckert E, Cho L, Dent R, Knusel B, Xue A, Scott R, Wasserman SM, Rocco M. Anti-PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: the GAUSS-2 randomized, placebo-controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol 2014; 63:2541-2548
  185. Moriarty PM, Thompson PD, Cannon CP, Guyton JR, Bergeron J, Zieve FJ, Bruckert E, Jacobson TA, Kopecky SL, Baccara-Dinet MT, Du Y, Pordy R, Gipe DA. Efficacy and safety of alirocumab vs ezetimibe in statin-intolerant patients, with a statin rechallenge arm: The ODYSSEY ALTERNATIVE randomized trial. J Clin Lipidol 2015; 9:758-769
  186. Sattar N, Preiss D, Robinson JG, Djedjos CS, Elliott M, Somaratne R, Wasserman SM, Raal FJ. Lipid-lowering efficacy of the PCSK9 inhibitor evolocumab (AMG 145) in patients with type 2 diabetes: a meta-analysis of individual patient data. Lancet Diabetes Endocrinol 2016; 4:403-410
  187. Abifadel M, Varret M, Rabes JP, Allard D, Ouguerram K, Devillers M, Cruaud C, Benjannet S, Wickham L, Erlich D, Derre A, Villeger L, Farnier M, Beucler I, Bruckert E, Chambaz J, Chanu B, Lecerf JM, Luc G, Moulin P, Weissenbach J, Prat A, Krempf M, Junien C, Seidah NG, Boileau C. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet 2003; 34:154-156
  188. Warden BA, Fazio S, Shapiro MD. Familial Hypercholesterolemia: Genes and Beyond. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  189. Cohen J, Pertsemlidis A, Kotowski IK, Graham R, Garcia CK, Hobbs HH. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat Genet 2005; 37:161-165
  190. Cohen JC, Boerwinkle E, Mosley TH, Jr., Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med 2006; 354:1264-1272
  191. Feingold KR. Introduction to Lipids and Lipoproteins. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  192. Horton JD, Cohen JC, Hobbs HH. PCSK9: a convertase that coordinates LDL catabolism. J Lipid Res 2009; 50 Suppl:S172-177
  193. Lambert G, Sjouke B, Choque B, Kastelein JJ, Hovingh GK. The PCSK9 decade. J Lipid Res 2012; 53:2515-2524
  194. Reyes-Soffer G, Pavlyha M, Ngai C, Thomas T, Holleran S, Ramakrishnan R, Karmally W, Nandakumar R, Fontanez N, Obunike J, Marcovina SM, Lichtenstein AH, Matthan NR, Matta J, Maroccia M, Becue F, Poitiers F, Swanson B, Cowan L, Sasiela WJ, Surks HK, Ginsberg HN. Effects of PCSK9 Inhibition With Alirocumab on Lipoprotein Metabolism in Healthy Humans. Circulation 2017; 135:352-362
  195. Watts GF, Chan DC, Dent R, Somaratne R, Wasserman SM, Scott R, Burrows S, Barrett PHR. Factorial Effects of Evolocumab and Atorvastatin on Lipoprotein Metabolism. Circulation 2017; 135:338-351
  196. Watts GF, Chan DC, Pang J, Ma L, Ying Q, Aggarwal S, Marcovina SM, Barrett PHR. PCSK9 Inhibition with alirocumab increases the catabolism of lipoprotein(a) particles in statin-treated patients with elevated lipoprotein(a). Metabolism 2020; 107:154221
  197. Ying Q, Chan DC, Pang J, Marcovina SM, Barrett PHR, Watts GF. PCSK9 inhibition with alirocumab decreases plasma lipoprotein(a) concentration by a dual mechanism of action in statin-treated patients with very high apolipoprotein(a) concentration. J Intern Med 2022; 291:870-876
  198. Raal FJ, Giugliano RP, Sabatine MS, Koren MJ, Blom D, Seidah NG, Honarpour N, Lira A, Xue A, Chirovolu P, Jackson S, Di M, Peach M, Somaratne R, Wasserman SM, Scott R, Stein EA. PCSK9 inhibition-mediated reduction in Lp(a) with evolocumab: an analysis of 10 clinical trials and the role of the LDL receptor. J Lipid Res 2016;
  199. Sabatine MS, Giugliano RP, Wiviott SD, Raal FJ, Blom DJ, Robinson J, Ballantyne CM, Somaratne R, Legg J, Wasserman SM, Scott R, Koren MJ, Stein EA, Open-Label Study of Long-Term Evaluation against LDLCI. Efficacy and safety of evolocumab in reducing lipids and cardiovascular events. N Engl J Med 2015; 372:1500-1509
  200. Sabatine MS, Leiter LA, Wiviott SD, Giugliano RP, Deedwania P, De Ferrari GM, Murphy SA, Kuder JF, Gouni-Berthold I, Lewis BS, Handelsman Y, Pineda AL, Honarpour N, Keech AC, Sever PS, Pedersen TR. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without diabetes and the effect of evolocumab on glycaemia and risk of new-onset diabetes: a prespecified analysis of the FOURIER randomised controlled trial. Lancet Diabetes Endocrinol 2017; 5:941-950
  201. Bonaca MP, Nault P, Giugliano RP, Keech AC, Pineda AL, Kanevsky E, Kuder J, Murphy SA, Jukema JW, Lewis BS, Tokgozoglu L, Somaratne R, Sever PS, Pedersen TR, Sabatine MS. Low-Density Lipoprotein Cholesterol Lowering With Evolocumab and Outcomes in Patients With Peripheral Artery Disease: Insights From the FOURIER Trial (Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects With Elevated Risk). Circulation 2018; 137:338-350
  202. Giugliano RP, Keech A, Murphy SA, Huber K, Tokgozoglu SL, Lewis BS, Ferreira J, Pineda AL, Somaratne R, Sever PS, Pedersen TR, Sabatine MS. Clinical Efficacy and Safety of Evolocumab in High-Risk Patients Receiving a Statin: Secondary Analysis of Patients With Low LDL Cholesterol Levels and in Those Already Receiving a Maximal-Potency Statin in a Randomized Clinical Trial. JAMA Cardiol 2017; 2:1385-1391
  203. Giugliano RP, Pedersen TR, Park JG, De Ferrari GM, Gaciong ZA, Ceska R, Toth K, Gouni-Berthold I, Lopez-Miranda J, Schiele F, Mach F, Ott BR, Kanevsky E, Pineda AL, Somaratne R, Wasserman SM, Keech AC, Sever PS, Sabatine MS. Clinical efficacy and safety of achieving very low LDL-cholesterol concentrations with the PCSK9 inhibitor evolocumab: a prespecified secondary analysis of the FOURIER trial. Lancet 2017; 390:1962-1971
  204. Sabatine MS, De Ferrari GM, Giugliano RP, Huber K, Lewis BS, Ferreira J, Kuder JF, Murphy SA, Wiviott SD, Kurtz CE, Honarpour N, Keech AC, Sever PS, Pedersen TR. Clinical Benefit of Evolocumab by Severity and Extent of Coronary Artery Disease. Circulation 2018; 138:756-766
  205. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Lecorps G, Mahaffey KW, Moryusef A, Pordy R, Quintero K, Roe MT, Sasiela WJ, Tamby JF, Tricoci P, White HD, Zeiher AM. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N Engl J Med 2018; 379:2097-2107
  206. Nicholls SJ, Puri R, Anderson T, Ballantyne CM, Cho L, Kastelein JJ, Koenig W, Somaratne R, Kassahun H, Yang J, Wasserman SM, Scott R, Ungi I, Podolec J, Ophuis AO, Cornel JH, Borgman M, Brennan DM, Nissen SE. Effect of Evolocumab on Progression of Coronary Disease in Statin-Treated Patients: The GLAGOV Randomized Clinical Trial. JAMA 2016; 316:2373-2384
  207. Raber L, Ueki Y, Otsuka T, Losdat S, Haner JD, Lonborg J, Fahrni G, Iglesias JF, van Geuns RJ, Ondracek AS, Radu Juul Jensen MD, Zanchin C, Stortecky S, Spirk D, Siontis GCM, Saleh L, Matter CM, Daemen J, Mach F, Heg D, Windecker S, Engstrom T, Lang IM, Koskinas KC. Effect of Alirocumab Added to High-Intensity Statin Therapy on Coronary Atherosclerosis in Patients With Acute Myocardial Infarction: The PACMAN-AMI Randomized Clinical Trial. JAMA 2022; 327:1771-1781
  208. Perez de Isla L, Diaz-Diaz JL, Romero MJ, Muniz-Grijalvo O, Mediavilla JD, Argueso R, Sanchez Munoz-Torrero JF, Rubio P, Alvarez-Banos P, Ponte P, Manas D, Suarez Gutierrez L, Cepeda JM, Casanas M, Fuentes F, Guijarro C, Angel Barba M, Saltijeral Cerezo A, Padro T, Mata P. Alirocumab and Coronary Atherosclerosis in Asymptomatic Patients with Familial Hypercholesterolemia: The ARCHITECT Study. Circulation 2023; 147:1436-1443
  209. Marston NA, Gurmu Y, Melloni GEM, Bonaca M, Gencer B, Sever PS, Pedersen TR, Keech AC, Roselli C, Lubitz SA, Ellinor PT, O'Donoghue ML, Giugliano RP, Ruff CT, Sabatine MS. The Effect of PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) Inhibition on the Risk of Venous Thromboembolism. Circulation 2020; 141:1600-1607
  210. Schwartz GG, Steg PG, Szarek M, Bittner VA, Diaz R, Goodman SG, Kim YU, Jukema JW, Pordy R, Roe MT, White HD, Bhatt DL. Peripheral Artery Disease and Venous Thromboembolic Events After Acute Coronary Syndrome: Role of Lipoprotein(a) and Modification by Alirocumab: Prespecified Analysis of the ODYSSEY OUTCOMES Randomized Clinical Trial. Circulation 2020; 141:1608-1617
  211. de Carvalho LSF, Campos AM, Sposito AC. Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Inhibitors and Incident Type 2 Diabetes: A Systematic Review and Meta-analysis With Over 96,000 Patient-Years. Diabetes Care 2018; 41:364-367
  212. Giugliano RP, Mach F, Zavitz K, Kurtz C, Im K, Kanevsky E, Schneider J, Wang H, Keech A, Pedersen TR, Sabatine MS, Sever PS, Robinson JG, Honarpour N, Wasserman SM, Ott BR. Cognitive Function in a Randomized Trial of Evolocumab. N Engl J Med 2017; 377:633-643
  213. Koren MJ, Giugliano RP, Raal FJ, Sullivan D, Bolognese M, Langslet G, Civeira F, Somaratne R, Nelson P, Liu T, Scott R, Wasserman SM, Sabatine MS. Efficacy and safety of longer-term administration of evolocumab (AMG 145) in patients with hypercholesterolemia: 52-week results from the Open-Label Study of Long-Term Evaluation Against LDL-C (OSLER) randomized trial. Circulation 2014; 129:234-243
  214. Robinson JG, Farnier M, Krempf M, Bergeron J, Luc G, Averna M, Stroes ES, Langslet G, Raal FJ, El Shahawy M, Koren MJ, Lepor NE, Lorenzato C, Pordy R, Chaudhari U, Kastelein JJ. Efficacy and safety of alirocumab in reducing lipids and cardiovascular events. N Engl J Med 2015; 372:1489-1499
  215. Shapiro MD, Feingold KR. Monogenic Disorders Causing Hypobetalipoproteinemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  216. LaRosa JC, Grundy SM, Kastelein JJ, Kostis JB, Greten H, Treating to New Targets Steering C, Investigators. Safety and efficacy of Atorvastatin-induced very low-density lipoprotein cholesterol levels in Patients with coronary heart disease (a post hoc analysis of the treating to new targets [TNT] study). Am J Cardiol 2007; 100:747-752
  217. Wiviott SD, Cannon CP, Morrow DA, Ray KK, Pfeffer MA, Braunwald E. Can low-density lipoprotein be too low? The safety and efficacy of achieving very low low-density lipoprotein with intensive statin therapy: a PROVE IT-TIMI 22 substudy. J Am Coll Cardiol 2005; 46:1411-1416
  218. Everett BM, Mora S, Glynn RJ, MacFadyen J, Ridker PM. Safety profile of subjects treated to very low low-density lipoprotein cholesterol levels (<30 mg/dl) with rosuvastatin 20 mg daily (from JUPITER). Am J Cardiol 2014; 114:1682-1689
  219. Sinning D, Landmesser U. Low-density Lipoprotein-Cholesterol Lowering Strategies for Prevention of Atherosclerotic Cardiovascular Disease: Focus on siRNA Treatment Targeting PCSK9 (Inclisiran). Curr Cardiol Rep 2020; 22:176
  220. Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, Bisch JA, Richardson T, Jaros M, Wijngaard PLJ, Kastelein JJP. Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. N Engl J Med 2020; 382:1507-1519
  221. Wright RS, Collins MG, Stoekenbroek RM, Robson R, Wijngaard PLJ, Landmesser U, Leiter LA, Kastelein JJP, Ray KK, Kallend D. Effects of Renal Impairment on the Pharmacokinetics, Efficacy, and Safety of Inclisiran: An Analysis of the ORION-7 and ORION-1 Studies. Mayo Clin Proc 2020; 95:77-89
  222. Ray KK, Troquay RPT, Visseren FLJ, Leiter LA, Scott Wright R, Vikarunnessa S, Talloczy Z, Zang X, Maheux P, Lesogor A, Landmesser U. Long-term efficacy and safety of inclisiran in patients with high cardiovascular risk and elevated LDL cholesterol (ORION-3): results from the 4-year open-label extension of the ORION-1 trial. Lancet Diabetes Endocrinol 2023; 11:109-119
  223. Raal FJ, Kallend D, Ray KK, Turner T, Koenig W, Wright RS, Wijngaard PLJ, Curcio D, Jaros MJ, Leiter LA, Kastelein JJP. Inclisiran for the Treatment of Heterozygous Familial Hypercholesterolemia. N Engl J Med 2020; 382:1520-1530
  224. Hovingh GK, Lepor NE, Kallend D, Stoekenbroek RM, Wijngaard PLJ, Raal FJ. Inclisiran Durably Lowers Low-Density Lipoprotein Cholesterol and Proprotein Convertase Subtilisin/Kexin Type 9 Expression in Homozygous Familial Hypercholesterolemia: The ORION-2 Pilot Study. Circulation 2020; 141:1829-1831
  225. Raal F, Durst R, Bi R, Talloczy Z, Maheux P, Lesogor A, Kastelein JJP. Efficacy, Safety, and Tolerability of Inclisiran in Patients With Homozygous Familial Hypercholesterolemia: Results From the ORION-5 Randomized Clinical Trial. Circulation 2024; 149:354-362
  226. Wright RS, Koenig W, Landmesser U, Leiter LA, Raal FJ, Schwartz GG, Lesogor A, Maheux P, Stratz C, Zang X, Ray KK. Safety and Tolerability of Inclisiran for Treatment of Hypercholesterolemia in 7 Clinical Trials. J Am Coll Cardiol 2023; 82:2251-2261
  227. Laufs U, Banach M, Mancini GBJ, Gaudet D, Bloedon LT, Sterling LR, Kelly S, Stroes ESG. Efficacy and Safety of Bempedoic Acid in Patients With Hypercholesterolemia and Statin Intolerance. J Am Heart Assoc 2019; 8:e011662
  228. Goldberg AC, Leiter LA, Stroes ESG, Baum SJ, Hanselman JC, Bloedon LT, Lalwani ND, Patel PM, Zhao X, Duell PB. Effect of Bempedoic Acid vs Placebo Added to Maximally Tolerated Statins on Low-Density Lipoprotein Cholesterol in Patients at High Risk for Cardiovascular Disease: The CLEAR Wisdom Randomized Clinical Trial. JAMA 2019; 322:1780-1788
  229. Ray KK, Bays HE, Catapano AL, Lalwani ND, Bloedon LT, Sterling LR, Robinson PL, Ballantyne CM. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N Engl J Med 2019; 380:1022-1032
  230. Lalwani ND, Hanselman JC, MacDougall DE, Sterling LR, Cramer CT. Complementary low-density lipoprotein-cholesterol lowering and pharmacokinetics of adding bempedoic acid (ETC-1002) to high-dose atorvastatin background therapy in hypercholesterolemic patients: A randomized placebo-controlled trial. J Clin Lipidol 2019; 13:568-579
  231. Ballantyne CM, Laufs U, Ray KK, Leiter LA, Bays HE, Goldberg AC, Stroes ES, MacDougall D, Zhao X, Catapano AL. Bempedoic acid plus ezetimibe fixed-dose combination in patients with hypercholesterolemia and high CVD risk treated with maximally tolerated statin therapy. Eur J Prev Cardiol 2019:2047487319864671
  232. Ballantyne CM, Banach M, Mancini GBJ, Lepor NE, Hanselman JC, Zhao X, Leiter LA. Efficacy and safety of bempedoic acid added to ezetimibe in statin-intolerant patients with hypercholesterolemia: A randomized, placebo-controlled study. Atherosclerosis 2018; 277:195-203
  233. Ridker PM, Lei L, Ray KK, Ballantyne CM, Bradwin G, Rifai N. Effects of bempedoic acid on CRP, IL-6, fibrinogen and lipoprotein(a) in patients with residual inflammatory risk: A secondary analysis of the CLEAR harmony trial. J Clin Lipidol 2023; 17:297-302
  234. Ray KK, Nicholls SJ, Li N, Louie MJ, Brennan D, Lincoff AM, Nissen SE. Efficacy and safety of bempedoic acid among patients with and without diabetes: prespecified analysis of the CLEAR Outcomes randomised trial. Lancet Diabetes Endocrinol 2024; 12:19-28
  235. Nissen SE, Nicholls SJ, Lincoff AM. Bempedoic Acid for Primary Prevention of Cardiovascular Events-Reply. JAMA 2023; 330:1696-1697
  236. Bays HE, Bloedon LT, Lin G, Powell HA, Louie MJ, Nicholls SJ, Lincoff AM, Nissen S. Safety of bempedoic acid in patients at high cardiovascular risk and with statin intolerance. J Clin Lipidol 2023;
  237. Burke AC, Telford DE, Huff MW. Bempedoic acid: effects on lipoprotein metabolism and atherosclerosis. Curr Opin Lipidol 2019; 30:1-9
  238. Ference BA, Ray KK, Catapano AL, Ference TB, Burgess S, Neff DR, Oliver-Williams C, Wood AM, Butterworth AS, Di Angelantonio E, Danesh J, Kastelein JJP, Nicholls SJ. Mendelian Randomization Study of ACLY and Cardiovascular Disease. N Engl J Med 2019; 380:1033-1042
  239. Nissen SE, Lincoff AM, Brennan D, Ray KK, Mason D, Kastelein JJP, Thompson PD, Libby P, Cho L, Plutzky J, Bays HE, Moriarty PM, Menon V, Grobbee DE, Louie MJ, Chen CF, Li N, Bloedon L, Robinson P, Horner M, Sasiela WJ, McCluskey J, Davey D, Fajardo-Campos P, Petrovic P, Fedacko J, Zmuda W, Lukyanov Y, Nicholls SJ. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N Engl J Med 2023; 388:1353-1364
  240. Neef D, Berthold HK, Gouni-Berthold I. Lomitapide for use in patients with homozygous familial hypercholesterolemia: a narrative review. Expert Rev Clin Pharmacol 2016; 9:655-663
  241. Gouni-Berthold I, Berthold HK. Mipomersen and lomitapide: Two new drugs for the treatment of homozygous familial hypercholesterolemia. Atheroscler Suppl 2015; 18:28-34
  242. Rader DJ, Kastelein JJ. Lomitapide and mipomersen: two first-in-class drugs for reducing low-density lipoprotein cholesterol in patients with homozygous familial hypercholesterolemia. Circulation 2014; 129:1022-1032
  243. Cuchel M, Meagher EA, du Toit Theron H, Blom DJ, Marais AD, Hegele RA, Averna MR, Sirtori CR, Shah PK, Gaudet D, Stefanutti C, Vigna GB, Du Plessis AM, Propert KJ, Sasiela WJ, Bloedon LT, Rader DJ, Phase 3 Ho FHLSi. Efficacy and safety of a microsomal triglyceride transfer protein inhibitor in patients with homozygous familial hypercholesterolaemia: a single-arm, open-label, phase 3 study. Lancet 2013; 381:40-46
  244. Blom DJ, Averna MR, Meagher EA, du Toit Theron H, Sirtori CR, Hegele RA, Shah PK, Gaudet D, Stefanutti C, Vigna GB, Larrey D, Bloedon LT, Foulds P, Rader DJ, Cuchel M. Long-Term Efficacy and Safety of the Microsomal Triglyceride Transfer Protein Inhibitor Lomitapide in Patients With Homozygous Familial Hypercholesterolemia. Circulation 2017; 136:332-335
  245. Samaha FF, McKenney J, Bloedon LT, Sasiela WJ, Rader DJ. Inhibition of microsomal triglyceride transfer protein alone or with ezetimibe in patients with moderate hypercholesterolemia. Nat Clin Pract Cardiovasc Med 2008; 5:497-505
  246. Hussain MM, Shi J, Dreizen P. Microsomal triglyceride transfer protein and its role in apoB-lipoprotein assembly. J Lipid Res 2003; 44:22-32
  247. Blom DJ, Cuchel M, Ager M, Phillips H. Target achievement and cardiovascular event rates with Lomitapide in homozygous Familial Hypercholesterolaemia. Orphanet J Rare Dis 2018; 13:96
  248. Sacks FM, Stanesa M, Hegele RA. Severe hypertriglyceridemia with pancreatitis: thirteen years' treatment with lomitapide. JAMA Intern Med 2014; 174:443-447
  249. Underberg JA, Cannon CP, Larrey D, Makris L, Blom D, Phillips H. Long-term safety and efficacy of lomitapide in patients with homozygous familial hypercholesterolemia: Five-year data from the Lomitapide Observational Worldwide Evaluation Registry (LOWER). J Clin Lipidol 2020; 14:807-817
  250. Larrey D, D'Erasmo L, O'Brien S, Arca M. Long-term hepatic safety of lomitapide in homozygous familial hypercholesterolaemia. Liver Int 2023; 43:413-423
  251. Agarwala A, Jones P, Nambi V. The role of antisense oligonucleotide therapy in patients with familial hypercholesterolemia: risks, benefits, and management recommendations. Curr Atheroscler Rep 2015; 17:467
  252. Raal FJ, Santos RD, Blom DJ, Marais AD, Charng MJ, Cromwell WC, Lachmann RH, Gaudet D, Tan JL, Chasan-Taber S, Tribble DL, Flaim JD, Crooke ST. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet 2010; 375:998-1006
  253. Stein EA, Dufour R, Gagne C, Gaudet D, East C, Donovan JM, Chin W, Tribble DL, McGowan M. Apolipoprotein B synthesis inhibition with mipomersen in heterozygous familial hypercholesterolemia: results of a randomized, double-blind, placebo-controlled trial to assess efficacy and safety as add-on therapy in patients with coronary artery disease. Circulation 2012; 126:2283-2292
  254. Santos RD, Duell PB, East C, Guyton JR, Moriarty PM, Chin W, Mittleman RS. Long-term efficacy and safety of mipomersen in patients with familial hypercholesterolaemia: 2-year interim results of an open-label extension. Eur Heart J 2015; 36:566-575
  255. Panta R, Dahal K, Kunwar S. Efficacy and safety of mipomersen in treatment of dyslipidemia: a meta-analysis of randomized controlled trials. J Clin Lipidol 2015; 9:217-225
  256. Duell PB, Santos RD, Kirwan BA, Witztum JL, Tsimikas S, Kastelein JJP. Long-term mipomersen treatment is associated with a reduction in cardiovascular events in patients with familial hypercholesterolemia. J Clin Lipidol 2016; 10:1011-1021
  257. Hashemi N, Odze RD, McGowan MP, Santos RD, Stroes ES, Cohen DE. Liver histology during Mipomersen therapy for severe hypercholesterolemia. J Clin Lipidol 2014; 8:606-611
  258. Raal FJ, Rosenson RS, Reeskamp LF, Hovingh GK, Kastelein JJP, Rubba P, Ali S, Banerjee P, Chan KC, Gipe DA, Khilla N, Pordy R, Weinreich DM, Yancopoulos GD, Zhang Y, Gaudet D. Evinacumab for Homozygous Familial Hypercholesterolemia. N Engl J Med 2020; 383:711-720
  259. Rosenson RS, Burgess LJ, Ebenbichler CF, Baum SJ, Stroes ESG, Ali S, Khilla N, Hamlin R, Pordy R, Dong Y, Son V, Gaudet D. Evinacumab in Patients with Refractory Hypercholesterolemia. N Engl J Med 2020; 383:2307-2319
  260. Rosenson RS, Burgess LJ, Ebenbichler CF, Baum SJ, Stroes ESG, Ali S, Khilla N, McGinniss J, Gaudet D, Pordy R. Longer-Term Efficacy and Safety of Evinacumab in Patients With Refractory Hypercholesterolemia. JAMA Cardiol 2023; 8:1070-1076
  261. Feingold KR. Triglyceride Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2024.
  262. Mattijssen F, Kersten S. Regulation of triglyceride metabolism by Angiopoietin-like proteins. Biochim Biophys Acta 2012; 1821:782-789
  263. Kersten S. New insights into angiopoietin-like proteins in lipid metabolism and cardiovascular disease risk. Curr Opin Lipidol 2019; 30:205-211
  264. Jaye M, Lynch KJ, Krawiec J, Marchadier D, Maugeais C, Doan K, South V, Amin D, Perrone M, Rader DJ. A novel endothelial-derived lipase that modulates HDL metabolism. Nat Genet 1999; 21:424-428
  265. Ishida T, Choi S, Kundu RK, Hirata K, Rubin EM, Cooper AD, Quertermous T. Endothelial lipase is a major determinant of HDL level. J Clin Invest 2003; 111:347-355
  266. Shimamura M, Matsuda M, Yasumo H, Okazaki M, Fujimoto K, Kono K, Shimizugawa T, Ando Y, Koishi R, Kohama T, Sakai N, Kotani K, Komuro R, Ishida T, Hirata K, Yamashita S, Furukawa H, Shimomura I. Angiopoietin-like protein3 regulates plasma HDL cholesterol through suppression of endothelial lipase. Arterioscler Thromb Vasc Biol 2007; 27:366-372
  267. Adam RC, Mintah IJ, Alexa-Braun CA, Shihanian LM, Lee JS, Banerjee P, Hamon SC, Kim HI, Cohen JC, Hobbs HH, Van Hout C, Gromada J, Murphy AJ, Yancopoulos GD, Sleeman MW, Gusarova V. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance. J Lipid Res 2020; 61:1271-1286
  268. Reeskamp LF, Millar JS, Wu L, Jansen H, van Harskamp D, Schierbeek H, Gipe DA, Rader DJ, Dallinga-Thie GM, Hovingh GK, Cuchel M. ANGPTL3 Inhibition With Evinacumab Results in Faster Clearance of IDL and LDL apoB in Patients With Homozygous Familial Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2021:ATVBAHA120315204
  269. Wang Y, Gusarova V, Banfi S, Gromada J, Cohen JC, Hobbs HH. Inactivation of ANGPTL3 reduces hepatic VLDL-triglyceride secretion. J Lipid Res 2015; 56:1296-1307
  270. Musunuru K, Pirruccello JP, Do R, Peloso GM, Guiducci C, Sougnez C, Garimella KV, Fisher S, Abreu J, Barry AJ, Fennell T, Banks E, Ambrogio L, Cibulskis K, Kernytsky A, Gonzalez E, Rudzicz N, Engert JC, DePristo MA, Daly MJ, Cohen JC, Hobbs HH, Altshuler D, Schonfeld G, Gabriel SB, Yue P, Kathiresan S. Exome sequencing, ANGPTL3 mutations, and familial combined hypolipidemia. N Engl J Med 2010; 363:2220-2227
  271. Dewey FE, Gusarova V, Dunbar RL, O'Dushlaine C, Schurmann C, Gottesman O, McCarthy S, Van Hout CV, Bruse S, Dansky HM, Leader JB, Murray MF, Ritchie MD, Kirchner HL, Habegger L, Lopez A, Penn J, Zhao A, Shao W, Stahl N, Murphy AJ, Hamon S, Bouzelmat A, Zhang R, Shumel B, Pordy R, Gipe D, Herman GA, Sheu WHH, Lee IT, Liang KW, Guo X, Rotter JI, Chen YI, Kraus WE, Shah SH, Damrauer S, Small A, Rader DJ, Wulff AB, Nordestgaard BG, Tybjaerg-Hansen A, van den Hoek AM, Princen HMG, Ledbetter DH, Carey DJ, Overton JD, Reid JG, Sasiela WJ, Banerjee P, Shuldiner AR, Borecki IB, Teslovich TM, Yancopoulos GD, Mellis SJ, Gromada J, Baras A. Genetic and Pharmacologic Inactivation of ANGPTL3 and Cardiovascular Disease. N Engl J Med 2017; 377:211-221
  272. Stitziel NO, Khera AV, Wang X, Bierhals AJ, Vourakis AC, Sperry AE, Natarajan P, Klarin D, Emdin CA, Zekavat SM, Nomura A, Erdmann J, Schunkert H, Samani NJ, Kraus WE, Shah SH, Yu B, Boerwinkle E, Rader DJ, Gupta N, Frossard PM, Rasheed A, Danesh J, Lander ES, Gabriel S, Saleheen D, Musunuru K, Kathiresan S. ANGPTL3 Deficiency and Protection Against Coronary Artery Disease. J Am Coll Cardiol 2017; 69:2054-2063
  273. Ahmad Z, Banerjee P, Hamon S, Chan KC, Bouzelmat A, Sasiela WJ, Pordy R, Mellis S, Dansky H, Gipe DA, Dunbar RL. Inhibition of Angiopoietin-Like Protein 3 With a Monoclonal Antibody Reduces Triglycerides in Hypertriglyceridemia. Circulation 2019; 140:470-486
  274. Feingold KR. Lipoprotein Apheresis. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  275. Awad K, Mikhailidis DP, Toth PP, Jones SR, Moriarty P, Lip GYH, Muntner P, Catapano AL, Pencina MJ, Rosenson RS, Rysz J, Banach M. Efficacy and Safety of Alternate-Day Versus Daily Dosing of Statins: a Systematic Review and Meta-Analysis. Cardiovasc Drugs Ther 2017; 31:419-431
  276. Kennedy SP, Barnas GP, Schmidt MJ, Glisczinski MS, Paniagua AC. Efficacy and tolerability of once-weekly rosuvastatin in patients with previous statin intolerance. J Clin Lipidol 2011; 5:308-315
  277. Banach M, Serban C, Sahebkar A, Ursoniu S, Rysz J, Muntner P, Toth PP, Jones SR, Rizzo M, Glasser SP, Lip GY, Dragan S, Mikhailidis DP, Lipid, Blood Pressure Meta-analysis Collaboration G. Effects of coenzyme Q10 on statin-induced myopathy: a meta-analysis of randomized controlled trials. Mayo Clin Proc 2015; 90:24-34
  278. Schaars CF, Stalenhoef AF. Effects of ubiquinone (coenzyme Q10) on myopathy in statin users. Curr Opin Lipidol 2008; 19:553-557
  279. Skarlovnik A, Janic M, Lunder M, Turk M, Sabovic M. Coenzyme Q10 supplementation decreases statin-related mild-to-moderate muscle symptoms: a randomized clinical study. Med Sci Monit 2014; 20:2183-2188
  280. Bogsrud MP, Langslet G, Ose L, Arnesen KE, Sm Stuen MC, Malt UF, Woldseth B, Retterstol K. No effect of combined coenzyme Q10 and selenium supplementation on atorvastatin-induced myopathy. Scand Cardiovasc J 2013; 47:80-87
  281. Caso G, Kelly P, McNurlan MA, Lawson WE. Effect of coenzyme q10 on myopathic symptoms in patients treated with statins. Am J Cardiol 2007; 99:1409-1412
  282. Fedacko J, Pella D, Fedackova P, Hanninen O, Tuomainen P, Jarcuska P, Lopuchovsky T, Jedlickova L, Merkovska L, Littarru GP. Coenzyme Q(10) and selenium in statin-associated myopathy treatment. Can J Physiol Pharmacol 2013; 91:165-170
  283. Hlatky MA, Gonzalez PE, Manson JE, Buring JE, Lee IM, Cook NR, Mora S, Bubes V, Stone NJ. Statin-Associated Muscle Symptoms Among New Statin Users Randomly Assigned to Vitamin D or Placebo. JAMA Cardiol 2023; 8:74-80
  284. Becker DJ, Gordon RY, Halbert SC, French B, Morris PB, Rader DJ. Red yeast rice for dyslipidemia in statin-intolerant patients: a randomized trial. Ann Intern Med 2009; 150:830-839, W147-839
  285. Halbert SC, French B, Gordon RY, Farrar JT, Schmitz K, Morris PB, Thompson PD, Rader DJ, Becker DJ. Tolerability of red yeast rice (2,400 mg twice daily) versus pravastatin (20 mg twice daily) in patients with previous statin intolerance. Am J Cardiol 2010; 105:198-204