Archives

Ambiguous Genitalia in the Newborn

ABSTRACT 

 

Ambiguous genitalia in a newborn are the clinical sign of atypical sexual development of the external genitalia in utero. This condition is rare and can result from various underlying factors, including certain disorders with potentially severe consequences, such as cortisol deficiency due to congenital adrenal hyperplasia. Therefore, it is crucial to promptly determine etiology when ambiguity is observed. The formation of typical male or female external genitalia is a complex process involving a cascade of genetic and physiological events that begin with sex determination and progress through the differentiation of internal and external reproductive structures. When this process is disrupted and does not occur in the typical manner, it is referred to as a difference or disorder of sex development (DSD). Not all DSD cases present with ambiguous genitalia at birth; for example, complete androgen insensitivity syndrome does not, but all cases of ambiguous genitalia are the result of a DSD. This chapter focuses on genital ambiguity associated with DSD in newborns who have either a 46,XY or 46,XX chromosomal sex. However, DSD with genital ambiguity may also be observed in newborns with other combinations of sex chromosomes, such as 45,X/46,XY. The chapter offers a comprehensive overview of the evaluation and management of newborns with ambiguous genitalia. It emphasizes the importance of a structured medical assessment of the external genitalia to diagnose and determine the underlying cause of genital ambiguity. It includes tables for differential diagnosis and step-by-step workup algorithms to guide medical professionals in their evaluation. It also includes additional information on structured physical assessments of external genitalia and tables with normative values for hormonal measurements, which are recommended in the diagnostic process. As the etiology of genital ambiguity in newborns is diverse and can have significant implications for management, the authors stress that obtaining an accurate diagnosis through a professional medical workup is crucial. The chapter highlights the recommendation for newborns with ambiguous genitalia (DSD) to be cared for by highly specialized, interdisciplinary DSD teams. These teams are equipped with medical and psychosocial expertise, and specialized psychologists are available to support parents and caregivers. The chapter recognizes that having a child with ambiguous genitalia can be very stressful for parents. It underscores the importance of early education, access to expert care through DSD network teams, psychological support, shared decision-making, and promoting acceptance and inclusivity. By providing comprehensive support and guidance, families can better navigate the challenges and uncertainties they may encounter when caring for a newborn with ambiguous genitalia.

 

INTRODUCTION

 

Typical male and female development commences with the presence of the typical 46,XY or 46,XX chromosomes, which play a pivotal role in determining the indifferent gonads. Around the 6th week of gestation, these gonads follow pathways to develop into male-typical or female-typical gonads (Figure 1). This gonadal determination is intricately regulated by a complex interplay of multiple genes, which guide the male gonads to become testes and the female gonads to become ovaries.

 

Subsequently, the gonads initiate the production of various hormones, including anti-Müllerian hormone (AMH) and testosterone. These hormones are responsible for driving the differentiation of the embryologic Wolffian and Müllerian structures into sex-typical internal reproductive organs (Figure 1).

 

Figure 1. Typical male and female sex determination and differentiation occur during fetal life. The presence of the Y chromosome in the karyotype and the SRY gene (sex-determining region Y gene) are the primary determinants of male sex development. The SRY gene initiates a cascade that leads to the development of testes from bipotential gonads. Testosterone and Anti Mullerian Hormone (AMH) are secreted by testicular cells, which masculinize the Wolffian (mesonephric) duct and cause the regression of the Mullerian (paramesonephric) duct, respectively. Dihydrotestosterone (DHT), produced by the 5-alpha reduction of testosterone, plays a major role in male external genital development and prostate formation. The absence of the SRY gene on the Y chromosome is the primary factor for the induction of the female sex, and the development of the uterus, fallopian tubes, cervix and upper vagina from the Mullerian duct.

 

The development of external genitalia begins with a neutral anlage, including a genital tubercule, genital folds, and a urogenital sinus. These structures are subsequently differentiated into typical male external genitalia under the influence of androgens, particularly dihydrotestosterone (DHT). Conversely, for the development of typical female external genitalia, the absence of androgens is crucial to prevent virilization.

 

Therefore, ambiguous genitalia at birth can arise from either an excessive or insufficient androgen effect on the neutral external genital anlage in males or females, respectively (Figure 2).

Figure 2. External genital differentiation. The development of external genitalia begins with a bipotential anlage which includes genital tubercle, urethral fold, urethral groove, and genital swellings. In a typical 46, XY, androgen secretion from the testes triggers the fusion of urethral folds, allowing the enclosure of the urethral tube. This, together with the cells from the genital swelling, forms the shaft of the penis. Genital swellings fuse in the midline to allow formation of the scrotum and genital Tubercle expands to give rise to the glans penis. Female external genital development is ensured by the absence of testosterone, it is independent of ovarian endocrine activity. Urethral folds and genital swellings remain separate to form the labia minora and majora. Genital tubercle forms the clitoris.

 

CHARACTERISTICS OF TYPICAL MALE AND FEMALE EXTERNAL GENITALIA

 

A full-term male infant is typically expected to have bilateral testicles that are descended, complete formation of scrotal folds with midline fusion, and a typical-sized penis, which includes well-formed corporal bodies and a urethral meatus located at the tip. The average penile length for a full-term newborn is approximately 3.5 ± 0.4 cm, but this measurement can vary with gestational age (Table 1) (1, 2). If an infant presents with bilateral cryptorchidism, a bifid scrotum, hypospadias, or isolated penoscrotal hypospadias, further investigations for a DSD are recommended. However, it’s important to note that isolated micropenis, as long as both testes are descended and normal in size, is not typically considered a manifestation of ambiguous genitalia. Similarly, in males, distal hypospadias with no other atypical genital features usually does not indicate a DSD (3).

 

Table 1. Stretched Penile Length in Preterm and Term Newborns According to Gestational Age (2)

Gestational age (week)

Number of cases

Median (cm)

Mean (cm)

SD

Min (cm)

Max (cm)

26

30

1.9

1.9

0.32

1.1

2.5

27

31

2.1

2.0

0.28

1.1

2.6

28

32

2.0

1.9

0.31

1.3

2.9

29

29

2.3

2.3

0.39

1.3

2.9

30

31

2.4

2.4

0.32

1.6

3.0

31

33

2.4

2.4

0.38

1.8

3.3

32

32

2.9

2.6

0.44

2.0

3.2

33

28

2.9

2.8

0.45

1.9

3.4

34

30

2.9

2.8

0.43

2.0

3.6

35

29

3.1

2.9

0.61

2.0

4.1

36

31

3.1

3.0

0.50

2.2

4.0

37

29

3.1

3.0

0.47

2.2

4.1

38

44

3.1

3.1

0.54

2.1

4.5

39

82

3.2

3.2

0.55

2.0

4.2

40

65

3.5

3.4

0.53

2.2

4.4

41

29

3.6

3.5

0.47

2.6

4.3

 

A full-term female infant typically presents with bilateral separated labial folds, no palpable gonads, and distinct urethral and vaginal openings (Figure 2). The average clitoral length and width for a full-term infant girl are approximately 6.1 mm and 4.2 mm, respectively (1, 4). In cases where clitoromegaly (clitoral size ˃10 mm), labial fusion or palpable gonads are observed in otherwise typical female genitalia, further investigation for a DSD is warranted. It’s important to note that perceived clitoromegaly is not typically associated with an underlying DSD in the event that the newborn girl was born prematurely (1). Please be aware that perceived clitoromegaly or pseudo-clitoromegaly is often seen in preterm newborns, where the clitoral size falls within the normal range, but the labia majora cannot completely cover labia minora, creating a misleading impression of clitoromegaly. Genital edema may also be observed in sick preterm babies or cases of ovarian hyperstimulation, which can cause pseudo-clitoromegaly.

 

Table 2. Clitoral Length and Width in Preterm and Term Newborns According to Gestational Age (4)

Gestational age (week)

Number of cases

Measure

Mean (mm)

+1 SD (mm)

+2 SD (mm)

+3 SD (mm)

28-30

30

Width

3.28

3.87

4.46

5.05

Length

5.03

5.91

6.79

7.67

30-32

57

Width

3.32

3.68

4.04

4.4

Length

5.03

5.52

6.01

6.5

32-34

74

Width

3.41

3.77

4.13

4.49

Length

5.14

5.65

6.16

6.67

34-36

104

Width

3.69

4.17

4.65

5.13

Length

5.53

6.12

6.71

7.3

36-38

128

Width

4.08

4.5

4.92

5.34

Length

5.83

6.29

6.75

7.21

>38

187

Width

4.21

4.64

5.07

5.5

Length

6.11

6.49

6.87

7.25

 

GENERAL THOUGHTS ON AMBIGUOUS GENITALIA AT BIRTH

 

The reported incidence of ambiguous genitalia in newborns is approximately one in 4,500. It is essential that a newborn with ambiguous genitalia and their parents are promptly referred to a specialized center for evaluation. This evaluation should be carried out by a multi-disciplinary team specializing in DSD in accordance with international recommendations on diagnostic and therapeutic strategies. This team should provide holistic care and treatment guidance, collaborating closely with relevant subspecialists and peer support groups (5). Key subspecialties involved in DSD teams typically include psychology, pediatric endocrinology, urology, neonatology, gynecology, andrology, nursing, social work, genetics, and medical ethics. It's worth noting that certain aspects of DSD management remain contentious or uncertain, resulting in recent changes in clinical practice that are ongoing. Notably, genital surgery has been banned in several countries, and it is increasingly avoided in young children to protect their rights to an open future and the integrity of their bodies.

 

Consequently, we can anticipate a growing number of children growing up with atypical-looking genitalia, requiring a careful assessment of the psychological impact (6). Providing psychological support to affected families as a standard component of care from the outset is, therefore, of utmost importance. Issues related to gender that may arise in the context of ambiguous genitalia should be openly and thoughtfully discussed.

 

A comprehensive evaluation of ambiguous genitalia in a newborn is essential for understanding the underlying cause and potential consequences. On one hand, this evaluation can lead to the detection of concomitant, treatable disorders, such as adrenal insufficiency that requires cortisol replacement. Also, it is important to note that in up to 30% of children with a Difference/Disorder of Sex Development (DSD), other organ system disorders may be identified, necessitating medical care and qualifying as syndromic DSD (7, 8). On the other hand, identifying the underlying cause of ambiguous genitalia forms the basis for making an informed decision regarding sex registration at birth, if necessary and desired by the parents. Gender assignment/sex registration at birth should take into account long-term satisfaction with the sex of rearing, sexual function, and fertility potential (1, 9, 10). Ideally, it should result from a shared decision-making process involving the child's parents and the DSD network team. This decision should be based on the individual's phenotype/genotype, existing knowledge from literature, databases, and patient representative experience (11-13). This process may require time, as parents need to grasp the complexity of their child's unique biology in order to participate in this decision. As a first step, the overall health of the child should be assessed, and parents should be educated about typical sexual development before explanations regarding the underlying cause and its implications for their child's ambiguous genitalia are provided. Even if the exact details of the underlying cause remain unresolved, it is essential to communicate what is known and what remains uncertain to the parents and make plans for the family's future in the best interests of the child.

 

Several websites are available for patients and their families to exchange information, share coping strategies, and discuss decision-making processes that can enhance outcomes for newborns with ambiguous genitalia. Such resources can be found on websites supported by organizations like the Accord Alliance (www.accordalliance.org), DSD families (www.dsdfamilies.org), AIS DSD Support Group (www.aisdsd.org), and the CARES Foundation (www.caresfoundation.org). However, the most current information will always be available from the responsible DSD team, which will also be aware of country-specific support services.

 

GENETICS OF SEX DETERMINATION AND DIFFERENTIATION 

 

Sex development is a sequential process that involves the coordinated action of numerous genes and pathways. This process culminates in the development of functional gonads, differentiated internal sex organs, external genitalia, and typical secondary sexual characteristics that manifest after puberty. Prenatal sex development can be divided into two distinct processes: sex determination and sex differentiation (Figure 3). Sex determination is the initial step that occurs shortly after conception, approximately at 6-7 weeks gestation. During this phase, the undifferentiated gonads evolve into either testes or ovaries. By contrast, sex differentiation occurs subsequently and involves hormones produced after the formation of the sex-specific gonads. These hormones play a critical role in shaping the further development of the dependent embryonic structures, leading to the formation of male and female internal and external phenotypes.

 

Figure 3. The genes and transcription factors required for gonadal development. The formation of the bipotential gonad requires at least seven genes including NR5A1 (Nuclear Receptor Subfamily 5 Group A Member 1), WT1 (WilmsTumor1 Transcription Factor), LHX9 (LIM Homeobox 9), EMX2 (Empty Spiracles Homeobox 2), CBX2 (Chromobox 2), PBX1 (Pre-B-Cell Leukemia Transcription Factor 1) and GATA4 (GATA Binding Protein 4). Adapted from (14).

 

The Male Specific Pathway

 

In the male specific pathway, the expression of the SRY gene in pre-Sertoli cells serves as a pivotal switch that directs the fate of the gonads toward testicular development. SRY, in conjunction with NR5A1, activates the SOX9 signaling pathway, initiating testis development (Figure 3). Other important genes for the early testis development are NR0B1(Nuclear Receptor subfamily 0 group B member 1), AMH (Anti-Mullerian Hormone) and GATA4. AMH secreted by Sertoli cells, plays a crucial role in promoting the regression of Müllerian structures and supporting the development of Leydig cells in the testes. Subsequently, androgens produced by the testes facilitate the development of male internal organs (epididymis, vas deferens, seminal vesicle) and external genitalia (penis, scrotum). The production of sex steroids relies on an intact steroidogenic pathway, with all steroid hormones originating from cholesterol. The initial steps of sex steroid biosynthesis overlap with the synthesis of mineralo-corticosteroids and gluco-corticosteroids (Figure 4). It is important to note that certain steroid disorders, often referred to as congenital adrenal hyperplasia (CAH), can affect both adrenal and gonadal steroidogenesis or result in androgen excess or deficiency that may impact fetal genital development. INSL3, produced in Leydig cells, plays a role in guiding the descent of the testes from the abdomen to the scrotal folds.

 

The process of masculinization of initially sex-neutral, undifferentiated external genitalia begins around 8-9 weeks of gestation when the potent androgen DHT (dihydrotestosterone) is produced by the testes. DHT is crucial for the fusion of the urethral and labioscrotal folds, elongation of the genital tubercle, regression of the urogenital sinus, and the development of the prostate (15). The enzyme 5α-reductase is responsible for converting testosterone produced by fetal Leydig cells into DHT. Complete masculinization of the external genitalia through DHT is typically achieved by week 14 of gestation (16).

 

Figure 4. Steroidogenesis of the adrenals and gonads. Adrenal and gonadal steroidogenesis starting from cholesterol and showing key steroids and intermediates of the classical and alternative pathways is illustrated. Cortisol and androgen biosynthesis are under control of hypothalamic-pituitary-adrenal (HPA) axis in a negative feedback loop. Androgen synthesis occurs via classical or aberrant and alternative pathways (i.e., backdoor pathway and 11oxC19 androgen pathway). Aberrant or alternative pathways are particularly active whenever cortisol with or without aldosterone biosynthesis is impaired and androgenic precursors accumulate proximal to enzymatic block. Abbreviations: StAR; Steroidogenic Acute Regulatory Protein defect, CYP11A1; P450 side-chain cleavage deficiency, CYP21A2; 21α-hydroxylase, HSD3B2; 3β-hydroxy steroid dehydrogenase 2, CYP11B1; 11β-hydroxylase, CYP11B2; Aldosterone synthase, DOC; 11-deoxycorticosterone, DHEA; Dehydroepiandrosterone, A4; Androstenedione, T; Testosterone, CYP17A1; 17α-hydroxylase/17,20-lyase, b5; CYP19A1; Cytochrome P450 aromatase, Cytochrome b5, POR; P450 oxidoreductase, 17β-HSD; 17β-hydroxysteroid oxidoreductase or 17-ketosteroid reductase; SRDA1/2; Steroid 5α -reductase 1/2, 11oxC19; 11-oxygenated 19-carbon (11oxC19) steroids; Fdx; Ferredoxin, FdR; Ferredoxin reductase, 11βOHA; 11β-hydroxyandrostenedione, 11βOHT; 11β-hydroxytestosterone, Pdione; 5α-Pregnane- 17α-ol-3,20-dione, Pdiol; 5α-Pregnane- 3α,17α-diol 20-one, AKR1C2,4; Aldo-Keto Reductase Family 1 Member C2,4, DHT; Dihydrotestosterone, HSD17B5,3,6= 17β-hydroxysteroid dehydrogenase 5,3,6. Adapted from (17).

 

The Female Specific Pathway

 

By contrast, the female-specific pathway is initiated in the absence of SRY and requires the suppression of SOX9 (Figure 3). Activation of Rspo1/Wnt4/b-catenin and Foxl2 (Forkhead Box L2) signaling pathways leads to the formation of typical ovaries. The absence of AMH allows the Müllerian structures to proliferate, resulting in the formation of the typical fallopian tube, uterus, cervix and the upper part of the vagina. In the absence of both testosterone secretion and 5α-reductase activity, the sex-neutral external genitalia develop along the female pathway. Specifically, without DHT, the labioscrotal and urethral folds give rise to the labia majora and minora, respectively. The genital tubercle develops into a clitoris, and the urogenital sinus contributes to the formation of the urethral opening and anterior portion of the vagina (16).

 

Any disruption in the intricate genetic and hormonal processes involved can lead to atypical sex development and result in atypical external genitalia at birth. Figure 3 provides a summary of these processes and highlights some key genes and hormones essential for typical development.

 

DIFFERENCES/DISORDERS OF SEX DEVELOPMENT (DSD) 

 

The discordance between genetic, gonadal, or anatomic sex is commonly referred to as DSD. In addition to candidate gene testing, the new era of molecular diagnostic tools, including whole exome/genome sequencing has uncovered numerous novel molecular etiologies in recent years. Accurate molecular diagnosis aids in managing affected individuals and provides families with information concerning prognosis and the risk of recurrence.  This information is often sourced from literature and comprehensive data registries, such as the I-DSD (18-24). For clinical purposes, DSD (including ambiguous genitalia in newborns) is typically classified based on the affected individual's karyotype. The consensus statement addressing the approach and care of DSD (originally published in 2006 and updated in 2016) suggests the following broad classification for DSD: (A) Sex Chromosome DSD, (B) 46,XY DSD, and (C) 46,XX DSD (Table 3) (1, 20).

 

Table 3. DSD Classification According to the Chicago Consensus.

Sex Chromosome DSD

46,XY DSD

46,XX DSD

A. 45,X0 (Turner syndrome and variants)

A. Disorders of gonadal (testicular) development

Complete gonadal dysgenesis (Swyer syndrome) or partial gonadal dysgenesis (e.g., SRY,SOX9, NR5A1, WT1, DHHgene mutations etc)

Testicular regression (e.g., DHX37 gene mutation)

Ovo-Testicular DSD

A. Disorder of gonadal (ovarian) development

Ovo-Testicular DSD

Testicular DSD (SRY (+))

Gonadal dysgenesis

B. 47,XXY (Klinefelter syndrome and variants)

B. Disorders of testicular hormone production or action

Impaired testosterone production

LH receptor mutations

CAH (3βHSD2 deficiency, 17OHD, POR, StAR, CYP11A1 deficiency)

HSD17B3 deficiency

 

Impaired testosterone action

5α-reductase deficiency

Androgen insensitivity syndrome (Complete/partial)

 

Impaired AMH production or action

Persistent Mullerian Duct Syndrome (AMH and AMHR2gene mutations)

B. Androgen excess

Fetal

CAH (21-hydroxylase, 11β-hydroxylase, 3βHSD2 deficiency)

Glucocorticoid resistance

 

Feto-placental

Aromatase deficiency

POR deficiency

 

Maternal

Luteoma, hilar cell tumors, arrhenoblastoma, lipoid cell tumors, Krukenberg tumors, androgen producing adrenal tumors,

External androgen exposure

C. 45,X0/46,XY (Mixed gonadal dysgenesis, ovotesticular DSD)

C. Other

Syndromic

Smith-Lemli-Opitz, Cloacal anomaly, Aarskog, Robinow, Meckel, Joubert, Hand-Foot-Genital, popliteal pterygium, CHARGE, VATER/VACTERL, IMAGe etc.

 

Drugs

Flutamide, ketoconazole, progestagens

 

Endocrine disruptors

Phthalate, BPA, paraben

 

Cryptorchidism

INSL3, GREAT gene mutations etc.

 

Isolated hypospadias

MAMLD1, HOXA4 gene mutations etc.

C. Other

Syndromic

Cloacal anomaly etc.

 

Mullerian anomalies

Mayer-Rokitanski-Küster-Hauser, MURCS, McKusick-Kaufmnann syndrome and variants, MODY5

 

D. 46,XX/46,XY (chimeric, ovotesticular DSD)

 

 

Abbreviations: StAR; Steroidogenic Acute Regulatory Protein defect, CYP11A1; P450 side-chain cleavage deficiency, 3βHSD2; 3β-hydroxy steroid dehydrogenase 2, CYP17A1; 17α-hydroxylase/17,20-lyase deficiency, 17OHD; 17α-hydroxylase/17,20-lyase deficiency, POR; P450 oxidoreductase, HSD17B3; 17β-hydroxysteroid oxidoreductase or 17-ketosteroid reductase; SF1/NR5A1; steroidogenic factor 1.

 

The 46,XY Newborn With Ambiguous Genitalia

 

Ambiguous genitalia in a 46XY newborn can result from abnormal formation of the early fetal testes (testicular dysgenesis), reduced production of testosterone or dihydrotestosterone (5α-reductase deficiency), or the inability to respond to androgens (androgen insensitivity syndrome, or AIS) (Table 3). Depending on the extent of androgen production defect or resistance, affected newborns may exhibit a range of external genitalia phenotype, varying from those that appear typically female to those that appear typically male but with a small phallus, hypospadias, and a bifid scrotum, with or without palpable testes. Here we focus primarily on causes that lead to ambiguous genitalia. Broadly, four groups of underlying causes of 46, XY DSD with ambiguous genitalia can be identified: (a) partial gonadal dysgenesis, (b) partial testosterone biosynthetic defects, (c) partial 5α-reductase deficiency, and (d) partial androgen insensitivity syndrome or PAIS.

 

PARTIAL GONADAL DYSGENESIS

 

In cases of partial gonadal dysgenesis, it is presumed that a gene mutation leads to a partial abnormality in the development of the early urogenital ridge or gonadal anlage. This category of DSD can also involve mutations in genes such as SRY and SOX9, which are essential for the differentiation of bipotential gonads into testes (Figure 3). Partial gonadal dysgenesis is typically associated with incomplete masculinization of the external genitalia, along with varying degrees of maintenance of Wolffian ducts and inhibition of Müllerian ducts. For a summary of known genetic variations associated with gonadal dysgenesis, please refer to Table 4.

 

Leydig cell aplasia or hypoplasia represents a variation of gonadal dysgenesis, characterized by inadequate Leydig cell differentiation in the male gonad, resulting in impaired androgen production.  Mutations in the LH receptor gene (LHCGR) as well as polymorphisms have been associated with this condition (25). The phenotype associated with Leydig cell hypoplasia includes incomplete masculinization of the external genitalia, along with incomplete development and maintenance of the Wolffian ducts. 

 

 

Table 4. Monogenic Disorders of Gonadal Development/Differentiation and Associated DSD Presentations in Humans, Both 46,XY and 46,XX

Gene defect

Locus

Mode of transmission

Clinical presentation

Reference

 

 

 

DSD phenotype

Other

 

 

 

 

46, XX

46, XY

 

 

ARX

Xp22.13

XL

 

Testicular dysgenesis

Lissencephaly, epilepsy, intellectual disability temperature instability

(26)

ATRX

Xq13.3

XL

 

Testicular dysgenesis

Dysmorphic features, intellectual disability, α-thalassemia

(27)

CBX2

17q25

AR

Ovarian dysgenesis

Testicular dysgenesis

 

(28)

DHH

12q13.12

AR

 

Testicular dysgenesis

Minifascicular neuropathy

(29)

DMRT1/DMRT2                       

9p24.3

AD

Ovarian dysgenesis

Testicular dysgenesis

Hypotonia, developmental delay, impaired intellectual development

(30)

DHX37

12q24.31

AD

 

Testicular dysgenesis, testicular regression

 

(31)

EMX2

10q26.11  

 

Ovarian dysgenesis

Testicular dysgenesis

Intellectual disability, kidney agenesis

(32)

ESR2

14q23.2-q23.3

AD,?

Ovarian dysgenesis

Testicular dysgenesis

Dysmorphic features, eye abnormalities, anal atresia, rectovestibular fistula

(33)

FGFR2

10q26.13

AD

 

Testicular dysgenesis, ovotesticular

Craniosynostosis

(34)

GATA4 

8p23.1

AD

 

Testicular dysgenesis

Congenital heart defects (atrial septum defects, ventricular septum defects, tetralogy of Fallot), diaphragmatic hernia

(35)

HHAT

1q32.2

AR

 

Testicular dysgenesis

Nivelon-Nivelon-Mabille syndrome. Dwarfism, chondrodysplasia, narrow, bell-shaped thorax, micromelia, brachydactyly, microcephaly with cerebellar vermis hypoplasia, facial anomalies, hypoplastic irides and coloboma of the optic discs

(36)

LHX9

1q31.3

AD

 

Testicular dysgenesis

Limb anomalies

 

(37)

MAMLD1

Xq28

XL

 

Severe hypospadias

Myotubular myopathy

 

(38)

MAP3K1

5q11.2

AD

 

Testicular dysgenesis

 

(39)

NR0B1 (Duplications, inversion

and upstream deletion)

X21.3

XL

 

 

Testicular dysgenesis

Congenital adrenal hypoplasia, hypogonadotropic hypogonadism, cleft palate, intellectual disability

 

(40)

NR2F2

15q26.2

AD

Ovarian dysgenesis, ovotesticular DSD

 

Congenital heart disease, somatic anomalies including

blepharophimosis-ptosis-epicanthus

inversus syndrome

(41)

NR5A1

9q33

AD

Ovarian dysgenesis, ovotesticular DSD

Testicular dysgenesis

Adrenocortical insufficiency,

spermatogenic failure, primary ovarian insufficiency, asplenia, polysplenia

(42)

PPP2R3C

14q13.2

AR

 

Testicular dysgenesis

Dysmorphic facies, retinal dystrophy, and

myopathy.

 (43)

RSPO1

1p34.3

AR

Testicular/

ovotesticular DSD

 

Palmoplantar hyperkeratosis

 (44)

SART3

12q23.3

AR

 

Testicular dysgenesis

Intellectual disability, global developmental delay and a subset of brain anomalies

 

(45)

SOX3 (Upstream deletion and

Duplication)

Xq27.1

XL

 

Testicular/ovotesticular DSD

 

Intellectual disability, hypopituitarism

(46)

SOX8 (SNV, upstream

duplication, inversion)

16p13.3

AD

 

 

Testicular dysgenesis

Variants may contribute to male and female infertility, primary ovarian insufficiency.

(47)

SOX9 (SNV, upstream

duplication, inversion)

17q24.3

AD

 

Testicular/ovotesticular DSD

Testicular dysgenesis

Campomelic dysplasia, Cooks syndrome, Pierre Robin sequence

 

 (48)

SOX10 (Duplication)

22q13.1

AD

Testicular/ovotesticular DSD

 

Peripheral and central demyelination, Waardenburg syndrome type IV, and Hirschsprung disease

 (49)

SRY (SNV, deletion,

Translocation)

Yp11.3

XL, YL

Testicular/ovotesticular DSD

Testicular dysgenesis

 

 (50, 51)

 

TSPYL1

6q22.1

AR

 

Testicular dysgenesis

Sudden infant death syndrome

(52)

WT1

11p13

AD

Testicular/ovotesticular DSD

Testicular dysgenesis

Denys-Drash syndrome; Frasier syndrome; WAGR syndrome; Meacham syndrome, Nephrotic syndrome; Wilms tumor

(53, 54)

 

 

Abbreviations: AD; autosomal dominant, AR; autosomal recessive, XL; X-linked, YL; Y-linked

 

DEFICIENCY OF TESTOSTERONE BIOSYNTHESIS

 

The inability to produce testosterone stems from defects in the activity of any of the enzymes required for testosterone biosynthesis from cholesterol (Figure 4able 5). Identified defects encompass genetic variants in Steroidogenic Acute Regulatory Protein (StAR), P450 side-chain cleavage (CYP11A1), 3β-hydroxy steroid dehydrogenase 2 (HSD3B2), 17α-hydroxylase/17,20-lyase (CYP17A1), and P450 oxidoreductase (POR) (59). In addition, apparent lyase deficiency due to variants in cytochrome b5 (CYB5) and variants in AKR1C2/4 have been very rarely described (60-62). Moreover, 17β-hydroxysteroid dehydrogenase deficiency (also known as 17β–hydroxysteroid oxidoreductase or 17-ketosteroid reductase, HSD17B3) is another cause of 46,XY DSD (63). Furthermore, variants in steroidogenic factor 1 (SF1/NR5A1), which regulates the transcription of several genes involved in steroidogenesis, may lead to ambiguous genitalia. Notably, variants in HSD3B2, POR, and NR5A1 can result in ambiguous genitalia in both 46,XY and 46,XX newborns (64-66).

 

Similar to partial gonadal dysgenesis, a partial testosterone biosynthesis defect leads to ambiguous external genitalia and variable degrees of Wolffian duct development. However, unlike partial gonadal dysgenesis, Müllerian ducts are not retained in 46,XY newborns with partial testosterone biosynthesis defects due to intact Sertoli cell function and normal AMH production.

 

Table 5. Adrenal and/or Gonadal Causes of Impaired Sex Steroid Biosynthesis Associated with Ambiguous Genitalia in 46,XX and 46,XY Individuals

Disorder

Gene/ OMIM

Adrenal Insufficiency

46,XY Gonadal Phenotype

(Testosterone Deficiency)

46,XX Gonadal Phenotype

(E2 Deficiency)

Fertility

Other Features

 

Lipoid congenital adrenal hyperplasia (LCAH)

StAR

201710

YES

Classic form: 46,XY DSD, gonadal insufficiency; ambiguous genitalia

Non-classic form: normal or NK

Classic: primary or secondary ovarian insufficiency (POI)

Non-classic: NK or normal

Classic:Absent in 46,XY; variable in 46,XX

 

P450 side chain cleavage syndrome (CAH)

CYP11A1

118485

YES

Classic form: 46,XY DSD, gonadal insufficiency; ambiguous genitalia

Non-classic form: normal

Classic: primary or secondary ovarian insufficiency (POI)

Non-classic: NK or normal

Reported in 46,XX

 

3β-hydroxysteroid dehydrogenase II deficiency (CAH)

HSD3B2

201810

YES

46,XY DSD, gonadal insufficiency; ambiguous genitalia

Non-classic form: normal, but premature adrenarche

46,XX DSD with atypical genital development; gonadal insufficiency; ambiguous genitalia

Non-classic form: normal, but premature adrenarche

Absent in 46,XY; reported in 46,XX

 

21-hydroxylase deficiency (CAH)

CYP21A2

201910

YES

Classic form: normal

Non-classic form: normal

46,XX DSD with atypical genital development;

Non-classic form: premature adrenarche, virilization, PCO

Normal in both 46,XX and 46,XY, if treated

Cave: Testicular adrenal rest tumor (m>>f)

CAH-X (when combined with Ehlers-Danlos syndrome with contiguous gene variants)

11-hydroxylase deficiency (CAH)

CYP11B1

202010

YES

Classic form: normal

Non-classic form: normal

46,XX DSD with atypical genital development;

Non-classic form: premature adrenarche, virilization, PCO

Normal in both 46,XX and 46,XY, if treated

Hypertension

Combined 17-hydroxylase, 17,20 lyase deficiency (CAH)

CYP17A1

202110

Rare

46,XY DSD, gonadal insufficiency; atypical genital development

Lack of pubertal development, POI

Possible in 46,XX with assisted fertility measures

Hypertension and hypokalemic alkalosis (not seen with isolated lyase deficiency)

P450 oxidoreductase deficiency (CAH)

POR

124015

201750

Variable

Mild to severe 46,XY DSD, gonadal insufficiency; atypical genital development

46,XX DSD with atypical genital development or

premature adrenarche, virilisation, POI, PCO

Reported

Maternal virilization during pregnancy; Antley-Bixler skeletal malformation syndrome; changes in drug metabolism

Cytochrome b5 deficiency

CYB5A

613218

NO

46,XY DSD; atypical genital development

NK

NK

Methemoglobinemia

17β-hydroxysteroid dehydrogenase III deficiency / 17-ketosteroid reductase deficiency

HSD17B3

264300

NO

46,XY DSD; atypical genital development; progressive virilization and gynecomastia at puberty

Normal

Decreased or absent in 46,XY

 

5α-reductase II deficiency

SRD5A2

607306

NO

46,XY DSD; atypical genital development; progressive virilisation and gynecomastia at puberty

Normal

Impaired in 46,XY

 

3α-hydroxysteroid dehydrogenase deficiency

AKR1C2/4

600450

600451

NO

46,XY DSD; gonadal insufficiency; atypical genital development

Normal

NK

 

Aromatase deficiency

CYP19A1

107910

NO

Normal

46,XX DSD with variable degree of virilisation at birth (ambiguous genitalia), gonadal insufficiency, POI

Impaired in 46,XX

Overgrowth and metabolic anomalies in males

Steroidogenic factor 1

NR5A1/

SF1

184757

Rare

Mild to severe 46,XY DSD; gonadal insufficiency – very variable; ambiguous genitalia

POI or normal; rarely 46,XX DSD with atypical genital development

Mostly impaired in 46,XY; variable in 46,XX

 

Abbreviations: NK; not known, POI; primary ovarian insufficiency, PCO; polycystic ovary.

 

5-ALPHA-REDUCTASE-2 DEFICIENCY (SRD5A2 VARIANTS)

 

Deficiency of the 5α-reductase enzyme arises from variants in the steroid 5α-reductase type 2 (SRD5A2) gene, which can show single point mutations to complete gene deletions. Newborns affected by this condition possess functioning Leydig and Sertoli cells, but due to their inability to convert testosterone to DHT, they may exhibit varying degrees of under-masculinized external genitalia, including genital ambiguity at birth in some cases (16, 67-69). The Müllerian ducts in affected individuals regress as expected, due to normal Sertoli cell function and AMH production. During puberty, significant virilization becomes possible in affected individuals, and the testes are capable of supporting spermatogenesis, as DHT is not required for germ cell maturation. Therefore, fertility is attainable in less severely affected individuals (70-72) or with the use of intrauterine insemination. Recent evidence also suggests that SRD5A2 activity may be even influenced by genetic polymorphisms of SRD5A2 (50).

 

GENETIC DEFECTS OF ANDROGEN ACTION

 

Complete or partial androgen insensitivity syndromes (CAIS/PAIS) are caused by genetic mutations affecting androgen receptor (AR) function (73, 74), and serum testosterone levels are typically elevated. Internationally, incidence of AIS is reported in 1 in 20,400 live born 46,XY infants, with CAIS occurring at a higher rate than PAIS (75).

 

The human androgen receptor (AR) is encoded by a single gene (AR) composed of 8 exons located in the q11-12 region of the X chromosome. The AR gene mutation database includes hundreds of AR variants that lead in varying degrees of atypical sex differentiation of 46,XY fetuses, also classified as AIS type I (76-78). A small number of complete AR gene deletions have been reported, as well as deletions that start at exons 2, 3 or 4 and extend to the terminus of the gene. In addition, a limited number of mutations resulting from premature terminations, base deletions and terminations have been identified. However, the most common type of AR gene mutation results from base substitutions (79). When mutations result in partial inactivation of AR activity (PAIS), the phenotypic variability in under-masculinization can be substantial and depends on the residual activity of the AR. In some cases, this can lead to ambiguous genitalia in newborns with a 46,XY chromosomal sex.

 

Unlike in CAIS, the underlying genetic defect in PAIS is only found in the AR gene in about 40% of cases (80). However, the clinical and biochemical phenotype is similar, and in vitro functional tests demonstrate androgen resistance in all of them. Thus, individuals with AIS without AR mutations are classified as AIS type II (73). In cases of AIS type II without AR variants, largely unidentified regulators or cofactors of the AR are responsible for the impaired AR signaling, as revealed by an AR-dependent bioassay using genital skin fibroblasts and the targeted apolipoprotein D as a biomarker (81). Given that AR activity can be regulated at various levels, the potential mechanisms of AIS type II are diverse.Recently, altered DNA methylation of the AR promoter has been observed in some individuals with PAIS (82). Additionally, mutations in the Disheveled associated activator of morphogenesis 2 gene (DAAM2) that impair nuclear actin assembly at AR have been associated with AIS type II (83).

 

Overall, individuals with PAIS experience variable degrees of end-organ unresponsiveness to androgens, resulting in varying degrees of Wolffian duct development and external genital ambiguity. Within families with the same AR mutation, phenotypic variability can occur due to variable degrees of insensitivity to androgens (84, 85). Consequently, solely relying on genetic information, it is challenging to predict whether a newborn with PAIS will identify as male, intersex, or female in later life and respond to future testosterone therapy. Similarly, genetic counselling is challenging.

 

The 46,XX Newborn With Ambiguous Genitalia

 

Ambiguous genitalia in a chromosomal 46,XX fetus invariably results from androgen excess during fetal development, and the degree of virilization (Figure 2) often provides clues about the timing and severity of the underlying disorder.

 

Similar to a 46,XY fetus, a 46,XX fetus may experience partial gonadal dysgenesis or develop testicular or ovo-testicular DSD due to pathogenic variants in genes involved in early sex determination (Table 3 and 4). Ovo-testicular DSD occurs when both ovarian and testicular tissue develop in the same individual (1). Most newborns with ovo-testicular DSD possess 46,XX chromosomes and present with ambiguous genitalia, although some affected individuals have 46,XY chromosomes or 46,XX/46,XY mosaicism. Just like in 46,XY DSD, the degree of testicular development determines the extent of masculinization/virilization of the external genitalia, Wolffian duct development, and Müllerian duct regression in affected newborns (79).  A summary of known genetic variations is given in Table 4.

 

In the majority of cases, ambiguous genitalia in a 46,XX fetus with typical ovarian organogenesis can be attributed to excessive androgen production/exposure early in utero, originating either from the fetus itself (Table 3 and Table 5) or from the mother or environment.

 

ABNORMAL FETAL ANDROGEN PRODUCTION BY CONGENITAL ADRENAL HYPERPLASIA (CAH)

 

The term congenital adrenal hyperplasia (CAH) encompasses several adrenal disorders, each linked to a mutation in one of the enzymes necessary for the biosynthesis of cortisol from cholesterol (86) (Figure 4 and Table 5). These abnormalities lead to increased ACTH secretion by the pituitary gland, which, in turn, results in the increased secretion of cortisol precursors, including adrenal androgens.

 

Deficiency of steroid acute regulatory protein (StAR) causes congenital lipoid adrenal hyperplasia, characterized by salt loss and a lack of cortisol, androgen, and estrogen secretion. Genetic 46,XX infants affected by StAR deficiency exhibit typical female external genitalia, while genetic 46,XY are born with ambiguous genitalia or may experience sex reversal in cases of severe loss-of-function variants. A similar phenotype is also observed with variants in CYP11A1.

 

Deficiency in 17α-hydroxylase/17,20-lyase leads to hypertension due to the hypersecretion of corticosterone and impaired androgen secretion. Similar to congenital lipoid adrenal hyperplasia, 46,XX fetus affected by 17α-hydroxylase/17, 20-lyase deficiency (CYP17A1) develop typical female external genitalia, while 46,XY fetus present with ambiguity.

 

Deficiency of 3β-hydroxysteroid dehydrogenase (HSD3B2) results in salt loss and impaired androgen synthesis. Affected 46,XX females may or may not exhibit minimal genital masculinization, whereas all genetic 46,XY males are variably under-masculinized (87).

 

CAH due to 11β-hydroxylase deficiency (CYP11B1) and 21-hydroxylase deficiency (CYP21A2) results in the most significant masculinization/virilization of external genitalia in 46,XX fetus compared to all other types of CAH. Additionally, 11β-hydroxylase deficiency can lead to hypertension in individuals of either sex in later life (88, 89).

 

21-hydroxylase deficiency CAH, caused by variants in CYP21A2, represents the most common form (accounting for more than 90% of cases) of CAH and is the most frequent genetic cause of DSD with ambiguous genitalia at birth in 46,XX newborns (59, 63). Cortisol deficiency necessitates replacement therapy in all classic forms of CAH. In the milder (simple-virilizing) presentation of classical 21-hydroxylase deficiency, salt loss is typically not an issue, whereas in the more severe (salt-wasting) form of classical 21-hydroxylase deficiency, salt-loss occurs and may pose a risk to the newborn shortly after birth if not treated with both mineralocorticoids and glucocorticoids (86, 90, 91).

 

The pathological mechanisms of 21-hydroxylase deficiency leading to 46,XX virilization in utero have been well described (92). It involves androgen excess due to lack of 21-hydroxylase activity during the critical time window around 6-12 weeks of gestation when the external genitalia are formed, and adrenal androgen production should be redirected to cortisol production to protect the female external genital anlage. In 46,XX CAH (Table 5), the resulting adrenal androgen excess exceeds the androgen metabolizing capacities of the fetal-placental unit, and alternative pathways of androgen production (Figure 4) are also activated, contributing to the overall androgen excess. Figure 5 illustrates in a scheme how the fetal adrenals, liver and placenta cooperate under normal conditions to metabolize fetal adrenal androgens into estrogens.

 

Figure 5. Steroid metabolic pathways in the fetal-placental-maternal unit, also known as the fetal-placental unit. The main function of the FZ of the fetal adrenal is the production of DHEA-S from cholesterol, which is transported to the placenta, desulfated to DHEA, and sequentially metabolized by placental 3β-hydroxysteroid dehydrogenase type 1 (3βHSD1), 17β-hydroxysteroid dehydrogenases (17βHSD) and cytochrome P450 aromatase (CYP19A1), to androstenedione (A4), testosterone (T) and estradiol (E2), respectively. E2 is then transported from the placenta into the maternal circulation. A4 is additionally also a substrate for placental CYP19A1, producing estrone (E1). Fetal adrenal DHEA-S is also metabolized by the fetal liver to 16α-hydroxy-DHEA-S by cytochrome P450 family 3 subfamily A member 7 (CYP3A7), which is metabolized to estriol (E3), the estrogen marker of pregnancy. 
Abbreviations: PREG, pregnenolone; 17OHPREG, 17α-hydroxypregnenolone; PROG, progesterone; DHEA, dehydroepiandrosterone; DHEA-S, dehydroepiandrosterone-sulfate; CYP11A1, cytochrome P450 cholesterol side chain cleavage; StAR, steroidogenic acute regulatory protein; CYP17A1, cytochrome P450 17α-hydroxylase/17,20-lyase; SULT2A1, sulfotransferase; CYP3A7, cytochrome P450 family 3 subfamily A member 7; CYP19A1, cytochrome P450 aromatase; HSD17B, 17β-hydroxysteroid dehydrogenase; HSD3B1, 3β-hydroxysteroid dehydrogenase type 1; HSD11B2, 11β-hydroxysteroid dehydrogenase type 2; STS, sulfatase.

Figure 5. Steroid metabolic pathways in the fetal-placental-maternal unit, also known as the fetal-placental unit. The main function of the FZ of the fetal adrenal is the production of DHEA-S from cholesterol, which is transported to the placenta, desulfated to DHEA, and sequentially metabolized by placental 3β-hydroxysteroid dehydrogenase type 1 (3βHSD1), 17β-hydroxysteroid dehydrogenases (17βHSD) and cytochrome P450 aromatase (CYP19A1), to androstenedione (A4), testosterone (T) and estradiol (E2), respectively. E2 is then transported from the placenta into the maternal circulation. A4 is additionally also a substrate for placental CYP19A1, producing estrone (E1). Fetal adrenal DHEA-S is also metabolized by the fetal liver to 16α-hydroxy-DHEA-S by cytochrome P450 family 3 subfamily A member 7 (CYP3A7), which is metabolized to estriol (E3), the estrogen marker of pregnancy. Abbreviations: PREG, pregnenolone; 17OHPREG, 17α-hydroxypregnenolone; PROG, progesterone; DHEA, dehydroepiandrosterone; DHEA-S, dehydroepiandrosterone-sulfate; CYP11A1, cytochrome P450 cholesterol side chain cleavage; StAR, steroidogenic acute regulatory protein; CYP17A1, cytochrome P450 17α-hydroxylase/17,20-lyase; SULT2A1, sulfotransferase; CYP3A7, cytochrome P450 family 3 subfamily A member 7; CYP19A1, cytochrome P450 aromatase; HSD17B, 17β-hydroxysteroid dehydrogenase; HSD3B1, 3β-hydroxysteroid dehydrogenase type 1; HSD11B2, 11β-hydroxysteroid dehydrogenase type 2; STS, sulfatase.

 

FETAL AND PLACENTAL AROMATASE DEFICIENCY

 

During fetal development, the adrenals produce substantial amounts of DHEA/S. These are either converted to 16-hydroxy-DHEA/S in the fetal liver and then transported or directly transferred to the placenta, where they are converted into estrogens (Figure 5). In case of an aromatase enzyme deficiency in the fetus (caused by variants in the CYP19A1 gene), androgen precursors accumulate and lead to virilization of 46,XX fetuses, typically leading to ambiguous genitalia at birth (93). Androgen excess due to fetal aromatase deficiency may also induce maternal virilization during pregnancy, as indicated by a deepening of the voice, oily skin and hair, and excessive hair growth (hirsutism).

 

EXCESS MATERNAL ANDROGEN PRODUCTION

 

Excessive androgen production can negatively impact fertility due to anovulation, therefore cases of maternal androgen production during pregnancy are exceedingly rare. Nonetheless, androgens originating from the mother can travers the placenta and lead to the masculinization of a female fetus. These maternal androgens usually originate from the ovaries or adrenal glands. Androgen-producing ovarian tumors include hilar cell tumors, arrhenoblastomas/androblastomas, lipoid cell tumors, and Krukenberg tumors. In contrast, pregnancy luteomas, rare non-neoplastic ovarian lesions occurring during pregnancy, are believed to be caused by hormonal effects related to pregnancy. Although exceptionally rare, androgen-secreting tumors of the adrenals can also occur during pregnancy.

 

DRUGS ADMINISTERED TO THE MOTHER DURING GESTATION

 

In 1958, Wilkins et al. reported that certain synthetic progestins administered to pregnant women, such as 17α-ethinyl-19-nortestosterone, can masculinize the external genitalia of female fetuses (94). Similarly, diethylstilbesterol (DHE), a nonsteroidal synthetic estrogen, has been linked with urogenital anomalies in both 46,XX and 46,XY fetuses (95, 96). More recently, numerous studies have indicated that fetal exposure to various endocrine disrupting chemicals (EDCs) such as pesticides, fungicides, herbicides, and plasticizers may adversely affect genital development (97-99).

 

Syndromes Associated with Ambiguous Genitalia

 

As previously mentioned in sections of 46,XX and 46,XY DSD, ambiguous genitalia can be associated with syndromes characterized by multiple congenital malformations, as summarized in Table 4. These associations arise because many of the transcription factors involved in sex development and differentiation also have roles in other aspects of development (Table 4).

 

For example, mutations of the WT-1 gene (11p3) can lead to several syndromes, including WAGR, Denys-Drash or Frasier syndromes (70-72). Mutations in SOX-9 (17q24-25) can result in campomelic dysplasia (100).

 

Mutations in DMRT1/DMRT2 (9p24.3), EMX2 (10q25.3q26.13), ATRX (Xq13.3) and WNT-4 (1p35) are often associated with developmental delay (101, 102). SF-1 (9q33) and DAX-1 (Xp21.3) play roles in the early formation of the adrenals, anterior pituitary, and parts of the hypothalamus.

 

Consequently, mutations can lead to abnormalities of these organs, in addition to those of the urogenital system. Abnormal development of the lower abdominal wall with pubic diastasis can result in urogenital anomalies observed in the VA(C)TER(L) and CHARGE syndromes, as well as bladder and cloacal exstrophy (103).

 

These examples underscore the complexities of the anomalies that may be associated with ambiguous genitalia in newborns and emphasize the importance of conducting a thorough physical examination and involving an experienced DSD team when faced with a child presenting ambiguous genitalia.

 

WORKUP OF NEWBORNS WITH AMBIGUOUS GENITALIA

 

The etiology of genital ambiguity in newborns is diverse and can have significant implications for management. Therefore, obtaining an accurate diagnosis through a medical workup is crucial. Some investigations are urgent, such as ruling out or confirming and treating potentially life-threatening adrenal insufficiency with mineralocorticoid and glucocorticoid replacement therapy in children with underlying CAH. Additionally, associated organ anomalies may require immediate attention to ensure the child’s survival, such as cardiac, pulmonary, or kidney malfunctions.

 

When the newborn is shown to be capable of excreting urine and stool without problems, and adrenal and other organ functions are found to be intact, investigations to understand genital ambiguity become primarily a psychosocial “emergency” for the family and caregivers. As a result, psychological support and holistic care by a DSD team as early as possible are essential to guide the parents and make decisions in the child’s best long-term interest. Obtaining an accurate diagnosis of the underlying cause of genital ambiguity early on is necessary for decision-making processes regarded to sex registration, hormonal and surgical treatments, and outcomes such as potential fertility and risk of gonadal malignancy. Therefore, it is recommended practice that a thorough workup should be performed for every child born with ambiguous genitalia, and guidelines for such assessments are available (22, 104-107).

 

Overall Assessment of the Child with Ambiguous Genitalia

 

Figure 6 provides an overview of the initial workup for a newborn with ambiguous genitalia. Several aspects are considered, including pregnancy and family history, as well as postnatal adaptation (such as feeding, weight gain, alertness, excretion).  In some cases, suspicion of genital ambiguity may arise during pregnancy through ultrasound surveillance. In such cases, first contact and information through a DSD team member (e.g., neonatologist, geneticist, pediatric endocrinologist) together with the DSD psychologist is recommended to guide the parents through pregnancy and alert the future care team early. Prenatal workup of ambiguous genitalia is generally not necessary if the fetus is otherwise developing normally without other organ anomalies that require attention. It is important to note that a fetus with CAH does not suffer from mineralocorticoid and cortisol deficiency in utero, as these hormones become essential only after birth (Figure 4). Genetic workup of a child with ambiguous genitalia during the prenatal period may only be recommended when there is a previous index case with a diagnosis, or when parental genetic data inform an abnormality, and when genetic counseling of the parents has led to a personalized action plan.

 

When taking the medical history of a newborn with ambiguous genitalia, asking specific questions can help guide the diagnostic workup and management. Figure 6 provides a list of important questions to consider. For example, androgenic drugs used in pregnancy may explain the virilization of a 46, XX newborns. Maternal virilization during pregnancy, for example, voice change and hirsutism may be important for the diagnoses of maternal virilizing tumors, P450 oxidoreductase (POR) deficiency, or placental aromatase deficiency. Questions about parental consanguinity are important, because many of the causes of ambiguous genitalia are recessively inherited. History of previously unexplained neonatal deaths, ambiguous genitalia, infertility or genital surgery in the family is important for identifying potential genetic etiologies.

 

In addition to the medical history, every newborn with ambiguous genitalia should undergo a comprehensive physical examination, with a special focus on signs of adrenal insufficiency (dehydration, lethargy, poor feeding) and any dysmorphic features (Figure 6). This examination may guide the diagnostic process.

 

Figure 6. Algorithm for initial workup of the child with ambiguous genitalia at birth.
Abbreviations: SDSST; Standard dose synthetic ACTH (synacthen) stimulation test, HCG; Human chorionic gonadotropin, WES; Whole exome sequencing, WGS; Whole genome sequencing, EMS; External masculinization score, EGS; External genital score, QF-PCR; quantitative fluorescence polymerase chain reaction, SRY FISH; SRY fluorescent in situ hybridization.

 

Physical Examination of the External Genital Area

 

Systematic assessment of the external genital area in newborns with ambiguous genitalia is crucial, and it should be carefully documented. Over the years, different scales and scoring systems have been used to evaluate the external genitalia. These assessments help provide a standardized way to document the physical characteristics, which is essential for comparing data over time and between different healthcare providers and patients. The use of these scales and scoring systems aids in follow-up and tracking of changes in the external genitalia.

 

Historically, the Prader or Quigley scales were commonly used for this purpose (108, 109) (Figure 7). Later, the external masculinization score (EMS) was introduced as a more comprehensive assessment tool (104) (Table 6). Nowadays, the external genital score (EGS) is recommended for evaluating and documenting the external genitalia (110) (Table 7). These tools provide a structured way to assess and record the physical findings, making it easier to monitor and communicate about the patient's condition.

 

The careful examination of the external genitalia in newborns with ambiguous genitalia provides valuable non-invasive, indirect information about prenatal exposure and response to DHT, and about the timing of this exposure. For instance, androgen exposure must have occurred in the first trimester to cause labial fusion.

 

During the genital exam, it’s important to measure the stretched phallic length, assess the quality of the corpora (erectile tissues), inspect of the labia, labio-scrotal folds, or scrotum, and document the position of the urethral opening (and vaginal opening, if applicable). Furthermore, the presence and location of palpable gonads should also be recorded. Any asymmetry in the gonads and external genitalia should be noted. Asymmetry in internal and/or external genital structures may suggest mixed gonadal dysgenesis or ovo-testicular DSD. All these observations can be documented using the external genital score (EGS), which provides a standardized way to assess and record the physical characteristics of the external genitalia (Table 7). In addition, the anogenital distance (AGD), the distance between the anus and the base of the genitalia, can be helpful in determining the extent of androgen exposure during fetal development, as a higher ratio is generally consistent with increased androgen exposure (111) (Table 8). In older infants, the anogenital index (AGI), which is the body weight standardized index of AGD [AGI/weight (mm/kg)], may be used for assessments to account for changes of the external genitalia through growth and postnatal development (111).

 

Overall, a thorough examination and documentation of the external genitalia provide important clinical information that can guide the diagnosis and management of newborns with ambiguous genitalia.

 

Figure 7. Prader and Quigley Scales for visual grading of external genital virilization. In Stage 3 a single urogenital orifice can be observed.

 

 

Table 6. External Masculinization Score (EMS)

External Masculinization Score (EMS).

Scrotal fusion

Micropenis

Urethral meatus

Right gonad

Left gonad

3

Yes

No

Normal

 

 

2

 

 

Distal

 

 

1.5

 

 

 

Labioscrotal

Labioscrotal

1

 

 

Mid

Inguinal

Inguinal

0.5

 

 

 

Abdominal

Abdominal

0

No

Yes

Proximal

Absent

Absent

An EMS score <11 out of 12 needs further clinical investigation for DSD in 46,XY.

 

Table 7. External Genital Score (EGS)

External genital score (EGS)

Labio-scrotal fusion

Genital tubercle size (mm)

Urethral meatus

Right gonad

Left gonad

3

Fused

≥31

Typical male

 

 

2.5

 

26-30

Coronal glandular

 

 

2

 

 

 Penile

 

 

1.5

Posterior fusion

21-25

Peno-scrotal

Labio-scrotal

Labio-scrotal

1

 

10-20

Scrotal

 Inguino-scrotal

Inguino-scrotal

0.5

   

Perineal

Inguinal

Inguinal

0

Unfused

≤ 10

Typical female

Impalpable

Impalpable

Median EGS (10th to 90th centile) in males < 28 weeks gestation is 10 (8.6-11.5); in males 28-32 weeks 11.5 (9.2-12); in males 33-36 weeks 11.5 (10.5-12) and in full-term males 12 (10.5-12). In all female babies, EGS is 0 (0-0).

 

Table 8. Reference Ranges for Anogenital Distance and Anogenital Index in Term Neonates

 

5th

10th

25th

50th

75th

90th

95th

Males

 

 

 

 

 

 

 

AGD (mm)

18.47

19.44

21.00

22.68

25.20

27.80

29.08

AGI (mm/kg)

5.59

5.83

6.30

6.93

7.62

8.22

8.57

Females

 

 

 

 

 

 

 

AGD (mm)

9.40

9.85

10.55

11.65

12.60

13.45

14.10

AGI (mm/kg)

3.04

3.15

3.38

3.59

3.80

4.00

4.19

Anogenital index (AGI): the body weight standardized index of AGD [AGI=AGD/weight (mm/kg)] and found that using AGI as parameter has a better correlation with age.

 

Basic Auxiliary Investigations in the First Days

 

In the first few days of life, basic investigations are essential to evaluate adrenal and gonadal development and function in otherwise healthy newborns with ambiguous genitalia. Some key aspects to address include serum electrolytes and glucose levels. They should be monitored daily as cortisol deficiency can manifest as hypoglycemia in newborns affected by CAH. Body weight and feeding characteristics should also be monitored as excessive weight loss may indicate dehydration and electrolyte disturbances.

 

KARYOTYPE

 

Besides careful clinical evaluation, first-line investigation of an individual with DSD involves confirming the chromosomal sex using quantitative fluorescence polymerase chain reaction (QF-PCR) and karyotype (22). QF-PCR detects a series of markers on the sex chromosomes and has a turn-around time of about 1–2 days. This technique has largely replaced fluorescence in situ hybridization (FISH) methods. The analysis of karyotype is a cytogenetic technique (involving G-banding) and is crucial for the initial classification of a newborn with ambiguous genitalia (Table 3). Some centers have replaced karyotyping by array-comparative genomic hybridization (aCGH) or SNP array, with faster turn-around times of 5–10 days. But these techniques will not detect structural chromosomal rearrangements and may be less effective at detecting sex chromosome mosaicism.

 

HORMONE STUDIES 

 

Detailed hormone studies may be indicated including serum gonadotropins (LH, FSH), androgens and androgen precursors (17-hydroxypregnenolone, 17-hydroxyprogesterone, androstenedione, testosterone, dihydrotestosterone), adrenal steroids and hormones (cortisol, aldosterone, and their precursors; ACTH, renin), and anti-Müllerian hormone (AMH) (1, 24).

 

Care must be taken when interpreting the results in premature babies, in whom these studies may need to be repeated at a later age and special normative value may apply. Even in term babies the timepoint and the clinical context (mode of delivery, drug effects, actual stress level etc.) need to be considered when evaluating hormonal data immediately after birth. Hormonal data change massively during delivery (e.g., surge) and within days thereafter. Additionally, it's essential to be aware that normative values for hormone levels can vary depending on the laboratory and the methods used for testing. The interpretation of hormonal data should be done by experienced healthcare providers and, if necessary, in consultation with a specialist in DSD.

 

IMAGING STUDIES

 

Ultrasound imaging of the abdominal-pelvic organs is a valuable diagnostic tool in the evaluation of newborns with ambiguous genitalia. It provides important insights into the internal reproductive and urinary structures, helping to determine the presence and characteristics of gonads, Müllerian structures (e.g., the uterus), and any associated abnormalities of the urinary tract. It can be performed at the first visit of a child with ambiguous genitalia. Of course, this investigation is not easy and may only be performed by an experienced ultra-sonographer to reveal meaningful results. Ultrasound can also detect abnormalities of the urinary tract (kidney, ureters, bladder) that sometimes occur in conjunction with genital ambiguity. Interestingly, data show that a child with a uterus and a female internal system that presents with ambiguous genitalia without gonads in the genital folds and has a 46,XX karyotype, will have in 80% of cases a diagnosis of a 46,XX CAH (most likely 21-hydroxylase deficiency). The information needed to come to this conclusion in a newborn with ambiguous genitalia can be available within 48 hours in institutions with an experienced DSD team.

 

In a second step, usually after months or years, additional imaging studies with MRI can be helpful in identifying both the type and extent of internal sex organ development in more details. An MRI may reveal further details on Müllerian structures (uterus remanent, fallopian tubes, upper portion of the vagina) than an ultrasound and can also help for localizing abdominal gonads. MRI may also be used for surveillance of the gonad at risk for cancer.

 

A genitogram had been used in the past (often early) to visualize the urinary tract, and to determine its position in relation to the vagina or vagino-utricular pouch but is no longer performed in most institutions.

 

However, in follow-up, cystoscopy and vaginoscopy may be used. These investigations are invasive procedures requiring general anesthesia, and therefore need to have a clear indication. Often, they are performed after complications such as urinary tract infections, to better understand the anatomy and its consequences, and in preparation for a surgical procedure (112).

 

Overall, the choice of imaging studies and procedures should be guided by the individual's clinical presentation, diagnosis, and the specific clinical questions that need to be addressed. In many cases, a multi-disciplinary DSD team collaborates to determine the most appropriate imaging approach to provide a comprehensive evaluation and guide treatment decisions.

 

GENETIC STUDIES

 

The rapid advancement of genetic technologies, particularly massive parallel sequencing methods like DSD panels, whole exome sequencing (WES), and whole genome sequencing (WGS), has significantly expanded our understanding of the genes involved in sex development. As a result, genetic workup has become an integral part of the recommended routine evaluation for individuals with DSD and thus ambiguous genitalia at birth (8, 22, 24, 113, 114).

 

While making a precise genetic diagnosis is still a challenge in some cases, the use of advanced genetic techniques has improved the diagnostic yield. Currently, a clear molecular-level diagnosis can be established in approximately half of individuals with 46,XY DSD who undergo genetic workup. However, as genetic technologies continue to evolve, and our understanding of sex development genes expands, it is likely that more cases, including those involving individuals with ambiguous genitalia at birth, will receive accurate and valid diagnoses. Therefore, re-review of genetically unsolved cases by a DSD team and genetic re-analysis by DSD-specialized teams is recommended to achieve a final diagnosis in as many as possible.

 

In summary, the integration of genetic workup, in combination with ongoing advancements in technology and genetic knowledge, is expected to enhance our ability to provide individuals with DSD, including those with ambiguous genitalia at birth, with more precise and comprehensive diagnoses in the future. This progress will lead to better-informed clinical decisions and management strategies for newborns with ambiguous genitalia.

 

DIFFERENTIAL DIAGNOSIS

 

Generally, the result from the karyotype plays a major role in the differential diagnosis and workup of DSD and thus ambiguous genitalia (Table 3). In most cases, the chromosomes in newborns with ambiguous genitalia will be either 46,XX or 46,XY. In rare instances, chromosomal aberration such as 45,X/46,XY or 46,XX/46,XY mosaicism may be the underlying cause explaining the phenotype. Additionally, measurements of specific hormones, such as serum luteinizing hormone (LH), follicle-stimulating hormone (FSH), sex steroids, AMH, inhibin B at birth and at follow-up, in the so-called mini-puberty of infancy period, can provide valuable information for the differential diagnosis of DSD and ambiguous genitalia.

 

In the following we provide two flowcharts and give some general information for the step-by-step workup of newborns with ambiguous genitalia and either 46,XX or 46,XY chromosomes.

 

46, XX DSD

 

A 46, XX karyotype in a newborn with ambiguous genitalia indicates that the child is a genetic female who was exposed to excessive amounts of androgens during fetal life. A flowchart for differential diagnosis is given in Figure 8.

 

Marked elevation of plasma 17-hydroxypregnenolone, 17-hydroxyprogesterone, 21-deoxycortisol and androstenedione, along with male-typical levels of testosterone, are characteristic of 21-hydroxylase deficiency (CYP21A2). High values of corticosterone and 11-deoxycortisol, along with elevated androgens, indicate 11β-hydroxylase deficiency (biallelic mutations in CYP11B1). In 3beta-HSD deficiency (biallelic mutations in HSD3B2), calculation of ratios of delta-5 (pregnenolone, 17OH-pregnenolone, DHEA) over delta-4 (progesterone, 17OH-progesterone, androstenedione) steroid metabolites (see Figure 4) will point towards the genetic defect.

 

Overall, comprehensive steroid profiling from blood or urine using chromatographic, mass spectrometric methods allow the diagnosis of most steroidogenic defects causing androgen excess CAH in 46,XX newborns (115-117) (see also Table 5). In addition, they have led to the description of novel alternate pathways to produce active androgens in the intermediate metabolism through the backdoor or 11-oxygenated pathways, especially in disorders such as 21-hydroxylase or POR deficiencies, thereby enhancing our understanding and diagnostic possibilities (115, 117, 118)(Figure 4). However, these methods are not (yet) widely available, and the interpretation of these data is not trivial. In addition, instruments and methods vary from lab to lab. Thus, method validation is tedious and comparison between labs difficult so far (119-121).

 

When excess maternal androgen production is the underlying cause for masculinization of a female fetus, the source of these steroids is eliminated postnatally. Thus, the various steroids studied in affected newborns will be in the female-typical range despite masculinization of the external genitalia. Then, detailed examination of the mother may provide the explanation.

 

Likewise, in cases of mild POR deficiency or aromatase deficiency, in which the feto-placental unit was not able to metabolize the fetal androgens and led to ambiguous genitalia, the problem will not (always) be visible in the newborn’s steroid metabolome, and the diagnosis may only be made by genetic testing. If not, only in later life, when aromatase activity is needed for estrogen production in puberty, or when ovarian cysts hint an underlying problem, the diagnosis may be made (Figure 8).

 

In individuals with ovo-testicular DSD and 46,XX chromosomes, masculinization arises from androgens secreted by the testicular portion of the differentiated gonads. Androgen production is similar to that produced by testes in unaffected males except that the amount is usually smaller. The degree of masculinization of the genitalia is thus linked to the amount of functioning testicular tissue, which can be determined by a serum AMH measurement (Table 9) (122).

 

Very low values of AMH and female-typical values for LH/FSH (Tables 9 and 10) are expected in 46,XX newborns with masculinized/virilized genitalia that is attributed to CAH or excess maternal androgen production during gestation (122, 123). AMH is higher in newborns with ovo-testicular DSD, due to Sertoli cell development in the testicular portion of their gonads where AMH is mainly produced.

 

On occasion, translocation of the pseudo-autosomal part of the Y chromosome along with a mutated SRY gene to an X chromosome occurs. The result is partial masculinization of the genitalia in a 46,XX newborn. With maturity, the phenotype of affected individuals closely resembles that of boys and men with Klinefelter syndrome.

 

Figure 8. 46,XX DSD decision tree diagram.
Abbreviations: USG: Ultrasound; 17OHP: 17OH-Progesterone; DSD: Differences in sex development; CAH: Congenital adrenal hyperplasia; 21OHD: 21α-Hydroxylase deficiency; 11β-OHD: 11β-Hydroxylase deficiency; 3β-HSD2: 3β-Hydroxysteroid dehydrogenase type 2; PORD: P450 oxidoreductase deficiency.

 

 

Table 9. Normal Values for AMH (ng/mL)* in Fetuses, Newborns and Infants Under 1 Year (122)

 

Female

Male

 

n

Median

5-95th percentile

n

Median

5-95th percentile

Fetal blood

25

<0.4

<0.4-0.5

23

44.4

13.4-86.2

Cord blood

53

<0.4

<0.4-1.1

48

30.7

7.4-610.7

1-30 days

70

<0.4

<0.4-1.3

73

57.2

23.8-124.0

31-120 days

31

0.8

<0.4-6.4

39

88.4

46.8-173.0

4-12 months

13

1.0

<0.4-2.7

53

124.7

67.4-197.0

* AMH/MIS enzyme immunoassay kit cat 2368, Immunotech-Beckman Coulter, France

 

Table 10. LH and FSH Values (means ± SD) From Female and Male Newborns. Modified from (123)

 

 

Female

Male

Age (days)

Hormone

n

Median

Mean±SD

n

Median

Mean±SD

1-5

FSH (IU/L)

31

1.80

2.00 ± 1.37

30

0.85

0.96 ± 0.60

LH (IU/L)

0.20

0.48 ± 0.66

0.20

0.39 ± 0.48

6-10

FSH (IU/L)

17

1.40

2.44 ± 2.52

15

1.40

2.91 ± 4.38

LH (IU/L)

0.30

0.45 ± 0.33

0.30

0.45 ± 0.33

11-15

FSH (IU/L)

8

8.95

8.16 ± 4.27

17

3.00

3.71 ± 2.69

LH (IU/L)

1.60

1.58 ± 1.28

2.90

3.55 ± 2.84

16-20

FSH (IU/L)

6

1.90

1.62 ± 1.05

14

2.15

2.63 ± 1.45

LH (IU/L)

0.35

1.03 ± 1.39

3.65

4.13 ± 2.76

21-25

FSH (IU/L)

3

3.90

7.07 ± 5.92

7

2.10

2.50 ± 1.51

LH (IU/L)

0.50

0.46 ± 0.25

2.70

2.86 ± 1.51

26-28

FSH (IU/L)

8

6.15

9.74 ± 9.89

8

2.40

2.25 ± 0.81

LH (IU/L)

2.80

2.75 ± 2.39

1.40

2.22 ± 2.37

Abbreviations: LH, luteinizing hormone; FSH, follicle-stimulating hormone.

 

46, XY DSD

 

A 46, XY karyotype reveals that one is dealing with a genetic male who was under-masculinized during fetal development. A flowchart for differential diagnosis is given in Figure 9.

 

Laboratory findings of normal or elevated testosterone and DHT with normal AMH (Figure 9) indicate a diagnosis of AIS. If testosterone levels are normal but DHT levels are low, a diagnosis of steroid 5α-reductase deficiency can be made. Low levels of testosterone and DHT, along with marked elevation of some androgen precursors, indicate a deficiency of one of the enzymes required for androgen biosynthesis (Figure 9). For example, if the elevated precursors include androstenedione and 17-hydroxyprogesterone, then the defective enzyme is 17-ketosteroid reductase (HSD17B3). In all cases of testosterone biosynthetic defects, AMH levels are similar to those observed in healthy 46,XY male infants with male-typical external genitalia (Figure 9). With early defects of steroidogenesis that affect both adrenal and gonadal function, it is though important to evaluate adrenal function, as it might be impaired with deficiencies of STAR, CYP11A1, HSD3B2, CYP17A1 and POR (Table 5, Figure 9).

 

Finally, when all androgens and their precursors are below normal levels, one is dealing with gonadal dysgenesis or 46,XY ovo-testicular DSD. In these cases, AMH values should also be low. In contrast, for babies affected by Leydig cell hypoplasia, androgens and their precursors are low, while AMH values should be in the normal male range.

 

Figure 9. 46,XY DSD decision tree diagram.
Abbreviations: USG: Ultrasound; 17OHP: 17OH-Progesterone; DSD: Differences in sex development; AMH: Anti-Müllerian hormone; T: Testosterone; N: Normal; PMDS: Persistent Müllerian duct syndrome; AIS: Androgen insensitivity syndrome; SRD5A2: 5α-reductase A2; HCG: Human chorionic gonadotropin; DHT: Dihydrotestosterone; 3β-HSD2: 3β-Hydroxysteroid dehydrogenase type 2; CYP17A1: 17α-Hydroxylase/17-20 lyase; POR: P450 oxidoreductase; StAR: Steroidogenic acute regulatory protein; CYP11A1; Cholesterol side-chain cleavage enzyme; 17β-HSD3: 17β-Hydroxysteroid dehydrogenase type 3; AKR1C2/4: Aldo-keto reductase family 1 member C2/4; CYB5: Cytochrome b5.

 

Testing Early Postnatal Gonadal Function and Mini-Puberty

 

In cases of ambiguous genitalia where a diagnosis may not be reached through first line investigations, and where a gonadal malfunction is suspected, functional testing and/or re-analysis of gonadal function at the timepoint of mini-puberty may be helpful, especially also with respect to having information when making considerations towards sex of registration (e.g., in utero effect of sex hormones on the brain) and prediction of later pubertal development and fertility potential.

 

Mini-puberty is the time window at day 30-100 after birth, when the hypothalamic-pituitary-gonadal (HPG) axis is re-activated for a short time frame in infancy, before going to inactivity until puberty. Little is known about the purpose of mini-puberty, but as the HPG axis is active and stimulates the gonads, it is a time window of opportunity to assess gonadal function without the necessity to stimulate with human chorionic gonadotrophin (hCG).

 

As a principle, in mini-puberty, the typical ovaries of a 46,XX newborn show little functional activity, while the typical testes of a 46,XY newborn are quite (re)active in testosterone (T), DHT, AMH and inhibin B production. While androgens are produced in the Leydig cells of the testes, AMH and inhibin B originate from the Sertoli cells. Table 11 gives an overview of the hormonal values observed during normal mini-puberty in healthy 46,XX and 46,XY infants (124).

 

Table 11. Evaluation of Reproductive Hormones During Mini-Puberty in Infants. Normative Values of Different Serum Hormones in 46,XX Female and 46,XY Male Infants. Modified from  (124)

 

 

Females

Males

 

Post-natal time (months)

n

GMean

2.5-97.5th percentile

n

GMean

2.5-97.5th percentile

LH (IU/L)

2-3.5

432

<LOD

<LOD-0.98

581

1.71

0.62-4.08

3.5-5

110

<LOD

<LOD-1.25

166

1.40

0.54-3.32

FSH (IU/L)

2-3.5

435

3.98

1.23-17.4

578

1.19

0.41- 3.02

3.5-5

111

3.93

1.30-17.7

165

1.11

0.42-2.68

Testosterone (RIA,nmol/L)

2-3.5

74

<LOD

<LOD-0.40

592

3.04

0.69-7.60

3.5-5

14

<LOD

<LOD-NA

168

1.97

<LOD-7.01

Testosterone

(LC-MS/MS, nmol/L)

2-3.5

165

<LOD

<LOD-0.21

251

4.75

1.35- 11.3

3.5-5

125

<LOD

<LOD-0.17

175

2.25

0.32- 9.65

Estradiol (pmol/L)

2-3.5

455

29

<LOD-79

571

<LOD

<LOD-47

3.5-5

113

31

<LOD-98

160

<LOD

<LOD-49

SHBG (nmol/L)

2-3.5

427

133

67-264

579

135

66-268

3.5-5

110

141

72-282

162

143

71-262

Inhibin B (pg/mL)

2-3.5

423

62

<LOD-184

571

379

229-631

3.5-5

106

67

<LOD-174

158

379

222-662

AMH (pmol/L)

2-3.5

339

11

<LOD-49

48

1013

425- 1810

3.5-5

67

15

2-46

12

1183

797-NA

Abbreviations: LOD, Limit of detection; GMean, geometric mean; LH, luteinizing hormone; FSH, follicle-stimulating hormone; T, testosterone; RIA, radioimmunoassay; SHBG, sex-hormone binding globulin; AMH, anti-Müllerian hormone; LC-MS/MS, liquid chromatography-tandem mass spectrometry; NA, not available.

 

HCG Testing of Gonadal Function in a DSD Newborn

 

The human chorionic gonadotrophin (hCG) test can assess whether in a newborn with ambiguous genitalia, functioning Leydig cells are present. The test may therefore be useful for diagnosing testosterone biosynthesis defects such as HSD17B3 or SRD5A2 deficiency (Table 12). Several protocols for the hCG test exist, but all baseline serum samples are taken for T, DHT and precursors. Then, one to three intramuscular injections of high dose hCG (500-1500 IU) are given at 24 hours interval, and finally repeat stimulated samples are taken for androgen measurements after 72 or 24 hours, respectively, after the last injection. The following Table 12 shows how results of the test may be interpreted in various DSD conditions that may lead to ambiguous genitalia (125, 126).  

 

Table 12. Interpretation of hCG Stimulation Test Results (125, 126)

Indication

Consideration

Abnormal result

Testosterone biosynthesis defects

T before and after hCG administration

T increase < 100-150 ng/dL (3.5-5 nmol/L)

5α-Reductase deficiency

T/DHT ratio after hCG administration

≥ 8.5 for minipuberty,

≥ 10 for prepuberty,

≥ 17 for puberty,

> 20 in genetically proven cases using LC-MS/MS

17β-HSD3 deficiency

T/A4 ratio after hCG administration

< 0.8

AIS

T, SHBG before and after hCG administration

T increases but SHBG does not change significantly

Abbreviations: T, testosterone; DHT, dihydrotestosterone; HCG, human chorionic gonadotropin; A4, androstenedione; SHBG, sex-hormone binding globulin; AMH, anti-Müllerian hormone; LC-MS/MS, liquid chromatography-tandem mass spectrometry; 17β-HSD3 deficiency, 17β-hydroxysteroid dehydrogenase type 3 deficiency; AIS, androgen insensitivity syndrome

 

RISK OF MALIGNANCY OF THE DSD GONAD

 

Individuals with certain disorders of sex development (DSD), especially those involving Y chromosome material, have an increased risk of developing gonadal tumors. These tumors are mainly gonadal germ cell tumors (GGCTs), which occur in about 15 % of all DSD cases, with the risk varying depending on the age and specific underlying diagnosis. Gonadal stromal tumors and smooth muscle leiomyomas/hamartomas are less common and are found in only about 0.9% of DSD cases (127). Risk factors for development of gonadal malignancy in DSD are shown in Figure 10. Several factors contribute to the increased risk of gonadal malignancy in individuals with DSD, including genetic factors, the presence of Y chromosome material, and the type of DSD. Therefore, ongoing monitoring and appropriate management are essential to minimize this risk. While gonadectomy (surgical removal of the gonads) is not recommended for every case of 46,XY DSD, it's crucial to consider the risk of malignant tumors in the decision-making process (1, 69). The consensus statement on the management of DSD provides guidelines and recommendations regarding the timing of gonadectomy for specific patients. These recommendations aim to balance the potential benefits of gonadectomy with the associated risks and individual patient needs.

 

In individuals with DSDs, germ cells typically undergo a process of apoptosis (programmed cell death) as the body grows and develops. This process eliminates germ cells that are not needed for reproduction. However, in some cases, certain germ cells may not undergo the normal maturation process and remain in an immature state. These immature germ cells can continue to express embryonic markers such as octamer-binding transcription factor 3/4 (OCT3/4) encoded by POU5F1; stem cell factor (SCF), also known as c-KIT ligand (encoded by KITLG); and placental alkaline phosphatase (PLAP). These cells are susceptible to developing into cancerous lesions, starting as pre-invasive lesions (gonado-blastoma) or as precursors to invasive GGCT (dysgerminoma) (Figure 10).

 

DSD patients who have Y chromosome material in their gonadal karyotype are at risk of GGCTs regardless of the degree of testicular differentiation. Y chromosome material associated with gonadal tumors is GBY locus which is around the centromere of the Y chromosome and includes the gene for ¨testis-specific protein on the Y chromosome¨ (TSPY, located in Yp11.2). Other potentially related genes are SRY and DYZ3.

 

The risk of developing gonadal tumors in gonadal dysgenesis may also vary according to the underlying molecular etiology. Patients with genetic defects that lead to early blockage of gonadal development such as mutation or deletions of SRY or WT1 have a higher risk. The risk of developing gonadal tumors ranges from 40% to 60% in gonadal dysgenesis due to WT1 gene mutations (Frasier syndrome and Denys-Drash syndrome). Early bilateral gonadectomy is therefore recommended. Similarly, early bilateral gonadectomy is recommended in patients with complete or partial gonadal dysgenesis due to SRY gene mutations due to the 20% to 52.5% risk of gonadal tumor development. Gonadal dysgenesis due to MAP3K1 gene mutations is another indication for early gonadectomy (127). Gonadal tumor risk is also present, though somewhat lower in gonadal dysgenesis due to 45,X0/46,XY mosaicism. The exact time of malignancy development and the prevalence of tumors in other molecular etiologies require further research for a comprehensive understanding.

 

Histologically the risk of gonadal tumor development increases with gonadal immaturity. Fully developed gonads such as the testes, ovary or ovotestis have lower risk of malignancy than streak or dysgenetic testes (Figure 10). In ovo-testicular DSD, the risk of germ cell tumors is low (3%) (1). Newborns with PAIS are at high risk (50%) for developing gonadal tumors, and bilateral orchiopexy or gonadectomy is recommended at the time of diagnosis, if the testes are located in the abdomen (1, 128).

 

The risk of developing germ cell malignancy in newborns with CYP11A1, HSD3B2 or CYP17A1 deficiency is unknown. Newborns with 17-ketosteroid reductase deficiency have a fairly low risk for malignancy (1). Tumor risk in newborns with Leydig cell hypoplasia is unclear, and thus the recommendation for gonadectomy is not well established (1, 25, 129, 130).

 

Overall, it remains challenging to predict the personal risk of gonadal neoplasia (mostly GGCT) for a DSD individual, because it depends on many other factors besides genetics. Stimulating gonadotropins and gonadal hormones during mini-puberty and after puberty onset are also regarded as risk factors increasing gonadal tumor risk in patients with DSD. Similarly, gonads located intra-abdominal carry a higher tumor risk. The management of gonadal tumor risk in individuals with DSD is a complex issue that requires careful consideration and a personalized approach, taking into account the specific DSD subtype, genetic factors, and patient preferences. Regular monitoring and discussions with a healthcare team experienced in DSD management are essential to make informed decisions about gonadectomy. If gonads are located in the scrotum, then periodic examination of the testes may be advised as an alternative to gonadectomy, while an intra-abdominal gonad at risk may only be monitored by imaging methods such as ultrasound or MRI.

 

Figure 10. Risk of malignancy development in the DSD gonad.
Abbreviations: (OCT3/4); Octamer-binding transcription factor 3/4, TSPY; testis-specific protein on the Y chromosome, c-KITL; c-KIT ligand also called stem cell factor (SCF).

 

SEX REGISTRATION OF A NEWBORN

 

Assigning or registering the sex of a newborn with atypical external genitalia is a complex and important decision that requires careful consideration, consultation, and a patient-centered approach. The parents and DSD network team should not rush but take the time needed to come to a consensus in the best interest of the child in its environment. Not knowing whether the newborn is a girl or boy causes mostly a lot of distress and one is tempted to call it an emergency, but if there are no medical reasons to make it an emergency (like e.g., suspected adrenal insufficiency or additional organ anomalies), it is important to declare it accordingly and inform that the newborn is not at risk and actually does not care whether it is a male or female or intersex person at this moment. Thus, the stress of ambiguity of the newborn is actually at this moment “only” an issue of the parents and the care team who mostly wish to know ASAP whether the child can be assigned/registered a “correct” sex that might also fit later in life.

 

To consider sex registration, not only is it important to take into account the underlying cause and knowledge on the possible outcome of the specific, often unique condition, but also the family, cultural, and societal preconditions and aspects. Most societies have a binary sex registration obligation by legislation, but some societies have more recently introduced a third sex category besides male and female. Although this seems to make things easier, it has been discussed that such categorization in a third or other or unclassified group might also stigmatize a person in a society that is mostly binary. Thus, in real life practice, there is rumor that a third sex category is rarely chosen for a newborn with ambiguous genitalia by the parents and the DSD team. Overall, a personalized approach for every newborn with ambiguous genitalia involving the parents and the DSD network team is recommended following the shared-decision-making process (12, 13).

 

Most important is that the team around the newborn with ambiguous genitalia works together in the best interest of the child with the latest knowledge of the literature and expertise when taking the decision of sex registration, and still remains humble and open minded for the child’s future that may follow a different path leading to sex reassignment later in life. Therefore, continuous support of the family and the child by a DSD psychologist is extremely helpful to recognize early signs of gender dysphoria, inform the child about the condition in an age-appropriate manner and follow its will, if case sex reassignment is desired.

 

Generally, people believe that the sex chromosomes indicate a person’s “true sex” and  laws exist supporting this idea, to the detriment of some individuals affected by DSD (131). Scientifically speaking, it is clear that the majority of genes on the X chromosome do not influence sex development and differentiation, although the AR gene is necessary for phenotypic masculinization. Concerning the Y chromosome, only the SRY gene contributes to testicular formation. In fact, most of the genes required for sex development and differentiation are found on the autosomes (1). Therefore, chromosomes do not dictate the sex of rearing in newborns affected by a DSD and are only one piece of a big puzzle (Figure 3).

 

Although there is no overall recommendation for sex registration at birth for a child with ambiguous genitalia, some helpful information is available from larger patients’ cohorts with a specific DSD diagnosis in the literature.

 

Newborns presenting with 46,XY chromosomes and female external genitalia at birth (thus actually not with ambiguous genitalia) due to complete androgen insensitivity syndromes (CAIS), complete gonadal dysgenesis (Swyer syndrome), or other complete loss of testosterone biosynthesis live successfully when assigned a female sex of rearing. Female assignment in such cases is widely accepted by patients throughout their lives despite challenges that they may experience regarding sexual dysfunction and infertility (1, 132-134).

 

For newborns with ambiguous genitalia affected by partial gonadal dysgenesis, no general recommendation can be made. Fewer procedures were historically required for surgical feminization compared to surgical masculinization of the genitalia. However, the functional outcome in individuals who received feminizing or masculinizing procedures is less than optimal (135, 136). Additionally, rates of satisfaction with sex of rearing are similar for affected individuals whether raised male or female (1, 20, 137). Thus, the decision should not be made on the basis of “surgical possibilities”, and overall, such surgical procedures should be executed with reservation. Clearly, further studies are needed to elucidate why some individuals with partial gonadal dysgenesis experience gender dysphoria, while others do not.

 

Likewise, no general recommendation can be made for patients with ambiguous genitalia due to a partial androgen biosynthesis defect, (e.g., partial 3βHSD2, CYP17A1, POR, StAR, CYP11A1 deficiencies). But some studies suggest that the degree of masculinization of the external genitalia may reflect the degree of virilization of the brain and thus the gender preference of this person (138). This might be true for 46,XY newborns with ambiguous genitalia that are manifesting with severe hypospadias, but not without exceptions. For patients with variants in the SRD5A2 or HSD17B3genes recognized with ambiguous genitalia at birth (or even missed because manifesting with a female-typical external genitalia), there is quite a strong body of data recommending male sex registration at birth as this is the preferred sex of these persons when asked in later life (139, 140). Normal virilization at puberty, coupled with intact fertility potential, are strong factors for this recommendation at birth. If registered as female at birth and diagnosed later, gender dysphoria and desire of sex reassignment later in life is very likely.

 

Similar to partial gonadal dysgenesis, consensus regarding an optimal sex of registration for newborns affected by PAIS does not exist. Although earlier data showed that fewer procedures were usually required for surgical feminization, compared to surgical masculinization, in a person with ambiguous external genitalia in the past, when larger series of genital surgery were analyzed, it is unclear if the functional outcome was better among newborns who received feminizing procedures compared to those who received masculinizing procedures. Rates of satisfaction with sex of rearing are similar for individuals with PAIS raised female or male (132, 141). While reports exist of impaired sexual function in people with PAIS raised male, it is highly suspected that sexual functional outcomes in affected people raised female are similarly poor (137). Reports of sexual satisfaction for people with PAIS who have not received genitoplasty in infancy, or at a later age, are limited. However, more information on outcome of newborns with ambiguous genitalia who had no genital surgery, at least not in infancy and childhood, will hopefully soon be added to the medical literature as current developments recommend abstaining from genital surgery.

 

By contrast, in 46,XX newborns with ambiguous genitalia due to CAH, it is generally recommended to register them a priori in the female sex, even if they are severely virilized (142-144). Masculinization due to 21-hydroxylase deficiency does not impair the development of the ovaries or the Müllerian ducts and thus fertility potential. Limited information is available about the medical and psychosexual outcome of 46,XX newborns affected by 21-hydroxylase deficiency and raised male (145, 146), although these patients do tend to have an increased risk of gender dysphoria compared to their female counterparts (147). Thus, a female sex of rearing is typically the decision for 46,XX newborns affected by 21-hydroxylase deficiency (145). Female sex of rearing in 46,XX newborns with ambiguous genitalia is also recommended for conditions resulting from maternal overexposure to androgens and placental aromatase deficiency.

 

FINAL REMARKS TO OVERALL MANAGEMENT

 

Only recently has the understanding of, and reaction to, having a child with ambiguous genitalia received systematic study. For some parents and caregivers, feelings of isolation and concern over what the future may hold for their affected child in terms of stigmatization and sexual dysfunction are paramount (148). One potential approach to ameliorate this issue is to provide information and management options to the family early in pregnancy or as soon as the child is born. Guidance by a DSD network team is therefore extremely important.

 

Some research has been done to examine the effects of prenatal diagnosis and treatment. In females with 21-hydroxylase deficiency CAH, one of the most common causes of ambiguous genitalia, dexamethasone treatment of the pregnant mother has been reported to reduce virilization of an affected female fetus by 80-85% (149), but this approach is no longer offered or recommended in guidelines because of potential adverse effects of dexamethasone on the developing fetus. Before the discovery of methods to measure fetal DNA in maternal blood, which enabled specific diagnosis of sex and whether the fetus inherited and was affected by the mutation, controversies existed because 7 unaffected fetuses would be treated unnecessarily (1:4 chance of inherited mutation and 1:2 chance of male vs female), So, on the one hand 7 unaffected fetuses and the mother would be exposed to an unnecessary drug, while on the other hand, even in affected fetuses long-term outcome showed adverse effects on neurocognitive parameters (150). Today, noninvasive prenatal diagnosis using massive parallel sequencing of cell-free fetal DNA can be used to determine a specific diagnosis, such as CAH, as early as 6 weeks of gestation (149, 151). Even preimplantation diagnostics is available (152). These techniques could be used to look at other autosomal dominant or X-linked conditions that cause ambiguous genitalia, especially when the fetus is known to be at risk (151). Further studies need to be done on this subject, but for families who already have an affected child or know that they are carriers, this may be an option that needs to be discussed with a DSD geneticist.

 

Patients for whom prenatal diagnosis and/or treatment is not an option, prognosis and management has improved significantly over time, but many open questions and controversies still exist. Our studies of parents of children with DSD reveal that the appearance of atypical genitalia can result in significant stress and maladaptive parenting strategies. For example, mothers of children with ambiguous genitalia experience greater stress if their child has not received “corrective” genital surgery (153) and some parents believe such surgeries eliminate stigmatization that may arise due to their child’s ambiguous genitalia However, as their children mature past infancy, some parents realize that their child’s DSD has not been ameliorated by genitoplasty, and concerns for their son or daughter resurface (148). To illustrate, among parents studied by our group, caregivers of adolescents with DSD experienced increased stress as their child matured despite the fact that many had received earlier surgery (154). Additionally, parents of children reared male reported the most perceived child vulnerability for their sons (155),and their reported levels of depression were directly associated with more atypical genital appearance for their sons (156). Overall, parents report that while early genitoplasty seems to “fix” some of their concerns for their child with DSD, this “fix” is not long-lasting. Instead, what needs to be emphasized to families is how to support their child who is different, but not damaged. Family centered, interdisciplinary care, with open and clear communication between patients, parents, and caregivers has been shown to be essential for optimal quality of life (157). In addition, networks of families who have the personal experience of parenting children with ambiguous genitalia can be an invaluable resource (158). Reservation towards early interventions needs to be considered,keeping in mind to preserve the personal right of the child to take his/her own decision towards body integrity. Any intervention that can be delayed without harming the newborn with ambiguous genitalia should be postponed until personal consent can be given. Finally, referring parents to support groups and introducing them to other caregivers of children born with ambiguous genitalia is extremely important to optimize parents’ understanding and acceptance of their child’s condition. With increased understanding and acceptance, optimal growth and development for children born with ambiguous genitalia may be obtained.

 

In conclusion, this Chapter give some insight into the current diagnostic evaluation and care of the newborn with ambiguous genitalia. We underscore the importance of a multidisciplinary and compassionate approach to the care of newborns with ambiguous genitalia, acknowledging the complex medical, psychological, and emotional aspects involved in such cases. We highlight the critical role of specialized teams and support in ensuring the well-being of both the child and his/her family.

 

REFERENCES

 

  1. Hughes IA, Houk C, Ahmed SF, Lee PA. Consensus statement on management of intersex disorders. Arch Dis Child. 2006;91(7):554-63.
  2. Halil H, Oguz SS. Establishment of normative data for stretched penile length in Turkish preterm and term newborns. Turk J Pediatr. 2017;59(3):269-73.
  3. Holmes NM, Miller WL, Baskin LS. Lack of defects in androgen production in children with hypospadias. J Clin Endocrinol Metab. 2004;89(6):2811-6.
  4. Alaei M, Rohani F, Norouzi E, Hematian Boroujeni N, Tafreshi RI, Salehiniya H, Soheilipour F. The Nomogram of Clitoral Length and Width in Iranian Term and Preterm Neonates. Front Endocrinol (Lausanne). 2020;11:297.
  5. Cools M, Nordenström A, Robeva R, Hall J, Westerveld P, Flück C, et al. Caring for individuals with a difference of sex development (DSD): a Consensus Statement. Nat Rev Endocrinol. 2018;14(7):415-29.
  6. Gorduza DB, Quigley CA, Caldamone AA, Mouriquand PDE. Surgery of Anomalies of Gonadal and Genital Development in the "Post-Truth Era". Urol Clin North Am. 2018;45(4):659-69.
  7. McElreavey K, Bashamboo A. Monogenic Forms of DSD: An Update. Horm Res Paediatr. 2023;96(2):144-68.
  8. O'Connell MA, Atlas G, Ayers K, Sinclair A. Establishing a Molecular Genetic Diagnosis in Children with Differences of Sex Development: A Clinical Approach. Horm Res Paediatr. 2023;96(2):128-43.
  9. Brown J, Warne G. Practical management of the intersex infant. J Pediatr Endocrinol Metab. 2005;18(1):3-23.
  10. Dayner JE, Lee PA, Houk CP. Medical treatment of intersex: parental perspectives. J Urol. 2004;172(4 Pt 2):1762-5; discussion 5.
  11. Lightfoot S, Carley M, Brinkman W, Gardner MD, Gruppen LD, Liang N, et al. Co-creating a suite of patient decision aids for parents of an infant or young child with differences of sex development: A methods roadmap. Front Urol. 2023;2.
  12. Légaré F, Witteman HO. Shared decision making: examining key elements and barriers to adoption into routine clinical practice. Health Aff (Millwood). 2013;32(2):276-84.
  13. Sandberg DE, Gardner M, Kopec K, Urbanski M, Callens N, Keegan CE, et al. Development of a decision support tool in pediatric Differences/Disorders of Sex Development. Semin Pediatr Surg. 2019;28(5):150838.
  14. Martinez de LaPiscina I, Fluck CE. Genetics of human sexual development and related disorders. Curr Opin Pediatr. 2021;33(6):556-63.
  15. Siiteri PK, Wilson JD. Testosterone formation and metabolism during male sexual differentiation in the human embryo. J Clin Endocrinol Metab. 1974;38(1):113-25.
  16. Wilson JD, Griffin JE, Russell DW. Steroid 5 alpha-reductase 2 deficiency. Endocr Rev. 1993;14(5):577-93.
  17. Güran T. Latest Insights on the Etiology and Management of Primary Adrenal Insufficiency in Children. J Clin Res Pediatr Endocrinol. 2017;9(Suppl 2):9-22.
  18. Ali SR, Lucas-Herald A, Bryce J, Ahmed SF. The Role of International Databases in Understanding the Aetiology and Consequences of Differences/Disorders of Sex Development. Int J Mol Sci. 2019;20(18).
  19. Wiesemann C, Ude-Koeller S, Sinnecker GH, Thyen U. Ethical principles and recommendations for the medical management of differences of sex development (DSD)/intersex in children and adolescents. Eur J Pediatr. 2010;169(6):671-9.
  20. Lee PA, Nordenström A, Houk CP, Ahmed SF, Auchus R, Baratz A, et al. Global Disorders of Sex Development Update since 2006: Perceptions, Approach and Care. Horm Res Paediatr. 2016;85(3):158-80.
  21. Krishnan S, Wisniewski AB. Ambiguous genitalia in newborns. Genetic Steroid Disorders. 2014:87-97.
  22. Audi L, Ahmed SF, Krone N, Cools M, McElreavey K, Holterhus PM, et al. GENETICS IN ENDOCRINOLOGY: Approaches to molecular genetic diagnosis in the management of differences/disorders of sex development (DSD): position paper of EU COST Action BM 1303 ‘DSDnet’. Eur J Endocrinol. 2018;179(4):R197-r206.
  23. Pyle LC, Nathanson KL. A practical guide for evaluating gonadal germ cell tumor predisposition in differences of sex development. Am J Med Genet C Semin Med Genet. 2017;175(2):304-14.
  24. Yatsenko SA, Witchel SF. Genetic approach to ambiguous genitalia and disorders of sex development: What clinicians need to know. Semin Perinatol. 2017;41(4):232-43.
  25. Zenteno JC, Canto P, Kofman-Alfaro S, Mendez JP. Evidence for genetic heterogeneity in male pseudohermaphroditism due to Leydig cell hypoplasia. J Clin Endocrinol Metab. 1999;84(10):3803-6.
  26. Ogata T, Matsuo N, Hiraoka N, Hata JI. X-linked lissencephaly with ambiguous genitalia: delineation of further case. Am J Med Genet. 2000;94(2):174-6.
  27. Reardon W, Gibbons RJ, Winter RM, Baraitser M. Male pseudohermaphroditism in sibs with the alpha-thalassemia/mental retardation (ATR-X) syndrome. Am J Med Genet. 1995;55(3):285-7.
  28. Biason-Lauber A, Konrad D, Meyer M, DeBeaufort C, Schoenle EJ. Ovaries and female phenotype in a girl with 46,XY karyotype and mutations in the CBX2 gene. Am J Hum Genet. 2009;84(5):658-63.
  29. Umehara F, Tate G, Itoh K, Yamaguchi N, Douchi T, Mitsuya T, Osame M. A novel mutation of desert hedgehog in a patient with 46,XY partial gonadal dysgenesis accompanied by minifascicular neuropathy. Am J Hum Genet. 2000;67(5):1302-5.
  30. Quinonez SC, Park JM, Rabah R, Owens KM, Yashar BM, Glover TW, Keegan CE. 9p partial monosomy and disorders of sex development: review and postulation of a pathogenetic mechanism. Am J Med Genet A. 2013;161a(8):1882-96.
  31. da Silva TE, Gomes NL, Lerário AM, Keegan CE, Nishi MY, Carvalho FM, et al. Genetic Evidence of the Association of DEAH-Box Helicase 37 Defects With 46,XY Gonadal Dysgenesis Spectrum. J Clin Endocrinol Metab. 2019;104(12):5923-34.
  32. Pellegrini M, Pantano S, Lucchini F, Fumi M, Forabosco A. Emx2 developmental expression in the primordia of the reproductive and excretory systems. Anat Embryol (Berl). 1997;196(6):427-33.
  33. Lang-Muritano M, Sproll P, Wyss S, Kolly A, Hürlimann R, Konrad D, Biason-Lauber A. Early-Onset Complete Ovarian Failure and Lack of Puberty in a Woman With Mutated Estrogen Receptor β (ESR2). J Clin Endocrinol Metab. 2018;103(10):3748-56.
  34. Bagheri-Fam S, Ono M, Li L, Zhao L, Ryan J, Lai R, et al. FGFR2 mutation in 46,XY sex reversal with craniosynostosis. Hum Mol Genet. 2015;24(23):6699-710.
  35. Lourenço D, Brauner R, Rybczynska M, Nihoul-Fékété C, McElreavey K, Bashamboo A. Loss-of-function mutation in GATA4 causes anomalies of human testicular development. Proc Natl Acad Sci U S A. 2011;108(4):1597-602.
  36. Callier P, Calvel P, Matevossian A, Makrythanasis P, Bernard P, Kurosaka H, et al. Loss of function mutation in the palmitoyl-transferase HHAT leads to syndromic 46,XY disorder of sex development by impeding Hedgehog protein palmitoylation and signaling. PLoS Genet. 2014;10(5):e1004340.
  37. Kunitomo M, Khokhar A, Kresge C, Edobor-Osula F, Pletcher BA. 46,XY DSD and limb abnormalities in a female with a de novo LHX9 missense mutation. Am J Med Genet A. 2020;182(12):2887-90.
  38. Fukami M, Wada Y, Miyabayashi K, Nishino I, Hasegawa T, Nordenskjöld A, et al. CXorf6 is a causative gene for hypospadias. Nat Genet. 2006;38(12):1369-71.
  39. Pearlman A, Loke J, Le Caignec C, White S, Chin L, Friedman A, et al. Mutations in MAP3K1 cause 46,XY disorders of sex development and implicate a common signal transduction pathway in human testis determination. Am J Hum Genet. 2010;87(6):898-904.
  40. Bardoni B, Zanaria E, Guioli S, Floridia G, Worley KC, Tonini G, et al. A dosage sensitive locus at chromosome Xp21 is involved in male to female sex reversal. Nat Genet. 1994;7(4):497-501.
  41. Bashamboo A, Eozenou C, Jorgensen A, Bignon-Topalovic J, Siffroi JP, Hyon C, et al. Loss of Function of the Nuclear Receptor NR2F2, Encoding COUP-TF2, Causes Testis Development and Cardiac Defects in 46,XX Children. Am J Hum Genet. 2018;102(3):487-93.
  42. Achermann JC, Ito M, Ito M, Hindmarsh PC, Jameson JL. A mutation in the gene encoding steroidogenic factor-1 causes XY sex reversal and adrenal failure in humans. Nat Genet. 1999;22(2):125-6.
  43. Guran T, Yesil G, Turan S, Atay Z, Bozkurtlar E, Aghayev A, et al. PPP2R3C gene variants cause syndromic 46,XY gonadal dysgenesis and impaired spermatogenesis in humans. Eur J Endocrinol. 2019;180(5):291-309.
  44. Parma P, Radi O, Vidal V, Chaboissier MC, Dellambra E, Valentini S, et al. R-spondin1 is essential in sex determination, skin differentiation and malignancy. Nat Genet. 2006;38(11):1304-9.
  45. Ayers KL, Eggers S, Rollo BN, Smith KR, Davidson NM, Siddall NA, et al. Variants in SART3 cause a spliceosomopathy characterised by failure of testis development and neuronal defects. Nat Commun. 2023;14(1):3403.
  46. Sutton E, Hughes J, White S, Sekido R, Tan J, Arboleda V, et al. Identification of SOX3 as an XX male sex reversal gene in mice and humans. J Clin Invest. 2011;121(1):328-41.
  47. Portnoi MF, Dumargne MC, Rojo S, Witchel SF, Duncan AJ, Eozenou C, et al. Mutations involving the SRY-related gene SOX8 are associated with a spectrum of human reproductive anomalies. Hum Mol Genet. 2018;27(7):1228-40.
  48. Croft B, Ohnesorg T, Hewitt J, Bowles J, Quinn A, Tan J, et al. Human sex reversal is caused by duplication or deletion of core enhancers upstream of SOX9. Nat Commun. 2018;9(1):5319.
  49. Falah N, Posey JE, Thorson W, Benke P, Tekin M, Tarshish B, et al. 22q11.2q13 duplication including SOX10 causes sex-reversal and peripheral demyelinating neuropathy, central dysmyelinating leukodystrophy, Waardenburg syndrome, and Hirschsprung disease. Am J Med Genet A. 2017;173(4):1066-70.
  50. Ahmad A, Siddiqui MA, Goyal A, Wangnoo SK. Is 46XX karyotype always a female? BMJ Case Rep. 2012;2012.
  51. Jäger RJ, Anvret M, Hall K, Scherer G. A human XY female with a frame shift mutation in the candidate testis-determining gene SRY. Nature. 1990;348(6300):452-4.
  52. Puffenberger EG, Hu-Lince D, Parod JM, Craig DW, Dobrin SE, Conway AR, et al. Mapping of sudden infant death with dysgenesis of the testes syndrome (SIDDT) by a SNP genome scan and identification of TSPYL loss of function. Proc Natl Acad Sci U S A. 2004;101(32):11689-94.
  53. Eozenou C, Gonen N, Touzon MS, Jorgensen A, Yatsenko SA, Fusee L, et al. Testis formation in XX individuals resulting from novel pathogenic variants in Wilms' tumor 1 (WT1) gene. Proc Natl Acad Sci U S A. 2020;117(24):13680-8.
  54. Hammes A, Guo JK, Lutsch G, Leheste JR, Landrock D, Ziegler U, et al. Two splice variants of the Wilms' tumor 1 gene have distinct functions during sex determination and nephron formation. Cell. 2001;106(3):319-29.
  55. Mandel H, Shemer R, Borochowitz ZU, Okopnik M, Knopf C, Indelman M, et al. SERKAL syndrome: an autosomal-recessive disorder caused by a loss-of-function mutation in WNT4. Am J Hum Genet. 2008;82(1):39-47.
  56. Biason-Lauber A, Konrad D, Navratil F, Schoenle EJ. A WNT4 mutation associated with Müllerian-duct regression and virilization in a 46,XX woman. N Engl J Med. 2004;351(8):792-8.
  57. Bashamboo A, Brauner R, Bignon-Topalovic J, Lortat-Jacob S, Karageorgou V, Lourenco D, et al. Mutations in the FOG2/ZFPM2 gene are associated with anomalies of human testis determination. Hum Mol Genet. 2014;23(14):3657-65.
  58. Harris A, Siggers P, Corrochano S, Warr N, Sagar D, Grimes DT, et al. ZNRF3 functions in mammalian sex determination by inhibiting canonical WNT signaling. Proc Natl Acad Sci U S A. 2018;115(21):5474-9.
  59. Miller WL. MECHANISMS IN ENDOCRINOLOGY: Rare defects in adrenal steroidogenesis. Eur J Endocrinol. 2018;179(3):R125-r41.
  60. Flück CE, Meyer-Böni M, Pandey AV, Kempná P, Miller WL, Schoenle EJ, Biason-Lauber A. Why boys will be boys: two pathways of fetal testicular androgen biosynthesis are needed for male sexual differentiation. Am J Hum Genet. 2011;89(2):201-18.
  61. Flück CE, Pandey AV. Steroidogenesis of the testis -- new genes and pathways. Ann Endocrinol (Paris). 2014;75(2):40-7.
  62. Kok RC, Timmerman MA, Wolffenbuttel KP, Drop SL, de Jong FH. Isolated 17,20-lyase deficiency due to the cytochrome b5 mutation W27X. J Clin Endocrinol Metab. 2010;95(3):994-9.
  63. Khattab A, Yuen T, Yau M, Domenice S, Frade Costa EM, Diya K, et al. Pitfalls in hormonal diagnosis of 17-beta hydroxysteroid dehydrogenase III deficiency. J Pediatr Endocrinol Metab. 2015;28(5-6):623-8.
  64. Burkhard FZ, Parween S, Udhane SS, Flück CE, Pandey AV. P450 Oxidoreductase deficiency: Analysis of mutations and polymorphisms. J Steroid Biochem Mol Biol. 2017;165(Pt A):38-50.
  65. Miller WL, Huang N, Pandey AV, Flück CE, Agrawal V. P450 oxidoreductase deficiency: a new disorder of steroidogenesis. Ann N Y Acad Sci. 2005;1061:100-8.
  66. Miller WL, Huang N, Flück CE, Pandey AV. P450 oxidoreductase deficiency. Lancet. 2004;364(9446):1663.
  67. Walsh PC, Madden JD, Harrod MJ, Goldstein JL, MacDonald PC, Wilson JD. Familial incomplete male pseudohermaphroditism, type 2. Decreased dihydrotestosterone formation in pseudovaginal perineoscrotal hypospadias. N Engl J Med. 1974;291(18):944-9.
  68. Imperato-McGinley J, Zhu YS. Androgens and male physiology the syndrome of 5alpha-reductase-2 deficiency. Mol Cell Endocrinol. 2002;198(1-2):51-9.
  69. Kang HJ, Imperato-McGinley J, Zhu YS, Rosenwaks Z. The effect of 5α-reductase-2 deficiency on human fertility. Fertil Steril. 2014;101(2):310-6.
  70. Nordenskjöld A, Ivarsson SA. Molecular characterization of 5 alpha-reductase type 2 deficiency and fertility in a Swedish family. J Clin Endocrinol Metab. 1998;83(9):3236-8.
  71. Bertelloni S, Baldinotti F, Baroncelli GI, Caligo MA, Peroni D. Paternity in 5α-Reductase-2 Deficiency: Report of Two Brothers with Spontaneous or Assisted Fertility and Literature Review. Sex Dev. 2019;13(2):55-9.
  72. Mendonca BB, Batista RL, Domenice S, Costa EM, Arnhold IJ, Russell DW, Wilson JD. Steroid 5α-reductase 2 deficiency. J Steroid Biochem Mol Biol. 2016;163:206-11.
  73. Hornig NC, Holterhus PM. Molecular basis of androgen insensitivity syndromes. Mol Cell Endocrinol. 2021;523:111146.
  74. Batista RL, Costa EMF, Rodrigues AS, Gomes NL, Faria JA, Jr., Nishi MY, et al. Androgen insensitivity syndrome: a review. Arch Endocrinol Metab. 2018;62(2):227-35.
  75. Shukla GC, Plaga AR, Shankar E, Gupta S. Androgen receptor-related diseases: what do we know? Andrology. 2016;4(3):366-81.
  76. Zhou ZX, Wong CI, Sar M, Wilson EM. The androgen receptor: an overview. Recent Prog Horm Res. 1994;49:249-74.
  77. The androgen receptor gene mutation world wide web server[Available from: http://androgendb.mcgill.ca/.
  78. Gottlieb B TM. Androgen Insensitivity Syndrome. 1999 Mar 24 [Updated 2017 May 11]In: GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2023. Available from: https://www.ncbi.nlm.nih.gov/books/NBK1429/.
  79. Gottlieb B, Beitel LK, Nadarajah A, Paliouras M, Trifiro M. The androgen receptor gene mutations database: 2012 update. Hum Mutat. 2012;33(5):887-94.
  80. Ahmed SF, Bashamboo A, Lucas-Herald A, McElreavey K. Understanding the genetic aetiology in patients with XY DSD. Br Med Bull. 2013;106:67-89.
  81. Hornig NC, Ukat M, Schweikert HU, Hiort O, Werner R, Drop SL, et al. Identification of an AR Mutation-Negative Class of Androgen Insensitivity by Determining Endogenous AR Activity. J Clin Endocrinol Metab. 2016;101(11):4468-77.
  82. Hornig NC, Rodens P, Dörr H, Hubner NC, Kulle AE, Schweikert HU, et al. Epigenetic Repression of Androgen Receptor Transcription in Mutation-Negative Androgen Insensitivity Syndrome (AIS Type II). J Clin Endocrinol Metab. 2018;103(12):4617-27.
  83. Knerr J, Werner R, Schwan C, Wang H, Gebhardt P, Grötsch H, et al. Formin-mediated nuclear actin at androgen receptors promotes transcription. Nature. 2023;617(7961):616-22.
  84. Mongan NP, Tadokoro-Cuccaro R, Bunch T, Hughes IA. Androgen insensitivity syndrome. Best Pract Res Clin Endocrinol Metab. 2015;29(4):569-80.
  85. Hughes IA, Werner R, Bunch T, Hiort O. Androgen insensitivity syndrome. Semin Reprod Med. 2012;30(5):432-42.
  86. El-Maouche D, Arlt W, Merke DP. Congenital adrenal hyperplasia. Lancet. 2017;390(10108):2194-210.
  87. Morel Y, Roucher F, Plotton I, Simard J, Coll M. 3β-hydroxysteroid dehydrogenase deficiency.Genetic steroid disorders: Elsevier; 2014. p. 99-110.
  88. White PC. Genetic Steroid Disorders: Chapter 3D. Steroid 11β-Hydroxylase Deficiency and Related Disorders: Elsevier Inc. Chapters; 2013.
  89. Khattab A, Haider S, Kumar A, Dhawan S, Alam D, Romero R, et al. Clinical, genetic, and structural basis of congenital adrenal hyperplasia due to 11β-hydroxylase deficiency. Proc Natl Acad Sci U S A. 2017;114(10):E1933-e40.
  90. Khattab A, Marshall I. Management of congenital adrenal hyperplasia: beyond conventional glucocorticoid therapy. Curr Opin Pediatr. 2019;31(4):550-4.
  91. Yau M, Gujral J, New MI. Congenital Adrenal Hyperplasia: Diagnosis and Emergency Treatment. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc. Copyright © 2000-2023, MDText.com, Inc.; 2000.
  92. Goto M, Piper Hanley K, Marcos J, Wood PJ, Wright S, Postle AD, et al. In humans, early cortisol biosynthesis provides a mechanism to safeguard female sexual development. J Clin Invest. 2006;116(4):953-60.
  93. Harada N. Genetic analysis of human placental aromatase deficiency. J Steroid Biochem Mol Biol. 1993;44(4-6):331-40.
  94. Wilkins L, Jones HW, Jr., Holman GH, Stempfel RS, Jr. Masculinization of the female fetus associated with administration of oral and intramuscular progestins during gestation: non-adrenal female pseudohermaphrodism. J Clin Endocrinol Metab. 1958;18(6):559-85.
  95. Troisi R, Hatch EE, Palmer JR, Titus L, Robboy SJ, Strohsnitter WC, et al. Prenatal diethylstilbestrol exposure and high-grade squamous cell neoplasia of the lower genital tract. Am J Obstet Gynecol. 2016;215(3):322.e1-8.
  96. Stillman RJ. In utero exposure to diethylstilbestrol: adverse effects on the reproductive tract and reproductive performance and male and female offspring. Am J Obstet Gynecol. 1982;142(7):905-21.
  97. Kalfa N, Philibert P, Baskin LS, Sultan C. Hypospadias: interactions between environment and genetics. Mol Cell Endocrinol. 2011;335(2):89-95.
  98. Jorgensen A, Svingen T, Miles H, Chetty T, Stukenborg JB, Mitchell RT. Environmental Impacts on Male Reproductive Development: Lessons from Experimental Models. Horm Res Paediatr. 2023;96(2):190-206.
  99. Hurtado-Gonzalez P, Mitchell RT. Analgesic use in pregnancy and male reproductive development. Curr Opin Endocrinol Diabetes Obes. 2017;24(3):225-32.
  100. Giordano J, Prior HM, Bamforth JS, Walter MA. Genetic study of SOX9 in a case of campomelic dysplasia. Am J Med Genet. 2001;98(2):176-81.
  101. Granata T, Freri E, Caccia C, Setola V, Taroni F, Battaglia G. Schizencephaly: clinical spectrum, epilepsy, and pathogenesis. J Child Neurol. 2005;20(4):313-8.
  102. Kremen J, Chan YM. Genetic evaluation of disorders of sex development: current practice and novel gene discovery. Curr Opin Endocrinol Diabetes Obes. 2019;26(1):54-9.
  103. Sofatzis JA, Alexacos L, Skouteli HN, Tiniakos G, Padiatellis C. Malformed female genitalia in newborns with the VATER association. Acta Paediatr Scand. 1983;72(6):923-4.
  104. Ahmed SF, Achermann JC, Arlt W, Balen A, Conway G, Edwards Z, et al. Society for Endocrinology UK guidance on the initial evaluation of an infant or an adolescent with a suspected disorder of sex development (Revised 2015). Clin Endocrinol (Oxf). 2016;84(5):771-88.
  105. Davies JH, Cheetham T. Recognition and assessment of atypical and ambiguous genitalia in the newborn. Arch Dis Child. 2017;102(10):968-74.
  106. Baidya A, Basu AK, Bhattacharjee R, Biswas D, Biswas K, Chakraborty PP, et al. Diagnostic approach in 46, XY DSD: an endocrine society of bengal (ESB) consensus statement. J Pediatr Endocrinol Metab. 2023;36(1):4-18.
  107. Bever YV, Brüggenwirth HT, Wolffenbuttel KP, Dessens AB, Groenenberg IAL, Knapen M, et al. Under-reported aspects of diagnosis and treatment addressed in the Dutch-Flemish guideline for comprehensive diagnostics in disorders/differences of sex development. J Med Genet. 2020;57(9):581-9.
  108. Prader A. [Genital findings in the female pseudo-hermaphroditism of the congenital adrenogenital syndrome; morphology, frequency, development and heredity of the different genital forms]. Helv Paediatr Acta. 1954;9(3):231-48.
  109. Quigley CA, De Bellis A, Marschke KB, el-Awady MK, Wilson EM, French FS. Androgen receptor defects: historical, clinical, and molecular perspectives. Endocr Rev. 1995;16(3):271-321.
  110. van der Straaten S, Springer A, Zecic A, Hebenstreit D, Tonnhofer U, Gawlik A, et al. The External Genitalia Score (EGS): A European Multicenter Validation Study. J Clin Endocrinol Metab. 2020;105(3).
  111. Cao W, Ding X, Dong Z, Tang H. Reference Values for and Correlation Analysis of the Anogenital Distance of Chinese Han Full-Term Singleton Neonates. Front Pediatr. 2022;10:905421.
  112. Vanderbrink BA, Rink RC, Cain MP, Kaefer M, Meldrum KK, Misseri R, Karmazyn B. Does preoperative genitography in congenital adrenal hyperplasia cases affect surgical approach to feminizing genitoplasty? J Urol. 2010;184(4 Suppl):1793-8.
  113. Eggers S, Sadedin S, van den Bergen JA, Robevska G, Ohnesorg T, Hewitt J, et al. Disorders of sex development: insights from targeted gene sequencing of a large international patient cohort. Genome Biol. 2016;17(1):243.
  114. Baxter RM, Arboleda VA, Lee H, Barseghyan H, Adam MP, Fechner PY, et al. Exome sequencing for the diagnosis of 46,XY disorders of sex development. J Clin Endocrinol Metab. 2015;100(2):E333-44.
  115. Bacila IA, Lawrence NR, Badrinath SG, Balagamage C, Krone NP. Biomarkers in congenital adrenal hyperplasia. Clin Endocrinol (Oxf). 2023.
  116. Ye L, Zhao Z, Ren H, Wang W, Zhou W, Zheng S, et al. A Multiclassifier System to Identify and Subtype Congenital Adrenal Hyperplasia Based on Circulating Steroid Hormones. J Clin Endocrinol Metab. 2022;107(8):e3304-e12.
  117. Kamrath C, Friedrich C, Hartmann MF, Wudy SA. Metabotypes of congenital adrenal hyperplasia in infants determined by gas chromatography-mass spectrometry in spot urine. J Steroid Biochem Mol Biol. 2023;231:106304.
  118. Jha S, Turcu AF, Sinaii N, Brookner B, Auchus RJ, Merke DP. 11-Oxygenated Androgens Useful in the Setting of Discrepant Conventional Biomarkers in 21-Hydroxylase Deficiency. J Endocr Soc. 2021;5(2):bvaa192.
  119. Bileck A, Fluck CE, Dhayat N, Groessl M. How high-resolution techniques enable reliable steroid identification and quantification. J Steroid Biochem Mol Biol. 2019;186:74-8.
  120. Dhayat NA, Frey AC, Frey BM, d'Uscio CH, Vogt B, Rousson V, et al. Estimation of reference curves for the urinary steroid metabolome in the first year of life in healthy children: Tracing the complexity of human postnatal steroidogenesis. J Steroid Biochem Mol Biol. 2015;154:226-36.
  121. Enver EO, Vatansever P, Guran O, Bilgin L, Boran P, Turan S, et al. Adrenal steroids reference ranges in infancy determined by LC-MS/MS. Pediatr Res. 2022;92(1):265-74.
  122. Guibourdenche J, Lucidarme N, Chevenne D, Rigal O, Nicolas M, Luton D, et al. Anti-Müllerian hormone levels in serum from human foetuses and children: pattern and clinical interest. Mol Cell Endocrinol. 2003;211(1-2):55-63.
  123. Schmidt H, Schwarz HP. Serum concentrations of LH and FSH in the healthy newborn. Eur J Endocrinol. 2000;143(2):213-5.
  124. 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.
  125. Bertelloni S, Russo G, Baroncelli GI. Human Chorionic Gonadotropin Test: Old Uncertainties, New Perspectives, and Value in 46,XY Disorders of Sex Development. Sex Dev. 2018;12(1-3):41-9.
  126. Gomes NL, Batista RL, Nishi MY, Lerário AM, Silva TE, de Moraes Narcizo A, et al. Contribution of Clinical and Genetic Approaches for Diagnosing 209 Index Cases With 46,XY Differences of Sex Development. J Clin Endocrinol Metab. 2022;107(5):e1797-e806.
  127. Guerrero-Fernández J, González-Peramato P, Rodríguez Estévez A, Alcázar Villar MJ, Audí Parera L, Azcona San Julián MC, et al. Consensus guide on prophylactic gonadectomy in different sex development. Endocrinol Diabetes Nutr (Engl Ed). 2022;69(8):629-45.
  128. Patel V, Casey RK, Gomez-Lobo V. Timing of Gonadectomy in Patients with Complete Androgen Insensitivity Syndrome-Current Recommendations and Future Directions. J Pediatr Adolesc Gynecol. 2016;29(4):320-5.
  129. Sircili MH, e Silva FA, Costa EM, Brito VN, Arnhold IJ, Dénes FT, et al. Long-term surgical outcome of masculinizing genitoplasty in large cohort of patients with disorders of sex development. J Urol. 2010;184(3):1122-7.
  130. Yan M, Dilihuma J, Luo Y, Reyilanmu B, Shen Y, Mireguli M. Novel Compound Heterozygous Variants in the LHCGR Gene in a Genetically Male Patient with Female External Genitalia. J Clin Res Pediatr Endocrinol. 2019;11(2):211-7.
  131. Berg JS, French SL, McCullough LB, Kleppe S, Sutton VR, Gunn SK, Karaviti LP. Ethical and legal implications of genetic testing in androgen insensitivity syndrome. J Pediatr. 2007;150(4):434-8.
  132. Wisniewski AB. Complete Androgen Insensitivity Syndrome: Long-Term Medical, Surgical, and Psychosexual Outcome. Journal of Clinical Endocrinology &amp; Metabolism. 2000;85(8):2664-9.
  133. Bianco S, Agrifoglio V, Mannino F, Cefalù E, Cittadini E. Successful pregnancy in a pure gonadal dysgenesis with karyotype 46,XY patient (Swyer's syndrome) following oocyte donation and hormonal treatment. Acta Eur Fertil. 1992;23(1):37-8.
  134. Hines M, Ahmed SF, Hughes IA. Archives of Sexual Behavior. 2003;32(2):93-101.
  135. Park S, Ha SH, Kim KS. Long-term follow-up after feminizing genital reconstruction in patients with ambiguous genitalia and high vaginal confluence. Journal of Korean medical science. 2011;26(3):399-403.
  136. Sircili MHP, de Queiroz e Silva FA, Costa EMF, Brito VN, Arnhold IJP, Dénes FT, et al. Long-Term Surgical Outcome of Masculinizing Genitoplasty in Large Cohort of Patients With Disorders of Sex Development. Journal of Urology. 2010;184(3):1122-7.
  137. Wisniewski AB, Migeon CJ, Gearhart JP, Rock JA, Berkovitz GD, Plotnick LP, et al. Congenital Micropenis: Long-Term Medical, Surgical and Psychosexual Follow-Up of Individuals Raised Male or Female. Hormone Research in Paediatrics. 2001;56(1-2):3-11.
  138. Mendonca BB, Costa EM, Belgorosky A, Rivarola MA, Domenice S. 46,XY DSD due to impaired androgen production. Best Pract Res Clin Endocrinol Metab. 2010;24(2):243-62.
  139. Batista RL, Mendonca BB. Integrative and Analytical Review of the 5-Alpha-Reductase Type 2 Deficiency Worldwide. Appl Clin Genet. 2020;13:83-96.
  140. Bonnet E, Winter M, Mallet D, Plotton I, Bouvattier C, Cartigny M, et al. Changes in the clinical management of 5α-reductase type 2 and 17β-hydroxysteroid dehydrogenase type 3 deficiencies in France. Endocr Connect. 2023;12(3).
  141. Duranteau L, Rapp M, van de Grift TC, Hirschberg AL, Nordenskjöld A. Participant- and Clinician-Reported Long-Term Outcomes After Surgery in Individuals with Complete Androgen Insensitivity Syndrome. J Pediatr Adolesc Gynecol. 2021;34(2):168-75.
  142. Dessens AB, Slijper FME, Drop SLS. Gender Dysphoria and Gender Change in Chromosomal Females with Congenital Adrenal Hyperplasia. Archives of Sexual Behavior. 2005;34(4):389-97.
  143. Long DN, Wisniewski AB, Migeon CJ. Gender Role Across Development in Adult Women with Congenital Adrenal Hyperplasia Due to 21-Hydroxylase Deficiency. Journal of Pediatric Endocrinology and Metabolism. 2004;17(10).
  144. Wisniewski AB, Migeon CJ, Malouf MA, Gearhart JP. PSYCHOSEXUAL OUTCOME IN WOMEN AFFECTED BY CONGENITAL ADRENAL HYPERPLASIA DUE TO 21-HYDROXYLASE DEFICIENCY. Journal of Urology. 2004;171(6 Part 1):2497-501.
  145. Lee PA, Houk CP. Outcome Studies Among Men with Micropenis. Journal of Pediatric Endocrinology and Metabolism. 2004;17(8).
  146. Lee PA, Houk CP, Husmann DA. Should Male Gender Assignment be Considered in the Markedly Virilized Patient With 46,XX and Congenital Adrenal Hyperplasia? Journal of Urology. 2010;184(4S):1786-92.
  147. de Jesus LE, Costa EC, Dekermacher S. Gender dysphoria and XX congenital adrenal hyperplasia: how frequent is it? Is male-sex rearing a good idea? Journal of Pediatric Surgery. 2019;54(11):2421-7.
  148. Crissman HP, Warner L, Gardner M, Carr M, Schast A, Quittner AL, et al. Children with disorders of sex development: A qualitative study of early parental experience. International journal of pediatric endocrinology. 2011;2011(1):10-.
  149. Yau M, Khattab A, New MI. Prenatal Diagnosis of Congenital Adrenal Hyperplasia. Endocrinology and Metabolism Clinics of North America. 2016;45(2):267-81.
  150. Van't Westeinde A, Karlsson L, Messina V, Wallensteen L, Brösamle M, Dal Maso G, et al. An update on the long-term outcomes of prenatal dexamethasone treatment in congenital adrenal hyperplasia. Endocr Connect. 2023;12(4).
  151. Kazmi D, Bailey J, Yau M, Abu-Amer W, Kumar A, Low M, Yuen T. New developments in prenatal diagnosis of congenital adrenal hyperplasia. The Journal of Steroid Biochemistry and Molecular Biology. 2017;165:121-3.
  152. Maher JY, Gomez-Lobo V, Merke DP. The management of congenital adrenal hyperplasia during preconception, pregnancy, and postpartum. Rev Endocr Metab Disord. 2023;24(1):71-83.
  153. Fedele D, Kirk K, Wolfe-Christensen C, Phillips T, Mazur T, Mullins L, et al. Primary Caregivers of Children Affected by Disorders of Sex Development: Mental Health and Caregiver Characteristics in the Context of Genital Ambiguity and Genitoplasty. International Journal of Pediatric Endocrinology. 2010;2010(1):690674.
  154. Hullmann SE, Fedele DA, Wolfe-Christensen C, Mullins LL, Wisniewski AB. Differences in adjustment by child developmental stage among caregivers of children with disorders of sex development. International journal of pediatric endocrinology. 2011;2011(1):16-.
  155. Kirk KD, Fedele DA, Wolfe-Christensen C, Phillips TM, Mazur T, Mullins LL, et al. Parenting Characteristics of Female Caregivers of Children Affected by Chronic Endocrine Conditions: A Comparison Between Disorders of Sex Development and Type 1 Diabetes Mellitus. Journal of Pediatric Nursing. 2011;26(6):e29-e36.
  156. Wolfe-Christensen C, Fedele DA, Kirk K, Phillips TM, Mazur T, Mullins LL, et al. Degree of External Genital Malformation at Birth in Children with a Disorder of Sex Development and Subsequent Caregiver Distress. Journal of Urology. 2012;188(4S):1596-600.
  157. Wisniewski AB, Batista RL, Costa EMF, Finlayson C, Sircili MHP, Dénes FT, et al. Management of 46,XY Differences/Disorders of Sex Development (DSD) Throughout Life. Endocrine Reviews. 2019;40(6):1547-72.
  158. Magritte E. Working together in placing the long term interests of the child at the heart of the DSD evaluation. Journal of Pediatric Urology. 2012;8(6):571-5.

 

Fibromyalgia

ABSTRACT

 

Fibromyalgia is a clinical entity characterized by the combination of chronic widespread pain and other non-pain symptoms, including fatigue, poor sleep, and cognitive disturbances, which can exhibit symptom variation not only between different patients, but also in the same patient during the course of the disease. These symptoms are relatively common and non-specific. They can be encountered in other disorders that may overlap with fibromyalgia, often without having clear boundaries, while their nature makes them difficult to be objectively defined and quantified. These issues have led to significant controversy over the definition and the diagnostic criteria of fibromyalgia. It has been suggested that the markers of physical and psychological distress have a continuous distribution in the general population with fibromyalgia patients being at the extreme end of this continuum. Genetic predisposition in combination with environmental factors, are responsible for each individual’s position in this this distribution. In recent years more knowledge has been obtained to better understand the environmental factors that seem to be important in triggering fibromyalgia. Most of them act as stressors superimposed onto a deranged stress-response system leading to dys-regulation of the nociceptive system and the appearance of clinical symptoms. The aim of the therapy is to relieve pain and motivate the patients to become more physically active using a multimodal individualized therapeutic strategy that includes education, exercise, cognitive-behavioral approaches and medications. The response to current therapeutic modalities varies significantly, with some patients responding adequately, while others do not seem to experience any long-term benefit. 

 

INTRODUCTION

 

Fibromyalgia is a clinical entity characterized by the combination of ill-defined symptoms including chronic widespread pain, with concomitant fatigue, sleeping disorders, and cognitive disturbances (1). The severity of these symptoms can vary significantly during the course of the disease. Fibromyalgia has been described as an arbitrarily created syndrome that lies at the extreme end of the spectrum of poly-symptomatic distress (2). The term poly-symptomatic was used to emphasize the variety of multiple different symptoms that can be found in fibromyalgia patients, while the distress can have a physical and/or a psychological component. This exact nature of fibromyalgia makes it difficult to be clearly defined, often overlapping with disorders that are characterized by similar symptoms. It is important to note that fibromyalgia is not an exclusion diagnosis as it can co-exist with other clinical conditions (3). 

 

CLINICAL FEATURES

 

The main presenting complains of patients with fibromyalgia include chronic widespread pain (also called multisite pain), fatigue, and poor sleep. Usually the pain is initially localized, but eventually it involves many muscle groups. It is characterized as persistent with varying intensity, while it can often be described as a sensation of burning, gnawing soreness, stiffness, or aching. Excessive sensitivity to normally painful stimuli, such as pressure or heat (hyperalgesia) and painful sensation to normally non-painful stimuli, such as touch (allodynia) are significant features of fibromyalgia. Often patients complain of swollen joints and paresthesias without though the presence of any objective clinical findings during physical examination. Pain is often aggravated by cold and humid weather, poor sleep, physical and mental stress. Additionally the patients may have a variety of less well understood pain symptoms, including abdominal pain, chest wall pain, symptoms suggestive of irritable bowel syndrome, pelvic pain, and bladder symptoms of frequency and urgency suggestive of interstitial cystitis (4–9).

 

Fatigue is present in almost all patients with fibromyalgia, while many complain of non-refreshing sleep, frequent awakening during the night, and difficulty falling back to sleep. Sleep apnea and nocturnal myoclonus can also be present along with a sensation of light-headedness, dizziness, and faintness. In addition, cognitive difficulties such as short-term memory loss, groping for words and poor vocabulary, are common among patients with fibromyalgia. Mood disturbances, including depression, anxiety and heightened somatic concern, may often also occur. Headaches, either muscular or migraine type, are also commonly present (6,7). Other often co-existing conditions include multiple chemical sensitivity, “allergic” symptoms, ocular dryness, palpitations, dyspnea, vulvodynia, dysmenorrhea, premenstrual syndrome, sexual dysfunction, weight fluctuations, night sweats, dysphagia, restless leg syndrome, temporomandibular joint pain, chronic fatigue syndrome (systemic exertion intolerance disease), Raynaud`s phenomenon, autonomic dysfunction,  and dysgeusia (6,8,9). These conditions cannot be used to support the diagnosis of fibromyalgia.

 

Approximately 40% of fibromyalgia patients have accompanying depression at the time of diagnosis, while 60% of patients have a lifetime history of depression. In addition, an anxiety disorder is present in 30% of the cases at the time of diagnosis while the lifetime prevalence of an anxiety disorder in fibromyalgia patients is approximately 60% (10–14). The levels of depression and anxiety in patients with fibromyalgia seem to be associated with the degree of cognitive impairment, as shown in a meta-analysis of 23 case-control studies (15). Based on the coexistence of depression and anxiety, fibromyalgia patients can be divided into 2 major groups. The first group comprises of patients without coexisting mood disorders, while the second of patients with concomitant depressive mood, often in combination with anxiety. According to the results of a study that intended to subgroup fibromyalgia patients based on: 1) mood status (evaluated by the Center for Epidemiologic Studies Depression Scale for depression and the State-Trait Personality Inventory for symptoms of trait-related anxiety), 2) cognition (by the catastrophizing and control of pain subscales of the Coping Strategies Questionnaire), and 3) hyperalgesia/tenderness (by dolorimetry and random pressure-pain applied at suprathreshold values), it was noted that fibromyalgia patients with depressive mood and anxiety are also ‘catastrophizing’. This term is used to indicate that such patients have a very negative, pessimistic view of what their pain is and what is causing, while they have no sense that they can control their pain. On the contrary fibromyalgia patients who are neither depressed nor anxious and therefore do not catastrophize, have a moderate sense that they can control their pain. These patients can be further divided into 2 subgroups based on the degree of hyperalgesia/tenderness, the first subgroup comprises of patients with high hyperalgesia/tenderness, while the second one of patients with moderate hyperalgesia/tenderness (11). In addition, it has been proposed that depressed fibromyalgia patients can also be divided into 2 subgroups, in the first one depression is a co-morbid condition, while in the second depression is the cause of fibromyalgia (16). All these fibromyalgia subgroups are illustrated in Figure 1.

 

Figure 1. Subgroups of fibromyalgia patients.

 

In another study fibromyalgia patients were classified as dysfunctional, inter-personally distressed, or adaptive copers, based on their responses to the Multidimensional Pain Inventory. The dysfunctional patients experienced more pain behaviors and overt expressions of pain, distress, and suffering, such as slowed movement, bracing, limping, and grimacing compared to the inter-personally distressed or the adaptive copers (17).

 

It is of interest that up to  25% of patients correctly diagnosed with a systemic rheumatic disease (e.g. rheumatoid arthritis, systemic lupus erythematosus) will also fulfill the classification criteria for fibromyalgia (18). This is also the case for many patients who experience persistent various forms of pain (including widespread myalgias, arthralgias, and headache), fatigue, neurocognitive dysfunction, and sleep disturbances after an infection with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) that caused a mild to moderate coronavirus disease 2019 (long COVID) (19,20). The most commonly encountered comorbid conditions in fibromyalgia patients are shown in Table 1.

 

-    Table 1. The Most Commonly Encountered Co-Morbid Conditions in Fibromyalgia

Sleep disorders

-    Non restorative sleep (alpha-delta sleep anomaly)

-    Sleep apnea

-    Restless leg syndrome

-    Nocturnal myoclonus

Chronic fatigue syndrome (systemic exertion intolerance disease)

 

Psychiatric disorders

 

-    Anxiety disorders

-    Depression

-    Obsessive compulsive disorder

Headache

-    Tension type headache

-    Migraine

Irritable bowel syndrome

 

Musculofascial pain syndrome

-    Temporomandibular disorders

-    Interstitial cystitis

Dysmenorrhea

 

Premenstrual syndrome

 

Non-cardiac chest pain

 

Raynaud’s phenomenon

 

Systemic autoimmune diseases

-    Rheumatoid arthritis

-    Systematic lupus erythematosus

-    Sjögren’s syndrome

-    Ankylosing spondylitis and other seronegative spondylarthritis

-    Polymyalgia rheumatica

Long COVID

 

 

ASSESSMENT

 

Fibromyalgia is a chronic illness, with a variety of symptoms that can change during the course of the disease and after treatment. Therefore, its core symptoms should be evaluated both in clinical practice and in treatment trials. A working group within OMERACT (Outcome Measures in Rheumatology) reached a consensus regarding the domains that need to be assessed in clinical trials for fibromyalgia, using Delphi exercises within patients and expert clinicians (21). The fibromyalgia core symptom domains include pain intensity, tenderness, fatigue, sleep disturbance, multidimensional function (including health related quality of life and physical function), patient global impression of change, cognitive dysfunction, and depression. However at the present time, there is no consensus on how to evaluate these domains, in order to quantify fibromyalgia disease activity state and/or response (22).

 

Pain intensity can be assessed using visual analog scales. It has been proposed to use the wording “please rate your pain by circling on the number that best describes your pain on average” and has anchors that vary from “no pain” to “pain as bad as you can imagine” (23).

 

Tenderness can be measured by evaluating the alteration of the severity of pain at the tender points, using visual or analog scales, but not by the change in their number. The change in the number of tender points is poorly correlated with improvement in fibromyalgia treatment trials. It has been noted that the tender points measure the combination of tenderness and distress an individual has, rendering them inadequate for the evaluation of tenderness per se (24).

 

Fatigue can be evaluated by the Fatigue Severity Scale (FSS), which measures the functional outcomes related to fatigue (25). It has been shown that FSS has the most robust psychometric properties of 19 reviewed fatigue measures, while it had the best ability to act as an outcome measure sensitive to change with treatment, in chronically ill patients (26). Other instruments that have been used to assess fatigue in fibromyalgia patients include the Multidimensional Fatigue Index and the Multidimensional Assessment of Fatigue.

 

Sleep disturbance in fibromyalgia patients has been evaluated by the Medical Outcome Studies (MOS) sleep scale, the Functional Outcomes of Sleep Questionnaire (FOSQ), and the Jenkins Sleep Scale (JSS). Of these instruments MOS sleep scale lacks validity to assess changes in sleep symptoms in fibromyalgia treatment trials, FOSQ has not been adequately validated in fibromyalgia patients, while JSS has been criticized for possible high-recall bias, since it requires the patients to rate the frequency of their symptoms over a period of a month (23).

 

Multidimensional function, including health related quality of life and physical function, can be evaluated using the fibromyalgia impact questionnaire (FIQ). This represents a useful tool in assessing functional abilities in daily life and measures patient status, progress and outcomes. FIQ is self-administered and is highly sensitive to changes during the course of the disease. However, its functional items are orientated toward high levels of disability, resulting in a possible floor effect, while its physical function items are addressed to women living in affluent countries, generating gender and ethnic bias. To address these issues the Revised Fibromyalgia Impact Questionnaire (FIQR) was developed, having modified physical function questions and including questions on memory, tenderness, balance and environmental sensitivity, while keeping questions that evaluate overall impact and symptoms (27). Other tools for assessing overall function and quality of life include the Health Assessment Questionnaire, the Symptom Interpretation Questionnaire, the Western Ontario and McMaster Universities Osteoarthritis Index, the Patient Global Impression of Change scale (PGIC), and psychometric scales (28).

 

Patient global impression of change can be evaluated by the Patient Global Impression of Change scale (PGIC), as recommended by the Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials (IMMPACT) (29).

 

Cognitive dysfunction can be evaluated by the Multiple Ability Self-report Questionnaire (MASQ), which is a self-report questionnaire measuring language, visuoperception, verbal memory and attention (30). However self-assessment is poorly correlated with objective measures of cognitive function and has poor discriminating ability for patients with mild cognitive impairment (31).

 

Depression can be evaluated using the Hospital Anxiety and Depression Scale (HADS) depression subscale (HADS-D). The use of the original Beck Depression Inventory (BDI) as well as the current BDI-II, should be avoided, since they tend to overestimate the presence of major depressive disorder in fibromyalgia patients by evaluating a number of non-depressive symptoms, which are often encountered in fibromyalgia (23). 

 

DIAGNOSIS

 

As already mentioned, fibromyalgia is characterized by symptoms that can vary in number and intensity not only between different patients, but also in the same patient during the course of the disease. These symptoms are also common in other disorders that can overlap with fibromyalgia, often without having clear boundaries. Additionally, the nature of the symptoms of fibromyalgia makes them difficult to be objectively defined and measured. All these issues have led to significant controversy over the definition and diagnosis of fibromyalgia. The clinical entity of fibromyalgia was first described in 1904, under the term “fibrositis”, after focusing on clinical evidence of muscle sensitivity (32). It was not until 1977 that specific criteria for the diagnosis of fibromyalgia were introduced (33). Since then a number of different criteria have been proposed, based on a combination of tender point examination and the presence of symptoms. In 1986 a committee of the American College of Rheumatology (ACR) started a multicenter study, trying to provide a definition of fibromyalgia and  establish classification criteria (34). In 2013, the Analgesic, Anesthetic, and Addiction Clinical Trial Translations Innovations Opportunities and Networks (ACTTION) public-private partnership with the US Food and Drug Administration (FDA) and American Pain Society (APS) initiated the ACTTION-APS Pain Taxonomy (AAPT) in an attempt to develop a diagnostic system that would be clinically useful and consistent across chronic pain disorders, including fibromyalgia (35).

 

In clinical practice either the 2016 revision of the 2010/2011 fibromyalgia diagnostic criteria or the AAPT criteria can be used to help physicians to diagnose fibromyalgia (36). However, these criteria should be viewed as an aid and not as a gold standard for diagnosing fibromyalgia in clinical practice. Good clinical judgment is necessary to interpret the findings of physical examination and to assess psychological factors and associated comorbidities so as to correctly identify the patients with fibromyalgia (37).

 

Relevant social, personal and family history can be helpful in establishing the diagnosis of fibromyalgia, since there is evidence that the symptoms of fibromyalgia can appear after a physical or emotional trauma, a medical illness, or asurgical operation, while a family history of fibromyalgia, makes the diagnosis of fibromyalgia more likely (38).

 

1990 ACR Classification Criteria

 

In 1990 the American College of Rheumatology (ACR) committee established criteria for the classification of fibromyalgia. According to these criteria fibromyalgia is defined as chronic widespread pain involving both sides of the body, above and below the waist, as well as the whole length of the spine, and excessive tenderness in the pressure of 11 of 18 specific muscle-tendon sites (9 pairs of tender points). The locations of the tender points are described in Figure2 and Table 2. Pressure equivalent of 4 kg/cm is applied to these points using the pulp of the thumb or the first two or three fingers. This can be accurately measured with a dolorimeter or it can be estimated, since 4 kg/cm is the pressure needed to be applied so as to whiten the examiner’s fingernail bed. These criteria specifically state that fibromyalgia is not an exclusionary diagnosis (34).

 

Figure 2. Fibromyalgia tender points.

 

 

Table 2. Description of the Location of Fibromyalgia Tender Points

Occiput:

at the insertions of one or more of the following muscles: trapezius, sternocleidomastoid, splenius capitus, semispinalis capitus

Trapezius:

at the midpoint of the upper border

Supraspinatus:

above the scapular spine near the medial border

Gluteal:

at the upper outer quadrant of the buttocks at the anterior edge of the gluteus maximus

Low cervical:

at the anterior aspect of the interspaces between the transverse processes of C5–C7

Second rib:

just lateral to the second costochondral junctions

Lateral epicondyle:

2 cm distal to the lateral epicondyle

Greater trochanter:

posterior to the greater trochanteric prominence

Knee:

at the medial fat pad proximal to the joint line

 

The tender points, which are examined in fibromyalgia, are not just areas that the patient feels pain. They are points that fibromyalgia patients are relatively more tender, compared to normal individuals, when pressure is applied on them. But fibromyalgia patients are more tender wherever you apply pressure, not only to some of these 18 specific tender points, including areas previously considered to be “control points” (39). There is evidence that these tender points are areas that everyone is generally more tender. In contrast, fibromyalgia patients are more tender not only to pressure but to other stimuli such as heat, cold and other sensory stimuli, most probably due to decreased pain threshold. The number of tender points an individual has is highly correlated with distress, as defined by the presence of anxiety, depression, sleep disturbance, fatigue and global severity. Tender points have been described as “a sedimentation rate of distress”. Consequently tender points measure the combination of tenderness and distress an individual has (24).

 

The use of the 1990 ACR classification criteria in clinical practice is surrounded by substantial controversy. Tender points were introduced as an objective physical finding. However, if the physician who performs the physical examination is not experienced enough, tender point counting is impossible to be performed accurately. Most of the physicians examining fibromyalgia patients did not know how to carry out the tender point examination. Consequently, tender point count was not routinely performed, and when performed it was performed inaccurately (40). Another shortcoming of the tender point counting is that it is not as objective as it was initially considered, since the physician can be biased by the patient’s interview that precedes the physical examination. These issues lead to a low inter-examiner reliability of the tender point count (41). 

 

Other more sophisticated measures of assessing tenderness, such as applying stimuli randomly, when the individual cannot anticipate what the next stimulus is going to be, are equally abnormal in fibromyalgia patients, but do not correlate with distress (39). These methods require special training and are more time consuming than the trigger point count. Other alternative assessment methods include functional magnetic resonance imaging (fMRI) and nociceptive flexion reflex (NFR) testing, which documents abnormal pain processing in fibromyalgia. Functional MRI demonstrates similar brain activation in regions involved in pain processing in fibromyalgia patients and normal individuals. However fibromyalgia patients have increased pain sensitivity and brain activation during comparable stimulus (42). Nociceptive flexion reflexes are sensory-motor responses elicited by electrical noxious stimuli, which involve activation of spinal and supraspinal neuronal circuits, providing an objective and quantitative assessment of the function of the pain-controlsystem. It has been demonstrated that the NFR threshold in patients with fibromyalgia is significantly decreased compared with that in controls (43). Of these methods fMRI is expensive and complex compared to NFR testing that appears to be more easily accessible and convenient, since standard electromyographic equipment can be used. This test also seems to eliminate subjective bias and dissimulation (44).

 

The 1990 ACR classification criteria, define fibromyalgia in terms of pain rather than its other features. However, patients with fibromyalgia apart from tenderness and pain, also have a number of other somatic symptoms. Although non-pain symptoms are important, there is no evidence to support the notion that they are more important than hyperalgesia and allodynia, which are key symptoms of fibromyalgia (44). Many clinicians with experience in fibromyalgia did not feel that the 1990 ACR classification criteria were sufficiently reliable for the diagnosis of fibromyalgia in clinical practice and were  considering other aspects of the disease in an attempt to reach a more accurate diagnosis (38).

 

2010 ACR Preliminary Diagnostic Criteria

 

To address the aforementioned issues the ACR in 2010 proposed the preliminary diagnostic criteria for fibromyalgia (Table 3), that were not meant to replace the 1990 ACR classification criteria, but to represent an alternative simple and easy method of diagnosis in clinical practice (45). These diagnostic criteria do not require a tender point count. Instead, they rely only on symptoms for the diagnosis of fibromyalgia. They introduced the widespread pain index (WPI), which counts the areas that the patient feels pain during one week preceding the examination, and the symptom severity (SS) scale, which describes the severity of fatigue, unrefreshing sleep, cognitive problems, and a number of associated somatic fibromyalgia symptoms. These symptoms need to be assessed and rated by a physician, therefore the 2010 ACR preliminary diagnostic criteria are in-adequate for patient self-diagnosis.

 

Two more conditions need to be fulfilled so as to diagnose fibromyalgia. The symptoms need to be present at a similar level for at least 3 months while alternate disorders that would otherwise explain the pain need to be excluded (Table 3). The authors of the 2010 ACR preliminary diagnostic criteria have clarified that the latter condition does not mean that fibromyalgia is an exclusion diagnosis according to these criteria. The diagnosis of fibromyalgia should not be made only when there is not another disease that could explain the pain that would otherwise be attributed to fibromyalgia. It should be noted that rheumatic diseases usually do not cause pain that can be confused with fibromyalgia (3).

 

Table 3. 2010 ACR Preliminary Diagnostic Criteria

Criteria:

A patient satisfies diagnostic criteria for fibromyalgia if the following 3 conditions are met:

1)    Widespread pain index (WPI) ≥7 and symptom severity (SS) scale score ≥5 or

Widespread pain index (WPI) 3-6 and symptom severity (SS) scale score ≥9.

2)    Symptoms have been present at a similar level for at least 3 months.

3)    The patient does not have a disorder that would otherwise explain the pain.

 

Ascertainment:

1)    WPI

Note the number of areas in which the patient has had pain over the last week. In how many areas has the patient had pain?

(Score will be between 0 and 19)

 

 

-Neck

-Upper arm, left

 

-Abdomen

 

-Upper leg, left

 

 

 

-Jaw, left

-Upper arm, right

 

-Upper back

 

-Upper leg, right

 

 

 

-Jaw, right

-Lower arm, left

 

-Lower back

 

-Lower leg, left

 

 

 

-Shoulder girdle, left

 

-Lower arm, right

 

-Hip (buttock, trochanter), left

 

-Lower leg, right

 

 

 

-Shoulder girdle, right

 

-Chest

 

-Hip (buttock, trochanter), right

 

 

 

 

2)    SS scale score

The SS scale score is the sum of the severity of the 3 symptoms (fatigue, waking unrefreshed, cognitive symptoms) plus the extent (severity) of somatic symptoms in general.

(The final score is between 0 and 12)

 

-For the each of the 3 symptoms below, indicate the level of severity over the past week using the following scale:

0 = no problem

1 = slight or mild problems, generally mild or intermittent

2 = moderate, considerable problems, often present and/or at a moderate level

3 = severe: pervasive, continuous, life-disturbing problems

Fatigue                     (0-3)

Waking unrefreshed (0-3)

Cognitive symptoms (0-3)

 

-Considering somatic symptoms in general, indicate whether the patient has: muscle pain, irritable bowel syndrome, fatigue/tiredness, thinking or remembering problem, muscle weakness, headache, pain/cramps in the abdomen, numbness/tingling, dizziness, insomnia, depression, constipation, pain in the upper abdomen, nausea, nervousness, chest pain, blurred vision, fever, diarrhea, dry mouth, itching, wheezing, Raynaud's phenomenon, hives/welts, ringing in ears, vomiting, heartburn, oral ulcers, loss of/change in taste, seizures, dry eyes, shortness of breath, loss of appetite, rash, sun sensitivity, hearing difficulties, easy bruising, hair loss, frequent urination, painful urination, and bladder spasms

0 = no symptoms

1 = few symptoms

2 = a moderate number of symptoms

3 = a great deal of symptoms

 

There is evidence that there is good agreement between the 1990 ACR classification criteria and the 2010 ACR preliminary diagnostic criteria (46–53). However, these criteria are expected not to agree completely, as the former are focused on the presence of tender points while the latter on the presence of symptoms. The 1990 criteria can diagnose fibromyalgia in patients who do not have sufficiently high symptom score according to the 2010 criteria, while the 2010 criteria can diagnose fibromyalgia in patients who do not have sufficient tender points according to the 1990 criteria.

 

The introduction of the 2010 ACR preliminary diagnostic criteria was surrounded by controversy too. In particular, they have been criticized for being completely symptom focused, ill-defined, and lucking some mechanistic features of fibromyalgia, such as hyperalgesia, central sensitization and dysfunctional pain modulation (54). Additionally, these diagnostic criteria are based on the subjective assessment of the patient’s somatic symptoms by the physician, adding ambiguity and influencing repeatability among different physicians (55). A self-reported version of the 2010 ACR preliminary diagnostic criteria was developed in 2011, so as to be used in survey research, and not in clinical practice (56). These criteria are known as the modified 2010 ACR preliminary diagnostic criteria or the 2011 ACR survey criteria. They introduced the fibromyalgia severity (FS) score (originally called fibromyalgianess scale) which is the sum of the self-reported WPI and SS score. This score can be used as an approximate measure of the severity of fibromyalgia. The FS score has also been called polysymptomatic distress (PSD) scale. It has been proposed that the markers of physical and psychological distress have a continuous distribution in the general population with fibromyalgia patients being at the extreme end of this distribution (57). The PSD scale could be useful to define the position of each individual in this continuum, without having to differentiate between patients with  fibromyalgia and those without, as this distinction can sometimes be unclear if not arbitrary (58).

 

2016 Revisions to the 2010/2011 Fibromyalgia Diagnostic Criteria

 

A limitation of the WPI is the fact that it counts the number of painful areas without considering their distribution in the body. Patients with regional pain disorders can fulfill the 2010 ACR preliminary diagnostic criteria since pain can be located in 3 or more areas in the same region (59). To overcome this issue the 2016 revision of the diagnostic criteria require the pain to be generalized (multisite pain). The areas WPI assesses are divided in 5 regions (Table 4) and the diagnosis of  fibromyalgia requires the distribution of pain in 4 out of 5 regions (60). The jaw, the chest and the abdomen area are problematic when they are used to define a region. In this way they are excluded from the definition of generalized pain (61). Since pain needs to be located in at least 4 areas according to the 2016 revision, the previous criterion for diagnosis, WPI of 3-6 and SS scale score ≥9 was changed to WPI of 4-6 and SS scale score ≥9.

 

The 2010 and 2011 ACR preliminary diagnostic criteria are extremely similar. Their difference is that the 2010 criteria are physician-based and can be used in clinical practice for the diagnosis of fibromyalgia, while the 2011 criteria are self-reported and can be used only in survey research. According to the 2010 criteria the SS scale assesses a wide range of somatic symptoms, which makes them impractical for use in questionnaires. With the 2016 revision the assessment of somatic symptoms that is included in the SS scale is limited to headaches, pain and cramps in the lower abdomen and depression. In this way, there is no longer need for different criteria for clinical practice and for survey research. The same criteria can be used in both settings having 2 different methods of administration.

 

One prerequisite for diagnosis of fibromyalgia according to the 2010 ACR preliminary diagnostic criteria is the patient not to have a condition that would otherwise explain the pain. The authors of these criteria clarified that this does not mean that the diagnosis of fibromyalgia is an exclusion diagnosis. However, this phrasing was not considered clear enough and caused significant misunderstanding. In this way this criterion was removed in the 2016 revision. The diagnosis of fibromyalgia can be valid even if there is another condition that can cause the pain that is attributed to fibromyalgia. According to this definition fibromyalgia can coexist with other clinically significant conditions that can cause pain.

 

Table 4. 2016 Revisions to the 2010/2011 Fibromyalgia Diagnostic Criteria

Criteria:

A patient satisfies diagnostic criteria for fibromyalgia if the following 3 conditions are met:

1)    Widespread pain index (WPI) ≥7 and symptom severity (SS) scale score ≥5 or

Widespread pain index (WPI) 4-6 and symptom severity (SS) scale score ≥9.

2)    Generalized pain: Pain must be present in at least 4 of 5 regions.

Jaw, chest, and abdominal pain are not included in generalized pain definition.

3)    Symptoms have been generally for at least 3 months.

4)    A diagnosis of fibromyalgia is valid irrespective of other diagnoses. A diagnosis of fibromyalgia does not exclude the presence of other clinically important illnesses.

 

Ascertainment:

1)    WPI

Note the number of areas in which the patient has had pain over the last week. In how many areas has the patient had pain?

       (Score will be between 0 and 19)

 

Region 1: Left Upper Region

-Jaw, left *

-Shoulder girdle, left

-Upper arm, left

-Lower arm, left

Region 2: Right Upper Region

-Jaw, right *

-Shoulder girdle, right

-Upper arm, right

-Lower arm, right

 

 

Region 5: Axial Region

-Neck

-Upper back

-Lower back

-Chest *

-Abdomen *

 

 

Region 3: Left Lower Region

-Hip (buttock, trochanter), left

-Upper leg, left

-Lower leg, left

Region 4: Right Lower Region

-Hip (buttock, trochanter), right

-Upper leg, right

-Lower leg, right

 

 

 

* Not included in generalized pain definition

 

2)    SS scale score

The SS scale score is the sum of the severity of the 3 symptoms (fatigue, waking unrefreshed, cognitive symptoms) plus the sum of the number of 3 symptoms (headaches, pain or cramps in lower abdomen, depression)

(The final score is between 0 and 12)

 

- For the each of the 3 symptoms below, indicate the level of severity over the past week using the following scale:

0 = no problem

1 = slight or mild problems, generally mild or intermittent

2 = moderate, considerable problems, often present and/or at a moderate level

3 = severe: pervasive, continuous, life-disturbing problems

Fatigue                          (0-3)

Waking unrefreshed     (0-3)

Cognitive symptoms     (0-3)

 

- During the previous 6 months indicate the number of the following symptoms the patient has been bothered by:

  - Headaches                                         (0-1)

  - Pain or cramps in lower abdomen     (0-1)

  - Depression                                         (0-1)

 

The fibromyalgia severity (FS) scale is the sum of the WPI and the SS scale

 

AAPT Diagnostic Criteria

 

In an attempt to improve the recognition of fibromyalgia in clinical practice, the AAPT fibromyalgia working group proposed new diagnostic criteria in 2018 (62). These criteria are similar to the ACR criteria as they require the pain to be generalized (multisite), require the presence of non-pain symptoms and require the symptoms to be present for at least 3 months (Table 5). These diagnostic criteria are more simple than the ACR criteria and they can be easily implemented in primary clinical practice, but some of their aspects have been criticized (63). According to the AAPT criteria the head, the abdomen and the chest are included in the areas that are assessed for the presence of generalized musculoskeletal pain. However, these regions are problematic since pain originating from the teeth, the heart and the bowel can be referred to these areas. Additionally, the AAPT criteria do not have the ability to quantify the severity of fibromyalgia as, apart from the generalized pain, they only assess the presence or absence of the 2 most common non-pain symptoms of fibromyalgia, abolishing all other somatic symptoms. Compared with the 2016 ACR diagnostic criteria, individuals with less symptom severity and fewer pain sites can be classified as fibromyalgia patients, when the AAPT diagnostic criteria are used (64).

 

Table 5. AAPT Diagnostic Criteria

Criteria:

1)    Multisite pain defined as 6 or more pain sites from a total of 9 possible sites:

- Head

- Left arm

- Right arm

- Chest

- Abdomen

- Upper back and spine

- Lower back and spine, including buttocks

- Left leg

- Right leg

2)    Moderate to severe sleep problems or fatigue

3)    Multisite pain plus fatigue or sleep problems must have been present for at least 3 months

 

NOTE. The presence of another pain disorder or related symptoms does not rule out a diagnosis of fibromyalgia. However, a clinical assessment is recommended to evaluate for any condition that could fully account for the patient’s symptoms or contribute to the severity of the symptoms.

 

EPIDEMIOLOGY

 

The prevalence of fibromyalgia depends on the criteria used to define it. Most studies use either the 1990 ACR classification criteria or the modified 2010 ACR preliminary diagnostic criteria (2011 ACR survey criteria) and the prevalence varies between 2-4% (65). Using the 2016 ACR diagnostic criteria the prevalence of fibromyalgia in the general population is 3-4% while with the AAPT diagnostic criteria the prevalence of fibromyalgia is 73% higher, ranging from 5% to 7% (64). In the general population the prevalence increases with age from 2% at the age of 20 to 8% at age of 70. Fibromyalgia appears more often in relatives of patients suffering from fibromyalgia (66), whereas there is a significant difference on the women to men ratio depending on the criteria used to define fibromyalgia. When the 1990 ACR classification criteria are used the women to men ratio is 7:1 while when the 2011 ACR survey criteria are used, that do not use the tender point count, the ratio ranges from 4:1 to 1:1 (58,67–69). Using the 2016 ACR or the AAPT diagnostic criteria there is no statistically significant difference in the prevalence of fibromyalgia between women and men (64).

 

DIFFERENTIAL DIAGNOSIS

 

Several conditions can mimic or overlap with fibromyalgia. In order to reach a differential diagnosis, careful history taking should be followed by a thorough physical examination. Careful neurologic and musculoskeletal examination needs to be performed in all fibromyalgia patients in order to exclude the presence of such conditions. Mood and functional impairment should also be evaluated. This can be easily performed using simple self-administered questionnaires. Patients with obvious mood disturbances should have a formal evaluation by a mental health professional. Baseline blood tests should be limited to a complete blood count, erythrocyte sedimentation rate, a comprehensive metabolic panel, and thyroid function tests. These tests are usually normal in fibromyalgia patients. Consequently, the identification of abnormalities in any of these examinations might suggest that a different condition is present. Additional tests are not recommended for a diagnosis, unless they are clinically indicated. The disorders that can mimic and/or overlap with fibromyalgia and the characteristic clinical features which differentiate them from fibromyalgia are described in Table 6. The clinical features of fibromyalgia, chronic fatigue syndrome (systemic exertion intolerance disease), depression, migraine, and irritable bowel syndrome often overlap being so interchangeable that some authors consider that these conditions should be approached as a “spectrum” of associated disorders (10). They are also considered as part of the spectrum of post-traumatic stress disorder (70,71).

 

 

Table 6. Disorders that can Mimic and/or Overlap with Fibromyalgia Along with Characteristic Clinical Features that Differentiate Them from Fibromyalgia

Disorders

Differentiating clinical features

Rheumatoid arthritis, Systematic Lupus Erythematosus, Sjögren’s syndrome

·      Characteristic synovitis and systemic features of connective tissue disease, apart from musculoskeletal pain, fatigue, Raynaud phenomenon, dry eyes and dry mouth, are usually not features of fibromyalgia.

·      Routine serologic tests are not recommended because of low positive predictive value.

Ankylosing spondylitis, other inflammatory back conditions

·      Generally, there is normal spinal motion in fibromyalgia.

·      Characteristic radiologic features of these disorders are not present in fibromyalgia.

Polymyalgia rheumatica

·      Tender points are not always present in polymyalgia rheumatica.

·      Stiffness is more prominent than pain in polymyalgia rheumatica.

·      Most patients with polymyalgia rheumatica have increased erythrocyte sedimentation rate, while it is normal in fibromyalgia.

·      Patients with polymyalgia rheumatica respond extremely well to modest doses of corticosteroids, in contrast to fibromyalgia patients.

Inflammatory myositis, metabolic myopathies

·      Myositis and myopathies can cause muscle weakness and muscle fatigue, but they are not usually associated with diffuse pain.

·      Patients with myositis or myopathies have abnormal muscle enzyme tests and specific histopathologic findings on muscle biopsy, in contrast to fibromyalgia patients (muscle biopsy should be limited to cases that there is clinical evidence suggestive of myositis or myopathy).

Statin myopathy

·      Statin myopathy symptoms are limited to muscle weakness and pain without other symptoms associated with fibromyalgia.

·      Statin myopathy pain is temporally associated with statin therapy.

·      Statin myopathy can be associated with abnormal muscle enzyme tests.

Infection:

chronic viral infection (e.g., infectious mononucleosis, HIV, HTLV, HBV, HCV, Lyme disease), long COVID

·      In fibromyalgia patients there is no objective evidence of inflammation or organ system dysfunction

Hypothyroidism

·      Although thyroid autoantibodies are common in fibromyalgia patients, thyroid function tests are usually normal.

Hyperparathyroidism

·      Hypercalcemia is not present in fibromyalgia.

Cushing’s syndrome

·      Cushing’s syndrome is associated with muscle weakness rather than pain.

·      The characteristic facial and skin signs of Cushing’s syndrome are not present in fibromyalgia.

Adrenal insufficiency

·      Adrenal insufficiency causes severe exhaustion, while it is not typically associated with chronic widespread pain.

Hypophosphatasia

·      Most hypophosphatasia patients have low alkaline phosphatase

Neurologic diseases:

peripheral neuropathies, cervical radiculopathy, entrapment syndromes (e.g., carpal tunnel syndrome), multiple sclerosis, myasthenia gravis

·      Multiple sclerosis and myasthenia gravis are associated with post-exercise muscle and generalized fatigue, but not with widespread pain.

·      Thorough neurologic examination can reveal neurologic signs characteristic of specific diseases.

Myofascial pain syndromes (they may include other common regional pain disorders such as tension headaches, occupational overuse syndrome, cumulative trauma disorder, work related musculoskeletal disorder,idiopathic low back and cervical strain disorders, chronic pelvic pain temporomandibular disorderand interstitial cystitis)

·      In myofascial pain syndromes the pain and the tenderness is confined in one anatomic region

Chronic fatigue syndrome

(systemic exertion intolerance disease)

·       Criteria for the diagnosis of chronic fatigue syndrome:

 

1.     According to the modified United States Centers for Disease Control and Prevention chronic fatigue syndrome is diagnosed when two criteria are fulfilled (72):

i)     Clinically evaluated, unexplained, persistent or relapsing fatigue that is of new or definite onset; is not the result of ongoing exertion; is not alleviated by rest; and results in substantial reduction in previous levels of occupational, educational, social, or personal activities

ii)    Four or more of the following symptoms that last six months or longer:

-    Impaired memory or concentration

-    Post-exertional malaise where physical or mental exertions bring on "extreme, prolonged exhaustion and sickness"

-    Unrefreshing sleep

-    Muscle pain

-    Arthralgia in multiple joints

-    Headaches of new kind or greater severity

-    Frequent or recurring sore throat

-    Tender cervical or axillary lymph nodes

 

2.     According to the proposed diagnostic criteria of the United States Institute of Medicine the chronic fatigue syndrome (systemic exertion intolerance disease) is diagnosed when two criteria are fulfilled (73):

i)    All of the following symptoms:

-    A substantial reduction or impairment in the ability to engage in pre-illness levels of occupational, educational, social, or personal activities that persists for more than 6 months and is accompanied by fatigue, which is often profound, is of new or definite onset (not lifelong), is not the result of ongoing excessive exertion, and is not substantially alleviated by rest

-    Post-exertional malaise*

-    Unrefreshing sleep*

ii)   Two or more of the following manifestations:

-    Cognitive impairment*

-    Orthostatic intolerance

-     

*   The diagnosis should be questioned if patients do not have these symptoms at least half of the time with moderate, substantial, or severe intensity.

 

·       Chronic widespread pain is not included in the criteria for diagnosis of chronic fatigue syndrome

Psychiatric disorders:

depression, anxiety disorders, posttraumatic stress disorder

·       In fibromyalgia patients with a concurrent psychiatric disorder, the attribution of symptoms to fibromyalgia or the psychiatric disorder is not always possible.

Sleep disorders:

obstructive sleep apnea, restless legs syndrome, periodic limb movement disorders

·       Detail history can identify the majority of the primary sleep disorders.

·       Chronic widespread pain is uncommon in primary sleep disorders.

Irritable bowel syndrome

·       According to the 2009 American College of Gastroenterology recommendations for the diagnosis of irritable bowel syndrome, it is defined by abdominal pain or discomfort that occurs in association with altered bowel habits over a period of at least three months (74).

Temporomandibular disorders

·       Temporomandibular disorders are characterized by recurrent facial/jaw pain and/or limitation in jaw opening occurring in the past six months.

Tension – Migraine headache

·       Tension – migraine headache is characterized by recurrent headaches (at least five for migraine, at least 10 for tension-type) lasting 30 minutes.

Interstitial cystitis

·       According to the American Urological Association guidelines interstitial cystitis is defined as an unpleasant sensation (pain, pressure, discomfort) perceived to be related to the urinary bladder, associated with lower urinary tract symptoms of more than six weeks duration, in the absence of infection or other identifiable causes (75).

HIV: human immunodeficiency virus, HTLV: human T-lymphotropic virus, HBV: hepatitis B virus, HCV: hepatitis C virus, COVID: coronavirus disease

 

PATHOPHYSIOLOGICAL MECHANISMS

 

Pain sensitivity in the population occurs over a wide continuum, forming a classic bell-shaped curve, just like any other physiologic variable. Genetic predisposition in combination with environmental factors, determine the place that each individual takes in this continuum. People who are placed in the right end of this curve are very sensitive to pain and they can probably develop pain even without having any inflammation or damage in the peripheral tissues. This pain can be either regional or widespread (39).

 

In the past fibromyalgia was thought to be a primary muscle disease. However, controlled studies found no evidence of significant pathologic or biochemical muscle abnormalities that can be the cause of chronic widespread pain and tenderness. Most investigators believe that any muscle pathology is secondary to chronic pain and inactivity, rather than primary in nature (76–79). Current research suggests that altered central nervous system (CNS) physiology might underlie the symptoms of fibromyalgia. Abnormal central sensory processing of pain signals seems to play a significant role in the pathogenesis of fibromyalgia. This dysregulation of the nociceptive system can arise from a combination of interactions between neurotransmitters, cytokines, hormones, the autonomic nervous system, behavioral constructs, and external stressors.

 

Abnormalities in Sensory Processing

 

Fibromyalgia overlaps with several other similar syndromes including chronic fatigue syndrome (systemic exertion intolerance disease) and myophasial pain syndrome (Table 6). It has been proposed that these disorders should be considered as members of the central sensitivity syndromes (Table 7). These similar and overlapping syndromes are bound by the common mechanism of central sensitization that involves hyper-excitement of the second-order neurons, especially the wide-dynamic-range neurons (WDR) in the dorsal horns of the spinal cord,  by various synaptic and neurotransmitter/neuromodulator activities (6). Central sensitization is clinically and physiologically characterized by hyperalgesia, allodynia, expansion of the receptive field (pain expanding beyond the area of the peripheral nerve supply, after the application of a nociceptive stimulus), prolonged electrophysiological discharge, and an after-stimulus unpleasant quality of the pain (e.g., burning, throbbing, tingling or numbness). Parallel to central sensitization, temporal summation takes place in the second-order neurons. It is characterized by a progressive increase in electrical discharges (and consequently increase in the perceived intensity of pain) in response to repetitive short noxious stimuli. Temporal summation involves the production of second pain, which is described as dull or burning, and leaves an after-stimulus unpleasant sensation (80).

 

Table 7. Central Sensitivity Syndromes

·       Fibromyalgia

·       Chronic fatigue syndrome (systemic exertion intolerance disease)

·       Irritable bowel syndrome

·       Tension type headaches

·       Migraine

·       Temporomandibular disorder

·       Myophasial pain syndrome

·       Restless leg syndrome

·       Periodic limb movements in sleep

·       Primary dysmenorrhea

·       Interstitial cystitis

·       Posttraumatic stress disorder

 

N-methyl-D-aspartate (NMDA) receptors are mostly responsible for escalation of hyperexcitability of the second-order nociceptive neurons. The role of the major neurotransmitters of the nociceptive system that participate in signal conduction at the level of the spinal cord is briefly illustrated in Figure 3 (6).

 

Figure 3. The role of the major neurotransmitters of the nociceptive system that participate in signal conduction at the level of the spinal cord. SP: Substance P, NGF: nerve growth factor, NMDA: N-methyl-D-aspartate, D: dopamine.

 

The second-order neurons have ascending projections to the thalamus, hypothalamus, the limbic system and the somatosensory cortex. These supraspinal structures are involved in the sensory, evaluative and affective dimensions of pain (e.g., unpleasantness, emotional reaction). Several descending pathways from the cortico-reticular system, locus ceruleus, hypothalamus, brain stem, and local spinal cord interneurons terminate to the dorsal horn cells. These pathways utilize neurotransmitters that include serotonin (5-HT), norepinephrine, γ-amino-butyric acid (GABA), enkephalins and adenosine (6). This descending system, once thought to be predominantly inhibitory, is now known to have a facilitatory potential (81). Evidence suggests that the 5-HT3 receptor has a facilitatory function, while the 5H-T1Areceptor is inhibitory. The ascending and descending pathways should not be considered as dichotomous in function. They are interactive and their functions are bidirectional. Both pathways can either facilitate or inhibit pain, depending on the site of action and the neurotransmitters that are used (6).

 

The dysregulation of the nociceptive system, either at the level of the dorsal horns of the spinal cord, or at the level of the ascending and descending pathways, can lead to its hyper-excitability. In other words, it can lead to central sensitization. Several factors may amplify and sustain central sensitization through interactive and synergistic actions. These factors are summarized in Table 8 (80). Central sensitization can become self-sustained, even when the event that triggered it no longer exists, due to long-term CNS plasticity.

 

Table 8. Factors that may Amplify and Sustain Central Sensitization

·       Genetics

·       Sympathetic over-activity

·       Endocrine dysfunctions

·       Viral infection

·       Peripheral nociception generators (e.g., arthritis)

·       Poor sleep

·       Environmental stimuli (e.g., weather, noise, chemicals)

·       Psychological distress (e.g., adverse childhood experience)

 

Neuroimaging studies provide moderate evidence for structural changes in the brain of patients with fibromyalgia. Gray matter volume appears to be reduced in areas related with pain processing, such as the cingulate, the insular, and the prefrontal cortices (82). Functional MRI studies reveal alterations in the functional connectivity of brain areas responsible for pain processing and provide support of functional dysregulation of the ascending and descending pain pathways in fibromyalgia patients (82–85). Additionally, alterations in neuronal activity between the ventral and the dorsal spinal cord have been demonstrated in fibromyalgia patients (86).

 

Although nearly all of the research on sensory processing in fibromyalgia has focused on the processing of pain, there are some data suggesting a more generalized disturbance in sensory processing. There is evidence that fibromyalgia patients have a hypersensitivity to unpleasant stimuli of other sensory systems. For example, many patients experience reduced tolerance to loud noises, bright lights, odors, drugs, and chemicals (87,88).

 

Neurotransmitters

 

The levels of Substance P (SP) in the cerebrospinal fluid (CSF) in patients with fibromyalgia are significantly increased compared to normal individuals, whereas CSF levels of serotonin metabolites are decreased, as are metabolites of dopamine and norepinephrine (89).

 

The first direct evidence that fibromyalgia patients may have abnormal dopamine response to pain originated from positron emission tomography (PET) competitive binding studies using the D2/D3 receptor antagonist [11C] raclopride. It was shown that dopamine is released in response to tonic toxic noxious muscle stimulation, but not after non-painful muscle stimulation in healthy human subjects. In contrast the dopamine response in fibromyalgia patients did not differ between painful and non-painful muscle stimulation (87). There are indications that disturbances of the opioidergic system occur in fibromyalgia patients, as there is an up-regulation of opioid receptors in the periphery, with a reduction of the brain opioid receptors (90,91). This implies an increased baseline endogenous opioidergic activity. Opioids can activate glial cells, via a non-stereoselective activation of toll-like receptor 4 (TLR4). Glial cells in turn can mediate pain by releasing neuroexcitatory, pro-inflammatory products (92).

 

In a study where fibromyalgia patients were evaluated for cortical excitability and intracortical modulation using transcranial magnetic stimulation of the motor cortex, it was shown that there were deficits in intracortical modulation of GABAergic and glutamatergic mechanisms (93). Diminished inhibitory neurotransmission resulting from lower concentrations of GABA within the right anterior insula of patients with fibromyalgia was documented using proton magnetic resonance spectroscopy (94). Evidence for enhanced glutaminergic neurotransmission in fibromyalgia patients is derived from studies that used magnetic resonance spectroscopy. It was shown that fibromyalgia patients have significantly higher levels of glutamine within the posterior insula and in the right amygdala (95,96). The levels of brain-derived neurotrophic factor, which is involved in neuronal survival and synaptic plasticity of the central and peripheral nervous system, have been found to be increased both in the brain and in the plasma of fibromyalgia patients (97).

 

Cytokines

 

Although fibromyalgia is not considered an inflammatory disorder, the interaction of immunological mechanisms with pain physiology, has led to the identification of alterations in the levels of various cytokines (98). However, it is not clear whether cytokine changes are the cause of pain in these patients, or just its consequence.

 

The serum levels of interleukin 1 receptor antibody (IL-1Ra), IL-6, and IL-8, and the plasma levels of IL-8 are higher in fibromyalgia patients, compared to controls (99). Inflammatory cytokines such as IL-1β, IL-6 and tumor necrosis factor alpha (TNFα) have been detected in skin biopsies taken from fibromyalgia patients, possibly indicating an element of neurogenic inflammation (100). Lower levels of the anti-inflammatory cytokines IL-4 and IL-10 have been reported in fibromyalgia patients compared to healthy controls (101,102). However, the interpretation of cytokine levels is not always easy. Most cytokines are expressed in low levels and a sensitive bioassay is needed for their detection. Consequently, a negative result can merely be the ramification of a not sensitive enough method. Additionally, cytokine levels can be vigorously influenced by a number of factors, including circadian rhythmicity, physical activity, and co-morbid conditions, including depression.

 

Inflammatory cytokines like IL-1β, IL-6 and TNFα can elicit pain, induce hyperalgesia and they are associated with neuropathic pain (103), although they do not appear to be involved in “normal” pain. Although serum cytokines do pass the blood brain barrier the release of pro-inflammatory cytokines by immune cells in the body leads, in turn, to release of pro-inflammatory cytokines by glial cells within the brain and spinal cord (104).Inflammatory cytokines like IL-1β, IL-6 and TNFα can also cause activation of the hypothalamic-pituitary-adrenal (HPA) axis alone, or in synergy with each other. There is evidence to suggest that IL-6, which is the main endocrine cytokine, plays the most significant role in the immune stimulation of the axis, especially in chronic inflammatory stress (105). IL-6 can stimulate the hypothalamic secretion of corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP), leading to the increase of serum adrenocorticotropic hormone (ACTH) and cortisol levels (106).

 

Hypothalamic-Pituitary-Adrenal Axis

 

There is substantial data supporting an abnormal function of the HPA axis in fibromyalgia. However, the results from studies on the HPA axis function in fibromyalgia patients are relatively heterogeneous and partially contradictory (107). The 24-hour urinary free cortisol (UFC) has been found to be reduced or normal (108). Findings regarding alterations in diurnal variation of cortisol secretion are also inconsistent. Although normal diurnal patterns of ACTH and cortisol have been reported, there is data demonstrating flattened cortisol diurnal rhythm with normal morning peak and higher evening cortisol, levels (109). It has been demonstrated that there is a significant decrease in the rate of decline from acrophase (peak) to nadir for diurnal cortisol levels in fibromyalgia patients, compared to controls, while there is no change in the ACTH to cortisol ratio (108). This implies a decreased ability of the HPA axis to return to baseline after a physiologic stimulation by meals, several other activities or even pain (108). Decreased morning cortisol release and reduced frequency of cortisol pulses over 24 hours have also been reported. There is evidence to suggest  that the reduced cortisol release in fibromyalgia patients is associated with depressive symptoms and experiences of childhood trauma (109).

 

In line with studies suggesting reduced adrenal output in fibromyalgia patients, reduced cortisol secretion has been reported in response to pharmacological challenge with synthetic ACTH1-24 and  insulin tolerance test (109). Patients with fibromyalgia exhibit increased ACTH, but normal cortisol response to CRH stimulation, compared to controls. This finding suggests a sensitization of the pituitary in combination with a degree of adrenal insufficiency (109). Arginine vasopressin (AVP), an ACTH secretagogue, has been found to be more increased in response to the postural challenge test in fibromyalgia patients, compared to controls (110). Alterations in the feedback regulation of HPA axis have also been reported in fibromyalgia patients, using the overnight dexamethasone suppression test (DST). Increased rates of non-suppression following the standard (1 mg) DST have been reported in fibromyalgia patients, compared to controls, but this finding was difficult to be interpreted as it was associated with depression. Interestingly, other studies have revealed lower rates of non-suppression in fibromyalgia patients (111).

 

There are indications that there is a dissociation between total and free cortisol levels in fibromyalgia patients, with normal salivary and plasma free cortisol despite diminished total cortisol levels. One possible explanation of this finding is a reduced concentration of the glucocorticoid binding globulin (CBG). Reduced levels of CBG have been reported in fibromyalgia patients compared to controls. It is of particular interest that chronic social stress might result in reduced CBG levels, whereas IL-6 and IL-1β that can also inhibit the production of CBG may contribute further (109). Apart from HPA axis abnormalities in fibromyalgia patients, abnormal levels of growth hormone have also been found in some, but not all reports (112). On the other hand the levels of sex hormones have not been clearly shown to differ between female fibromyalgia patients and controls (113).

 

Autonomic Nervous System

 

Sympathetic hyperactivity, often associated with sympathetic hypo-activity in response to stressors, or parasympathetic underactivity has been described in fibromyalgia. Attenuated sympathetic and parasympathetic activity was demonstrated in a study where fibromyalgia patients and healthy controls were assessed for 24 hours in a controlled hospital setting, including relaxation, a test with prolonged mental stress and sleep. The urinary catecholamine levels were found to be lower in fibromyalgia patients compared to controls. Patients with fibromyalgia had significantly lower adrenaline levels during the night and the second day of the study, and significantly lower dopamine levels during the first day, the night, and the second day. Furthermore, heart rate during relaxation and sleep was significantly higher in patients than in controls (114). In another study, plasma catecholamines, ACTH, and cortisol levels were reduced in 16 fibromyalgia patients compared to 16 healthy controls while performing static knee extension until exhaustion (115). Nocturnal heart rate variability indices have been shown to be significantly different in fibromyalgia women compared to healthy individuals, indicating a sympathetic predominance (116).  In addition, orthostatic hypotension and increased pain in response to tilt table test have been described along with increased resting supine heart rate and decreased heart rate variability  (117,118). IL-6 administration causes exaggerated norepinephrine responses and increases in heart rate, as well as delayed ACTH release, suggesting an incapacitated stress-regulating system (119). In-vitro testing of beta adrenergic receptor mediated cyclic AMP generation has revealed decreased responsiveness to beta-adrenergic stimulation (120).

 

Overall, it has been suggested that sympathetic dysfunction can not only cause diffuse pain, but also contribute to other symptoms like sleep disturbances, due to sustained nocturnal sympathetic activity, and fatigue, due to deranged sympathetic response to stress (6).

 

Psychological, Cognitive, and Behavioral Factors

 

Pain apart from a sensory-discriminative dimension, which includes the location and the intensity of pain, has a very significant psychological component. This includes the affective dimension of pain, the emotional valance of pain in other words, as well as attention and cognitive aspects, which are based on CNS mechanisms. Emotion and selective attention can enhance pain perception, with the involvement of the descending pathways that have a facilitatory effect on the spinal cord dorsal horn neurons (80). Catastrophizing has been shown to be related to decreased pain threshold and tolerance to heat stimuli in fibromyalgia patients. However there is a subgroup of fibromyalgia patients that is very tender, despite the fact that they do not catastrophize and they have a moderate control over their pain (11). A fMRI study has shown that although depression is associated with the magnitude of neuronal activation in brain regions that process the affective-motivational dimension of pain, neither the extent of depression nor the presence of comorbid major depression modulated the sensory-discriminative aspects of pain processing in fibromyalgia patients (121). Catastrophizing, has been associated with increased activity in brain areas related to anticipation, attention and the emotional aspects of pain, as shown by fMRI in response to pressure stimuli. This study also revealed an association between catastrophizing and increased activity in the secondary somatosensory cortex, indicating that the way patients think about their pain might actually influence its sensory processing (80).

 

Genetic Predisposition

 

It is currently well established that familial aggregation is a characteristic of fibromyalgia. First degree relatives of fibromyalgia patients are 8.5 times more likely to have fibromyalgia than relatives of patients with rheumatoid arthritis (12). As with other complex and multifactorial syndromes, the occurrence of familial aggregation in the case of fibromyalgia does not necessarily imply a genetic basis. Shared environmental factors and learned patterns of behavior that may evolve within families are equally valid targets of investigation.

 

Genome-wide association studies have shown significant differences in allele frequencies between fibromyalgia patients and controls. However many of the results are inconsistent, without being replicated. Additionally the small sample size of these studies limits the genetic variants that can be identified only to those with large effects (122–126). The coexistence of other comorbidities in fibromyalgia patients further obscures these results. In a large scale genome-wide association study of 26,749 individuals the overall estimated heritability of fibromyalgia was 14%. There was a significant difference between age groups, with the heritability in individuals less than 50 years of age to be 23.5%, while in those over 60 years of age it was only 7.5% (126).

 

Although no specific candidate gene has been identified, the following genes have been associated with fibromyalgia:

 

SEROTONIN TRANSPORTER (5-HTT) GENE

 

An increased frequency of the S/S genotype of the 5-HTT gene has been found in fibromyalgia patients compared to controls (127,128). However this putative association may be limited to patients with concomitant affective disorders, since it was not confirmed in fibromyalgia patients without depression or anxiety (129).

 

D4 RECEPTOR GENE

 

Polymorphisms affecting the number of repeats in the third cytoplasmic loop of the dopamine D4 receptor gene have been shown to be significantly decreased in frequency in fibromyalgia patients (130).

 

CATECHOL-O-METHYL TRANSFERASE (COMT) GENE

 

The homozygous low activity (met/met) and the heterozygous low activity (val/met) COMT genotypes occur more often in fibromyalgia patients than in controls, whereas the homozygous high activity (val/val) genotype is less frequent (131). However in a meta-analysis COMT gene val(158)met polymorphism was not associated with an increased risk for fibromyalgia (132). The met/met genotype has been associated with greater fibromyalgia illness severity across the domains of pain, fatigue, sleep disturbance, and psychological distress, while fibromyalgia patients with the met/met polymorphism experienced a greater decline in exhibiting a positive attitude on days when pain was elevated than did patients with the val/met or val/val genotype (133).

 

OPIOID RECEPTOR μ 1 GENE (OPRM1)

 

The 118G allele frequency has been described to be significantly lower in patients with fibromyalgia than in the control group (134).

 

ADRENERGIC RECEPTOR GENES

 

The presence fibromyalgia and its symptom severity is associated with various adrenergic receptor gene polymorphisms (135).

 

Other genes associated with the regulation of nociceptive and analgesic neuronal pathways

Specific variants of trace amine-associated receptor 1 (TAAR1) gene, regulator of G-protein signaling 4 (RGS4) gene, cannabinoid receptor 1 (CNR1) gene, and glutamate receptor, ionotrophic, AMPA 4 (GRIA4) gene, have been associated with fibromyalgia (123).  

 

External Stressors

 

Almost all diseases are caused by a combination of genetic predisposition and the effect of environmental factors. We are now beginning to better understand the environmental factors that seem to be important in triggering fibromyalgia. Most of them act as “stressors” that when superimposed onto a deranged stress-response system can lead to the dysregulation of the nociceptive system.

 

PERIPHERAL PAIN SYNDROME

 

Pain due to damage or inflammation of peripheral tissues may trigger fibromyalgia. Additionally, small fiber neuropathy can be associated with fibromyalgia (136–138). Chronic localized – regional pain can lead to central sensitization and pain dis-inhibition, causing pain hypersensitivity and widespread pain. Systematic autoimmune diseases can be associated with fibromyalgia too. Approximately 20-25% of patients with rheumatoid arthritis, systemic lupus erythematosus and ankylosing spondylitis, have co-morbid fibromyalgia (18). In such cases, it is important to realize that many symptoms may be attributed to fibromyalgia rather than the underlying disorder. This recognition has significant clinical implications.

 

INFECTIONS

 

Various infections have been linked to the development of fibromyalgia and chronic fatigue syndrome (systemic exertion intolerance disease). Epstein-Barr virus, parvovirus, Lyme disease, Q fever, HIV and hepatitis C virus (HCV), have been suggested as triggers of fibromyalgia or chronic fatigue syndrome (systemic exertion intolerance disease), but more robust evidence is needed. The role of vaccination in precipitating fibromyalgia and related syndromes is still not clear (139,140).

 

PHYSICAL TRAUMA

 

Various forms of physical trauma have been considered as culprits of triggering the pathogenesis of fibromyalgia. Many patients report the initiation or the exacerbation of their symptoms after a traumatic event such as whiplash injury, while increased rates of fibromyalgia have been demonstrated among patients undergoing cervical trauma during motor vehicle accidents (141,142).

 

PSYCHOLOGICAL DISTRESS

 

It has been considered that psychological factors that give rise to chronic stress may initiate the chain of events that leads to fibromyalgia. The chronic stress can be a result of the accumulation of daily stress events. Emotional stress, catastrophic events such as war, job loss, marital discord and excess family responsibilities such as caring for sick elders, have been implicated as triggers of fibromyalgia (143). However the data that supports the notion that psychological stress and distress directly causes fibromyalgia is rather weak (39).

 

MANAGEMENT

 

The treatment of fibromyalgia is challenging because of our limited understanding of its pathogenesis and the poor response of patients to conventional pain treatments. The aim of the therapy is to relieve pain and increase function using a multimodal individualized therapeutic strategy which, in most cases, includes pharmacologic and non-pharmacologic interventions. Current clinical-based evidence supports the use of a multimodal program that includes education, exercise, cognitive-behavioral approaches and medications. The treatment should be individualized based on the symptoms, the comorbidities and the preferences of the patient, who should be encouraged to participate in the decision-making process of selecting the optimal therapies (144,145). Coexisting disorders are common in fibromyalgia patients. Their identification and effective treatment can have beneficial effects on fibromyalgia symptoms. It is also important to assure that adequate adherence to both pharmacological and non-pharmacological treatment is maintained, so as to achieve the optimal benefit from these treatments.

 

Non-Pharmacological Management

 

PATIENT EDUCATION

 

The first step should be the education of the patient. The patients with fibromyalgia need to understand their illness before any treatment modality is used (146). Providing a diagnosis, “labeling” the patient with fibromyalgia, may have beneficial effects. It has been shown that fewer symptoms and an improvement in health status is noted after the patients are informed of their diagnosis (147,148). The physician should clarify that fibromyalgia is a real illness and the symptoms the patient experiences are not imaginary. The role of neurotransmitters and neuromodulators in pain perception, fatigue, abnormal sleep and mood disturbances should be discussed, so as the patient to understand the rationale of the pharmacologic therapy, especially when antidepressant drugs are used. The significance of good sleep hygiene should be reviewed and poor sleep habits should be addressed. Fibromyalgia patients who are overweight or obese should be informed for the adverse effect of increase body mass index to fibromyalgia symptoms and quality of life (149). For these patients weight reduction should be encouraged. The patient also needs to acknowledge that fibromyalgia is a chronic relapsing condition without though being life-threatening nor deforming.

 

EXERCISE

 

Another potent non-pharmacological treatment for fibromyalgia is exercise. It has been reported that an exercise program incorporating aerobic, strengthening, and flexibility elements can lead to greater benefits than a relaxation program. Exercise in fibromyalgia patients should have two major components: strengthening to increase soft-tissue length and joint mobility, and aerobic conditioning to increase fitness and function, reduce fibromyalgia symptoms and improve quality of life (144,150–153). Exercise should be of low impact and of sufficient intensity so as to be able to change aerobic capacity (28). Successful interventions include fast walking, biking, swimming, water aerobics, tai chi, and yoga. Land and aquatic training appears to be equally beneficial (154). An improvement in the severity of fibromyalgia symptoms has also been achieved with web-based exercise programs (155). A gradual incremental increase in physical activity should be encouraged as it is common for fibromyalgia patients to initially perceive an aggravation of their pain and fatigue at the beginning of a training program. It has been suggested that in the presence of exercise-induced pain, the intensity and duration of exercise should be reduced, while its frequency should be maintained, so as to avoid any further decrease in exercise tolerance (144). The type and intensity of the exercise program should be individualized and should be based upon patient preference and the presence of any other cardiovascular, pulmonary, or musculoskeletal comorbidities.

 

COGNITIVE-BEHAVIORAL APPROACHES

 

One of the goals of the management should be to help patients understand the effect of thoughts, beliefs and expectations on their symptoms. This can help them to abolish the perception of helplessness and the catastrophizing thoughts that can adversely influence their condition. Patients with greater self-efficacy are more likely to have a good response to treatment programs and experience better outcomes. The beneficial effect of cognitive-behavioral therapies in fibromyalgia patients with anxiety and depression disorders is limited to a reduction of negative mood, while the rest of the patients also demonstrate a reduction of pain and fatigue. Psychologically based interventions, have been proven to be useful when they are compared to no treatment or treatment other than aerobic exercise (156). In a 2021 systematic review and meta-analysis there was high quality evidence that cognitive-behavioral therapy can significantly reduce the pain intensity in fibromyalgia patients for 3 months (157). Preliminary data from functional MRI studies suggest that cognitive-behavioral therapies have the ability to restore the alterations in the functional connectivity of brain areas responsible for pain processing observed in fibromyalgia patients (158,159).

 

COMPLEMENTARY AND ALTERNATIVE APPROACHES

 

Acupuncture

 

Acupuncture is the insertion of needles in the human body. There are different styles of acupuncture depending on the location and the depth the needles are inserted. The inserted needles can be stimulated by heat, electrical current (electro-acupuncture), mechanical pressure (acupressure), or laser (laser acupuncture). The most common type of acupuncture involves skin penetration without stimulation (manual acupuncture).  Sham or fake acupuncture is a research tool to control the effects of real acupuncture. It can involve skin contact with the needles without actual penetration or needle insertion in areas other than the ones usually targeted.

 

In a high quality meta-analysis it was demonstrated that the effects of manual acupuncture on pain, sleep quality and global well-being did not differ significantly from the effects of sham acupuncture. On the contrary electro-acupuncture significantly reduced pain, fatigue, and stiffness, while it improved sleep quality and global well-being when compared to sham acupuncture. Additionally, electro-acupuncture significantly improved pain, stiffness, and global well-being when compared to non-acupuncture. The beneficial effects of acupuncture could be observed at 1 month after treatment, but they were not maintained at 6-7 months (160).

 

Other

 

The effectiveness of meditative movement therapies (qigong, yoga, tai chi) on sleep and fatigue improvement and of hydrotherapy on pain reduction has been supported by some studies (161,162). A number of other modalities has also been utilized for the treatment of fibromyalgia including biofeedback, chiropractic therapy, massage therapy, hypnotherapy, guided imagery, electrothermal therapy, phototherapeutic therapy, music therapy, journaling / storytelling, static magnet therapy, transcutaneous electrical nerve stimulation, and transcranial direct current stimulation. However there are no well-designed studies to advocate their general use (145).

 

Pharmacologic Treatment

 

A wide range of drugs has been used in the treatment of fibromyalgia including antidepressants, sedatives, muscle relaxants and antiepileptic drugs. The choice of medication is influenced by patient preference; prominence of particular symptoms, including fatigue, insomnia, and depression; potential adverse effects; patient tolerance of individual medications; cost and regulatory limitations on prescription choice (163,164). Nonsteroidal anti-inflammatory drugs and opioids, although often prescribed for fibromyalgia, are not an effective form of treatment (39,165). However analgesics and anti-inflammatory medications can be useful in case of coexisting conditions that cause regional pain, like arthritis, which can aggravate or trigger the fibromyalgia symptoms. Regarding opioids, with the exception of tramadol, apart from not being effective for the treatment of fibromyalgia symptoms, their long-term use also curries a dose-dependent risk for serious adverse effects, including overdose, abuse, fractures, myocardial infarction and sexual dysfunction (166). Additionally opioids in fibromyalgia patients can reduce the effectiveness of psychological therapy (167), while their long-term use can cause sleep disturbances (168).

 

Patients should be informed that for most pharmacologic therapies several weeks may be needed until they experience a benefit. Initially a single drug should be administered. However, in the case of non-responsiveness combination therapy should be considered. Since therapeutic responses are rarely durable, physicians should not be surprised when the initial efficacy of a medication is abolished. Successful treatment of fibromyalgia may require regular reassessment and possible rotation or combination of medications (169). Adequate dose prescription and patient adherence are significant for the effectiveness and tolerability of pharmacologic treatment (170). The doses of the most commonly used medications with strong and moderate evidence of effectiveness are shown in Table 9.

 

ANTIDEPRESSANTS

 

Tricyclic Antidepressants (TCAs)

 

TCAs are often used as initial treatment for fibromyalgia. Their analgesic effect is independent of their antidepressant action and is thought to be mediated by inhibition of norepinephrine (rather than serotonin) reuptake at spinal dorsal horn synapses, with secondary activity at the sodium channels. The most widely studied drugs of this group are amitriptylineand cyclobenzaprine. They should be administered at lower doses than those required for the treatment of depression, a few hours before bedtime, and their dose should be escalated very slowly. A clinically important improvement is observed in 25-45% of patients treated with TCAs compared to 20% in those taking placebo (171–175). However their use is limited by the fact that they are ineffective or intolerable in 60-70% of patients (144), while their efficacy may decrease over time (171,176).

 

Amitriptyline is more efficient compared to the serotonin-norepinephrine reuptake inhibitors duloxetine and milnacipran in reducing pain, sleep disturbance, and fatigue, without differences in acceptability, as it was shown in a systematic review and meta-analysis (177). In a 2022 network meta-analysis comparing amitriptyline, duloxetine and pregabalin it was shown that treatment with amitriptyline 25 mg was superior to duloxetine and pregabalin for the reduction of pain intensity for at least 50% (178). The combination of 20 mg of fluoxetine in the morning with 25 mg of amitriptyline at bedtime has been shown to be more effective than either medication alone (179). Side effects of amitriptyline include dry mouth, constipation, fluid retention, weight gain, difficulty in concentrating and possibly cardiotoxicity.

 

Cyclobenzaprine has a similar tricyclic structure and presumed mode of action with amitriptyline in fibromyalgia, but is thought to have minimal antidepressant effect (163). A meta-analysis of five placebo-controlled trials has revealed improvement of the global functioning, with a similar effect size as this reported for amitriptyline. The group that received cyclobenzaprine had a significant decrease in pain for 4 weeks, compared to those treated with placebo, but the decrease in pain was not significantly different after 8 and 12 weeks. Sleep was improved at all time points in both cyclobenzaprine and placebo groups, while no effect was noted on fatigue (171–173). It has been demonstrated that the use of very low-dose cyclobenzaprine (1 to 4 mg at bedtime) can improve the symptoms of fibromyalgia, including pain, fatigue, and depression, compared to symptoms at baseline and to placebo. Significantly more patients who received the very low-dose of cyclobenzaprine experienced improved restorative sleep, based upon analysis of cyclic alternating pattern of sleep by electroencephalography. The increase in nights with improved sleep by this measure correlated with improvements in fatigue and depression (180).

 

Desipramine has fewer anticholinergic and sedative effects than other TCAs, which can make it a possible alternative, although its efficacy is not well studied in fibromyalgia.

 

Serotonin-Norepinephrine Reuptake Inhibitors (SNRIs)

 

SNRIs are similar to TCAs in their ability to inhibit the reuptake of both serotonin and norepinephrine, but they differ from TCAs in being devoid of significant activity at other receptor systems, resulting in diminished side effects and increased tolerance. Venlafaxine, duloxetine and milnacipran have been shown to be effective in diminishing fibromyalgia symptoms (147,181,182). These drugs can be used in fibromyalgia patients who do not respond to a trial of low-dose TCAs or who have intolerable side effects. They can also be administered as an alternative to amitriptyline for initial therapy especially for patients with significant fatigue or depression. Of these medications duloxetine and milnacipranare better studied and they are preferred to be administered to patients with fibromyalgia. There are more limited data regarding the efficacy of venlafaxine for fibromyalgia, while withdrawal symptoms if a dose is missed occur more often, because of the short half-life of this medication (183). A meta-analysis has shown that fibromyalgia patients treated with duloxetine at 60mg daily are more likely to have more than 50% reduction in pain, compared to patients taking placebo (184). However, duloxetine at 30mg daily does not significantly reduce pain (185). The efficacy of duloxetine can be maintained at 3 and 6 months of treatment (186). In a 2018 systematic review and meta-analysis it was shown that duloxetine and milnacipran were not superior to placebo in the frequency of pain relief of at least 50%, but there was a benefit in reducing the pain at least by 30% and in the patient's global impression to be much or very much improved. Additionally, there was not a significant difference in the reduction of fatigue, in the reduction of sleep problems, nor in the improvement of health-related quality of life (187). Another meta-analysis has shown that duloxetine, pregabalin and milnacipran were superior to placebo for pain relief, while duloxetine and pregabalin were superior to milnacipran. These drugs also differed in their effects on sleep disturbances, depression and fatigue (188). A fMRI study in fibromyalgia patients treated with milnacipran and placebo demonstrated that pain reduction with milnacipran treatment was associated with decreased functional connectivity between the insular cortex and the rostral part of the anterior cingulate cortex as well as the periaqueductal gray, while these changes were not demonstrated with the placebo (189).Regarding their side effects, headaches and nausea are more common with duloxetine and milnacipran treatment, while diarrhea is more common with duloxetine treatment. Other side effects related to SNRIs include dry mouth, constipation, somnolence, dizziness and insomnia (188).

 

Monoamine Oxidase Inhibitors

 

Monoamine oxidase inhibitors block the catabolism of serotonin, increasing its levels in the brain. It has been indicated that pirlindole and moclobemide have a significant beneficial effect on pain, without a significant effect on sleep nor fatigue. Their use for the treatment of fibromyalgia patients is limited (190).

 

ANTICONVULSANTS

 

Antiepileptic medications useful for the treatment of fibromyalgia patients include pregabalin and gabapentin. Both of these medications are structurally related to GABA and they bind with high affinity to the alpha2-delta subunit site of cellular voltage-dependent calcium channels. Although their exact mechanism of action is unknown their therapeutic effects can be mediated by blocking the release of various neurotransmitters. They can be used in cases where other medications initially administered to the fibromyalgia patients become intolerable or ineffective, or as the initial treatment for patients with significant sleep disturbance in addition to pain.  

 

Pregabalin has been reported to be efficient against pain, sleep disturbances and fatigue in fibromyalgia. In a recent meta-analysis a reduction of at least 50% in pain intensity was found in 22-24% of patients taking pregabalin 300-600 mg per day, approximately 9% higher compared to the placebo group. A reduction in pain intensity of at least 30% was found in 39-43% of patients on pregabalin 300-600 mg per day, compared to 28% of patients taking placebo (191). In addition to pain, pregabalin at doses 300-600 mg per day can improve sleep and patient function, as it is demonstrated in a 2018 review of clinical trials, meta-analyses, combination studies and post-hoc analyses (192). The improvement in pain and sleep can be apparent as early as 1-2 days after the onset of treatment (193). In a 2022 network meta-analysis it was shown that treatment with pregabalin 450 mg per day was superior to duloxetine 30 mg for the reduction of pain intensity of at least 30% (178). A randomized placebo-controlled neuroimaging study demonstrated that the reduction in pain intensity from pregabalin was associated with a reduction in connectivity between the posterior insula and the default mode network (DMN) and that pregabalin but not placebo can reduce the response of the DMN to experimental pain (194). It is of interest that baseline patterns of brain connectivity have been used in a machine-learning model to successfully distinguish fibromyalgia patients who have a favourable response to pain intensity after the treatment with milnacipran from those who achieve a reduction of pain intensity after the treatment with pregabalin (195). Common side effects of pregabalin include somnolence, dizziness, weight gain and peripheral oedema. Discontinuation due to side effects is approximately 10% higher in patients treated with pregabalin compared to placebo, while discontinuation due to inefficiency of treatment is 6% lower (191). The intensity of adverse effects and the frequency of discontinuation of the treatment due to adverse effects is dose dependent. It is important for pregabalin to be titrated to the maximally tolerated therapeutic dose for each patient (192).  

 

Gabapentin has been shown to be efficient in treating fibromyalgia associated pain, while it was well tolerated (196). Side effects include dizziness, sedation, lightheadedness, and weight gain. Its efficacy and tolerability is not well studied in fibromyalgia patients, however it can be considered as an acceptable alternative in case pregabalin cannot be administered due to its cost or due to regulatory limitations (197).

 

SEDATIVE HYPNOTIC AGENTS

 

Zopiclone and zolpidem have been used in fibromyalgia. It has been suggested that they can improve the sleep and perhaps fatigue, without any significant effects on pain (144).

 

Sodium oxibate, a precursor of GABA with powerful sedative properties has been shown to improve pain, fatigue and sleep architecture in fibromyalgia (198). However, in view of safety concerns the European Medicines Agency and the US Food and Drug Administration have not approved it for use in fibromyalgia patients.

 

TRAMADOL

 

Tramadol has multiple analgesic effects, since it inhibits norepinephrine and serotonin reuptake, and its major metabolite binds weakly to opioid μ receptors (144). The use of tramadol (with or without acetaminophen) is both effective and well tolerated for the management of pain in fibromyalgia (199,200). There are some concerns regarding the long-term potential of abuse of tramadol, although the risk is less than that of more potent narcotic analgesics that have also been used in fibromyalgia.

 

Table 9. The Doses of the Most Commonly used Medications with Strong and Moderate Evidence of Effectiveness in Fibromyalgia

Drugs

Doses

Tricyclic antidepressants

Amitriptyline

Start 5-10 mg at bedtime, increase up to 25-50 mg

Cyclobenzaprine

Start 10 mg at bedtime, increase up to 30-40mg,

decrease to 5mg if 10mg too sedating

Serotonin-norepinephrine reuptake inhibitors

Duloxetine

Start 10-15mg twice daily,

gradually increased to 30 mg twice daily

Milnacipran

Start 12.5mg in the morning,

gradually increase to 50mg twice daily

Venlafaxine

167 mg per day

Anticonvulsants

Gabapentin

Start 100mg at bedtime,

increase to 1200-2400 mg per day

Pregabalin

Start 25-50mg at bedtime,

increase to 300-450 mg/day

Other

Tramadol

37.5 mg four times daily

 

CONCLUSION

 

Fibromyalgia is a common disease that is often underdiagnosed. Genetic predisposition, in combination with exposure to external stressors may lead to dysregulation of the nociceptive system and to the appearance of clinical symptoms. Fibromyalgia patients do not form a homogenous group with some patients responding adequately to current therapeutic modalities, and some others not experiencing any long-term benefit. Patients treated by primary care physicians in the community have a much better prognosis, compared to patients treated in tertiary referral centers. Certain psychological factors, such as an increased sense of control over pain, a belief that one is not disabled, that pain is not a sign of damage, and behaviors like seeking help from others, decreased guarding during examination, exercising more and having pacing activities are associated with better prognosis. Conversely, catastrophizing is associated with increased awareness of pain and with worsening symptoms.

 

REFERENCES

 

  1. Bennett RM. Clinical manifestations and diagnosis of fibromyalgia. Rheum. Dis. Clin. North Am. 2009;35(2):215–232.
  2. Wessely S, Hotopf M. Is fibromyalgia a distinct clinical entity? Historical and epidemiological evidence. Baillieres Best Pract Res Clin Rheumatol 1999;13(3):427–436.
  3. Wolfe F. Editorial: the status of fibromyalgia criteria. Arthritis & Rheumatology (Hoboken, N.J.) 2015;67(2):330–333.
  4. Weir PT, Harlan GA, Nkoy FL, Jones SS, Hegmann KT, Gren LH, Lyon JL. The incidence of fibromyalgia and its associated comorbidities: a population-based retrospective cohort study based on International Classification of Diseases, 9th Revision codes. J Clin Rheumatol 2006;12(3):124–128.
  5. Mease P, Arnold LM, Bennett R, Boonen A, Buskila D, Carville S, Chappell A, Choy E, Clauw D, Dadabhoy D, Gendreau M, Goldenberg D, Littlejohn G, Martin S, Perera P, Russell IJ, Simon L, Spaeth M, Williams D, Crofford L. Fibromyalgia syndrome. J Rheumatol 2007;34(6):1415–1425.
  6. Yunus MB. Fibromyalgia and overlapping disorders: the unifying concept of central sensitivity syndromes. Semin. Arthritis Rheum. 2007;36(6):339–356.
  7. Marcus DA, Bernstein C, Rudy TE. Fibromyalgia and headache: an epidemiological study supporting migraine as part of the fibromyalgia syndrome. Clin. Rheumatol. 2005;24(6):595–601.
  8. Rhodus NL, Fricton J, Carlson P, Messner R. Oral symptoms associated with fibromyalgia syndrome. J Rheumatol2003;30(8):1841–1845.
  9. Aydin G, Başar MM, Keleş I, Ergün G, Orkun S, Batislam E. Relationship between sexual dysfunction and psychiatric status in premenopausal women with fibromyalgia. Urology 2006;67(1):156–161.
  10. Aaron LA, Burke MM, Buchwald D. Overlapping conditions among patients with chronic fatigue syndrome, fibromyalgia, and temporomandibular disorder. Arch. Intern. Med. 2000;160(2):221–227.
  11. Giesecke T, Williams DA, Harris RE, Cupps TR, Tian X, Tian TX, Gracely RH, Clauw DJ. Subgrouping of fibromyalgia patients on the basis of pressure-pain thresholds and psychological factors. Arthritis & Rheumatism2003;48(10):2916–2922.
  12. Arnold LM, Hudson JI, Hess EV, Ware AE, Fritz DA, Auchenbach MB, Starck LO, Keck PE Jr. Family study of fibromyalgia. Arthritis Rheum. 2004;50(3):944–952.
  13. Løge-Hagen JS, Sæle A, Juhl C, Bech P, Stenager E, Mellentin AI. Prevalence of depressive disorder among patients with fibromyalgia: Systematic review and meta-analysis. J Affect Disord 2019;245:1098–1105.
  14. Kleykamp BA, Ferguson MC, McNicol E, Bixho I, Arnold LM, Edwards RR, Fillingim R, Grol-Prokopczyk H, Turk DC, Dworkin RH. The Prevalence of Psychiatric and Chronic Pain Comorbidities in Fibromyalgia: an ACTTION systematic review. Semin Arthritis Rheum 2021;51(1):166–174.
  15. Wu Y-L, Huang C-J, Fang S-C, Ko L-H, Tsai P-S. Cognitive Impairment in Fibromyalgia: A Meta-Analysis of Case-Control Studies. Psychosom Med 2018;80(5):432–438.
  16. Müller W, Schneider EM, Stratz T. The classification of fibromyalgia syndrome. Rheumatol. Int. 2007;27(11):1005–1010.
  17. Thieme K, Spies C, Sinha P, Turk DC, Flor H. Predictors of pain behaviors in fibromyalgia syndrome. Arthritis Rheum. 2005;53(3):343–350.
  18. Goldenberg DL. Fibromyalgia syndrome a decade later: What have we learned? Arch Intern Med1999;159(8):777–785.
  19. Ursini F, Ciaffi J, Mancarella L, Lisi L, Brusi V, Cavallari C, D’Onghia M, Mari A, Borlandelli E, Faranda Cordella J, La Regina M, Viola P, Ruscitti P, Miceli M, De Giorgio R, Baldini N, Borghi C, Gasbarrini A, Iagnocco A, Giacomelli R, Faldini C, Landini MP, Meliconi R. Fibromyalgia: a new facet of the post-COVID-19 syndrome spectrum? Results from a web-based survey. RMD Open 2021;7(3):e001735.
  20. Clauw DJ, Calabrese L. Rheumatology and Long COVID: lessons from the study of fibromyalgia. Ann Rheum Dis2023:ard-2023-224250.
  21. Mease P, Arnold LM, Choy EH, Clauw DJ, Crofford LJ, Glass JM, Martin SA, Morea J, Simon L, Strand CV, Williams DA. Fibromyalgia syndrome module at OMERACT 9: domain construct. J. Rheumatol. 2009;36(10):2318–2329.
  22. Mease P, Clauw D, Christensen R, Crofford L, Gendreau M, Martin S, Simon L, Strand V, Williams D, Arnold L.Toward Development of a Fibromyalgia Responder Index and Disease Activity Score: OMERACT Module Update. J Rheumatol 2011;38(7):1487–1495.
  23. Boomershine CS. A Comprehensive Evaluation of Standardized Assessment Tools in the Diagnosis of Fibromyalgia and in the Assessment of Fibromyalgia Severity. Pain Research and Treatment 2012;2012:1–11.
  24. Endresen GKM. Fibromyalgia: a rheumatologic diagnosis? Rheumatol Int 2007;27(11):999–1004.
  25. Taylor RR, Jason LA, Torres A. Fatigue rating scales: an empirical comparison. Psychol Med 2000;30(4):849–856.
  26. Whitehead L. The measurement of fatigue in chronic illness: a systematic review of unidimensional and multidimensional fatigue measures. J Pain Symptom Manage 2009;37(1):107–128.
  27. Bennett RM, Friend R, Jones KD, Ward R, Han BK, Ross RL. The Revised Fibromyalgia Impact Questionnaire (FIQR): validation and psychometric properties. Arthritis Res. Ther. 2009;11(4):R120.
  28. Chakrabarty S, Zoorob R. Fibromyalgia. Am Fam Physician 2007;76(2):247–254.
  29. Dworkin RH, Turk DC, Wyrwich KW, Beaton D, Cleeland CS, Farrar JT, Haythornthwaite JA, Jensen MP, Kerns RD, Ader DN, Brandenburg N, Burke LB, Cella D, Chandler J, Cowan P, Dimitrova R, Dionne R, Hertz S, Jadad AR, Katz NP, Kehlet H, Kramer LD, Manning DC, McCormick C, McDermott MP, McQuay HJ, Patel S, Porter L, Quessy S, Rappaport BA, Rauschkolb C, Revicki DA, Rothman M, Schmader KE, Stacey BR, Stauffer JW, von Stein T, White RE, Witter J, Zavisic S. Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. J Pain 2008;9(2):105–121.
  30. Seidenberg M, Haltiner A, Taylor MA, Hermann BB, Wyler A. Development and validation of a Multiple Ability Self-Report Questionnaire. J Clin Exp Neuropsychol 1994;16(1):93–104.
  31. Farias ST, Mungas D, Jagust W. Degree of discrepancy between self and other-reported everyday functioning by cognitive status: dementia, mild cognitive impairment, and healthy elders. Int J Geriatr Psychiatry 2005;20(9):827–834.
  32. Gowers WR. A Lecture on Lumbago: Its Lessons and Analogues: Delivered at the National Hospital for the Paralysed and Epileptic. Br Med J 1904;1(2246):117–121.
  33. Smythe HA, Moldofsky H. Two contributions to understanding of the “fibrositis” syndrome. Bull Rheum Dis1977;28(1):928–931.
  34. Wolfe F, Smythe HA, Yunus MB, Bennett RM, Bombardier C, Goldenberg DL, Tugwell P, Campbell SM, Abeles M, Clark P. The American College of Rheumatology 1990 Criteria for the Classification of Fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis Rheum. 1990;33(2):160–172.
  35. Fillingim RB, Bruehl S, Dworkin RH, Dworkin SF, Loeser JD, Turk DC, Widerstrom-Noga E, Arnold L, Bennett R, Edwards RR, Freeman R, Gewandter J, Hertz S, Hochberg M, Krane E, Mantyh PW, Markman J, Neogi T, Ohrbach R, Paice JA, Porreca F, Rappaport BA, Smith SM, Smith TJ, Sullivan MD, Verne GN, Wasan AD, Wesselmann U. The ACTTION-American Pain Society Pain Taxonomy (AAPT): an evidence-based and multidimensional approach to classifying chronic pain conditions. J Pain 2014;15(3):241–249.
  36. Clauw D. Time to Stop the Fibromyalgia Criteria Wars and Refocus on Identifying and Treating Individuals With This Type of Pain Earlier in Their Illness. Arthritis Care Res (Hoboken) 2021;73(5):613–616.
  37. Goldenberg DL. Diagnosing Fibromyalgia as a Disease, an Illness, a State, or a Trait? Arthritis Care Res (Hoboken) 2019;71(3):334–336.
  38. Perrot S, Dickenson AH, Bennett RM. Fibromyalgia: harmonizing science with clinical practice considerations. Pain Pract 2008;8(3):177–189.
  39. Clauw DJ. Fibromyalgia: update on mechanisms and management. J Clin Rheumatol 2007;13(2):102–109.
  40. Fitzcharles M-A, Boulos P. Inaccuracy in the diagnosis of fibromyalgia syndrome: analysis of referrals. Rheumatology (Oxford) 2003;42(2):263–267.
  41. Wolfe F, Walitt BT, Häuser W. What is fibromyalgia, how is it diagnosed, and what does it really mean? Arthritis Care Res (Hoboken) 2014;66(7):969–971.
  42. Gracely RH, Petzke F, Wolf JM, Clauw DJ. Functional magnetic resonance imaging evidence of augmented pain processing in fibromyalgia. Arthritis Rheum. 2002;46(5):1333–1343.
  43. Desmeules JA, Cedraschi C, Rapiti E, Baumgartner E, Finckh A, Cohen P, Dayer P, Vischer TL. Neurophysiologic evidence for a central sensitization in patients with fibromyalgia. Arthritis Rheum. 2003;48(5):1420–1429.
  44. Harth M, Nielson WR. The fibromyalgia tender points: use them or lose them? A brief review of the controversy. J. Rheumatol. 2007;34(5):914–922.
  45. Wolfe F, Clauw DJ, Fitzcharles M-A, Goldenberg DL, Katz RS, Mease P, Russell AS, Russell IJ, Winfield JB, Yunus MB. The American College of Rheumatology Preliminary Diagnostic Criteria for Fibromyalgia and Measurement of Symptom Severity. Arthritis Care & Research 2010;62(5):600–610.
  46. Kim SM, Lee SH, Kim HR. Applying the ACR Preliminary Diagnostic Criteria in the Diagnosis and Assessment of Fibromyalgia. Korean J Pain 2012;25(3):173–182.
  47. Ferrari R, Russell AS. A questionnaire using the modified 2010 American College of Rheumatology criteria for fibromyalgia: specificity and sensitivity in clinical practice. J. Rheumatol. 2013;40(9):1590–1595.
  48. Marcus DA, Bernstein C, Albrecht KL. Brief, self-report fibromyalgia screener evaluated in a sample of chronic pain patients. Pain Med 2013;14(5):730–735.
  49. Usui C, Hatta K, Aratani S, Yagishita N, Nishioka K, Kanazawa T, Itoh K, Yamano Y, Nakamura H, Nakajima T, Nishioka K. The Japanese version of the modified ACR preliminary diagnostic criteria for fibromyalgia and the fibromyalgia symptom scale: reliability and validity. Mod Rheumatol 2013;23(5):846–850.
  50. Bennett RM, Friend R, Marcus D, Bernstein C, Han BK, Yachoui R, Deodhar A, Kaell A, Bonafede P, Chino A, Jones KD. Criteria for the diagnosis of fibromyalgia: validation of the modified 2010 preliminary American College of Rheumatology criteria and the development of alternative criteria. Arthritis Care Res (Hoboken)2014;66(9):1364–1373.
  51. Segura-Jiménez V, Aparicio VA, Álvarez-Gallardo IC, Soriano-Maldonado A, Estévez-López F, Delgado-Fernández M, Carbonell-Baeza A. Validation of the modified 2010 American College of Rheumatology diagnostic criteria for fibromyalgia in a Spanish population. Rheumatology (Oxford) 2014;53(10):1803–1811.
  52. Carrillo-de-la-Peña MT, Triñanes Y, González-Villar A, Romero-Yuste S, Gómez-Perretta C, Arias M, Wolfe F.Convergence between the 1990 and 2010 ACR diagnostic criteria and validation of the Spanish version of the Fibromyalgia Survey Questionnaire (FSQ). Rheumatol. Int. 2015;35(1):141–151.
  53. Jones GT, Atzeni F, Beasley M, Flüß E, Sarzi-Puttini P, Macfarlane GJ. The prevalence of fibromyalgia in the general population: a comparison of the American College of Rheumatology 1990, 2010, and modified 2010 classification criteria. Arthritis & Rheumatology (Hoboken, N.J.) 2015;67(2):568–575.
  54. Staud R, Price DD, Robinson ME. The provisional diagnostic criteria for fibromyalgia: One step forward, two steps back: Comment on the article by Wolfe et al. Arthritis Care & Research 2010;62(11):1675–1676.
  55. Toda K. Preliminary diagnostic criteria for fibromyalgia should be partially revised: Comment on the article by Wolfe et al. Arthritis Care & Research 2011;63(2):308–309.
  56. Wolfe F, Clauw DJ, Fitzcharles M-A, Goldenberg DL, Häuser W, Katz RS, Mease P, Russell AS, Russell IJ, Winfield JB. Fibromyalgia criteria and severity scales for clinical and epidemiological studies: a modification of the ACR Preliminary Diagnostic Criteria for Fibromyalgia. J. Rheumatol. 2011;38(6):1113–1122.
  57. Häuser W, Schmutzer G, Brähler E, Glaesmer H. A cluster within the continuum of biopsychosocial distress can be labeled “fibromyalgia syndrome”--evidence from a representative German population survey. J. Rheumatol.2009;36(12):2806–2812.
  58. Wolfe F, Brähler E, Hinz A, Häuser W. Fibromyalgia prevalence, somatic symptom reporting, and the dimensionality of polysymptomatic distress: results from a survey of the general population. Arthritis Care Res (Hoboken) 2013;65(5):777–785.
  59. Egloff N, von Känel R, Müller V, Egle UT, Kokinogenis G, Lederbogen S, Durrer B, Stauber S. Implications of proposed fibromyalgia criteria across other functional pain syndromes. Scand. J. Rheumatol. 2015;44(5):416–424.
  60. Wolfe F, Egloff N, Häuser W. Widespread Pain and Low Widespread Pain Index Scores among Fibromyalgia-positive Cases Assessed with the 2010/2011 Fibromyalgia Criteria. J. Rheumatol. 2016;43(9):1743–1748.
  61. Wolfe F, Clauw DJ, Fitzcharles M-A, Goldenberg DL, Häuser W, Katz RL, Mease PJ, Russell AS, Russell IJ, Walitt B. 2016 Revisions to the 2010/2011 fibromyalgia diagnostic criteria. Semin. Arthritis Rheum. 2016;46(3):319–329.
  62. Arnold LM, Bennett RM, Crofford LJ, Dean LE, Clauw DJ, Goldenberg DL, Fitzcharles M-A, Paiva ES, Staud R, Sarzi-Puttini P, Buskila D, Macfarlane GJ. AAPT Diagnostic Criteria for Fibromyalgia. J Pain 2019;20(6):611–628.
  63. Wolfe F. Letter to the editor, “Fibromyalgia Criteria.” J Pain 2019;20(6):739–740.
  64. Häuser W, Brähler E, Ablin J, Wolfe F. Modified 2016 American College of Rheumatology Fibromyalgia Criteria, the Analgesic, Anesthetic, and Addiction Clinical Trial Translations Innovations Opportunities and Networks-American Pain Society Pain Taxonomy, and the Prevalence of Fibromyalgia. Arthritis Care Res (Hoboken)2021;73(5):617–625.
  65. Queiroz LP. Worldwide epidemiology of fibromyalgia. Curr Pain Headache Rep 2013;17(8):356.
  66. Neumann L, Buskila D. Epidemiology of fibromyalgia. Curr Pain Headache Rep 2003;7(5):362–368.
  67. Wolfe F, Ross K, Anderson J, Russell IJ, Hebert L. The prevalence and characteristics of fibromyalgia in the general population. Arthritis Rheum. 1995;38(1):19–28.
  68. Vincent A, Lahr BD, Wolfe F, Clauw DJ, Whipple MO, Oh TH, Barton DL, St Sauver J. Prevalence of fibromyalgia: a population-based study in Olmsted County, Minnesota, utilizing the Rochester Epidemiology Project. Arthritis Care Res (Hoboken) 2013;65(5):786–792.
  69. Nakamura I, Nishioka K, Usui C, Osada K, Ichibayashi H, Ishida M, Turk DC, Matsumoto Y, Nishioka K. An epidemiologic internet survey of fibromyalgia and chronic pain in Japan. Arthritis Care Res (Hoboken)2014;66(7):1093–1101.
  70. Amital D, Fostick L, Polliack ML, Segev S, Zohar J, Rubinow A, Amital H. Posttraumatic stress disorder, tenderness, and fibromyalgia syndrome: are they different entities? J Psychosom Res 2006;61(5):663–669.
  71. Cohen H, Jotkowitz A, Buskila D, Pelles-Avraham S, Kaplan Z, Neumann L, Sperber AD. Post-traumatic stress disorder and other co-morbidities in a sample population of patients with irritable bowel syndrome. Eur. J. Intern. Med. 2006;17(8):567–571.
  72. Fukuda K, Straus SE, Hickie I, Sharpe MC, Dobbins JG, Komaroff A. The chronic fatigue syndrome: a comprehensive approach to its definition and study. International Chronic Fatigue Syndrome Study Group. Ann. Intern. Med. 1994;121(12):953–959.
  73. Committee on the Diagnostic Criteria for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome, Board on the Health of Select Populations, Institute of Medicine.Beyond Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Redefining an Illness. Washington (DC): National Academies Press (US); 2015. Available at: http://www.ncbi.nlm.nih.gov/books/NBK274235/. Accessed December 16, 2019.
  74. Brandt LJ, Chey WD, Foxx-Orenstein AE, Schiller LR, Schoenfeld PS, Spiegel BM, Talley NJ, Quigley EMM. An evidence-based position statement on the management of irritable bowel syndrome. Am. J. Gastroenterol.2009;104 Suppl 1:S1-35.
  75. Hanno PM, Burks DA, Clemens JQ, Dmochowski RR, Erickson D, Fitzgerald MP, Forrest JB, Gordon B, Gray M, Mayer RD, Newman D, Nyberg L Jr, Payne CK, Wesselmann U, Faraday MM. AUA guideline for the diagnosis and treatment of interstitial cystitis/bladder pain syndrome. J. Urol. 2011;185(6):2162–2170.
  76. Abeles AM, Pillinger MH, Solitar BM, Abeles M. Narrative review: the pathophysiology of fibromyalgia. Ann. Intern. Med. 2007;146(10):726–734.
  77. Simms RW, Roy SH, Hrovat M, Anderson JJ, Skrinar G, LePoole SR, Zerbini CA, de Luca C, Jolesz F. Lack of association between fibromyalgia syndrome and abnormalities in muscle energy metabolism. Arthritis Rheum.1994;37(6):794–800.
  78. Häkkinen A, Häkkinen K, Hannonen P, Alen M. Force production capacity and acute neuromuscular responses to fatiguing loading in women with fibromyalgia are not different from those of healthy women. J. Rheumatol.2000;27(5):1277–1282.
  79. Lund E, Kendall SA, Janerot-Sjøberg B, Bengtsson A. Muscle metabolism in fibromyalgia studied by P-31 magnetic resonance spectroscopy during aerobic and anaerobic exercise. Scand. J. Rheumatol. 2003;32(3):138–145.
  80. Yunus MB. Central sensitivity syndromes: a new paradigm and group nosology for fibromyalgia and overlapping conditions, and the related issue of disease versus illness. Semin. Arthritis Rheum. 2008;37(6):339–352.
  81. Suzuki R, Rygh LJ, Dickenson AH. Bad news from the brain: descending 5-HT pathways that control spinal pain processing. Trends Pharmacol. Sci. 2004;25(12):613–617.
  82. Cagnie B, Coppieters I, Denecker S, Six J, Danneels L, Meeus M. Central sensitization in fibromyalgia? A systematic review on structural and functional brain MRI. Semin. Arthritis Rheum. 2014;44(1):68–75.
  83. Truini A, Tinelli E, Gerardi MC, Calistri V, Iannuccelli C, La Cesa S, Tarsitani L, Mainero C, Sarzi-Puttini P, Cruccu G, Caramia F, Di Franco M. Abnormal resting state functional connectivity of the periaqueductal grey in patients with fibromyalgia. Clin. Exp. Rheumatol. 2016;34(2 Suppl 96):S129-133.
  84. Fallon N, Chiu Y, Nurmikko T, Stancak A. Functional Connectivity with the Default Mode Network Is Altered in Fibromyalgia Patients. PLoS ONE 2016;11(7):e0159198.
  85. Kaplan CM, Schrepf A, Vatansever D, Larkin TE, Mawla I, Ichesco E, Kochlefl L, Harte SE, Clauw DJ, Mashour GA, Harris RE. Functional and neurochemical disruptions of brain hub topology in chronic pain. Pain2019;160(4):973–983.
  86. Martucci KT, Weber KA, Mackey SC. Altered Cervical Spinal Cord Resting-State Activity in Fibromyalgia. Arthritis & Rheumatology (Hoboken, N.J.) 2019;71(3):441–450.
  87. Schweinhardt P, Sauro KM, Bushnell MC. Fibromyalgia: A Disorder of the Brain? Neuroscientist 2008. doi:10.1177/1073858407312521.
  88. Staud R, Godfrey MM, Robinson ME. Fibromyalgia Patients Are Not Only Hypersensitive to Painful Stimuli But Also to Acoustic Stimuli. J Pain 2021;22(8):914–925.
  89. Clauw DJ, Crofford LJ. Chronic widespread pain and fibromyalgia: what we know, and what we need to know. Best Pract Res Clin Rheumatol 2003;17(4):685–701.
  90. Salemi S, Aeschlimann A, Wollina U, Gay RE, Michel BA, Gay S, Sprott H. Up-regulation of delta-opioid receptors and kappa-opioid receptors in the skin of fibromyalgia patients. Arthritis Rheum. 2007;56(7):2464–2466.
  91. Harris RE, Clauw DJ, Scott DJ, McLean SA, Gracely RH, Zubieta J-K. Decreased central mu-opioid receptor availability in fibromyalgia. J. Neurosci. 2007;27(37):10000–10006.
  92. Watkins LR, Hutchinson MR, Rice KC, Maier SF. The “toll” of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia. Trends Pharmacol. Sci. 2009;30(11):581–591.
  93. Mhalla A, de Andrade DC, Baudic S, Perrot S, Bouhassira D. Alteration of cortical excitability in patients with fibromyalgia. Pain 2010;149(3):495–500.
  94. Foerster BR, Petrou M, Edden RAE, Sundgren PC, Schmidt-Wilcke T, Lowe SE, Harte SE, Clauw DJ, Harris RE.Reduced insular γ-aminobutyric acid in fibromyalgia. Arthritis Rheum. 2012;64(2):579–583.
  95. Harris RE, Sundgren PC, Craig AD, Kirshenbaum E, Sen A, Napadow V, Clauw DJ. Elevated insular glutamate in fibromyalgia is associated with experimental pain. Arthritis Rheum. 2009;60(10):3146–3152.
  96. Valdés M, Collado A, Bargalló N, Vázquez M, Rami L, Gómez E, Salamero M. Increased glutamate/glutamine compounds in the brains of patients with fibromyalgia: a magnetic resonance spectroscopy study. Arthritis Rheum.2010;62(6):1829–1836.
  97. Haas L, Portela LVC, Böhmer AE, Oses JP, Lara DR. Increased plasma levels of brain derived neurotrophic factor (BDNF) in patients with fibromyalgia. Neurochem. Res. 2010;35(5):830–834.
  98. O’Mahony LF, Srivastava A, Mehta P, Ciurtin C. Is fibromyalgia associated with a unique cytokine profile? A systematic review and meta-analysis. Rheumatology (Oxford) 2021;60(6):2602–2614.
  99. Uçeyler N, Häuser W, Sommer C. Systematic review with meta-analysis: cytokines in fibromyalgia syndrome. BMC Musculoskelet Disord 2011;12:245.
  100. Salemi S, Rethage J, Wollina U, Michel BA, Gay RE, Gay S, Sprott H. Detection of interleukin 1beta (IL-1beta), IL-6, and tumor necrosis factor-alpha in skin of patients with fibromyalgia. J. Rheumatol. 2003;30(1):146–150.
  101. Üçeyler N, Valenza R, Stock M, Schedel R, Sprotte G, Sommer C. Reduced levels of antiinflammatory cytokines in patients with chronic widespread pain. Arthritis & Rheumatism 2006;54(8):2656–2664.
  102. Sturgill J, McGee E, Menzies V. Unique cytokine signature in the plasma of patients with fibromyalgia. J Immunol Res 2014;2014:938576.
  103. Sommer C, Kress M. Recent findings on how proinflammatory cytokines cause pain: peripheral mechanisms in inflammatory and neuropathic hyperalgesia. Neurosci. Lett. 2004;361(1–3):184–187.
  104. Staud R. Fibromyalgia pain: do we know the source? Curr Opin Rheumatol 2004;16(2):157–163.
  105. Tsigos C, Chrousos GP. Hypothalamic-pituitary-adrenal axis, neuroendocrine factors and stress. J Psychosom Res2002;53(4):865–871.
  106. Mastorakos G, Pavlatou M, Diamanti-Kandarakis E, Chrousos GP. Exercise and the stress system. Hormones (Athens) 2005;4(2):73–89.
  107. Beiner E, Lucas V, Reichert J, Buhai D-V, Jesinghaus M, Vock S, Drusko A, Baumeister D, Eich W, Friederich H-C, Tesarz J. Stress biomarkers in individuals with fibromyalgia syndrome: a systematic review with meta-analysis. Pain 2023;164(7):1416–1427.
  108. Crofford LJ, Young EA, Engleberg NC, Korszun A, Brucksch CB, McClure LA, Brown MB, Demitrack MA. Basal circadian and pulsatile ACTH and cortisol secretion in patients with fibromyalgia and/or chronic fatigue syndrome. Brain Behav. Immun. 2004;18(4):314–325.
  109. Wingenfeld K, Heim C, Schmidt I, Wagner D, Meinlschmidt G, Hellhammer DH.HPA axis reactivity and lymphocyte glucocorticoid sensitivity in fibromyalgia syndrome and chronic pelvic pain. Psychosom Med2008;70(1):65–72.
  110. Crofford LJ, Pillemer SR, Kalogeras KT, Cash JM, Michelson D, Kling MA, Sternberg EM, Gold PW, Chrousos GP, Wilder RL. Hypothalamic-pituitary-adrenal axis perturbations in patients with fibromyalgia. Arthritis Rheum.1994;37(11):1583–1592.
  111. Wingenfeld K, Wagner D, Schmidt I, Meinlschmidt G, Hellhammer DH, Heim C. The low-dose dexamethasone suppression test in fibromyalgia. J Psychosom Res 2007;62(1):85–91.
  112. Jones KD, Deodhar P, Lorentzen A, Bennett RM, Deodhar AA. Growth hormone perturbations in fibromyalgia: a review. Semin. Arthritis Rheum. 2007;36(6):357–379.
  113. Ablin J, Neumann L, Buskila D. Pathogenesis of fibromyalgia - a review. Joint Bone Spine 2008;75(3):273–279.
  114. Riva R, Mork PJ, Westgaard RH, Okkenhaug Johansen T, Lundberg U. Catecholamines and heart rate in female fibromyalgia patients. J Psychosom Res 2012;72(1):51–57.
  115. Kadetoff D, Kosek E. Evidence of reduced sympatho-adrenal and hypothalamic-pituitary activity during static muscular work in patients with fibromyalgia. J Rehabil Med 2010;42(8):765–772.
  116. Lerma C, Martinez A, Ruiz N, Vargas A, Infante O, Martinez-Lavin M. Nocturnal heart rate variability parameters as potential fibromyalgia biomarker: correlation with symptoms severity. Arthritis Res. Ther. 2011;13(6):R185.
  117. Cohen H, Neumann L, Shore M, Amir M, Cassuto Y, Buskila D. Autonomic dysfunction in patients with fibromyalgia: application of power spectral analysis of heart rate variability. Semin. Arthritis Rheum.2000;29(4):217–227.
  118. Cohen H, Neumann L, Alhosshle A, Kotler M, Abu-Shakra M, Buskila D. Abnormal sympathovagal balance in men with fibromyalgia. J. Rheumatol. 2001;28(3):581–589.
  119. Torpy DJ, Papanicolaou DA, Lotsikas AJ, Wilder RL, Chrousos GP, Pillemer SR. Responses of the sympathetic nervous system and the hypothalamic-pituitary-adrenal axis to interleukin-6: a pilot study in fibromyalgia. Arthritis Rheum. 2000;43(4):872–880.
  120. Maekawa K, Twe C, Lotaif A, Chiappelli F, Clark GT. Function of beta-adrenergic receptors on mononuclear cells in female patients with fibromyalgia. J. Rheumatol. 2003;30(2):364–368.
  121. Giesecke T, Gracely RH, Williams DA, Geisser ME, Petzke FW, Clauw DJ. The relationship between depression, clinical pain, and experimental pain in a chronic pain cohort. Arthritis Rheum. 2005;52(5):1577–1584.
  122. Lee YH, Choi SJ, Ji JD, Song GG. Candidate gene studies of fibromyalgia: a systematic review and meta-analysis. Rheumatol Int 2012;32(2):417–426.
  123. Smith SB, Maixner DW, Fillingim RB, Slade G, Gracely RH, Ambrose K, Zaykin DV, Hyde C, John S, Tan K, Maixner W, Diatchenko L. Large candidate gene association study reveals genetic risk factors and therapeutic targets for fibromyalgia. Arthritis Rheum 2012;64(2):584–593.
  124. Arnold LM, Fan J, Russell IJ, Yunus MB, Khan MA, Kushner I, Olson JM, Iyengar SK. The fibromyalgia family study: a genome-wide linkage scan study. Arthritis Rheum 2013;65(4):1122–1128.
  125. Docampo E, Escaramís G, Gratacòs M, Villatoro S, Puig A, Kogevinas M, Collado A, Carbonell J, Rivera J, Vidal J, Alegre J, Estivill X, Rabionet R. Genome-wide analysis of single nucleotide polymorphisms and copy number variants in fibromyalgia suggest a role for the central nervous system. Pain 2014;155(6):1102–1109.
  126. Dutta D, Brummett CM, Moser SE, Fritsche LG, Tsodikov A, Lee S, Clauw DJ, Scott LJ. Heritability of the Fibromyalgia Phenotype Varies by Age. Arthritis Rheumatol 2020;72(5):815–823.
  127. Offenbaecher M, Bondy B, de Jonge S, Glatzeder K, Krüger M, Schoeps P, Ackenheil M. Possible association of fibromyalgia with a polymorphism in the serotonin transporter gene regulatory region. Arthritis Rheum.1999;42(11):2482–2488.
  128. Cohen H, Buskila D, Neumann L, Ebstein RP.Confirmation of an association between fibromyalgia and serotonin transporter promoter region (5- HTTLPR) polymorphism, and relationship to anxiety-related personality traits. Arthritis Rheum. 2002;46(3):845–847.
  129. Gursoy S. Absence of association of the serotonin transporter gene polymorphism with the mentally healthy subset of fibromyalgia patients. Clin. Rheumatol. 2002;21(3):194–197.
  130. Buskila D, Dan B, Cohen H, Hagit C, Neumann L, Lily N, Ebstein RP. An association between fibromyalgia and the dopamine D4 receptor exon III repeat polymorphism and relationship to novelty seeking personality traits. Mol. Psychiatry 2004;9(8):730–731.
  131. Gürsoy S, Erdal E, Herken H, Madenci E, Alaşehirli B, Erdal N. Significance of catechol-O-methyltransferase gene polymorphism in fibromyalgia syndrome. Rheumatol. Int. 2003;23(3):104–107.
  132. Zhang L, Zhu J, Chen Y, Zhao J. Meta-analysis reveals a lack of association between a common catechol-O-methyltransferase (COMT) polymorphism val158met and fibromyalgia. Int J Clin Exp Pathol 2014;7(12):8489–8497.
  133. Finan PH, Zautra AJ, Davis MC, Lemery-Chalfant K, Covault J, Tennen H. Genetic influences on the dynamics of pain and affect in fibromyalgia. Health Psychol 2010;29(2):134–142.
  134. Solak Ö, Erdoğan MÖ, Yıldız H, Ulaşlı AM, Yaman F, Terzi ESA, Ulu S, Dündar Ü, Solak M. Assessment of opioid receptor μ1 gene A118G polymorphism and its association with pain intensity in patients with fibromyalgia. Rheumatol. Int. 2014;34(9):1257–1261.
  135. Vargas-Alarcón G, Fragoso J-M, Cruz-Robles D, Vargas A, Martinez A, Lao-Villadóniga J-I, García-Fructuoso F, Vallejo M, Martínez-Lavín M. Association of adrenergic receptor gene polymorphisms with different fibromyalgia syndrome domains. Arthritis Rheum. 2009;60(7):2169–2173.
  136. Üçeyler N, Zeller D, Kahn A-K, Kewenig S, Kittel-Schneider S, Schmid A, Casanova-Molla J, Reiners K, Sommer C. Small fibre pathology in patients with fibromyalgia syndrome. Brain 2013;136(Pt 6):1857–1867.
  137. Giannoccaro MP, Donadio V, Incensi A, Avoni P, Liguori R. Small nerve fiber involvement in patients referred for fibromyalgia. Muscle Nerve 2014;49(5):757–759.
  138. Grayston R, Czanner G, Elhadd K, Goebel A, Frank B, Üçeyler N, Malik RA, Alam U. A systematic review and meta-analysis of the prevalence of small fiber pathology in fibromyalgia: Implications for a new paradigm in fibromyalgia etiopathogenesis. Semin Arthritis Rheum 2019;48(5):933–940.
  139. Clauw DJ, Chrousos GP. Chronic pain and fatigue syndromes: overlapping clinical and neuroendocrine features and potential pathogenic mechanisms. Neuroimmunomodulation 1997;4(3):134–153.
  140. Buskila D, Atzeni F, Sarzi-Puttini P. Etiology of fibromyalgia: the possible role of infection and vaccination. Autoimmun Rev 2008;8(1):41–43.
  141. Buskila D, Neumann L, Vaisberg G, Alkalay D, Wolfe F. Increased rates of fibromyalgia following cervical spine injury. A controlled study of 161 cases of traumatic injury. Arthritis Rheum. 1997;40(3):446–452.
  142. Wynne-Jones G, Jones GT, Wiles NJ, Silman AJ, Macfarlane GJ. Predicting new onset of widespread pain following a motor vehicle collision. J. Rheumatol. 2006;33(5):968–974.
  143. Peterson EL. Fibromyalgia--management of a misunderstood disorder. J Am Acad Nurse Pract 2007;19(7):341–348.
  144. Sarzi-Puttini P, Buskila D, Carrabba M, Doria A, Atzeni F. Treatment strategy in fibromyalgia syndrome: where are we now? Semin. Arthritis Rheum. 2008;37(6):353–365.
  145. Macfarlane GJ, Kronisch C, Dean LE, Atzeni F, Häuser W, Fluß E, Choy E, Kosek E, Amris K, Branco J, Dincer F, Leino-Arjas P, Longley K, McCarthy GM, Makri S, Perrot S, Sarzi-Puttini P, Taylor A, Jones GT. EULAR revised recommendations for the management of fibromyalgia. Ann. Rheum. Dis. 2017;76(2):318–328.
  146. Burckhardt CS, Bjelle A. Education programmes for fibromyalgia patients: description and evaluation. Baillieres Clin Rheumatol 1994;8(4):935–955.
  147. White KP, Nielson WR, Harth M, Ostbye T, Speechley M. Does the label “fibromyalgia” alter health status, function, and health service utilization? A prospective, within-group comparison in a community cohort of adults with chronic widespread pain. Arthritis Rheum. 2002;47(3):260–265.
  148. Ehrlich GE. Pain is real; fibromyalgia isn’t. J. Rheumatol. 2003;30(8):1666–1667.
  149. Atzeni F, Alciati A, Salaffi F, Di Carlo M, Bazzichi L, Govoni M, Biasi G, Di Franco M, Mozzani F, Gremese E, Dagna L, Batticciotto A, Fischetti F, Giacomelli R, Guiducci S, Guggino G, Bentivegna M, Gerli R, Salvarani C, Bajocchi G, Ghini M, Iannone F, Giorgi V, Farah S, Bonazza S, Barbagli S, Gioia C, Marino NG, Capacci A, Cavalli G, Cappelli A, Carubbi F, Nacci F, Riccucci I, Cutolo M, Sinigaglia L, Sarzi-Puttini P. The association between body mass index and fibromyalgia severity: data from a cross-sectional survey of 2339 patients. Rheumatol Adv Pract2021;5(1):rkab015.
  150. Busch AJ, Webber SC, Brachaniec M, Bidonde J, Bello-Haas VD, Danyliw AD, Overend TJ, Richards RS, Sawant A, Schachter CL. Exercise therapy for fibromyalgia. Curr Pain Headache Rep 2011;15(5):358–367.
  151. McDowell CP, Cook DB, Herring MP. The Effects of Exercise Training on Anxiety in Fibromyalgia Patients: A Meta-analysis. Med Sci Sports Exerc 2017;49(9):1868–1876.
  152. Andrade A, Dominski FH, Sieczkowska SM. What we already know about the effects of exercise in patients with fibromyalgia: An umbrella review. Semin Arthritis Rheum 2020;50(6):1465–1480.
  153. Estévez-López F, Maestre-Cascales C, Russell D, Álvarez-Gallardo IC, Rodriguez-Ayllon M, Hughes CM, Davison GW, Sañudo B, McVeigh JG. Effectiveness of Exercise on Fatigue and Sleep Quality in Fibromyalgia: A Systematic Review and Meta-analysis of Randomized Trials. Arch Phys Med Rehabil 2021;102(4):752–761.
  154. Bidonde J, Busch AJ, Webber SC, Schachter CL, Danyliw A, Overend TJ, Richards RS, Rader T. Aquatic exercise training for fibromyalgia. Cochrane Database Syst Rev 2014;(10):CD011336.
  155. Salaffi F, Di Carlo M, Farah S, Marotto D, Giorgi V, Sarzi-Puttini P. Exercise therapy in fibromyalgia patients: comparison of a web-based intervention with usual care. Clin Exp Rheumatol 2020;38 Suppl 123(1):86–93.
  156. Bernardy K, Klose P, Busch AJ, Choy EHS, Häuser W. Cognitive behavioural therapies for fibromyalgia. Cochrane Database Syst Rev 2013;(9):CD009796.
  157. Mascarenhas RO, Souza MB, Oliveira MX, Lacerda AC, Mendonça VA, Henschke N, Oliveira VC. Association of Therapies With Reduced Pain and Improved Quality of Life in Patients With Fibromyalgia: A Systematic Review and Meta-analysis. JAMA Intern Med 2021;181(1):104–112.
  158. Jensen KB, Kosek E, Wicksell R, Kemani M, Olsson G, Merle JV, Kadetoff D, Ingvar M. Cognitive Behavioral Therapy increases pain-evoked activation of the prefrontal cortex in patients with fibromyalgia. Pain2012;153(7):1495–1503.
  159. Lazaridou A, Kim J, Cahalan CM, Loggia ML, Franceschelli O, Berna C, Schur P, Napadow V, Edwards RR.Effects of Cognitive-Behavioral Therapy (CBT) on Brain Connectivity Supporting Catastrophizing in Fibromyalgia. Clin J Pain 2017;33(3):215–221.
  160. Deare JC, Zheng Z, Xue CCL, Liu JP, Shang J, Scott SW, Littlejohn G. Acupuncture for treating fibromyalgia. Cochrane Database Syst Rev 2013;(5):CD007070.
  161. Langhorst J, Klose P, Dobos GJ, Bernardy K, Häuser W. Efficacy and safety of meditative movement therapies in fibromyalgia syndrome: a systematic review and meta-analysis of randomized controlled trials. Rheumatol. Int.2013;33(1):193–207.
  162. Langhorst J, Musial F, Klose P, Häuser W. Efficacy of hydrotherapy in fibromyalgia syndrome--a meta-analysis of randomized controlled clinical trials. Rheumatology (Oxford) 2009;48(9):1155–1159.
  163. Schmidt-Wilcke T, Clauw DJ. Fibromyalgia: from pathophysiology to therapy. Nat Rev Rheumatol 2011;7(9):518–527.
  164. Boomershine CS, Crofford LJ. A symptom-based approach to pharmacologic management of fibromyalgia. Nat Rev Rheumatol 2009;5(4):191–199.
  165. Goldenberg DL BC. MAnagement of fibromyalgia syndrome. JAMA 2004;292(19):2388–2395.
  166. Chou R, Turner JA, Devine EB, Hansen RN, Sullivan SD, Blazina I, Dana T, Bougatsos C, Deyo RA. The effectiveness and risks of long-term opioid therapy for chronic pain: a systematic review for a National Institutes of Health Pathways to Prevention Workshop. Ann Intern Med 2015;162(4):276–286.
  167. Hwang J-M, Lee B-J, Oh TH, Park D, Kim C-H. Association between initial opioid use and response to a brief interdisciplinary treatment program in fibromyalgia. Medicine (Baltimore) 2019;98(1):e13913.
  168. Curtis AF, Miller MB, Rathinakumar H, Robinson M, Staud R, Berry RB, McCrae CS. Opioid use, pain intensity, age, and sleep architecture in patients with fibromyalgia and insomnia. Pain 2019;160(9):2086–2092.
  169. Abeles M, Solitar BM, Pillinger MH, Abeles AM. Update on fibromyalgia therapy. Am. J. Med. 2008;121(7):555–561.
  170. Liu Y, Qian C, Yang M. Treatment Patterns Associated with ACR-Recommended Medications in the Management of Fibromyalgia in the United States. J Manag Care Spec Pharm 2016;22(3):263–271.
  171. Tofferi JK, Jackson JL, O’Malley PG. Treatment of fibromyalgia with cyclobenzaprine: A meta-analysis. Arthritis Rheum. 2004;51(1):9–13.
  172. O’Malley PG, Balden E, Tomkins G, Santoro J, Kroenke K, Jackson JL. Treatment of fibromyalgia with antidepressants: a meta-analysis. J Gen Intern Med 2000;15(9):659–666.
  173. Arnold LM, Keck PE Jr, Welge JA.Antidepressant treatment of fibromyalgia. A meta-analysis and review. Psychosomatics 2000;41(2):104–113.
  174. Carette S, Bell MJ, Reynolds WJ, Haraoui B, McCain GA, Bykerk VP, Edworthy SM, Baron M, Koehler BE, Fam AG. Comparison of amitriptyline, cyclobenzaprine, and placebo in the treatment of fibromyalgia. A randomized, double-blind clinical trial. Arthritis Rheum. 1994;37(1):32–40.
  175. Carette S, Oakson G, Guimont C, Steriade M. Sleep electroencephalography and the clinical response to amitriptyline in patients with fibromyalgia. Arthritis Rheum. 1995;38(9):1211–1217.
  176. Uçeyler N, Häuser W, Sommer C. A systematic review on the effectiveness of treatment with antidepressants in fibromyalgia syndrome. Arthritis Rheum. 2008;59(9):1279–1298.
  177. Häuser W, Petzke F, Üçeyler N, Sommer C. Comparative efficacy and acceptability of amitriptyline, duloxetine and milnacipran in fibromyalgia syndrome: a systematic review with meta-analysis. Rheumatology (Oxford)2011;50(3):532–543.
  178. Alberti FF, Becker MW, Blatt CR, Ziegelmann PK, da Silva Dal Pizzol T, Pilger D. Comparative efficacy of amitriptyline, duloxetine and pregabalin for treating fibromyalgia in adults: an overview with network meta-analysis. Clin Rheumatol 2022;41(7):1965–1978.
  179. Goldenberg D, Mayskiy M, Mossey C, Ruthazer R, Schmid C. A randomized, double-blind crossover trial of fluoxetine and amitriptyline in the treatment of fibromyalgia. Arthritis Rheum. 1996;39(11):1852–1859.
  180. Moldofsky H, Harris HW, Archambault WT, Kwong T, Lederman S. Effects of bedtime very low dose cyclobenzaprine on symptoms and sleep physiology in patients with fibromyalgia syndrome: a double-blind randomized placebo-controlled study. J. Rheumatol. 2011;38(12):2653–2663.
  181. Arnold LM, Gendreau RM, Palmer RH, Gendreau JF, Wang Y. Efficacy and safety of milnacipran 100 mg/day in patients with fibromyalgia: results of a randomized, double-blind, placebo-controlled trial. Arthritis Rheum.2010;62(9):2745–2756.
  182. Branco JC, Zachrisson O, Perrot S, Mainguy Y. A European multicenter randomized double-blind placebo-controlled monotherapy clinical trial of milnacipran in treatment of fibromyalgia. J. Rheumatol. 2010;37(4):851–859.
  183. Sayar K, Aksu G, Ak I, Tosun M. Venlafaxine treatment of fibromyalgia. Ann Pharmacother 2003;37(11):1561–1565.
  184. Lunn MPT, Hughes RAC, Wiffen PJ. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database Syst Rev 2014;1:CD007115.
  185. Arnold LM, Zhang S, Pangallo BA. Efficacy and safety of duloxetine 30 mg/d in patients with fibromyalgia: a randomized, double-blind, placebo-controlled study. Clin J Pain 2012;28(9):775–781.
  186. Russell IJ, Mease PJ, Smith TR, Kajdasz DK, Wohlreich MM, Detke MJ, Walker DJ, Chappell AS, Arnold LM.Efficacy and safety of duloxetine for treatment of fibromyalgia in patients with or without major depressive disorder: Results from a 6-month, randomized, double-blind, placebo-controlled, fixed-dose trial. Pain 2008;136(3):432–444.
  187. Welsch P, Üçeyler N, Klose P, Walitt B, Häuser W. Serotonin and noradrenaline reuptake inhibitors (SNRIs) for fibromyalgia. Cochrane Database Syst Rev 2018;2:CD010292.
  188. Häuser W, Petzke F, Sommer C. Comparative efficacy and harms of duloxetine, milnacipran, and pregabalin in fibromyalgia syndrome. J Pain 2010;11(6):505–521.
  189. Schmidt-Wilcke T, Ichesco E, Hampson JP, Kairys A, Peltier S, Harte S, Clauw DJ, Harris RE. Resting state connectivity correlates with drug and placebo response in fibromyalgia patients. Neuroimage Clin 2014;6:252–261.
  190. Häuser W, Bernardy K, Uçeyler N, Sommer C. Treatment of fibromyalgia syndrome with antidepressants: a meta-analysis. JAMA 2009;301(2):198–209.
  191. Derry S, Cording M, Wiffen PJ, Law S, Phillips T, Moore RA. Pregabalin for pain in fibromyalgia in adults. Cochrane Database Syst Rev 2016;9(9):CD011790.
  192. Arnold LM, Choy E, Clauw DJ, Oka H, Whalen E, Semel D, Pauer L, Knapp L. An evidence-based review of pregabalin for the treatment of fibromyalgia. Curr Med Res Opin 2018;34(8):1397–1409.
  193. Arnold LM, Emir B, Pauer L, Resnick M, Clair A. Time to improvement of pain and sleep quality in clinical trials of pregabalin for the treatment of fibromyalgia. Pain Med 2015;16(1):176–185.
  194. Harris RE, Napadow V, Huggins JP, Pauer L, Kim J, Hampson J, Sundgren PC, Foerster B, Petrou M, Schmidt-Wilcke T, Clauw DJ. Pregabalin rectifies aberrant brain chemistry, connectivity, and functional response in chronic pain patients. Anesthesiology 2013;119(6):1453–1464.
  195. Ichesco E, Peltier SJ, Mawla I, Harper DE, Pauer L, Harte SE, Clauw DJ, Harris RE. Prediction of Differential Pharmacologic Response in Chronic Pain Using Functional Neuroimaging Biomarkers and a Support Vector Machine Algorithm: An Exploratory Study. Arthritis Rheumatol 2021;73(11):2127–2137.
  196. Arnold LM, Goldenberg DL, Stanford SB, Lalonde JK, Sandhu HS, Keck PE Jr, Welge JA, Bishop F, Stanford KE, Hess EV, Hudson JI. Gabapentin in the treatment of fibromyalgia: a randomized, double-blind, placebo-controlled, multicenter trial. Arthritis Rheum. 2007;56(4):1336–1344.
  197. Cooper TE, Derry S, Wiffen PJ, Moore RA. Gabapentin for fibromyalgia pain in adults. Cochrane Database Syst Rev 2017;1(1):CD012188.
  198. Perrot S, Russell IJ. More ubiquitous effects from non-pharmacologic than from pharmacologic treatments for fibromyalgia syndrome: a meta-analysis examining six core symptoms. Eur J Pain 2014;18(8):1067–1080.
  199. Roskell NS, Beard SM, Zhao Y, Le TK. A meta-analysis of pain response in the treatment of fibromyalgia. Pain Pract 2011;11(6):516–527.
  200. MacLean AJB, Schwartz TL. Tramadol for the treatment of fibromyalgia. Expert Rev Neurother 2015;15(5):469–475.

 

Immune System Effects on the Endocrine System

ABSTRACT

 

Among the most important and complex systems in the human body are the endocrine and immune systems. Emerging research over the last decade has shed light on their remarkable interplay, revealing a multitude of bidirectional communication pathways and reciprocal regulation mechanisms. Endocrine diseases, such as autoimmune thyroiditis, diabetes mellitus type 1 and type 2, osteoporosis, and disorders of the hypothalamic-pituitary-adrenal (HPA) axis, as well as endocrine malignancies, such as thyroid cancer, are highly interconnected with dysregulations of the immune system.Thus, multiple cytokines, chemokines, and evolving inflammatory processes are involved in the pathogenesis of immune-related endocrine disorders, providing potential targets for immune-based therapeutic approaches. In this chapter, we provide a comprehensive overview of the molecular mechanisms underlying these complex endocrine-immune interactions, and discuss the implications of immune system function or dysfunction in endocrine disorders.

 

INTRODUCTION

 

The immune system is a host defense system that comprises numerous biological structures and processes to defend the human body against potentially harmful substances and invading pathogens. It functions through a series of coordinated mechanisms, including innate and adaptive immunity. The innate immune response consists of i) phagocytosis by macrophages, neutrophils, monocytes, and dendritic cells, and ii) cytotoxicity by natural killer cells, providing an immediate, nonspecific defense against a wide range of invaders by recognizing common patterns shared by many pathogens (1).

 

Adaptive immunity, on the other hand, is an acquired, specially designed defense system; it develops over time and utilizes highly specialized immune cells involving antibody-dependent complement or cell-mediated cytotoxicity produced by T cells that recognize injurious agents, such as heat shock proteins or microbial antigens. During adaptive immunity, antigens taken up by antigen-presenting cells (APCs) are presented to T cells through binding with major histocompatibility complex (MHC) molecules on the surface of these cells. Activated CD4 helper T cells stimulate the release of cytokines, such as interleukin (IL)-2, which i) induce T cell proliferation and activation, ii) stimulate killer cell activity by CD8 suppressor T cells, and iii) activate B cells to differentiate into plasma cells and produce antibodies. Naïve T cells differentiate mainly into two main subsets that produce a different set of cytokines and regulate distinct immune functions. T-helper 1 (Th1) cells produce mainly interferon-γ (IFN)-γ, tumor necrosis factor-α (TNF)-, and IL-12 to regulate cell-mediated responses, while T-helper 2 (Th2) cells secrete IL-4, IL-5, and IL-13, to stimulate antibody production. In addition, three other subsets of T helper cells have been identified: Th22, which secrete IL-22, Th17, which secrete IL-17, and Treg, which secrete transforming growth factor (TGF)- b, all of which also play a role in the pathogenesis of autoimmune diseases (2).

 

Multiple regulatory mechanisms are involved in maintaining central and peripheral T and B cell tolerance (1). Defects in the processes that ensure immune cell tolerance may induce a maladaptive immune response to a self-antigen and lead to the development of autoimmune diseases.

 

Emerging research of the last few decades has shed light on the remarkable interplay between endocrine and immune systems, revealing a multitude of bidirectional communication pathways and reciprocal regulation.

 

Autoimmune endocrine diseases, such as Hashimoto thyroiditis, diabetes mellitus type 1 (DM1), and Addison disease, as well as endocrine malignancies, such as differentiated, anaplastic, and medullary thyroid cancer (3), and other endocrine disorders, including diabetes mellitus type 2 (DM2), osteoporosis, as well as a dysfunctional hypothalamic-pituitary-adrenal (HPA) axis response to stress and inflammation, are highly interconnected with dysregulations of the immune system.

 

This chapter seeks to elucidate the interplay between the endocrine and immune systems, exploring their interconnections and highlighting the impact of their crosstalk on health and disease. We present a comprehensive overview of the molecular mechanisms underlying this interaction and discuss the potential therapeutic implications of targeting the immune system for the management of endocrine diseases.

 

IMMUNE SYSTEM AND THYROID DISEASE

 

Autoimmune Thyroid Disease

 

Autoimmune thyroid disease (AITD) results from a dysregulation of the immune system that leads to loss of tolerance to thyroid antigens and to an autoimmune attack on the thyroid gland. The most common clinical manifestations of AITD are Hashimoto's thyroiditis and Graves’ disease, while less prevalent manifestations are drug-induced thyroiditis, postpartum thyroiditis, or thyroiditis associated with polyglandular syndromes (i.e., autoimmune polyglandular syndromes type 1 and type 2). The underlying molecular mechanisms of AITD involve both circulating autoantibodies and T cell immune mechanisms, while genetic background, as well as cross-reactivity to external antigens (4-6), are also implicated.

 

There are three major thyroid autoantigens that are targeted during autoimmune thyroid attack and are critical for thyroid homeostasis, namely, thyroglobulin (Tg), thyroid peroxidase (TPO), and the thyrotropin receptor (TSHR) (Figure 1).

 

Figure 1. Thyroid proteins that serve as autoantigens. Thyroglobulin (Tg) function as storage protein in thyroid cells, playing a critical role in the synthesis and release of thyroid hormones. Thyroid peroxidase (TPO) catalyzes iodination of tyrosines in thyroglobulin, which attaches one or two iodine molecules to form monoiodotyrosine (MIT) or diiodotyrosine (DIT), respectively. In addition, thyroid peroxidase catalyzes the coupling of iodotyrosine residues to form triiodothyronine (T3) and thyroxine (T4) attached to thyroglobulin. Thyrotropin receptor (TSHR) is a transmembrane G-protein coupled receptor that upon stimulation by circulating TSH activates the expression of downstream effector genes to regulate thyroid growth, thyrocyte differentiation, and thyroid hormone synthesis. Sodium/iodide symporter (NIS) is a membrane glycoprotein, which actively cotransports two sodium cations per each iodide anion, using the electrochemical sodium gradient generated by the Na+/K+-ATPase. Pendrin is involved in the apical iodide efflux in thyroid cells. It can also exchange chloride and bicarbonate. [Modified by Boguslawska et al (7)].

 

Thyroglobulin is a soluble glycoprotein homodimer composed of two subunits of ~330 kDa in size and is the most abundant glycoprotein in the thyroid gland. It is the scaffold for the synthesis of thyroid hormones and the storage-form of thyroid hormones inside the gland. Recently, researchers described the first atomic structure of full-length Tg and identified its hormone-forming tyrosine residues (8). Anti-Tg antibodies (TgAb) act mainly through antibody-dependent cytotoxicity cells rather than through complement fixation (7). In AITD, the prevalent TgAb species recognize native rather than denatured antigens and bind to a number of overlapping epitopic domains located mainly in the central region and C-terminal end of Tg. Of note, TgAb in the serum of healthy subjects have a different epitopic pattern (9).

 

Another major thyroid antigen is TPO, a glycosylated heme-containing homodimer of two 107-kDa transmembrane subunits located in the apical membrane of thyrocytes (Figure 1). It catalyzes the iodination of tyrosyl residues in Tg and the coupling of iodotyrosine residues to form iodothyronines attached to Tg (10). Both humoral and cellular immune responses are directed against TPO. TPO autoantibodies (TPOAb) occur in almost all patients with Hashimoto thyroiditis and approximately 75% of individuals with Graves’ disease (11). In addition, TPOAb may be involved in autoimmune thyroid cell death via antibody-dependent cytotoxic cells and C3 complement-mediated cytotoxicity (7). Specific patterns of TPOAb recognition remain stable in an individual over time and are genetically transmitted through family lineages (12).

 

Thyrotropin receptor is primarily expressed on the basolateral membrane of thyrocytes (Figure 1) and belongs to the transmembrane G protein-coupled receptor family. The intracellular signaling pathway that is activated by the interaction of TSH with TSHR is indispensable for the synthesis of thyroid hormones and the proliferation of follicular epithelial cells. TSHR-stimulating antibodies (TSAb) act as TSHR agonists and stimulate thyroid growth and production of thyroid hormone in an autonomous and unregulated manner. On the other hand, TSHR-blocking antibodies (TBAb) act as antagonists that block the intracellular signaling of TSH, leading to decrease of thyroid hormone synthesis and subsequently hypothyroidism. Neutral antibodies to TSHR, which may also be detected in the serum of patients with Graves’ disease, bind to the receptor but do not alter its activity (13). The exact antigenic sites of TSHR-specific TBAb and TSAb overlap and are mostly directed to the extracellular A subunit of the receptor (Figure 1) (14,15).

 

Other thyroid antigens include the iodide transporters, sodium iodide symporter (NIS), and pendrin (Figure 1). The presence of NIS antibodies is increased in AITDs, especially in patients with Graves’ disease, whereas their expression in euthyroid individuals is rare (16). Pendrin is an apical membrane-bound iodide transporter, but the diagnostic value of antibodies against pendrin is rather low (16).

 

Although the above-described circulating autoantibodies are useful markers of thyroid autoimmunity, it is the T cell immune mechanism, i.e., the loss of immune self-tolerance, that is the core of AITD pathophysiology. Loss of immune self-tolerance may result from either: i) the loss of central tolerance (i.e., disturbed deletion of autoreactive T cells in the thymus), ii) dysfunction of peripheral tolerance (i.e., impaired apoptosis of self-reactive T cells and inhibition of the activity of T-regulatory cells), or iii) disturbed energy (i.e., disturbance of the functional inactivation that prevents the lymphocytes from activating an immune reaction against the antigen).

 

Patients with AITD express IFN-γ-induced MHC class II molecules, which promotes the presentation of thyroid autoantigens and activates T cells (4). The subsequent infiltration of the thyroid gland by APCs (dendritic cells and macrophages) may be triggered by inflammation resulting from either viral or bacterial infection or exposure to toxins. The common mechanism involved in the initiation and perturbation of the inflammatory processes in AITDs and in other autoimmune endocrine diseases (e.g., type 1 diabetes and Addison disease) is the Th1-cytokine/chemokine axis (17). Upon activation, Th1 lymphocytes produce IFN-γ -and TNF-α, which stimulate thyrocytes and retroorbital cells (in Graves ophthalmopathy) to secrete chemokines (CXCL10, CXCL9, and CXCL11). Chemokines, in turn, bind and activate the CXCR3 receptor on Th1 cells and further enhance IFN-γ and TNF-α secretion in a positive feedback loop which aggravates recruitment and activation of inflammatory cells in the affected organs (Figure 2). In advanced thyroiditis, the thyroid gland is infiltrated by B cells (representing up to 50% of the infiltrating immune cells), as well as cytotoxic T lymphocytes and CD4+ cells (7).

 

Figure 2. Depiction of the molecular mechanism involved in the inflammatory processes in autoimmune thyroid diseases. Activation of Th1 lymphocytes by antigen presenting cells produce INF-γ -and IL-2 and 12, which stimulate thyrocytes and retroorbital cells to secrete chemokines (CXCL10, CXCL9, and CXCL11). Chemokines bind and activate the CXCR3 receptor on Th1 cells, further enhancing IFN-γ and ILs secretion. APC, antigen presenting cells; IFΝ-γ, interferon gamma; IL-2, interleukin 2; IL-12, interleukin-12; CXCR3, C-X-C Motif Chemokine Receptor 3.

 

In Hashimoto thyroiditis, the humoral immune response is characterized by the presence of autoantibodies to TPO or Tg and is related to thyroid lymphocytic infiltration. These autoantibodies are themselves cytotoxic or may affect antigen processing or presentation to T cells. Th1 cells are the predominant T cell clones found in patients with Hashimoto thyroiditis: they promote apoptosis of thyrocytes through secretion of IL-12, TNF-α, and INF-γ, which activate cytotoxic T lymphocytes and macrophages (18). In addition, the Toll-like receptor-3 protein is overexpressed in human thyrocytes surrounded by immune cells in all patients with Hashimoto thyroiditis, but not in Graves’ disease or in euthyroid individuals (17).

 

In patients with Graves’ disease, the predominant antibodies are directed against the TSHR. T cells are activated through the presentation of TSHR peptides and, in turn, trigger B-cells and plasma cells that infiltrate the thyroid to produce autoantibodies directed against the TSHR. Activity of B-cells and plasma cells is also regulated by liver-produced insulin growth factor 1 (IGF1). In contrast to TPO and Tg, TSHR is widely expressed in extrathyroidal tissues and cells, including lymphocytes, adipose tissue, and fibroblasts. In consequence, the presence of TSHR antibodies contributes to the extrathyroidal manifestations of Graves’ disease, such as Graves ophthalmopathy, Graves dermopathy, and Graves-associated thymus hyperplasia (19).

 

Particularly in Graves ophthalmopathy, the TSHR autoantigen is presented by macrophages and B cells recruited to the orbit (20). Activated T cells, in turn, initiate an immunological attack on the orbital fibroblasts expressing TSHR. In response to the cytokines released by Th cells, orbital fibroblasts and adipocytes —both expressing TSHR— produce and deposit large amounts of glycosaminoglycans (e.g., hyaluronan), leading to increased osmotic pressure and water uptake, swelling of the extraorbital muscles, and increased accumulation of orbital adipose tissue. The process is enhanced by the IGF1/IGF1R signaling pathway in fibroblasts and adipocytes, as well as the crosstalk between TSHR and IGF1R intracellular signaling (21). A similar molecular pathophysiology may underlie Graves dermopathy (19).

 

NON-CODING RNAs

 

In recent years, non-coding RNAs, which are known to modulate gene transcription at the post-transcriptional level, have attracted a great deal of attention as potential biomarkers in various endocrine diseases. Although the study of microRNAs and circular RNAs is still in its infancy, these newly discovered non-coding, single-stranded RNA molecules have been implicated in the development and progression of AITDs.

 

A cluster of biomarkers consisting of miR-205/miR-20a-3p/miR-375/miR-296/miR-451/miR-500a/miR-326 has been reported to be differentially expressed in patients with Hashimoto thyroiditis (22,23). Similarly, multi-miRNA-based biomarkers, such as miR-762/miR-144-3p or miR-210/miR-155/miR-146, were differentially expressed in the serum of patients with Graves’ disease compared to healthy individuals. These dysregulated miRNAs can target key genes involved in the immune response and thyroid function. Regarding their prognostic role, higher miR-21-5p expression is associated with a worse prognosis for patients with Graves’ disease, whereas impaired expression of miR-155 correlates with the size of the goiter (22,24). Further understanding of the role of miRNAs in AITDs could provide valuable insights into disease mechanisms and potentially identify novel therapeutic targets. Data on other non-coding RNAs (such as long-noncoding RNAs and circular RNAs) are scarcer. Impaired expression of n335641, TCONS-00022357-xloc-010919, and n337845 was found in B cells of patients with Graves’ disease (25), while altered expression of 627 circRNAs in PBMCs of patients with Hashimoto thyroiditis has been tested for their potential prognostic value (26).

 

GUT MICROBIOME

 

Emerging evidence suggests that the composition and function of the gut microbiome may be involved in AITDs. The gut microbiome refers to the complex community of microorganisms residing in the gastrointestinal tract, which plays a vital role in various aspects of human health, such as prevention of intestinal colonization by pathogenic bacteria, fermentation/degradation of food debris, and production of nutrients. Studies have indicated that alterations in the gut microbiome can influence immune responses and contribute to the development of autoimmune diseases (7). In this context, dysbiosis, or an imbalance in the gut microbiota, has been observed in patients with Hashimoto thyroiditis and Graves’ disease, thus distinguishing them from healthy controls, while it was associated with the stage of disease, the level of thyroid autoantibodies, and the response to therapy.

 

Targeting the microbiome through dietary interventions or probiotics may represent a promising potential therapeutic avenue. It is therefore of interest to note that in hypothyroid patients treated with LT4, symbiotic supplementation for 8 weeks resulted in decrease of TSH concentration and LT4 dose (27). Moreover, the results of a large clinical study, INDIGO, reported a significant effect of LAB4 probiotics on the gut microbiota composition of patients with Graves’ disease and a temporary reduction in the serum level of IgG and IgA antibodies (28). Further research involving patients from different populations is required to fully understand the relationship between the gut microbiome and AITDs and to explore potential strategies for intervention.

 

Postpartum Thyroiditis

 

Postpartum thyroiditis may occur up to 12 months after delivery. Usually it presents as transient hyperthyroidism (median time of onset, 13 weeks post-delivery), followed by transient hypothyroidism (median time of onset, 19 weeks post-delivery). In the majority of patients, restoration of normal thyroid function occurs. Pregnancy triggers hormonal changes and immune system alterations, such as a shift from Th1 to Th2 cytokine production followed by a "rebound" shift back to Th1 after delivery, and fluctuations in transforming growth factor-beta1 TGF-β1) serum levels (29,30).

 

Anti-TPO and anti-Tg antibodies are found in almost all patients with postpartum thyroiditis. Notably, up to 50% of women who had anti-TPO antibodies at the end of the first trimester of gestation (i.e., before thyroid antibody titers start to decline during pregnancy) developed postpartum thyroiditis. Furthermore, there is evidence that the anti-TPO antibody titer at 16 weeks of gestation is related to the severity of postpartum thyroiditis (31), while activation of the complement is also involved in the pathogenesis (23).

 

Euthyroid Sick Syndrome

 

Euthyroid sick syndrome (ESS) is a condition characterized by alterations in thyroid hormone levels despite normal thyroid gland function. The characteristic laboratory abnormalities of the ESS include low T3 and/or fT3, elevated reverse T3 (rT3), normal or low TSH, and normal or low serum T4 or fT4 concentrations. Clinical conditions that trigger the development of ESS include systemic inflammation, myocardial infarction, starvation, sepsis, surgery, trauma, chronic degenerative diseases, malignancy, and every other condition associated with severe illness.

 

During illness or periods of severe physiological stress, the immune system releases various proinflammatory cytokines, such as in IL-6, TNF-α, IL-1β, IFN-γ, and TGF-β2. These cytokines disrupt the hypothalamic-pituitary-thyroid axis and interfere with the normal synthesis, secretion, and metabolism of thyroid hormones (Figure 3), while the more severe the illness, the more extensive the hormonal alterations.

 

Figure 3. Secretion of proinflammatory cytokines during severe illness or stress inhibits the activity of hepatic deiodinase type 1- suppressing the peripheral conversion of T4 to T3. They also suppress intrathyroidal hormone synthesis, TRH release and TSH secretion from the pituitary. IL-6, interleukin-6; IL-1, interleukin-1; TNF-α, tumor necrosis factor alpha; INF-γ, interferon gamma; TGF-β, tumor growth factor beta; TG, thyroglobulin; NIS, sodium/iodide symporter; TSH, thyroid stimulating hormone; TRH, Thyrotropin-releasing hormone.

 

Proinflammatory cytokines suppress the peripheral conversion of T4 to T3, resulting in low T3 levels and increase rT3 levels, by inhibiting the activity of hepatic deiodinase type 1, which promotes conversion of T4 to T3 and of rT3 to diiodothyronine (32). They also suppress TRH release and inhibit TSH response to TRH stimulation, leading to a decrease in TSH levels. Thus, the cause of the decreased T3 concentration in ESS is decreased T3 production, whereas the cause of the increased rT3 concentration is the result of impaired degradation. Prolonged and severe illness is marked by a decrease in circulating total T4 along with low T3 and high rT3, with very low T4 levels displaying a poor prognosis and having been associated with an increased mortality rate (32). In addition, cytokines reduce iodine uptake by inhibiting sodium-iodine symporter protein expression (25-27), and decrease thyrocyte growth (33), iodide organification (34,35), and thyroglobulin synthesis (36,37).

 

The central role of cytokines in the pathophysiology of ESS has been further elucidated in studies involving cytokine administration to humans. TNF-α administration in healthy volunteers caused a decrease in serum T3 and an increase in serum rT3 concentration (38). Unlike IL-6, serum TNF-α levels did not correlate with any of the typical thyroid parameters, such as low T3, increased rT3, or decreased TSH levels seen in ESS (39,40), suggesting that the changes of thyroid hormone profile following TNF-α administration might be indirect (i.e., through TNF-α increase in circulating IL-6 levels). Furthermore, both IL-6 and TNF-α can upregulate type 2 deiodinase in the anterior pituitary, affecting TSH release and contributing to the development of the non-thyroidal illness syndrome (41,42).

 

Leptin, a hormone primarily known for its role in regulating appetite and energy balance, has also been implicated in the development of EES. During acute illness or chronic inflammation, leptin levels are usually elevated. A link between leptin and the proinflammatory cytokines TNF-α and IL-6 in chronic inflammatory diseases, such as chronic obstructive pulmonary disease (43) and ankylosing spondylitis (44), respectively, has also been proposed previously.

 

The primary effect exerted by leptin on the hypothalamic-pituitary-thyroid axis is alteration of the setpoint for feedback sensitivity of hypophysiotropic TRH-producing neurons in the paraventricular nucleus of the hypothalamus to thyroid hormones (mainly T3) by lowering of the setpoint when leptin levels are suppressed during fasting (45). Two anatomically distinct and functionally antagonistic populations of neurons in the arcuate nucleus of the hypothalamus, α-melanocortin-stimulating hormone (α-MSH)-producing neurons that co-express cocaine- and amphetamine-regulated transcript and neuropeptide Y (NPY)-producing neurons that co-express agouti-related peptide (AGRP), are responsible for the effects of leptin on hypophysiotropic TRH. It has also been proposed that leptin directly affects hypophysiotropic TRH neurons (46). Leptin has been found to inhibit the conversion of T4 to T3 in peripheral tissues and increase the activity of the enzyme type 3 deiodinase, which converts T4 to rT3. These data suggest that leptin can disturb thyroid function in seriously ill patients via two different independent mechanisms (cytokine-dependent and directly).

 

Amiodarone-Induced Thyroid Disease

 

Amiodarone, a benzofuran derivative with a similar structure to that of thyroid hormones, is a highly effective antiarrhythmic agent widely used in the treatment of various types of tachyarrhythmias (supraventricular and ventricular arrhythmias). Amiodarone contains two iodine atoms per molecule, which is approximately 37.5% iodine by molecular weight (47).

 

Treatment with amiodarone may be related to an increase in lymphocyte subsets leading to an exacerbation of pre-existing autoimmunity (48,49). The relative proportion of patients developing either thyrotoxicosis or hypothyroidism depends on the iodine content of the local diet and pre-existing thyroid autoimmunity. In relatively iodine-replete areas, approximately 25% of patients with amiodarone-induced thyroid dysfunction become thyrotoxic, accounting for approximately 3% of amiodarone-treated individuals (50).

 

Amiodarone-induced hypothyroidism is attributed to an increased susceptibility to the inhibitory effect of iodide on thyroid hormone synthesis and/or to a failure to escape the Wolff-Chaikoff effect (49). Hashimoto thyroiditis is the most common risk factor for amiodarone-induced hypothyroidism and it is considered the most likely reason for the female preponderance of this clinical entity (51). Female patients with positive anti-TPO and anti-Tg autoantibodies have a relative risk of 13.5% for developing amiodarone-induced hypothyroidism compared to men without thyroid autoantibodies (20).

 

The pathogenesis of amiodarone-induced thyrotoxicosis is complex, although two distinct forms, type 1 and type 2, are recognized. Type 1 develops in patients with latent thyroid disease, predominantly nodular goiter, in whom the amiodarone iodine load triggers increased synthesis of thyroid hormones. Type 2 is the result of a destructive thyroiditis in a previously normal gland, with leakage of preformed thyroid hormones despite a reduction in hormone synthesis (47,50,52,53).

 

Differentiating between the two types of amiodarone-induced thyrotoxicosis is an essential step in their management, as treatment of each type is different (50). Type 1 usually responds to thionamide therapy, which blocks hormone synthesis, and perchlorate, which blocks active transport of iodine into the thyroid, whereas type 2 responds to high-dose glucocorticoids (50,53-55). Nevertheless, several studies now suggest that these two types should be treated concomitantly; thus, currently, patients with amiodarone-induced thyrotoxicosis receive both antithyroid drugs and prednisolone. In cases resistant to medical treatment and/or in patients with severe cardiac diseases who cannot interrupt amiodarone or require quick amiodarone reintroduction, total thyroidectomy may be offered after rapid correction of thyrotoxicosis following combination treatment with thionamides, KClO4, glucocorticoids, and a short course of iopanoic acid (56).

 

Thyroid Cancer

 

Thyroid cancer is the most common endocrine cancer, the incidence of which has steadily increased over the past few decades (57).

 

The association of chronic inflammation induced by Hashimoto thyroiditis and thyroid cancer has been long recognized (58). However, the immune response triggered against thyroid cancer and AITDs differs significantly. In thyroid cancer, the immune response is more tolerant and allows tumor growth, whereas in AITDs, the response is more aggressive, triggering cell destruction and reduction of the function of the gland (59). Hashimoto thyroiditis is considered both a risk factor for the development of thyroid cancer (60) and a favorable prognostic factor due to chronic lymphocytic infiltration, which can downregulate tumor aggressiveness (60,61). In Graves’ disease, the presence of a strong humoral immune response appears to be protective against thyroid cancer. Patients with increased anti-TPO and anti-Tg levels show lower distant metastasis rates than patients without thyroid autoantibodies (62,63), suggesting their potentially protective role (59).

 

Immune infiltrates in the tumor microenvironment differ between the different thyroid neoplasm subtypes (Figure 4). In general, differentiated thyroid cancer (DTC) has a higher number of tumor-associated lymphocytes and regulatory T cells (Tregs), while anaplastic thyroid cancer (ATC) and medullary thyroid cancer (MTC) display a high density of tumor-associated macrophages (64). The number of tumor-associated macrophages has been associated with dedifferentiation, lymph node metastases, and reduced survival (65). It is important to note, however, that most of the studies analyzing the immune milieu of DTC have used papillary thyroid cancer (PTC) tumor samples (66-69). Myeloid-derived suppressor cells are associated with aggressive characteristics of differentiated thyroid cancer and are related to poor prognosis (65).

 

Figure 4. Immune infiltrates differ in different types of thyroid cancer [Modified by Garcia-Alvarez et al, (70)]. TAM, tumor-associated macrophages; MDSC, myeloid-derived suppressor cell.

 

The dendritic cells, which play a critical role in antigen presentation, are increased in PTC, while neutrophils are found in more aggressive thyroid cancers (such as poorly differentiated or anaplastic). In addition, natural killer cells that play an important role in immunosurveillance are also increased in PTC and are negatively correlated with tumor stage, while lymphocytic infiltration is associated with better overall survival and low recurrence rate (71,72).

 

Cytokines, which may be produced by thyroid follicular cells and by immune cells infiltrating thyroid tumors, are also related to tumor development. IL-1 and IL-6 stimulate thyroid cell proliferation and tumor growth, while TGF-β, which is a suppressive cytokine, is overexpressed in aggressive cancers (73). In addition, multiple chemokines may be secreted by thyroid cancer or immune cells and affect chemiotaxis, angiogenesis, and lymphangiogenesis (73).

 

Immunomodulatory proteins, such as programmed death-ligand 1 (PDL1), cytotoxic T-lymphocyte antigen 4 (CTLA-4), T cell immunoglobulin and mucin-domain containing-3 (TIM-3), lymphocyte activation gene-3 (LAG-3), and T cell immunoglobulin and ITIM domain (TIGIT), which are considered major immune coinhibitory receptors and promising immunotherapeutic targets in cancer treatment, are also expressed in thyroid cancer, being associated with more aggressive tumor characteristics and a poor prognosis (64). Programmed death-ligand 1 staining by immunohistochemistry has shown higher expression in ATC than in other subtypes (70). Six cohort studies have been published to-date reporting positive PD-L1 expression, varying between 22% and 65%, this being higher compared to that detected in DTC and poorly differentiated thyroid cancer (74-79). TIM-3 expression was observed in 48% of patients with MTC, and in the majority of cases (84.4%) its expression was restricted to tumor cells (80). Other coinhibitory receptors, such as LAG-3 and T cell TIGIT, were observed in a lesser percentage of cases (approximately 3%) (80). Nevertheless, results from clinical trials with immunotherapy as monotherapy or combinations have shown limited efficacy (70). In one phase Ib KEYNOTE-028 trial assessing the efficacy of pembrolizumab in patients with PD-L1+ (membranous staining on ≥1%) locally advanced or metastatic follicular or papillary thyroid cancer, pembrolizumab achieved an objective response rate of 9% and a median progression-free survival of 7 months (81). Several clinical trials further investigating the efficacy of combination therapy of immune checkpoint inhibitors are currently ongoing.

 

IMMUNE SYSTEM AND DIABETES MELLITUS

 

The immune system plays a crucial role in the pathogenesis of both type 1 and type 2 diabetes.

Diabetes type 1, which is immune-mediated in more than 95% of cases, is an organ-specific autoimmune disease characterized by lymphocytic infiltration and inflammation that leads to pancreatic β-cell destruction and absolute insulin deficiency (82). The immune system’s attack on pancreatic β-cells is usually triggered by a number of factors, including genetic predisposition and environmental triggers such as viral infections (e.g., Coxsackie B4, mumps, and rubella) or dietary compounds (e.g., cow’s milk) (82). The process involves the activation of immune cells, particularly T cells, which recognize self-autoantigens in pancreatic β-cells and initiate an immune response.

 

On the other hand, in type 2 diabetes, the immune system also plays a different role from that in DM1. Chronic low-grade inflammation, often associated with obesity, leads to immune cell activation and the release of proinflammatory cytokines. These cytokines interfere with the normal functioning of insulin and promote insulin resistance. Macrophages infiltrate adipose tissue and release inflammatory molecules, further exacerbating insulin resistance with increasing adiposity. The immune system, thus, contributes to the development and progression of insulin resistance and eventually promotes the onset of DM2.

 

Understanding the intricate relations between the immune system and diabetes pathogenesis is essential for the development of effective treatments and interventions for the management of both types of diabetes.

 

Diabetes Type 1

 

The susceptibility to develop DM1 is associated with multiple alleles of the major histocompatibility complex MHC I and II locus. More than 90% of patients with DM1 express either HLA DR3, DQ2 or DR4, or DQ8 (83), whereas HLA haplotype DR2, DQ6 is protective against DM1 development.

 

The primary pathological presentation of DM1 is inflammation of the pancreatic islets, also known as insulitis, caused by infiltration of immune cells, including CD4 and CD8 T cells along with B cells (84-86). Although the initial events triggering autoreactive responses remain unclear, presentation of pancreatic islet autoantigens by the associated MHC class II molecules contribute to priming and expansion of pathogenic T cells.

 

CD4 T helper cells are required for the development of the autoimmune process in the pancreatic islets, while CD8 cytotoxic T cells are the cells responsible for β-cell destruction (Figure 5). T cell receptors recognize peptides bound to MHC molecules on the surface of antigen-presenting cells (B-cells, dendritic cells, and macrophages). Each T cell then generates a unique receptor for the recognition of an autoantigen presented in the MHC molecule. The interaction between T cell receptor/autoantigen/MHC leads to activation of the T cells.

 

Figure 5. T cell mediated destruction of β- cells in diabetes type 1. CD4+ effector T cells recognize an insulin or islet peptide presented by diabetes-risk conferring HLA-DQ8 or DR4 on APCs, migrate to the pancreas and promote pancreatic β-cell destruction. Inside the pancreatic islets CD8+ effector T cells recognize islet antigens presented by HLA-A2 leading to the destruction of the β-cells. [Modified by Mitchel et al (87)]. TCR, T cell receptor; APC, antigen presenting cell; HLA-A2, human leukocyte antigen-A2.

 

Three different mechanisms have been proposed to explain T cell activation in DM1. One mechanism is thought to involve molecular mimicry-activated T cell proliferation. The hypothesis for this mechanism is based on the assumption that epitopes of proteins expressed by infectious agents can be shared by unrelated molecules encoded by host genes (88). A second mechanism that may trigger molecular mimicry-activated T cell proliferation is “bystander” T cell proliferation. This mechanism involves the stimulation of non-antigen-specific T cells by various cytokines during infection, simply because they are in the area. The cytokines thought to be involved in this nonspecific stimulation are IFN-α and IFN-β (89). The 3rd mechanism might involve a superantigen-mediated T cell proliferation mechanism: this theory proposes that autoreactive T cells can be inappropriately primed to react against self-structures through an encounter with a superantigen (90).

 

Multiple autoantigens in the pancreatic islets have been identified, including non-specific islet cell autoantigens (ICA), insulin, glutamic acid decarboxylase 65 (GAD65), insulinoma antigen-2 (IA-2), heat shock protein (HSP), islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), imogen-38, zinc transporter-8 (ZnT8), and the most recently identified pancreatic duodenal homeobox factor 1 (PDX1), chromogranin A (CHGA), and islet amyloid polypeptide (IAPP) (91). Autoantibodies against these autoantigens may be detected long before the onset of hyperglycemia and usually decline during the course of the disease (92).

 

Most patients diagnosed with DM1 have circulating islet cell autoantibodies directed against pancreatic islet autoantigens. However, although the detection of autoantibodies may be useful for DM1 diagnosis and prediction, it is the cellular immune system that eventually infiltrates the pancreatic islets and causes β-cell destruction. In turn, further β-cell destruction leads to more self-antigen presentation and ensuing amplification of the immune response (82,93,94).

 

Among several proinflammatory cytokines, IL-21, produced by CD8 T cells, is required for the development of DM1, while TNF-α may also be involved (95,96). IL-6 plays an important role in the pathogenesis of vitiligo-associated DM1 (97). In contrast, in vivo experiments with non-obese diabetic mice have shown that IL-4, produced by Th2 cells, may be protective against developing diabetes (95,96,98,99).

 

Understanding both the pathophysiology and the regulatory mechanisms involved in DM1 is a critical step towards the development of antigen-specific, β-cell-directed, immunomodulatory or cellular treatment modalities (100).

 

Investigations into the efficacy and safety of various immunotherapeutic strategies against the development of DM1 have been carried out in recent clinical trials and are still ongoing in current trials (101). Among them, T cell-directed therapies that aim at a favorable balance between effector T cell depletion and regulatory T cell preservation have shown the most promising results. Teplizumab, an anti-CD3-directed monoclonal antibody, was the first immunomodulatory agent to demonstrate a significant delay in disease progression in high-risk individuals before clinical onset (102) and has recently (November 17, 2022) been approved by the FDA as the first disease-modifying therapy for DM1 in adults and in children aged 8 years and older.

 

The Enigma of Pancreatic α-Cell Resistance in Diabetes Mellitus Type 1

 

Although both insulin-producing β-cells and glucagon-producing α-cells of the pancreatic islets share a similar embryonic origin and are directly exposed to the deleterious immune signals in DM1, it appears that the immune system selectively destroys β-cells, while α-cells survive even in long-term DM1 (103). α-Cells are located in close proximity to β-cells in human pancreatic islets (104), creating a closed communication loop that regulates their function and secretory capacity (105).  Dysfunction of α-cells plays a significant role in the pathogenesis of both types of diabetes (106). It has long been proposed that the reduced functional β-cell mass in DM1 and the consequent hypoglycemia were the key mechanisms indirectly inducing dysfunction of α-cells in diabetes (107). Advanced molecular technology of the last decade has, however, challenged this notion, showing that dysfunction of α-cells in diabetes is not secondary to β-cell pathology but is instead directly immune-induced (108-110). Despite this dysfunction, α-cells exhibit a remarkable autoimmune resistance that enables them to survive over β-cells in long-standing diabetes (103,111). Recent studies using single-cell RNA sequencing (109,112) have demonstrated important differences between these two cell types in terms of expression of: i) anti-apoptotic genes, ii) endoplasmic reticulum (ER) stress-related genes, III) innate immune response genes, and iv) antiviral response genes, all of which render α-cells less immunogenic and more resistant to viral infections and ER stress (113). In addition, CD8+ T cells invading the islets in DM1 are reactive to preproinsulin but not to glucagon. Furthermore, although β-cells are essential to life (neither humans nor animal models can survive without them), mice with 98% α-cell ablation retain near-normal glucose homeostasis (114): this points to the knowledge gap we have, from an evolutionary point of view, regarding the question of why β-cells are more fragile than α-cells. Certainly, greater understanding of the underlying mechanisms responsible for the autoimmune resistance of α-cells is critical since it is likely to reveal intracellular pathways amenable to therapeutic interventions that would increase the resistance of the β-cells themselves to the immune attack of the host’s immune system.

 

Diabetes Type 2

 

DM2 accounts for 90% of cases of diabetes worldwide (115). The increasing prevalence of DM2 around the world has been largely attributed to an unhealthy lifestyle and resultant development of obesity and overweight (116). Obesity is strongly related to DM2 mainly through inducing insulin resistance in the insulin sensitive tissues in the periphery.

 

The concept that a smoldering inflammatory process plays an important part in the pathogenesis of DM2 (117) has attracted much attention and is supported by evidence of inflammation in islets, adipose tissue, liver, and muscle that can provoke insulin resistance and β-cell dysfunction (118-120). Adipose tissue is characterized by infiltration by macrophages and other immune cells that produce cytokines and chemokines and contribute to the development of local and systemic chronic low-grade inflammation, this inflammatory milieu being the link between obesity, insulin resistance, and diabetes mellitus (121,122).

 

Earlier in vivo studies demonstrated that levels of TNF-α (123-125), IL-6, C-reactive protein, plasminogen activator inhibitor, and other inflammation mediators were elevated in adipose tissue and plasma of obese mice (126,127). It was also observed that these inflammatory mediators, together with saturated free fatty acids and reactive oxygen species (ROS), inhibited serine phosphorylation of the insulin receptor substrates (IRS-1 and 2) (128-130) in insulin sensitive tissues, such as adipose tissue and the liver (131), promoting insulin resistance (Figure 6). TNF-α is associated with increased release of free fatty acids by adipose tissue and leads to impaired insulin secretion and signaling (123,132).

 

Figure 6. Insulin resistance and inflammation in diabetes type 2. Insulin binds to insulin receptor (IR) in insulin sensitive tissues, and autophosphorylates tyrosine molecules of IRS-1 and -2 substrates. In the presence of obesity, and hyperlipidemia, the influx of free fatty acids, inflammatory cytokines and glucose activates IKKβ and JNK, which are the mediators for stress and inflammatory. In turn IKKβ and JNK inhibit tyrosine phosphorylation of IRS1 and 2 and promote transcriptional activation of genes related to inflammatory and stress responses resulting in insulin resistance. [Modified by Berbudi et al (133)]. IRS 1 -2, insulin receptor substrates; ER, Endoplasmic reticulum; IKKβ, inhibitory kappa B kinase β; JNK1 and c-Jun N-terminal kinase I.

 

In line with these early in vivo studies, studies in humans have shown that elevated levels of i) nonspecific indicators of inflammation, such as white cell count, fibrinogen, and CRP levels (134-136), ii) markers of reduced fibrinolysis, such as plasminogen activator inhibitor-1 (PAI-1), and tissue plasminogen activator (tPA), iii) von Willebrand factor (vWf), which is a marker of endothelial injury, and iv) early markers of inflammation, such as monocyte chemotactic protein-1 (MCP-1), IL-8, and interferon-γ-inducible protein-10 (137), were predictive of DM2 development.

 

Furthermore, CRP and/or IL-6 were associated with the incidence of DM2 independently of adiposity or insulin resistance (136,138,139). Visceral adipose tissue appears to be a major source of circulating IL-6 in humans, and obese people with insulin resistance display high levels of plasma IL-6 concentration, also predictive of DM2 development (113). TNF-α is also increased in obese individuals with insulin resistance (124,140) and it plays a major role in the pathogenesis of obesity-linked DM2 (141).

 

At the cellular level, chronic exposure of adipocytes to low doses of TNF-α led to a dramatic decrease in insulin-stimulated auto-phosphorylation of the IRS 1-2 (142). Treatment of cultured murine adipocytes with TNF-α induced serine phosphorylation of IRS-1 and convert IRS-1 into an inhibitor of IR tyrosine kinase activity in vitro. TNF-α has also been shown to downregulate glucose transporter GLUT4 mRNA levels in adipocyte and myocyte cultures as well (125,143,144).

 

Oxidative stress, as a result of increased cytokine levels in DM2, is also thought to play an important role in activating inflammatory genes (145,146) (Figure 6). It is possible that oxidative stress markers do not adequately reflect the impact of increased ROS on β-cells or insulin signaling, while inflammatory, procoagulant or endothelial dysfunction markers are more specific to the pathophysiology of hyperglycemia (145,146). Hasnain et al. showed that islet-endogenous and exogenous IL-22 suppressed oxidative and ER stress caused by cytokines or glucolipotoxicity in mouse and human β-cells by regulating oxidative stress pathways. In obese mice, antibody neutralization of IL-23 or IL-24 partially reduced β-cell ER stress and improved glucose tolerance, whereas IL-22 administration modulated oxidative stress regulatory genes in islets, suppressed ER stress and inflammation, promoted secretion of high-quality efficacious insulin, and fully restored glucose homeostasis, followed by reinstitution of insulin sensitivity (147).

 

The chemokine system is also associated with obesity and insulin resistance. MCP-1, which acts on monocytes, macrophages, T cells and NK cells, is increased in obese compared to lean subjects and is related to non-alcoholic fatty liver disease and other lipid overload states (148-153). A short-term program of 4-month lifestyle modification significantly decreases MCP-1 levels, with favorable effects on the glycemic and lipid profile (151).

 

Collectively, these findings supported the investigation of new therapeutic approaches that target inflammation to ameliorate diabetes and its complications. Regarding the latter, it is important to note that multiple sources of evidence support a pathogenic connection between rheumatoid arthritis (RA) and the mechanisms of DM2, via formation of a vicious circle that is perpetuated by impaired glucose metabolism and inflammation. In this context, ongoing clinical studies have shown that the inhibition of interleukin IL-1 and IL-6 may allow the treatment of RA and concomitant T2D at the same time (154).

 

Treatment with anakinra, a recombinant form of human IL-1 receptor antagonist that works as a competitive inhibitor of IL-1β, achieved significant improvements of glycemia and secretory function of β-cells (155,156), which was maintained after anakinra withdrawal during a 39-week follow-up (157). Canakinumab, a monoclonal antibody against IL-1β, significantly reduced inflammatory proteins, such as CRP, IL-6, and fibrinogen, in patients with DM2 and high cardiovascular risk (158,159), while regarding glycemic control, it reduced values of HbA1c during the first 6–9 months of treatment, without, however, consistent long-term benefits (159). Antagonists of IL-6 receptor tocilizumab and sarilumab have also been investigated in patients with RA with or without concomitant DM2. Tocilizumab showed a positive effect on insulin resistance in some studies (160-162), while other studies failed to report any beneficial effect on glycemic control (163,164). The efficacy of sarilumab was assessed in a post hoc analysis (165) of three randomized clinical trials in patients with RA with or without DM2 (166-168). Sarilumab, as monotherapy or in combination, significantly reduced HbA1c compared to adalimumab monotherapy or placebo plus methotrexate/convectional DMARDs in patients with RA and DM2 (165).

 

Several studies have analyzed the effects of TNF inhibitors (i.e., adalimumab, etanercept, and infliximab) on glucose metabolism, demonstrating a potential favorable effect on insulin resistance and insulin sensitivity (169,170). In a meta-analysis combining data from 22 randomized controlled trials and three cohort studies (171), new-onset DM2 was delayed in RA patients treated with TNF inhibitors.

 

The Effect of Diabetes on the Immune System

 

Like a mirror image, chronic hyperglycemia in diabetic patients impairs the host’s immune response, which in turn fails to control the spread of invading pathogens, rendering diabetic patients more susceptible to infections (133,172). Both innate immune response defects and defective adaptive immune response are implicated in this incapacity of the immune system to defend against invading pathogens in patients with diabetes. Several mechanisms have been proposed by experimental and human studies including: i) leukocyte recruitment inhibition (173,174), ii) defective pathogen recognition (175), iii) dysfunction of neutrophils (176-178), macrophages resulting in impairment of phagocytosis (179), iv) functional defects in natural killer cells (180), and v) dysfunction of complement activation (181).

 

OSTEOPOROSIS AND THE IMMUNE SYSTEM

 

Osteoporosis is a clinical condition characterized by low bone mass and impaired bone microarchitecture associated with an increased risk of fragility fractures. Growing evidence of the last few decades has demonstrated the effect of the immune system on bone metabolism, leading to the emergence of the new field of osteoimmunology (182-185) and immunology of osteoporosis (named as immunoporosis) (184-188).

 

States of immune dysfunction such as immunodeficiency, inflammatory diseases, or immune response to infections are associated with increased osteoclastic bone resorption and, therefore, increased bone loss and increased fracture risk.

 

Bone Remodeling and Bone Cells

 

Bone remodeling is a dynamic and continuous process that is responsible for the maintenance of skeletal health throughout life. It involves three consecutive phases: i) osteoclast-mediated bone resorption; ii) the reversal phase, during which mesenchymal derived osteoblasts are recruited to the bone site of bone resorption; and iii) osteoblast-mediated bone formation. The two processes of bone resorption and bone formation are tightly coupled and under control of the matrix-embedded osteocytes, capable of sensing and integrating mechanical and chemical signals from their environment to regulate bone formation and resorption at the bone surfaces.

 

Osteoclasts originate from the same myeloid precursor that derives macrophage and dendritic cells and are specialized in bone degradation (186). Osteoblasts are the main bone-forming cells and are derived from mesenchymal stem cells. Osteoclast formation and differentiation is regulated by macrophage colony-stimulating factor (M-CSF) and the receptor activator of nuclear factor-kB (RANK) ligand (RANKL) produced by osteoblasts and osteocytes (189,190). Osteocytes are a significant source of RANKL and its decoy receptor, osteoprotegerin (OPG), which binds to RANKL, preventing its interaction with RANK, while they also secrete the Wnt signaling inhibitor sclerostin, which regulates bone formation (189). RANKL is additionally expressed by fibroblasts and immune cells, including antigen-stimulated T cells and dendritic cells (191-193), while OPG is also produced by B lymphocytes and dendritic cells (194).

 

Inflammatory Diseases

 

Activated T cells increase the production of TNF-α and RANKL and stimulate osteoclastogenesis during inflammation (182-185,192-195). Multiple cytokines may promote osteoclastogenesis mainly by regulating the RANK/RANKL/OPG axis. TNF-α, IL-1, IL-6, IL-7, IL-11, Il-17, and IL-23 promote osteoclast differentiation, while IFN-α, IFN-γ, IL-3, IL-4, IL-10, IL-27, and IL-33 are considered anti-osteoclastogenic cytokines that protect bone integrity (195). Th17 cells are considered an osteoclastogenic subset of T cells as they enhance osteoclastogenesis by secreting IL-1, IL-6, Il-17, RANKL, TNF-α, and IFN-γ. Activation of Th2 leads to enhanced production of PTH and promotes the anabolic activity of osteoblasts in several inflammatory states. Furthermore, Th2 lymphocytes are associated with a low RANKL/OPG ratio and inhibition of bone loss (196). In addition, β-cells produce RANKL and OPG and may influence bone formation and absorption, while it has been observed that in HIV infected patients, there is an altered β-cell RANKL/OPG ratio that is inversely correlated with BMD (197).

 

Interleukin 6, produced by both stromal and osteoblastic cells (198) in response to stimulation by systemic hormones such as PTH, PTH-related peptide (PTH-rP), thyroid hormones, and 1,25-dihydroxyvitamin D3 and other cytokines (i.e., TGF-β, IL-1, and TNF-α), plays a major role in osteoclast development and function. Increased IL-6 levels contribute significantly to the abnormal bone resorption observed in patients with multiple myeloma (199), Paget’s disease of bone (200), rheumatoid arthritis (201), and Langerhans cell histiocytosis (202). Effects of increased osteoclast-induced bone resorption are not solely attributable to IL-6, but to all IL-6 family cytokines (203).

 

TNF-α has also been shown to induce bone resorption and plays an important part in inflammatory bone diseases (192). TNF-α promotes RANKL expression in osteoclast precursors and the formation of multinucleated osteoclasts in the presence of M-CSF. Furthermore, TNF-α increases RANKL and M-CSF expression in osteoblasts, stromal cells, and T lymphocytes, while RANKL can also enhance TNF-α mediated osteoclastogenesis (195). IL-1β increases RANKL expression and stimulates osteoclast formation and bone resorption while also promoting TNF-α induced osteoclastogenesis (204,205).

 

Postmenopausal Osteoporosis

 

Estrogen deficiency is a state of increased bone remodeling associated with an increased rate of bone resorption relative to bone formation, resulting in net bone loss. It has been shown that estrogen deficiency-induced bone loss has a complex mechanism mainly involving the immune system rather than a direct effect of estrogen on bone cells (206). Estrogen loss during menopause is associated with an expansion of T and B lymphocytes (207,208) leading to increased production of RANKL (209). In addition, an increased level of proinflammatory cytokines, including TNF-a and IL-1b, is observed in postmenopausal women (210,211). In addition, B lymphocytes may partially contribute to trabecular bone loss in postmenopausal osteoporosis (212). In the absence of estrogens, dendritic cells live longer, increasing their expression of IL-7 and IL-15. In turn, IL-7 and IL-15 induce IL-17 and TNF-a production in a subset of memory T cells, independent of antigen activation (213). These proinflammatory cytokines contribute to inflammation-induced bone loss in postmenopausal osteoporosis by activating low-grade inflammation. In contrast, Treg cells have a bone-protective role in postmenopausal osteoporosis (210). However, it has been shown that Th17/Treg balance is disturbed in estrogen deficiency. Treg cells lose their immunosuppressive function and convert to Th17 cells, which explains the imbalance of Th17/Treg in postmenopausal osteoporosis (214).

 

Senile Osteoporosis

 

Senile osteoporosis, on the other hand, is a low-bone turnover disease with decreased bone resorption and significantly reduced bone formation (215), which commonly occurs in older people, above 65, and affects both males and females. In recent years, it was demonstrated that aging is usually accompanied by systemic low-grade chronic inflammation and enhanced inflammatory mediators, such as IL-6 and TNF-a (216). A recent study found that senescent immune cells, such as macrophages and neutrophils, accumulate in bone marrow during aging in rats and mice (217). The senescent macrophages and neutrophils repress osteogenesis by promoting bone marrow mesenchymal stromal cell adipogenesis. In addition to directly inhibiting osteogenesis, the senescent immune cells contribute to chronic inflammation, thus leading to inflammatory bone resorption (217). Aging can tilt the balance of Th1/Th2 toward Th2 cells, resulting in an increased inflammatory response (218) and low-level chronic inflammation, ultimately leading to continuous bone loss.

 

Thyrotoxicosis-Induced Osteoporosis

 

IL-6 and IL-8 play a major role in thyrotoxicosis-induced osteoporosis and are increased in patients with thyrotoxicosis due to Graves’ disease or toxic multinodular goiter (219). In addition, patients with thyroid carcinoma on TSH suppressive therapy have significantly increased circulating levels of IL-6 and IL-8 compared to controls (219), which are tightly associated with serum T3 and fT4 concentrations. Both IL-6 and IL-8 have also been shown to be released by human bone marrow stromal cell cultures containing osteoblast progenitor cells in response to T3 (196). TNF-α elevations due to low TSH signaling in human hyperthyroidism also contribute to the bone loss that has traditionally been attributed solely to high thyroid hormone levels (220). Hyperthyroid mice lacking TSHR had greater bone resorption than hyperthyroid wild-type mice, demonstrating that the absence of TSH signaling contributes to low bone mass (221) in the hyperthyroid state.

 

Primary Hyperparathyroidism-Induced Osteoporosis

 

Bone resorption in primary hyperparathyroidism (PHP) also appears to be related to immune system effects. Circulating levels of IL-6 and TNF-α, which are significantly increased in patients with PHP, are strongly correlated with biochemical markers of resorption, returning to normal after successful parathyroidectomy (222). The hypothesis that IL-6 mediates the catabolic effects of parathyroid hormone (PTH) on the skeleton has been further strengthened by the finding that neutralizing IL-6 in vivo attenuates PTH-induced bone resorption in mice, while the resorptive response to PTH was also reduced in IL-6 knockout mice (223). Furthermore, it has been observed that transplantation of parathyroid from humans with hyperparathyroidism to mice lacking T cells was not associated with bone loss, suggesting a possible role of T lymphocytes in PTH-related osteoporosis (224). A direct action of PTH on T lymphocytes may also contribute, as deletion of the PTH receptor from T cells failed to induce bone loss (225). It has been proposed that PTH action on T cells results in secretion of TNF-α and, in combination with RANKL increase and OPG suppression, guides their differentiation to Th17 subsets, with subsequent IL-17 secretion and further RANKL amplification (226).

 

Drug-Induced Osteoporosis

 

The immune cells are also involved in drug-induced osteoporosis, such as glucocorticoid and chemotherapy-induced osteoporosis. A recent study demonstrated that glucocorticoid-induced osteoporosis could not be induced in T cell-deficient mice, while re-establishment was found to be possible by transferring splenic T cells from wild-type mice (227).

 

Cyclophosphamide, is a chemotherapy drug that causes immunosuppression and is associated with increased risk of osteoporosis (228-230). By improving the functional status of immune cells in an immunosuppressive mouse model induced by cyclophosphamide, bone loss was dramatically reduced (231), pointing to the possible contribution of immune cells in cyclophosphamide-induced osteoporosis.

 

EFFECTS OF THE IMMUNE SYSTEM ON THE STRESS SYSTEM

 

The Hypothalamic-Pituitary-Adrenal (HPA) Axis

 

The relations between the immune and the stress systems are complex and bidirectional, denoting that while stress can affect immune function, immune responses can also influence stress levels through various ways and mechanisms.

 

INFLAMMATION

 

During acute inflammation, the immune system is activated in response to infection or injury and releases proinflammatory cytokines and other inflammation-related factors into the central nervous system (CNS), plasma, and endocrine glands. Inflammatory cytokines, such as TNF-α, IL-1, and IL-6, produced by a variety of cells, including monocytes, macrophages, astrocytes, endothelial cells, and fibroblasts, activate the HPA axis leading to an increase of corticotropin-releasing hormone (CRH), adrenocorticotrophic hormone (ACTH) and, finally, glucocorticoids (38, 184). In turn, increased circulating levels of glucocorticoids exert suppressive effects on the inflammatory reaction, controlling the immune response and helping the organism to reach its prior healthy homeostasis (232). Similarly, in acute stress, the amplitude and synchronization of CRH secretory pulses is increased, and this is reflected in the levels of ACTH and cortisol in the systemic circulation (233).

 

The proinflammatory cytokine IL-1, especially its β form, is probably the most important molecule capable of modulating cerebral functions during systemic and localized inflammation. Systemic IL-1β injection activates the neurons involved in the control of autonomic functions, and neutralizing antibodies or IL-1 receptor antagonists are capable of preventing numerous responses during inflammatory stimuli (234). Similarly to IL-1β, intravenous IL-6 stimulates the hypothalamic-pituitary unit, leading to the secretion of cortisol by the adrenal glands and subsequent termination of the inflammatory cascade (235). All three inflammatory cytokines (IL-1, IL-6, and TNF-α) have the capacity to activate the HPA-axis, but it appears that IL-6 is the most critical component of this cascade. Studies in rats have demonstrated that immunoneutralization of IL-6 abolishes the effects of the other two cytokines on the HPA-axis (236). TNF-α and IL-1, on the other hand, stimulate the production of IL-6, which in turn stimulates the HPA-axis. The final end-product of HPA activation, glucocorticoids, inhibit IL-6 secretion at the transcriptional level through interaction of the ligand-activated glucocorticoid receptor with nuclear factor-kappa B, creating a negative feedback loop. In this way, IL-6 stimulates glucocorticoid release to control inflammation, and glucocorticoids subsequently inhibit IL-6 release through a negative feedback loop preventing uncontrolled and potentially harmful sequalae of inflammatory mechanisms, including tissue damage (237,238).

 

In an older study involving patients with Cushing disease studied before and after transsphenoidal adenectomy, plasma IL-6 concentration was increased when patients were hypocortisolemic, experiencing symptoms of glucocorticoid deficiency, as part of the “steroid withdrawal syndrome” (i.e., pyrexia, headache, anorexia, nausea, fatigue, malaise, arthralgias, myalgias, and somnolence of variable degree). Notably, IL-6 levels did not increase in patients who did not become hypocortisolemic after surgery, while following glucocorticoid replacement, a dramatic decrease of IL-6 levels concomitantly with relief of the observed symptoms was reported (239).

 

While acute stimulation with IL-6 activates the HPA axis mainly through the hypothalamic CRH neurons, chronic exposure to IL-6 may also directly stimulate the pituitary corticotropic cells and the adrenal cells via CRH receptor-independent mechanisms (240).

 

In vivo experiments have shown a stimulatory effect of IL-6 on cortisol production by the adrenal cortex in the absence of CRH and subsequent activation of the HPA axis (240) in cytomegalovirus-infected mice (241), as well as in murine colitis (242). In addition, experiments in mice deficient in CRH (CRH KO) and deficient in both CRH and IL-6 (CRH KO)/IL-6 KO) have demonstrated that IL-6 during prolonged immunological challenge may surpass CRH in augmentation of adrenal function (240). Protein expression of IL-6 and IL-6 receptor has been detected in primary cultures of human adrenocortical cells depleted of macrophages (CD68-positive cells), predominantly in the zona reticularis but also in the zona fasciculata and in single cells within the zona glomerulosa and the medulla (243). Moreover, IL-6 was able to induce adrenal steroidogenesis in vitro in a time- and dose-dependent manner in the absence of macrophages, suggesting that IL-6 may be a long-term stimulator of steroidogenesis but with no acute effects (243).

 

In humans, IL-6 may stimulate cortisol release directly at the level of the adrenal gland in long-term stress situations (244), and the same may also apply in chronic inflammatory diseases, although direct evidence is still lacking (245). In a recent clinical study, increased daily, and especially evening, saliva cortisol secretion, in the context of the acute viral infection COVID-19, appeared to be mostly driven by hypersecretion of IL-6, independently of ACTH (246). However, the hypothesis that IL-6 may partially replace ACTH when there is an acute requirement for increased cortisol secretion has yet to be tested.

 

PROLONGED OR CHRONIC STRESS AND INFLAMMATION

 

Acute versus chronic stress and inflammation are distinct conditions that exert extremely different effects on the immune system, each altering its function in a distinct manner. Chronic stress is associated with a blunted circadian cortisol rhythm, a suppressed inflammatory response, and a shift from Th1   to Th 2 and a Th reg to Th 17 immunity. It has clearly been shown that in chronic stress, endogenous glucocorticoids fail to terminate the stress response and, in fact, cause body composition changes reminiscent of those in hypercortisolism, such as visceral adiposity, sarcopenia, and osteoporosis (247-249).

 

On the other hand, following a period of intense stress, there may be glucocorticoid-induced suppression of the HPA axis (238). In this case, long-term “inadequate” cortisol secretion may unleash its inhibitory effect on immune system activation, resulting in immune dysregulation and expression of sickness-syndrome manifestations. In this case, the post-stress sustained HPA axis suppression and hypocortisolism is reminiscent of the clinical picture of the intrinsic hypocortisolemia of primary adrenal insufficiency (Addison’s disease), associated with immune perturbations due to failure of cortisol to appropriately suppress the increased secretion of proinflammatory cytokines (239,250).

 

The subsequent protracted lack of axis recovery due to a post-illness state of hypocortisolism in probably predisposed individuals may explain the underlying pathophysiologic mechanisms responsible for some post-viral infection sickness syndromes (232,251), such as long COVID syndrome (252).

 

AUTOIMMUNE DISORDERS  

 

In some cases, chronic stress can contribute to the development or exacerbation of autoimmune disorders. Without appropriate cortisol regulation in cases of chronic stress and chronic inflammation, the organism fails to downregulate inflammatory processes, contributing to a vicious cycle where stress, inflammation, and compromised HPA axis function may result in the development of chronic immune-mediated inflammatory diseases, such as rheumatoid arthritis. Stress can potentially trigger or worsen these conditions by dysregulating the immune response.

 

Cortisol diurnal secretion displays a circadian rhythm in healthy individuals, with the highest levels in the morning and a gradual decline throughout the day, reaching the lowest levels around midnight (253). During acute stress (254-256) or infection (237,257), the rhythm is disturbed and the afternoon and/or midnight cortisol levels do not drop. A disturbed circadian cortisol rhythm with abnormally high afternoon and night cortisol levels was found in patients with even mild or moderate COVID-19 compared to healthy controls (246). However, although not systematically studied, the adrenal response to chronic versus acute exposure to proinflammatory cytokines appears to follow a different pattern.

 

In patients with rheumatoid arthritis, symptoms follow circadian rhythms with impaired function due to pain and joint stiffness being most severe in the early morning (258,259) because of increased proinflammatory cytokines, such as TNF-a and IL-6, that occur during late night hours (260,261). This explains the inverse relation between diurnal variation of circulating IL-6 and glucocorticoid levels (262). Increased endogenous nocturnal secretion of cortisol could alleviate these morning symptoms, but this is not the case in most patients with rheumatoid arthritis (263-270). To clarify this issue, several studies have been carried out reporting that the optimal time for delivery of glucocorticoid treatment would be during the night in order to target the effects of nocturnal proinflammatory stimuli (271-274).

 

PSYCHOLOGICAL WELL-BEING

 

The immune system also plays a role in maintaining overall psychological well-being. Chronic stress and its impact on the immune system can increase susceptibility to mental health problems such as anxiety and depression. Conversely, psychological well-being can improve the human body’s immune responses, enhance resistance to diseases (including infectious diseases), and improve mental health (275,276). Changes in psychological status of patients with Alzheimer’s induced significant differences in their immune response (277).

In addition, long-term practice of meditation was shown to decrease stress reactivity and exert a favorable therapeutic effect in chronic inflammatory conditions characterized by neurogenic inflammation (278), while joyful activities such as singing were able to boost the immune response in cancer patients and family members (279).

 

ADRENAL MEDULLA

 

The chromaffin cells of the adrenal medulla play a role in stress response by secreting catecholamines and various biologically active peptides (238). As the stress response starts, rapidly augmented secretion of norepinephrine and epinephrine is initiated, followed by activation of the HPA axis and increased release of CRH and ACTH and secretion of glucocorticoids (280). CRH and norepinephrine stimulate the secretion of each other through CRH-R1 and α1-noradrenergic receptors, respectively (281).

 

Cytokines TNF-α, IL-1, and IFN-γ act directly on chromaffin cells (282-285). It has also been demonstrated that cytokines regulate the secretion of various peptides that are co-secreted with catecholamines, such as vasoactive intestinal peptide (VIP), galanin and secretogranin II, enkephalin and neuropeptide Y (283,285). IL-6 directly modulates the secretion of catecholamines and neuropeptides by chromaffin cells and therefore influences the adrenal stress response. It has been hypothesized that medullary peptides may serve as paracrine modulators of glucocorticoid production (286). It has also been shown that IL-6 increases intracellular Ca2+ concentration and induces catecholamine secretion in rat carotid body glomus cells, a finding which has finally confirmed the relations between IL-6 and catecholamine secretion (287). Furthermore, IL-10 is a critical target downstream of epinephrine and norepinephrine which limits inflammation (288). On the other hand, norepinephrine may act directly on macrophages and dendritic cells to suppress inflammatory cytokine secretion through primarily the β2-adrenergic receptors that are expressed by both innate and adaptive immune cells (289,290).

 

Although scientific advances of the last decade have shed light on the previously unrecognized major role of the catecholamines epinephrine and norepinephrine in controlling the immune system, much remains to be discovered, and further revelations will reveal new therapeutic targets in the management of inflammation.

 

ACKNOWLEDGEMENTS

 

This chapter is an update of a previous chapter and the authors would like to thank Professor Gregory Kaltsas, an author of the prior chapter.

 

REFERENCES

 

  1. Poland GA, Quill H, Togias A. Understanding the human immune system in the 21st century: the Human Immunology Project Consortium. Vaccine. 2013;31(28):2911-2912.
  2. Li Q, Wang B, Mu K, Zhang JA. The pathogenesis of thyroid autoimmune diseases: New T lymphocytes - Cytokines circuits beyond the Th1-Th2 paradigm. J Cell Physiol. 2019;234(3):2204-2216.
  3. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21(2):137-148.
  4. McLachlan SM, Rapoport B. Breaking tolerance to thyroid antigens: changing concepts in thyroid autoimmunity. Endocr Rev. 2014;35(1):59-105.
  5. Colaci M, Malatino L, Antonelli A, Fallahi P, Giuggioli D, Ferri C. Endocrine disorders associated with hepatitis C virus chronic infection. Rev Endocr Metab Disord. 2018;19(4):397-403.
  6. Wang B, Shao X, Song R, Xu D, Zhang JA. The Emerging Role of Epigenetics in Autoimmune Thyroid Diseases. Front Immunol. 2017;8:396.
  7. Boguslawska J, Godlewska M, Gajda E, Piekielko-Witkowska A. Cellular and molecular basis of thyroid autoimmunity. Eur Thyroid J. 2022;11(1).
  8. Coscia F, Taler-Vercic A, Chang VT, Sinn L, O'Reilly FJ, Izore T, Renko M, Berger I, Rappsilber J, Turk D, Lowe J. The structure of human thyroglobulin. Nature. 2020;578(7796):627-630.
  9. McLachlan SM, Rapoport B. Thyroid Autoantibodies Display both "Original Antigenic Sin" and Epitope Spreading. Front Immunol. 2017;8:1845.
  10. Godlewska M, Banga PJ. Thyroid peroxidase as a dual active site enzyme: Focus on biosynthesis, hormonogenesis and thyroid disorders of autoimmunity and cancer. Biochimie. 2019;160:34-45.
  11. Godlewska M, Gawel D, Buckle AM, Banga JP. Thyroid Peroxidase Revisited - What's New? Horm Metab Res. 2019;51(12):765-769.
  12. Jaume JC, Burek CL, Hoffman WH, Rose NR, McLachlan SM, Rapoport B. Thyroid peroxidase autoantibody epitopic 'fingerprints' in juvenile Hashimoto's thyroiditis: evidence for conservation over time and in families. Clin Exp Immunol. 1996;104(1):115-123.
  13. Morshed SA, Ando T, Latif R, Davies TF. Neutral antibodies to the TSH receptor are present in Graves disease and regulate selective signaling cascades. Endocrinology. 2010;151(11):5537-5549.
  14. Sanders J, Miguel RN, Bolton J, Bhardwaja A, Sanders P, Nakatake N, Evans M, Furmaniak J, Smith BR. Molecular interactions between the TSH receptor and a Thyroid-stimulating monoclonal autoantibody. Thyroid. 2007;17(8):699-706.
  15. Sanders P, Young S, Sanders J, Kabelis K, Baker S, Sullivan A, Evans M, Clark J, Wilmot J, Hu X, Roberts E, Powell M, Nunez Miguel R, Furmaniak J, Rees Smith B. Crystal structure of the TSH receptor (TSHR) bound to a blocking-type TSHR autoantibody. J Mol Endocrinol. 2011;46(2):81-99.
  16. Eleftheriadou AM, Mehl S, Renko K, Kasim RH, Schaefer JA, Minich WB, Schomburg L. Re-visiting autoimmunity to sodium-iodide symporter and pendrin in thyroid disease. Eur J Endocrinol. 2020;183(6):571-580.
  17. Fallahi P, Ferrari SM, Ragusa F, Ruffilli I, Elia G, Paparo SR, Antonelli A. Th1 Chemokines in Autoimmune Endocrine Disorders. J Clin Endocrinol Metab. 2020;105(4).
  18. Meeusen EN, Premier RR, Brandon MR. Tissue-specific migration of lymphocytes: a key role for Th1 and Th2 cells? Immunol Today. 1996;17(9):421-424.
  19. Davies TF, Andersen S, Latif R, Nagayama Y, Barbesino G, Brito M, Eckstein AK, Stagnaro-Green A, Kahaly GJ. Graves disease. Nat Rev Dis Primers. 2020;6(1):52.
  20. Wiersinga WM. Advances in treatment of active, moderate-to-severe Graves ophthalmopathy. Lancet Diabetes Endocrinol. 2017;5(2):134-142.
  21. Neumann S, Krieger CC, Gershengorn MC. Targeting TSH and IGF-1 Receptors to Treat Thyroid Eye Disease. Eur Thyroid J. 2020;9(Suppl 1):59-65.
  22. Taheri M, Eghtedarian R, Dinger ME, Ghafouri-Fard S. Dysregulation of non-coding RNAs in autoimmune thyroid disease. Exp Mol Pathol. 2020;117:104527.
  23. Liu Y, Cui X, Wang S, Liu J, Zhao N, Huang M, Qin J, Li Y, Shan Z, Teng W. Elevated MicroRNA-326 Levels Regulate the IL-23/IL-23R/Th17 Cell Axis in Hashimoto's Thyroiditis by Targeting a Disintegrin and Metalloprotease 17. Thyroid. 2020;30(9):1327-1337.
  24. Zheng L, Zhuang C, Wang X, Ming L. Serum miR-146a, miR-155, and miR-210 as potential markers of Graves disease. J Clin Lab Anal. 2018;32(2).
  25. Jiang X, Wang Y, Li X, He L, Yang Q, Wang W, Liu J, Zha B. Microarray profile of B cells from Graves disease patients reveals biomarkers of proliferation. Endocr Connect. 2020;9(5):405-417.
  26. Xiong S, Peng H, Ding X, Wang X, Wang L, Wu C, Wang S, Xu H, Liu Y. Circular RNA Expression Profiling and the Potential Role of hsa_circ_0089172 in Hashimoto's Thyroiditis via Sponging miR125a-3p. Mol Ther Nucleic Acids. 2019;17:38-48.
  27. Talebi S, Karimifar M, Heidari Z, Mohammadi H, Askari G. The effects of synbiotic supplementation on thyroid function and inflammation in hypothyroid patients: A randomized, double‑blind, placebo‑controlled trial. Complement Ther Med. 2020;48:102234.
  28. Biscarini F, Masetti G, Muller I, Verhasselt HL, Covelli D, Colucci G, Zhang L, Draman MS, Okosieme O, Taylor P, Daumerie C, Burlacu MC, Marino M, Ezra DG, Perros P, Plummer S, Eckstein A, Salvi M, Marchesi JR, Ludgate M. Gut Microbiome Associated With Graves Disease and Graves Orbitopathy: The INDIGO Multicenter European Study. J Clin Endocrinol Metab. 2023;108(8):2065-2077.
  29. Olivieri A, De Angelis S, Vaccari V, Valensise H, Magnani F, Stazi MA, Cotichini R, Gilardi E, Cordeddu V, Sorcini M, Boirivant M. Postpartum thyroiditis is associated with fluctuations in transforming growth factor-beta1 serum levels. J Clin Endocrinol Metab. 2003;88(3):1280-1284.
  30. Weetman AP, Bennett GL, Wong WL. Thyroid follicular cells produce interleukin-8. J Clin Endocrinol Metab. 1992;75(1):328-330.
  31. Premawardhana LD, Parkes AB, Ammari F, John R, Darke C, Adams H, Lazarus JH. Postpartum thyroiditis and long-term thyroid status: prognostic influence of thyroid peroxidase antibodies and ultrasound echogenicity. J Clin Endocrinol Metab. 2000;85(1):71-75.
  32. Papanicolaou DA. Euthyroid Sick Syndrome and the role of cytokines. Rev Endocr Metab Disord. 2000;1(1-2):43-48.
  33. Simons PJ, Delemarre FG, Drexhage HA. Antigen-presenting dendritic cells as regulators of the growth of thyrocytes: a role of interleukin-1beta and interleukin-6. Endocrinology. 1998;139(7):3148-3156.
  34. Asakawa H, Hanafusa T, Kobayashi T, Takai S, Kono N, Tarui S. Interferon-gamma reduces the thyroid peroxidase content of cultured human thyrocytes and inhibits its increase induced by thyrotropin. J Clin Endocrinol Metab. 1992;74(6):1331-1335.
  35. Ashizawa K, Yamashita S, Nagayama Y, Kimura H, Hirayu H, Izumi M, Nagataki S. Interferon-gamma inhibits thyrotropin-induced thyroidal peroxidase gene expression in cultured human thyrocytes. J Clin Endocrinol Metab. 1989;69(2):475-477.
  36. Poth M, Tseng YC, Wartofsky L. Inhibition of TSH activation of human cultured thyroid cells by tumor necrosis factor: an explanation for decreased thyroid function in systemic illness? Thyroid. 1991;1(3):235-240.
  37. Yamashita S, Kimura H, Ashizawa K, Nagayama Y, Hirayu H, Izumi M, Nagataki S. Interleukin-1 inhibits thyrotrophin-induced human thyroglobulin gene expression. J Endocrinol. 1989;122(1):177-183.
  38. van der Poll T, Romijn JA, Wiersinga WM, Sauerwein HP. Tumor necrosis factor: a putative mediator of the sick euthyroid syndrome in man. J Clin Endocrinol Metab. 1990;71(6):1567-1572.
  39. Chopra IJ, Sakane S, Teco GN. A study of the serum concentration of tumor necrosis factor-alpha in thyroidal and nonthyroidal illnesses. J Clin Endocrinol Metab. 1991;72(5):1113-1116.
  40. Girvent M, Maestro S, Hernandez R, Carajol I, Monne J, Sancho JJ, Gubern JM, Sitges-Serra A. Euthyroid sick syndrome, associated endocrine abnormalities, and outcome in elderly patients undergoing emergency operation. Surgery. 1998;123(5):560-567.
  41. Davis PJ. Cytokines and growth factors and thyroid hormone. Curr Opin Endocrinol Diabetes Obes. 2008;15(5):428.
  42. Baur A, Bauer K, Jarry H, Kohrle J. Effects of proinflammatory cytokines on anterior pituitary 5'-deiodinase type I and type II. J Endocrinol. 2000;167(3):505-515.
  43. Calikoglu M, Sahin G, Unlu A, Ozturk C, Tamer L, Ercan B, Kanik A, Atik U. Leptin and TNF-alpha levels in patients with chronic obstructive pulmonary disease and their relationship to nutritional parameters. Respiration. 2004;71(1):45-50.
  44. Park MC, Lee SW, Choi ST, Park YB, Lee SK. Serum leptin levels correlate with interleukin-6 levels and disease activity in patients with ankylosing spondylitis. Scand J Rheumatol. 2007;36(2):101-106.
  45. Lechan RM, Fekete C. Feedback regulation of thyrotropin-releasing hormone (TRH): mechanisms for the non-thyroidal illness syndrome. J Endocrinol Invest. 2004;27(6 Suppl):105-119.
  46. Harris M, Aschkenasi C, Elias CF, Chandrankunnel A, Nillni EA, Bjoorbaek C, Elmquist JK, Flier JS, Hollenberg AN. Transcriptional regulation of the thyrotropin-releasing hormone gene by leptin and melanocortin signaling. J Clin Invest. 2001;107(1):111-120.
  47. Harjai KJ, Licata AA. Effects of amiodarone on thyroid function. Ann Intern Med. 1997;126(1):63-73.
  48. Cohen-Lehman J, Dahl P, Danzi S, Klein I. Effects of amiodarone therapy on thyroid function. Nat Rev Endocrinol. 2010;6(1):34-41.
  49. Goundan PN, Lee SL. Thyroid effects of amiodarone: clinical update. Curr Opin Endocrinol Diabetes Obes. 2020;27(5):329-334.
  50. Newman CM, Price A, Davies DW, Gray TA, Weetman AP. Amiodarone and the thyroid: a practical guide to the management of thyroid dysfunction induced by amiodarone therapy. Heart. 1998;79(2):121-127.
  51. Narayana SK, Woods DR, Boos CJ. Management of amiodarone-related thyroid problems. Ther Adv Endocrinol Metab. 2011;2(3):115-126.
  52. Loh KC. Amiodarone-induced thyroid disorders: a clinical review. Postgrad Med J. 2000;76(893):133-140.
  53. Martino E, Bartalena L, Bogazzi F, Braverman LE. The effects of amiodarone on the thyroid. Endocr Rev. 2001;22(2):240-254.
  54. Bartalena L, Brogioni S, Grasso L, Bogazzi F, Burelli A, Martino E. Treatment of amiodarone-induced thyrotoxicosis, a difficult challenge: results of a prospective study. J Clin Endocrinol Metab. 1996;81(8):2930-2933.
  55. Mosher MC. Amiodarone-induced hypothyroidism and other adverse effects. Dimens Crit Care Nurs. 2011;30(2):87-93.
  56. Piga M, Serra A, Boi F, Tanda ML, Martino E, Mariotti S. Amiodarone-induced thyrotoxicosis. A review. Minerva Endocrinol. 2008;33(3):213-228.
  57. Kent WD, Hall SF, Isotalo PA, Houlden RL, George RL, Groome PA. Increased incidence of differentiated thyroid carcinoma and detection of subclinical disease. CMAJ. 2007;177(11):1357-1361.
  58. Cunha LL, Marcello MA, Ward LS. The role of the inflammatory microenvironment in thyroid carcinogenesis. Endocr Relat Cancer. 2014;21(3):R85-R103.
  59. Zeng R, Lyu Y, Zhang G, Shou T, Wang K, Niu H, Yan X. Positive effect of RORgammat on the prognosis of thyroid papillary carcinoma patients combined with Hashimoto's thyroiditis. Am J Transl Res. 2018;10(10):3011-3024.
  60. Kim EY, Kim WG, Kim WB, Kim TY, Kim JM, Ryu JS, Hong SJ, Gong G, Shong YK. Coexistence of chronic lymphocytic thyroiditis is associated with lower recurrence rates in patients with papillary thyroid carcinoma. Clin Endocrinol (Oxf). 2009;71(4):581-586.
  61. Zivancevic-Simonovic S, Mihaljevic O, Majstorovic I, Popovic S, Markovic S, Milosevic-Djordjevic O, Jovanovic Z, Mijatovic-Teodorovic L, Mihajlovic D, Colic M. Cytokine production in patients with papillary thyroid cancer and associated autoimmune Hashimoto thyroiditis. Cancer Immunol Immunother. 2015;64(8):1011-1019.
  62. Veit F, Graf D, Momberger S, Helmich-Kapp B, Ruschenburg I, Peters A, Kussmann J, Saeger W, Schmidt KW, Toetsch M, Nestler K, Mann K. Papillary Thyroid Cancer and Coexisting Autoimmune Thyroiditis. Horm Metab Res. 2017;49(11):869-872.
  63. Chowdhury S, Veyhl J, Jessa F, Polyakova O, Alenzi A, MacMillan C, Ralhan R, Walfish PG. Programmed death-ligand 1 overexpression is a prognostic marker for aggressive papillary thyroid cancer and its variants. Oncotarget. 2016;7(22):32318-32328.
  64. Dias Lopes NM, Mendonca Lens HH, Armani A, Marinello PC, Cecchini AL. Thyroid cancer and thyroid autoimmune disease: A review of molecular aspects and clinical outcomes. Pathol Res Pract. 2020;216(9):153098.
  65. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20(11):651-668.
  66. Na KJ, Choi H. Immune landscape of papillary thyroid cancer and immunotherapeutic implications. Endocr Relat Cancer. 2018;25(5):523-531.
  67. Ryder M, Ghossein RA, Ricarte-Filho JC, Knauf JA, Fagin JA. Increased density of tumor-associated macrophages is associated with decreased survival in advanced thyroid cancer. Endocr Relat Cancer. 2008;15(4):1069-1074.
  68. French JD, Kotnis GR, Said S, Raeburn CD, McIntyre RC, Jr., Klopper JP, Haugen BR. Programmed death-1+ T cells and regulatory T cells are enriched in tumor-involved lymph nodes and associated with aggressive features in papillary thyroid cancer. J Clin Endocrinol Metab. 2012;97(6):E934-943.
  69. Gogali F, Paterakis G, Rassidakis GZ, Kaltsas G, Liakou CI, Gousis P, Neonakis E, Manoussakis MN, Liapi C. Phenotypical analysis of lymphocytes with suppressive and regulatory properties (Tregs) and NK cells in the papillary carcinoma of thyroid. J Clin Endocrinol Metab. 2012;97(5):1474-1482.
  70. Garcia-Alvarez A, Hernando J, Carmona-Alonso A, Capdevila J. What is the status of immunotherapy in thyroid neoplasms? Front Endocrinol (Lausanne). 2022;13:929091.
  71. Mould RC, van Vloten JP, AuYeung AWK, Karimi K, Bridle BW. Immune responses in the thyroid cancer microenvironment: making immunotherapy a possible mission. Endocr Relat Cancer. 2017;24(12):T311-T329.
  72. Pan J, Ye F, Yu C, Zhu Q, Li J, Zhang Y, Tian H, Yao Y, Zhu M, Shen Y, Zhu F, Wang Y, Zhou X, Guo G, Wu Y. Papillary Thyroid Carcinoma Landscape and Its Immunological Link With Hashimoto Thyroiditis at Single-Cell Resolution. Front Cell Dev Biol. 2021;9:758339.
  73. Galdiero MR, Varricchi G, Marone G. The immune network in thyroid cancer. Oncoimmunology. 2016;5(6):e1168556.
  74. Ahn S, Kim TH, Kim SW, Ki CS, Jang HW, Kim JS, Kim JH, Choe JH, Shin JH, Hahn SY, Oh YL, Chung JH. Comprehensive screening for PD-L1 expression in thyroid cancer. Endocr Relat Cancer. 2017;24(2):97-106.
  75. Cameselle-Garcia S, Abdulkader-Sande S, Sanchez-Ares M, Rodriguez-Carnero G, Garcia-Gomez J, Gude-Sampedro F, Abdulkader-Nallib I, Cameselle-Teijeiro JM. PD-L1 expression and immune cells in anaplastic carcinoma and poorly differentiated carcinoma of the human thyroid gland: A retrospective study. Oncol Lett. 2021;22(1):553.
  76. Cantara S, Bertelli E, Occhini R, Regoli M, Brilli L, Pacini F, Castagna MG, Toti P. Blockade of the programmed death ligand 1 (PD-L1) as potential therapy for anaplastic thyroid cancer. Endocrine. 2019;64(1):122-129.
  77. Chintakuntlawar AV, Rumilla KM, Smith CY, Jenkins SM, Foote RL, Kasperbauer JL, Morris JC, Ryder M, Alsidawi S, Hilger C, Bible KC. Expression of PD-1 and PD-L1 in Anaplastic Thyroid Cancer Patients Treated With Multimodal Therapy: Results From a Retrospective Study. J Clin Endocrinol Metab. 2017;102(6):1943-1950.
  78. Zwaenepoel K, Jacobs J, De Meulenaere A, Silence K, Smits E, Siozopoulou V, Hauben E, Rolfo C, Rottey S, Pauwels P. CD70 and PD-L1 in anaplastic thyroid cancer - promising targets for immunotherapy. Histopathology. 2017;71(3):357-365.
  79. Wu H, Sun Y, Ye H, Yang S, Lee SL, de las Morenas A. Anaplastic thyroid cancer: outcome and the mutation/expression profiles of potential targets. Pathol Oncol Res. 2015;21(3):695-701.
  80. Strickler JH, Hanks BA, Khasraw M. Tumor Mutational Burden as a Predictor of Immunotherapy Response: Is More Always Better? Clin Cancer Res. 2021;27(5):1236-1241.
  81. Mehnert JM, Varga A, Brose MS, Aggarwal RR, Lin CC, Prawira A, de Braud F, Tamura K, Doi T, Piha-Paul SA, Gilbert J, Saraf S, Thanigaimani P, Cheng JD, Keam B. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced, PD-L1-positive papillary or follicular thyroid cancer. BMC Cancer. 2019;19(1):196.
  82. Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010;464(7293):1293-1300.
  83. Erlich H, Valdes AM, Noble J, Carlson JA, Varney M, Concannon P, Mychaleckyj JC, Todd JA, Bonella P, Fear AL, Lavant E, Louey A, Moonsamy P, Type 1 Diabetes Genetics C. HLA DR-DQ haplotypes and genotypes and type 1 diabetes risk: analysis of the type 1 diabetes genetics consortium families. Diabetes. 2008;57(4):1084-1092.
  84. Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG. Analysis of islet inflammation in human type 1 diabetes. Clin Exp Immunol. 2009;155(2):173-181.
  85. Vandamme C, Kinnunen T. B cell helper T cells and type 1 diabetes. Scand J Immunol. 2020;92(4):e12943.
  86. Pugliese A. Insulitis in the pathogenesis of type 1 diabetes. Pediatr Diabetes. 2016;17 Suppl 22(Suppl Suppl 22):31-36.
  87. Mitchell AM, Michels AW. Self-Antigens Targeted by Regulatory T Cells in Type 1 Diabetes. Int J Mol Sci. 2022;23(6).
  88. Atkinson MA, Bowman MA, Campbell L, Darrow BL, Kaufman DL, Maclaren NK. Cellular immunity to a determinant common to glutamate decarboxylase and coxsackie virus in insulin-dependent diabetes. J Clin Invest. 1994;94(5):2125-2129.
  89. Tough DF, Borrow P, Sprent J. Induction of bystander T cell proliferation by viruses and type I interferon in vivo. Science. 1996;272(5270):1947-1950.
  90. Katz JD, Benoist C, Mathis D. T helper cell subsets in insulin-dependent diabetes. Science. 1995;268(5214):1185-1188.
  91. Han S, Donelan W, Wang H, Reeves W, Yang LJ. Novel autoantigens in type 1 diabetes. Am J Transl Res. 2013;5(4):379-392.
  92. Ziegler AG, Nepom GT. Prediction and pathogenesis in type 1 diabetes. Immunity. 2010;32(4):468-478.
  93. Burton AR, Vincent E, Arnold PY, Lennon GP, Smeltzer M, Li CS, Haskins K, Hutton J, Tisch RM, Sercarz EE, Santamaria P, Workman CJ, Vignali DA. On the pathogenicity of autoantigen-specific T-cell receptors. Diabetes. 2008;57(5):1321-1330.
  94. Serreze DV, Fleming SA, Chapman HD, Richard SD, Leiter EH, Tisch RM. B lymphocytes are critical antigen-presenting cells for the initiation of T cell-mediated autoimmune diabetes in nonobese diabetic mice. J Immunol. 1998;161(8):3912-3918.
  95. Sutherland AP, Van Belle T, Wurster AL, Suto A, Michaud M, Zhang D, Grusby MJ, von Herrath M. Interleukin-21 is required for the development of type 1 diabetes in NOD mice. Diabetes. 2009;58(5):1144-1155.
  96. Lechleitner M, Koch T, Herold M, Hoppichler F. Relationship of tumor necrosis factor-alpha plasma levels to metabolic control in type 1 diabetes. Diabetes Care. 1999;22(10):1749.
  97. Farhan J, Al-Shobaili HA, Zafar U, Al Salloom A, Meki AR, Rasheed Z. Interleukin-6: a possible inflammatory link between vitiligo and type 1 diabetes. Br J Biomed Sci. 2014;71(4):151-157.
  98. Zhang J, Huang Z, Sun R, Tian Z, Wei H. IFN-gamma induced by IL-12 administration prevents diabetes by inhibiting pathogenic IL-17 production in NOD mice. J Autoimmun. 2012;38(1):20-28.
  99. Arif S, Moore F, Marks K, Bouckenooghe T, Dayan CM, Planas R, Vives-Pi M, Powrie J, Tree T, Marchetti P, Huang GC, Gurzov EN, Pujol-Borrell R, Eizirik DL, Peakman M. Peripheral and islet interleukin-17 pathway activation characterizes human autoimmune diabetes and promotes cytokine-mediated beta-cell death. Diabetes. 2011;60(8):2112-2119.
  100. Jacobsen LM, Newby BN, Perry DJ, Posgai AL, Haller MJ, Brusko TM. Immune Mechanisms and Pathways Targeted in Type 1 Diabetes. Curr Diab Rep. 2018;18(10):90.
  101. Smigoc Schweiger D. Recent Advances in Immune-based Therapies for Type 1 Diabetes. Horm Res Paediatr. 2022.
  102. An Anti-CD3 Antibody, Teplizumab, in Relatives at Risk for Type 1 Diabetes. N Engl J Med. 2020;382(6):586.
  103. Eizirik DL, Szymczak F, Mallone R. Why does the immune system destroy pancreatic beta-cells but not alpha-cells in type 1 diabetes? Nat Rev Endocrinol. 2023;19(7):425-434.
  104. Bosco D, Armanet M, Morel P, Niclauss N, Sgroi A, Muller YD, Giovannoni L, Parnaud G, Berney T. Unique arrangement of alpha- and beta-cells in human islets of Langerhans. Diabetes. 2010;59(5):1202-1210.
  105. Campbell JE, Newgard CB. Mechanisms controlling pancreatic islet cell function in insulin secretion. Nat Rev Mol Cell Biol. 2021;22(2):142-158.
  106. Gromada J, Chabosseau P, Rutter GA. The alpha-cell in diabetes mellitus. Nat Rev Endocrinol. 2018;14(12):694-704.
  107. Brissova M, Haliyur R, Saunders D, Shrestha S, Dai C, Blodgett DM, Bottino R, Campbell-Thompson M, Aramandla R, Poffenberger G, Lindner J, Pan FC, von Herrath MG, Greiner DL, Shultz LD, Sanyoura M, Philipson LH, Atkinson M, Harlan DM, Levy SE, Prasad N, Stein R, Powers AC. alpha Cell Function and Gene Expression Are Compromised in Type 1 Diabetes. Cell Rep. 2018;22(10):2667-2676.
  108. Shapira SN, Naji A, Atkinson MA, Powers AC, Kaestner KH. Understanding islet dysfunction in type 2 diabetes through multidimensional pancreatic phenotyping: The Human Pancreas Analysis Program. Cell Metab. 2022;34(12):1906-1913.
  109. Fasolino M, Schwartz GW, Patil AR, Mongia A, Golson ML, Wang YJ, Morgan A, Liu C, Schug J, Liu J, Wu M, Traum D, Kondo A, May CL, Goldman N, Wang W, Feldman M, Moore JH, Japp AS, Betts MR, Consortium H, Faryabi RB, Naji A, Kaestner KH, Vahedi G. Single-cell multi-omics analysis of human pancreatic islets reveals novel cellular states in type 1 diabetes. Nat Metab. 2022;4(2):284-299.
  110. Kaestner KH, Powers AC, Naji A, Consortium H, Atkinson MA. NIH Initiative to Improve Understanding of the Pancreas, Islet, and Autoimmunity in Type 1 Diabetes: The Human Pancreas Analysis Program (HPAP). Diabetes. 2019;68(7):1394-1402.
  111. Doliba NM, Rozo AV, Roman J, Qin W, Traum D, Gao L, Liu J, Manduchi E, Liu C, Golson ML, Vahedi G, Naji A, Matschinsky FM, Atkinson MA, Powers AC, Brissova M, Kaestner KH, Stoffers DA, Consortium H. alpha Cell dysfunction in islets from nondiabetic, glutamic acid decarboxylase autoantibody-positive individuals. J Clin Invest. 2022;132(11).
  112. Colli ML, Ramos-Rodriguez M, Nakayasu ES, Alvelos MI, Lopes M, Hill JLE, Turatsinze JV, Coomans de Brachene A, Russell MA, Raurell-Vila H, Castela A, Juan-Mateu J, Webb-Robertson BM, Krogvold L, Dahl-Jorgensen K, Marselli L, Marchetti P, Richardson SJ, Morgan NG, Metz TO, Pasquali L, Eizirik DL. An integrated multi-omics approach identifies the landscape of interferon-alpha-mediated responses of human pancreatic beta cells. Nat Commun. 2020;11(1):2584.
  113. Chen CW, Guan BJ, Alzahrani MR, Gao Z, Gao L, Bracey S, Wu J, Mbow CA, Jobava R, Haataja L, Zalavadia AH, Schaffer AE, Lee H, LaFramboise T, Bederman I, Arvan P, Mathews CE, Gerling IC, Kaestner KH, Tirosh B, Engin F, Hatzoglou M. Adaptation to chronic ER stress enforces pancreatic beta-cell plasticity. Nat Commun. 2022;13(1):4621.
  114. Thorel F, Damond N, Chera S, Wiederkehr A, Thorens B, Meda P, Wollheim CB, Herrera PL. Normal glucagon signaling and beta-cell function after near-total alpha-cell ablation in adult mice. Diabetes. 2011;60(11):2872-2882.
  115. Shaw JE, Sicree RA, Zimmet PZ. Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract. 2010;87(1):4-14.
  116. Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW, Malanda B. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:271-281.
  117. Donath MY, Storling J, Berchtold LA, Billestrup N, Mandrup-Poulsen T. Cytokines and beta-cell biology: from concept to clinical translation. Endocr Rev. 2008;29(3):334-350.
  118. Varma V, Yao-Borengasser A, Rasouli N, Nolen GT, Phanavanh B, Starks T, Gurley C, Simpson P, McGehee RE, Jr., Kern PA, Peterson CA. Muscle inflammatory response and insulin resistance: synergistic interaction between macrophages and fatty acids leads to impaired insulin action. Am J Physiol Endocrinol Metab. 2009;296(6):E1300-1310.
  119. Ehses JA, Boni-Schnetzler M, Faulenbach M, Donath MY. Macrophages, cytokines and beta-cell death in Type 2 diabetes. Biochem Soc Trans. 2008;36(Pt 3):340-342.
  120. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J Clin Invest. 2006;116(7):1793-1801.
  121. Sell H, Habich C, Eckel J. Adaptive immunity in obesity and insulin resistance. Nat Rev Endocrinol. 2012;8(12):709-716.
  122. Nikolajczyk BS, Jagannathan-Bogdan M, Shin H, Gyurko R. State of the union between metabolism and the immune system in type 2 diabetes. Genes Immun. 2011;12(4):239-250.
  123. Hotamisligil GS. The role of TNFalpha and TNF receptors in obesity and insulin resistance. J Intern Med. 1999;245(6):621-625.
  124. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest. 1995;95(5):2409-2415.
  125. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259(5091):87-91.
  126. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab. 2004;89(6):2548-2556.
  127. Halberg N, Wernstedt-Asterholm I, Scherer PE. The adipocyte as an endocrine cell. Endocrinol Metab Clin North Am. 2008;37(3):753-768, x-xi.
  128. Gao Z, He Q, Peng B, Chiao PJ, Ye J. Regulation of nuclear translocation of HDAC3 by IkappaBalpha is required for tumor necrosis factor inhibition of peroxisome proliferator-activated receptor gamma function. J Biol Chem. 2006;281(7):4540-4547.
  129. Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, Shoelson SE. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta. Science. 2001;293(5535):1673-1677.
  130. Aguirre V, Uchida T, Yenush L, Davis R, White MF. The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser(307). J Biol Chem. 2000;275(12):9047-9054.
  131. Ye J. Regulation of PPARgamma function by TNF-alpha. Biochem Biophys Res Commun. 2008;374(3):405-408.
  132. Ruan H, Miles PD, Ladd CM, Ross K, Golub TR, Olefsky JM, Lodish HF. Profiling gene transcription in vivo reveals adipose tissue as an immediate target of tumor necrosis factor-alpha: implications for insulin resistance. Diabetes. 2002;51(11):3176-3188.
  133. Berbudi A, Rahmadika N, Tjahjadi AI, Ruslami R. Type 2 Diabetes and its Impact on the Immune System. Curr Diabetes Rev. 2020;16(5):442-449.
  134. Duncan BB, Schmidt MI, Offenbacher S, Wu KK, Savage PJ, Heiss G. Factor VIII and other hemostasis variables are related to incident diabetes in adults. The Atherosclerosis Risk in Communities (ARIC) Study. Diabetes Care. 1999;22(5):767-772.
  135. Schmidt MI, Duncan BB, Sharrett AR, Lindberg G, Savage PJ, Offenbacher S, Azambuja MI, Tracy RP, Heiss G. Markers of inflammation and prediction of diabetes mellitus in adults (Atherosclerosis Risk in Communities study): a cohort study. Lancet. 1999;353(9165):1649-1652.
  136. Festa A, D'Agostino R, Jr., Tracy RP, Haffner SM. Elevated levels of acute-phase proteins and plasminogen activator inhibitor-1 predict the development of type 2 diabetes: the insulin resistance atherosclerosis study. Diabetes. 2002;51(4):1131-1137.
  137. Herder C, Baumert J, Thorand B, Koenig W, de Jager W, Meisinger C, Illig T, Martin S, Kolb H. Chemokines as risk factors for type 2 diabetes: results from the MONICA/KORA Augsburg study, 1984-2002. Diabetologia. 2006;49(5):921-929.
  138. Lee CC, Adler AI, Sandhu MS, Sharp SJ, Forouhi NG, Erqou S, Luben R, Bingham S, Khaw KT, Wareham NJ. Association of C-reactive protein with type 2 diabetes: prospective analysis and meta-analysis. Diabetologia. 2009;52(6):1040-1047.
  139. Lin JD, Chao TC, Weng HF, Lin KD. The roles of cytokines and retinoic acid in the regulation of human thyroid cancer cell growth. Cytokine. 1998;10(7):536-539.
  140. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am J Physiol Endocrinol Metab. 2001;280(5):E745-751.
  141. Kirwan JP, Hauguel-De Mouzon S, Lepercq J, Challier JC, Huston-Presley L, Friedman JE, Kalhan SC, Catalano PM. TNF-alpha is a predictor of insulin resistance in human pregnancy. Diabetes. 2002;51(7):2207-2213.
  142. Vilcek J, Lee TH. Tumor necrosis factor. New insights into the molecular mechanisms of its multiple actions. J Biol Chem. 1991;266(12):7313-7316.
  143. Cornelius P, Lee MD, Marlowe M, Pekala PH. Monokine regulation of glucose transporter mRNA in L6 myotubes. Biochem Biophys Res Commun. 1989;165(1):429-436.
  144. Stephens JM, Pekala PH. Transcriptional repression of the GLUT4 and C/EBP genes in 3T3-L1 adipocytes by tumor necrosis factor-alpha. J Biol Chem. 1991;266(32):21839-21845.
  145. Meigs JB, Larson MG, Fox CS, Keaney JF, Jr., Vasan RS, Benjamin EJ. Association of oxidative stress, insulin resistance, and diabetes risk phenotypes: the Framingham Offspring Study. Diabetes Care. 2007;30(10):2529-2535.
  146. Rosen P, Nawroth PP, King G, Moller W, Tritschler HJ, Packer L. The role of oxidative stress in the onset and progression of diabetes and its complications: a summary of a Congress Series sponsored by UNESCO-MCBN, the American Diabetes Association and the German Diabetes Society. Diabetes Metab Res Rev. 2001;17(3):189-212.
  147. Hasnain SZ, Borg DJ, Harcourt BE, Tong H, Sheng YH, Ng CP, Das I, Wang R, Chen AC, Loudovaris T, Kay TW, Thomas HE, Whitehead JP, Forbes JM, Prins JB, McGuckin MA. Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress. Nat Med. 2014;20(12):1417-1426.
  148. Huma ZE, Sanchez J, Lim HD, Bridgford JL, Huang C, Parker BJ, Pazhamalil JG, Porebski BT, Pfleger KDG, Lane JR, Canals M, Stone MJ. Key determinants of selective binding and activation by the monocyte chemoattractant proteins at the chemokine receptor CCR2. Sci Signal. 2017;10(480).
  149. Catalan V, Gomez-Ambrosi J, Ramirez B, Rotellar F, Pastor C, Silva C, Rodriguez A, Gil MJ, Cienfuegos JA, Fruhbeck G. Proinflammatory cytokines in obesity: impact of type 2 diabetes mellitus and gastric bypass. Obes Surg. 2007;17(11):1464-1474.
  150. Franca CN, Izar MCO, Hortencio MNS, do Amaral JB, Ferreira CES, Tuleta ID, Fonseca FAH. Monocyte subtypes and the CCR2 chemokine receptor in cardiovascular disease. Clin Sci (Lond). 2017;131(12):1215-1224.
  151. Gokulakrishnan K, Ranjani H, Weber MB, Pandey GK, Anjana RM, Balasubramanyam M, Prabhakaran D, Tandon N, Narayan KM, Mohan V. Effect of lifestyle improvement program on the biomarkers of adiposity, inflammation and gut hormones in overweight/obese Asian Indians with prediabetes. Acta Diabetol. 2017;54(9):843-852.
  152. Townsend SA, Newsome PN. Review article: new treatments in non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2017;46(5):494-507.
  153. Chistiakov DA, Melnichenko AA, Grechko AV, Myasoedova VA, Orekhov AN. Potential of anti-inflammatory agents for treatment of atherosclerosis. Exp Mol Pathol. 2018;104(2):114-124.
  154. Di Muzio C, Cipriani P, Ruscitti P. Rheumatoid Arthritis Treatment Options and Type 2 Diabetes: Unravelling the Association. BioDrugs. 2022;36(6):673-685.
  155. Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, Mandrup-Poulsen T, Donath MY. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356(15):1517-1526.
  156. Malozowski S, Sahlroot JT. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;357(3):302-303; author reply 303.
  157. Larsen CM, Faulenbach M, Vaag A, Ehses JA, Donath MY, Mandrup-Poulsen T. Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes. Diabetes Care. 2009;32(9):1663-1668.
  158. Ridker PM, Howard CP, Walter V, Everett B, Libby P, Hensen J, Thuren T, Group CPI. Effects of interleukin-1beta inhibition with canakinumab on hemoglobin A1c, lipids, C-reactive protein, interleukin-6, and fibrinogen: a phase IIb randomized, placebo-controlled trial. Circulation. 2012;126(23):2739-2748.
  159. Everett BM, Donath MY, Pradhan AD, Thuren T, Pais P, Nicolau JC, Glynn RJ, Libby P, Ridker PM. Anti-Inflammatory Therapy With Canakinumab for the Prevention and Management of Diabetes. J Am Coll Cardiol. 2018;71(21):2392-2401.
  160. Otsuka Y, Kiyohara C, Kashiwado Y, Sawabe T, Nagano S, Kimoto Y, Ayano M, Mitoma H, Akahoshi M, Arinobu Y, Niiro H, Akashi K, Horiuchi T. Effects of tumor necrosis factor inhibitors and tocilizumab on the glycosylated hemoglobin levels in patients with rheumatoid arthritis; an observational study. PLoS One. 2018;13(4):e0196368.
  161. Ogata A, Morishima A, Hirano T, Hishitani Y, Hagihara K, Shima Y, Narazaki M, Tanaka T. Improvement of HbA1c during treatment with humanised anti-interleukin 6 receptor antibody, tocilizumab. Ann Rheum Dis. 2011;70(6):1164-1165.
  162. Castaneda S, Remuzgo-Martinez S, Lopez-Mejias R, Genre F, Calvo-Alen J, Llorente I, Aurrecoechea E, Ortiz AM, Triguero A, Blanco R, Llorca J, Gonzalez-Gay MA. Rapid beneficial effect of the IL-6 receptor blockade on insulin resistance and insulin sensitivity in non-diabetic patients with rheumatoid arthritis. Clin Exp Rheumatol. 2019;37(3):465-473.
  163. Makrilakis K, Fragiadaki K, Smith J, Sfikakis PP, Kitas GD. Interrelated reduction of chemerin and plasminogen activator inhibitor-1 serum levels in rheumatoid arthritis after interleukin-6 receptor blockade. Clin Rheumatol. 2015;34(3):419-427.
  164. Tournadre A, Pereira B, Dutheil F, Giraud C, Courteix D, Sapin V, Frayssac T, Mathieu S, Malochet-Guinamand S, Soubrier M. Changes in body composition and metabolic profile during interleukin 6 inhibition in rheumatoid arthritis. J Cachexia Sarcopenia Muscle. 2017;8(4):639-646.
  165. Genovese MC, Burmester GR, Hagino O, Thangavelu K, Iglesias-Rodriguez M, John GS, Gonzalez-Gay MA, Mandrup-Poulsen T, Fleischmann R. Interleukin-6 receptor blockade or TNFalpha inhibition for reducing glycaemia in patients with RA and diabetes: post hoc analyses of three randomised, controlled trials. Arthritis Res Ther. 2020;22(1):206.
  166. Genovese MC, Fleischmann R, Kivitz AJ, Rell-Bakalarska M, Martincova R, Fiore S, Rohane P, van Hoogstraten H, Garg A, Fan C, van Adelsberg J, Weinstein SP, Graham NM, Stahl N, Yancopoulos GD, Huizinga TW, van der Heijde D. Sarilumab Plus Methotrexate in Patients With Active Rheumatoid Arthritis and Inadequate Response to Methotrexate: Results of a Phase III Study. Arthritis Rheumatol. 2015;67(6):1424-1437.
  167. Burmester GR, Lin Y, Patel R, van Adelsberg J, Mangan EK, Graham NM, van Hoogstraten H, Bauer D, Ignacio Vargas J, Lee EB. Efficacy and safety of sarilumab monotherapy versus adalimumab monotherapy for the treatment of patients with active rheumatoid arthritis (MONARCH): a randomised, double-blind, parallel-group phase III trial. Ann Rheum Dis. 2017;76(5):840-847.
  168. Fleischmann R, van Adelsberg J, Lin Y, Castelar-Pinheiro GD, Brzezicki J, Hrycaj P, Graham NM, van Hoogstraten H, Bauer D, Burmester GR. Sarilumab and Nonbiologic Disease-Modifying Antirheumatic Drugs in Patients With Active Rheumatoid Arthritis and Inadequate Response or Intolerance to Tumor Necrosis Factor Inhibitors. Arthritis Rheumatol. 2017;69(2):277-290.
  169. Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, Garcia-Porrua C, Sanchez-Andrade A, Martin J, Llorca J. Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol. 2006;24(1):83-86.
  170. Solomon DH, Massarotti E, Garg R, Liu J, Canning C, Schneeweiss S. Association between disease-modifying antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA. 2011;305(24):2525-2531.
  171. Lin C, Ji H, Cai X, Yang W, Lv F, Ji L. The association between the biological disease-modifying anti-rheumatic drugs and the incidence of diabetes: A systematic review and meta-analysis. Pharmacol Res. 2020;161:105216.
  172. Tessaro FHG, Ayala TS, Nolasco EL, Bella LM, Martins JO. Insulin Influences LPS-Induced TNF-alpha and IL-6 Release Through Distinct Pathways in Mouse Macrophages from Different Compartments. Cell Physiol Biochem. 2017;42(5):2093-2104.
  173. Kumar M, Roe K, Nerurkar PV, Orillo B, Thompson KS, Verma S, Nerurkar VR. Reduced immune cell infiltration and increased pro-inflammatory mediators in the brain of Type 2 diabetic mouse model infected with West Nile virus. J Neuroinflammation. 2014;11:80.
  174. Martinez N, Ketheesan N, Martens GW, West K, Lien E, Kornfeld H. Defects in early cell recruitment contribute to the increased susceptibility to respiratory Klebsiella pneumoniae infection in diabetic mice. Microbes Infect. 2016;18(10):649-655.
  175. Gupta S, Maratha A, Siednienko J, Natarajan A, Gajanayake T, Hoashi S, Miggin S. Analysis of inflammatory cytokine and TLR expression levels in Type 2 Diabetes with complications. Sci Rep. 2017;7(1):7633.
  176. Chao WC, Yen CL, Wu YH, Chen SY, Hsieh CY, Chang TC, Ou HY, Shieh CC. Increased resistin may suppress reactive oxygen species production and inflammasome activation in type 2 diabetic patients with pulmonary tuberculosis infection. Microbes Infect. 2015;17(3):195-204.
  177. Stegenga ME, van der Crabben SN, Blumer RM, Levi M, Meijers JC, Serlie MJ, Tanck MW, Sauerwein HP, van der Poll T. Hyperglycemia enhances coagulation and reduces neutrophil degranulation, whereas hyperinsulinemia inhibits fibrinolysis during human endotoxemia. Blood. 2008;112(1):82-89.
  178. Joshi MB, Lad A, Bharath Prasad AS, Balakrishnan A, Ramachandra L, Satyamoorthy K. High glucose modulates IL-6 mediated immune homeostasis through impeding neutrophil extracellular trap formation. FEBS Lett. 2013;587(14):2241-2246.
  179. Restrepo BI, Twahirwa M, Rahbar MH, Schlesinger LS. Phagocytosis via complement or Fc-gamma receptors is compromised in monocytes from type 2 diabetes patients with chronic hyperglycemia. PLoS One. 2014;9(3):e92977.
  180. Berrou J, Fougeray S, Venot M, Chardiny V, Gautier JF, Dulphy N, Toubert A, Peraldi MN. Natural killer cell function, an important target for infection and tumor protection, is impaired in type 2 diabetes. PLoS One. 2013;8(4):e62418.
  181. Mauriello CT, Hair PS, Rohn RD, Rister NS, Krishna NK, Cunnion KM. Hyperglycemia inhibits complement-mediated immunological control of S. aureus in a rat model of peritonitis. J Diabetes Res. 2014;2014:762051.
  182. Ponzetti M, Rucci N. Updates on Osteoimmunology: What's New on the Cross-Talk Between Bone and Immune System. Front Endocrinol (Lausanne). 2019;10:236.
  183. Huang F, Wong P, Li J, Lv Z, Xu L, Zhu G, He M, Luo Y. Osteoimmunology: The correlation between osteoclasts and the Th17/Treg balance in osteoporosis. J Cell Mol Med. 2022;26(13):3591-3597.
  184. Srivastava RK, Dar HY, Mishra PK. Immunoporosis: Immunology of Osteoporosis-Role of T Cells. Front Immunol. 2018;9:657.
  185. Ahmad SS, Ahmed F, Ali R, Ghoneim MM, Alshehri S, Najmi AK, Ahmad S, Ahmad MZ, Ahmad J, Khan MA. Immunology of osteoporosis: relevance of inflammatory targets for the development of novel interventions. Immunotherapy. 2022;14(10):815-831.
  186. Srivastava RK, Sapra L. The Rising Era of "Immunoporosis": Role of Immune System in the Pathophysiology of Osteoporosis. J Inflamm Res. 2022;15:1667-1698.
  187. Saxena Y, Routh S, Mukhopadhaya A. Immunoporosis: Role of Innate Immune Cells in Osteoporosis. Front Immunol. 2021;12:687037.
  188. Zhang W, Gao R, Rong X, Zhu S, Cui Y, Liu H, Li M. Immunoporosis: Role of immune system in the pathophysiology of different types of osteoporosis. Front Endocrinol (Lausanne). 2022;13:965258.
  189. Xiong J, Piemontese M, Onal M, Campbell J, Goellner JJ, Dusevich V, Bonewald L, Manolagas SC, O'Brien CA. Osteocytes, not Osteoblasts or Lining Cells, are the Main Source of the RANKL Required for Osteoclast Formation in Remodeling Bone. PLoS One. 2015;10(9):e0138189.
  190. Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000;21(2):115-137.
  191. Walsh MC, Choi Y. Biology of the RANKL-RANK-OPG System in Immunity, Bone, and Beyond. Front Immunol. 2014;5:511.
  192. Pietschmann P, Mechtcheriakova D, Meshcheryakova A, Foger-Samwald U, Ellinger I. Immunology of Osteoporosis: A Mini-Review. Gerontology. 2016;62(2):128-137.
  193. Lorenzo J. Cytokines and Bone: Osteoimmunology. Handb Exp Pharmacol. 2020;262:177-230.
  194. Amarasekara DS, Yun H, Kim S, Lee N, Kim H, Rho J. Regulation of Osteoclast Differentiation by Cytokine Networks. Immune Netw. 2018;18(1):e8.
  195. Rauner M, Sipos W, Pietschmann P. Osteoimmunology. Int Arch Allergy Immunol. 2007;143(1):31-48.
  196. Weitzmann MN, Cenci S, Rifas L, Brown C, Pacifici R. Interleukin-7 stimulates osteoclast formation by up-regulating the T-cell production of soluble osteoclastogenic cytokines. Blood. 2000;96(5):1873-1878.
  197. Titanji K, Vunnava A, Sheth AN, Delille C, Lennox JL, Sanford SE, Foster A, Knezevic A, Easley KA, Weitzmann MN, Ofotokun I. Dysregulated B cell expression of RANKL and OPG correlates with loss of bone mineral density in HIV infection. PLoS Pathog. 2014;10(10):e1004497.
  198. Manolagas SC, Jilka RL, Girasole G, Passeri G, Bellido T. Estrogen, cytokines, and the control of osteoclast formation and bone resorption in vitro and in vivo. Osteoporos Int. 1993;3 Suppl 1:114-116.
  199. Klein B, Wijdenes J, Zhang XG, Jourdan M, Boiron JM, Brochier J, Liautard J, Merlin M, Clement C, Morel-Fournier B, et al. Murine anti-interleukin-6 monoclonal antibody therapy for a patient with plasma cell leukemia. Blood. 1991;78(5):1198-1204.
  200. Roodman GD, Kurihara N, Ohsaki Y, Kukita A, Hosking D, Demulder A, Smith JF, Singer FR. Interleukin 6. A potential autocrine/paracrine factor in Paget's disease of bone. J Clin Invest. 1992;89(1):46-52.
  201. Kotake S, Sato K, Kim KJ, Takahashi N, Udagawa N, Nakamura I, Yamaguchi A, Kishimoto T, Suda T, Kashiwazaki S. Interleukin-6 and soluble interleukin-6 receptors in the synovial fluids from rheumatoid arthritis patients are responsible for osteoclast-like cell formation. J Bone Miner Res. 1996;11(1):88-95.
  202. Devlin RD, Bone HG, 3rd, Roodman GD. Interleukin-6: a potential mediator of the massive osteolysis in patients with Gorham-Stout disease. J Clin Endocrinol Metab. 1996;81(5):1893-1897.
  203. Martin TJ, Allan EH, Evely RS, Reid IR. Leukaemia inhibitory factor and bone cell function. Ciba Found Symp. 1992;167:141-150; discussion 150-145.
  204. Wei S, Kitaura H, Zhou P, Ross FP, Teitelbaum SL. IL-1 mediates TNF-induced osteoclastogenesis. J Clin Invest. 2005;115(2):282-290.
  205. Ruscitti P, Cipriani P, Carubbi F, Liakouli V, Zazzeroni F, Di Benedetto P, Berardicurti O, Alesse E, Giacomelli R. The role of IL-1beta in the bone loss during rheumatic diseases. Mediators Inflamm. 2015;2015:782382.
  206. D'Amelio P. The immune system and postmenopausal osteoporosis. Immunol Invest. 2013;42(7):544-554.
  207. Cenci S, Toraldo G, Weitzmann MN, Roggia C, Gao Y, Qian WP, Sierra O, Pacifici R. Estrogen deficiency induces bone loss by increasing T cell proliferation and lifespan through IFN-gamma-induced class II transactivator. Proc Natl Acad Sci U S A. 2003;100(18):10405-10410.
  208. Miyaura C, Onoe Y, Inada M, Maki K, Ikuta K, Ito M, Suda T. Increased B-lymphopoiesis by interleukin 7 induces bone loss in mice with intact ovarian function: similarity to estrogen deficiency. Proc Natl Acad Sci U S A. 1997;94(17):9360-9365.
  209. Jabbar S, Drury J, Fordham JN, Datta HK, Francis RM, Tuck SP. Osteoprotegerin, RANKL and bone turnover in postmenopausal osteoporosis. J Clin Pathol. 2011;64(4):354-357.
  210. Fischer V, Haffner-Luntzer M. Interaction between bone and immune cells: Implications for postmenopausal osteoporosis. Semin Cell Dev Biol. 2022;123:14-21.
  211. Zha L, He L, Liang Y, Qin H, Yu B, Chang L, Xue L. TNF-alpha contributes to postmenopausal osteoporosis by synergistically promoting RANKL-induced osteoclast formation. Biomed Pharmacother. 2018;102:369-374.
  212. Onal M, Xiong J, Chen X, Thostenson JD, Almeida M, Manolagas SC, O'Brien CA. Receptor activator of nuclear factor kappaB ligand (RANKL) protein expression by B lymphocytes contributes to ovariectomy-induced bone loss. J Biol Chem. 2012;287(35):29851-29860.
  213. Wu D, Cline-Smith A, Shashkova E, Perla A, Katyal A, Aurora R. T-Cell Mediated Inflammation in Postmenopausal Osteoporosis. Front Immunol. 2021;12:687551.
  214. Lai N, Zhang Z, Wang B, Miao X, Guo Y, Yao C, Wang Z, Wang L, Ma R, Li X, Jiang G. Regulatory effect of traditional Chinese medicinal formula Zuo-Gui-Wan on the Th17/Treg paradigm in mice with bone loss induced by estrogen deficiency. J Ethnopharmacol. 2015;166:228-239.
  215. Duque G, Troen BR. Understanding the mechanisms of senile osteoporosis: new facts for a major geriatric syndrome. J Am Geriatr Soc. 2008;56(5):935-941.
  216. De Maeyer RPH, Chambers ES. The impact of ageing on monocytes and macrophages. Immunol Lett. 2021;230:1-10.
  217. Li CJ, Xiao Y, Sun YC, He WZ, Liu L, Huang M, He C, Huang M, Chen KX, Hou J, Feng X, Su T, Guo Q, Huang Y, Peng H, Yang M, Liu GH, Luo XH. Senescent immune cells release grancalcin to promote skeletal aging. Cell Metab. 2022;34(1):184-185.
  218. Sandmand M, Bruunsgaard H, Kemp K, Andersen-Ranberg K, Pedersen AN, Skinhoj P, Pedersen BK. Is ageing associated with a shift in the balance between Type 1 and Type 2 cytokines in humans? Clin Exp Immunol. 2002;127(1):107-114.
  219. Siddiqi A, Monson JP, Wood DF, Besser GM, Burrin JM. Serum cytokines in thyrotoxicosis. J Clin Endocrinol Metab. 1999;84(2):435-439.
  220. Sun L, Zhu LL, Lu P, Yuen T, Li J, Ma R, Baliram R, Moonga SS, Liu P, Zallone A, New MI, Davies TF, Zaidi M. Genetic confirmation for a central role for TNFalpha in the direct action of thyroid stimulating hormone on the skeleton. Proc Natl Acad Sci U S A. 2013;110(24):9891-9896.
  221. Baliram R, Sun L, Cao J, Li J, Latif R, Huber AK, Yuen T, Blair HC, Zaidi M, Davies TF. Hyperthyroid-associated osteoporosis is exacerbated by the loss of TSH signaling. J Clin Invest. 2012;122(10):3737-3741.
  222. Grey A, Mitnick MA, Shapses S, Ellison A, Gundberg C, Insogna K. Circulating levels of interleukin-6 and tumor necrosis factor-alpha are elevated in primary hyperparathyroidism and correlate with markers of bone resorption--a clinical research center study. J Clin Endocrinol Metab. 1996;81(10):3450-3454.
  223. Grey A, Mitnick MA, Masiukiewicz U, Sun BH, Rudikoff S, Jilka RL, Manolagas SC, Insogna K. A role for interleukin-6 in parathyroid hormone-induced bone resorption in vivo. Endocrinology. 1999;140(10):4683-4690.
  224. Hory BG, Roussanne MC, Rostand S, Bourdeau A, Drueke TB, Gogusev J. Absence of response to human parathyroid hormone in athymic mice grafted with human parathyroid adenoma, hyperplasia or parathyroid cells maintained in culture. J Endocrinol Invest. 2000;23(5):273-279.
  225. Bedi B, Li JY, Tawfeek H, Baek KH, Adams J, Vangara SS, Chang MK, Kneissel M, Weitzmann MN, Pacifici R. Silencing of parathyroid hormone (PTH) receptor 1 in T cells blunts the bone anabolic activity of PTH. Proc Natl Acad Sci U S A. 2012;109(12):E725-733.
  226. Weitzmann MN. Bone and the Immune System. Toxicol Pathol. 2017;45(7):911-924.
  227. Song L, Cao L, Liu R, Ma H, Li Y, Shang Q, Zheng Z, Zhang L, Zhang W, Han Y, Zhang X, Yang H, Wang Y, Melino G, Shao C, Shi Y. The critical role of T cells in glucocorticoid-induced osteoporosis. Cell Death Dis. 2020;12(1):45.
  228. Ponnapakkam T, Katikaneni R, Nichols T, Tobin G, Sakon J, Matsushita O, Gensure RC. Prevention of chemotherapy-induced osteoporosis by cyclophosphamide with a long-acting form of parathyroid hormone. J Endocrinol Invest. 2011;34(11):e392-397.
  229. Wang W, Gao Y, Liu H, Feng W, Li X, Guo J, Li M. Eldecalcitol, an active vitamin D analog, effectively prevents cyclophosphamide-induced osteoporosis in rats. Exp Ther Med. 2019;18(3):1571-1580.
  230. Zhao D, Wang C, Zhao Y, Shu B, Jia Y, Liu S, Wang H, Chang J, Dai W, Lu S, Shi Q, Yang Y, Zhang Y, Wang Y. Cyclophosphamide causes osteoporosis in C57BL/6 male mice: suppressive effects of cyclophosphamide on osteoblastogenesis and osteoclastogenesis. Oncotarget. 2017;8(58):98163-98183.
  231. Chen X, Wang S, Chen G, Wang Z, Kan J. The immunomodulatory effects of Carapax Trionycis ultrafine powder on cyclophosphamide-induced immunosuppression in Balb/c mice. J Sci Food Agric. 2021;101(5):2014-2026.
  232. Filippa MG, Tektonidou MG, Mantzou A, Kaltsas GA, Chrousos GP, Sfikakis PP, Yavropoulou MP. Adrenocortical dysfunction in rheumatoid arthritis: Alpha narrative review and future directions. Eur J Clin Invest. 2022;52(1):e13635.
  233. Nicolaides NC, Kyratzi E, Lamprokostopoulou A, Chrousos GP, Charmandari E. Stress, the stress system and the role of glucocorticoids. Neuroimmunomodulation. 2015;22(1-2):6-19.
  234. Laflamme N, Lacroix S, Rivest S. An essential role of interleukin-1beta in mediating NF-kappaB activity and COX-2 transcription in cells of the blood-brain barrier in response to a systemic and localized inflammation but not during endotoxemia. J Neurosci. 1999;19(24):10923-10930.
  235. Nadeau S, Rivest S. Regulation of the gene encoding tumor necrosis factor alpha (TNF-alpha) in the rat brain and pituitary in response in different models of systemic immune challenge. J Neuropathol Exp Neurol. 1999;58(1):61-77.
  236. Perlstein RS, Whitnall MH, Abrams JS, Mougey EH, Neta R. Synergistic roles of interleukin-6, interleukin-1, and tumor necrosis factor in the adrenocorticotropin response to bacterial lipopolysaccharide in vivo. Endocrinology. 1993;132(3):946-952.
  237. Chrousos GP, Kaltsas G. Post-SARS sickness syndrome manifestations and endocrinopathy: how, why, and so what? Clin Endocrinol (Oxf). 2005;63(4):363-365.
  238. Chrousos GP, Gold PW. The concepts of stress and stress system disorders. Overview of physical and behavioral homeostasis. JAMA. 1992;267(9):1244-1252.
  239. Papanicolaou DA, Tsigos C, Oldfield EH, Chrousos GP. Acute glucocorticoid deficiency is associated with plasma elevations of interleukin-6: does the latter participate in the symptomatology of the steroid withdrawal syndrome and adrenal insufficiency? J Clin Endocrinol Metab. 1996;81(6):2303-2306.
  240. Bethin KE, Vogt SK, Muglia LJ. Interleukin-6 is an essential, corticotropin-releasing hormone-independent stimulator of the adrenal axis during immune system activation. Proc Natl Acad Sci U S A. 2000;97(16):9317-9322.
  241. Silverman MN, Miller AH, Biron CA, Pearce BD. Characterization of an interleukin-6- and adrenocorticotropin-dependent, immune-to-adrenal pathway during viral infection. Endocrinology. 2004;145(8):3580-3589.
  242. Franchimont D, Bouma G, Galon J, Wolkersdorfer GW, Haidan A, Chrousos GP, Bornstein SR. Adrenal cortical activation in murine colitis. Gastroenterology. 2000;119(6):1560-1568.
  243. Path G, Bornstein SR, Ehrhart-Bornstein M, Scherbaum WA. Interleukin-6 and the interleukin-6 receptor in the human adrenal gland: expression and effects on steroidogenesis. J Clin Endocrinol Metab. 1997;82(7):2343-2349.
  244. Path G, Scherbaum WA, Bornstein SR. The role of interleukin-6 in the human adrenal gland. Eur J Clin Invest. 2000;30 Suppl 3:91-95.
  245. Filippa MG, Tektonidou MG, Mantzou A, Kaltsas GA, Chrousos GP, Sfikakis PP, Yavropoulou MP. Adrenocortical dysfunction in rheumatoid arthritis: Alpha narrative review and future directions. Eur J Clin Invest. 2021:e13635.
  246. Yavropoulou MP, Filippa MG, Mantzou A, Ntziora F, Mylona M, Tektonidou MG, Vlachogiannis NI, Paraskevis D, Kaltsas GA, Chrousos GP, Sfikakis PP. Alterations in cortisol and interleukin-6 secretion in patients with COVID-19 suggestive of neuroendocrine-immune adaptations. Endocrine. 2022;75(2):317-327.
  247. de Kloet ER. Stress in the brain. Eur J Pharmacol. 2000;405(1-3):187-198.
  248. Lightman SL, Wiles CC, Atkinson HC, Henley DE, Russell GM, Leendertz JA, McKenna MA, Spiga F, Wood SA, Conway-Campbell BL. The significance of glucocorticoid pulsatility. Eur J Pharmacol. 2008;583(2-3):255-262.
  249. Stavreva DA, Wiench M, John S, Conway-Campbell BL, McKenna MA, Pooley JR, Johnson TA, Voss TC, Lightman SL, Hager GL. Ultradian hormone stimulation induces glucocorticoid receptor-mediated pulses of gene transcription. Nat Cell Biol. 2009;11(9):1093-1102.
  250. Hochberg Z, Pacak K, Chrousos GP. Endocrine withdrawal syndromes. Endocr Rev. 2003;24(4):523-538.
  251. Yavropoulou MP, Filippa MG, Panopoulos S, Spanos E, Spanos G, Tektonidou MG, Sfikakis PP. Impaired adrenal cortex reserve in patients with rheumatic and musculoskeletal diseases who relapse upon tapering of low glucocorticoid dose. Clin Exp Rheumatol. 2022.
  252. Yavropoulou MP, Tsokos GC, Chrousos GP, Sfikakis PP. Protracted stress-induced hypocortisolemia may account for the clinical and immune manifestations of Long COVID. Clin Immunol. 2022;245:109133.
  253. Kirschbaum C, Hellhammer DH. Salivary cortisol in psychobiological research: an overview. Neuropsychobiology. 1989;22(3):150-169.
  254. Sephton SE, Sapolsky RM, Kraemer HC, Spiegel D. Diurnal cortisol rhythm as a predictor of breast cancer survival. J Natl Cancer Inst. 2000;92(12):994-1000.
  255. Pervanidou P, Kolaitis G, Charitaki S, Margeli A, Ferentinos S, Bakoula C, Lazaropoulou C, Papassotiriou I, Tsiantis J, Chrousos GP. Elevated morning serum interleukin (IL)-6 or evening salivary cortisol concentrations predict posttraumatic stress disorder in children and adolescents six months after a motor vehicle accident. Psychoneuroendocrinology. 2007;32(8-10):991-999.
  256. Bougea AM, Spandideas N, Alexopoulos EC, Thomaides T, Chrousos GP, Darviri C. Effect of the emotional freedom technique on perceived stress, quality of life, and cortisol salivary levels in tension-type headache sufferers: a randomized controlled trial. Explore (NY). 2013;9(2):91-99.
  257. Lopez-Acevo CA, Arrendondo-Loza E, Salinas-Carmona MC, Rendon A, Martinez-Castilla AM, Vazquez-Marmolejo AV, Munoz-Maldonado G, Rosas-Taraco AG. Cortisol and perceived stress are associated with cytokines levels in patients infected with influenza B virus. Cytokine. 2021;138:155400.
  258. Cutolo M, Villaggio B, Otsa K, Aakre O, Sulli A, Seriolo B. Altered circadian rhythms in rheumatoid arthritis patients play a role in the disease's symptoms. Autoimmun Rev. 2005;4(8):497-502.
  259. Straub RH, Cutolo M. Circadian rhythms in rheumatoid arthritis: implications for pathophysiology and therapeutic management. Arthritis Rheum. 2007;56(2):399-408.
  260. Cutolo M, Masi AT. Circadian rhythms and arthritis. Rheum Dis Clin North Am. 2005;31(1):115-129, ix-x.
  261. Cutolo M. Circadian rhythms and rheumatoid arthritis. Joint Bone Spine. 2019;86(3):327-333.
  262. Sothern RB, Roitman-Johnson B, Kanabrocki EL, Yager JG, Fuerstenberg RK, Weatherbee JA, Young MR, Nemchausky BM, Scheving LE. Circadian characteristics of interleukin-6 in blood and urine of clinically healthy men. In Vivo. 1995;9(4):331-339.
  263. Chikanza IC, Petrou P, Kingsley G, Chrousos G, Panayi GS. Defective hypothalamic response to immune and inflammatory stimuli in patients with rheumatoid arthritis. Arthritis Rheum. 1992;35(11):1281-1288.
  264. Gudbjornsson B, Skogseid B, Oberg K, Wide L, Hallgren R. Intact adrenocorticotropic hormone secretion but impaired cortisol response in patients with active rheumatoid arthritis. Effect of glucocorticoids. J Rheumatol. 1996;23(4):596-602.
  265. Crofford LJ, Kalogeras KT, Mastorakos G, Magiakou MA, Wells J, Kanik KS, Gold PW, Chrousos GP, Wilder RL. Circadian relationships between interleukin (IL)-6 and hypothalamic-pituitary-adrenal axis hormones: failure of IL-6 to cause sustained hypercortisolism in patients with early untreated rheumatoid arthritis. J Clin Endocrinol Metab. 1997;82(4):1279-1283.
  266. Gutierrez MA, Garcia ME, Rodriguez JA, Mardonez G, Jacobelli S, Rivero S. Hypothalamic-pituitary-adrenal axis function in patients with active rheumatoid arthritis: a controlled study using insulin hypoglycemia stress test and prolactin stimulation. J Rheumatol. 1999;26(2):277-281.
  267. Cutolo M, Foppiani L, Prete C, Ballarino P, Sulli A, Villaggio B, Seriolo B, Giusti M, Accardo S. Hypothalamic-pituitary-adrenocortical axis function in premenopausal women with rheumatoid arthritis not treated with glucocorticoids. J Rheumatol. 1999;26(2):282-288.
  268. Cutolo M, Foppiani L, Minuto F. Hypothalamic-pituitary-adrenal axis impairment in the pathogenesis of rheumatoid arthritis and polymyalgia rheumatica. J Endocrinol Invest. 2002;25(10 Suppl):19-23.
  269. Straub RH, Paimela L, Peltomaa R, Scholmerich J, Leirisalo-Repo M. Inadequately low serum levels of steroid hormones in relation to interleukin-6 and tumor necrosis factor in untreated patients with early rheumatoid arthritis and reactive arthritis. Arthritis Rheum. 2002;46(3):654-662.
  270. Imrich R, Vigas M, Rovensky J, Aldag JC, Masi AT. Adrenal plasma steroid relations in glucocorticoid-naive premenopausal rheumatoid arthritis patients during insulin-induced hypoglycemia test compared to matched normal control females. Endocr Regul. 2009;43(2):65-73.
  271. De Silva M, Binder A, Hazleman BL. The timing of prednisolone dosage and its effect on morning stiffness in rheumatoid arthritis. Ann Rheum Dis. 1984;43(6):790-793.
  272. Buttgereit F, Doering G, Schaeffler A, Witte S, Sierakowski S, Gromnica-Ihle E, Jeka S, Krueger K, Szechinski J, Alten R. Efficacy of modified-release versus standard prednisone to reduce duration of morning stiffness of the joints in rheumatoid arthritis (CAPRA-1): a double-blind, randomised controlled trial. Lancet. 2008;371(9608):205-214.
  273. Buttgereit F, Doering G, Schaeffler A, Witte S, Sierakowski S, Gromnica-Ihle E, Jeka S, Krueger K, Szechinski J, Alten R. Targeting pathophysiological rhythms: prednisone chronotherapy shows sustained efficacy in rheumatoid arthritis. Ann Rheum Dis. 2010;69(7):1275-1280.
  274. Buttgereit F, Mehta D, Kirwan J, Szechinski J, Boers M, Alten RE, Supronik J, Szombati I, Romer U, Witte S, Saag KG. Low-dose prednisone chronotherapy for rheumatoid arthritis: a randomised clinical trial (CAPRA-2). Ann Rheum Dis. 2013;72(2):204-210.
  275. Nicolaides NC, Chrousos GP. Impact of Stress on Health in Childhood and Adolescence. Horm Res Paediatr. 2023;96(1):5-7.
  276. Tzelepi I, Bacopoulou F, Chrousos GP, Sotiropoulou L, Vlachakis D, Darviri C. Mindfulness and Academic Performance of College and University Students: A Systematic Review. Adv Exp Med Biol. 2023;1425:207-215.
  277. Redwine L, Mills PJ, Sada M, Dimsdale J, Patterson T, Grant I. Differential immune cell chemotaxis responses to acute psychological stress in Alzheimer caregivers compared to non-caregiver controls. Psychosom Med. 2004;66(5):770-775.
  278. Rosenkranz MA, Lutz A, Perlman DM, Bachhuber DR, Schuyler BS, MacCoon DG, Davidson RJ. Reduced stress and inflammatory responsiveness in experienced meditators compared to a matched healthy control group. Psychoneuroendocrinology. 2016;68:117-125.
  279. Fancourt D, Perkins R, Ascenso S, Carvalho LA, Steptoe A, Williamon A. Effects of Group Drumming Interventions on Anxiety, Depression, Social Resilience and Inflammatory Immune Response among Mental Health Service Users. PLoS One. 2016;11(3):e0151136.
  280. Sapolsky RM, Romero LM, Munck AU. How do glucocorticoids influence stress responses? Integrating permissive, suppressive, stimulatory, and preparative actions. Endocr Rev. 2000;21(1):55-89.
  281. Kiss A, Aguilera G. Participation of alpha 1-adrenergic receptors in the secretion of hypothalamic corticotropin-releasing hormone during stress. Neuroendocrinology. 1992;56(2):153-160.
  282. Bunn SJ, Ait-Ali D, Eiden LE. Immune-neuroendocrine integration at the adrenal gland: cytokine control of the adrenomedullary transcriptome. J Mol Neurosci. 2012;48(2):413-419.
  283. Ait-Ali D, Turquier V, Tanguy Y, Thouennon E, Ghzili H, Mounien L, Derambure C, Jegou S, Salier JP, Vaudry H, Eiden LE, Anouar Y. Tumor necrosis factor (TNF)-alpha persistently activates nuclear factor-kappaB signaling through the type 2 TNF receptor in chromaffin cells: implications for long-term regulation of neuropeptide gene expression in inflammation. Endocrinology. 2008;149(6):2840-2852.
  284. Douglas SA, Bunn SJ. Interferon-alpha signalling in bovine adrenal chromaffin cells: involvement of signal-transducer and activator of transcription 1 and 2, extracellular signal-regulated protein kinases 1/2 and serine 31 phosphorylation of tyrosine hydroxylase. J Neuroendocrinol. 2009;21(3):200-207.
  285. Rosmaninho-Salgado J, Araujo IM, Alvaro AR, Mendes AF, Ferreira L, Grouzmann E, Mota A, Duarte EP, Cavadas C. Regulation of catecholamine release and tyrosine hydroxylase in human adrenal chromaffin cells by interleukin-1beta: role of neuropeptide Y and nitric oxide. J Neurochem. 2009;109(3):911-922.
  286. Jenkins DE, Sreenivasan D, Carman F, Samal B, Eiden LE, Bunn SJ. Interleukin-6-mediated signaling in adrenal medullary chromaffin cells. J Neurochem. 2016;139(6):1138-1150.
  287. Fan J, Zhang B, Shu HF, Zhang XY, Wang X, Kuang F, Liu L, Peng ZW, Wu R, Zhou Z, Wang BR. Interleukin-6 increases intracellular Ca2+ concentration and induces catecholamine secretion in rat carotid body glomus cells. J Neurosci Res. 2009;87(12):2757-2762.
  288. Sharma D, Farrar JD. Adrenergic regulation of immune cell function and inflammation. Semin Immunopathol. 2020;42(6):709-717.
  289. Takenaka MC, Guereschi MG, Basso AS. Neuroimmune interactions: dendritic cell modulation by the sympathetic nervous system. Semin Immunopathol. 2017;39(2):165-176.
  290. Guyot M, Simon T, Panzolini C, Ceppo F, Daoudlarian D, Murris E, Macia E, Abelanet S, Sridhar A, Vervoordeldonk MJ, Glaichenhaus N, Blancou P. Apical splenic nerve electrical stimulation discloses an anti-inflammatory pathway relying on adrenergic and nicotinic receptors in myeloid cells. Brain Behav Immun. 2019;80:238-246.

Type 2 Diabetes In Children and Adolescents – A Focus On Diagnosis and Treatment

ABSTRACT

Nearly three decades have passed since the first publications on type 2 diabetes (T2D) in children and adolescents, and it is now well established as a global problem. As the prevalence of obesity continues to increase within the general population, diagnosing T2D in adolescents presents a multifaceted challenge. In this chapter, we shall delve into the epidemiological aspects, as well as the distinct characteristics inherent to various types of diabetes. Cohort studies with long-term follow-ups have illuminated our understanding of the alarming incidence of complications in adolescents diagnosed with T2D. Recent approvals of novel pharmaceutical interventions for teenagers have ushered in a new era of hope. Hopefully, in the forthcoming decade, we anticipate a decline in the prevalence of these complications. However, in the interim, a proactive and assertive approach remains essential for addressing the complications associated with T2D in adolescents.

 

SHORT HISTORY

Youth-onset type 2 diabetes (T2D) was initially described in Pima Indian children and adolescents (1). This tribe, also known as “the pathfinders,” has a notable history of obesity and early-onset diabetes. In the mid-nineties, studies from various clinics in the United States reported cases of T2D in children from diverse ethnic backgrounds, primarily non-Hispanic Blacks and Hispanics (2-4). While these reports were initially met with suspicion, it soon became clear that a new disease had emerged among children. Subsequently, similar reports emerged from various parts of the world. In 2000, the SEARCH for Diabetes Study was launched in the US. This prospective nationwide, multi-center study aimed at understanding the epidemiology of both type 1 diabetes (T1D) and T2D in children. A few years later, in 2004, the Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) study was initiated. The TODAY study was a prospective, randomized treatment trial, designed to explore treatment regimens and the clinical course of T2D in youth. Registries from Germany, Diabetes Prospective Follow-up Registry (DPV) (5), Hong Kong (6), China (7), India (Registry of People with Diabetes with Youth Age at Onset (YDR)) (8) and Israel (9), contributed to the current knowledge. Importantly, while the US is leading in the scope of the T2D epidemic, similar trends have been observed worldwide.

 

EPIDEMIOLOGY

 

Epidemiology in the USA

 

According to the most recent data from the SEARCH study published in 2023, the adjusted incidence of T2DM among children and adolescents nearly doubled, from 9.0 to 17.9 cases per 100,000 persons per year, from 2002-03 to 2017-18 (10).

 

EPIDEMIOLOGY ACCORDING TO ETHNICITY

 

The incidence of T2D varies largely based on ethnicity, the highest incidence of T2D, 50.1 per 100,000 children age 10-20 years, was recorded in non-Hispanic Blacks, followed by Pima Indians (46), Hispanics (25.8), and Asian/Pacific Islanders (16.6). Among non-Hispanic Whites, the incidence is only 5.5 per 100,000. An annual increase was observed in all groups, but the highest increase was observed in Asian/Pacific Islanders (8.92%), followed by 7.17% in Hispanics and 5.99% in non-Hispanic Blacks, compared to 1.83% in non-Hispanic Whites.

 

EPIDEMIOLOGY ACCORDING TO GENDER AND AGE

The current incidence in females is 21.6 per 100,000 and in males 14.2; the temporal increase was similar in both genders. The incidence increases with age. One of the most remarkable findings in the SEARCH study is that among individuals 15-19 years of age, the incidence of T2D in 2017-2018 exceeded that of T1D (19.7 vs. 14.6 per 100,000). This is the first time the incidence of T2D surpassed that of T1D among youth, sounding alarming for other countries, which may have a lower incidence than the USA, but are showing steady increases.

Epidemiology in Europe

Although the overall incidence of T2D in Europe is much lower than in the US, a similar change over time has been reported. In Germany, a three-fold increase in the prevalence of T2D was reported for 10- to 19-year-olds between 2002 and 2020 (3.4 to 10.8 per 100,000) (5). Similar to the US, the estimated standardized prevalence of T2D was 1.4 times higher among girls (12.8) than boys (9.0). In the UK, the number of children registered as having T2D and being treated in pediatric diabetes units has risen by more than 50% in the past five years (11).

Epidemiology in Asia

Data retrieved from the Hong Kong Childhood Diabetes Registry revealed a threefold increase in T2D in children, from 1.27 per 100,000 in 1997-2007 to 3.42 per 100,000 in 2008-2018 (6). Data from China demonstrated an average annual increase of 26.6% in youth aged 10-19 years (12). There was no statistically significant difference in incidence between boys and girls. However, the risk of T2D was 1.49 times higher in urban areas than in rural areas.

 

RISK FACTORS FOR T2D IN CHILDREN

 

The increased risk of developing T2D in children is associated with genetics, an obesogenic environment, and obesity, along with the interactions among these factors (Figure 1). This visual representation emphasizes the multifaceted nature of risk factors for T2D in children and adolescents, illustrating the intricate connections between genetics, environmental factors, obesity, and the eventual development of T2D.

 

Figure 1. Risk Factors for T2D in Children and Adolescents. Risk factors for T2D in children and adolescents can be visualized as a series of interconnected circles, illustrating the complex interplay of various influences:
Environment (Outer Circle): The outermost circle represents the broader environmental factors that encompass everything from the intrauterine environment to residential neighborhood.
Genetics (Second Circle): Within the environment circle, genetics forms an important inner circle. It signifies the genetic predisposition that some individuals may have towards developing obesity and, consequently, T2D.
Obesity (Third Circle): Nested within the genetics circle is the obesity circle. It is influenced by multiple factors such as immigration, sedentary lifestyle, diet, microbiome, medication use, depression, and abuse. Of note, not everyone exposed to the obesogenic environment will become obese, indicating the influence of other factors.
Type 2 Diabetes (Innermost Circle): The innermost circle represents the development of T2D. Importantly, not all obese individuals will go on to develop early-onset T2D, highlighting the complexity of these interrelated risk factors.

Obesity

 

Severe obesity stands out as one of the most significant risk factors for youth-onset of T2D. Obesity is closely linked to the worsening of insulin resistance, a primary feature of the pathophysiology of T2D, along with progressive β-cell and α-cell dysfunction (13). In a recent meta-analysis comprising data from 30 studies involving 4688 children and adolescents, the prevalence of obesity at the time of T2D diagnosis was found to be 77 % (95% CI, 71%-83%) (14). Male participants exhibited higher odds of obesity than their female counterparts (odds ratio, 2.10; 95% CI, 1.3-3.3). These findings were sustained after several sensitivity analyses that excluded studies with uncertain or unspecified T2D diagnostic criteria, studies involving individuals with positive pancreatic autoantibodies, individuals presenting with weight loss, or those with genetically proven monogenic diabetes. While acknowledging the limitations of using BMI-based measures as a surrogate for obesity and considering the retrospective nature of the included studies, these data suggest that approximately 20-25% of adolescents with diabetes but without obesity may still have T2D. Furthermore, the definition of obesity needs to consider body composition variations among certain populations, such as East Asian and South Asian populations.

 

Family History of T2D

 

Most adolescents with T2D have a family member with the disease. In the TODAY study, 59.6% of adolescents with T2D had a first-degree family member with a history of diabetes, and 89% had a grandparent affected with diabetes (15). This increased risk of T2D associated with family history reflects genetic influences, the impact of the environment, and the intrauterine environment.

Maternal Obesity and Maternal T2D During Pregnancy

 

Exposure to maternal obesity has been linked to an increased risk of childhood obesity in offspring (16) and 2.8-fold higher odds of T2D. Exposure to maternal gestational diabetes was associated with 5.7-fold higher odds of T2D (17). Moreover, individuals exposed to maternal diabetes during pregnancy were diagnosed with diabetes at a younger age and exhibited worse β-cell function. Exposure in utero to maternal diabetes and obesity accounted for 47% of the T2D risk in this population (17,18), and, importantly, it sets up a vicious cycle for future generations. Suggested mechanisms affecting the developmental programming of offspring towards T2D include epigenetic modifications, alterations in stem cell differentiation, variation in the metabolome and microbiome, and immune dysregulation (19). Of note, assistedreproductive technology was not found to be a risk factor for early-onset T2D (20).

Pregnancies complicated by pre-existing diabetes are associated with extreme birth weights. In a large study, small for gestational age (SGA) babies, defined as below the 10th percentile for birth weight, were reported in 14.1% of pregnancies of women with T2D, while 26.2% had newborns with a large birth weight, defined as above the 90th percentile (21). Both SGA and high birth weight are associated with an increased risk of a history of early-onset T2D among adults. Of note, a lower birthweight was associated with an increased risk of developing T2D independently of adult BMI and the genetic risk of T2D (22).

 

Pregnancies complicated by pre-existing diabetes are associated with preterm delivery and birth weight extremes. In a national cohort study involving over 4 million singletons born in Sweden from 1973 to 2014, preterm birth (occurring before 37 weeks) was associated with a 1.3-fold increased risk of developing T2D before age 18 years (23). Notably, the association between preterm birth and T2D was significantly stronger among females (23).

 

Mental Health and Treatment

 

The association between psychiatric disorders in adolescents and T2D is bidirectional, complex, and not well studied in the pediatric population (24). On the one hand, youth with chronic illnesses such as obesity are more likely to develop depression, depressive symptoms, and anxiety than those without chronic illnesses. On the other hand, youth who have mental health morbidities are at increased risk for isolation, sedentary lifestyle, weight gain, and the development of T2D. Furthermore, some psychotropic medications, particularly atypical antipsychotics, are associated with weight gain and increased risk for T2D in adults. These associations have not been systematically studied in youth, but clinical experience suggests a contribution.

 

Immigration

 

Globally, immigration is on the rise. Studies conducted among adults have indicated that immigrants, when compared to non-immigrants, exhibit higher rates of obesity, insulin-resistance, and hypertension. In a recent review (25) focusing on European countries, non-European migrant children face a greater risk of being overweight or obese compared to their native counterparts; the prevalence of obesity in migrant and native children ranged from 1.2 to 15.4% and from 0.6 to 11.6%, respectively. The increased rates of overweight and obesity among migrating children and adolescents render them more susceptible to developing T2D and other metabolic abnormalities. An Israeli study found that among adolescents of Ethiopian origin, the prevalence of overweight and obesity increased two-and-a-half fold and fourfold in males and females, respectively, during the study period, compared to a 1.5-fold increase among native Israeli-bornmales and females (26).

 

Socioeconomic Status

 

A discernible disparity exists in the socioeconomic status (SES) of children and adolescents with T2D. In developed countries, such as the United States, low SES is a recognized risk factor for the development of obesity and T2D. This is attributed to factors such as limited access to healthy foods and reduced physical inactivity. However, in developing countries, higher SES groups tend to have greater levels of physical inactivity and consume higher quantities of fat, salt, and processed foods compared to their lower SES counterparts (Figure 2). When data from 384 youth with T2D who completed a baseline research visit as part of the SEARCH study and 227 youth with T2D from the Registry of People with Diabetes with Youth Age at Onset (YDR) in India who completed a baseline visit were compared, only 23.6% of SEARCH youth belonged to a high SES group, in contrast to 88.5% of YDR youths (P < .001) (8).

Figure 2. Global Disparities in Type 2 Diabetes Risk Factors. This illustrative figure offers insights into the diversity in risk factors for T2D among children across the world. It highlights how these risk factors vary across different regions and populations.
Gender Disparities (Upper Part of Figure): In many Western countries, the risk of developing T2D in children is higher in girls compared to boys. However, this pattern is not consistent globally. In other parts of the world, an equal or even higher risk of T2D has been documented in males. The upper portion of the figure visually represents these variations in risk, depicting the female-to-male ratios in different regions.
Socioeconomic Status (SES) Variation: (Lower Part of Figure) SES plays a significant role in T2D risk. In Western countries, lower SES is associated with an increased risk of T2D in children. Conversely, in developing countries, a contrary trend is observed, with a higher risk of T2D observed in populations with higher SES. These complex socioeconomic variations are highlighted in the figure, showcasing how T2D risk is influenced by economic factors on a global scale.

 

Data from 747 children and youths with T2D under 19 years of age, collected between 2009 and 2016 (from the population-based National Pediatric Diabetes Audit, covering over >95% of diabetes cases in England and Wales), revealed that under half of those with T2D reside in the most disadvantaged areas of England and Wales, and 41% fall into the most disadvantaged SES quintile (27). In contrast, in China, in Zhejiang, one of the most economically prosperous coastal provinces marked by industrialization and urbanization, changes in diet and decreased physical activity have led to a higher incidence of pediatric obesity. The mean annual incidence was 2.32 per 100,000 person-years in urban area compared with 1.44 in urban and rural area (12).


COVID-19 Pandemic


In a study conducted in the United States, the incidence of T2DM at 24 diabetes centers during the first year of the COVID-19 pandemic was examined (28). The average number of new diagnoses per year in the two years before the pandemic was 825 but rose to 1463 during the first pandemic year. This increase of 77% is significantly higher than the 5% expected annual increase in incidence observed in the two previous years. Similarly, a retrospective cross-sectional review of youths (age ≤ 21) diagnosed with T2D during the COVID-19 pandemic (from May 1, 2020, to April 30, 2021) and the five years preceding it (from May 1, 2015, to April 30, 2020) at a tertiary diabetes center revealed an increase of 293% (29).  Similar trends were reported from Germany (30). Data on T2D in adolescents during 2 years of the COVID-19 pandemic (2020–2021) were compared with the control period 2011–2019 in children aged 6 to <18 years were obtained from the DPV (German Diabetes Prospective Follow-up) Registry. The incidence of youth-onset T2D increased from 0.75 per 100,000 patient-years in 2011 to 1.25 per 100,000 in 2019, an annual increase of 6.8%. However, in 2021, the observed incidence was 1.95, significantly higher than expected. Of note, the observed incidence was significantly higher in boys (2.16), leading to a reversal of the sex ratio of pediatric T2D incidence (30).  The surge in T2D incidence during the pandemic can be attributed to several factors. First, there was an increase in obesity among young people during the COVID-19 pandemic, accompanied by increased consumption of processed foods and reduced physical activity, both of which contribute to the risk of developing T2D (31). In addition, there was increased psychosocial stress, which is emerging as an important contributor to the risk of T2D. Furthermore, viral mediated non-autoimmune β-cell destruction, resulting in reduced β-cell function in predisposed adolescents, has been suggested as a contributing factor (28).    

 

Genetics


The genetics of T2D in children and adolescents have largely remained unexplored. ProDiGY is a multiethnic collaboration encompassing three studies (TODAY, SEARCH, and T2D-GENES) involving 3,006 youth subjects with T2D, diagnosed at a mean age of 15.1±2.9 years, along with 6,061 diabetes-free adult control subjects. Association analyses were conducted on approximately 10 million imputed variants, employing a generalized linear mixed model incorporating a genetic relationship matrix to account for population structure. The analysis was further adjusted for sex. This comprehensive study identified seven genome-wide significant loci that included a novel locus in PHF2, along with TCF7L2, MC4R, CDC123, KCNQ1, IGF2BP2, and SLC16A11. A secondary analysis involving 856 diabetes-free youth subjects uncovered an additional locus in CPEB2, and consistent directional effects for diabetes risk were observed (32). 

CLINICAL PRESENTATION

Diabetes Related Signs

The classical signs at the onset of diabetes - polydipsia and polyuria - are observed in about two-thirds of youth at the diagnosis of T2D. Only about one-third are diagnosed through routine screening of asymptomatic youth with obesity. Furthermore, the prevalence of diabetic ketoacidosis (DKA) among youth with T2D in the SEARCH study and the YDR in India was 5.5% and 6.6%, respectively. Importantly, the incidence of DKA at the onset of T2D was higher during the SARS-CoV-2 pandemic (33). Hyperglycemic hyperosmolar state is present at diagnosis in 2% of youth with T2D (34).  

 

Seasonal Diagnosis of T2D

 

There is significant seasonal variation in the onset of T2D in children and young people in the United States; diagnoses increased in August. Possible explanations for an August peak in diagnoses include weight gain during the summer vacation and an increase in physical exams for school athletic programs that may detect asymptomatic hyperglycemia. The greater proportion of diagnoses made during routine health visits rather than following symptoms is consistent with this latter possibility. Similar seasonal variations in incidence are beginning to emerge in other countries.

 

Age of Onset of T2D

 

The incidence of T2D rises gradually during puberty, primarily due to the physiological insulin resistance characteristic of this period. However, illustrating the heterogeneity of T2D in children and adolescents, there was a significant difference between the mean age of onset of youth in the United States (Pediatric Diabetes Consortium, PDC) databases who were diagnosed at the mean age of 12 years compared to the mean age in Germany of 13 years (35).

 

PREPUBERTAL T2D

With the worldwide epidemic of childhood obesity on the rise, there are increasing reports of T2D occurring in prepubertal children. Initially, there were case reports on prepubertal children with T2D, including cases from Nigeria (36), and a 5-year-old Indigenous girl from Australia (37). Lately, several reports have described the onset of T2D in prepubertal children. A total of 42 children ≤ 10 years with T2DM were reported from Alabama (38), 12 from Australia (39), 35 prepubertal children from Houston, Texas (40), from San Antonio, Texas (41) and India (42). The common denominator of all reports of prepubertal children is extreme obesity, with a disproportionate impact on females and ethnic minorities as shown in Table 1. Additionally, there is a high prevalence of comorbidities.

Table 1. Characteristics of Prepubertal Children Diagnosed with T2D

 

San Antonio, Texas

N=20

Alabama, US

N=42

Australia

N=12

Houston, Texas.

N=35

Chennai, Tamil Nadu India

N=4

Age, years

Range

8.1

4-9

 

 

7-10

10.6± 2.5

 

5-8

10

8

 

 

 

9-10

10

34

 

 

 

Ethnicity

Hispanic 80%

African American 88(%)

 Caucasian 12%

Aboriginal Australians 92

Maori 8

 Hispanic 71

Black 23

Non Hispanic White 3%

India

Females (%)

75

88

75

51

100

BMI Z score

2.72

(1.7-5.0)

2.5 ± 0.4

2.38± 0.64

2.4±0.4

>97%ile

Hypertension (%)

NA

21

25

14

 

Dyslipidemia

 

NA

58

58%

100

 

NAFLD

NA

 

 

20

 

Microalbuminuria

NA

 

16.6

5

 

 

These reports suggest that there are unique features and pathophysiologic drivers distinct from the influence of pubertal hormones, which are often implicated in the mechanism underlying T2D in older children and adolescents. Furthermore, the emergence of T2D in prepubertal children and the high prevalence of comorbidities among them serve as a warning for an impending public health challenge.

Sex

 

In the Western world, youth-onset T2D is almost twice as common in girls as in boys, whereas Asian countries report no differences in incidence by sex. For T2D, SEARCH had a higher proportion of females, while studies from China show no difference between females and males, and there are reports of increased prevalence in males in the Middle East; in the UAE (43), the female to male ratio was 0.5:1, in Iraq (44) 0.6:1, and Kuwait (45) 0.8:1 (Figure 2). This may reflect the higher prevalence of obesity in males in these countries (46). Indeed, in the UAE, among children aged 11 to 14 years, the prevalence of obesity was reported as 24.3% and extreme obesity (BMI ≥99th percentile) as 5.7%; the rate of extreme obesity was 9.6-fold higher in boys than girls (47). In Kuwait, 41.4% of boys were classified as obese, compared to 28.9% of girls of the same age (46).

 

DIAGNOSIS

 

Early diagnosis of T2D is likely to bring several benefits, enabling prompt multifactorial treatment and management of cardiovascular risk factors. Considering the increasing prevalence of T2D among children and adolescents, clinical symptoms have become less distinct, and age, sex, and weight are no longer definitive criteria for a clear diagnosis. Therefore, the most important diagnostic feature is the presence or absence of pancreatic autoantibodies in any child diagnosed with hyperglycemia; positive antibodies indicate T1D even if the child is overweight or has a family history of T2D. If the antibodies are negative, a genetic test for monogenic diabetes (previously referred to as Maturity-Onset Diabetes of the Young (MODY) should be considered if available.

TREATMENT

 

Initial treatment for T2D includes a focus on lifestyle changes, including healthy diet, increasing regular physical activity, achieving weight loss, and receiving emotional support. While lifestyle change is recommended as a practical approach,there is limited evidence that lifestyle intervention alone has a sustained impact on glycemic control in youth with T2D (48).

 

The Impact of Lifestyle Changes

 

The impact of lifestyle intervention, as determined by changes in diet and cardiovascular fitness, on glycemic control in youth with T2D was assessed in the TODAY clinical trial cohort spanning 15 centers across the United States. The study included 699 youth aged 10 to 17 years with T2D for less than 2 years. Those participants randomly assigned to the lifestyle intervention participated in a family-based behavioral program aimed at promoting weight loss. Each family was assigned a dedicated leader responsible for guiding them through their journey of physical activity and nutrition improvement. The intervention was structured across three stages, initially there were weekly meetings for 6 to 8 months, focusing on physical activity, setting individual calorie intake goals, self-monitoring, and problem-solving. In the subsequent stage, biweekly meetings were held for 12 to 16 months. Lastly, meetings were held monthly for 24 to 28 months, concentrating on maintaining a healthy lifestyle. Dietary data were collected through an interviewer-administered food frequency questionnaire, and cardiovascular fitness was assessed using a submaximal cycle ergometer test at baseline, 6 months, and 24 months (49). At 6 months, approximately 25% of females and 33% of males improved cardiovascular fitness. The authors concluded that a minority of youth improved fitness and/or diet over time, although those who did showed a beneficial impact on glycemic outcomes. However, the positive lifestyle behavior changes did not persist after 24 months, providing a sobering perspective on the challenges of implementing lasting lifestyle changes in this population.

 

Pharmacotherapy

 

The beginning of the third decade of the 21st century will be marked by the introduction of novel medications for children with T2D. There are several groups of medications for T2D, biguanides, sulfonylureas, GLP1 receptor agonists, SGLT2 inhibitors, dipeptidyl peptidase inhibitors, and combinations of the dual incretin analogs and the new triple G agents (dual incretin and glucagon). However, only biguanides, GLP1 receptor agonists, and SGLT2 inhibitors are currently FDA-approved for children. Doses and modes of administration are presented in Table 2.

 

Table 2. FDA-Approved Medications for Treating Type 2 Diabetes in Children Aged 10 Years and Older

Biguanide

Metformin

Glucophage

p.o daily

850, 1000. up to 3000 day

GLP receptor agonist

Liraglutide

Victoza

s.c. daily

0.6mg
1.2mg
 1.8mg

Exenatide

Bydureon

s.c weekly

2 mg

Dulaglutide

Trulicity

s.c weekly

0.75mg/0.5mL

1.5mg/0.5mL

3mg/0.5mL

4.5mg/0.5mL

SGLT2 inhibitor

Empagliflozin

Jardiance

p.o daily

10 mg

25 mg

Dapagliflozin

Forxiga

p.o daily

5mg
10 mg

 

Five randomized control studies assessed the impact of these drugs in children with T2D. The inclusion criteria were consistent across all studies and included an age above 10 years, a BMI above the 85th percentile, and minor variations in HbA1c levels (between 7.0 and 11.0%, or 6.5% and 10%). The characteristics of the participants and outcomes are detailed in Table 3. Figure 3 depicts HbA1c results vs. placebo for each medication, and Figure 4 depicts the percent of individuals with HbA1c less than 7% at the end of each study.

 

Table 3. Effects of Various Medications on Glycemic and Anthropometric Parameters in Adolescents with Type 2 Diabetes After 26 Weeks of Treatment

 

Liraglutide

 

Exenatide

Dulaglutide
0.75 mg

Dulaglutide

1.5 mg

Empagliflozin

Dapagliflozin

Linagliptin

Sitagliptin

 

Tx

Pl

Tx

Pl

Tx

PL

Tx

PL

Tx

PL

Tx

PL

Tx

Pl

Tx

Pl

Number

66

68

59

24

52

51

52

51

52

5

39

33

53

53

95

95

Age (years)

14.6

14.6

15

16

14.7

14.2

14.7

14.2

14.6

14.4

16.1

14.4

14.6

14.4

14.3

13.7

Baseline HbA1c )%(

7.9

7.7

8.1

8.1

7.9

8.1

8.2

8.1

8.0

8.05

7.95

7.85

8.05

8.05

7.4

7.6

HbA1c change

-0.64

0.46

-0.36

0.49

-0.6

0.6

-0.9

0.6

-0.17

0.68

-0.25

0.5

0.33

0.68

-0.01

0.18

HbA1c ETD 

1.06

0.85

1.2

1.5

0.84

0.75

0.35

0.19

% HbA1c <7%

63.7

36.5

48

35.1

55

14

48

14

34.6

24.5

25

4

26.5

24.5

49.5

36.8

Baseline BMI (kg/m2)

34.6

33.3

36.9

35.4

33.6

34.3

34.3

34.3

35.5

36.1

31.3

33.6

36.5

36.1

0.0

-0.7

BMI ETD

-0.25

-0.21

ND

ND

-0.2

0

-0.1

0

ND

ND

-0.08

-0.11

ND

ND

-0.7

Weight change (kg)

-2.3

-0.99

-0.59

0.63

0.3

0.1

0.2

0.1

-0.79

-0.04

ND

ND

1.42

-0.04

-1.1

0.06

Weight ETD

-1.3

-1.2

0.2

0.1

-0.75

 

1.46

1.04

Systolic BP ETD

0.03

-2.9

3.8

2.5

-1.42

1.9

0.91

 

Diastolic BP

ETD

-1.08

ND

-0.8

-2.6

0.02

-0.5

1.5

 

Pl=placebo, Tx= treatment, ETD = estimated treatment difference, ND= no data, placebo group for Empagliflozin and Linagliptin was the same.

 

Figure 3. Change in HbA1c percentage points in adolescents with T2D under medication treatment compared with untreated control group. The differences in HbA1c percentage points from the baseline for adolescents diagnosed with T2D who received medication treatment (depicted in black) compared to control groups (shown in white with data points). In the medication-treated group, there is a significant decrease in HbA1c levels, denoted by the downward trend in the black bars. Conversely, in the control groups of all the studies, HbA1c levels increased, as indicated by the upward trend in the white bars with data points.

Figure 4. HbA1c Levels below 7% in adolescents with T2D. The percentage of adolescents who achieved HbA1c levels below 7% at the end of the follow-up period. A comparison is drawn between those treated with medication (depicted in black) and the control group (depicted in white).

 

METFORMIN

 

Metformin is a biguanide antihyperglycemic used in conjunction with diet and exercise to control glycemia. Metformin is considered the preferred first-line agent for treating T2D in pediatric patients 10 or older (50). Its onset of action is around 1.5 hours, and its total duration of action is 16-20 hours. Metformin exerts its glucose-lowering effect through various mechanisms, including inhibiting hepatic gluconeogenesis, increasing glucose uptake in skeletal muscles, reducing adipogenesis, and activating brown adipose tissue, leading to enhanced thermogenesis. Additionally, metformin decreases the absorption of glucose from the intestine and downregulates inflammation.

Although it is the first line of treatment for children and adolescents with T2D, prospective, randomized, controlled, long-term studies are limited. In a randomized control study involving 82 children and adolescents, 42 received metformin at doses up to 2000 mg/day, and 40 received placebo (51). After 16 weeks, mean HbA1c levels were lower in the metformin group compared to the placebo (7.5% and 8.6%, respectively). The metformin group also experienced a mean weight decrease of -1.5 kg compared with a mean increase of 0.9 kg in the placebo group. The mean BMI changed by -0.5 units in the treatment group vs. -0.4 units in the placebo group. In another study, metformin (500–1000 mg twice daily) was compared to glimepiride, a sulfonylurea (1–8 mg once daily), for 24 weeks (52). Both metformin (-0.71%) and glimepiride (-0.54%) groups showed significant reductions from baseline HbA1c levels. A total of 48.1% of metformin-treated and 42.4% of glimepride-treated participants achieved HbA1c levels below 7.0% at week 24. While the incidence of hypoglycemia was similar in both groups, metformin resulted in less weight gain compared to glimepiride. In a large observational study (53), 927 youth aged 13.7±2.0 years old, who had been diagnosed with T2D for a median of 2 months and had a baseline HbA1c of 7.7±2.2%, were treated with metformin. After a median of 71 days, the mean HbA1c change was -1.33, and the mean weight change -0.43 kg (53,54).

 

Metformin-related side effects are gastrointestinal, including diarrhea, abdominal pain, bloating, nausea, and decreased appetite, which occur in about 50% of users. Gastrointestinal side effects are usually transient and improve over time. Another potential side effect of metformin is vitamin B12 deficiency, and regular monitoring is indicated, though no cases of vitamin B12 deficiency were identified in the TODAY study. Lactic acidosis is a concerning side effect associated with metformin use in adults. It occurs in the presence of hypoperfusion or hypoxemia; however, it has rarely been reported in children and adolescents.

GLUCAGON-LIKE-PEPTIDE 1 (GLP1) 

 

Glucagon-like peptide-1 (GLP-1) is a peptide hormone primarily produced by intestinal enteroendocrine cells at low basal levels that rapidly increases within minutes of food consumption. GLP-1 plays a vital role in regulating meal-related glycemic excursions by enhancing insulin secretion and inhibiting glucagon secretion. Additionally, GLP-1 slows gastric emptying and reduces food intake through a central effect on appetite, which contributes to weight loss. However, GLP-1 has a short half-life of approximately 2 minutes, as it is rapidly degraded by dipeptidyl peptidase-4 (DPP-4). To enhance GLP-1 activity, medications such as DPP-4 inhibitors and GLP-1 receptor agonists have been developed.


The story of the development of GLP1 agonists is truly fascinating. It begins with the venomous Gila monster (Heloderma suspectum), native to New Mexico and Arizona. H. suspectum, a long-lived and reclusive species, spends a significant portion of its life underground. Although its venomous bite causes pain and weakness, it is rarely fatal to adult humans. In 1990, endocrinologist Dr. John Eng, while analyzing the venom to identify new hormones, identified a peptide named exendin-4. He discovered that exendin-4 had a remarkable ability to stimulate the synthesis and release of insulin from β-cells in the pancreas. Interestingly, exendin-4 closely resembled GLP-1. However, while GLP-1 remains active for about two minutes, the effect of exendin-4 persists for several hours. Preclinical studies demonstrated that a single daily injection of exendin-4 normalized blood glucose concentrations in mice with diabetes. Following extensive clinical testing, exenatide, an analog of exendin-4, was found to be safe and effective, leading to its FDA approval for adults in 2005.

Of note, in a 2-year rat carcinogenicity study involving prolonged-release exenatide, an increased incidence of thyroid adenoma and C-cell carcinoma was observed at doses ≥2-fold the human systemic exposure. The clinical relevance of these adverse findings is currently unknown. However, GLP-1 analogs are contraindicated in patients with a personal or family history of medullary thyroid carcinoma (MTC) or in patients with multiple endocrine neoplasia type 2 (MEN 2), as well as in patients with a serious hypersensitivity reaction. Patients should be counseled about the potential risk of MTC associated with the GLP1 receptor agonists, and it is important to inform them about the symptoms that may indicate thyroid tumors, such as a neck mass, dysphagia, dyspnea, and persistent hoarseness. Routine monitoring of serum calcitonin levels or use of thyroid ultrasound for the detection of MTC is of uncertain value.

 

Side effects include serious hypersensitivity reactions, including anaphylactic reactions and angioedema. Additionally, acute pancreatitis, including fatal and nonfatal hemorrhagic or necrotizing pancreatitis, and the acute onset of gallbladder disease, such as cholelithiasis or cholecystitis, have been reported in GLP-1 receptor agonists trials and post-marketing surveys (55).

 

Liraglutide-Victoza® 

 

Children aged 10 to less than 17 years with T2D were randomly assigned 1:1 to receive subcutaneous liraglutide (up to 1.8 mg per day) or placebo for a 26-week double-blind period, followed by a 26-week open-label extension (56). At the 26-week visit, the mean HbA1c had decreased by 0.64 percentage points with liraglutide and increased by 0.42 percentage points with placebo, resulting in an estimated treatment difference of -1.06 percentage points. By 52 weeks, the difference had increased to -1.30 percentage points (Table 3). In the liraglutide group, 63.7% of patients achieved HbA1c levels of less than 7.0%, compared with 36.5% in the placebo group. On the other hand, there was no statistical benefit of liraglutide over placebo in lowering BMI or blood pressure (Table 3). The overall rate of adverse events, including gastrointestinal adverse events, was higher among patients receiving liraglutide. The U.S. Food and Drug Administration approved liraglutide injection for the treatment of pediatric patients 10 years of age or older with T2D. Liraglutide was the first non-insulin drug approved to treat T2D in pediatric patients since metformin was approved for pediatric use in 2000.

Mode of administration: The recommended initial dose is 0.6 mg subcutaneously once daily for the first week, followed by an increase to 1.2 mg once daily. It’s important to note that the initial dose of 0.6 mg once daily is primarily intended to mitigate gastrointestinal adverse effects and does not provide glycemic control. If adequate glycemic control is not achieved, the dose can be further increased to 1.8 mg once daily.

 

Exenatide-Bydureon  

 

The effectiveness of once-weekly exenatide 2 mg (Bydureon AstraZeneca) in youth with suboptimally controlled T2D was evaluated (57). At 24 weeks, exenatide demonstrated superiority over the placebo in reducing HbA1c levels (a decrease of -0.36% with exenatide compared to +0.49% with placebo), resulting in a significant between-group difference of 0.85 percentage points. Notably, no major hypoglycemic events were reported. and the most frequently reported adverse events included gastrointestinal symptoms such as nausea, diarrhea, and vomiting, along with injection site reactions such as pruritus, erythema, and nodules. Clinically meaningful differences were not observed in change from baseline in BMI Z score, body weight, or waist circumference, nor were there significant differences in blood pressure. The prolonged-release suspension of exenatide administered via injection with a pre-filled pen has received approval from the European Union and the FDA for the treatment of T2DM in children and adolescents aged 10 years and older.

Mode of administration: Bydureon (2 mg per dose) should be administered once every 7 days. The dose can be administered at any time of day, with or without meals. Bydureon is not recommended as first-line therapy for patients with inadequate glycemic control on diet and exercise because of the uncertain relevance of the rat thyroid C-cell tumor findings to humans.

Dulaglutide-Trulicity

 

Dulaglutide (Trulicity) is a GLP-1 receptor agonist administered via subcutaneous injections once a week. In a randomized, double-blind, phase 3 trial involving youth with inadequately controlled T2DM, the efficacy and safety of dulaglutide treatment in reducing HbA1c levels at 26 weeks were assessed (58). Remarkably, there was 99% adherence to treatment in this trial. Treatment resulted in a significant reduction in HbA1c relative to placebo; at week 26, HbA1c decreased by 0.8 percentage points but increased by 0.6 percentage points in the placebo group (1.4 percentage point difference). Gastrointestinal symptoms were among the most common adverse events, but they were primarily mild and were most likely to occur soon after the initiation of therapy. There were no clinically meaningful differences in the incidence or annual rate of hypoglycemia between the dulaglutide groups and the placebo group. Body weight did not decrease significantly following treatment.

Mode of administration: The recommended initiating dose of Trulicity is 0.75 mg once weekly, administered subcutaneously. The dose may be increased to 1.5 mg once a week for additional glycemic control. Trulicity can be administered any time of day, with or without meals, and should be injected subcutaneously in the abdomen, thigh, or upper arm. The day of weekly administration can be changed, if necessary, as long as the last dose was administered 3 days (72 hours) or more before. Trulicity and insulin should not be mixed in the same syringe and must be administered as two separate injections at two different injection sites.

 

SODIUM-GLUCOSE TRANSPORT PROTEIN 2 - SGLT2 INHIBITORS

 

Sodium-glucose transport protein 2 (SGLT2) inhibitors belong to a class of medications that modulate sodium-glucose transport proteins in the nephron. These inhibitors primarily target the SGLT2 proteins, expressed in the renal proximal convoluted tubules, to reduce the reabsorption of filtered glucose and sodium (59). Apart from their role in controlling glucose concentrations, SGLT2 inhibitors have demonstrated substantial cardiovascular benefits, particularly reducing heart failure events, for adults with T2D. In adults, the FDA-approved indications for SGLT2 inhibitors include the reduction of major adverse cardiovascular events in individuals with T2D and established cardiovascular disease, as well as the reduction of the risk of eGFR decline in patients with chronic kidney disease at risk of progression. The most common reported adverse events associated with SGLT2 inhibitors include female genital mycotic infections, urinary tract infections (including urosepsis and pyelonephritis), as well as nausea and constipation. Most importantly, SGLT2 inhibitors are associated with an almost three-fold increased risk for DKA in adults. The risk for DKA is highest for canagliflozin, followed by empagliflozin and dapagliflozin. Furthermore, individuals may develop euglycemic DKA, i.e., a triad of increased anion gap acidosis, ketosis, and a serum glucose level below 250 mg/dL. The association between SGLT-2 inhibitors and euglycemic DKA appears to be secondary to their noninsulin-dependent glucose clearance, compensatory hyperglucagonemia, and volume depletion. Currently, euglycemic DKA has not been reported among youth with T2D taking SLGT2 inhibitors.

 

Empagliflozin - Jardiance® 

 

A total of 105 children and adolescents were randomly assigned 1:1 to receive empagliflozin 10 mg or placebo (60). The adjusted mean change in HbA1c from baseline at week 26 was significantly greater in the empagliflozin group (–0·84%). Changes in body weight and blood pressure did not reach statistical significance. Adverse events occurred in 64% of participants in the placebo group and 77% in the empagliflozin group up to week 26, including severe adverse events in 4% of participants in the placebo group and 2% in the empagliflozin group. Mild and moderate hypoglycemia were the most frequently reported adverse events, with higher rates for those on active drug treatment compared with placebo; no severe hypoglycemic events were reported, and there were no episodes of DKA or necrotizing fasciitis associated with empagliflozin treatment. An increased incidence of bone fractures has been reported in adults, leading the American Diabetes Association to recommend avoiding SGLT-2 inhibitors in patients with a risk factor for fractures. Furthermore, SGLT2 inhibitors are not recommended for glycemic lowering in patients with T2D with an eGFR less than 30 mL/min/1.73 m2.

Mode of administration: Empagliflozin is an oral medication dosed at either 10 mg daily or 25 mg daily. The recommended dose is 10 mg once daily in the morning, taken with or without food. If tolerated initially, dosing may increase up to 25 mg. 

 

Dapagliflozin - Farxiga®

 

Participants aged 10–24 years with T2D and HbA1c concentrations ranging from 6.5 to 11% were given oral dapagliflozin 10 mg or placebo during a 24-week double-blind period (61); 39 participants were assigned to dapagliflozin and 33 were assigned to placebo. After 24 weeks, the mean change in HbA1c concentration was −0·25% for dapagliflozin and 0·50% for the placebo group; the between-group difference was −0·75%, which did not reach statistical significance. However, in a sub-analysis, excluding participants with poor compliance, the HbA1c change was -0.51%, while it was 0.62% for the placebo group, resulting in a between group significant difference of 1.13%.

Adverse events occurred in 69% of participants assigned to dapagliflozin and 58% of those assigned to placebo over a 24-week period. Over 52 weeks, adverse events occurred in 74% of participants who received dapagliflozin. Hypoglycemia was noted in 28% of participants assigned to dapagliflozin and in 18% of those assigned to placebo, none were considered serious adverse events. Notably, there were no reported episodes of DKA.

Mode of administration: To improve glycemic control the recommended starting dose is 5 mg once daily, taken in the morning. Increase dose to 10 mg once daily in patients tolerating 5 mg who require additional glycemic control. Dapagliflozin is not recommended for use in patients receiving loop diuretics or who are volume depleted, e.g., due to acute illness (such as gastrointestinal illness).

 

DIPEPTIDYL PEPTIDASE 4 (DPP 4) INHIBITORS

 

DPP-4 is a ubiquitous enzyme that degrades GLP-1 and GIP (gastric inhibitory peptide), resulting in a short half-life. By inhibiting the DPP-4 enzyme, DPP-4 inhibitors increase the levels of GLP-1 and GIP. This, in turn, enhances beta-cell insulin secretion, thus reducing postprandial and fasting hyperglycemia (62).


Linagliptin (Tradjenta)

 

105 children and adolescents were randomly assigned 1:1 to oral linagliptin (5mg daily) or placebo (60).  The adjusted mean HbA1c change from baseline at week 26 was 0.33% for linagliptin and 0·68% for the placebo group; the difference was not statistically or clinically significant. None of the adverse events of special interest occurred with DPP-4 inhibitors, such as hypersensitivity reactions, angioedema, angioedema-like events and anaphylaxis, skin lesions, pancreatitis, or hepatic injury, were present. Linagliptin is not approved for adolescents with T2D (60).

 

Sitagliptin (Januvia)

 

A total of 190 children and adolescents were randomly assigned 1:1 to oral sitagliptin 100 mg daily or placebo (63). The adjusted mean change in HbA1c from baseline at week 20 was -0.01% for sitagliptin and 0·18% for the placebo group, resulting in a between-group difference of 0.19%, which was not significant. There was no significant difference in the percentage of participants with HbA1c below 7%, with 49.5% in the sitagliptin group and 36.8% in the placebo group. Notably, after 54 weeks, the HbA1c in the sitagliptin group decreased from 7.4% at baseline to 7.1%. Sitagliptin is not approved for adolescents with T2D (50).

 

ANTI-OBESITY PHARMACOTHERAPY FOR TREATMENT OF PEDIATRIC TYPE 2 DIABETES

 

As obesity is a significant risk factor for the development of T2D, the American Diabetes Association (ADA) recommends the use of anti-obesity medications as adjuvant therapy for adults with both T2D and overweight/obesity. In adults, adding anti-obesity medications to a diabetes regimen can improve glycemic control, reduce weight, and decrease the use of anti-diabetes medication (64). Semaglutide and tirzepatide have shown promise in reducing weight in adults with obesity and T2DM compared to liraglutide (65). Tirzepatide is a novel dual glucose-dependent insulinotropic polypeptide (GIP) and GLP-1 receptor agonist. A study to evaluate tirzepatide in pediatric and adolescent participants with T2D, who are inadequately controlled with metformin, or basal Insulin, or both (SURPASS-PEDS) is currently ongoing.

Liraglutide 3.0mg/day 

 

A higher dose of liraglutide of 3.0 mg/day (Saxenda®) is FDA-approved for obesity in youth aged 12 years and older who weigh ≥ 60kg or have an initial BMI ≥ 30kg/m2. The Satiety and Clinical Adipose-Liraglutide Evidence Trial in Adolescents with and without T2D (SCALE-Teens) was a double-blind, placebo-controlled RCT that randomized 251 adolescents to either liraglutide 3.0 mg/day or placebo. However, only two adolescents with T2D were enrolled. Consequently, no conclusive findings can be drawn regarding the impact of high-dose liraglutide on reducing BMI and improving HbA1c in young individuals with T2D. Since almost all adolescents with T2DM have overweight or obesity and could potentially meet the prescribing criteria, it is reasonable to speculate that the results might resemble those observed in adult trials for T2D, potentially leading to reductions in weight and BMI as well as improvements in hyperglycemia.

 

Semaglutide 2.4mg Weekly

 

Semaglutide in its once-weekly injectable GLP-1RA form at doses up to 2.4 mg weekly (Wegovy®) has shown significant weight loss in obese children and has now been approved for treatment of obesity in adolescents aged 12 years and older (66). The mean change in BMI from baseline to week 68 was −16.1% with semaglutide and 0.6% with placebo. Of note, HbA1c decreased 0.4 percentage points in the treatment group compared with 0.1 in the placebo. However, the impact on youth with T2D has not yet been studied.

 

Semaglutide injection up to 2.0 mg (Ozempic®) and oral semaglutide up to 14 mg (Rybelsus®) are approved for adults with T2D, but studies in youth with T2D have not yet been reported.

SUMMARY: WHICH TREATMENT DO I RECOMMEND TO MY PATIENT?

 

The answer to this pivotal question hinges on the art of personalized medicine. First, it is crucial to recognize that 50% of youth with T2D can maintain good glycemic control on metformin monotherapy, making this agent still an excellent choice for initial therapy. However, several measures can predict the likelihood of glycemic deterioration, including decreased insulin secretion, impaired insulin processing (elevated proinsulin/insulin ratio), and HbA1c concentration after a few months on monotherapy. Longitudinally, a rising proinsulin and an increase in HbA1c of more than 0.5 percentage points over any 6-month period are predictive of loss of glycemic control on monotherapy (67).

 

If these parameters suggest a risk for deterioration, the next step is to consider the therapeutic objective(s). Is it primarily elevated HbA1c? If so, the best approach may involve selecting a medication known for its efficacy in reducing HbA1c. Is obesity the central concern for the patient? In such cases, exploring pharmaceutical interventions tailored for weight management may be prudent. Does the individual have concerns about needle-based treatments? In such instances, favoring orally administered medications could offer a more suitable solution. If there is an elevated risk of cardiac or renal complications, it would be wise to consider options from the SGLT2 inhibitor group or GLP1 agonist group. In essence, treatment needs to be tailored to the unique needs and circumstances of each individual, ensuring that treatment decisions align with their specific health challenges and objectives.

 

BARIATRIC SURGERY

 

Weight reduction plays a crucial role in the management of T2D because weight loss is associated with improved insulin resistance and glycemic status. Considering the limited efficacy of lifestyle and pharmacologic interventions in treating severe obesity and obesity-related T2D, along with the remarkable weight reduction and remission of T2D observed in youth who undergo bariatric surgery the ADA recommends considering bariatric surgery for adolescents with T2D who have a BMI >35 kg/m2 (68). The two most common bariatric surgeries performed in adolescents with T2D are laparoscopic sleeve gastrectomy (LSG) and laparoscopic Roux-en-Y gastric bypass (LRYGB).

 

Impact of Bariatric Surgery on Glycemic Control

 

Studies have demonstrated improvement in HbA1c across all age groups. In one study, sixty-four pediatric patients diagnosed with morbid obesity and T2D underwent LSG surgery. The patients’ ages ranged from 5 to 14 years old. Their BMI decreased from 44.6±9.3 to 34.8 ± 9.6 kg/m2.Their HbA1c decreased from 6.0±0.8% prior to LSG surgery to 5.4±0.4%, 12 months post-surgery, a change of 10.9% (p = 0.001) (69). There was no significant difference in post-operative HbA1c between the age groups.

 

Impact of Bariatric Surgery on T2D Remission

 

Bariatric surgery in adolescents with T2D has a better outcome compared to medical treatment with metformin alone or in combination with rosiglitazone or intensive lifestyle intervention with insulin therapy given for glycemic progression (70). In a multicenter, nonrandomized, retrospective study of 202 obese adolescents with T2D (before the approval of new drugs for adolescents with T2D), 109 adolescents underwent surgery, and 93 adolescents received nonsurgical treatment (71(. In the surgery group, the remission rate for diabetes was 76%, compared to 6.5% in the medical treatment group. The remission was sustained for the two years of follow-up. Of note, LRYGB had better effects on weight loss and glycemic control than LSG.


Impact of Bariatric Surgery on Kidney Outcomes

 

In a three-year longitudinal study, kidney outcomes in youth following bariatric surgery were assessed. Improvement in albuminuria and GFR was observed in those with pre-operative kidney impairment. Among participants with a preoperative eGFR < 90 ml/min/1/73m2, eGFR improved from a mean of 76 ml/min/1.73m2 to 102 ml/min/1.73m2. Similarly, in those with an elevated albumin-to-creatinine ratio at baseline (ACR ≥30mg/g), the median ACR decreased from 74 mg/g to 17 mg/g. Estimated GFR and albuminuria remained stable in those without any evidence of pre-operative impairment. The effect of bariatric surgery on diabetic kidney disease was also investigated in adolescents with severe obesity and T2D five years after surgery compared to adolescents receiving medical management in the TODAY study (72). Elevated albuminuria was present in 21% of those receiving medical management at baseline, and it increased to 43% at 5 years. Conversely, albuminuria decreased from 27% prior to surgery to 5% at the 5-year follow-up. In adjusted analyses, the medical group had 27-fold higher odds of diabetic kidney disease at 5 years compared to Teen-LABS participants (72-73).


Impact of Bariatric Surgery on CVD Events

 

In another analysis, the risk for CVD events in two cohorts of adolescents with T2D and severe obesity undergoing medical or surgical treatment of T2D was assessed. Thirty adolescents who underwent bariatric surgery were matched to 63 who had received metformin alone or in combination with rosiglitazone or an intensive lifestyle intervention, with insulin therapy given for glycemic progression. The BMI of those who underwent bariatric surgery was 54.4±9.5 kg/m2, and that of the medical group was BMI 40.5±4.9 kg/m2. Although the baseline likelihood of CVD events was higher in the group that underwent bariatric surgery, one year after the surgery, the event risk was significantly lower and sustained at 5-year follow-up, whereas medical therapy was associated with an increase in risk among adolescents with T2D and severe obesity (74).

 

Adverse Effects of Bariatric Surgery

Bariatric surgery in adolescents can lead to various potential complications beyond the first postoperative month. The most commonly reported include gastroesophageal reflux, incisional hernia, treatment failure requiring operative revision, and nutritional deficits (50). Less frequent complications include stomach cancer, liver necrosis, gallbladder disorders, pancreatic disorders, acute kidney failure, neuromuscular complications, skin complications, and rarely mortality (51).

 

Summary

 

In summary, bariatric surgery currently outperforms medical management of T2D in adolescents and is the most efficacious treatment for youth-onset T2D, albeit with significant risks and economic implications. However, given the rapidly emerging medical therapies for weight reduction in adults and youth, many of which (e.g., dual-incretin analogs, triple-G agonists (75)) are now achieving degrees of weight loss approaching those seen following surgery, the individual roles of surgical and medical treatment for youth-onset T2D will need continued reassessment in the coming years.

 

COMPLICATIONS AND COMORBIDITIES AMONG ADOLESCENTS WITH T2D

 

 Large cohort studies investigating the incidence of complications in children and teenagers with T2D unequivocally demonstrated a higher incidence of complications, both compared to the incidence of complications in T2D in adults and in T1D in children. In a multicenter observational study conducted from 2011 to 2020, the cumulative incidence of diabetic complications was assessed in 500 adolescents who had participated in the TODAY study (76). After only 13 years from diagnosis, higher complication rates were observed compared to those reported for pediatric patients with T1DM or for adults with T2D. The first striking finding from this report is the participants’ poor glycemic control despite participation in a well-resourced clinical trial. While glycemic control in adolescents with T1DM tends to improve with age, a gradual deterioration occurred in adolescents with T2D throughout the follow-up period. Approximately 45.0% had HbA1c of at least 10%, and an additional 20% were in the range of 8-10%. Their BMI remained consistently in the range of 35.0 to 37.5 kg/m2. It is important to note that poor glycemic control and obesity are associated with early complications. The complications associated with T2D in children affect various systems (Figure 5).

 

Figure 5. Comprehensive Overview of Type 2 Diabetes (T2D) Complications in Adolescents. A comprehensive visual representation of the diverse range of complications affecting major physiological systems that can arise in adolescents with T2D.

 

On average, thirteen years after the initial diagnosis of T2D, incidence rates for hypertension, dyslipidemia, diabetic kidney disease, and neuropathy were 67.5%, 51.6%, 54.8%, and 32.4%, respectively. Additionally, retinopathy was present in 51% of the participants. One-fifth of the cohort (21.3%) had two complications, and 7.1% had three. There were seventeen serious cardiovascular events, including myocardial infarctions, six cases of congestive heart failure, three cases of coronary artery disease, and four stroke events. Additionally, six deaths were reported. This rapid development of complications was associated with severe insulin resistance and poor socioeconomic circumstances.

Hypertension

 

Hypertension, defined as blood pressure ≥ 95th percentile for age, sex, and height or systolic BP ≥ 130/80 mm Hg, is often present at the time of diagnosis in adolescents with T2D. In a study of 391 children from Hong Kong under 18 years of age, 22.5% had hypertension at diagnosis. In the TODAY study, 11.6% were hypertensive at the time of diagnosis (77) and the cumulative incidence of hypertension was 67.5% at a mean age of 26.4±2.8 years and 13.3±1.8 years from diagnosis (78). A systematic review and meta-analysis of 31 international studies on 4363 children with T2D aged 6.5 to 21 years at diagnosis found a pooled prevalence of hypertension of 25.33% (95% CI, 19.57%-31.53%) (79).Male participants had a higher hypertension risk than female participants (odds ratio 1.42 [95% CI, 1.10-1.83]).

 

Initial treatment involves lifestyle modification, aiming to not only enhance glycemic control but also incorporate a diet that is low in sodium and abundant in fruits, vegetables, whole grains, low-fat dairy products, and lean proteins. The Dietary Approaches to Stop Hypertension (DASH diet) is highly recommended for its effectiveness in reducing high blood pressure and supporting weight loss. In addition to lifestyle modification, the ADA recommends starting Angiotensin-Converting Enzyme inhibitors (ACEis) or Angiotensin Receptor Blockers (ARBs). Due to the potential teratogenic effects, the use of reliable contraception should be encouraged in individuals of childbearing age (68). There are no agents specifically approved for use in youth with T2D, however, a once-daily medication is recommended for better compliance (80,81).   

 

Nephropathy

 

The primary microvascular diabetic complication in adolescents with T2D is diabetic kidney disease (DKD), which develops in 25–40% of T2D patients and is associated with rapid progression and poor prognosis. The severity and risk of rapid deterioration in young people with T2D are reflected in the fact that microalbuminuria is reported at diagnosis among adolescents with T2D. In a recent systematic review and meta-analysis (79) that included 14 studies of 2250 children and adolescents with T2D, the prevalence of albuminuria was 22.17% (95%CI, 17.34%-27.38%), and the pooled prevalence of macroalbuminuria among 730 children and adolescents was 3.85% (79). In the SEARCH study (82) after a duration of 8 years, the prevalence was 19.9% among adolescents with T2D compared with 5.8% in those with T1D.

The accelerated development of kidney damage is attributed to inadequate glycemic control and the presence of various risk factors, including obesity, dyslipidemia, insulin resistance, hypertension, elevated serum uric acid, female sex, and the presence of chronic inflammation (83).

While albuminuria is the earliest clinical sign of diabetic kidney disease (DKD), the natural history of DKD begins with hyperfiltration, characterized by an increase in glomerular filtration rate (GFR) >120 mL/min/1.73 m² as a consequence of obesity and impaired glucose tolerance (84).  This increase predicts deterioration before other clinical signs appear. The second stage in the evolution of kidney dysfunction, still without clinical manifestations, is a mild reduction in GFR (60-89 mL/min/1.73 m²) (Figure 6). Structural changes of the kidney are typical at this stage, yet they are often reversible, making this a critical time for risk factor reduction (84).

 

Figure 6. Heat map depicting diabetes kidney disease (DKD) staging by GFR and albuminuria and suggested treatments. Adapted from Naaman SC, Bakris GLJDC. Diabetic Nephropathy: Update on Pillars of Therapy Slowing Progression. 2023;46:1574-86.

 

Treatment in these early stages involves achieving adequate glycemic control along with lifestyle modification, including increasing physical activity, improving sleep quality, and quitting smoking. Additionally, a healthy diet with lower salt and potassium intake, along with a low protein intake of 0.8 g/kg/day, is recommended (68). Pharmacological treatment includes control of blood pressure and managing dyslipidemia. Theoretically, medications that improve glycemic control would also provide benefits for preventing and/or treating DKD. GLP1 receptor agonists are reported to have a renal protective effect in adults with T2D. A recent meta-analysis of approximately 60,000 adults with T2DM treated with GLP-1 receptor agonist found a significant reduction in the composite kidney outcome (development of macroalbuminuria, doubling of serum creatinine, 40% or greater decline in eGFR, kidney replacement therapy, or death from kidney disease) when compared to placebo, as well as a trend towards a reduction in worsening kidney function (84). Similarly, the hemodynamic and natriuretic effects of SGLT2 inhibition are also postulated as protective mechanisms for DKD (85).However, studies in adolescents with T2D did not show an effect of systolic or diastolic blood pressure (Table 3). Of note, these studies involved relatively small groups of youth, and were short-term. Larger, long-term studies are needed, as well as additional therapeutic options for reducing the risk of DKD, such as mineralocorticoid receptor antagonists (MRAs), and endothelin receptor antagonists (ERAs).

Dyslipidemia

 

Screening for dyslipidemia should be conducted once optimal glycemic control has been achieved or within 3 months after T2D diagnosis. Initial cholesterol screening can be performed non-fasting. The recommended target values are triglycerides <150 mg/dL (1.7 mmol/L), HDL cholesterol >35 mg/dL (0.91 mmol/L), and LDL cholesterol <100 mg/dL (2.6 mmol/L). If these levels are outside the normal range, medical nutritional therapy (MNT) should be initiated, including restricting caloric intake from fat to 20–30% and daily cholesterol intake to <200 mg/day, avoiding trans fatty acids, limiting saturated fats to <7%, and controlling mono and poly unsaturated fats to 10-15%. If, after 6 months of dietary intervention, the LDL-C remains >130 mg/dL, statin therapy should be initiated, with a goal of achieving LDL-C <100 mg/dL. Currently, there are 7 approved statins for use in children and adolescents: Lovastatin, Simvastatin, Atorvastatin, and Fluvastatin [children ≥10 years old], Pitavastatin and Pravastatin [> 8 years], and Rosuvastatin [>6 years old]. It is important to note that due to potential teratogenic effects, individuals of childbearing age should receive reproductive counseling, and statins should be avoided in individuals of childbearing age who are not using reliable contraception. If triglyceride levels are greater than 400 mg/dL (4.7 mmol/L) in a fasting state or exceed 1,000 mg/dL (11.6 mmol/L) in a non-fasting state, it is imperative to optimize glycemic control and initiate fibrate therapy. This is essential for reducing the risk of pancreatitis.

 

In a study assessing whether clinicians are sufficiently aggressive in treating diabetes-related hyperlipidemia in youth, among 278 youth with T2D <10 years, 57% had LDL-C exceeding 100 mg/dL, 24% had LDL-C at or above 130 mg/dL, and 9% had LDL-C surpassing 160 mg/dL (86). Additionally, 29% had hypertriglyceridemia, and 44% had HDL-C levels below 40 mg/dL. However, only 5% of these youth were prescribed lipid-lowering medications.

 

Polycystic Ovarian Syndrome (PCOS)

 

There is a strong association between T2D in adolescent girls and polycystic ovary syndrome (PCOS). A systematic review and meta-analysis comprising 6 studies that involved 470 girls diagnosed with T2D, with an age range at diagnosis between 12.9 and 16.1 years, revealed that the prevalence of PCOS was estimated at 19.58% (87). PCOS is associated with a range of metabolic diseases, including hypertension and dyslipidemia. Furthermore, it is associated with a higher prevalence of cardiovascular risk factors, such as higher carotid intima thickness, β stiffness index, and reduced arterial compliance. Additionally, adolescent girls with PCOS have an increased likelihood of experiencing psychiatric comorbidities, such as anxiety and depression, which can negatively impact their health-related quality of life. A menstrual history should be taken on every girl with T2D at the diagnosis and at every follow-up visit. Metformin, in addition to lifestyle modification, is likely to improve menstrual cyclicity and hyperandrogenism in female individuals with T2D.

 

Pregnancy and Contraception

 

In the TODAY study, the maternal and offspring outcomes of young women with youth-onset T2D who experienced one or more pregnancies were evaluated (88). Over a span of up to 15 years, a total of 260 pregnancies were reported by 141 women. Their average age was 21.5±3.2 years, a mean BMI of 35.6±7.2 kg/m2, and an average diabetes duration of 8.1±3.2 years (88). Complications during pregnancy were identified in 65% of the women. Elevated HbA1c levels of ≥8% were observed in 31.9% of the pregnancies, and chronic hypertension complicated 35% of them. Pregnancy loss was observed in 25.3% and preterm birth occurred in 32.6% of pregnancies. Among the offspring, 7.8% of the babies were classified as small for gestational age, 26.8% as large for gestational age, and 17.9% fell into the macrosomic range. Other complications and congenital anomalies were noted in 10% of infants, including anencephaly, renal anomalies, and complications related to prematurity. The rate of known miscarriage for the entire study was 12.3%. The rate of known stillbirths in the cohort was 3%, which is more than triple the reported national rates.

 

The impact of T2D during pregnancy extends beyond the immediate prenatal and birth periods, exerting long-term effects on offsprings. Data pertaining to the diagnosis of diabetes in biological mothers were available for 621 participants in the TODAY study, of whom 301 had never been diagnosed with diabetes, 218 were diagnosed either before or during pregnancy, and 102 were diagnosed after pregnancy (89). For biological fathers, the data were available for 519 participants, with 352 having no history of diabetes and 167 having paternal diabetes. Notably, it was found that maternal diabetes, but not paternal diabetes, was associated with lower beta-cell function and deterioration of glycemic control over time. These effects remained significant even after accounting for variables such as age, sex, race/ethnicity, and household income (89).

 

Contraception is indicated in adolescent girls with T2D for several compelling reasons. Firstly, it is a recommendedtreatment for managing the symptoms of PCOS. Secondly, these young women are at increased risk of unintended pregnancy, which can lead to poor outcomes. Lastly, it is imperative for those treated with potentially teratogenic medications such as ACEis, ARBs, and statins. Unfortunately, despite the evident need, preconception counseling was reported only in 16.3% of women prior to their first pregnancy and only 14.9% used any method of contraception prior to the first pregnancy (88). This gap in reproductive health care highlights the importance of healthcare providers and support staff addressing this issue promptly and effectively.

 

Although in theory, adolescent girls with T2D without severe obesity, micro- or macrovascular disease, or other cardiovascular risk factors can consider a wide range of contraceptive methods, the practical reality often differs. A significant proportion of these individuals are afflicted by severe obesity, and a considerable number also exhibit elevated blood pressure. Consequently, when morbid obesity, severe hypertension, micro- or macrovascular disease, or multiple cardiovascular risk factors are present, it is advisable to prioritize nonhormonal or progestin-only contraceptive methods (Figure 7) (90).

 

Figure 7. Heat map of contraceptive treatment options for adolescent girls with T2D based on disease duration and complications. Adapted from Merino PM, Codner E. Contraception for Adolescents and Young Women with Type 2 Diabetes-Specific Considerations. Current diabetes reports 2022;22:77-84.

 

Diabetic Retinopathy 

 

Diabetic retinopathy (DR) is one of the most important causes of visual loss worldwide. DR is often asymptomatic until the very late stages. Given the potential for a rapid rate of progression and the effectiveness of new therapies in slowing deterioration, it is imperative to routinely screen individuals with diabetes for the early detection of retinal changes. It is important to note that direct fundoscopy may be less sensitive than 7-field stereoscopic fundus photography for detecting retinopathy (91). Diabetic retinopathy (DR) is divided into two major forms: non-proliferative and proliferative, according to the presence of abnormal new blood vessels emanating from the retina. Non-proliferative DR consists of microvascular abnormalities (including microaneurysms, occluded vessels, and dilated or tortuous vessels) primarily in the macula and posterior retina, intraretinal hemorrhages, hard exudates; and the variable presence of nerve-fiber layer infarcts (cotton wool spots). Visual loss is mainly due to the development of macular edema. Proliferative DR is marked by the presence of neovascularization arising from the disc and/or retinal vessels, resulting in preretinal and vitreous hemorrhage, subsequent fibrosis, and traction retinal detachment resulting in visual loss.

 

In a meta-analysis that included 27 studies involving 5924 children and adolescents (91) with T2D duration between 6.5-21.0 years, 1.11% had DR less than 2.5 years after T2D diagnosis. The prevalence of DR increased over time, with rates of 9.04% at 2.5 to 5.0 years after T2D diagnosis, and 28.14% more than 5 years after T2D diagnosis. The global prevalence of diabetic retinopathy in pediatric T2D was found to be 6.99%. Optical coherence tomography (OCT)performed in adolescents with T2D in the TODAY study revealed that changes in retinal thickness correlated with HbA1c, fasting glucose, and blood pressure, illustrating the importance of intensive control of hyperglycemia, hypertension, and hyperlipidemia.(92)

 

Among the systemic strategies for the prevention and treatment of diabetic retinopathy, two randomized clinical trials, "The Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study and the Action to Control Cardiovascular Risk in Diabetes (ACCORD)-Eye Study, documented a reduction in the risk of non-proliferative DR progressing in individuals using fenofibrate (93). The protective effects of fenofibrate have been attributed to its antioxidative, anti-inflammatory, anti-apoptotic, and anti-angiogenic properties. Other treatments include pan-retinal photocoagulation, intravitreal anti-vascular endothelial growth factor (VEGF) therapy, integrin antagonists, and anti-inflammatory agents. Corticosteroids remain important second-line therapies for patients with macular edema.

 

Diabetic Neuropathy

 

DIABETIC PERIPHERAL NEUROPATHY

 

Diabetic peripheral neuropathy (DPN) is a cause of significant disability and poor quality of life. Signs and symptoms include sensory loss, paresthesia, and pain. However, subclinical signs of DPN may precede the development of frank neuropathic symptoms, and systematic screening is required to identify DPN in its earliest stages. Studies from USA (78,94), Canada (95), and India (96) reported the prevalence of DPN in adolescents with T2D assessed by examination for foot abnormalities, distal vibration perception, and ankle reflexes (97). In the SEARCH study, the prevalence of peripheral neuropathy assessed after 8 years in 258 adolescents with T2D was 22%, compared with 7% in T1DM (98). Data from the TODAY Study Group revealed that at baseline, 1.0% of the participants had nerve disease, and the cumulative incidence at 15 years was 32.4% (78). The risk factors associated with peripheral neuropathy in youth are longer duration of diabetes, older age, male sex, and smoking.  

Calcium channel a2δ ligands (gabapentin, pregabalin= Lyrica), serotonin- norepinephrine reuptake inhibitors, and tricyclic antidepressants are the most widely used medications for painful DN in adults. These medications are used to treat various presentations of neuropathic pain in children, but there have been no clinical studies devoted to painful DPN and no agents are licensed specifically for painful DN in childhood and youth (99). “Fortunately, painful DPN is relatively rare in children and youth.

AUTONOMIC NEUROPATHY 

 

Diabetic autonomic neuropathy (DAN) is a common form of neuropathy in individuals with diabetes mellitus characterized by dysfunction due to impairment of peripheral autonomic nerves, with a wide spectrum of manifestations (100). DAN that involves autonomic fibers of the enteric nervous system may cause upper and lower gastrointestinal symptoms, including gastroesophageal reflux disease, gastroparesis, and constipation. Additional manifestations include bladder and sexual dysfunction. DAN also involves the cardiovascular system. Cardiovascular autonomic neuropathy (CAN), secondary to the pathology of the autonomic nerve fibers that innervate the heart and blood vessels, results in resting tachycardia, exercise intolerance, orthostatic hypotension, syncope, and silent myocardial infarction and ischemia(101). A reduction in heart rate variability is an early sign of CAN and was reported in 47% of youth with T2D after a mean disease duration of only 1.7 years (102). In the SEARCH study, the prevalence of CAN assessed after duration of 8 years in 252 adolescents with T2D was 17% compared with 12% in T1DM (103). CAN was associated with elevated triglycerides and increased urinary albumin excretion.

 

Cardiovascular Complications 

 

T2D is a major risk factor for cardiovascular disease (CVD), including myocardial infarction and stroke. Markers for early CVD include increased arterial stiffness, carotid intima-media thickness, an alteration in the vascular endothelium, and cardiac autonomic neuropathy.
Arterial stiffness prevalence was significantly higher among youth with T2D than those with T1D (104). Arterial stiffness (measured by pulse wave velocity from carotid-femoral, femoral-foot, and carotid-radial), was measured in 388 youth with T2D at a mean age of 21 years and a diabetes duration of 7.7±1.5 years (105). Higher arterial stiffness was associated with an adverse change in left ventricular diastolic function. Similarly, cardiac parameters were assessed in 177 individuals with T2D, with a mean age of 24.6±4.2 years and a disease duration of 10.3±3.5 years (106). Of these, 72% were females, and their HbA1c levels averaged 8.9±1.9. Their parameters were compared to those of individuals with T1D. The prevalence of increased atrial stiffness was significantly higher in those with T2D, at 75.7%, compared to only 23.4% in individuals with T1D. In addition, the prevalence of cardiac autonomic neuropathy was significantly higher in those with T2D, at 16.9%, compared to 9.0% in individuals with T1D. Participants with T2D exhibited a higher left ventricular mass index, lower systolic function, and lower diastolic function compared to those with T1D.

Adolescents with T2DM have increased carotid intima-media thickness (cIMT) compared to obese and normal weight groups; the group difference was detected early at 14 years of age (107).  

Vascular endothelial function was lower in adolescents with T2D compared to normal weight and compared to adolescents with T1D (108).  These changes are of importance as the rate of all adjudicated heart, vascular, and cerebrovascular events was 3.73 per 1000 person-years (78). There were 17 serious cardiovascular events, myocardial infarction [4 events], congestive heart failure [6 events], coronary artery disease [3 events], and stroke [4 events].

 

Decreased Bone Mineral Density (BMD) 

 

In a study involving 17 adolescents newly diagnosed with T2D and 59 age, sex, and BMI-matched controls, bone mineral density (BMD) was assessed at multiple sites while accounting for potential confounding factors such as age, sex, Tanner stage, and BMI (109). The results revealed that BMD Z-scores for the femoral neck and bone mineral apparent density Z-scores of the lumbar spine were notably lower in individuals with T2D compared to their healthy counterparts.In another cross-sectional study of BMD in 180 youths aged 10 to 23 years, the participants were categorized into three groups: those with T2D were compared to those with obesity (n=226) and those with a healthy weight (BMI <85th percentile; n=238) (110). An age-dependent pattern emerged, wherein the BMD Z-score in children was higher in the T2D group compared to the obese group. However, in adolescents and young adults, the BMD Z-scores were lower in the T2D group when compared to the obese group. These findings suggest that T2D may have a detrimental impact on bone density, particularly around the age when individuals reach peak bone mass. Considering the elevated fracture risk observed in adults with T2D, the decrease in bone density at a young age raises concerns about potential long-term morbidity and fracture risk in adulthood.

Mental Health Comorbidity

 

DEPRESSION  

 

As part of routine diabetes care, a total of 197 adolescents and youth diagnosed with T2D, with a mean age of 16.9 years and 57% male, completed the Patient Health Questionnaire (PHQ) (111). 19.3% reported elevated depressive symptoms (PHQ score ≥10). Furthermore, in a sample of 53, 18.9% acknowledged thoughts of self-harm. Despite the prevalence of depressive symptoms, only 50.0% of those with depressive symptoms had a documented referral for mental health treatment in the electronic health record after the positive screening outcome. Older age, shorter diabetes duration, a higher HbA1c level, more blood glucose checks per day, and being prescribed oral medications were significantly associated with more depressive symptoms. Results from the TODAY study revealed that older teen girls had the highest rates of clinically significant depressive symptoms (112). Increased number of stressful life events were associated with elevated depressive symptoms. Depressive symptoms were correlated with low adherence to diabetes treatment, lower psychosocial functioning, and impaired health-related quality of life (QOL). In another study comparing PHQ between adolescents with T2D and T1D, the median PHQ-9 score in females with T2D was significantly greater than in females with T1D, but did not differ between males with T2D and T1D (113). The association between depression and T2D has been reviewed elsewhere (114).


DISORDERED EATING BEHAVIORS

 

In the TODAY study, out of 678 adolescents with T2D at a mean age of 14.0 years, 6% had clinical and 20% had subclinical levels of binge eating (115). Moreover, 50.3% of adolescents with T2D who are receiving insulin therapy had disordered eating behaviors (DEB) compared with 21.2% of those with T1DM (116). Disordered eating behavior is associated with poorer clinical outcomes and psychosocial well-being.

 

Mortality

 

In the SEARCH study, over a median follow-up of 5.3 years, the mortality rate among individuals with T1D was 70.6 deaths per 100,000 patient-years, compared with 185.6 deaths per 100,000 patients-years for those with T2D (117)..When compared to the general populations of US states, it was observed that the mortality rate for individuals with T2D was significantly higher than expected while this was not the case for individuals with T1DM. Females had higher mortality rates than males. Among adolescents with T2D, the leading underlying cause of death was attributed to transport/motor vehicle accidents followed by accidental poisoning, and intentional self-harm. According to a life expectancy model, it was predicted that youth with T2DM lose approximately 15 years (118).

 

CONCLUSION

 

A summary of the key points is presented in Table 4.

 

Table 4. Key Points

•           The prevalence of type 2 diabetes (T2D) varies across different countries and regions globally, but there is a consistent upward trend in its occurrence. Large-scale national studies in countries like Sweden, Germany, and India have reported similar increasing trends.

•           Contrary to common perception, not all adolescents with T2D are severely obese. Only 70-80% fall into this category.

•           Socioeconomic status plays a significant role in the prevalence of T2D, with lower socioeconomic status being associated with higher incidence in Western countries, while the opposite is observed in low-income countries.

•           Although adolescents are at higher risk of developing T2D, there are growing reports of T2D cases in children younger than 10 years old.

•           The gender distribution of T2D varies, with a higher proportion of cases in females in Western countries but a higher proportion in males in the Middle East and China.

•           Promisingly, there are new medications designed for weight loss, which hold the potential to reduce the high rates of complications associated with T2D in children.

•           There are low prescription rates for statins and contraception in the management of T2D in adolescents, highlighting potential gaps in medical care.

•           Many complications of T2D can potentially be reversed if detected and treated aggressively in their early stages.

 

 

ACKNOWLEGEMENTS

 

The authors extend their heartfelt gratitude to the gifted illustrator, Mrs Noah Levek, for her exceptional eye-opening illustrations.

 

REFERENCES

 

  1. Knowler WC, Pettitt DJ, Savage PJ, Bennett PH. Diabetes incidence in Pima indians: contributions of obesity and parental diabetes. Am J Epidemiol 1981; 113:144-156
  2. Pinhas-Hamiel O, Dolan LM, Daniels SR, Standiford D, Khoury PR, Zeitler P. Increased incidence of non-insulin-dependent diabetes mellitus among adolescents. J Pediatr 1996; 128:608-615
  3. Glaser NS, Jones KL. Non-insulin dependent diabetes mellitus in Mexican-American children. West J Med 1998; 168:11-16
  4. Neufeld ND, Raffel LJ, Landon C, Chen YD, Vadheim CM. Early presentation of type 2 diabetes in Mexican-American youth. Diabetes Care 1998; 21:80-86
  5. Stahl-Pehe A, Kamrath C, Prinz N, Kapellen T, Menzel U, Kordonouri O, Schwab KO, Bechtold-Dalla Pozza S, Rosenbauer J, Holl RW. Prevalence of type 1 and type 2 diabetes in children and adolescents in Germany from 2002 to 2020: A study based on electronic health record data from the DPV registry. J Diabetes 2022; 14:840-850
  6. Tung JY, Kwan EY, But BW, Wong WH, Fu AC, Pang G, Tsang JW, Yau HC, Belaramani K, Wong LM, Wong SM, Lo P, Ng KL, Yeung WK, Chan KT, Chan AM, Wong SW, Tay MK, Chung J, Lee CY, Lam YY, Cheung PT. Incidence and clinical characteristics of pediatric-onset type 2 diabetes in Hong Kong: The Hong Kong childhood diabetes registry 2008 to 2017. Pediatr Diabetes 2022; 23:556-561
  7. Urakami T, Miyata M, Yoshida K, Mine Y, Kuwabara R, Aoki M, Suzuki J. Changes in annual incidence of school children with type 2 diabetes in the Tokyo Metropolitan Area during 1975-2015. Pediatr Diabetes 2018; 19:1385-1392
  8. Hockett CW, Praveen PA, Ong TC, Amutha A, Isom SP, Jensen ET, D'Agostino RB, Jr., Hamman RF, Mayer-Davis EJ, Lawrence JM, Pihoker C, Kahn MG, Mohan V, Tandon N, Dabelea D. Clinical profile at diagnosis with youth-onset type 1 and type 2 diabetes in two pediatric diabetes registries: SEARCH (United States) and YDR (India). Pediatr Diabetes 2021; 22:22-30
  9. Zuckerman Levin N, Cohen M, Phillip M, Tenenbaum A, Koren I, Tenenbaum-Rakover Y, Admoni O, Hershkovitz E, Haim A, Mazor Aronovitch K, Zangen D, Strich D, Brener A, Yeshayahu Y, Schon Y, Rachmiel M, Ben-Ari T, Levy-Khademi F, Tibi R, Weiss R, Lebenthal Y, Pinhas-Hamiel O, Shehadeh N. Youth-onset type 2 diabetes in Israel: A national cohort. Pediatr Diabetes 2022; 23:649-659
  10. Wagenknecht LE, Lawrence JM, Isom S, Jensen ET, Dabelea D, Liese AD, Dolan LM, Shah AS, Bellatorre A, Sauder K, Marcovina S, Reynolds K, Pihoker C, Imperatore G, Divers J, study SfDiY. Trends in incidence of youth-onset type 1 and type 2 diabetes in the USA, 2002-18: results from the population-based SEARCH for Diabetes in Youth study. The lancet Diabetes & endocrinology 2023; 11:242-250
  11. Wise J. Type 2 diabetes: Charity warns of "perfect storm" putting more children at risk. BMJ (Clinical research ed)2022; 377:o1451
  12. Wu H, Zhong J, Yu M, Wang H, Gong W, Pan J, Fei F, Wang M, Yang L, Hu R. Incidence and time trends of type 2 diabetes mellitus in youth aged 5-19 years: a population-based registry in Zhejiang, China, 2007 to 2013. BMC pediatrics 2017; 17:85
  13. Biondi G, Marrano N, Borrelli A, Rella M, Palma G, Calderoni I, Siciliano E, Lops P, Giorgino F, Natalicchio A. Adipose Tissue Secretion Pattern Influences &beta;-Cell Wellness in the Transition from Obesity to Type 2 Diabetes. 2022; 23:5522
  14. Cioana M, Deng J, Nadarajah A, Hou M, Qiu Y, Chen SSJ, Rivas A, Banfield L, Toor PP, Zhou F, Guven A, Alfaraidi H, Alotaibi A, Thabane L, Samaan MC. The Prevalence of Obesity Among Children With Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Network Open 2022; 5:e2247186-e2247186
  15. Copeland KC, Zeitler P, Geffner M, Guandalini C, Higgins J, Hirst K, Kaufman FR, Linder B, Marcovina S, McGuigan P, Pyle L, Tamborlane W, Willi S. Characteristics of Adolescents and Youth with Recent-Onset Type 2 Diabetes: The TODAY Cohort at Baseline. The Journal of Clinical Endocrinology & Metabolism 2011; 96:159-167
  16. Lowe WL, Jr., Lowe LP, Kuang A, Catalano PM, Nodzenski M, Talbot O, Tam WH, Sacks DA, McCance D, Linder B, Lebenthal Y, Lawrence JM, Lashley M, Josefson JL, Hamilton J, Deerochanawong C, Clayton P, Brickman WJ, Dyer AR, Scholtens DM, Metzger BE. Maternal glucose levels during pregnancy and childhood adiposity in the Hyperglycemia and Adverse Pregnancy Outcome Follow-up Study. Diabetologia 2019; 62:598-610
  17. Dabelea D, Mayer-Davis EJ, Lamichhane AP, D'Agostino RB, Jr., Liese AD, Vehik KS, Narayan KM, Zeitler P, Hamman RF. Association of intrauterine exposure to maternal diabetes and obesity with type 2 diabetes in youth: the SEARCH Case-Control Study. Diabetes Care 2008; 31:1422-1426
  18. Shah AS, Nadeau KJ, Dabelea D, Redondo MJ. Spectrum of Phenotypes and Causes of Type 2 Diabetes in Children. Annual review of medicine 2022; 73:501-515
  19. Rughani A, Friedman JE, Tryggestad JB. Type 2 Diabetes in Youth: the Role of Early Life Exposures. Current diabetes reports 2020; 20:45
  20. Norrman E, Petzold M, Gissler M, Spangmose AL, Opdahl S, Henningsen AK, Pinborg A, Tiitinen A, Rosengren A, Romundstad LB, Wennerholm UB, Bergh C. Cardiovascular disease, obesity, and type 2 diabetes in children born after assisted reproductive technology: A population-based cohort study. PLoS medicine 2021; 18:e1003723
  21. Murphy HR, Howgate C, O'Keefe J, Myers J, Morgan M, Coleman MA, Jolly M, Valabhji J, Scott EM, Knighton P, Young B, Lewis-Barned N. Characteristics and outcomes of pregnant women with type 1 or type 2 diabetes: a 5-year national population-based cohort study. The lancet Diabetes & endocrinology 2021; 9:153-164
  22. Wibaek R, Andersen GS, Linneberg A, Hansen T, Grarup N, Thuesen ACB, Jensen RT, Wells JCK, Pilgaard KA, Brøns C, Vistisen D, Vaag AA. Low birthweight is associated with a higher incidence of type 2 diabetes over two decades independent of adult BMI and genetic predisposition. Diabetologia 2023;
  23. Crump C, Sundquist J, Sundquist K. Preterm birth and risk of type 1 and type 2 diabetes: a national cohort study. Diabetologia 2020; 63:508-518
  24. Nouwen A, Adriaanse MC, van Dam K, Iversen MM, Viechtbauer W, Peyrot M, Caramlau I, Kokoszka A, Kanc K, de Groot M, Nefs G, Pouwer F. Longitudinal associations between depression and diabetes complications: a systematic review and meta-analysis. Diabet Med 2019; 36:1562-1572
  25. Omer Gilon M, Balmakov Y, Gelman S, Twig G. Adolescent Immigration and Type-2 Diabetes. Current diabetes reports 2021; 21:60
  26. Hamiel U, Pinhas-Hamiel O, Vivante A, Bendor C, Bardugo A, Afek A, Beer Z, Derazne E, Tzur D, Behar D, Itzhak A, Skorecki K, Tirosh A, Grossman E, Twig G. Impact of Immigration on Body Mass Index and Blood Pressure Among Adolescent Males and Females: A Nationwide Study. Hypertension 2019; 74:1316-1323
  27. Khanolkar AR, Amin R, Taylor-Robinson D, Viner RM, Warner J, Stephenson T. Inequalities in glycemic control in childhood onset type 2 diabetes in England and Wales-A national population-based longitudinal study. Pediatr Diabetes 2019; 20:821-831
  28. Magge SN, Wolf RM, Pyle L, Brown EA, Benavides VC, Bianco ME, Chao LC, Cymbaluk A, Balikcioglu PG, Halpin K, Hsia DS, Huerta-Saenz L, Kim JJ, Kumar S, Levitt Katz LE, Marks BE, Neyman A, O'Sullivan KL, Pillai SS, Shah AS, Shoemaker AH, Siddiqui JAW, Srinivasan S, Thomas IH, Tryggestad JB, Yousif MF, Kelsey MM. The Coronavirus Disease 2019 Pandemic is Associated with a Substantial Rise in Frequency and Severity of Presentation of Youth-Onset Type 2 Diabetes. J Pediatr 2022; 251:51-59.e52
  29. DeLacey S, Arzu J, Levin L, Ranganna A, Swamy A, Bianco ME. Impact of SARS-CoV2 on youth onset type 2 diabetes new diagnoses and severity. J Diabetes 2022; 14:532-540
  30. Denzer C, Rosenbauer J, Klose D, Körner A, Reinehr T, Baechle C, Schröder C, Wiegand S, Holl RW, Prinz N. Is COVID-19 to Blame? Trends of Incidence and Sex Ratio in Youth-Onset Type 2 Diabetes in Germany. Diabetes Care 2023; 46:1379-1387
  31. Chang TH, Chen YC, Chen WY, Chen CY, Hsu WY, Chou Y, Chang YH. Weight Gain Associated with COVID-19 Lockdown in Children and Adolescents: A Systematic Review and Meta-Analysis. Nutrients 2021; 13
  32. Srinivasan S, Chen L, Todd J, Divers J, Gidding S, Chernausek S, Gubitosi-Klug RA, Kelsey MM, Shah R, Black MH, Wagenknecht LE, Manning A, Flannick J, Imperatore G, Mercader JM, Dabelea D, Florez JC. The First Genome-Wide Association Study for Type 2 Diabetes in Youth: The Progress in Diabetes Genetics in Youth (ProDiGY) Consortium. Diabetes 2021; 70:996-1005
  33. Hao S, Cossen K, Westbrook AL, Umpierrez GE, Vellanki P. Diabetic Ketoacidosis and Long-term Insulin Requirements in Youths with Newly Diagnosed Type 2 Diabetes During the SARS-CoV-2 Pandemic. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 2023; 29:754-761
  34. Klingensmith GJ, Connor CG, Ruedy KJ, Beck RW, Kollman C, Haro H, Wood JR, Lee JM, Willi SM, Cengiz E, Tamborlane WV. Presentation of youth with type 2 diabetes in the Pediatric Diabetes Consortium. Pediatr Diabetes 2016; 17:266-273
  35. Klingensmith GJ, Lanzinger S, Tamborlane WV, Hofer SE, Cheng P, de Beaufort C, Gal RL, Reinehr T, Kollman C, Holl RW. Adolescent type 2 diabetes: Comparing the Pediatric Diabetes Consortium and Germany/Austria/Luxemburg Pediatric Diabetes Prospective registries. Pediatr Diabetes 2018; 19:1156-1163
  36. Otaigbe BE, Imafidon EE. Type 2 diabetes mellitus in a Nigerian child: a case report. African health sciences2011; 11:454-456
  37. Kevat D, Wilson D, Sinha A. A 5-year-old girl with type 2 diabetes. Lancet 2014; 383:1268
  38. Greenup E, Sunil B, Barr MM, Ashraf AP. Glycaemic control and outcomes in children with type 2 diabetes diagnosed at or before 10 years of age. Endocrinology, diabetes & metabolism 2021; 4:e00192
  39. Curran JA, Haynes A, Davis EA. Clinical characteristics of Western Australian children diagnosed with type 2 diabetes before 10 years of age. The Medical journal of Australia 2020; 212:95-95.e91
  40. Astudillo M, Tosur M, Castillo B, Rafaey A, Siller AF, Nieto J, Sisley S, McKay S, Nella AA, Balasubramanyam A, Bacha F, Redondo MJ. Type 2 diabetes in prepubertal children. Pediatr Diabetes 2021; 22:946-950
  41. Hutchins J, Barajas RA, Hale D, Escaname E, Lynch J. Type 2 diabetes in a 5-year-old and single center experience of type 2 diabetes in youth under 10. Pediatr Diabetes 2017; 18:674-677
  42. Amutha A, Unnikrishnan R, Anjana RM, Mohan V. Prepubertal Childhood Onset Type 2 Diabetes Mellitus: Four Case Reports. The Journal of the Association of Physicians of India 2017; 65:43-46
  43. Al Amiri E, Abdullatif M, Abdulle A, Al Bitar N, Afandi EZ, Parish M, Darwiche G. The prevalence, risk factors, and screening measure for prediabetes and diabetes among Emirati overweight/obese children and adolescents. BMC Public Health 2015; 15:1298-1298
  44. Alsaffar Y, Hussain AM, Selman NAJAAVdFyT. Prevalence of type 2 diabetes in pediatrics and adolescents newly diagnosed with diabetes in Babylon Governorate, Iraq. 2020; 39
  45. Moussa MA, Alsaeid M, Abdella N, Refai TM, Al-Sheikh N, Gomez JE. Prevalence of type 2 diabetes mellitus among Kuwaiti children and adolescents. Medical principles and practice : international journal of the Kuwait University, Health Science Centre 2008; 17:270-275
  46. Elkum N, Al-Arouj M, Sharifi M, Shaltout A, Bennakhi A. Prevalence of childhood obesity in the state of Kuwait. Pediatr Obes 2016; 11:e30-e34
  47. AlBlooshi A, Shaban S, AlTunaiji M, Fares N, AlShehhi L, AlShehhi H, AlMazrouei A, Souid AK. Increasing obesity rates in school children in United Arab Emirates. Obesity science & practice 2016; 2:196-202
  48. Pearson ER. Treating type 2 diabetes in youth: a depressing picture. The journal of the Royal College of Physicians of Edinburgh 2012; 42:228
  49. Kriska A, El Ghormli L, Copeland KC, Higgins J, Ievers-Landis CE, Levitt Katz LE, Trief PM, Wauters AD, Yasuda PM, Delahanty LM. Impact of lifestyle behavior change on glycemic control in youth with type 2 diabetes. Pediatr Diabetes 2018; 19:36-44
  50. Alfaraidi H, Samaan MC. Metformin therapy in pediatric type 2 diabetes mellitus and its comorbidities: A review. Frontiers in endocrinology 2022; 13:1072879
  51. Jones KL, Arslanian S, Peterokova VA, Park JS, Tomlinson MJ. Effect of metformin in pediatric patients with type 2 diabetes: a randomized controlled trial. Diabetes Care 2002; 25:89-94
  52. Gottschalk M, Danne T, Vlajnic A, Cara JF. Glimepiride versus metformin as monotherapy in pediatric patients with type 2 diabetes: a randomized, single-blind comparative study. Diabetes Care 2007; 30:790-794
  53. Laffel L, Chang N, Grey M, Hale D, Higgins L, Hirst K, Izquierdo R, Larkin M, Macha C, Pham T, Wauters A, Weinstock RS. Metformin monotherapy in youth with recent onset type 2 diabetes: experience from the prerandomization run-in phase of the TODAY study. Pediatr Diabetes 2012; 13:369-375
  54. Kelsey MM, Geffner ME, Guandalini C, Pyle L, Tamborlane WV, Zeitler PS, White NH. Presentation and effectiveness of early treatment of type 2 diabetes in youth: lessons from the TODAY study. Pediatr Diabetes2016; 17:212-221
  55. Filippatos TD, Panagiotopoulou TV, Elisaf MS. Adverse Effects of GLP-1 Receptor Agonists. The review of diabetic studies : RDS 2014; 11:202-230
  56. Tamborlane WV, Barrientos-Pérez M, Fainberg U, Frimer-Larsen H, Hafez M, Hale PM, Jalaludin MY, Kovarenko M, Libman I, Lynch JL, Rao P, Shehadeh N, Turan S, Weghuber D, Barrett T. Liraglutide in Children and Adolescents with Type 2 Diabetes. N Engl J Med 2019; 381:637-646
  57. Tamborlane WV, Bishai R, Geller D, Shehadeh N, Al-Abdulrazzaq D, Vazquez EM, Karoly E, Troja T, Doehring O, Carter D, Monyak J, Sjostrom CD. Once-Weekly Exenatide in Youth With Type 2 Diabetes. Diabetes Care 2022; 45:1833-1840
  58. Arslanian SA, Hannon T, Zeitler P, Chao LC, Boucher-Berry C, Barrientos-Perez M, Bismuth E, Dib S, Cho JI, Cox D, Investigators A-P. Once-Weekly Dulaglutide for the Treatment of Youths with Type 2 Diabetes. N Engl J Med 2022; 387:433-443
  59. Padda IS, Mahtani AU, Parmar M. Sodium-Glucose Transport Protein 2 (SGLT2) Inhibitors. StatPearls. Treasure Island (FL) ineligible companies. Disclosure: Arun Mahtani declares no relevant financial relationships with ineligible companies. Disclosure: Mayur Parmar declares no relevant financial relationships with ineligible companies.: StatPearls Publishing, Copyright © 2023, StatPearls Publishing LLC.; 2023.
  60. Laffel LM, Danne T, Klingensmith GJ, Tamborlane WV, Willi S, Zeitler P, Neubacher D, Marquard J. Efficacy and safety of the SGLT2 inhibitor empagliflozin versus placebo and the DPP-4 inhibitor linagliptin versus placebo in young people with type 2 diabetes (DINAMO): a multicentre, randomised, double-blind, parallel group, phase 3 trial. The lancet Diabetes & endocrinology 2023; 11:169-181
  61. Tamborlane WV, Laffel LM, Shehadeh N, Isganaitis E, Van Name M, Ratnayake J, Karlsson C, Norjavaara E. Efficacy and safety of dapagliflozin in children and young adults with type 2 diabetes: a prospective, multicentre, randomised, parallel group, phase 3 study. The lancet Diabetes & endocrinology 2022; 10:341-350
  62. Mulvihill EE, Drucker DJ. Pharmacology, physiology, and mechanisms of action of dipeptidyl peptidase-4 inhibitors. Endocrine reviews 2014; 35:992-1019
  63. Shankar RR, Zeitler P, Deeb A, Jalaludin MY, Garcia R, Newfield RS, Samoilova Y, Rosario CA, Shehadeh N, Saha CK, Zhang Y, Zilli M, Scherer LW, Lam RLH, Golm GT, Engel SS, Kaufman KD. A randomized clinical trial of the efficacy and safety of sitagliptin as initial oral therapy in youth with type 2 diabetes. Pediatr Diabetes 2022; 23:173-182
  64. Bensignor MO, Kelly AS, Arslanian S. Anti-obesity pharmacotherapy for treatment of pediatric type 2 diabetes: Review of the literature and lessons learned from adults. Frontiers in endocrinology 2022; 13:1043650
  65. Stretton B, Kovoor J, Bacchi S, Chang S, Ngoi B, Murray T, Bristow TC, Heng J, Gupta A, Ovenden C, Maddern G, Thompson CH, Heilbronn L, Boyd M, Rayner C, Talley NJ, Horowtiz M. Weight loss with subcutaneous semaglutide versus other glucagon-like peptide 1 receptor agonists in type 2 diabetes: a systematic review. Internal medicine journal 2023; 53:1311-1320
  66. Weghuber D, Barrett T, Barrientos-Pérez M, Gies I, Hesse D, Jeppesen OK, Kelly AS, Mastrandrea LD, Sørrig R, Arslanian S. Once-Weekly Semaglutide in Adolescents with Obesity. N Engl J Med 2022; 387:2245-2257
  67. Zeitler P, El Ghormli L, Arslanian S, Caprio S, Isganaitis E, Kelsey MK, Weinstock RS, White NH, Drews K. Deterioration of Glycemic Control in Youth-Onset Type 2 Diabetes: What Are the Early and Late Predictors? J Clin Endocrinol Metab 2022; 107:e3384-e3394
  68. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 14. Children and Adolescents: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S230-S253
  69. Asiri A, Alzahrani F, Alghamdi H, Alamri Z. Effect of Laparoscopic Sleeve Gastrectomy on HbA1C Level in Children with Type 2 Diabetes Mellitus. Medicina (Kaunas, Lithuania) 2022; 58
  70. Inge TH, Laffel LM, Jenkins TM, Marcus MD, Leibel NI, Brandt ML, Haymond M, Urbina EM, Dolan LM, Zeitler PS. Comparison of Surgical and Medical Therapy for Type 2 Diabetes in Severely Obese Adolescents. JAMA pediatrics 2018; 172:452-460
  71. Zhu X, Zhou G, Gu X, Jiang X, Huang H, You S, Zhang G. Comparison of bariatric surgery and medical therapy for obese adolescents with type 2 diabetes. Asian journal of surgery 2022;
  72. Bjornstad P, Hughan K, Kelsey MM, Shah AS, Lynch J, Nehus E, Mitsnefes M, Jenkins T, Xu P, Xie C, Inge T, Nadeau K. Effect of Surgical Versus Medical Therapy on Diabetic Kidney Disease Over 5 Years in Severely Obese Adolescents With Type 2 Diabetes. Diabetes Care 2020; 43:187-195
  73. Bjornstad P, Nehus E, van Raalte D. Bariatric surgery and kidney disease outcomes in severely obese youth. Seminars in pediatric surgery 2020; 29:150883
  74. Ryder JR, Xu P, Nadeau KJ, Kelsey MM, Xie C, Jenkins T, Inge TH, Bjornstad P. Effect of surgical versus medical therapy on estimated cardiovascular event risk among adolescents with type 2 diabetes and severe obesity. Surgery for obesity and related diseases : official journal of the American Society for Bariatric Surgery 2021; 17:23-33
  75. Jastreboff AM, Kaplan LM, Frías JP, Wu Q, Du Y, Gurbuz S, Coskun T, Haupt A, Milicevic Z, Hartman ML. Triple-Hormone-Receptor Agonist Retatrutide for Obesity - A Phase 2 Trial. N Engl J Med 2023; 389:514-526
  76. Medicine TSGJNEJo. Long-term complications in youth-onset type 2 diabetes. 2021; 385:416-426
  77. Rapid rise in hypertension and nephropathy in youth with type 2 diabetes: the TODAY clinical trial. Diabetes Care2013; 36:1735-1741
  78. Bjornstad P, Drews KL, Caprio S, Gubitosi-Klug R, Nathan DM, Tesfaldet B, Tryggestad J, White NH, Zeitler P. Long-Term Complications in Youth-Onset Type 2 Diabetes. N Engl J Med 2021; 385:416-426
  79. Cioana M, Deng J, Hou M, Nadarajah A, Qiu Y, Chen SSJ, Rivas A, Banfield L, Chanchlani R, Dart A, Wicklow B, Alfaraidi H, Alotaibi A, Thabane L, Samaan MC. Prevalence of Hypertension and Albuminuria in Pediatric Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e216069
  80. Chu PY, Campbell MJ, Miller SG, Hill KD. Anti-hypertensive drugs in children and adolescents. World journal of cardiology 2014; 6:234-244
  81. Siddiqi N, Shatat IF. Antihypertensive agents: a long way to safe drug prescribing in children. Pediatric nephrology (Berlin, Germany) 2020; 35:2049-2065
  82. Dabelea D, Sauder KA, Jensen ET, Mottl AK, Huang A, Pihoker C, Hamman RF, Lawrence J, Dolan LM, Agostino R, Jr., Wagenknecht L, Mayer-Davis EJ, Marcovina SM. Twenty years of pediatric diabetes surveillance: what do we know and why it matters. Ann N Y Acad Sci 2021;
  83. Amatruda M, Gembillo G, Giuffrida AE, Santoro D, Conti G. The Aggressive Diabetic Kidney Disease in Youth-Onset Type 2 Diabetes: Pathogenetic Mechanisms and Potential Therapies. Medicina (Kaunas, Lithuania) 2021; 57
  84. Naaman SC, Bakris GL. Diabetic Nephropathy: Update on Pillars of Therapy Slowing Progression. Diabetes Care 2023; 46:1574-1586
  85. Blazek O, Bakris GL. Slowing the Progression of Diabetic Kidney Disease. Cells 2023; 12
  86. Jackson S, Creo A, Kumar S. Are Clinicians Aggressive Enough in Treating Diabetes-Related Hyperlipidemia in Youth? Current atherosclerosis reports 2022; 24:471-481
  87. Cioana M, Deng J, Nadarajah A, Hou M, Qiu Y, Chen SSJ, Rivas A, Banfield L, Alfaraidi H, Alotaibi A, Thabane L, Samaan MC. Prevalence of Polycystic Ovary Syndrome in Patients With Pediatric Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Network Open 2022; 5:e2147454-e2147454
  88. Pregnancy Outcomes in Young Women With Youth-Onset Type 2 Diabetes Followed in the TODAY Study. Diabetes Care 2021; 45:1038-1045
  89. Shah RD, Chernausek SD, El Ghormli L, Geffner ME, Keady J, Kelsey MM, Farrell R, Tesfaldet B, Tryggestad JB, Van Name M, Isganaitis E. Maternal Diabetes in Youth-Onset Type 2 Diabetes Is Associated With Progressive Dysglycemia and Risk of Complications. J Clin Endocrinol Metab 2023; 108:1120-1131
  90. Merino PM, Codner E. Contraception for Adolescents and Young Women with Type 2 Diabetes-Specific Considerations. Current diabetes reports 2022; 22:77-84
  91. Cioana M, Deng J, Nadarajah A, Hou M, Qiu Y, Chen SSJ, Rivas A, Toor PP, Banfield L, Thabane L, Chaudhary V, Samaan MC. Global Prevalence of Diabetic Retinopathy in Pediatric Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Netw Open 2023; 6:e231887
  92. Mititelu M, Uschner D, Doherty L, Bjornstad P, Domalpally A, Drews KL, Gubitosi-Klug R, Levitsky LL, Pak JW, White NH, Blodi BA. Retinal Thickness and Morphology Changes on OCT in Youth with Type 2 Diabetes: Findings from the TODAY Study. Ophthalmol Sci 2022; 2:100191
  93. Wright AD, Dodson PM. Medical management of diabetic retinopathy: fenofibrate and ACCORD Eye studies. Eye (London, England) 2011; 25:843-849
  94. Dabelea D, Stafford JM, Mayer-Davis EJ, D'Agostino R, Jr., Dolan L, Imperatore G, Linder B, Lawrence JM, Marcovina SM, Mottl AK, Black MH, Pop-Busui R, Saydah S, Hamman RF, Pihoker C. Association of Type 1 Diabetes vs Type 2 Diabetes Diagnosed During Childhood and Adolescence With Complications During Teenage Years and Young Adulthood. Jama 2017; 317:825-835
  95. Dart AB, Martens PJ, Rigatto C, Brownell MD, Dean HJ, Sellers EA. Earlier Onset of Complications in Youth With Type 2 Diabetes. Diabetes Care 2014; 37:436-443
  96. Amutha A, Datta M, Unnikrishnan R, Anjana RM, Mohan V. Clinical profile and complications of childhood- and adolescent-onset type 2 diabetes seen at a diabetes center in south India. Diabetes Technol Ther 2012; 14:497-504
  97. Jaiswal M, Lauer A, Martin CL, Bell RA, Divers J, Dabelea D, Pettitt DJ, Saydah S, Pihoker C, Standiford DA, Rodriguez BL, Pop-Busui R, Feldman EL. Peripheral neuropathy in adolescents and young adults with type 1 and type 2 diabetes from the SEARCH for Diabetes in Youth follow-up cohort: a pilot study. Diabetes Care 2013; 36:3903-3908
  98. Jaiswal M, Divers J, Dabelea D, Isom S, Bell RA, Martin CL, Pettitt DJ, Saydah S, Pihoker C, Standiford DA, Dolan LM, Marcovina S, Linder B, Liese AD, Pop-Busui R, Feldman EL. Prevalence of and Risk Factors for Diabetic Peripheral Neuropathy in Youth With Type 1 and Type 2 Diabetes: SEARCH for Diabetes in Youth Study. Diabetes Care 2017; 40:1226-1232
  99. Akinci G, Savelieff MG, Gallagher G, Callaghan BC, Feldman EL. Diabetic neuropathy in children and youth: New and emerging risk factors. Pediatr Diabetes 2021; 22:132-147
  100. Vinik AI, Maser RE, Mitchell BD, Freeman R. Diabetic autonomic neuropathy. Diabetes Care 2003; 26:1553-1579
  101. Duque A, Mediano MFF, De Lorenzo A, Rodrigues LF, Jr. Cardiovascular autonomic neuropathy in diabetes: Pathophysiology, clinical assessment and implications. World J Diabetes 2021; 12:855-867
  102. Varley BJ, Gow ML, Cho YH, Benitez-Aguirre P, Cusumano J, Pryke A, Chan A, Velayutham V, Donaghue KC, Craig ME. Higher frequency of cardiovascular autonomic neuropathy in youth with type 2 compared to type 1 diabetes: Role of cardiometabolic risk factors. Pediatr Diabetes 2022; 23:1073-1079
  103. Jaiswal M, Divers J, Urbina EM, Dabelea D, Bell RA, Pettitt DJ, Imperatore G, Pihoker C, Dolan LM, Liese AD, Marcovina S, Linder B, Feldman EL, Pop-Busui R. Cardiovascular autonomic neuropathy in adolescents and young adults with type 1 and type 2 diabetes: The SEARCH for Diabetes in Youth Cohort Study. Pediatr Diabetes2018; 19:680-689
  104. Wadwa RP, Urbina EM, Anderson AM, Hamman RF, Dolan LM, Rodriguez BL, Daniels SR, Dabelea D, Group ftSS. Measures of Arterial Stiffness in Youth With Type 1 and Type 2 Diabetes: The SEARCH for Diabetes in Youth study. Diabetes Care 2010; 33:881-886
  105. Shah AS, Gidding SS, El Ghormli L, Tryggestad JB, Nadeau KJ, Bacha F, Levitt Katz LE, Willi SM, Lima J, Urbina EM. Relationship between Arterial Stiffness and Subsequent Cardiac Structure and Function in Young Adults with Youth-Onset Type 2 Diabetes: Results from the TODAY Study. Journal of the American Society of Echocardiography : official publication of the American Society of Echocardiography 2022; 35:620-628.e624
  106. Urbina EM, Isom S, Dabelea D, D'Agostino R, Daniels SR, Dolan LM, Imperatore G, Lustigova E, Marcovina S, Mottl A, Pihoker C, Shah AS. Association of Elevated Arterial Stiffness With Cardiac Target Organ Damage and Cardiac Autonomic Neuropathy in Young Adults With Diabetes: The SEARCH for Diabetes in Youth Study. Diabetes Care 2023; 46:786-793
  107. Urbina EM, Kimball TR, McCoy CE, Khoury PR, Daniels SR, Dolan LM. Youth with obesity and obesity-related type 2 diabetes mellitus demonstrate abnormalities in carotid structure and function. Circulation 2009; 119:2913-2919
  108. Shah AS, Urbina EM. Vascular and Endothelial Function in Youth with Type 2 Diabetes Mellitus. Current diabetes reports 2017; 17:36-36
  109. Lee HS, Yoon JS, Park KJ, Lim JS, Hwang JS. The Relationship Between Bone Mineral Density and Type 2 Diabetes in Obese Children and Adolescents at the Time of Initial Diagnosis. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme 2019; 51:42-46
  110. Kindler JM, Kelly A, Khoury PR, Levitt Katz LE, Urbina EM, Zemel BS. Bone Mass and Density in Youth With Type 2 Diabetes, Obesity, and Healthy Weight. Diabetes Care 2020; 43:2544-2552
  111. Monaghan M, Mara CA, Kichler JC, Westen SC, Rawlinson A, Jacobsen LM, Adams RN, Stone JY, Hood KK, Mulvaney SA. Multisite Examination of Depression Screening Scores and Correlates Among Adolescents and Young Adults With Type 2 Diabetes. Can J Diabetes 2021; 45:411-416
  112. McVoy M, Hardin H, Fulchiero E, Caforio K, Briggs F, Neudecker M, Sajatovic M. Mental health comorbidity and youth onset type 2 diabetes: A systematic review of the literature. International journal of psychiatry in medicine2023; 58:37-55
  113. Hoffman RP, Damilano CP, Hong KMC, Glick BA, Kamboj MK. Glycemic control, depression, diabetes distress among adolescents with type 2 diabetes: effects of sex, race, insurance, and obesity. Acta Diabetol 2022; 59:1083-1089
  114. Gulley LD, Shomaker LB. Depression in Youth-Onset Type 2 Diabetes. Current diabetes reports 2020; 20:51
  115. Wilfley D, Berkowitz R, Goebel-Fabbri A, Hirst K, Ievers-Landis C, Lipman TH, Marcus M, Ng D, Pham T, Saletsky R, Schanuel J, Van Buren D. Binge eating, mood, and quality of life in youth with type 2 diabetes: baseline data from the today study. Diabetes Care 2011; 34:858-860
  116. Nip ASY, Reboussin BA, Dabelea D, Bellatorre A, Mayer-Davis EJ, Kahkoska AR, Lawrence JM, Peterson CM, Dolan L, Pihoker C. Disordered Eating Behaviors in Youth and Young Adults With Type 1 or Type 2 Diabetes Receiving Insulin Therapy: The SEARCH for Diabetes in Youth Study. Diabetes Care 2019; 42:859-866
  117. Reynolds K, Saydah SH, Isom S, Divers J, Lawrence JM, Dabelea D, Mayer-Davis EJ, Imperatore G, Bell RA, Hamman RF. Mortality in youth-onset type 1 and type 2 diabetes: The SEARCH for Diabetes in Youth study. Journal of diabetes and its complications 2018; 32:545-549
  118. Rhodes ET, Prosser LA, Hoerger TJ, Lieu T, Ludwig DS, Laffel LM. Estimated morbidity and mortality in adolescents and young adults diagnosed with Type 2 diabetes mellitus. Diabetic medicine : a journal of the British Diabetic Association 2012; 29:453-463

Diabetes in People Living with HIV

ABSTRACT

 

People living with HIV (PLWH) are living longer and also have unique risk factors for developing several metabolic diseases, including diabetes. This has been observed in both high income and low to middle income countries. Risk factors for diabetes in PLWH consist of specific antiretroviral therapies (ART), including older generation protease inhibitors and nucleoside reverse transcriptase inhibitors, lipodystrophy, and hepatitis C co-infection. In addition, obese and overweight states are common in PLWH, and an increased risk of incident diabetes has been noted with weight gain after ART initiation in PLWH, compared to individuals without HIV. Inflammation associated with HIV has also been linked to incident diabetes. This chapter reviews points to consider in the diagnosis and monitoring of diabetes in PLWH and discusses interactions that may occur between specific ART agents and glucose-lowering medications. Moreover, PLWH have risk factors for complications involving organ systems that are also affected by microvascular disease in diabetes. Because PLWH have a greater risk for cardiovascular disease (CVD) than individuals without HIV, modifiable risk factors of CVD should be addressed in the care of PLWH, considering that dyslipidemia, hypertension, and cigarette smoking are all highly prevalent in PLWH.

 

BACKGROUND

           

HIV infection is prevalent in 38.4 million people worldwide as of 2021 (1) and 1.2 million people in the United States as of 2019. The incidence of HIV infection has decreased from 2015 to 2019 in the United States but has remained stable in some demographic groups, including African-Americans and Latinos (2). Untreated, HIV infection can lead to opportunistic infections including cytomegalovirus disease and pneumocystosis (3) associated with AIDS, which is defined as a CD4+ cell count < 200 cells/mL. With advances in antiretroviral therapy (ART), however, HIV infection has been transformed from a disease strongly linked to AIDS and opportunistic infections into a chronic disease that is associated with several cardiometabolic consequences, including diabetes (4), heart disease (5), and other non-AIDS related illnesses such as osteoporosis (6). In part, this phenomenon is secondary to the fact that people living with HIV (PLWH) are living longer and are more susceptible to diseases of aging.

 

An important aspect to consider in the understanding of HIV associated cardiometabolic diseases is their growing global presence. The same factors that are associated with cardiometabolic disease prevalence in PLWH in high-income countries are also present in low-income countries, in addition to unique factors including industrialization, with resultant decreased physical activity and increased energy consumption (7) (8). As such, the care of PLWH needs to focus not only on ART but also on the management of chronic co-morbidities. This chapter will focus primarily on diabetes in PLWH.

 

EPIDEMIOLOGY AND RISK FACTORS FOR DIABETES IN PLWH

 

Epidemiology           

 

PLWH have a unique set of risk factors that may increase their likelihood of developing diabetes. Illustrating the greater burden of diabetes in PLWH on ART, a study from 2005 on the Multicenter AIDS Cohort Study (MACS) of gay and bisexual men with (MWH) and without HIV (MWoH) found that the incidence of diabetes was found to be more than fourfold higher in MWH and that the prevalence of diabetes was 14% in MWH on ART and 7% in MWH not on ART, compared to 5% in MWoH. These differences were significant even after adjusting for age and body mass index (BMI). Of note, the majority of MWH on ART in the study were on first generation protease inhibitor (PI) therapy (discussed further in the section Protease Inhibitors) (9). Other studies have described incidence rates of diabetes in PLWH of 4.4 cases per 1000 person-years of follow-up in the Swiss HIV Cohort Study (10) and 5.72 cases per 1000 person-years of follow-up in the Data Collection on Adverse Events of Anti-HIV Drugs (D:A:D) Study of participants from Europe, the US, Australia, and Argentina (11).

 

The effect of HIV disease on developing diabetes is also growing in low- and middle-income countries (LMIC), where the majority of the people with diabetes live (12). Prevalence of estimates of diabetes in PLWH in LMIC range from 6.8% in Chile (13) to 26% in Cameroon (14). PLWH and diabetes in LMIC are younger than those in high income countries (15). The wide range of prevalence estimates of diabetes in PLWH in LMIC may be in part because of differences in the definitions used to identifying diabetes. In addition to factors common to PLWH globally, urbanization may be a contributing factor to the development of diabetes in PLWH in LMIC (16).

 

Antiretroviral Therapy (ART)

 

PROTEASE INHIBITORS (PI)

 

One risk factor for developing diabetes in PLWH is PI use. In 1995, saquinavir became the first FDA approved PI (17). Incidence rates of hyperglycemia as high as fivefold have been reported in the setting of PI use (18). In addition, PIs, including ritonavir, have been associated with hypertriglyceridemia (19,20) and lipodystrophy (21). Mechanisms that may account for these effects are multifold. In one study, insulin sensitivity, as measured by glucose infusion rate during hyperglycemic clamp, decreased significantly after 12 weeks of PI therapy compared to baseline. A defect in beta cell function, in particular, a decrease in the disposition index, was also observed after PI therapy (22). An in vitro study demonstrated that indinavir may lower the function of the glucose transporter GLUT4 (23). Additional mechanisms may include changes in the hormone adiponectin, which is associated with improved insulin sensitivity, although in vivo and in vitro effects of PIs on adiponectin have differed (24,25).

 

The prescribing patterns of PI use have evolved. One study found that in the Veterans Affairs system, the prevalence of PI based regimen use decreased from 1997 to         2004 (26). In addition, the effects of all PIs, namely older generation versus newer generation PIs, including atazanavir and darunavir, on glucose homeostasis are not equal. In an in vitro study, atazanavir, which received FDA approval in 2003 (27), did not inhibit either GLUT1 or GLUT4 glucose transporters (28). In a clinical study of HIV- participants, atazanavir in combination with ritonavir had a significantly lower effect on insulin sensitivity compared to lopinavir/ritonavir, as measured by glucose disposal rate during hyperinsulinemic euglycemic clamp. (29). Moreover, the newer generation PIs have not been associated with an increased incidence of diabetes, with one study showing a lower risk of diabetes in individuals on these medications (30).

 

NUCLEOSIDE REVERSE TRANSCRIPTASE INHIBITORS (NRTIs)

 

In addition to PIs, certain nucleoside reverse transcriptase inhibitors (NRTIs) have been associated with diabetes. In the D:A:D study, incident diabetes was associated with exposure to the NRTIs stavudine and zidovudine, both thymidine analogs, and didanosine, after adjusting for age, sex, race, and BMI (11). NRTIs have also been implicated in mitochondrial dysfunction, which may be another mechanism by which NRTIs are associated with diabetes. One study found that one month of treatment with stavudine was associated with significant decreases in mitochondrial DNA from muscle biopsies and insulin sensitivity, as measured by the glucose infusion rate during a hyperinsulinemic euglycemic clamp (31). Because of serious adverse effects linked to the use of these NRTIs, stavudine, zidovudine, and didanosine are not advised for use in the United States (32).

 

Similar to the newer generation of PIs, the use of newer NRTIs, including emtricitabine, abacavir, and tenofovir, have been associated with a lower risk of diabetes (30). Supporting this finding, tenofovir disoproxil fumarate (TDF), in a study of participants without HIV or diabetes, did not significantly affect insulin sensitivity, as measured by hyperinsulinemic euglycemic clamp (33).

 

Tenofovir alafenamide (TAF) is a newer prodrug of tenofovir and is associated with less renal and bone toxicity than TDF (33a). However, TAF has been associated with weight gain in PLWH, compared with TDF (33b). Weight gain leading to an overweight state or obesity is a risk factor for the development of type 2 diabetes. In a retrospective study of the ADVANCE trial, which included ART naïve PLWH randomized to 3 treatment arms, 2 of which included TDF and 1 of which included TAF, an increase in the 10-year predicted risk of type 2 diabetes was observed for participants in the TAF arm, compared to participants in one of the TDF arms, although the risk calculator that was used had not been validated for the population in the study (33c). On the other hand, in a cohort study of more than 4,000 PWH on a TDF-based ART regimen for at least 6 months, of whom about 80% switched to a TAF-based ART regimen and about 20% continued on a TDF-based ART regimen, TAF was associated with a significant weight gain, compared to TDF, after 18 months, but it was not associated with incident diabetes.

 

INTEGRASE STRAND TRANSFER INHIBITORS (INSTIs)

           

Integrase strand transfer inhibitor (INSTI) based regimens are among the recommended initial ART regimens for PLWH (32) (34). Reasons for this include the effectiveness of and fewer adverse effects associated with INSTIs in studies of treatment-naïve patients (34-37). However, studies have discovered some metabolic effects of INSTIs. These include weight gain on average of 2.9 kilograms over 18 months, as observed in a retrospective study of patients who switched to an INSTI based regimen from previously being on a non-NRTI (NNRTI)-based regimen. This was in contrast to an average 0.7 kg weight gain in those participants who switched to a PI-based regimen and 0.9 kg weight gain in those who continued on a NNRTI-based regimen (38). In addition to weight gain, case reports of new onset diabetes have been reported in conjunction with INSTI use (39,40). One clinical trial compared the INSTI raltegravir to 2 boosted PI regimens and found that the increases in homeostasis model assessment-insulin resistance (HOMA-IR) in all 3 arms were not significantly different from one another, were noted by 4 weeks, and appeared to be independent of changes in visceral adipose tissue (41). However, other studies have not demonstrated an association between diabetes and INSTI use (41a) or have found that the association between INSTI use and incident diabetes varies by individual INSTI (41b).

 

The exact mechanisms by which INSTIs may be related to weight gain and diabetes are not known. One proposed mechanism by which INSTIs may cause hyperglycemia is their effect on magnesium, which is a necessary cation for insulin action (40). In mice, INSTI use leads to an increase in fat mass and decreases brown and beige adipocyte differentiation, leading to lower energy expenditure and weight gain (41c).

 

Lipodystrophy

 

Lipodystrophy associated with ART is characterized by either lipoatrophy in the face, arms, and legs, or lipoaccumulation that can lead to gynecomastia, dorsocervical fat tissue, and increased intra-abdominal fat,  (42,43), or mixed lipodystrophy, in which lipoatrophy and lipoaccumulation occur together. As noted above, lipodystrophy has been associated with PI use (21) and with older generation NRTI use, including stavudine (44), but less commonly with the newer generation NRTI tenofovir (45). In a cross-sectional study, HIV+ men with lipodystrophy had a greater insulin resistance than men without lipodystrophy (21). Lipodystrophy has also been associated with incident diabetes (46,47).

 

In one study that measured body fat changes longitudinally using dual-energy x-ray absorptiometry (DXA) in PLWH on ART (specifically zidovudine and lamivudine or didanosine and stavudine in combination with nelfinavir, efavirenz, or both), 32% of participants had discordant changes in trunk and limb fat (48).

 

Lipodystrophy may also be persistent in patients with exposure to thymidine analogs. Although improvements in limb fat mass were reported in patients who switched from taking the thymidine analog zidovudine to the NRTI abacavir, no significant improvement was noted in self-assessment of dorsocervical fat (49).

 

Hepatitis C Co-Infection

 

25% of PLWH in the U.S. are co-infected with hepatitis C (50). Hepatitis C infection is associated with a higher risk of developing diabetes. Treatments for hepatitis C, including direct-acting antiviral treatment and pegylated interferon/ribivirin, have been found to lower the incidence of diabetes, with a larger effect in those patients with advanced fibrosis/cirrhosis. In addition among individuals who received treatment for hepatitis C, those with a sustained virologic response were less likely to develop diabetes than those without a sustained virologic response (51).

 

Weight Gain, Overweight, and Obesity

 

Weight gain that is observed after the initiation of ART in those patients with wasting associated with untreated HIV has been characterized as a “return to health” phenomenon. However, because of an emphasis on early ART initiation, the wasting that was previously seen with advanced HIV infection is less common in countries with access to ART (52). In one 2012 study conducted in Alabama, more than 40% of patients were overweight or obese at the time of starting ART. After 2 years on ART, the percentages of underweight and normal weight participants decreased significantly, and significant increases in the percentages of overweight and obese participants were observed. Having a lower baseline CD4 count and the use of a PI as a third drug were risk factors associated with a greater increase in BMI (53).

 

The consequences of weight gain in PLWH may be different from those in the general population. In a study of U.S. Veterans, the risk of incident diabetes with 10 or more pounds of weight gain during the first year after ART initiation in HIV+ Veterans was significantly greater than in HIV- Veterans (54).

 

Among cohorts of PLWH from different countries, prevalence estimates of an overweight state or obesity range from 25% to 68% (55,56). In one study of PLWH, risk factors for being overweight or obese in PLWH included increasing age and either no evidence of hepatitis C infection or evidence of cleared hepatitis C infection (57).

 

Inflammation

 

In addition to the risk factors for diabetes in PLWH outlined above, inflammation may play a role in the development of diabetes in PLWH. Systemic inflammation results from several factors: viral replication, immune activation, which leads to T cell depletion, as well as T cell loss specifically in the gastrointestinal tract, with associated translocation of microbial factors including lipopolysaccharide (58). Co-infection with viruses, including hepatitis C virus (as discussed above), hepatitis B virus, Epstein-Barr virus, and cytomegalovirus, perpetuate systemic inflammation (59).

 

Although ART reduces some inflammatory biomarkers in PLWH (60), there is evidence of residual inflammation, with levels of other inflammatory biomarkers in PLWH on ART that do not decrease to levels seen in HIV- individuals (61). Persistent inflammation measured months after ART initiation has been associated with incident diabetes in PLWH (62).

 

DIAGNOSIS OF AND MONOTORING DIABETES IN PLWH

 

The American Diabetes Association (ADA) recommends the use of a fasting plasma glucose, hemoglobin A1c, or a 2-hour plasma glucose after a 75-gram oral glucose tolerance test to establish the diagnosis of diabetes in the general population. However, there is a caveat that for certain populations of patients, including PLWH, fasting plasma glucose is preferable to hemoglobin A1c (63). Hemoglobin A1c has been found to underestimate glycemia in PLWH (64,65). One reason for this is the use of medications that cause hemolysis, including dapsone and trimethoprim-sulfamethoxazole, which are used for prophylaxis of Pneumocystis jiroveci, an opportunistic infection (65). However, another reason for the discrepancy between glucose levels and hemoglobin A1c is NRTI use. NRTIs, especially the thymidine analogs, are associated with an increased risk of macrocytosis (66), which can lower hemoglobin A1c. Finally, lower CD4 cell count (< 500 cells/mm3) was associated with a significant discordance between expected and measured hemoglobin A1c in MWoH in the MACS (67). As such, self-monitoring of blood glucose in PLWH may be preferable to using hemoglobin A1c for monitoring glycemic control (64).

 

The most recent Infectious Diseases Society of America guidelines on the primary care of PLWH recommends obtaining either a fasting plasma glucose and/or hemoglobin A1c at baseline and at 1 to 3 months after starting ART. In addition, hemoglobin A1c is preferred to fasting glucose for diagnosing diabetes because of the relative ease of obtaining a hemoglobin A1c over a fasting glucose. However, a hemoglobin A1c level ≥ 5.8% can be considered to diagnose diabetes in PLWH, instead of the ADA recommendation of ≥ 6.5%, as this improves the sensitivity of the test (68,69). Moreover, a hemoglobin A1c is recommended to be obtained every 6 months in PLWH and diabetes, with a goal hemoglobin A1c of < 7% (68).

 

TREATMENT OF DIABETES IN PLWH

           

Special considerations should be taken into account in the treatment of diabetes in PLWH. PLWH may not respond to treatment for diabetes in the same manner as HIV- individuals. Part of this observation may be because treatment responses were measured using hemoglobin A1c, which may be an inaccurate indicator of glycemia in PLWH (70), as noted above.

Interactions between several diabetes medications and ART are known. In addition, some diabetes medications may present advantages or disadvantages in PLWH. These conditions are described in further detail below and are organized by diabetes medication.

 

Lifestyle Interventions

 

Both the 2023 ADA Standards of Care on Pharmacologic Approaches to Glycemic Treatment and the 2023 updated American Association of Clinical Endocrinology algorithm of the management of type 2 diabetes highlight lifestyle interventions as part of the recommended treatment of type 2 diabetes (70a, 70b). Lifestyle interventions include not only a healthy diet and exercise but also smoking cessation and moderating alcohol intake (70b).

 

Physical activity has benefits for PLWH, including decreasing waist circumference (70c) and improvements in glucose and high-density lipoprotein cholesterol levels (70d). However, low physical activity has been reported in some studies of PLWH (70e, 70f).

 

Metformin

 

The 2023 ADA Standards of Care on Pharmacologic Approaches to Glycemic Treatment includes metformin as one first-line treatment to consider in patients in the general population with type 2 diabetes (70a) (71).

 

In PLWH with lipodystrophy and insulin resistance but without diabetes, 3 months of metformin 1000 mg twice daily was found to significantly decrease insulin area under the curve after an oral glucose tolerance test. Although no increased incidence in lactic acidosis was noted in the participants who received metformin, the study was not powered for this outcome, and liver and kidney dysfunction were exclusion criteria (72).

 

The commonly used integrase transfer strand inhibitor, dolutegravir, increases plasma levels of metformin, and thus adjusting the dose of metformin upon dolutegravir initiation has been recommended (73,74). However, one retrospective study found that there was no significant difference in glycemic control before and after starting dolutegravir in PLWH and diabetes on metformin (75).

 

In summary, there are no guidelines to suggest that metformin should not be a possible first-line treatment of type 2 diabetes in PLWH, after consideration of liver and kidney function and dolutegravir treatment.

 

Sulfonylureas

 

Sulfonylureas are a substrate of the CYP2C9 enzyme. The PIs ritonavir and nelfinavir are CYP2C9 inducers and can decrease sulfonylurea levels (76,77). In comparison with initial use of metformin in PLWH, no significant difference in glycemia after one year of therapy was noted with initial use of a sulfonylurea (78).

 

The 2023 ADA guidelines note that sulfonylureas are a high efficacy drug class for glucose management and are less cost-prohibitive compared to many other drug classes (70a) (71). However, adverse effects of sulfonylureas include hypoglycemia and weight gain. In PLWH, consideration should be made if a patient is on ritonavir or nelfinavir and the possible loss of efficacy of sulfonylurea treatment.

 

Thiazolidinediones

 

The effect of thiazolidinediones on glycemic control in PLWH with diabetes specifically has not been studied, although some trials found that rosiglitazone lowered serum insulin levels in PLWH without diabetes (79) and improved insulin sensitivity in PLWH with hyperinsulinemia (80). Several studies have focused on the effect of thiazolidinediones (TZDs) on body fat in PLWH with lipodystrophy, with equivocal findings. One 24-week study on the effect of rosiglitazone on PLWH, all of whom were on a PI, found no significant difference in arm fat between the treatment and placebo groups (81). However, the study did not have enough power to detect a difference in this outcome (82). On the other hand, other studies demonstrated that rosiglitazone increased visceral and subcutaneous abdominal fat in MWH with lipodystrophy over 6 months (83) and subcutaneous leg fat in PLWH with lipodystrophy over 3 months (80).

 

Adverse side effects of TZDs should be considered and discussed prior to initiation. These include fluid retention, osteoporosis, edema, and potential liver injury (84).

 

Dipeptidyl Peptidase 4 Inhibitors

 

Some clinical studies have demonstrated that dipeptidyl peptidase 4 (DPP4) inhibitors exert an anti-inflammatory effect in patients with type 2 diabetes (85,86). This idea was further examined in PLWH in a pilot study of 20 PLWH on ART randomized to either the DPP4 inhibitor sitagliptin or placebo for 24 weeks. A significant decrease was noted in the chemokine SDF-1α in the treatment group. In addition, an improvement in glucose tolerance was noted at week 8 in the treatment group, but the difference in glucose tolerance between the two groups was no longer significant at the end of the study (87). In another study of PLWH with impaired glucose tolerance, sitagliptin resulted in a significant improvement in glucose tolerance and decreases in the inflammatory markers hsCRP and CXCL10 from baseline after 8 weeks of treatment, compared to placebo (88). Similarly, in a larger study of 84 PLWH on ART with viral suppression and without diabetes who were randomized to 16 weeks of sitagliptin versus placebo, a significant decrease from baseline was seen at week 15 in CXCL10 in participants in the treatment group (89).

 

Glucagon-Like Peptide-1 Receptor Agonists

           

Several glucagon-like peptide-1 (GLP-1) receptor agonists have been shown to improve weight and cardiovascular outcomes, albeit in patient populations not specific to PLWH (90,91). Liraglutide, dulaglutide, and semaglutide have been shown to lower the risk of a composite outcome of nonfatal stroke, nonfatal myocardial infarction, and cardiovascular death in patients with type 2 diabetes (90,91). Liraglutide, semaglutide, and dulaglutide have indications from the Food and Drug Administration for lowering cardiovascular event risk in patients with cardiovascular disease and type 2 diabetes, and dulaglutide is also approved for lowering cardiovascular event risk in patients with multiple cardiovascular risk factors and type 2 diabetes. The 2023 ADA guidelines recommend the use of a GLP-1 receptor agonist as a first-line treatment in patients with cardiovascular disease and type 2 diabetes (70a).

 

One case report described a single PLWH who was able to discontinue insulin treatment (insulin glargine 60 units daily) and who experienced improvements in weight and hemoglobin A1c after starting liraglutide therapy (92). Another case report described improvements in hemoglobin A1c and fasting glucose on dulaglutide in a PLWH. Relatively scant literature exists on the effects of GLP-1 receptors agonists in PLWH and diabetes (93,94).

 

Sodium-Glucose Co-Transporter-2 Inhibitors

           

Similar to the GLP-1 receptor agonists, sodium-glucose co-transporter-2 (SGLT2) inhibitors have demonstrated reductions in adverse cardiovascular outcomes, including cardiovascular death and hospitalization for heart failure in general population studies (95,96). In addition, both dapagliflozin and canagliflozin have an FDA indication to reduce the risk of end-stage renal disease in patients with type 2 diabetes and diabetic nephropathy with albuminuria (96a). As such, the 2023 ADA guidelines on pharmacologic treatment of diabetes recommend that in those patients with type 2 diabetes and chronic kidney disease, a SGLT2 inhibitor that has been shown to reduce risk of renal disease progression should be used (70a).

 

SGLT2 inhibitors are also approved for reduction of adverse cardiovascular events and heart failure hospitalization in people with diabetes and have cardiovascular benefits for patients with either systolic or diastolic heart failure (96a). Diastolic dysfunction is more common in PLWH compared with people without HIV. As such, the 2023 ADA guidelines recommend the use of an SGLT2 inhibitor with demonstrated benefit in patients with type 2 diabetes and heart failure (70a).

 

One small trial studied canagliflozin for 24 weeks in 8 obese PLWH with type 2 diabetes and hemoglobin A1c > 7% and observed improvements in weight and hemoglobin A1c at the end of the study compared to baseline (97).

 

Adverse side effects of SGLT2 inhibitors include genital mycotic and urinary tract infections. In addition, 55 post-marketing cases of Fournier’s gangrene have been reported between March 2013 to January 2019 (98). A risk factor for Fournier’s gangrene is HIV infection (99-101), especially a CD4 cell count < 200 cells/μL (102).

 

Insulin

 

The 2023 ADA guidelines recommend considering insulin initiation in the patient 1) with a hemoglobin A1c > 11%, 2) with signs and symptoms of catabolism, 3) who has a presentation concerning for type 1 diabetes, and/or 4) whose hemoglobin A1c is not at goal despite taking 2 to 3 medications for diabetes, in addition to a GLP-1 receptor agonist.

 

As with all patients with diabetes, side effects of insulin therapy to consider in a discussion with a patient with HIV disease and diabetes include the risks of hypoglycemia and weight gain.

 

Sequence of Initiating Diabetes Medications in PLWH

 

There are no specific guidelines for the treatment of diabetes in PLWH. The 2023 ADA guidelines recommend the treatment of type 2 diabetes be tailored to the individual patient and consider factors including cost of medications, weight loss goals, and comorbidities including chronic kidney disease, atherosclerotic cardiovascular disease, and heart failure. Healthy lifestyle interventions should be incorporated alongside the pharmacologic treatment of type 2 diabetes.

 

For those patients with type 2 diabetes and ASCVD or who are at high risk for ASCVD, treatment with either a GLP-1 receptor agonist or SGLT2 inhibitor is recommended. If A1c is not at goal, then adding an additional agent, 1) such as a SGLT2 inhibitor, if the patient is already on a GLP-1 receptor agonist, or a GLP-1 receptor agonist, if the patient is already on a SGLT2 inhibitor, or 2) a thiazolidinedione, is recommended (70a). For patients with type 2 diabetes and heart failure, a SGLT2 inhibitor is recommended, as noted above. For patients with type 2 diabetes and chronic kidney disease, a SGLT2 inhibitor, or if contraindicated, a GLP-1 receptor agonist is recommended. Finally, for those patients with type 2 diabetes for whom weight management is a priority, a medication with weight loss or weight neutral effects can be considered. With regards to weight loss, semaglutide and tirzepatide have the most effect, whereas metformin and DPP4 inhibitors are weight neutral. Dulaglutide and liraglutide have a high weight loss efficacy, and SGLT2 inhibitors have intermediate weight loss efficacy (70a).

In PLWH, an additional factor to consider is the possible interaction of a diabetes medication with specific ART.

 

MICROVASCULAR COMPLICATIONS ASSOCIATED WITH DIABETES IN PLWH

 

Peripheral Neuropathy

 

HIV is associated with multiple peripheral neuropathies (PNs), including a distal, symmetric polyneuropathy (DSPN) (103). The prevalence of HIV-associated PN among PLWH varies < 10% to upwards of 50%, with the wide variability in prevalence estimates secondary in part to differences in methods used to assess PN (104). Risk factors for HIV-associated DSPN include use of the NRTIs zalcitabine, didanosine, and stavudine (105). In addition, HIV-associated DSPN has been observed in PLWH within 1 year of HIV transmission and is significantly associated with evidence of immune activation present in the central nervous system (106).

 

HIV-associated PN, in addition to the risk of developing PN secondary to diabetes, may place PLWH and diabetes at increased risk of sequelae including falls, particularly in those PLWH with a detectable viral load (107), foot ulceration, and amputations.

 

In summary, in addition to considering the risk of diabetic peripheral neuropathy, the risk of HIV-associated PNs should be kept in mind in the assessment and treatment of PLWH with diabetes.

 

Nephropathy

 

The prevalence estimates of chronic kidney disease (CKD) in PLWH vary depending on geographic region, with 7.9% of PLWH affected in Africa (with the highest prevalence among African regions in West Africa at 22%), compared to 3.7% in Europe. In addition, prevalence estimates of CKD in PLWH are significantly greater among those individuals with co-morbid diabetes (108). In the MACS, two independent risk factors associated with greater odds of proteinuria were HIV+ serostatus with ART use, compared to HIV- serostatus, and a history of diabetes (109). Moreover, glomerular hyperfiltration, or a supranormal estimated glomerular filtration rate, is more prevalent among HIV+ men without CKD than HIV- men without CKD (110). Glomerular hyperfiltration has been reported to be an initial state of dysfunction seen in patients with diabetic kidney disease and proteinuria (111).

 

Other risk factors for CKD in PLWH include the following: recurrent acute kidney injury, African-American race, in part because of risk variants of the APOL1 gene, and persistent inflammation, even in the setting of ART (112-114). In addition, an HIV-associated nephropathy (HIVAN), which is characterized by several insults to the kidney, including focal and segmental glomerulosclerosis, exists. Certain ART drugs within the NRTI, NNRTI, and PI classes are also associated with renal injury (114). Among these are the NRTI tenofovir, which is associated with adverse kidney disease outcomes independent of diabetes (115), and the PI indinavir (116).

           

Retinopathy

 

Ocular opportunistic infections in PLWH include CMV retinitis and ischemic HIV retinopathy (117). In addition, patients with a history of CMV retinitis should be monitored periodically for retinitis recurrence by an ophthalmologist (118). However, these ocular opportunistic infections are less common in the setting of ART. However, retinal disease has been noted in PLWH on ART as part of a larger syndrome of the HIV-associated neuroretinal disorder (117), which has an incidence of more than 50% at 20 years after a diagnosis of AIDS (119). One study found that among HIV+ men with a median duration of ART use of 12 years and suppressed viremia did have a significant difference in total peripheral retinal thickness from HIV- men, although the long-term clinical relevance of this is unknown (117).

 

There is limited literature on the effect of co-morbid diabetes on HIV-associated retinal disease.

 

MODIFIABLE FACTORS OF MACROVASCULAR COMPLICATIONS ASSOCIATED WITH DIABETES IN PLWH

 

PLWH are at increased risk of developing atherosclerotic cardiovascular disease (ASCVD) compared to individuals without HIV, despite controlling for traditional cardiovascular (CV) risk factors such as diabetes (5,120). In addition, PLWH have a greater burden of traditional CV risk factors (120). Other non-traditional risk factors include some ART such as older generation PIs (121) and inflammation (122). As such, calculators developed for the general population to calculate ASCVD risk may not accurately capture risk in PLWH (123).

 

Aspirin

 

Recent American College of Cardiology/American Heart Association (ACC/AHA) recommend the use of aspirin for primary prevention of ASCVD in general population patients age 40 to 70 years with high ASCVD risk (124), and similarly the American Diabetes Association (ADA) guidelines also recommend aspirin use in select patients with diabetes < 70 years of age with high ASCVD risk and low bleeding risk (125). However, evidence has shown that aspirin use is lower in PLWH with CV risk factors than in HIV- individuals (126), and PLWH are less likely to be prescribed aspirin for primary prevention than HIV- individuals (127).

 

Blood Pressure

 

The prevalence of hypertension globally in PLWH is substantial. PLWH have several risk factors for developing hypertension, including a greater prevalence of smoking (as noted below) (128,129) and ART use (130). The relationship of ART to hypertension is thought to be in part from its association to weight gain. Moreover, in the MACS, ART use was associated with greater systolic hypertension but not diastolic hypertension. A mechanism for this could be a change in arterial compliance as a consequence of ART use (131).

 

The 2023 ADA guidelines recommend a goal blood pressure of < 130/80 (132).

 

Cholesterol

 

Dyslipidemia is seen in both untreated PLWH and PLWH on ART (133,134). The decision to initiate a statin in a patient living with HIV for primary prevention should take into account the patient’s HIV serostatus, especially in those patients with a 10-year ASCVD risk of 5 to < 20% (124).

 

The primary prevention of atherosclerotic cardiovascular disease is a clinically important topic. The Evaluating the Use of Pitavastatin to Reduce Cardiovascular Disease in HIV-Infected Adults (REPRIEVE) study is a multicenter, international, randomized clinical trial on PWH on ART, in which participants with a low to moderate risk of ASCVD were randomized to either pitavastatin daily or placebo (134a). In March 2023, the Data Safety and Monitoring Board recommended premature closure of the study based on the observed efficacy of the study treatment to reduce the primary endpoint of major adverse cardiovascular events (MACE) by 35% relative to the placebo. The incidence of a major adverse cardiovascular event was 4.81 per 1000 person-years in the pitavastatin group and 7.32 per 1000 person-years in the placebo group (hazard ratio, 0.65; 95% confidence interval, 0.48 to 0.90; P = 0.002). Of note, similar to other studies the risk of diabetes was increased in the pitavastatin group (diabetes mellitus occurred in 5.3% in pitavastatin group and 4.0% in the control group. These results suggest that many PLWH should be on statin therapy.

 

Cigarette Smoking

 

Cigarette smoking is more prevalent in PLWH than in individuals without HIV (129), and life expectancy is estimated to be lower in HIV+ men and women who are current smokers at the time of HIV care initiation than in HIV+ men and women who are former or never smokers, with a 2.9 year gain in life expectancy at 10 years after HIV care initiation (135). The ADA guidelines recommend that people with diabetes not smoke cigarettes and that smoking cessation treatment be offered for those patients with diabetes who do smoke (136).

 

SUMMARY

 

In summary, PLWH have unique risk factors that increase their risk of diabetes. Factors to take into consideration in the treatment of PLWH include the following: type of ART, evidence of lipodystrophy, co-infection with other viruses, and overweight or obese state. A caveat is that HbA1c may be inaccurate in diagnosing and monitoring diabetes in PLWH. PLWH have additional risk factors for developing microvascular complications of diabetes. Some glycemic agents may interact with ART, and other glycemic agents may have unwanted effects, including weight gain, that should be addressed in a patient-provider discussion. Finally, additional modifiable cardiovascular risk factors, including hypertension and smoking, should be addressed in the comprehensive treatment of diabetes in PLWH.

           

REFERENCES

 

  1. World Health Organization. https://www.who.int/hiv/data/en/. Accessed July 3, 2019.
  2. Centers for Disease Control and Prevention. https://www.cdc.gov/nchhstp/newsroom/docs/factsheets/HIV-Incidence-Fact-Sheet_508.pdf. Accessed July 3, 2019.
  3. Chu C, Selwyn PA. An epidemic in evolution: the need for new models of HIV care in the chronic disease era. J Urban Health. 2011;88(3):556-566.
  4. Duncan AD, Goff LM, Peters BS. Type 2 diabetes prevalence and its risk factors in HIV: A cross-sectional study. PLoS One. 2018;13(3):e0194199.
  5. Freiberg MS, Chang CC, Kuller LH, Skanderson M, Lowy E, Kraemer KL, et al. HIV infection and the risk of acute myocardial infarction. JAMA Intern Med. 2013;173(8):614-622.
  6. Mallon PW. Aging with HIV: osteoporosis and fractures. Curr Opin HIV AIDS. 2014;9(4):428-435.
  7. Mohan V DM. The metabolic syndrome in developing countries. Diabetes Voice. 2006;51:15-17.
  8. Naidu S, Ponnampalvanar S, Kamaruzzaman SB, Kamarulzaman A. Prevalence of Metabolic Syndrome Among People Living with HIV in Developing Countries: A Systematic Review. AIDS Patient Care STDS. 2017;31(1):1-13.
  9. Brown TT, Cole SR, Li X, Kingsley LA, Palella FJ, Riddler SA, et al. Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study. Arch Intern Med. 2005;165(10):1179-1184.
  10. Ledergerber B, Furrer H, Rickenbach M, Lehmann R, Elzi L, Hirschel B, et al. Factors associated with the incidence of type 2 diabetes mellitus in HIV-infected participants in the Swiss HIV Cohort Study. Clin Infect Dis. 2007;45(1):111-119.
  11. De Wit S, Sabin CA, Weber R, Worm SW, Reiss P, Cazanave C, et al. Incidence and risk factors for new-onset diabetes in HIV-infected patients: the Data Collection on Adverse Events of Anti-HIV Drugs (D:A:D) study. Diabetes Care. 2008;31(6):1224-1229.
  12. Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW, et al. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:271-281.
  13. Cahn P, Leite O, Rosales A, Cabello R, Alvarez CA, Seas C, et al. Metabolic profile and cardiovascular risk factors among Latin American HIV-infected patients receiving HAART. Braz J Infect Dis. 2010;14(2):158-166.
  14. Ngatchou W, Lemogoum D, Ndobo P, Yagnigni E, Tiogou E, Nga E, et al. Increased burden and severity of metabolic syndrome and arterial stiffness in treatment-naive HIV+ patients from Cameroon. Vasc Health Risk Manag. 2013;9:509-516.
  15. Ali MK, Magee MJ, Dave JA, Ofotokun I, Tungsiripat M, Jones TK, et al. HIV and metabolic, body, and bone disorders: what we know from low- and middle-income countries. J Acquir Immune Defic Syndr. 2014;67 Suppl 1:S27-39.
  16. Patel P, Rose CE, Collins PY, Nuche-Berenguer B, Sahasrabuddhe VV, Peprah E, et al. Noncommunicable diseases among HIV-infected persons in low-income and middle-income countries: a systematic review and meta-analysis. AIDS. 2018;32 Suppl 1:S5-S20.
  17. HIV/AIDS Historical Time Line 1995-1999. https://www.fda.gov/patients/hiv-timeline-and-history-approvals/hivaids-historical-time-line-1995-1999. Accessed July 5, 2019.
  18. Tsiodras S, Mantzoros C, Hammer S, Samore M. Effects of protease inhibitors on hyperglycemia, hyperlipidemia, and lipodystrophy: a 5-year cohort study. Arch Intern Med. 2000;160(13):2050-2056.
  19. Markowitz M, Saag M, Powderly WG, Hurley AM, Hsu A, Valdes JM, et al. A preliminary study of ritonavir, an inhibitor of HIV-1 protease, to treat HIV-1 infection. N Engl J Med. 1995;333(23):1534-1539.
  20. Cameron DW, Heath-Chiozzi M, Danner S, Cohen C, Kravcik S, Maurath C, et al. Randomised placebo-controlled trial of ritonavir in advanced HIV-1 disease. The Advanced HIV Disease Ritonavir Study Group. Lancet. 1998;351(9102):543-549.
  21. Carr A, Samaras K, Burton S, Law M, Freund J, Chisholm DJ, et al. A syndrome of peripheral lipodystrophy, hyperlipidaemia and insulin resistance in patients receiving HIV protease inhibitors. AIDS. 1998;12(7):F51-58.
  22. Woerle HJ, Mariuz PR, Meyer C, Reichman RC, Popa EM, Dostou JM, et al. Mechanisms for the deterioration in glucose tolerance associated with HIV protease inhibitor regimens. Diabetes. 2003;52(4):918-925.
  23. Hertel J, Struthers H, Horj CB, Hruz PW. A structural basis for the acute effects of HIV protease inhibitors on GLUT4 intrinsic activity. J Biol Chem. 2004;279(53):55147-55152.
  24. Lee GA, Mafong DD, Noor MA, Lo JC, Mulligan K, Schwarz JM, et al. HIV protease inhibitors increase adiponectin levels in HIV-negative men. J Acquir Immune Defic Syndr. 2004;36(1):645-647.
  25. Xu A, Yin S, Wong L, Chan KW, Lam KS. Adiponectin ameliorates dyslipidemia induced by the human immunodeficiency virus protease inhibitor ritonavir in mice. Endocrinology. 2004;145(2):487-494.
  26. Holodniy M, Hornberger J, Rapoport D, Robertus K, MaCurdy TE, Lopez J, et al. Relationship between antiretroviral prescribing patterns and treatment guidelines in treatment-naive HIV-1-infected US veterans (1992-2004). J Acquir Immune Defic Syndr. 2007;44(1):20-29.
  27. Research CfDEa. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2003/21-567_Reyataz_Approv.pdf. Accessed July 5, 2019.
  28. Hresko RC, Hruz PW. HIV protease inhibitors act as competitive inhibitors of the cytoplasmic glucose binding site of GLUTs with differing affinities for GLUT1 and GLUT4. PLoS One. 2011;6(9):e25237.
  29. Noor MA, Flint OP, Maa JF, Parker RA. Effects of atazanavir/ritonavir and lopinavir/ritonavir on glucose uptake and insulin sensitivity: demonstrable differences in vitro and clinically. AIDS. 2006;20(14):1813-1821.
  30. Spagnuolo V, Galli L, Poli A, Salpietro S, Gianotti N, Piatti P, et al. Associations of statins and antiretroviral drugs with the onset of type 2 diabetes among HIV-1-infected patients. BMC Infect Dis. 2017;17(1):43.
  31. Fleischman A, Johnsen S, Systrom DM, Hrovat M, Farrar CT, Frontera W, et al. Effects of a nucleoside reverse transcriptase inhibitor, stavudine, on glucose disposal and mitochondrial function in muscle of healthy adults. Am J Physiol Endocrinol Metab. 2007;292(6):E1666-1673.
  32. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV. https://aidsinfo.nih.gov/guidelines/html/1/adult-and-adolescent-arv-guidelines/11/what-to-start. Accessed July 5, 2019.
  33. Randell PA, Jackson AG, Zhong L, Yale K, Moyle GJ. The effect of tenofovir disoproxil fumarate on whole-body insulin sensitivity, lipids and adipokines in healthy volunteers. Antivir Ther. 2010;15(2):227-233.

33A.  Wang H, Lu X, Yang X, Xu N. The efficacy and safety of tenofovir alafenamide versus tenofovir disoproxil fumarate in antiretroviral regimens for HIV-1 therapy: Meta-analysis. Medicine (Baltimore). 2016 Oct;95(41):e5146.

33B.  Sax, P. E. , Erlandson, K. M. , Lake, J. E. , Mccomsey, G. A. , Orkin, C. , Esser, S. , Brown, T. T. , Rockstroh, J. K. , Wei, X. , Carter, C. C. , Zhong, L. , Brainard, D. M. , Melbourne, K. , Das, M. , Stellbrink, H. , Post, F. A. , Waters, L. & Koethe, J. R. 2020. Weight Gain Following Initiation of Antiretroviral Therapy. Clinical Infectious Diseases, 71 (6), 1379-1389.

33C.  McCann K, Shah S, Hindley L, Hill A, Qavi A, Simmons B, Serenata C, Sokhela S, Venter WDF. Implications of weight gain with newer anti-retrovirals: 10-year predictions of cardiovascular disease and diabetes. AIDS. 2021 Aug 1;35(10):1657-1665.

  1. Hulgan T. Factors Associated With Insulin Resistance in Adults With HIV Receiving Contemporary Antiretroviral Therapy: a Brief Update. Curr HIV/AIDS Rep. 2018;15(3):223-232.
  2. Lennox JL, DeJesus E, Lazzarin A, Pollard RB, Madruga JV, Berger DS, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet. 2009;374(9692):796-806.
  3. DeJesus E, Rockstroh JK, Henry K, Molina JM, Gathe J, Ramanathan S, et al. Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate versus ritonavir-boosted atazanavir plus co-formulated emtricitabine and tenofovir disoproxil fumarate for initial treatment of HIV-1 infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet. 2012;379(9835):2429-2438.
  4. Raffi F, Rachlis A, Stellbrink HJ, Hardy WD, Torti C, Orkin C, et al. Once-daily dolutegravir versus raltegravir in antiretroviral-naive adults with HIV-1 infection: 48 week results from the randomised, double-blind, non-inferiority SPRING-2 study. Lancet. 2013;381(9868):735-743.
  5. Norwood J, Turner M, Bofill C, Rebeiro P, Shepherd B, Bebawy S, et al. Brief Report: Weight Gain in Persons With HIV Switched From Efavirenz-Based to Integrase Strand Transfer Inhibitor-Based Regimens. J Acquir Immune Defic Syndr. 2017;76(5):527-531.
  6. McLaughlin M, Walsh S, Galvin S. Dolutegravir-induced hyperglycaemia in a patient living with HIV. J Antimicrob Chemother. 2018;73(1):258-260.
  7. Fong PS, Flynn DM, Evans CD, Korthuis PT. Integrase strand transfer inhibitor-associated diabetes mellitus: A case report. Int J STD AIDS. 2017;28(6):626-628.
  8. Dirajlal-Fargo S, Moser C, Brown TT, Kelesidis T, Dube MP, Stein JH, et al. Changes in Insulin Resistance After Initiation of Raltegravir or Protease Inhibitors With Tenofovir-Emtricitabine: AIDS Clinical Trials Group A5260s. Open Forum Infect Dis. 2016;3(3):ofw174.

41A.  Calmy A, Tovar Sanchez T, Kouanfack C, Mpoudi-Etame M, Leroy S, Perrineau S, Lantche Wandji M, Tetsa Tata D, Omgba Bassega P, Abong Bwenda T, Varloteaux M, Tongo M, Mpoudi-Ngolé E, Montoyo A, Mercier N, LeMoing V, Peeters M, Reynes J, Delaporte E; Dolutegravir-based and low-dose efavirenz-based regimen for the initial treatment of HIV-1 infection (NAMSAL): week 96 results from a two-group, multicentre, randomised, open label, phase 3 non-inferiority trial in Cameroon. New Antiretroviral and Monitoring Strategies in HIV-infected Adults in Low-Income Countries (NAMSAL) ANRS 12313 Study Group. Lancet HIV. 2020 Oct;7(10):e677-e687.

41B.  Rebeiro PF, Jenkins CA, Bian A, Lake JE, Bourgi K, Moore RD, Horberg MA, Matthews WC, Silverberg MJ, Thorne J, Mayor AM, Lima VD, Palella FJ, Saag MS, Althoff KN, Gill MJ, Wong C, Klein MB, Crane HM, Marconi VC, Shepherd BE, Sterling TR, Koethe JR. Risk of Incident Diabetes Mellitus, Weight Gain, and Their Relationships With Integrase Inhibitor-Based Initial Antiretroviral Therapy Among Persons With Human Immunodeficiency Virus in the United States and Canada. Clin Infect Dis. 2021 Oct 5;73(7):e2234-e2242.

41C.  Jung I, Tu-Sekine B, Jin S, Anokye-Danso F, Ahima RS, Brown TT, Kim SF. Dolutegravir Suppresses Thermogenesis via Disrupting Uncoupling Protein 1 Expression and Mitochondrial Function in Brown/Beige Adipocytes in Preclinical Models. J Infect Dis. 2022 Nov 1;226(9):1626-1636.

  1. Baril JG, Junod P, Leblanc R, Dion H, Therrien R, Laplante F, et al. HIV-associated lipodystrophy syndrome: A review of clinical aspects. Can J Infect Dis Med Microbiol. 2005;16(4):233-243.
  2. Carr A. HIV protease inhibitor-related lipodystrophy syndrome. Clin Infect Dis. 2000;30 Suppl 2:S135-142.
  3. Mallal SA, John M, Moore CB, James IR, McKinnon EJ. Contribution of nucleoside analogue reverse transcriptase inhibitors to subcutaneous fat wasting in patients with HIV infection. AIDS. 2000;14(10):1309-1316.
  4. Gallant JE, Staszewski S, Pozniak AL, DeJesus E, Suleiman JM, Miller MD, et al. Efficacy and safety of tenofovir DF vs stavudine in combination therapy in antiretroviral-naive patients: a 3-year randomized trial. JAMA. 2004;292(2):191-201.
  5. Nansseu JR, Bigna JJ, Kaze AD, Noubiap JJ. Incidence and Risk Factors for Prediabetes and Diabetes Mellitus Among HIV-infected Adults on Antiretroviral Therapy: A Systematic Review and Meta-analysis. Epidemiology. 2018;29(3):431-441.
  6. Putcharoen O, Wattanachanya L, Sophonphan J, Siwamogsatham S, Sapsirisavat V, Gatechompol S, et al. New-onset diabetes in HIV-treated adults: predictors, long-term renal and cardiovascular outcomes. AIDS. 2017;31(11):1535-1543.
  7. Mulligan K, Parker RA, Komarow L, Grinspoon SK, Tebas P, Robbins GK, et al. Mixed patterns of changes in central and peripheral fat following initiation of antiretroviral therapy in a randomized trial. J Acquir Immune Defic Syndr. 2006;41(5):590-597.
  8. Martin A, Smith DE, Carr A, Ringland C, Amin J, Emery S, et al. Reversibility of lipoatrophy in HIV-infected patients 2 years after switching from a thymidine analogue to abacavir: the MITOX Extension Study. AIDS. 2004;18(7):1029-1036.
  9. Prevention CfDCa. Coinfection with HIV and Viral Hepatitis. https://www.cdc.gov/hepatitis/hiv-hepatitis-coinfection.htm. Accessed July 12, 2019.
  10. Butt AA, Aslam S, Yan P, Shaikh OS, Abou-Samra A-B. Incident Diabetes and Glucose Control after HCV Treatment with DAAs in Erchives. Paper presented at: Conference on Retroviruses and Opportunistic Infections 2019; Seattle, Washington.
  11. Kumar S, Samaras K. The Impact of Weight Gain During HIV Treatment on Risk of Pre-diabetes, Diabetes Mellitus, Cardiovascular Disease, and Mortality. Front Endocrinol (Lausanne). 2018;9:705.
  12. Tate T, Willig AL, Willig JH, Raper JL, Moneyham L, Kempf MC, et al. HIV infection and obesity: where did all the wasting go? Antivir Ther. 2012;17(7):1281-1289.
  13. Herrin M, Tate JP, Akgun KM, Butt AA, Crothers K, Freiberg MS, et al. Weight Gain and Incident Diabetes Among HIV-Infected Veterans Initiating Antiretroviral Therapy Compared With Uninfected Individuals. J Acquir Immune Defic Syndr. 2016;73(2):228-236.
  14. Levy ME, Greenberg AE, Hart R, Powers Happ L, Hadigan C, Castel A, et al. High burden of metabolic comorbidities in a citywide cohort of HIV outpatients: evolving health care needs of people aging with HIV in Washington, DC. HIV Med. 2017;18(10):724-735.
  15. Semu H, Zack RM, Liu E, Hertzmark E, Spiegelman D, Sztam K, et al. Prevalence and Risk Factors for Overweight and Obesity among HIV-Infected Adults in Dar es Salaam, Tanzania. J Int Assoc Provid AIDS Care. 2016;15(6):512-521.
  16. Obry-Roguet V, Bregigeon S, Cano CE, Lions C, Zaegel-Faucher O, Laroche H, et al. Risk factors associated with overweight and obesity in HIV-infected people: Aging, behavioral factors but not cART in a cross-sectional study. Medicine (Baltimore). 2018;97(23):e10956.
  17. Douek DC, Roederer M, Koup RA. Emerging concepts in the immunopathogenesis of AIDS. Annu Rev Med. 2009;60:471-484.
  18. Zicari S, Sessa L, Cotugno N, Ruggiero A, Morrocchi E, Concato C, et al. Immune Activation, Inflammation, and Non-AIDS Co-Morbidities in HIV-Infected Patients under Long-Term ART. Viruses. 2019;11(3).
  19. Wada NI, Jacobson LP, Margolick JB, Breen EC, Macatangay B, Penugonda S, et al. The effect of HAART-induced HIV suppression on circulating markers of inflammation and immune activation. AIDS. 2015;29(4):463-471.
  20. Neuhaus J, Jacobs DR, Jr., Baker JV, Calmy A, Duprez D, La Rosa A, et al. Markers of inflammation, coagulation, and renal function are elevated in adults with HIV infection. J Infect Dis. 2010;201(12):1788-1795.
  21. Brown TT, Tassiopoulos K, Bosch RJ, Shikuma C, McComsey GA. Association between systemic inflammation and incident diabetes in HIV-infected patients after initiation of antiretroviral therapy. Diabetes Care. 2010;33(10):2244-2249.
  22. American Diabetes A. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019;42(Suppl 1):S13-S28.
  23. Diop ME, Bastard JP, Meunier N, Thevenet S, Maachi M, Capeau J, et al. Inappropriately low glycated hemoglobin values and hemolysis in HIV-infected patients. AIDS Res Hum Retroviruses. 2006;22(12):1242-1247.
  24. Polgreen PM, Putz D, Stapleton JT. Inaccurate glycosylated hemoglobin A1C measurements in human immunodeficiency virus-positive patients with diabetes mellitus. Clin Infect Dis. 2003;37(4):e53-56.
  25. Geene D, Sudre P, Anwar D, Goehring C, Saaidia A, Hirschel B. Causes of macrocytosis in HIV-infected patients not treated with zidovudine. Swiss HIV Cohort Study. J Infect. 2000;40(2):160-163.
  26. Slama L, Palella FJ, Jr., Abraham AG, Li X, Vigouroux C, Pialoux G, et al. Inaccuracy of haemoglobin A1c among HIV-infected men: effects of CD4 cell count, antiretroviral therapies and haematological parameters. J Antimicrob Chemother. 2014;69(12):3360-3367.
  27. Aberg JA, Gallant JE, Ghanem KG, Emmanuel P, Zingman BS, Horberg MA, et al. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis. 2014;58(1):1-10.
  28. Eckhardt BJ, Holzman RS, Kwan CK, Baghdadi J, Aberg JA. Glycated Hemoglobin A(1c) as screening for diabetes mellitus in HIV-infected individuals. AIDS Patient Care STDS. 2012;26(4):197-201.
  29. Han JH, Crane HM, Bellamy SL, Frank I, Cardillo S, Bisson GP, et al. HIV infection and glycemic response to newly initiated diabetic medical therapy. AIDS. 2012;26(16):2087-2095.

70A.  ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes-2023. Diabetes Care. 2023 Jan 1;46(Suppl 1):S140-S157.

70B.  Samson SL, Vellanki P, Blonde L, Christofides EA, Galindo RJ, Hirsch IB, Isaacs SD, Izuora KE, Low Wang CC, Twining CL, Umpierrez GE, Valencia WM. American Association of Clinical Endocrinology Consensus Statement: Comprehensive Type 2 Diabetes Management Algorithm - 2023 Update. Endocr Pract. 2023 May;29(5):305-340.

70C.  Jaggers JR, Prasad VK, Dudgeon WD, Blair SN, Sui X, Burgess S, Hand GA. Associations between physical activity and sedentary time on components of metabolic syndrome among adults with HIV. AIDS Care. 2014;26(11):1387-92.

70D.  Roos R, Myezwa H, van Aswegen H, Musenge E. Effects of an education and home-based pedometer walking program on ischemic heart disease risk factors in people infected with HIV: a randomized trial. J Acquir Immune Defic Syndr. 2014 Nov 1;67(3):268-76.

70E.  Rehm KE, Konkle-Parker D. Physical activity levels and perceived benefits and barriers to physical activity in HIV-infected women living in the deep south of the United States. AIDS Care. 2016 Sep;28(9):1205-10.

70F.  Vancampfort D, Mugisha J, Richards J, De Hert M, Lazzarotto AR, Schuch FB, Probst M, Stubbs B. Dropout from physical activity interventions in people living with HIV: a systematic review and meta-analysis. AIDS Care. 2017 May;29(5):636-643.

  1. American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019;42(Suppl 1):S90-S102.
  2. Hadigan C, Corcoran C, Basgoz N, Davis B, Sax P, Grinspoon S. Metformin in the treatment of HIV lipodystrophy syndrome: A randomized controlled trial. JAMA. 2000;284(4):472-477.
  3. Song IH, Zong J, Borland J, Jerva F, Wynne B, Zamek-Gliszczynski MJ, et al. The Effect of Dolutegravir on the Pharmacokinetics of Metformin in Healthy Subjects. J Acquir Immune Defic Syndr. 2016;72(4):400-407.
  4. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/204790lbl.pdf. Tdpi. Accessed July 8, 2019.
  5. Gervasoni C, Minisci D, Clementi E, Rizzardini G, Cattaneo D. How Relevant is the Interaction Between Dolutegravir and Metformin in Real Life? J Acquir Immune Defic Syndr. 2017;75(1):e24-e26.
  6. May M, Schindler C. Clinically and pharmacologically relevant interactions of antidiabetic drugs. Ther Adv Endocrinol Metab. 2016;7(2):69-83.
  7. Fichtenbaum CJ, Gerber JG. Interactions between antiretroviral drugs and drugs used for the therapy of the metabolic complications encountered during HIV infection. Clin Pharmacokinet. 2002;41(14):1195-1211.
  8. Han JH, Gordon K, Womack JA, Gibert CL, Leaf DA, Rimland D, et al. Comparative Effectiveness of Diabetic Oral Medications Among HIV-Infected and HIV-Uninfected Veterans. Diabetes Care. 2017;40(2):218-225.
  9. Sutinen J, Hakkinen AM, Westerbacka J, Seppala-Lindroos A, Vehkavaara S, Halavaara J, et al. Rosiglitazone in the treatment of HAART-associated lipodystrophy--a randomized double-blind placebo-controlled study. Antivir Ther. 2003;8(3):199-207.
  10. Hadigan C, Yawetz S, Thomas A, Havers F, Sax PE, Grinspoon S. Metabolic effects of rosiglitazone in HIV lipodystrophy: a randomized, controlled trial. Ann Intern Med. 2004;140(10):786-794.
  11. Cavalcanti RB, Raboud J, Shen S, Kain KC, Cheung A, Walmsley S. A randomized, placebo-controlled trial of rosiglitazone for HIV-related lipoatrophy. J Infect Dis. 2007;195(12):1754-1761.
  12. Grinspoon S. Use of thiazolidinediones in HIV-infected patients: what have we learned? J Infect Dis. 2007;195(12):1731-1733.
  13. van Wijk JP, de Koning EJ, Cabezas MC, op't Roodt J, Joven J, Rabelink TJ, et al. Comparison of rosiglitazone and metformin for treating HIV lipodystrophy: a randomized trial. Ann Intern Med. 2005;143(5):337-346.
  14. Pioglitazone - FDA label. https://wwwaccessdatafdagov/drugsatfda_docs/label/2011/021073s043s044lblpdf. Accessed July 10, 2019.
  15. Makdissi A, Ghanim H, Vora M, Green K, Abuaysheh S, Chaudhuri A, et al. Sitagliptin exerts an antinflammatory action. J Clin Endocrinol Metab. 2012;97(9):3333-3341.
  16. Tremblay AJ, Lamarche B, Deacon CF, Weisnagel SJ, Couture P. Effects of sitagliptin therapy on markers of low-grade inflammation and cell adhesion molecules in patients with type 2 diabetes. Metabolism. 2014;63(9):1141-1148.
  17. Goodwin SR, Reeds DN, Royal M, Struthers H, Laciny E, Yarasheski KE. Dipeptidyl peptidase IV inhibition does not adversely affect immune or virological status in HIV infected men and women: a pilot safety study. J Clin Endocrinol Metab. 2013;98(2):743-751.
  18. Best C, Struthers H, Laciny E, Royal M, Reeds DN, Yarasheski KE. Sitagliptin Reduces Inflammation and Chronic Immune Cell Activation in HIV+ Adults With Impaired Glucose Tolerance. J Clin Endocrinol Metab. 2015;100(7):2621-2629.
  19. Dube MP, Chan ES, Lake JE, Williams B, Kinslow J, Landay A, et al. A Randomized, Double-Blinded, Placebo-Controlled Trial of Sitagliptin for Reducing Inflammation and Immune Activation in Treated and Suppressed HIV Infection. Clin Infect Dis. 2018.
  20. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2016;375(4):311-322.
  21. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, et al. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med. 2016;375(19):1834-1844.
  22. Diamant M, van Agtmael M. Liraglutide treatment in a patient with HIV and uncontrolled insulin-treated type 2 diabetes. Diabetes Care. 2012;35(5):e34.
  23. Gutierrez AD, Balasubramanyam A. Dysregulation of glucose metabolism in HIV patients: epidemiology, mechanisms, and management. Endocrine. 2012;41(1):1-10.
  24. Paik IJ, Kotler DP. The prevalence and pathogenesis of diabetes mellitus in treated HIV-infection. Best Pract Res Clin Endocrinol Metab. 2011;25(3):469-478.
  25. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2019;380(4):347-357.
  26. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med. 2015;373(22):2117-2128.

96A.  Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes. 2022 Aug 26. 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, Laferrère 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, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–97.  

  1. Garcia de Lucas MD, Olalla J. Experience of using ISGTL-2 in patients with DM2 and HIV infection. Eur J Intern Med. 2017;41:e29.
  2. Bersoff-Matcha SJ, Chamberlain C, Cao C, Kortepeter C, Chong WH. Fournier Gangrene Associated With Sodium-Glucose Cotransporter-2 Inhibitors: A Review of Spontaneous Postmarketing Cases. Ann Intern Med. 2019.
  3. Elem B, Ranjan P. Impact of immunodeficiency virus (HIV) on Fournier's gangrene: observations in Zambia. Ann R Coll Surg Engl. 1995;77(4):283-286.
  4. Ngugi P, Magoha G, Nyaga P. Fournier's ganrene in the HIV era. Afr Health Sci. 2014;14(4):1063-1068.
  5. Chazan B, Chen Y, Raz R, Kopelman D, Katz Z, Kniznik D, et al. Fournier's gangrene as the initial presentation of HIV infection. Int J Infect Dis. 2007;11(2):184-185.
  6. Chalya PL, Igenge JZ, Mabula JB, Simbila S. Fournier's gangrene at a tertiary health facility in northwestern Tanzania: a single centre experiences with 84 patients. BMC Res Notes. 2015;8:481.
  7. Saylor D. Neurologic Complications of Human Immunodeficiency Virus Infection. Continuum (Minneap Minn). 2018;24(5, Neuroinfectious Disease):1397-1421.
  8. Ghosh S, Chandran A, Jansen JP. Epidemiology of HIV-related neuropathy: a systematic literature review. AIDS Res Hum Retroviruses. 2012;28(1):36-48.
  9. Simpson DM, Tagliati M. Nucleoside analogue-associated peripheral neuropathy in human immunodeficiency virus infection. J Acquir Immune Defic Syndr Hum Retrovirol. 1995;9(2):153-161.
  10. Wang SX, Ho EL, Grill M, Lee E, Peterson J, Robertson K, et al. Peripheral neuropathy in primary HIV infection associates with systemic and central nervous system immune activation. J Acquir Immune Defic Syndr. 2014;66(3):303-310.
  11. Erlandson KM, Zhang L, Ng DK, Althoff KN, Palella FJ, Jr., Kingsley LA, et al. Risk Factors for Falls, Falls With Injury, and Falls With Fracture Among Older Men With or at Risk of HIV Infection. J Acquir Immune Defic Syndr. 2019;81(4):e117-e126.
  12. Ekrikpo UE, Kengne AP, Bello AK, Effa EE, Noubiap JJ, Salako BL, et al. Chronic kidney disease in the global adult HIV-infected population: A systematic review and meta-analysis. PLoS One. 2018;13(4):e0195443.
  13. Palella FJ, Jr., Li X, Gupta SK, Estrella MM, Phair JP, Margolick JB, et al. Long-term kidney function, proteinuria, and associated risks among HIV-infected and uninfected men. AIDS. 2018;32(10):1247-1256.
  14. Ng DK, Jacobson LP, Brown TT, Palella FJ, Jr., Martinson JJ, Bolan R, et al. HIV therapy, metabolic and cardiovascular health are associated with glomerular hyperfiltration among men with and without HIV infection. AIDS. 2014;28(3):377-386.
  15. Tonneijck L, Muskiet MH, Smits MM, van Bommel EJ, Heerspink HJ, van Raalte DH, et al. Glomerular Hyperfiltration in Diabetes: Mechanisms, Clinical Significance, and Treatment. J Am Soc Nephrol. 2017;28(4):1023-1039.
  16. Wyatt CM, Arons RR, Klotman PE, Klotman ME. Acute renal failure in hospitalized patients with HIV: risk factors and impact on in-hospital mortality. AIDS. 2006;20(4):561-565.
  17. Naicker S, Aboud O, Gharbi MB. Epidemiology of acute kidney injury in Africa. Semin Nephrol. 2008;28(4):348-353.
  18. Hou J, Nast CC. Changing concepts of HIV infection and renal disease. Curr Opin Nephrol Hypertens. 2018;27(3):144-152.
  19. Scherzer R, Estrella M, Li Y, Choi AI, Deeks SG, Grunfeld C, et al. Association of tenofovir exposure with kidney disease risk in HIV infection. AIDS. 2012;26(7):867-875.
  20. Mocroft A, Kirk O, Gatell J, Reiss P, Gargalianos P, Zilmer K, et al. Chronic renal failure among HIV-1-infected patients. AIDS. 2007;21(9):1119-1127.
  21. Demirkaya N, Wit FW, van Den Berg TJ, Kooij KW, Prins M, Schlingemann RO, et al. HIV-Associated Neuroretinal Disorder in Patients With Well-Suppressed HIV-Infection: A Comparative Cohort Study. Invest Ophthalmol Vis Sci. 2016;57(3):1388-1397.
  22. Masur H, Brooks JT, Benson CA, Holmes KK, Pau AK, Kaplan JE, et al. Prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: Updated Guidelines from the Centers for Disease Control and Prevention, National Institutes of Health, and HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis. 2014;58(9):1308-1311.
  23. Jabs DA, Drye L, Van Natta ML, Thorne JE, Holland GN, Studies of the Ocular Complications of ARG. Incidence and long-term outcomes of the human immunodeficiency virus neuroretinal disorder in patients with AIDS. Ophthalmology. 2015;122(4):760-768.
  24. Triant VA, Lee H, Hadigan C, Grinspoon SK. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J Clin Endocrinol Metab. 2007;92(7):2506-2512.
  25. Worm SW, Sabin C, Weber R, Reiss P, El-Sadr W, Dabis F, et al. Risk of myocardial infarction in patients with HIV infection exposed to specific individual antiretroviral drugs from the 3 major drug classes: the data collection on adverse events of anti-HIV drugs (D:A:D) study. J Infect Dis. 2010;201(3):318-330.
  26. Duprez DA, Neuhaus J, Kuller LH, Tracy R, Belloso W, De Wit S, et al. Inflammation, coagulation and cardiovascular disease in HIV-infected individuals. PLoS One. 2012;7(9):e44454.
  27. Thompson-Paul AM, Lichtenstein KA, Armon C, Palella FJ, Jr., Skarbinski J, Chmiel JS, et al. Cardiovascular Disease Risk Prediction in the HIV Outpatient Study. Clin Infect Dis. 2016;63(11):1508-1516.
  28. Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, et al. 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. J Am Coll Cardiol. 2019.
  29. American Diabetes A. 9. Cardiovascular Disease and Risk Management: Standards of Medical Care in Diabetes-2018. Diabetes Care. 2018;41(Suppl 1):S86-S104.
  30. Suchindran S, Regan S, Meigs JB, Grinspoon SK, Triant VA. Aspirin Use for Primary and Secondary Prevention in Human Immunodeficiency Virus (HIV)-Infected and HIV-Uninfected Patients. Open Forum Infect Dis. 2014;1(3):ofu076.
  31. Ladapo JA, Richards AK, DeWitt CM, Harawa NT, Shoptaw S, Cunningham WE, et al. Disparities in the Quality of Cardiovascular Care Between HIV-Infected Versus HIV-Uninfected Adults in the United States: A Cross-Sectional Study. J Am Heart Assoc. 2017;6(11).
  32. Elf JL, Variava E, Chon S, Lebina L, Motlhaoleng K, Gupte N, et al. Prevalence and Correlates of Smoking Among People Living With HIV in South Africa. Nicotine Tob Res. 2018;20(9):1124-1131.
  33. Mdodo R, Frazier EL, Dube SR, Mattson CL, Sutton MY, Brooks JT, et al. Cigarette smoking prevalence among adults with HIV compared with the general adult population in the United States: cross-sectional surveys. Ann Intern Med. 2015;162(5):335-344.
  34. Xu Y, Chen X, Wang K. Global prevalence of hypertension among people living with HIV: a systematic review and meta-analysis. J Am Soc Hypertens. 2017;11(8):530-540.
  35. Seaberg EC, Munoz A, Lu M, Detels R, Margolick JB, Riddler SA, et al. Association between highly active antiretroviral therapy and hypertension in a large cohort of men followed from 1984 to 2003. AIDS. 2005;19(9):953-960.
  36. American Diabetes A. 10. Cardiovascular Disease and Risk Management: Standards of Medical Care in Diabetes-2019. Diabetes care. 2019;42(Suppl 1):S103-S123.
  37. Grunfeld C, Pang M, Doerrler W, Shigenaga JK, Jensen P, Feingold KR. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J Clin Endocrinol Metab. 1992;74(5):1045-1052.
  38. Riddler SA, Smit E, Cole SR, Li R, Chmiel JS, Dobs A, et al. Impact of HIV infection and HAART on serum lipids in men. JAMA. 2003;289(22):2978-2982.

134A. 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; REPRIEVE Investigators. Pitavastatin to Prevent Cardiovascular Disease in HIV Infection. N Engl J Med. 2023 Aug 24;389(8):687-699.

  1. Reddy KP, Parker RA, Losina E, Baggett TP, Paltiel AD, Rigotti NA, et al. Impact of Cigarette Smoking and Smoking Cessation on Life Expectancy Among People With HIV: A US-Based Modeling Study. J Infect Dis. 2016;214(11):1672-1681.
  2. American Diabetes A. 5. Lifestyle Management: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019;42(Suppl 1):S46-S60.

 

Dyslipidemia in Patients With Diabetes

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in both men and women with T1DM and T2DM. In patients with T1DM in good glycemic control, the lipid profile is very similar to the general population. In contrast, in patients with T2DM, even with good glycemic control, there are frequently lipid abnormalities (elevated TG and non-HDL-C, decreased HDL-C, and an increase in small dense LDL). In both T1DM and T2DM, poor glycemic control increases TG levels and decreases HDL-C levels with modest effects on LDL-C levels. Extensive studies have demonstrated that statins decrease ASCVD in patients with diabetes. Treatment with high doses of potent statins reduces ASCVD events to a greater extent than low dose statin therapy. Adding fibrates or niacin to statin therapy has not been shown to further decrease ASCVD events. In contrast, studies have shown that the combination of a statin and ezetimibe or a statin and a PCSK9 inhibitor result in a greater decrease in ASCVD events than statins alone. Studies have suggested that EPA, an omega-3-fatty acid, when added to statins also reduces ASCVD events but this result is controversial. In statin intolerant patients with T2DM bempedoic acid decreases ASCVD events. Current recommendations state that most patients with diabetes should be on statin therapy. In certain patients with diabetes ezetimibe, PCSK9 inhibitors, and bempedoic acid can play a role in reducing ASCVD.  

 

INTRODUCTION

 

Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in both men and women with diabetes (1-5). In addition to coronary disease, ASCVD includes stroke and peripheral arterial disease (PAD).  PAD is common in diabetes, may be the first presentation of ASCVD and should be recognized as needing aggressive treatment of risk factors. The risk of ASCVD is increased approximately 2-fold in men and 3-4-fold in women (2-4,6,7). In the Framingham study, the annual rate of ASCVD was similar in men and women with diabetes, emphasizing that woman with diabetes need as aggressive preventive treatment as men with diabetes (2,6). In addition, several but not all studies, have shown that patients with diabetes with no history of ASCVD have a similar risk of having a myocardial infarction as non-diabetic patients who have a history of ASCVD, i.e., diabetes is an equivalent risk factor as a history of a previous cardiovascular event (8,9). The duration of diabetes and the presence of other risk factors or complications of diabetes likely determine whether a patient with diabetes has a risk equivalent to patients with a history of previous ASCVD events (10,11). In one study patients with T2DM who had the following risk factors within the target range, HbA1c, LDL-C, albuminuria, smoking, and blood pressure, the risk of an acute myocardial infarction or stroke was similar to individuals without diabetics (12). Moreover, numerous studies have shown that patients with diabetes who have ASCVD are at a very high risk of having another event, indicating that this population of patient’s needs especially aggressive preventive measures (1,8). This increased risk for the development of ASCVD in patients with diabetes is seen both in populations where the prevalence of ASCVD is high (Western societies) and low (for example, Japan) (2). However, in societies where the prevalence of ASCVD is low, the contribution of ASCVD as a cause of morbidity and mortality in patients with diabetes is relatively low compared to Western societies (2).

 

While the database is not as robust, the evidence indicates that patients with T1DM are also at high risk for the development of ASCVD (1,13-15). Interestingly, women with T1DM have twice the excess risk of fatal and nonfatal vascular events compared to men with T1DM (16,17). Additionally, developing T1DM at a young age increases the risk of ASCVD to a greater degree than late onset T1DM (17). Approximately 50% of patients with T1DM are obese or overweight and between 8% and 40% meet the criteria for the metabolic syndrome, which increases their risk of developing ASCVD (18).

 

While the development of diabetes at a young age increases the risk of ASCVD in patients with both T1DM and T2DM the deleterious impact is greater in patients with T2DM (19). Lastly, in patients with both T1DM and T2DM the presence of renal disease increases the risk of ASCVD (4,14). Of note is that the risk of developing ASCVD events in patients with diabetes has decreased recently, most likely due to better lipid and blood pressure control, which again reinforces the need to aggressively treat these risk factors in patients with diabetes (5,7,20). 

 

ROLE OF RISK FACTORS IN ASCVD

 

Numerous studies have demonstrated that the traditional risk factors for ASCVD play an important role in patients with diabetes (2,4,5,110). Patients with diabetes without other risk factors have a relatively low risk of ASCVD (in most studies higher than similar non-diabetic patients), whereas the increasing prevalence of other risk factors markedly increases the risk of developing ASCVD (2). The major reversible traditional risk factors are hypertension, cigarette smoking, and lipid abnormalities (2,4,5,14,111). Other risk factors include obesity (particularly visceral obesity), insulin resistance, small dense LDL, elevated TG, low HDL-C, procoagulant state (increased PAI-1, fibrinogen), family history of early ASCVD, homocysteine, Lp (a), renal disease, albuminuria, and inflammation (C-reactive protein, SAA, cytokines) (2,4,5,110,111). In the last decade, it has become clear that to reduce the risk of ASCVD in patients with diabetes, one will not only need to improve glycemic control but also address these other cardiovascular risk factors. In the remainder of this chapter, I will focus on the dyslipidemia that occurs in patients with diabetes.

 

ROLE OF LIPIDS IN ASCVD

 

As in non-diabetic populations, epidemiological studies have shown that increased LDL-C and non-HDL-C levels and decreased HDL-C levels are associated with an increased risk of ASCVD in patients with diabetes (2,4,110,111). In the UKPDS cohort LDL-C levels were the strongest predictor of coronary artery disease (112). While it is universally accepted that elevated levels of LDL-C and non-HDL-C cause atherosclerosis and ASCVD the role of HDL-C is uncertain. Genetic studies and studies of drugs that raise HDL-C have not supported low HDL-C levels as a causative factor for atherosclerosis (113). Rather it is currently thought that HDL function is associated with atherosclerosis risk and that this does not precisely correlate with HDL-C levels (113). In patients with diabetes, elevations in serum triglyceride (TG) levels also are associated with an increased risk of ASCVD (4,111,114). With regard to TG, it is not clear whether they are a causative factor for ASCVD or whether the elevation in TG is a marker for other abnormalities (4,111,114,115). Recent Mendelian randomization studies have provided support for the hypothesis that elevated TG levels play a causal role in atherosclerosis (115,116). Unfortunately, as will be discussed later in this chapter lowering TG levels in patients on statin therapy has not decreased cardiovascular events.

 

LIPID ABNORMALITIES IN PATIENTS WITH DIABETES

 

In patients with T1DM in good glycemic control, the lipid profile is very similar to lipid profiles in the general population (110). In some studies HDL-C levels are modestly increased in patients with T1DM (117). In contrast, in patients with T2DM, even when in good glycemic control, there are abnormalities in lipid levels (118-121). It is estimated that 30-60% of patients with T2DM have dyslipidemia (5,122). Specifically, patients with T2DM often have an increase in serum TG levels, increased VLDL and IDL, and decreased HDL-C levels. Non-HDL-C levels are increased due to the increase in VLDL and IDL. LDL-C levels are typically not markedly different than in normal subjects but there is an increase in small dense LDL, a lipoprotein particle that may be particularly pro-atherogenic (123). As a consequence, there are more LDL particles, which coupled with the increases in VLDL and IDL, leads to an increase in apolipoprotein B levels (118-121). Additionally, the postprandial increase in serum TG is accentuated and elevations in postprandial lipids may increase the risk of ASCVD (118-121).

 

It should be recognized that the lipid changes in patients with T2D are characteristic of the alterations in lipid profile seen in obesity and the metabolic syndrome (insulin resistance syndrome) (124). Since a high percentage of patients with T2DM are obese, insulin resistant, and have the metabolic syndrome, it is not surprising that the prevalence of increased TG and small dense LDL and decreased HDL-C is common in patients with T2DM even when these patients are in good glycemic control. Obesity is also accompanied by increased systemic inflammation. The increasing prevalence of obesity/overweight in patients with T1D will likely result in an increased prevalence of dyslipidemia in this population.

 

Studies have shown that the anti-oxidant and anti-inflammatory functions of HDL isolated from patients with T1DM and T2DM are reduced (117,125). Additionally, the ability of HDL to facilitate cholesterol efflux is reduced in patients with T1DM and T2DM (126,127). Together these findings indicate that HDL-C levels per se may not fully reflect risk of ASCVD in patients with diabetes and that HDL function is perturbed in patients with diabetes.

 

In both T1DM and T2DM, poor glycemic control increases serum TG levels, VLDL, and IDL, and decreases HDL-C levels (119). Poor glycemic control can also result in a modest increase in LDL-C, which because of the elevation in TG is often in the small dense LDL subfraction. It is therefore important to optimize glycemic control in patients with diabetes because this will have secondary beneficial effects on lipid levels.

 

Lp(a) levels are usually within the normal range in patients with T1DM and T2DM (128). Some studies have observed no impact of diabetes mellitus on Lp(a) concentrations while other studies reported an elevation or a decrease in Lp(a) concentrations (128). The development of microalbuminuria and the onset of renal disease are associated with an increase in Lp (a) levels (129). Of note low Lp(a) levels are associated with an increased risk of developing T2DM (128). A recent very large case control study found that an Lp(a) concentration in the bottom 10% increases T2DM risk (130).

 

Table 1. Lipid Abnormalities in Patients with Diabetes

T1DM

Lipid profile is similar to controls if glycemic control is good

T2DM

Increased TG, VLDL, IDL, and non-HDL-C. Decreased HDL-C. Normal LDL-C but increase in small dense LDL, LDL particle number, and apolipoprotein B.

Poor glycemic control

Increased TG, VLDL, IDL, and non-HDL-C.  Decreased HDL-C. Modest increase in LDL-C with increase in small dense LDL, LDL particle number, and apolipoprotein B.

 

EFFECT OF GLUCOSE LOWERING DRUGS ON LIPIDS

 

Some therapies used to improve glycemic control may have an impact on lipid levels above and beyond their effects on glucose metabolism. In reviewing the literature, it is often very difficult to separate improvements in glycemic control vs. direct effects of drugs. Additionally, many of the changes induced by drug therapy result in only small changes in LDL-C, HDL-C, and TG levels, are variable from study to study, and are of questionable clinical significance. Insulin, sulfonylureas, meglinitides, DPP4 inhibitors, and alpha-glucosidase inhibitors do not appear to markedly alter fasting lipid profiles other than by improving glucose control (there are data indicating that DPP4 inhibitors and acarbose decrease postprandial triglyceride excursions, but they do not markedly alter fasting lipid levels) (131). In contrast, metformin, thiazolidinediones, GLP1 receptor agonists, bromocriptine-QR, and SGLT2 inhibitors have effects independent of glycemic control on serum lipid levels (table 2).

 

Metformin may decrease serum TG levels and LDL-C levels without altering HDL-C levels (131). In a meta-analysis of 37 trials with 2,891 patients, metformin decreased TG by 11.4mg/dL when compared with control treatment (p=0.003) (132). In an analysis of 24 trials with 1,867 patients, metformin decreased LDL-C by 8.4mg/dL compared to control treatment (p<0.001) (132). In contrast, metformin did not significantly alter HDL-C levels (132). It should be noted that in the Diabetes Prevention Program 3,234 individuals with impaired glucose metabolism were randomized to placebo, intensive lifestyle, or metformin therapy. In the metformin therapy group no significant changes were noted in TG, LDL-C, or HDL-C levels compared to the placebo group (133). Thus, metformin may have small effects on lipid levels.    

 

The effect of thiazolidinediones depends on which agent is used. Rosiglitazone increases serum LDL-C levels, increases HDL-C levels, and only decreases serum TG if the baseline TG levels are high (131). In contrast, pioglitazone has less impact on LDL-C levels, but increases HDL-C levels, and decreases TG (131). In the PROactive study, a large randomized cardiovascular outcome study, pioglitazone decreased TG levels by approximately 10%, increased HDL-C levels by approximately 10%, and increased LDL-C by 1-4% (134). It should be noted that reductions in the small dense LDL subfraction and an increase in the large buoyant LDL subfraction are seen with both thiazolidinediones (131). In a randomized head-to-head trial, it was shown that pioglitazone decreased TG levels and increased serum HDL-C levels to a greater degree than rosiglitazone treatment (135,136). Additionally, pioglitazone increased LDL-C levels less than rosiglitazone. In contrast to the differences in lipid parameters, both rosiglitazone and pioglitazone decreased A1c and C-reactive protein to a similar extent. The mechanism by which pioglitazone induces more favorable changes in lipid levels than rosiglitazone despite similar changes in glucose levels is unclear, but differential actions of ligands for nuclear hormone receptors are well described.

 

Treatment with SGLT2 inhibitors results in a small increase in LDL-C and HDL-C levels (131). In a meta-analysis of 48 randomized controlled trials SGLT2 inhibitors significantly increased LDL-C (3.8mg/dL, p < 0.00001), HDL-C (2.3mg/dL, p < 0.00001), and decreased TG levels (8.8mg/dL, p < 0.00001) (137). The mechanism for these increases in LDL and HDL cholesterol is unknown but could be due to a decrease in plasma volume. The decrease in TG levels could be secondary to weight loss.

 

Bromocriptine-QR (Cycloset) treatment decreases TG levels but has no significant effect on LDL-C or HDL-C levels (138,139). The decrease in TG levels is thought to be due to a decrease in hepatic TG synthesis, likely due to a decrease in adipose tissue lipolysis resulting in decreased blood free fatty acid levels and reduced delivery of fatty acids to the liver for TG synthesis (140).

 

Colesevelam, a bile acid sequestrant that is approved for glucose lowering, lowers LDL-C levels by 15-20% and has only a modest effect on HDL-C levels (101,141). The effect of bile acid sequestrants on TG levels varies (141). In patients with normal TG levels, bile acid sequestrants increase TG levels by a small amount. However, as baseline TG levels increase, the effect of bile acid sequestrants on TG levels becomes greater, and can result in substantial increases in TG levels (141). In patients with TG > 500mg/dL the use of bile acid sequestrants is contraindicated (141).

 

Finally, GLP-1 receptor agonists can favorably affect the lipid profile by inducing weight loss (decreasing TG and very modestly decreasing LDL-C levels) (131). In a review by Nauck and colleagues it was noted that GLP-1 receptor agonists lowered TG levels by 18 to 62mg/dL depending upon the specific GLP-1 receptor agonist while decreasing LDL-C by 3-8mg/dL and increasing HDL-C by less than 1mg/dL (142). Additionally, GLP-1 receptor agonists reduce postprandial TG by reducing circulating chylomicrons by decreasing intestinal lipoprotein production (131,142). DPP4 inhibitors have a similar effect on postprandial TG levels as GLP-1 receptor agonists while having minimal effects on fasting lipid levels (142).

 

In the SURPASS trials, tirzepatide studies TG levels were consistently decreased by 13-25% (83,143). In most studies with the exception of SURPASS 5, HDL cholesterol levels increased by 3-11% (83,143). Total cholesterol and LDL cholesterol levels were modestly decreased in most studies (83,143). Not unexpectedly given the decrease in TG levels small LDL particles were decreased. For details see the Endotext chapter Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes (83).

 

Table 2. Effect of Glucose Lowering Drugs on Lipid Levels

Metformin

Modestly decrease TG and LDL-C

Sulfonylureas

No effect

DPP4 inhibitors

Decrease postprandial TG

GLP1 analogues

Decrease fasting and postprandial TG, modestly decrease LDL-C

Tirzepatide

Decrease TG, modestly decrease LDL-C, increase HDL-C

Acarbose

Decrease postprandial TG

Pioglitazone

Rosiglitazone

Decrease TG and increase HDL-C. Small increase LDL-C but a decrease in small dense LDL

SGLT2 inhibitors

Small increase in LDL-C and HDL-C

Colesevelam

Decrease LDL-C. May increase TG

Bromocriptine-QR

Decrease TG

Insulin

No effect

 

PATHOPHYSIOLOGY OF THE DYSLIPIDEMIA OF DIABETES

 

Figure 1. Pathophysiology of the Dyslipidemia of Diabetes

 

Multiple mechanisms account for the dyslipidemia seen in patients with T2DM, which are affected both by the level of glucose control and by factors such as obesity and inflammation that also contribute to dyslipidemia.

 

Increase in TG

 

There are a number of different abnormalities that contribute to the dyslipidemia seen in patients with T2DM and obesity (figure 1) (119-122,144-146).

 

OVERPRODUCTION OF VLDL BY THE LIVER

 

A key abnormality is the overproduction of VLDL by the liver, which is a major contributor to the elevations in serum TG levels. The rate of secretion of VLDL is highly dependent on TG availability, which is determined by the levels of fatty acids available for the synthesis of TG in the liver. An abundance of TG prevents the intra-hepatic degradation of Apo B-100 allowing for increased VLDL formation and secretion. There are three major sources of fatty acids in the liver all of which may be altered in patients with T2DM. First, the flux of fatty acids from adipose tissue to the liver is increased. An increased mass of adipose tissue, particularly visceral stores, results in increased fatty acid delivery to the liver. Additionally, insulin suppresses the lipolysis of TG to free fatty acids in adipose tissue; thus, in patients with either poorly controlled diabetes due to a decrease in insulin or a decrease in insulin activity due to insulin resistance, the inhibition of TG lipolysis is blunted and there is increased TG breakdown leading to increased fatty acid deliver to the liver. A second source of fatty acids in the liver is de novo fatty acid synthesis. Numerous studies have shown that fatty acid synthesis is increased in the liver in patients with T2DM. This increase may be mediated by the hyperinsulinemia seen in patients with insulin resistance. While the liver is resistant to the effects of insulin on carbohydrate metabolism, the liver remains sensitive to the effects of insulin stimulating lipid synthesis. Specifically, insulin stimulates the activity of SREBP-1c, a transcription factor that increases the expression of the enzymes required for the synthesis of fatty acids. Thus, while the liver is resistant to the effects of insulin on carbohydrate metabolism the liver remains sensitive to the effects of insulin stimulating lipid synthesis. Additionally, in the presence of hyperglycemia, glucose can induce another transcription factor, carbohydrate responsive element binding protein (ChREBP), which also stimulates the transcription of the enzymes required for fatty acid synthesis. The third source of fatty acids is the uptake of TG rich lipoproteins by the liver. Studies have shown an increase in intestinal fatty acid synthesis and the enhanced secretion of chylomicrons in animal models of T2DM. This increase in chylomicrons leads to the increased delivery of fatty acids to the liver. The increase in hepatic fatty acids produced by these three pathways results in an increase in the synthesis of TG in the liver and the protection of Apo B-100 from degradation resulting in the increased formation and secretion of VLDL. Finally, insulin stimulates the post translational degradation of Apo B-100 in the liver and a decrease in insulin activity in patients with T2DM also allows for the enhanced survival of Apo B-100 promoting increased VLDL formation.

 

DECREASED DEGRADATION OF TRIGLYCERIDE RICH LIPOPROTEINS

 

While the overproduction of triglyceride rich lipoproteins by the liver and intestine are important contributors to the elevations in serum TG levels in patients with T2DM, there are also abnormalities in the metabolism of these TG rich lipoproteins. First, there is a modest decrease in lipoprotein lipase activity, the key enzyme that metabolizes TG rich lipoproteins. The expression of lipoprotein lipase is stimulated by insulin and decreased insulin activity in patients with T2DM results in a decrease in lipoprotein lipase, which plays a key role in the hydrolysis of the TG carried in chylomicrons and VLDL. Additionally, patients with T2DM have an increase in Apo C-III levels, a key regulator of TG rich lipoprotein clearance. Glucose stimulates and insulin suppresses Apo C-III expression; thus, diabetes with hyperglycemia and either insulin deficiency or insulin resistance contribute to an increase in Apo C-III. Apo C-III is an inhibitor of lipoprotein lipase activity and thereby reduces the clearance of TG rich lipoproteins. In addition, Apo C-III also inhibits the cellular uptake of lipoproteins. Studies have shown that loss of function mutations in Apo C-III lead to lower serum TG levels and a reduced risk of ASCVD (147,148). Interestingly, inhibition of Apo C-III expression results in a decrease in serum TG levels even in patients deficient in lipoprotein lipase, indicating that the ability of Apo C-III to modulate serum TG levels is not dependent solely on regulating lipoprotein lipase activity (149). Lastly, insulin resistance is associated with an increase in Angptl3, an inhibitor of LPL (150). Thus, in patients with diabetes, a decrease in clearance of TG rich lipoproteins also contributes to the elevation in serum triglyceride levels.  

 

Mechanism for the Increase in Small Dense LDL and Decrease in HDL

 

The elevation in TG rich lipoproteins in turn has effects on other lipoproteins. Specifically, cholesterol ester transfer protein (CETP) mediates the exchange of TG from TG rich VLDL and chylomicrons to LDL and HDL. The increase in TG rich lipoproteins per se leads to an increase in CETP mediated exchange, increasing the TG content of both LDL and HDL. The TG on LDL and HDL is then hydrolyzed by hepatic lipase and lipoprotein lipase leading to the production of small dense LDL and small HDL. Notably hepatic lipase activity is increased in patients with T2DM, which will also facilitate the removal of TG from LDL and HDL resulting in small lipoprotein particles. The affinity of Apo A-I for small HDL particles is reduced, leading to the disassociation of Apo A-I, which in turn leads to the accelerated clearance and breakdown of Apo A-I by the kidneys. Additionally, the production of Apo A-I may be reduced in patients with diabetes. High glucose levels can activate ChREBP and this transcription factor inhibits Apo A-I expression. Furthermore, insulin stimulates Apo A-I expression and a reduction in insulin activity due to insulin resistance or decreased insulin levels may also lead to a decrease in Apo A-I expression. The net result is lower levels of Apo A-I and HDL-C levels in patients with T2DM.

 

Role of Poor Glycemic Control

 

The above-described changes lead to the typical dyslipidemia observed in patients with T2DM (increased TG, decreased HDL-C, and an abundance of small dense LDL and small HDL). In patients with both Type 1 and T2DM, poor glycemic control can further adversely affect lipid and lipoprotein metabolism. As noted above the expression of lipoprotein lipase is stimulated by insulin. If insulin activity is very low the expression of lipoprotein lipase is severely suppressed and the metabolism of TG rich lipoproteins is markedly impaired. This leads to the delayed clearance of both chylomicrons and VLDL and elevations of TG rich lipoproteins. Additionally, insulinopenia results in a marked increase in lipolysis in adipose tissue, leading to the release of free fatty acids into the circulation. This increase in serum fatty acids results in the increased delivery of fatty acids to the liver, enhanced TG synthesis in the liver, and the increased production and secretion of VLDL. Whereas patients with T1DM who are well controlled and not obese or overweight typically have normal serum lipid profiles, if their control deteriorates, they will develop hypertriglyceridemia. In patients with T2DM deterioration of glycemic control will further exacerbate their underlying dyslipidemia resulting in greater increases in TG levels. If the synthesis of new VLDL is increased sufficiently this can result in an increase in LDL-C levels. HDL-C levels may decrease due to the formation of small HDL that are more susceptible to accelerated clearance. Improvements in glycemic control can markedly lower TG levels and may increase serum HDL-C levels. In patients with poorly controlled diabetes improvements in glycemic control may also lower LDL-C levels.

 

Role of Obesity and Inflammation

 

Most patients with T2DM and many patients with T1D are obese or overweight. Obesity is a pro-inflammatory state due to the macrophages that infiltrate adipose tissue. The cytokines produced by these macrophages and the adipokines that are produced by fat cells also alter lipid metabolism (151,152). The pro-inflammatory cytokines, TNF and IL-1, decrease the expression of lipoprotein lipase and increase the expression of angiopoietin like protein 4, an inhibitor of lipoprotein lipase. Together these changes decrease lipoprotein lipase activity, thereby delaying the clearance of TG rich lipoproteins. In addition, pro-inflammatory cytokines stimulate lipolysis in adipocytes increasing circulating free fatty acid levels, which will provide substrate for hepatic TG synthesis. In the liver, pro-inflammatory cytokines stimulate de novo fatty acid and TG synthesis. These alterations will lead to the increased production and secretion of VLDL. Thus, increases in the levels of pro-inflammatory cytokines will stimulate the production of TG rich lipoproteins and delay the clearance of TG rich lipoproteins, which together will contribute to the increase in serum TG that occurs in obese patients.

 

Obesity and the increase in pro-inflammatory cytokines may also affect HDL-C levels (153-155). First, pro-inflammatory cytokines inhibit the production of Apo A-I, the main protein constituent of HDL. Second, in many tissues pro-inflammatory cytokines decrease the expression of ABCA1 and ABCG1, which will lead to a decrease in the efflux of phospholipids and cholesterol from the cell to HDL decreasing the formation of mature HDL. Third, pro-inflammatory cytokines inhibit the production and activity of LCAT, which will limit the conversion of cholesterol to cholesterol esters in HDL. This conversion step is required for the formation of a normal spherical HDL particle and is crucial for the ability of HDL to increase the efflux of cholesterol from cells (including macrophages). Together these effects may lead to a decrease in HDL-C levels and a decrease in reverse cholesterol transport. Reverse cholesterol transport plays an important role in preventing cholesterol accumulation in macrophages and thereby reduces atherosclerosis.

 

Inflammation also decreases other important functions of HDL, such as its ability to prevent LDL oxidation (156). This reduction in the ability of HDL to protect from oxidation may be mediated in part by inflammation inducing lower levels of the enzyme paraoxonase, which is commonly seen in patients with diabetes (151,157). In parallel inflammation increases the oxidation of LDL and the amount of small dense LDL that is more susceptible to oxidation.

 

Role of Adipokines

 

Adipokines, such as leptin, adiponectin, and resistin, regulate lipid metabolism and the levels are altered in obese patients. Obesity increases serum leptin levels and leptin stimulates lipolysis in adipocytes which will increase serum free fatty acid levels (158). The circulating levels of adiponectin are decreased in subjects who are obese (159). Decreased adiponectin levels are associated with elevations in serum TG levels and decreases in HDL-C levels (159). This association is thought to be causal as studies in mice have shown that overexpressing adiponectin (transgenic mice) decreases TG and increases HDL-C levels while conversely, adiponectin knock-out mice have increased TG and decreased HDL-C levels (159). The adiponectin induced decrease in TG levels is mediated by an increased catabolism of TG rich lipoproteins due to an increase in lipoprotein lipase activity and a decrease Apo C-III, an inhibitor of lipoprotein lipase (159). The increase in HDL-C levels induced by adiponectin is mediated by an increase in hepatic Apo A-I and ABCA1, which results in the increased production of HDL particles (159).

 

Resistin is increased in subjects who are obese and the levels of resistin directly correlate with plasma TG levels (160). Moreover, resistin has been shown to stimulate hepatic VLDL production and secretion due to an increase in the synthesis of Apo B, TG, and cholesterol (160,161). Finally, resistin is associated with a decrease in HDL-C and Apo A-I levels (160).

 

EFFECT OF LIPID LOWERING ON ASCVD EVENTS IN PATIENTS WITH DIABETES

 

Monotherapy Studies

 

STATINS

 

The Cholesterol Treatment Trialists analyzed data from 18,686 subjects with diabetes (mostly T2DM) from 14 randomized trials (162). 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 39mg/dL (1mmol/L) 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. Thus, these studies indicate that statins are beneficial in reducing ASCVD in patients with diabetes. Because of the large number of patients with diabetes included in the Heart Protection Study (HPS) and CARDS these two studies will be discussed in greater depth.

 

The HPS was a double-blind randomized trial that focused on patients at high risk for the development of cardiovascular events, including patients with a history of MIs, other atherosclerotic lesions, diabetes, and/or hypertension (163,164). Patients were between 40 and 80 years of age and had to have total serum cholesterol levels greater than 135mg/dL (thus very few patients were excluded because they did not have a high enough cholesterol level). The major strength of this trial was the large number of patients studied (>20,000). The diabetes subgroup included 5,963 subjects and thus was as large as many other prevention trials. The study was a 2x2 study design comparing simvastatin 40mg a day vs. placebo and anti-oxidant vitamins (vitamin E 600mg, vitamin C 250mg, and beta-carotene 20mg) vs. placebo and lasted approximately 5 years. Analysis of the group randomized to the anti-oxidant vitamins revealed no beneficial or harmful effects. In contrast, simvastatin therapy (40mg per day) reduced cardiovascular events, including MIs and strokes, by approximately 25% in all participants and to a similar degree in the diabetic subjects (total ASCVD reduced 27%, coronary mortality 20%, MI 37%, stroke 24%). Further analysis of the subjects with diabetes revealed that the reduction in cardiovascular events with statin therapy was similar in individuals with diabetes diagnosed for a short duration (<6 years) and for a long duration (>13 years). Similarly, subjects with diabetes in good control (HbA1c <7%) and those not in ideal control (HbA1c >7%) also benefited to a similar degree with statin therapy. Moreover, both T1DM and T2DM patients had a comparable reduction in ASCVD with simvastatin therapy. The decrease in cardiovascular events in patients with T1DM was not statistically significant because of the small number of subjects. Nevertheless, this is the only trial that included patients with T1DM and suggests that patients with T1DM will benefit from statin therapy similar to T2DM. In general, statin therapy reduced ASCVD in all subgroups of subjects with diabetes (females, males, older age, renal disease, hypertension, high TG, low HDL, ASA therapy, etc.) i.e., statin therapy benefits all patients with diabetes (note this study did not include patients with end stage renal disease but other studies have failed to show benefits of statin therapy in patients with diabetes and end stage renal disease (165)).

 

The CARDS trial specifically focused on subjects with diabetes (166). The subjects in this trial were males and females with T2DM between the ages of 40 to 75 years of age who were at high risk of developing ASCVD based on the presence of hypertension, retinopathy, renal disease, or current smoking. Of particular note, the subjects did not have any evidence of clinical atherosclerosis (myocardial disease, stroke, peripheral vascular disease) at entry and hence this study is a primary prevention trial. Inclusion criteria included LDL-C levels less than 160mg/dL and TG levels less than 600mg/dL. It is important to recognize that the average LDL-C in this trial was approximately 118mg/dL, indicating relatively low LDL-C levels. A total of 2,838 T2DM subjects were randomized to either placebo or atorvastatin 10mg a day. Atorvastatin therapy resulted in a 40% decrease in LDL-C levels with over 80% of patients achieving LDL-C levels less than 100mg/dL. Most importantly, atorvastatin therapy resulted in a 37% reduction in cardiovascular events. In addition, strokes were reduced by 48% and coronary revascularization by 31%. As seen in the HPS, subjects with relatively low LDL-C levels (LDL <120mg/dL) benefited to a similar extent as subjects with higher LDL-C levels (>120mg/dL).

 

HPS and CARDS, in combination with the other statin trials, provide conclusive evidence that statin therapy will reduce cardiovascular events in patients with diabetes. Importantly, the benefits of statin therapy are seen in patients with diabetes in both primary and secondary prevention trials. 

 

Effect of Aggressive LDL-C Lowering with Statins

 

Studies have compared reductions of LDL-C to approximately 100mg/dL to more aggressive reductions in LDL-C on atheroma volume. The Reversal Trial studied 502 symptomatic coronary artery disease patients with an average LDL-C of 150mg/dL (167). Approximately 19% of the patients in this trial had diabetes. Patients were randomized to moderate LDL lowering therapy with pravastatin 40mg per day or to aggressive lipid lowering with atorvastatin 80mg per day. As expected, LDL-C levels were considerably lower in the atorvastatin treated group (pravastatin LDL= 110mg/dL vs. atorvastatin LDL= 79mg/dL). Most importantly, when one analyzed the change in atheroma volume determined after 18 months of therapy using intravascular ultrasound, the group treated aggressively with atorvastatin had a much lower progression rate than the group treated with pravastatin. Compared with baseline values, patients treated with atorvastatin had no change in atheroma burden (there was a very slight regression of lesions), whereas patients treated with pravastatin showed progression of lesions. When one compares the extent of the reduction in LDL-C to the change in atheroma volume, a 50% reduction in LDL (LDL-C levels of approximately 75mg/dL) resulted in the absence of lesion progression. This study suggests that lowering the LDL-C to levels well below 100mg/dL is required to prevent disease progression as measured by intravascular ultrasound. Other studies, such as Asteroid, have shown that marked reductions in LDL-C (in Asteroid the mean LDL-C levels were 61mg/dL) can also result in the regression of coronary artery atherosclerosis determined by intravascular ultrasound measurements (168). Additionally, the Saturn trial demonstrated that aggressive lipid lowering with either atorvastatin 80mg or rosuvastatin 40mg would induce regression of coronary artery atherosclerosis to a similar degree in patients with and without diabetes if the LDL-C levels were reduced to less than 70mg/dL (169). Together these trials indicate that aggressive lowering of LDL-C levels to below 70mg/dL can induce regression of atherosclerotic lesions.

 

The Prove-It trial determined in patients recently hospitalized for an acute coronary syndrome whether aggressively lowering of LDL-C with atorvastatin 80mg per day vs. moderate LDL-C lowering with pravastatin 40mg per day would have a similar effect on cardiovascular end points such as death, MI, documented unstable angina requiring hospitalization, revascularization, or stroke (170,171). In this trial, approximately 18% of the patients were diabetic. As expected, the on-treatment LDL-C levels were significantly lower in patients aggressively treated with atorvastatin compared to the moderate treated pravastatin group (atorvastatin LDL-C = approximately 62 vs. pravastatin LDL-C = approximately 95mg/dL). Of great significance, death or major cardiovascular events was reduced by 16% over the two years of the study in the group aggressively treated with atorvastatin. Moreover, the risk reduction in the patients with diabetes in the aggressive treatment group was similar to that observed in non-diabetics.

 

In the treating to new targets trial (TNT) patients with stable coronary heart disease and LDL-C levels less than 130mg/dL were randomized to either 10mg or 80mg atorvastatin and followed for an average of 4.9 years (172,173). Approximately 15% of the patients had diabetes. As expected, LDL-C levels were lowered to a greater extent in the patients treated with 80mg atorvastatin than with 10mg atorvastatin (77mg/dL vs. 101mg/dL). Impressively, the occurrence of major cardiovascular events was reduced by 22% in the group treated with atorvastatin 80mg (p<0.001). In the patients with diabetes events were reduced by 25% in the high dose statin group.

 

Finally, the IDEAL trial was a randomized study that compared atorvastatin 80mg vs. simvastatin 20-40mg in 8,888 patients with a history of ASCVD (174). Approximately 12% of the patients had diabetes. As expected, LDL-C levels were reduced to a greater extent in the atorvastatin treated group than the simvastatin treated group (approximately 81mg/dL vs. 104mg/dL). Once again, the greater reduction in LDL-C levels was associated with a greater reduction in cardiovascular events. Specifically, major coronary events defined as coronary death, nonfatal MI, or cardiac arrest was reduced by 11% (p=0.07), while nonfatal acute MI were reduced by 17% (p=0.02).

 

Combining the results of the Heart Protection Study, CARDS, Reversal, Saturn, Asteroid, Prove-It, TNT, and IDEAL leads one to the conclusion that aggressive lowering of LDL-C with statin therapy will be beneficial and suggests that in high-risk patients lowering the LDL to levels well below 100mg/dL is desirable. Moreover, the Cholesterol Treatment Trialists reviewed five trials with 39,612 subjects that were designed to determine the effect of usual vs. aggressive reductions in LDL-C (175). They reported that intensive control (approximately a 19mg/dL difference in LDL-C) resulted in a 15% decrease in major vascular events, a 13% reduction in coronary death or non-fatal MI, a 19% decrease in coronary revascularization, and a 16% decrease in strokes. As will be discussed below treatment guidelines reflect the results of these studies. Additionally, as described in detail below, studies of the addition of either ezetimibe or PCSK9 inhibitors to statins further demonstrates that aggressive lowering of LDL-C levels further reduces cardiovascular events

 

FIBRATES

 

The beneficial effect of monotherapy with fibrates (e.g., gemfibrozil, fenofibrate) on ASCVD in patients with diabetes is shown in Table 3. The results of these randomized trials suggest that monotherapy with this class of drug might reduce cardiovascular events in patients with diabetes, but the data is not very robust. The largest trial was the Field Trial (176). In this trial, 9,795 patients with T2DM between the ages of 50 and 75 not taking statin therapy were randomized to fenofibrate or placebo and followed for approximately 5 years. Fenofibrate therapy resulted in a 12% decrease in LDL-C, a 29% decrease in TG, and a 5% increase in HDL-C levels. The primary outcome was coronary events (coronary heart disease death and non-fatal MI), which were reduced by 11% in the fenofibrate group but did not reach statistical significance (p= 0.16). However, there was a 24% decrease in non-fatal MI in the fenofibrate treated group (p=0.01) and a non-significant increase in coronary heart disease mortality. Total ASCVD events (coronary events plus stroke and coronary or carotid revascularization) were reduced 11% (p=0.035). These beneficial effects of fenofibrate therapy on ASCVD were observed in patients without a previous history of ASCVD. In patients with a previous history of ASCVD no benefits were observed. Additionally, the beneficial effect of fenofibrate therapy was seen only in those subjects less than 65 years of age. The beneficial effects of fenofibrate in this study may have been muted by the increased use of statins in the placebo group, which reduced the differences in lipid levels between the placebo and fenofibrate groups. If one adjusted for the addition of lipid-lowering therapy, fenofibrate reduced the risk of coronary heart disease events by 19% (p=0.01) and of total ASCVD events by 15% (p=0.004). Thus, while the results of this large trial are intriguing they do not clearly show a benefit of fibrate therapy reducing ASCVD events. The number of patients with diabetes in the other fibrate trials are relatively small (table 3).

 

While the results of the monotherapy fibrate trials have been very heterogeneous it should be noted that fibrate trials in patients with elevated TG levels have reported a greater reduction of cardiovascular events (177). Additionally, subgroup analysis of several fibrate trials has also suggested that the benefit of fibrates was greatest in patients with elevated TG levels (177,178).

 

The mechanism by which fibrates may reduce cardiovascular events is unclear. These drugs lower serum TG levels and increase HDL-C, but it should be recognized that the beneficial effects of fibrates could be due to other actions of these drugs. Specifically, these drugs activate the nuclear hormone receptor PPAR alpha, which is present in the cells that comprise the atherosclerotic lesions, and it is possible that these compounds directly affect lesion formation and development. In addition, fibrates are anti-inflammatory. In fact, analysis of the VA-HIT study suggested that much of the benefit of fibrate therapy was not due to changes in serum lipoprotein levels (179,180).

 

To summarize, while in general the studies suggest that monotherapy with fibrates may reduce ASCVD in patients with diabetes, the results are not very robust or consistent as seen in the statin trials. Of note fibrate therapy appeared to be most effective in patients with increased TG levels and decreased HDL levels, a lipid profile typically seen in patients with T2DM. However, as will be presented in detail below (combination therapy section) the addition of fibrates to statins does not reduce ASCVD.

 

Table 3. Effect of Fibrate Monotherapy on Cardiovascular Outcomes

Study

Drug

#Diabetic subjects

%Decrease controls

% Decrease diabetics

Helsinki Heart Study (181)

Gemfibrozil

135

34

60*

VA-HIT (180)

Gemfibrozil

620

24

24

DIAS (182)

Fenofibrate

418

-

23*

Sendcap (183)

Bezafibrate

164

-

70

Field (176)

Fenofibrate

9795

-

11*

* Not statistically significant

 

NIACIN

 

A single randomized trial, the Coronary Drug Project, has examined the effect of niacin monotherapy on cardiovascular outcomes (184). This trial was carried out from 1966 to 1974 (before the introduction of statin therapy) in men with a history of a prior MI and demonstrated that niacin therapy reduced cardiovascular events. The results of this study were re-analyzed to determine the effect of niacin therapy in subjects with varying baseline fasting and 1-hour post meal glucose levels (185). It was noted that 6 years of niacin therapy reduced the risk of coronary heart disease death or nonfatal MI by approximately 15-25% regardless of baseline fasting or 1-hour post glucose challenge glucose levels. Particularly notable is that reductions in events were seen in the subjects who had a fasting glucose level >126mg/dL or 1-hour glucose levels >220mg/dL (i.e., patients with diabetes). Thus, based on this single study, niacin monotherapy reduces cardiovascular events both in normal subjects and patients with diabetes. However, as will be presented in detail below (combination therapy section) the addition of niacin to statins does not reduce ASCVD.

 

EZETIMIBE

 

A multicenter, randomized trial in Japan examined the efficacy of ezetimibe in patients aged ≥75 years with elevated LDL-C (≥140 mg/dL) without a history of coronary artery disease who were not taking lipid lowering drugs (186). Patients were randomized to ezetimibe (n=1716) or usual care (n=1695) and followed for 4.1 years. The primary outcome was a composite of sudden cardiac death, MI, 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. Approximately 25% of the patients in this trial had diabetes and the beneficial effects were similar in the diabetic and non-diabetic subjects. 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 suggestive evidence that ezetimibe monotherapy may reduce cardiovascular events in patients with diabetes.

 

BEMPEDOIC ACID

 

A multicenter study of bempedoic acid in statin intolerant patients with ASCVD or at high risk for ASCVD was recently reported (187). Patients were randomized to bempedoic acid, 180 mg daily (n=6992), or placebo (n=6978) and the primary end point was death from cardiovascular causes, nonfatal MI, nonfatal stroke, or coronary revascularization. Bempedoic acid therapy reduced LDL-C and hsCRP levels by approximately 22% compared to the placebo group. The primary composite endpoint was reduced by 13% in the bempedoic acid group (HR 0.87; 95% CI, 0.79 to 0.96; P = 0.004). The four individual components of the primary endpoint were also significantly reduced in the bempedoic acid treatment group. In this trial approximately 45% of the patients had diabetes. In an analysis of the patients without clinical ASCVD, (i.e., primary prevention) (bempedoic acid n = 2100 or placebo n = 2106), there was a 30% decrease in cardiovascular events (HR 0.70: 95% CI, 0.55-0.89; P = .002) (188). In this subgroup analysis 66% of the patients had diabetes. This study clearly indicates that monotherapy with bempedoic acid will reduce cardiovascular events.

 

OTHER DRUGS

 

With regard to PCSK9 inhibitors and bile acid sequestrants there have been no randomized monotherapy studies that have examined the effect of these drugs on cardiovascular end points in subjects with diabetes. In non-diabetic subjects, monotherapy with bile acid sequestrants have reduced cardiovascular events (102,103). Since bile acid sequestrants have a similar beneficial impact on serum lipid levels in diabetic and non-diabetic subjects one would anticipate that these drugs would also result in a reduction in events in the diabetic population. Additionally, bile acid sequestrants improve glycemic control (101). However, bile acid sequestrants can raise TG levels and therefore must be used with caution in hypertriglyceridemic patients. There are no outcome studies with PCSK9 inhibitor monotherapy in patients with diabetes but given that these drugs reduce LDL-C levels and in combination with statins reduce cardiovascular events one would anticipate that PCSK9 inhibitor monotherapy will also reduce cardiovascular events.

 

Combination Therapy

 

The studies with statins have been so impressive that most patients with diabetes over the age of 40 are routinely treated with statin therapy and younger patients with diabetes at high risk for ASCVD are also typically on statin therapy (see Current Guidelines Section). Therefore, a key issue is whether the addition of other lipid lowering drugs to statins will result in a further reduction in cardiovascular events. A difficulty with such studies is that the reduction in cardiovascular events induced by statin therapy is so robust that very large trials may be required to see additional benefit.

 

STATINS + FIBRATES

 

The ACCORD-LIPID trial was designed to determine if the addition of fenofibrate to aggressive statin therapy would result in a further reduction in ASCVD in patients with T2DM (189). In this trial, 5,518 patients on statin therapy were randomized to placebo or fenofibrate therapy. The patients had diabetes for approximately 10 years and either had pre-existing ASCVD or were at high risk for developing ASCVD. During the trial, LDL-C levels were approximately 80mg/dL in both groups. There was only a small difference in HDL-C with the fenofibrate groups having a mean HDL-C of 41.2mg/dL while the control group had an HDL-C of 40.5mg/dL. Differences in TG levels were somewhat more impressive with the fenofibrate group having a mean TG level of 122mg/dL while the control group had a TG level of 144mg/dL. First occurrence of nonfatal MI, nonfatal stroke, or death from cardiovascular causes was the primary outcome and there was no statistical difference between the fenofibrate treated group and the placebo group. Additionally, there were also no statistically significant differences between the groups with regards to any of the secondary outcome measures of ASCVD. Of note, the addition of fenofibrate to statin therapy did not result in an increase in either muscle or liver side effects. On further analysis, there was a possible benefit of fenofibrate therapy in the patients in whom the baseline TG levels were elevated (>204mg/dL) and HDL-C levels decreased (<34mg/dL). Finally, similar to what has been reported in other trials, fenofibrate had beneficial effects on the progression of microvascular disease (190,191).

 

The PROMINENT trial studied the effect of pemafibrate, a new selective PPAR-alpha activator, in reducing cardiovascular outcomes in 10,497 patients (66.9% with previous ASCVD) with diabetes (192). This was a double-blind, randomized, controlled trial, in patients with T2DM, with mild-to-moderate hypertriglyceridemia (TG level, 200 to 499 mg/dL), LDL-C < 100mg/dL, and HDL-C levels < 40 mg/dL) who received either pemafibrate (0.2-mg tablets twice daily) or placebo in addition to statin therapy (96% on statins). The primary end point was a composite of nonfatal MI, ischemic stroke, coronary revascularization, or death from cardiovascular causes. Baseline fasting TG was 271 mg/dL, HDL-C 33 mg/dL, and LDL-C 78 mg/dL. Compared with placebo, pemafibrate decreased TG by 26.2%, while HDL-C increased 5.1% and LDL-C increased 12.3%. Notably non-HDL-C levels were unchanged and Apo B levels increased 4.8%. The primary endpoint was similar in the pemafibrate and placebo group (HR 1.03; 95% CI 0.91 to 1.15). The increase in LDL-C and Apo B levels likely account for the failure to reduce cardiovascular events.

 

Taken together the ACCORD study and the PROINENT trial indicate that the addition of fibrate therapy to statin therapy will not result in a reduction in cardiovascular events in patients with diabetes.

 

STATIN + NIACIN

 

The AIM-HIGH trial was designed to determine if the addition of Niaspan to aggressive statin therapy would result in a further reduction in cardiovascular events in patients with pre-existing ASCVD (193). In this trial 3,314 patients were randomized to Niaspan vs. placebo. Approximately 33% of the patients had diabetes. On trial, LDL-C levels were in the 60-70mg/dL range in both groups. As expected, HDL-C levels were increased in the Niaspan treated group (approximately 44mg/dL vs. 38mg/dL), while TG were decreased (approximately 121mg/dL vs. 155mg/dL). However, there were no differences in the primary endpoint between the control and Niaspan treated groups (Primary endpoint consisted of the first event of death from coronary heart disease, nonfatal MI, ischemic stroke, hospitalization for an acute coronary syndrome, or symptom-driven coronary or cerebral revascularization). There were also no differences in secondary endpoints except for a possible increase in strokes in the Niaspan treated group. The addition of Niaspan to statin therapy did not result in a significant increase in either muscle or liver toxicity. Thus, this study does not provide support for the addition of niacin to statins. However, it should be recognized that this was a relatively small study and a considerable number of patients stopped taking the Niaspan during the course of the study (25.4% of patients discontinued Niaspan therapy). In addition, most of the patients included in this study did not have a lipid profile that one would typically consider treating with niacin therapy. In the subset of patients with TG > 198mg/dL and HDL-C < 33mg/dL niacin showed a trend towards benefit (hazard ratio 0.74; p=0.073), suggesting that if the appropriate patient population was studied the results may have been positive (194).

 

HPS 2 Thrive also studied the effect of niacin added to statin therapy (195). This trial utilized extended-release niacin combined with laropiprant, a prostaglandin D2 receptor antagonist that reduces the flushing side effect of niacin treatment. HPS 2 Thrive was a very large trial with over 25,000 patients randomized to either niacin therapy or placebo. Approximately 32% of the patients in this trial had diabetes. The LDL-C level was 63mg/dL, the HDL-C 44mg/dL, and the TG 125mg/dL at baseline. As expected, niacin therapy resulted in a modest reduction in LDL-C (10mg/dL), a modest increase in HDL-C (6mg/dL), and a larger reduction in TG (33mg/dL). However, despite these lipid changes there were no significant differences in major cardiovascular events between the niacin and control group (risk ratio 0.96 CI 0.90- 1.03). It is unknown whether laropiprant, the prostaglandin D2 receptor antagonist, might have effects that worsen atherosclerosis and increase event rates. Similar to the AIM-HIGH study, the group of patients included in the HPS 2 Thrive trial were not the ideal patient population to test for the beneficial effects of niacin treatment added to statin therapy. Ideally, patients with high TG and high non-HDL-C levels coupled with low HDL-C levels should be studied. Nevertheless, the results of the AIM-HIGH and HPS 2 Thrive trials do not provide support for the addition of niacin to statin therapy in patients with diabetes.

 

STATIN + EZETIMIBE

 

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 (196). This was a large trial with over 18,000 patients randomized to statin therapy vs. statin therapy + ezetimibe. Approximately 27% of the patients in this trial had diabetes. On treatment LDL-C levels were 70mg/dL in the statin alone group vs. 53mg/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 re-hospitalization, 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). These beneficial effects were particularly pronounced in the patients with diabetes (Primary endpoint hazard ratio, 0.85; 95% confidence interval, 0.78-0.94) (197,198). This trial provides evidence that the combination of a statin + ezetimibe that results in a greater reduction in LDL-C levels will lead to a larger decrease in cardiovascular events than statin alone. It should be noted that the observed reduction in events was in the range expected based on the decrease in LDL-C levels.

 

The RACING trial compared rosuvastatin 10 mg plus ezetimibe 10 mg (combination therapy) vs. rosuvastatin 20mg in 3,780 patients (1,398 patients (37.0%) with diabetes) at 26 centers in South Korea (199). In the patients with diabetes the baseline LDL-C levels was 74mg/dL and during the study the median LDL-C was 53mg/dL in the combination therapy group and 61mg/dL in the high-intensity statin group (P < 0.001). After a median follow-up of 3 years the rate of cardiovascular events in patients with diabetes was 10.0% in the combination therapy group and 11.3% in the high-intensity statin group (HR: 0.89; 95% CI: 0.64–1.22; P = 0.460). Interestingly the rate of discontinuation or dose reduction of the study drug due to intolerance was lower in the combination therapy group than in the high-intensity statin group (5.2 vs. 8.7%; P = 0.014). This study demonstrates that cardiovascular outcomes were comparable between those receiving combination therapy vs. high-intensity statin monotherapy and that combination therapy significantly reduced the rate of drug discontinuation or dose reduction due to intolerance.

 

STATIN + PCSK9 INHIBITORS

 

The FOURIER trial was a randomized, double-blind, placebo-controlled trial of evolocumab vs. placebo in 27,564 patients with atherosclerotic ASCVD and an LDL-C level of 70 mg/dL or higher who were on statin therapy (200). Approximately 40% of the patients had diabetes (201). The primary end point was cardiovascular death, MI, stroke, hospitalization for unstable angina, or coronary revascularization and the key secondary end point was cardiovascular death, MI, 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-C levels (LDL-C level on treatment approximately 30mg/dL). Evolocumab treatment significantly reduced the risk of the primary end point (HR 0.85; 95% CI 0.79 to 0.92; P<0.001) and the key secondary end point (HR 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 (202). 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 (203). Finally, 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 ASCVD (204).

 

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 Apo B level of at least 80 mg/dL while on high intensity statin therapy or the maximum tolerated statin dose (205). Approximately 29% of the patients had diabetes. 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 MI, 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 > 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 similar to the FOURIER trial 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).

 

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 takes time with only modest effects observed during the first year of treatment. In the FOURIER trial the reduction of cardiovascular death, MI, 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.

 

Additional support for the benefits of further lowering of LDL-C levels with a PCSK9 inhibitor added to statin therapy is seen in the GLAGOV trial (206). This trial was a double-blind, placebo-controlled, randomized trial of evolocumab vs. placebo in 968 patients presenting for coronary angiography. Approximately 20-21% of the patients had diabetes. 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). The results in the patients with diabetes were similar to the non-diabetic patients.

 

Taken together these trials demonstrate that further lowering LDL-C levels with PCSK9 monoclonal antibodies in patients taking statins will have beneficial effects on ASCVD outcomes. A study of the effect of inclisiran on ASCVD endpoints is currently in progress.

 

The results of the ezetimibe and PCSK9 trials have several important implications. First, it demonstrates that combination therapy may have benefits above and beyond statin therapy alone. Second, it provides further support for the hypothesis that lowering LDL per se will reduce cardiovascular events. Third, it suggests that lowering LDL levels to much lower levels than usual will have significant benefits. These results have implications for determining goals of therapy.

 

STATINS + LOW DOSE OMEGA-3-FATTY ACIDS

 

ORIGIN was a double-blind study in 12,536 patients at high risk for ASCVD who had impaired fasting glucose, impaired glucose tolerance, or diabetes (207).  Patients were randomized to receive a 1-gram capsule containing at least 900mg of ethyl esters of omega-3 fatty acids (EPA 465mg and DHA 375mg) or placebo for approximately 6 years. Greater than 50% of the patients were on statin therapy. The primary outcome was death from cardiovascular causes. TG levels were reduced by 14.5mg/dL in the group receiving omega-3-fatty acids compared to the placebo group (P<0.001), without a significant effect on other lipids. The incidence of the primary outcome was not significantly decreased among patients receiving omega-3-fatty acids as compared with those receiving placebo. The use of omega-3-fatty acids also had no significant effect on the rates of major vascular events, death from any cause, or death from arrhythmia.

 

A Study of Cardiovascular Events in Diabetes (ASCEND) was a randomized, placebo controlled, double blind trial of 1-gram omega-3-fattys acids (400mg EPA and 300mg DHA ethyl esters) vs. olive oil placebo in 15,480 patients with diabetes without a history of ASCVD (primary prevention trial) (208). Approximately 75% of patients were on statin therapy. The primary end point was serious vascular events (non-fatal MI, non-fatal stroke, transient ischemic attack, or vascular death). Total cholesterol, HDL-C, and non-HDL-C levels were not significantly altered by omega-3-fatty acid treatment (changes in TG levels were not reported). After a mean follow-up of 7.4 years the composite outcome of a serious vascular event or revascularization occurred in 882 patients (11.4%) on omega-3-fatty acids and 887 patients (11.5%) on placebo (rate ratio, 1.00; 95% CI, 0.91 to 1.09). Serious adverse events were similar in placebo and omega-3-fatty acid treated groups.

 

Taken together these studies indicate that low dose omega-3-fatty acids do not reduce cardiovascular events in patients with diabetes. Studies in non-diabetics have also found little effect of low dose omega-3-fatty acids on ASCVD (209).

 

STATINS + HIGH DOSE OMEGA-3-FATTY ACIDS

 

The Japan EPA Lipid Intervention Study (JELIS) was an open label non-placebo controlled study in patients on statin therapy with total cholesterol levels > 254mg/dL with (n= 3664) or without ASCVD (n=14,981) who were randomly assigned to be treated with 1800 mg of EPA (Vascepa) + statin (n=9326) or statin alone (n= 9319) with a 5 year follow-up (210). Approximately 16% of the patients had diabetes. The mean baseline TG level was 153mg/dL. The primary endpoint was any major coronary event, including sudden cardiac death, fatal and non-fatal MI, and other non-fatal events including unstable angina pectoris, angioplasty, stenting, or coronary artery bypass grafting. On treatment total cholesterol, LDL-C, and HDL-C levels were similar in the two groups but plasma TG were modestly decreased in the EPA treated group (5% decrease in EPA group compared to controls; p = 0.0001). In the EPA + 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). Unstable angina and non-fatal coronary events were also significantly reduced in the EPA 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 MI, stroke, or cardiovascular death. A subjective endpoint has the potential to be an unreliable endpoint in an open label study and is a major limitation of the JELIS Study. The reduction in events was similar in the subgroup of patients with diabetes. In patients with TG levels >150mg/dL and HDL-C levels < 40mg/dL there was a 53% decrease in events (211). In the EPA group, small increases in the occurrence of bleeding (1.1% vs. 0.6%, p=0.0006), gastrointestinal disturbance (3.8%% vs. 1.7%, p<0.0001) and skin abnormalities (1.7 vs. 0.7%, p<0.0001) were seen. 

 

The Reduction of Cardiovascular Events with EPA – Intervention Trial (REDUCE-IT) was a randomized, double blind trial of 2 grams twice per day of EPA ethyl ester (icosapent ethyl) (Vascepa) vs. placebo (mineral oil) in 8,179 patients with hypertriglyceridemia (135mg/dL to 499mg/dL) and established ASCVD or high ASCVD risk (diabetes plus one risk factor) who were on stable statin therapy (212). Approximately 60% of the patients in this trial had diabetes. The primary end point was a composite of cardiovascular death, nonfatal MI, 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; 95% CI 0.68 to 0.83; P<0.001), indicating a 25% decrease in events. The beneficial effects were similar in patients with and without diabetes. The number needed to treat to avoid one primary end-point event was 21. The reduction in cardiovascular events was noted after approximately 2 years of EPA treatment. Additionally, the risk of cardiovascular death was decreased by 20% in the EPA group (HR 0.80; 95% CI, 0.66 to 0.98; P=0.03). The cardiovascular benefits of EPA were similar across baseline levels of TG (<150, ≥150 to <200, and ≥200 mg/dL). 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 cardiovascular risk reduction was not associated with attainment of a normal TG levels. An increase in hospitalization for atrial fibrillation or flutter (3.1% vs. 2.1%, P=0.004) occurred in the EPA group. In addition, serious bleeding events occurred in 2.7% of the patients in the EPA group and in 2.1% in the placebo group (P=0.06). 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.

 

These results demonstrate that EPA treatment reduces ASCVD events. Of note the reduction in TG levels is relatively modest and would not be expected to result in the magnitude of the decrease in ASCVD observed in the JELIS and REDUCE-IT trials. 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 cardiovascular events (213). It is likely that the beneficial effects of EPA seen in the JELIS and REDUCE-IT trials are multifactorial.

 

The Statin Residual Risk Reduction with Epanova in High Risk Patients with Hypertriglyceridemia (STRENGTH) trial was a randomized, placebo controlled, double blind trial of 4 grams per day of omega-3-fatty acids (Epanova) (carboxylic acid formulation of EPA and DHA) vs. placebo (corn oil) in 13,000 patients on statins with hypertriglyceridemia (180-500mg/dL), optimal LDL-C levels (< 100mg/dL or on maximal statin therapy), low HDL-C (<42mg/dL in men and < 47mg/dL in women), and either ASCVD or high risk for ASCVD (214). The primary outcome was major atherosclerotic cardiovascular events (cardiovascular death, MI, stroke, coronary revascularization or hospitalization for unstable angina). The primary end point occurred in 785 patients (12.0%) treated with omega-3 CA vs 795 (12.2%) treated with corn oil (HR, 0.99: [95% CI, 0.90-1.09]; P = .84) (215). Thus, in contrast to EPA alone this omega-3-fatty acid formulation failed to show benefits despite reducing TG levels (18% decrease) to a similar degree as in the REDUCE-IT trial.

 

Whether EPA has special properties that resulted in the reduction in cardiovascular events in the REDUCE-IT trial or there were flaws in the trial design (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% (LDL-C by approximately 9mg/dL and non-HDL-C by approximately 10mg/dL) in the mineral oil placebo group (212). Additionally, Apo B levels were increased by 7% (6mg/dL) by mineral oil (212). 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 (212,216). 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 (215). 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 (217,218). Ideally, another large randomized cardiovascular trial with EPA ethyl ester (icosapent ethyl) (Vascepa) using a placebo other than mineral oil would resolve this controversy.

 

CURRENT GUIDELINES FOR SERUM LIPIDS

 

There are several different guidelines for treating lipids in patients with diabetes. While they all focus on lowering LDL-C there are differences between the various guidelines.

 

American Diabetes Association Guidelines

 

The 2023 American Diabetes Association (ADA) recommends that adult patients with diabetes have their lipid profile determined at the time of diabetes diagnosis and at least every 5 years thereafter or more frequently if indicated (219). This profile includes total cholesterol, HDL-C, TG, and calculated LDL-C. A lipid panel should be obtained immediately prior to initiating statin therapy. Once a patient is on statin therapy testing should be carried out 4-12 weeks after initiating therapy and annually thereafter to monitor adherence and efficacy. Lifestyle modifications including a reduction in saturated fat, trans fat, and cholesterol intake, weight loss if indicated, an increase in omega-3-fatty acids, viscous fiber, and plant stanols /sterol intake, and increased physical activity is indicated in all patients with diabetes. A focus on a Mediterranean style diet or Dietary Approaches to Stop Hypertension (DASH) diet should be encouraged. In patients with elevated TG levels glycemic control is beneficial and dietary changes and lifestyle changes including weight loss and abstinence from alcohol should be undertaken. Secondary disorders and medications that raise TG levels should be evaluated. Optimize glycemic control to improve TG and HDL-C levels. The recommendations for lipid lowering therapy are shown in table 4. If one follows these recommendations almost all patients with diabetes over the age of 40 will be on statin therapy and many under the age of 40 will also be treated with statins. The addition of ezetimibe should be considered to further lower LDL-C levels in high-risk primary prevention patients. In very high-risk patients with ASCVD if the LDL-C level on statin therapy is greater than 70mg/dL the use of ezetimibe or a PCSK9 inhibitor should be considered. The use of fibrates or niacin with statins were generally not recommended as there is no evidence of benefit. However, in patients with ASCVD or other cardiovascular risk factors on a statin with controlled LDL-C but elevated TG levels (135-499mg/dL) the addition of icosapent ethyl can be considered. Finally, in patients with fasting TG levels greater than 500mg/dL an evaluation for secondary causes of hypertriglyceridemia should be initiated and consideration of drug therapy to reduce the risk of pancreatitis.

 

Table 4. ADA Recommendations for Lipid Lowering Therapy

Primary Prevention

Age 20-39: With additional risk factors may be reasonable to initiate statin therapy

Age 40-75: Use moderate-intensity statin therapy* in addition to lifestyle therapy

Age 40-75: If at higher cardiovascular risk, including those with one or more ASCVD risk factors, it is recommended to use high intensity statin therapy to reduce LDL cholesterol by >50% and to target an LDL-C <70 mg/dL

Age 40-75: If at higher cardiovascular risk, especially those with multiple ASCVD risk factors and an LDL-C >70 mg/dL, it may be reasonable to add ezetimibe or a PCSK9 inhibitor to maximum tolerated statin therapy

Age > 75: Initiating moderate intensity statin therapy is reasonable after discussion and in patient already on statin therapy it is reasonable to continue statin therapy

Secondary Prevention

All ages: High intensity statin therapy**/maximally tolerated stain

For people with diabetes and ASCVD, treatment with high intensity statin therapy is recommended to target an LDL-C reduction of >50% and an LDL-C l goal of <55 mg/dL. Addition of ezetimibe or a PCSK9 inhibitor is recommended if this goal is not achieved

*Moderate intensity statin- atorvastatin 10-20mg, rosuvastatin 5-10mg, simvastatin 20-40mg, pravastatin 40-80mg, lovastatin 40mg, Fluvastatin XL 80mg, pitavastatin 3-4mg.

**High Intensity statin- atorvastatin 40-80mg, rosuvastatin 20-40mg.

 

American College of Cardiology and American Heart Association Guidelines

 

The 2018 American College of Cardiology and American Heart Association (ACC/AHA) guidelines recommend the following (220). “In patients 40 to 75 years of age with diabetes mellitus and LDL-C ≥70 mg/dL (≥1.8 mmol/L), start moderate-intensity statin therapy without calculating 10-year ASCVD risk. In patients with diabetes mellitus at higher risk, especially those with multiple risk factors or those 50 to 75 years of age, it is reasonable to use a high-intensity statin to reduce the LDL-C level by ≥50%.” In patients with diabetes and ASCVD they recommend “In patients with clinical ASCVD, reduce LDL-C with high-intensity statin therapy or maximally tolerated statin therapy. The more LDL-C is reduced on statin therapy, the greater will be subsequent risk reduction. Use a maximally tolerated statin to lower LDLC levels by ≥50%. In very high-risk ASCVD, use an LDL-C threshold of 70 mg/dL (1.8 mmol/L) to consider addition of non-statins to statin therapy. Very high-risk includes a history of multiple major ASCVD events or 1 major ASCVD event and multiple high-risk conditions. In very high-risk ASCVD patients, it is reasonable to add ezetimibe to maximally tolerated statin therapy when the LDL-C level remains ≥70 mg/dL (≥1.8 mmol/L). In patients at very high risk whose LDL-C level remains ≥70 mg/dL (≥1.8 mmol/L) on maximally tolerated statin and ezetimibe therapy, adding a PCSK9 inhibitor is reasonable, although the long-term safety (>3 years) is uncertain and cost effectiveness is low at mid-2018 list prices.” With regards to testing they recommend “Assess adherence and percentage response to LDL-C–lowering medications and lifestyle changes with repeat lipid measurement 4 to 12 weeks after statin initiation or dose adjustment, repeated every 3 to 12 months as needed”. Finally, there are several diabetes specific risk enhancers that are independent of other risk factors that should be considered in deciding the risk of cardiovascular events in a patient with diabetes (Table 5).

 

Table 5. Diabetes Specific Risk Enhancers That are Independent of Other Risk Factors in Diabetes

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

Albuminuria ≥30 mcg of albumin/mg creatinine

eGFR <60 mL/min/1.73 m2

Retinopathy

Neuropathy

ABI <0.9

ABI indicates ankle-brachial index

 

American Association of Clinical Endocrinologists/American College of Endocrinology Guidelines

 

The American Association of Clinical Endocrinologists and American College of Endocrinology guidelines consider individuals with T2DM to be at high, very high, or extreme risk for ASCVD (221,222). Patients with T1DM and a duration of diabetes of more than 15 years or two or more risk factors, poorly controlled A1c, or insulin resistance with metabolic syndrome should be considered to have an equivalent risk to patients with T2DM (221). The recommended treatment goals are shown in Table 6.

 

Table 6. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dL

Non-HDL-C mg/dL

Apo B mg/dL

TG

mg/dL

Extreme Risk

Diabetes and clinical ASCVD

<55

<80

<70

<150

Very High Risk

Diabetes with one or more risk factors

<70

<100

<80

<150

High Risk

Diabetes and no other risk factors

<100

<130

<90

<150

 

European Society of Cardiology and European Atherosclerosis Society Guidelines

 

The European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS) has 2019 guidelines for the treatment of lipids in patients with diabetes (223). These guidelines classify patients with diabetes as very high risk, high risk, or moderate risk (table 7). The recommended goals of therapy based on risk classification are shown in table 8. As with other guidelines intensification of statin therapy should be considered before the introduction of combination therapy. If the goal is not reached, statin combination with ezetimibe should be considered next.

 

Table 7. ESC/EAS Classification of Risk in Patients with Diabetes

Very High Risk- target organ damage, or at least three major risk factors, or early onset of T1DM of long duration (>20 years)

High Risk- without target organ damage, with DM duration >10 years or another additional risk factor

Moderate Risk- Young patients (T1DM <35 years; T2DM <50 years) with DM duration <10 years, without other risk factors. Calculated SCORE >1 % and <5% for 10-year risk of fatal CVD

 

Table 8. ESC/EAS Goals of Therapy in Patients with Diabetes

 

LDL-C

Non-HDL-C

Apo B

Very High Risk

>50% reduction and <55mg/dL (<1.4mmol/L)

<85mg/d;

<65mg/dL

High Risk

>50% reduction and <70mg/dL (<1.8mmol/L)

<100mg/dL

<80mg/dL

Moderate Risk

<100mg/dL

<130mg/dL

<100mg/dL

 

European Society of Cardiology Guidelines

 

The ESC has updated their guidelines in 2023 (224). An important new recommendation is that in patients with T2DM without symptomatic ASCVD or severe target organ damage, it is recommended to estimate 10-year CVD risk using SCORE2-Diabetes (225). This has resulted in a new classification of risk in patients with T2DM (table 9). The LDL-C goals for each category are shown in table 9.

 

Table 9. Cardiovascular Risk Categories and Goals in Patients with Type 2 Diabetes

Very high risk

Clinically established ASCVD or

Severe target organ damage or

10-year risk of CVD> 20% using SCORE2-Diabetes

LDL-C < 55mg/dl

Non-HDL-C <85mg/dL

High risk*

10-year risk of CVD 10% to < 20% using SCORE2-Diabetes

LDL-C < 70mg/dL

Non-HDL-C <100mg/dL

Moderate risk*

10-year risk of CVD 5% to <10% using SCORE2-Diabetes

LDL-C < 100mg/dL

Low Risk*

10-year risk of CVD <5% using SCORE2-Diabetes

no recommendations

*Patients not meeting the very high-risk category.

Severe target organ damage is defined as eGFR <45 mL/min/1.73 m2 irrespective of albuminuria; or eGFR 45–59 mL/min/1.73 m2 and microalbuminuria (UACR 30–300 mg/g; stage A2); or proteinuria (UACR>300 mg/g; stage A3), or presence of microvascular disease in at least three different sites [e.g., microalbuminuria (stage A2) plus retinopathy plus neuropathy].

 

My Guideline Recommendations

 

Thus, different organizations have proposed somewhat different recommendations for the treatment of lipids in patients with diabetes. Despite these differences it is clear that the vast majority of patients with diabetes will need to be treated with statins regardless of which guidelines one elects to follow.

 

The approach I use is to combine these recommendations (Tables 10 and 11). In patients with diabetes who have pre-existing ASCVD I initiate intensive statin therapy. I prefer LDL or non-HDL-C goals over percent reduction goals. Given the extensive data showing that the lower the LDL-C the greater the reduction in cardiovascular events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering without markedly increasing costs (226). In patients with diabetes 40-75 years of age without pre-existing ASCVD I calculate the 10-year risk of developing ASCVD (http://www.cvriskcalculator.com/) and identify risk enhancing factors (Table 5) and other factors that increase risk that are not included in the calculator (for example family history, inflammatory disorders, etc.). I initiate intensive statin therapy if the 10-year risk is > 7.5% or if there are multiple risk factors/risk enhancers. I initiate moderate statin therapy if the risk is < 7.5% without multiple risk factors/enhancers. Four to twelve weeks after initiating statin therapy I obtain a lipid panel to determine if the LDL and non-HDL-C levels are at goal. In patients with pre-existing ASCVD or multiple risk factors/risk enhancers (i.e., very high-risk patients) my goal is an LDL-C < 55mg/dL and a non-HDL-C < 80mg/dL. In patients that are at high-risk the goal my goal is an LDL-C < 70mg/dL and a non-HDL-C < 100mg/dL. In patients with moderate risk an LDL-C goal of < 100mg/dL and a non-HDL c < 130mg/dL is appropriate. If the levels are not at goal, I first adjust the statin dose until the patient is taking the maximally tolerated statin dose and then consider adding additional medications.  In patients with diabetes who are less than 40 years of age I initiate statin therapy if the patient has overt ASCVD, long standing diabetes, or risk factors/risk enhancers for ASCVD and the LDL and non-HDL-C levels are not at goal. In these younger patients I also calculate the life time risk of ASCVD events to start a discussion of beginning early therapy given the abundance of data indicating that initiating LDL-C lowering therapy early has great potential in markedly lowering ASCVD risk (226,227). In patients over 75 years of age with a reasonable life expectancy I begin moderate statin therapy and adjust based on response. When there is difficulty classifying a patient’s risk, I will obtain a coronary calcium score and use the score to help stratify the patient’s risk. In all cases the benefits and risks of lipid lowering therapy needs to be discussed with patients and the patient’s personnel preferences taken into account.

 

Table 10. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dL

Non-HDL-C mg/dL

Very High Risk

Diabetes and clinical ASCVD, multiple risk factors, 10-year risk > 20%

<55

<85

High Risk

Diabetes with one or more risk factors, 10-year risk >7.5% to <20%

<70

<100

Moderate Risk

Diabetes and no other risk factors. 10-year risk <7.5%

<100

<130

 

Table 11. Drug Therapy According to Risk Category that is Typically Required

Very High Risk

Intensive statin therapy + ezetimibe. Add PCSK9 if not close to goal

High Risk

Intensive statin therapy. Add ezetimibe if not at goal

Moderate Risk

Moderate statin therapy. Increase to intensive statin therapy if not at goal

 

TREATMENT OF LIPID ABNORMALITIES IN PATIENT WITH DIABETES

 

Life Style Changes and Weight Loss

 

Initial treatment of lipid disorders should focus on lifestyle changes. There is little debate that exercise is beneficial and that all patients with diabetes should, if possible, exercise for at least 150 minutes per week (for example 30 minutes 5 times per week). Exercise will decrease serum TG levels and increase HDL-C levels (an increase in HDL-C requires vigorous exercise) (124). Exercise increases fitness and improves insulin resistance even with limited weight loss; reductions in obesity are even more beneficial. It should be noted that many patients with diabetes may have substantial barriers to participating in exercise programs, such as comorbidities that limit exercise tolerance, risk of hypoglycemia, and presence of microvascular complications (visual impairment, neuropathy) that make exercise difficult.

 

Diet is debated to a greater extent and for detailed information on nutrition therapy for adults with diabetes see the consensus report by the American Diabetes Association (228). Everyone agrees that weight loss in obese patients is essential (124). But how this can be achieved is hotly debated with many different "experts" advocating different dietary approaches. The wide diversity of approach is likely due to the failure of any approach to be effective in the long term for the majority of obese patients with diabetes. If successful, weight loss will decrease serum TG levels, increase HDL-C levels, and modestly reduce LDL-C (124,229). To reduce LDL-C levels, it is important that the diet decrease saturated fat, trans fatty acids, and cholesterol intake. Increasing soluble fiber is also helpful.

 

It is debated whether a low fat, high complex carbohydrate diets vs. a high monounsaturated fat  diet is ideal for obese patients with diabetes (124). One can find "experts" in favor of either of these approaches and there are pros and cons to each approach. It is essential to recognize that both approaches reduce simple sugars, saturated fat, trans fatty acids, and cholesterol intake. The high complex carbohydrate diet will increase serum TG levels in some patients and if the amount of fat in the diet is markedly reduced serum HDL-C levels may decrease. In obese patients, it has been postulated that a diet high in monounsaturated fats, because of the increase in caloric density, will lead to an increase in weight gain. Both diets reduce saturated fat and cholesterol intake that will result in reductions in LDL-C levels. Additionally, both diets also reduce trans-fatty acid intake, which will have a beneficial effect on LDL and HDL-C levels and simple sugars, which will have a beneficial effect on TG levels. Very high levels of TG (>1000mg/dL), require diets that are very low in fat.

 

The available data do not indicate that any particular diet is best for inducing weight loss and it is essential to adapt the diet to fit the food preferences of the patient. Ultimately no weight loss diet will be successful if the patient cannot follow the diet for the long term and therefore the diet needs to be tailored to the specific preferences of the patient. For more detailed information on the effect of diet on lipid and lipoprotein levels and cardiovascular disease see the Endotext chapter “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” (229).

 

While it is widely accepted that lifestyle changes will decrease ASCVD events it should be recognized that the Look Ahead trial failed to demonstrate a reduction in ASCVD events (230). In this trial, over 5,000 overweight or obese patients with T2DM were randomized to either an intensive lifestyle intervention group that promoted weight loss through decreased caloric intake and increased physical activity or to a group that received diabetes support and education (control group). After a median follow-up of 9.6 years there was no difference in cardiovascular events (hazard ratio in the intervention group, 0.95; 95% CI 0.83 to 1.09; P=0.51). A major limitation of this study was that while the weight difference between groups was impressive during the first year of the trial, over time the differences greatly narrowed such that at the end of the trial the intensive group had a 6.0% weight loss while the control group had a 3.5% weight loss. This very modest weight difference demonstrates the difficulty in sustaining long term lifestyle changes. Thus, while weight loss and diet therapy are likely to be beneficial in reducing cardiovascular events, in clinical practice they are seldom sufficient because long-term life style changes are very difficult for most patients to maintain.

 

In contrast to the failure of lifestyle therapy in the Look Ahead trial to reduce cardiovascular events, the PREDIMED trial employing a Mediterranean diet (increased monounsaturated fats) did reduce the incidence of major ASCVD events (231,232). In this multicenter trial center 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. Approximately 50% of the patients in this trial had T2DM. 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 with and without diabetes. 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 (233).

 

The CORDIOPREV study was a single center randomized trial that compared a Mediterranean diet to a low-fat diet in 1,002 patients with ASCVD (234). Approximately 50% of the patients had diabetes. 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 is unknown.

 

Finally, another secondary prevention trial of a Mediterranean diet has also demonstrated a reduction in cardiovascular events. The Lyon Diet Heart Study randomized 584 patients who had a MI within 6 months to a Mediterranean type diet vs usual diet (235,236). There was a marked reduction in events in the group of patients randomized to the Mediterranean diet (cardiac death and nonfatal MI rate was 4.07 per 100 patient years in the control diet vs. 1.24 in the Mediterranean diet; p<0.0001). Unfortunately, there is no indication of the number of patients with diabetes in the Lyon Diet Heart Study or whether patients with diabetes responded similar to the entire group. Lipid levels were similar in both groups in this trial (235).

 

The results of these three randomized trials indicate that we should be encouraging our patients to follow a Mediterranean type diet. 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.

 

For additional information on the effect of diets on lipid levels and ASCVD see the chapter entitled “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” in the Lipids and Lipoproteins section of Endotext (229).

 

Bariatric surgery can have profound effects on weight and can result in marked improvements in lipid profiles with a decrease in TG and LDL-C and an increase in HDL-C (124,229) Additionally, observational studies have shown a decrease in cardiovascular events following bariatric surgery in patients with and without diabetes (237-241). For additional information see the chapter entitled “Obesity and Dyslipidemia” (124).

 

Ethanol and simple sugars, in particular fructose, increase serum TG levels in susceptible patients. In patients with hypertriglyceridemia efforts should be made to reduce the intake of ethanol, simple sugars, and fructose (229).

 

Lastly, in the past some "experts" advocated the addition of fish oil supplements to reduce cardiovascular events. However, both the Origin Trial and the ASCEND Trial did not demonstrate that fish oil supplements were beneficial in patients with T2DM or patients at high risk for the development of T2DM (207,208). It should be recognized that higher doses of fish oil are required to lower serum triglyceride levels (~ 3-4 grams of DHA/EPA per day) and are useful in treating patients with high TG levels (242). Most studies of fish oil supplements in patients with diabetes have demonstrated that this is a safe approach and that worsening of glycemic control does not occur in patients with diabetes treated with fish oil supplements (242). Additionally, in some patient's high dose fish oil increases LDL-C levels, particularly when serum TG levels are very high (242). For additional information on fish oil see the chapter on Triglyceride Lowering Drugs (209).

 

Drug Therapy

 

The effect of statins, fibrates, niacin, ezetimibe, omega-3-fatty acids, bile acid sequestrants, bempedoic acid, and PCSK9 inhibitors on lipid levels in patients with diabetes is virtually identical to that seen in non-diabetic patients (Table 12). Below we will highlight issues particularly relevant to the use of these drugs in patients with diabetes. For detailed information on lipid lowering drugs see the chapters on Triglyceride Lowering Drugs and Cholesterol Lowering Drugs (141,209).

 

STATINS

 

Statins are easy to use and generally well tolerated by patients with diabetes. However, statins can adversely affect glucose homeostasis. In patients without diabetes the risk of developing diabetes is increased by approximately 10% with higher doses of statin causing a greater risk than more moderate doses (243,244). The mechanism for this adverse effect is unknown but older, obese patients with higher baseline glucose levels are at greatest risk. 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 HbA1c than the placebo group indicating that statin therapy is associated with only a very small increase in HbA1c levels in patients with diabetes, which is unlikely to be clinically significant (245). Individual studies such as CARDS and the Heart Protection Study have also shown only a very modest effect of statins on HbA1c levels in patients with diabetes (163,166,246). Muscle symptoms occur in patients with diabetes similar to what is observed in patients without diabetes.

 

EZETIMIBE

 

Ezetimibe is easy to use and generally well tolerated by patients with diabetes. Ezetimibe does not appear to increase the risk of new onset diabetes (199,247,248).

 

FIBRATES

 

Fibrates are easy to use and generally well tolerated by patients with diabetes. When combining fibrates with statin therapy it is best to use fenofibrate as the risk of inducing myositis is much less than when statins are used in combination with gemfibrozil, which can inhibit statin metabolism (249). In the ACCORD-LIPID Trial the incidence of muscle disorders was not increased in the statin + fenofibrate group compared to statin alone (189). The dose of fenofibrate needs to be adjusted in patients with renal disease and fenofibrate itself can induce a reversible increase in serum creatinine levels. It should be noted that marked reductions in HDL-C levels can occur in some patients treated with both fenofibrate and a TZD (250).

 

Diabetic Retinopathy

 

Fenofibrate has been shown to have beneficial effects on diabetic eye disease. The FIELD study, described earlier, was a randomized trial of fenofibrate vs. placebo in patients with T2DM. Laser treatment for retinopathy was significantly lower in the fenofibrate group than in the placebo group (3.4% patients on fenofibrate vs 4.9% on placebo; p=0.0002) (191). Fenofibrate therapy reduced the need for laser therapy to a similar extent for maculopathy (31% decrease) and for proliferative retinopathy (30% decrease). In the ophthalmology sub-study (n=1012), the primary endpoint of 2-step progression of retinopathy grade did not differ significantly between the fenofibrate and control groups (9.6% patients on fenofibrate vs 12.3% on placebo; p=0.19). In patients without pre-existing retinopathy there was no difference in progression (11.4% vs 11.7%; p=0.87). However, in patients with pre-existing retinopathy, significantly fewer patients on fenofibrate had a 2-step progression than did those on placebo (3.1% patients vs 14.6%; p=0.004). A composite endpoint of 2-step progression of retinopathy grade, macular edema, or laser treatments was significantly reduced in the fenofibrate group (HR 0.66, 95% CI 0.47-0.94; p=0.022).

 

In the ACCORD Study a subgroup of participants was evaluated for the progression of diabetic retinopathy by 3 or more steps on the Early Treatment Diabetic Retinopathy Study Severity Scale or the development of diabetic retinopathy necessitating laser photocoagulation or vitrectomy over a four-year period (190). At 4 years, the rates of progression of diabetic retinopathy were 6.5% with fenofibrate therapy (n=806) vs. 10.2% with placebo (n=787) (adjusted odds ratio, 0.60; 95% CI, 0.42 to 0.87; P = 0.006). Of note, this reduction in the progression of diabetic retinopathy was of a similar magnitude as intensive glycemic treatment vs. standard therapy.

 

Taken together these results indicate that fibrates have beneficial effects on the progression of diabetic retinopathy. The mechanisms by which fibrates decrease diabetic retinopathy are unknown.

 

Diabetic Nephropathy

 

The Diabetes Atherosclerosis Intervention Study (DAIS) evaluated the effect of fenofibrate therapy (n= 155) vs. placebo (n=159) on changes in urinary albumin excretion in patients with T2DM (251). Fenofibrate significantly reduced the worsening of albumin excretion (fenofibrate 8% vs. placebo 18%; P < 0.05). This effect was primarily due to reduced progression from normal albumin excretion to microalbuminuria (fenofibrate 3% vs. 18% placebo; P < 0.001).

 

In the FIELD trial, fenofibrate reduced urine albumin/creatinine ratio by 24% vs 11% in placebo group (p < 0.001), with 14% less progression and 18% more albuminuria regression (p < 0.001) in the fenofibrate group than in participants on placebo (252). As expected, fenofibrate therapy acutely increased plasma creatinine levels and decreased eGFR but over the long term, the increase in plasma creatinine was decreased in the fenofibrate group compared to the placebo group (14% decrease; p=0.01). Similarly, there was a slower annual decrease in eGFR in the fenofibrate group (1.19 vs 2.03 mL/min/1.73m2   annually, p < 0.001). The effect of fenofibrate on kidney function was greater in those with higher TG and lower HDL levels. End-stage renal disease, dialysis, renal transplant, and renal death were similar in the fenofibrate and placebo groups, but the incidence was low.

 

In the ACCORD-LIPID trial the post-randomization incidence of microalbuminuria was 38.2% in the fenofibrate group and 41.6% in the placebo group (p=0.01) and post-randomization incidence of macroalbumuria was 10.5% in the fibrate group and 12.3% in the placebo group (p=0.04) indicating a modest reduction in the development of proteinuria in patients treated with fenofibrate (189). There was no significant difference in the incidence of end-stage renal disease or need for dialysis between the fenofibrate group and the placebo group.

 

These studies suggest that fibrates may have a beneficial effect on diabetic kidney disease. One should recognize that reducing proteinuria is a surrogate marker and may not indicate a reduction in the development of end stage renal disease. The mechanisms accounting for decreased in proteinuria are unknown.

 

Amputations

 

In the FIELD study the risks of first amputation were decreased by 36% (p=0.02) and minor amputation events without known large-vessel disease by 47% (p=0.027) in the fenofibrate treated group (253). The reduction in amputations was independent of glucose control or dyslipidemia. No difference between the risks of major amputations was seen in the placebo and fenofibrate groups. The basis for this reduction in amputations is unknown.

 

Do Fibrates have an Independent Effect on Microvascular Disease?

 

Multiple studies cited above have now shown that fenofibrate decreases retinopathy, nephropathy, and amputation in the absence of large vessel disease. The effects are independent of blood glucose control. Given that there also was no effect of fenofibrate on cardiovascular (macrovascular) disease, these results may suggest that fenofibrate has an independent effect on microvascular disease.  Further studies are warranted, but these results should be taken into account when considering treatment of marked hypertriglyceridemia in patients with diabetes. 

 

BILE ACID SEQUESTRANTS

 

Bile acid sequestrants are relatively difficult to take due to GI toxicity (mainly constipation) (141). Diabetic subjects have an increased prevalence of constipation, which may be exacerbated by the use of bile acid sequestrants. On the other hand, in diabetic patients with diarrhea, the use of bile acid sequestrants may be advantageous. Bile acid sequestrants may also increase serum TG levels, which can be a problem in patients with diabetes who are already hypertriglyceridemic (141). An additional difficulty in using bile acid sequestrants is their potential for binding other drugs (141). Many drugs should be taken either two hours before or four hours after taking bile acid sequestrants to avoid the potential of decreased drug absorption. Patients with diabetes are frequently on multiple drugs for glycemic control, hypertension, etc., and it can sometimes be difficult to time the ingestion of bile resin sequestrants to avoid these other drugs. Colesevelam (Welchol) is a bile acid sequestrant that comes in pill, powder, or chewable bars and causes fewer side effects and has fewer interactions with other drugs than other preparations (254). The usual dose is3.75 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). Of particular note is that a number of studies have shown that colesevelam improves glycemic control in patients with diabetes resulting in an approximately 0.5% decrease in A1c levels (255).

 

NIACIN

 

Niacin is well known to cause skin flushing and itching and GI upset (256). Additionally, niacin reduces insulin sensitivity (i.e., causes insulin resistance), which can worsen glycemic control (256). Studies have shown that niacin is usually well tolerated in diabetic subjects who are in good glycemic control (257,258). In patients with poor glycemic control, niacin is more likely to adversely impact glucose levels. In the HPS2-Thrive trial, niacin therapy significantly worsened glycemic control in patients with diabetes and induced new onset diabetes in 1.3% of subjects that were non-diabetic (195). High doses of niacin are more likely to adversely affect glycemic control. Niacin can also increase serum uric acid levels and induce gout, both of which are already common in obese patients with T2DM (256). Additionally recent trials have reported an increased incidence of infection and bleeding with niacin therapy (256). However, niacin is the most effective drug in increasing HDL-C levels, which are frequently low in patients with diabetes. 

 

OMEGA-3-FATTY ACIDS

 

A Cochrane review of fish oil in patients with diabetes have demonstrated that this is a safe approach and does not result in worsening of glycemic control in patients with diabetes (242). Fish oil effectively lowers TG levels but, in some patients, particularly those with significant hypertriglyceridemia, high dose fish oil increases LDL-C levels (242). It should be noted that fish oil products that contain just EPA (Vascepa) do not adversely affect LDL-C levels (259). When using fish oil to lower serum TG levels it is important to recognize that one is aiming to provide 3-4 grams of DHA/EPA per day. The quantity of these active omega-3-fatty acids can vary greatly from product to product. Prescription fish oil products contain large amounts of these active ingredients whereas the amount of DHA/EPA in food supplements can vary greatly and in some products levels are very low. Additionally, while prescription omega-3-fatty acid preparations have high levels of quality control, omega-3-fish oil food supplements may have contaminants and the dosage may not be precisely controlled.

 

PCSK9 INHIBITORS

 

The beneficial effects of PCSK9 inhibitors in patients with diabetes is similar to what is observed in non-diabetic patients. Additionally, except for local reactions at the injection sites PCSK9 inhibitors do not seem to cause major side effects. PCSK9 inhibitors do not appear to increase the risk of developing new-onset diabetes (260,261).

 

BEMPEDOIC ACID

 

The effect of bempedoic acid on LDL-C levels in patients with diabetes are similar to the decreases seen on non-diabetics. Patients with T2DM often have elevated uric acid levels and an increased risk of gouty attacks and a major side effect of bempedoic acid is elevating uric acid levels (141). 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. Elevations in blood uric acid levels may lead to the development of gout and 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). 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.

 

In a meta-analysis, bempedoic acid therapy was associated with a decrease in the onset of diabetes and worsening of diabetes  (RR 0.65, p = 0.03) (7/100 vs 6.4/100 patient years) (262). In a study focusing solely on the development of new onset diabetes it was reported that new-onset diabetes/hyperglycemia occurred less frequently with bempedoic acid vs placebo (263). In the bempedoic acid cardiovascular outcome trial (CLEAR Outcomes) the development of diabetes and worsening of pre-existing diabetes was similar in the bempedoic acid and placebo groups (187).  

 

Table 12. Effect of Lipid Lowering Drugs

 

LDL-C

HDL-C

TG

 

Statins

↓ 20-60%

↑ 5-15%

↓ 0-35%*

Bile acid sequestrants

↓ 10-30%

↑ 0-10%

↑ 0-10%**

Fibrates

↓ 0-15%***

↑ 5-15%

↓ 20-50%

Niacin

↓ 10-25%

↑ 10-30%

↓ 20-50%

Ezetimibe

↓ 15-25%

↑ 1-3%

↓ 10-20%

PCSK9 Inhibitors

↓ 50-60%

↑ 5-15%

↓ 5-20%

Bempedoic Acid

↓ 15-25%

↓ 5-6%

No change

High Dose Fish Oil

↑ 0- 50%***

↑ 4- 9%

↓ 20- 50%*

 *Patients with elevated TG have largest decrease

** In patients with high TG may cause marked increase

*** In patients with high TG may increase LDL

 

Therapeutic Approach

 

FIRST PRIORITY- LDL-C

 

The first priority in treating lipid disorders in patients with diabetes is to lower the LDL-C levels to goal, unless TG are markedly elevated (> 500- 1000mg/dL), which increases the risk of pancreatitis. LDL-C is the first priority because the database linking lowering LDL-C with reducing ASCVD is extremely strong and we now have the ability to markedly decrease LDL-C levels. Dietary therapy is the initial step but, in almost all patients, will not be sufficient to achieve the LDL-C goals. If patients are willing and able to make major changes in their diet it is possible to achieve significant reductions in LDL-C levels but this seldom occurs in clinical practice (264).

 

Statins are the first-choice drugs to lower LDL-C levels and the vast majority of diabetic patients will require statin therapy. There are several statins currently available in the US and they are available as generic drugs and therefore relatively inexpensive. The particular statin used may be driven by price, ability to lower LDL-C levels, and potential drug interactions. Patients with ASCVD (secondary prevention patients) should be started on intensive statin therapy (atorvastatin 40-80mg per day or rosuvastatin 20-40mg per day). Given the extensive data showing that the lower the LDL-C the greater the reduction in ASCVD events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering. Ezetimibe is now a generic drug and therefore this strategy will not markedly increase costs. Similarly, primary prevention patients who are at high risk for cardiovascular events will also benefit from the use of high intensity statin therapy in combination with ezetimibe. Primary prevention patients at moderate risk can be started on moderate intensity statin therapy.

 

If a patient is unable to tolerate statins or statins as monotherapy are not sufficient to lower LDL-C to goal the second-choice drug is either ezetimibe or a PCSK9 inhibitor. Ezetimibe can be added to any statin. PCSK9 inhibitors can also be added to any statin and are the drug of choice if a large decrease in LDL-C is required to reach goal (PCSK9 inhibitors will lower LDL-C levels by 50-60% when added to a statin, whereas ezetimibe will only lower LDL-C by approximately 20%).  Bile acid sequestrants and bempedoic acid are alternatives with the use of a bile acid sequestrant particularly useful if a reduction in A1c level is also needed. It should be noted that in statin intolerant patients with ASCVD or at high risk (approximately 45% with diabetes), bempedoic acid has been shown to reduce cardiovascular events by 13% (HR 0.87; 95% CI 0.79 to 0.96; P = 0.004) (187). Ezetimibe, PCSK9 inhibitors, bempedoic acid, and bile acid sequestrants additively lower LDL-C levels when used in combination with a statin, because these drugs increase hepatic LDL receptor levels by different mechanisms, thereby resulting in a reduction in serum LDL-C levels (141). Niacin and the fibrates also lower LDL-C levels but are not usually employed to lower LDL-C levels. 

 

SECOND PRIORITY- NON-HDL-C

 

The second priority should be non-HDL-C (non-HDL-C = total cholesterol – HDL-C), which is particularly important in patients with elevated TG levels (>150mg/dL). Non-HDL-C is a measure of all the pro-atherogenic apolipoprotein B containing particles. Numerous studies have shown that non-HDL-C is a strong risk factor for the development of ASCVD (265). The non-HDL-C goals are approximately 30mg/dL greater than the LDL-C goals. For example, if the LDL goal is <100mg/dL then the non-HDL-C goal would be <130mg/dL. Drugs that reduce either LDL-C or TG levels will reduce non-HDL-C levels. To lower TG levels initial therapy should focus on glycemic control and lifestyle changes including weight loss if indicated and a decrease in simple sugars and ethanol intake. Additionally, if possible, discontinue medications that increase triglyceride levels. As discussed above, studies with the omega-3-fatty acid icosapent ethyl (EPA; Vascepa) added to statin therapy have reduced the risk of cardiovascular events. The National Lipid Association has recommended “that for patients aged ≥45 years with clinical ASCVD, or aged ≥50 years with diabetes mellitus requiring medication plus ≥1 additional risk factor, with fasting TGs 135 to 499 mg/dL on high-intensity or maximally tolerated statin therapy (±ezetimibe), treatment with icosapent ethyl is recommended for ASCVD risk reduction” (266). As noted earlier in this chapter there is controversy regarding the benefits of icosapent ethyl on cardiovascular outcomes with some experts interpreting the beneficial results of the REDUCE-IT trial as being due to the adverse effects of the mineral oil placebo (217).

 

VERY HIGH TG

 

Patients with very high TG levels (> 500-1000 mg/dL) are at risk of pancreatitis and therefore lifestyle interventions including diet, exercise, and weight loss if indicated should be initiated early. Treatment is a low-fat diet and glycemic control. Additionally, decreasing simple sugars and avoiding alcohol is beneficial. When the TG levels are very elevated (> 1000mg/dL) a very low-fat diet (5-20% of calories as fat) should be the primary treatment until the TG levels decrease to < 1000mg/dL. Treating secondary disorders that raise TG levels and when possible, stopping drugs that increase TG levels is essential. If the TG levels remain above 500mg/dL the addition of fenofibrate or omega-3-fatty acids is indicated.

 

LOW HDL-C

 

While there is strong epidemiologic data linking low HDL-C levels with ASCVD there is no clinical trials demonstrating that increasing HDL-C levels reduce ASCVD. Thus, the use of drugs such as niacin to raise HDL-C levels is not recommended.

 

CONCLUSION

 

Patients with diabetes, particularly T2DM, often have dyslipidemia. Modern therapy of patients with diabetes demands that we aggressively treat lipids to reduce the high risk of ASCVD in this susceptible population and in those with very high TG to reduce the risk of pancreatitis.

 

REFERENCES

 

  1. Milicevic Z, Raz I, Beattie SD, Campaigne BN, Sarwat S, Gromniak E, Kowalska I, Galic E, Tan M, Hanefeld M. Natural history of cardiovascular disease in patients with diabetes: role of hyperglycemia. Diabetes Care 2008; 31 Suppl 2:S155-160
  2. Feingold KR, Siperstein MD. Diabetic vascular disease. Adv Intern Med 1986; 31:309-340
  3. Regensteiner JG, Golden S, Huebschmann AG, Barrett-Connor E, Chang AY, Chyun D, Fox CS, Kim C, Mehta N, Reckelhoff JF, Reusch JE, Rexrode KM, Sumner AE, Welty FK, Wenger NK, Anton B. Sex Differences in the Cardiovascular Consequences of Diabetes Mellitus: A Scientific Statement From the American Heart Association. Circulation 2015; 132:2424-2447
  4. Fox CS, Golden SH, Anderson C, Bray GA, Burke LE, de Boer IH, Deedwania P, Eckel RH, Ershow AG, Fradkin J, Inzucchi SE, Kosiborod M, Nelson RG, Patel MJ, Pignone M, Quinn L, Schauer PR, Selvin E, Vafiadis DK. Update on Prevention of Cardiovascular Disease in Adults With Type 2 Diabetes Mellitus in Light of Recent Evidence: A Scientific Statement From the American Heart Association and the American Diabetes Association. Diabetes Care 2015; 38:1777-1803
  5. Low Wang CC, Hess CN, Hiatt WR, Goldfine AB. Clinical Update: Cardiovascular Disease in Diabetes Mellitus: Atherosclerotic Cardiovascular Disease and Heart Failure in Type 2 Diabetes Mellitus - Mechanisms, Management, and Clinical Considerations. Circulation 2016; 133:2459-2502
  6. Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA 1979; 241:2035-2038
  7. Members Writing Group, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB. Executive Summary: Heart Disease and Stroke Statistics--2016 Update: A Report From the American Heart Association. Circulation 2016; 133:447-454
  8. Haffner SM, Lehto S, Ronnemaa T, Pyorala K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229-234
  9. Evans JM, Wang J, Morris AD. Comparison of cardiovascular risk between patients with type 2 diabetes and those who had had a myocardial infarction: cross sectional and cohort studies. BMJ 2002; 324:939-942
  10. Wannamethee SG, Shaper AG, Whincup PH, Lennon L, Sattar N. Impact of diabetes on cardiovascular disease risk and all-cause mortality in older men: influence of age at onset, diabetes duration, and established and novel risk factors. Arch Intern Med 2011; 171:404-410
  11. Howard BV, Best LG, Galloway JM, Howard WJ, Jones K, Lee ET, Ratner RE, Resnick HE, Devereux RB. Coronary heart disease risk equivalence in diabetes depends on concomitant risk factors. Diabetes Care 2006; 29:391-397
  12. Rawshani A, Rawshani A, Franzen S, Sattar N, Eliasson B, Svensson AM, Zethelius B, Miftaraj M, McGuire DK, Rosengren A, Gudbjornsdottir S. Risk Factors, Mortality, and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2018; 379:633-644
  13. Lind M, Svensson AM, Kosiborod M, Gudbjornsdottir S, Pivodic A, Wedel H, Dahlqvist S, Clements M, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med 2014; 371:1972-1982
  14. de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Diabetes Care 2014; 37:2843-2863
  15. Maahs DM, Daniels SR, de Ferranti SD, Dichek HL, Flynn J, Goldstein BI, Kelly AS, Nadeau KJ, Martyn-Nemeth P, Osganian SK, Quinn L, Shah AS, Urbina E. Cardiovascular disease risk factors in youth with diabetes mellitus: a scientific statement from the American Heart Association. Circulation 2014; 130:1532-1558
  16. Huxley RR, Peters SA, Mishra GD, Woodward M. Risk of all-cause mortality and vascular events in women versus men with type 1 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol 2015; 3:198-206
  17. Rawshani A, Sattar N, Franzen S, Rawshani A, Hattersley AT, Svensson AM, Eliasson B, Gudbjornsdottir S. Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 2018; 392:477-486
  18. Chillaron JJ, Flores Le-Roux JA, Benaiges D, Pedro-Botet J. Type 1 diabetes, metabolic syndrome and cardiovascular risk. Metabolism 2014; 63:181-187
  19. Constantino MI, Molyneaux L, Limacher-Gisler F, Al-Saeed A, Luo C, Wu T, Twigg SM, Yue DK, Wong J. Long-term complications and mortality in young-onset diabetes: type 2 diabetes is more hazardous and lethal than type 1 diabetes. Diabetes Care 2013; 36:3863-3869
  20. Preis SR, Hwang SJ, Coady S, Pencina MJ, D'Agostino RB, Sr., Savage PJ, Levy D, Fox CS. Trends in all-cause and cardiovascular disease mortality among women and men with and without diabetes mellitus in the Framingham Heart Study, 1950 to 2005. Circulation 2009; 119:1728-1735
  21. Matuleviciene-Anangen V, Rosengren A, Svensson AM, Pivodic A, Gudbjornsdottir S, Wedel H, Kosiborod M, Haraldsson B, Lind M. Glycaemic control and excess risk of major coronary events in persons with type 1 diabetes. Heart 2017; 103:1687-1695
  22. Miller RG, Orchard TJ, Costacou T. 30-Year Cardiovascular Disease in Type 1 Diabetes: Risk and Risk Factors Differ by Long-term Patterns of Glycemic Control. Diabetes Care 2022; 45:142-150
  23. Abraira C, Colwell JA, Nuttall FQ, Sawin CT, Nagel NJ, Comstock JP, Emanuele NV, Levin SR, Henderson W, Lee HS. Veterans Affairs Cooperative Study on glycemic control and complications in type II diabetes (VA CSDM). Results of the feasibility trial. Veterans Affairs Cooperative Study in Type II Diabetes. Diabetes Care1995; 18:1113-1123
  24. Goldner MG, Knatterud GL, Prout TE. Effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. 3. Clinical implications of UGDP results. JAMA 1971; 218:1400-1410
  25. Meinert CL, Knatterud GL, Prout TE, Klimt CR. A study of the effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. II. Mortality results. Diabetes 1970; 19:Suppl:789-830
  26. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 1993; 329:977-986
  27. Ohkubo Y, Kishikawa H, Araki E, Miyata T, Isami S, Motoyoshi S, Kojima Y, Furuyoshi N, Shichiri M. Intensive insulin therapy prevents the progression of diabetic microvascular complications in Japanese patients with non-insulin-dependent diabetes mellitus: a randomized prospective 6-year study. Diabetes Res Clin Pract 1995; 28:103-117
  28. Shichiri M, Kishikawa H, Ohkubo Y, Wake N. Long-term results of the Kumamoto Study on optimal diabetes control in type 2 diabetic patients. Diabetes Care 2000; 23 Suppl 2:B21-29
  29. Lachin JM, Orchard TJ, Nathan DM, Group DER. Update on cardiovascular outcomes at 30 years of the diabetes control and complications trial/epidemiology of diabetes interventions and complications study. Diabetes Care 2014; 37:39-43
  30. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, Raskin P, Zinman B, Diabetes C, Complications Trial/Epidemiology of Diabetes I, Complications Study Research Group. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 2005; 353:2643-2653
  31. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:837-853
  32. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359:1577-1589
  33. Malmberg K. Prospective randomised study of intensive insulin treatment on long term survival after acute myocardial infarction in patients with diabetes mellitus. DIGAMI (Diabetes Mellitus, Insulin Glucose Infusion in Acute Myocardial Infarction) Study Group. BMJ 1997; 314:1512-1515
  34. Mellbin LG, Malmberg K, Norhammar A, Wedel H, Ryden L, Investigators D. The impact of glucose lowering treatment on long-term prognosis in patients with type 2 diabetes and myocardial infarction: a report from the DIGAMI 2 trial. Eur Heart J 2008; 29:166-176
  35. Action to Control Cardiovascular Risk in Diabetes Study Group, Gerstein HC, Miller ME, Byington RP, Goff DC, Jr., Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail-Beigi F, Grimm RH, Jr., Probstfield JL, Simons-Morton DG, Friedewald WT. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545-2559
  36. Gerstein HC, Miller ME, Ismail-Beigi F, Largay J, McDonald C, Lochnan HA, Booth GL, ACCORD STUDY Group. Effects of intensive glycaemic control on ischaemic heart disease: analysis of data from the randomised, controlled ACCORD trial. Lancet 2014; 384:1936-1941
  37. ACCORD STUDY Group. Nine-Year Effects of 3.7 Years of Intensive Glycemic Control on Cardiovascular Outcomes. Diabetes Care 2016; 39:701-708
  38. Advance Collaborative Group, Patel A, MacMahon S, Chalmers J, Neal B, Billot L, Woodward M, Marre M, Cooper M, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Liu L, Mancia G, Mogensen CE, Pan C, Poulter N, Rodgers A, Williams B, Bompoint S, de Galan BE, Joshi R, Travert F. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560-2572
  39. Zoungas S, Chalmers J, Neal B, Billot L, Li Q, Hirakawa Y, Arima H, Monaghan H, Joshi R, Colagiuri S, Cooper ME, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Lisheng L, Mancia G, Marre M, Matthews DR, Mogensen CE, Perkovic V, Poulter N, Rodgers A, Williams B, MacMahon S, Patel A, Woodward M, Group A-OC. Follow-up of blood-pressure lowering and glucose control in type 2 diabetes. N Engl J Med 2014; 371:1392-1406
  40. Duckworth W, Abraira C, Moritz T, Reda D, Emanuele N, Reaven PD, Zieve FJ, Marks J, Davis SN, Hayward R, Warren SR, Goldman S, McCarren M, Vitek ME, Henderson WG, Huang GD, Investigators V. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129-139
  41. Hayward RA, Reaven PD, Wiitala WL, Bahn GD, Reda DJ, Ge L, McCarren M, Duckworth WC, Emanuele NV, Investigators V. Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. N Engl J Med2015; 372:2197-2206
  42. Reaven PD, Emanuele NV, Wiitala WL, Bahn GD, Reda DJ, McCarren M, Duckworth WC, Hayward RA, Investigators V. Intensive Glucose Control in Patients with Type 2 Diabetes - 15-Year Follow-up. N Engl J Med2019; 380:2215-2224
  43. Buehler AM, Cavalcanti AB, Berwanger O, Figueiro M, Laranjeira LN, Zazula AD, Kioshi B, Bugano DG, Santucci E, Sbruzzi G, Guimaraes HP, Carvalho VO, Bordin SA. Effect of tight blood glucose control versus conventional control in patients with type 2 diabetes mellitus: a systematic review with meta-analysis of randomized controlled trials. Cardiovasc Ther 2013; 31:147-160
  44. Control Group, Turnbull FM, Abraira C, Anderson RJ, Byington RP, Chalmers JP, Duckworth WC, Evans GW, Gerstein HC, Holman RR, Moritz TE, Neal BC, Ninomiya T, Patel AA, Paul SK, Travert F, Woodward M. Intensive glucose control and macrovascular outcomes in type 2 diabetes. Diabetologia 2009; 52:2288-2298
  45. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:854-865
  46. Kooy A, de Jager J, Lehert P, Bets D, Wulffele MG, Donker AJ, Stehouwer CD. Long-term effects of metformin on metabolism and microvascular and macrovascular disease in patients with type 2 diabetes mellitus. Arch Intern Med 2009; 169:616-625
  47. Hong J, Zhang Y, Lai S, Lv A, Su Q, Dong Y, Zhou Z, Tang W, Zhao J, Cui L, Zou D, Wang D, Li H, Liu C, Wu G, Shen J, Zhu D, Wang W, Shen W, Ning G. Effects of metformin versus glipizide on cardiovascular outcomes in patients with type 2 diabetes and coronary artery disease. Diabetes Care 2013; 36:1304-1311
  48. Goldberg RB, Orchard TJ, Crandall JP, Boyko EJ, Budoff M, Dabelea D, Gadde KM, Knowler WC, Lee CG, Nathan DM, Watson K, Temprosa M. Effects of Long-term Metformin and Lifestyle Interventions on Cardiovascular Events in the Diabetes Prevention Program and Its Outcome Study. Circulation 2022; 145:1632-1641
  49. Goldberg RB, Aroda VR, Bluemke DA, Barrett-Connor E, Budoff M, Crandall JP, Dabelea D, Horton ES, Mather KJ, Orchard TJ, Schade D, Watson K, Temprosa M. Effect of Long-Term Metformin and Lifestyle in the Diabetes Prevention Program and Its Outcome Study on Coronary Artery Calcium. Circulation 2017; 136:52-64
  50. Fitch K, Abbara S, Lee H, Stavrou E, Sacks R, Michel T, Hemphill L, Torriani M, Grinspoon S. Effects of lifestyle modification and metformin on atherosclerotic indices among HIV-infected patients with the metabolic syndrome. AIDS 2012; 26:587-597
  51. Rosenstock J, Kahn SE, Johansen OE, Zinman B, Espeland MA, Woerle HJ, Pfarr E, Keller A, Mattheus M, Baanstra D, Meinicke T, George JT, von Eynatten M, McGuire DK, Marx N. Effect of Linagliptin vs Glimepiride on Major Adverse Cardiovascular Outcomes in Patients With Type 2 Diabetes: The CAROLINA Randomized Clinical Trial. JAMA 2019;
  52. Navigator Study Group, Holman RR, Haffner SM, McMurray JJ, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, Buckley BM, Chacra AR, Chiang FT, Charbonnel B, Chow CC, Davies MJ, Deedwania P, Diem P, Einhorn D, Fonseca V, Fulcher GR, Gaciong Z, Gaztambide S, Giles T, Horton E, Ilkova H, Jenssen T, Kahn SE, Krum H, Laakso M, Leiter LA, Levitt NS, Mareev V, Martinez F, Masson C, Mazzone T, Meaney E, Nesto R, Pan C, Prager R, Raptis SA, Rutten GE, Sandstroem H, Schaper F, Scheen A, Schmitz O, Sinay I, Soska V, Stender S, Tamas G, Tognoni G, Tuomilehto J, Villamil AS, Vozar J, Califf RM. Effect of nateglinide on the incidence of diabetes and cardiovascular events. N Engl J Med 2010; 362:1463-1476
  53. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefebvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L, Laakso M, Mokan M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schernthaner G, Schmitz O, Skrha J, Smith U, Taton J. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet 2005; 366:1279-1289
  54. Kernan WN, Viscoli CM, Furie KL, Young LH, Inzucchi SE, Gorman M, Guarino PD, Lovejoy AM, Peduzzi PN, Conwit R, Brass LM, Schwartz GG, Adams HP, Jr., Berger L, Carolei A, Clark W, Coull B, Ford GA, Kleindorfer D, O'Leary JR, Parsons MW, Ringleb P, Sen S, Spence JD, Tanne D, Wang D, Winder TR. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N Engl J Med 2016; 374:1321-1331
  55. Vaccaro O, Masulli M, Nicolucci A, Bonora E, Del Prato S, Maggioni AP, Rivellese AA, Squatrito S, Giorda CB, Sesti G, Mocarelli P, Lucisano G, Sacco M, Signorini S, Cappellini F, Perriello G, Babini AC, Lapolla A, Gregori G, Giordano C, Corsi L, Buzzetti R, Clemente G, Di Cianni G, Iannarelli R, Cordera R, La Macchia O, Zamboni C, Scaranna C, Boemi M, Iovine C, Lauro D, Leotta S, Dall'Aglio E, Cannarsa E, Tonutti L, Pugliese G, Bossi AC, Anichini R, Dotta F, Di Benedetto A, Citro G, Antenucci D, Ricci L, Giorgino F, Santini C, Gnasso A, De Cosmo S, Zavaroni D, Vedovato M, Consoli A, Calabrese M, di Bartolo P, Fornengo P, Riccardi G. Effects on the incidence of cardiovascular events of the addition of pioglitazone versus sulfonylureas in patients with type 2 diabetes inadequately controlled with metformin (TOSCA.IT): a randomised, multicentre trial. Lancet Diabetes Endocrinol 2017; 5:887-897
  56. Vaccaro O, Lucisano G, Masulli M, Bonora E, Del Prato S, Rivellese AA, Giorda CB, Mocarelli P, Squatrito S, Maggioni AP, Riccardi G, Nicolucci A. Cardiovascular Effects of Pioglitazone or Sulfonylureas According to Pretreatment Risk: Moving Toward Personalized Care. J Clin Endocrinol Metab 2019; 104:3296-3302
  57. Mazzone T, Meyer PM, Feinstein SB, Davidson MH, Kondos GT, D'Agostino RB, Sr., Perez A, Provost JC, Haffner SM. Effect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes: a randomized trial. JAMA 2006; 296:2572-2581
  58. Pfutzner A, Marx N, Lubben G, Langenfeld M, Walcher D, Konrad T, Forst T. Improvement of cardiovascular risk markers by pioglitazone is independent from glycemic control: results from the pioneer study. J Am Coll Cardiol2005; 45:1925-1931
  59. Nissen SE, Nicholls SJ, Wolski K, Nesto R, Kupfer S, Perez A, Jure H, De Larochelliere R, Staniloae CS, Mavromatis K, Saw J, Hu B, Lincoff AM, Tuzcu EM. Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial. JAMA2008; 299:1561-1573
  60. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 2007; 356:2457-2471
  61. Singh S, Loke YK, Furberg CD. Long-term risk of cardiovascular events with rosiglitazone: a meta-analysis. JAMA 2007; 298:1189-1195
  62. Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet 2009; 373:2125-2135
  63. Home PD, Pocock SJ, Beck-Nielsen H, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes--an interim analysis. N Engl J Med 2007; 357:28-38
  64. Mahaffey KW, Hafley G, Dickerson S, Burns S, Tourt-Uhlig S, White J, Newby LK, Komajda M, McMurray J, Bigelow R, Home PD, Lopes RD. Results of a reevaluation of cardiovascular outcomes in the RECORD trial. Am Heart J 2013; 166:240-249 e241
  65. Bach RG, Brooks MM, Lombardero M, Genuth S, Donner TW, Garber A, Kennedy L, Monrad ES, Pop-Busui R, Kelsey SF, Frye RL. Rosiglitazone and outcomes for patients with diabetes mellitus and coronary artery disease in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial. Circulation 2013; 128:785-794
  66. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for Type 2 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)2022.
  67. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, Ohman P, Frederich R, Wiviott SD, Hoffman EB, Cavender MA, Udell JA, Desai NR, Mosenzon O, McGuire DK, Ray KK, Leiter LA, Raz I. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 2013; 369:1317-1326
  68. Scirica BM, Braunwald E, Raz I, Cavender MA, Morrow DA, Jarolim P, Udell JA, Mosenzon O, Im K, Umez-Eronini AA, Pollack PS, Hirshberg B, Frederich R, Lewis BS, McGuire DK, Davidson J, Steg PG, Bhatt DL. Heart failure, saxagliptin, and diabetes mellitus: observations from the SAVOR-TIMI 53 randomized trial. Circulation 2014; 130:1579-1588
  69. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Cushman WC, Zannad F. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med 2013; 369:1327-1335
  70. Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Lam H, White WB. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet 2015; 385:2067-2076
  71. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, Josse R, Kaufman KD, Koglin J, Korn S, Lachin JM, McGuire DK, Pencina MJ, Standl E, Stein PP, Suryawanshi S, Van de Werf F, Peterson ED, Holman RR. Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2015; 373:232-242
  72. Rosenstock J, Perkovic V, Johansen OE, Cooper ME, Kahn SE, Marx N, Alexander JH, Pencina M, Toto RD, Wanner C, Zinman B, Woerle HJ, Baanstra D, Pfarr E, Schnaidt S, Meinicke T, George JT, von Eynatten M, McGuire DK. Effect of Linagliptin vs Placebo on Major Cardiovascular Events in Adults With Type 2 Diabetes and High Cardiovascular and Renal Risk: The CARMELINA Randomized Clinical Trial. JAMA 2019 Jan 1;321(1):69-79.
  73. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med 2015; 373:2117-2128
  74. Fitchett D, Inzucchi SE, Cannon CP, McGuire DK, Scirica BM, Johansen OE, Sambevski S, Kaspers S, Pfarr E, George JT, Zinman B. Empagliflozin Reduced Mortality and Hospitalization for Heart Failure Across the Spectrum of Cardiovascular Risk in the EMPA-REG OUTCOME Trial. Circulation 2019; 139:1384-1395
  75. Fitchett D, Butler J, van de Borne P, Zinman B, Lachin JM, Wanner C, Woerle HJ, Hantel S, George JT, Johansen OE, Inzucchi SE. Effects of empagliflozin on risk for cardiovascular death and heart failure hospitalization across the spectrum of heart failure risk in the EMPA-REG OUTCOME(R) trial. Eur Heart J 2018; 39:363-370
  76. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med 2017; 377:644-657
  77. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, de Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med 2019; 380:2295-2306
  78. Jardine MJ, Zhou Z, Mahaffey KW, Oshima M, Agarwal R, Bakris G, Bajaj HS, Bull S, Cannon CP, Charytan DM, de Zeeuw D, Di Tanna GL, Greene T, Heerspink HJL, Levin A, Neal B, Pollock C, Qiu R, Sun T, Wheeler DC, Zhang H, Zinman B, Rosenthal N, Perkovic V. Renal, Cardiovascular, and Safety Outcomes of Canagliflozin by Baseline Kidney Function: A Secondary Analysis of the CREDENCE Randomized Trial. J Am Soc Nephrol2020; 31:1128-1139
  79. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med2018;
  80. Furtado RHM, Bonaca MP, Raz I, Zelniker TA, Mosenzon O, Cahn A, Kuder J, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Nicolau JC, Gause-Nilsson IAM, Fredriksson M, Langkilde AM, Sabatine MS, Wiviott SD. Dapagliflozin and Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus and Previous Myocardial Infarction. Circulation 2019; 139:2516-2527
  81. Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B, Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N Engl J Med 2020; 383:1425-1435
  82. Cosentino F, Cannon CP, Cherney DZI, Masiukiewicz U, Pratley R, Dagogo-Jack S, Frederich R, Charbonnel B, Mancuso J, Shih WJ, Terra SG, Cater NB, Gantz I, McGuire DK. Efficacy of Ertugliflozin on Heart Failure-Related Events in Patients With Type 2 Diabetes Mellitus and Established Atherosclerotic Cardiovascular Disease: Results of the VERTIS CV Trial. Circulation 2020; 142:2205-2215
  83. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 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)2022.
  84. Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Kober LV, Lawson FC, Ping L, Wei X, Lewis EF, Maggioni AP, McMurray JJ, Probstfield JL, Riddle MC, Solomon SD, Tardif JC. Lixisenatide in Patients with Type 2 Diabetes and Acute Coronary Syndrome. N Engl J Med 2015; 373:2247-2257
  85. 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. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2016; 375:311-322
  86. Mann JFE, Fonseca V, Mosenzon O, Raz I, Goldman B, Idorn T, von Scholten BJ, Poulter NR. Effects of Liraglutide Versus Placebo on Cardiovascular Events in Patients With Type 2 Diabetes Mellitus and Chronic Kidney Disease. Circulation 2018; 138:2908-2918
  87. Verma S, Poulter NR, Bhatt DL, Bain SC, Buse JB, Leiter LA, Nauck MA, Pratley RE, Zinman B, Orsted DD, Monk Fries T, Rasmussen S, Marso SP. Effects of Liraglutide on Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus With or Without History of Myocardial Infarction or Stroke. Circulation 2018; 138:2884-2894
  88. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsboll T. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2016;
  89. Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, Jeppesen OK, Lingvay I, Mosenzon O, Pedersen SD, Tack CJ, Thomsen M, Vilsboll T, Warren ML, Bain SC. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2019; 381:841-851
  90. Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB, Chan JC, Choi J, Gustavson SM, Iqbal N, Maggioni AP, Marso SP, Ohman P, Pagidipati NJ, Poulter N, Ramachandran A, Zinman B, Hernandez AF. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2017; 377:1228-1239
  91. Hernandez AF, Green JB, Janmohamed S, D'Agostino RB, Sr., Granger CB, Jones NP, Leiter LA, Rosenberg AE, Sigmon KN, Somerville MC, Thorpe KM, McMurray JJV, Del Prato S. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial. Lancet 2018; 392:1519-1529
  92. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Riesmeyer JS, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019; 394:121-130
  93. Gerstein HC, Hart R, Colhoun HM, Diaz R, Lakshmanan M, Botros FT, Probstfield J, Riddle MC, Ryden L, Atisso CM, Dyal L, Hall S, Avezum A, Basile J, Conget I, Cushman WC, Hancu N, Hanefeld M, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Munoz EGC, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. The effect of dulaglutide on stroke: an exploratory analysis of the REWIND trial. Lancet Diabetes Endocrinol 2020; 8:106-114
  94. Kristensen SL, Rorth R, Jhund PS, Docherty KF, Sattar N, Preiss D, Kober L, Petrie MC, McMurray JJV. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol 2019; 7:776-785
  95. Sposito AC, Berwanger O, de Carvalho LSF, Saraiva JFK. GLP-1RAs in type 2 diabetes: mechanisms that underlie cardiovascular effects and overview of cardiovascular outcome data. Cardiovasc Diabetol 2018; 17:157
  96. Sattar N, McGuire DK, Pavo I, Weerakkody GJ, Nishiyama H, Wiese RJ, Zoungas S. Tirzepatide cardiovascular event risk assessment: a pre-specified meta-analysis. Nat Med 2022; 28:591-598
  97. Chiasson JL, Josse RG, Gomis R, Hanefeld M, Karasik A, Laakso M. Acarbose treatment and the risk of cardiovascular disease and hypertension in patients with impaired glucose tolerance: the STOP-NIDDM trial. JAMA 2003; 290:486-494
  98. Yun P, Du AM, Chen XJ, Liu JC, Xiao H. Effect of Acarbose on Long-Term Prognosis in Acute Coronary Syndromes Patients with Newly Diagnosed Impaired Glucose Tolerance. J Diabetes Res 2016; 2016:1602083
  99. Holman RR, Coleman RL, Chan JCN, Chiasson JL, Feng H, Ge J, Gerstein HC, Gray R, Huo Y, Lang Z, McMurray JJ, Ryden L, Schroder S, Sun Y, Theodorakis MJ, Tendera M, Tucker L, Tuomilehto J, Wei Y, Yang W, Wang D, Hu D, Pan C. Effects of acarbose on cardiovascular and diabetes outcomes in patients with coronary heart disease and impaired glucose tolerance (ACE): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2017; 5:877-886
  100. Gaziano JM, Cincotta AH, O'Connor CM, Ezrokhi M, Rutty D, Ma ZJ, Scranton RE. Randomized clinical trial of quick-release bromocriptine among patients with type 2 diabetes on overall safety and cardiovascular outcomes. Diabetes Care 2010; 33:1503-1508
  101. Younk LM, Davis SN. Evaluation of colesevelam hydrochloride for the treatment of type 2 diabetes. Expert Opin Drug Metab Toxicol 2012; 8:515-525
  102. The Lipid Research Clinics Coronary Primary Prevention Trial results. I. Reduction in incidence of coronary heart disease. JAMA 1984; 251:351-364
  103. 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
  104. Investigators OT, Gerstein HC, Bosch J, Dagenais GR, Diaz R, Jung H, Maggioni AP, Pogue J, Probstfield J, Ramachandran A, Riddle MC, Ryden LE, Yusuf S. Basal insulin and cardiovascular and other outcomes in dysglycemia. N Engl J Med 2012; 367:319-328
  105. Origin Trial Investigators. Cardiovascular and Other Outcomes Postintervention With Insulin Glargine and Omega-3 Fatty Acids (ORIGINALE). Diabetes Care 2016; 39:709-716
  106. Marso SP, McGuire DK, Zinman B, Poulter NR, Emerson SS, Pieber TR, Pratley RE, Haahr PM, Lange M, Brown-Frandsen K, Moses A, Skibsted S, Kvist K, Buse JB. Efficacy and Safety of Degludec versus Glargine in Type 2 Diabetes. N Engl J Med 2017; 377:723-732
  107. Chaitman BR, Hardison RM, Adler D, Gebhart S, Grogan M, Ocampo S, Sopko G, Ramires JA, Schneider D, Frye RL. The Bypass Angioplasty Revascularization Investigation 2 Diabetes randomized trial of different treatment strategies in type 2 diabetes mellitus with stable ischemic heart disease: impact of treatment strategy on cardiac mortality and myocardial infarction. Circulation 2009; 120:2529-2540
  108. Bari D. Study Group, Frye RL, August P, Brooks MM, Hardison RM, Kelsey SF, MacGregor JM, Orchard TJ, Chaitman BR, Genuth SM, Goldberg SH, Hlatky MA, Jones TL, Molitch ME, Nesto RW, Sako EY, Sobel BE. A randomized trial of therapies for type 2 diabetes and coronary artery disease. N Engl J Med 2009; 360:2503-2515
  109. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S140-S157
  110. de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Circulation 2014; 130:1110-1130
  111. Martin-Timon I, Sevillano-Collantes C, Segura-Galindo A, Del Canizo-Gomez FJ. Type 2 diabetes and cardiovascular disease: Have all risk factors the same strength? World J Diabetes 2014; 5:444-470
  112. Turner RC, Millns H, Neil HA, Stratton IM, Manley SE, Matthews DR, Holman RR. Risk factors for coronary artery disease in non-insulin dependent diabetes mellitus: United Kingdom Prospective Diabetes Study (UKPDS: 23). BMJ 1998; 316:823-828
  113. Hovingh GK, Rader DJ, Hegele RA. HDL re-examined. Curr Opin Lipidol 2015; 26:127-132
  114. Brunzell JD, Davidson M, Furberg CD, Goldberg RB, Howard BV, Stein JH, Witztum JL. Lipoprotein management in patients with cardiometabolic risk: consensus statement from the American Diabetes Association and the American College of Cardiology Foundation. Diabetes Care 2008; 31:811-822
  115. 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
  116. Nordestgaard BG. Triglyceride-Rich Lipoproteins and Atherosclerotic Cardiovascular Disease: New Insights From Epidemiology, Genetics, and Biology. Circ Res 2016; 118:547-563
  117. Ganjali S, Dallinga-Thie GM, Simental-Mendia LE, Banach M, Pirro M, Sahebkar A. HDL functionality in type 1 diabetes. Atherosclerosis 2017; 267:99-109
  118. Ginsberg HN, MacCallum PR. The obesity, metabolic syndrome, and type 2 diabetes mellitus pandemic: Part I. Increased cardiovascular disease risk and the importance of atherogenic dyslipidemia in persons with the metabolic syndrome and type 2 diabetes mellitus. J Cardiometab Syndr 2009; 4:113-119
  119. Goldberg IJ. Clinical review 124: Diabetic dyslipidemia: causes and consequences. J Clin Endocrinol Metab2001; 86:965-971
  120. Krauss RM. Lipids and lipoproteins in patients with type 2 diabetes. Diabetes Care 2004; 27:1496-1504
  121. Wu L, Parhofer KG. Diabetic dyslipidemia. Metabolism 2014; 63:1469-1479
  122. Taskinen MR, Boren J. New insights into the pathophysiology of dyslipidemia in type 2 diabetes. Atherosclerosis2015; 239:483-495
  123. Feingold KR, Grunfeld C, Pang M, Doerrler W, Krauss RM. LDL subclass phenotypes and triglyceride metabolism in non-insulin-dependent diabetes. Arterioscler Thromb 1992; 12:1496-1502
  124. Feingold KR. Obesity and Dyslipidemia. 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)2023.
  125. Morgantini C, Natali A, Boldrini B, Imaizumi S, Navab M, Fogelman AM, Ferrannini E, Reddy ST. Anti-inflammatory and antioxidant properties of HDLs are impaired in type 2 diabetes. Diabetes 2011; 60:2617-2623
  126. Apro J, Tietge UJ, Dikkers A, Parini P, Angelin B, Rudling M. Impaired Cholesterol Efflux Capacity of High-Density Lipoprotein Isolated From Interstitial Fluid in Type 2 Diabetes Mellitus-Brief Report. Arterioscler Thromb Vasc Biol 2016; 36:787-791
  127. Manjunatha S, Distelmaier K, Dasari S, Carter RE, Kudva YC, Nair KS. Functional and proteomic alterations of plasma high density lipoproteins in type 1 diabetes mellitus. Metabolism 2016; 65:1421-1431
  128. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res 2016; 57:1111-1125
  129. Durrington PN, Schofield JD, Siahmansur T, Soran H. Lipoprotein (a): gene genie. Curr Opin Lipidol 2014; 25:289-296
  130. Gudbjartsson DF, Thorgeirsson G, Sulem P, Helgadottir A, Gylfason A, Saemundsdottir J, Bjornsson E, Norddahl GL, Jonasdottir A, Jonasdottir A, Eggertsson HP, Gretarsdottir S, Thorleifsson G, Indridason OS, Palsson R, Jonasson F, Jonsdottir I, Eyjolfsson GI, Sigurdardottir O, Olafsson I, Danielsen R, Matthiasson SE, Kristmundsdottir S, Halldorsson BV, Hreidarsson AB, Valdimarsson EM, Gudnason T, Benediktsson R, Steinthorsdottir V, Thorsteinsdottir U, Holm H, Stefansson K. Lipoprotein(a) Concentration and Risks of Cardiovascular Disease and Diabetes. J Am Coll Cardiol 2019; 74:2982-2994
  131. Ferrannini E, DeFronzo RA. Impact of glucose-lowering drugs on cardiovascular disease in type 2 diabetes. Eur Heart J 2015; 36:2288-2296
  132. Wulffele MG, Kooy A, de Zeeuw D, Stehouwer CD, Gansevoort RT. The effect of metformin on blood pressure, plasma cholesterol and triglycerides in type 2 diabetes mellitus: a systematic review. J Intern Med 2004; 256:1-14
  133. Ratner R, Goldberg R, Haffner S, Marcovina S, Orchard T, Fowler S, Temprosa M, Diabetes Prevention Program Research Group. Impact of intensive lifestyle and metformin therapy on cardiovascular disease risk factors in the diabetes prevention program. Diabetes Care 2005; 28:888-894
  134. Spanheimer R, Betteridge DJ, Tan MH, Ferrannini E, Charbonnel B. Long-term lipid effects of pioglitazone by baseline anti-hyperglycemia medication therapy and statin use from the PROactive experience (PROactive 14). Am J Cardiol 2009; 104:234-239
  135. Deeg MA, Buse JB, Goldberg RB, Kendall DM, Zagar AJ, Jacober SJ, Khan MA, Perez AT, Tan MH. Pioglitazone and rosiglitazone have different effects on serum lipoprotein particle concentrations and sizes in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2007; 30:2458-2464
  136. Goldberg RB, Kendall DM, Deeg MA, Buse JB, Zagar AJ, Pinaire JA, Tan MH, Khan MA, Perez AT, Jacober SJ. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2005; 28:1547-1554
  137. Sanchez-Garcia A, Simental-Mendia M, Millan-Alanis JM, Simental-Mendia LE. Effect of sodium-glucose co-transporter 2 inhibitors on lipid profile: A systematic review and meta-analysis of 48 randomized controlled trials. Pharmacol Res 2020; 160:105068
  138. Lamos EM, Levitt DL, Munir KM. A review of dopamine agonist therapy in type 2 diabetes and effects on cardio-metabolic parameters. Prim Care Diabetes 2016; 10:60-65
  139. Holt RI, Barnett AH, Bailey CJ. Bromocriptine: old drug, new formulation and new indication. Diabetes Obes Metab 2010; 12:1048-1057
  140. Raskin P, Cincotta AH. Bromocriptine-QR therapy for the management of type 2 diabetes mellitus: developmental basis and therapeutic profile summary. Expert Rev Endocrinol Metab 2016; 11:113-148
  141. Feingold KR. Cholesterol Lowering Drugs. 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.
  142. Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular Actions and Clinical Outcomes With Glucagon-Like Peptide-1 Receptor Agonists and Dipeptidyl Peptidase-4 Inhibitors. Circulation 2017; 136:849-870
  143. Kanbay M, Copur S, Siriopol D, Yildiz AB, Gaipov A, van Raalte DH, Tuttle KR. Effect of tirzepatide on blood pressure and lipids: A meta-analysis of randomized controlled trials. Diabetes Obes Metab 2023;
  144. Ginsberg HN. Diabetic dyslipidemia: basic mechanisms underlying the common hypertriglyceridemia and low HDL cholesterol levels. Diabetes 1996; 45 Suppl 3:S27-30
  145. Ginsberg HN, Zhang YL, Hernandez-Ono A. Metabolic syndrome: focus on dyslipidemia. Obesity (Silver Spring)2006; 14 Suppl 1:41S-49S
  146. Klop B, Elte JW, Cabezas MC. Dyslipidemia in obesity: mechanisms and potential targets. Nutrients 2013; 5:1218-1240
  147. Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med 2014; 371:32-41
  148. Tg,Hdl Working Group of the Exome Sequencing Project, National Heart LungBlood, Institute, Crosby J, Peloso GM, Auer PL, Crosslin DR, Stitziel NO, Lange LA, Lu Y, Tang ZZ, Zhang H, Hindy G, Masca N, Stirrups K, Kanoni S, Do R, Jun G, Hu Y, Kang HM, Xue C, Goel A, Farrall M, Duga S, Merlini PA, Asselta R, Girelli D, Olivieri O, Martinelli N, Yin W, Reilly D, Speliotes E, Fox CS, Hveem K, Holmen OL, Nikpay M, Farlow DN, Assimes TL, Franceschini N, Robinson J, North KE, Martin LW, DePristo M, Gupta N, Escher SA, Jansson JH, Van Zuydam N, Palmer CN, Wareham N, Koch W, Meitinger T, Peters A, Lieb W, Erbel R, Konig IR, Kruppa J, Degenhardt F, Gottesman O, Bottinger EP, O'Donnell CJ, Psaty BM, Ballantyne CM, Abecasis G, Ordovas JM, Melander O, Watkins H, Orho-Melander M, Ardissino D, Loos RJ, McPherson R, Willer CJ, Erdmann J, Hall AS, Samani NJ, Deloukas P, Schunkert H, Wilson JG, Kooperberg C, Rich SS, Tracy RP, Lin DY, Altshuler D, Gabriel S, Nickerson DA, Jarvik GP, Cupples LA, Reiner AP, Boerwinkle E, Kathiresan S. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med 2014; 371:22-31
  149. Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, Geary RS, Baker BF, Graham MJ, Crooke RM, Witztum JL. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med 2014; 371:2200-2206
  150. Arca M, D'Erasmo L, Minicocci I. Familial combined hypolipidemia: angiopoietin-like protein-3 deficiency. Curr Opin Lipidol 2020; 31:41-48
  151. Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, Grunfeld C. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res2004; 45:1169-1196
  152. Lara-Castro C, Fu Y, Chung BH, Garvey WT. Adiponectin and the metabolic syndrome: mechanisms mediating risk for metabolic and cardiovascular disease. Curr Opin Lipidol 2007; 18:263-270
  153. Feingold KR, Grunfeld C. The acute phase response inhibits reverse cholesterol transport. J Lipid Res 2010; 51:682-684
  154. Feingold KR, Grunfeld C. Effect of inflammation on HDL structure and function. Curr Opin Lipidol 2022; 27:521-530
  155. Lu B, Moser A, Shigenaga JK, Grunfeld C, Feingold KR. The acute phase response stimulates the expression of angiopoietin like protein 4. Biochem Biophys Res Commun 2010; 391:1737-1741
  156. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. 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.
  157. Rosenblat M, Karry R, Aviram M. Paraoxonase 1 (PON1) is a more potent antioxidant and stimulant of macrophage cholesterol efflux, when present in HDL than in lipoprotein-deficient serum: relevance to diabetes. Atherosclerosis 2006; 187:74-81
  158. Ahima RS. Adipose tissue as an endocrine organ. Obesity (Silver Spring) 2006; 14 Suppl 5:242S-249S
  159. Christou GA, Kiortsis DN. Adiponectin and lipoprotein metabolism. Obes Rev 2013; 14:939-949
  160. Rashid S, Kastelein JJ. PCSK9 and resistin at the crossroads of the atherogenic dyslipidemia. Expert Rev Cardiovasc Ther 2013; 11:1567-1577
  161. Yu YH, Ginsberg HN. Adipocyte signaling and lipid homeostasis: sequelae of insulin-resistant adipose tissue. Circ Res 2005; 96:1042-1052
  162. Cholesterol Treatment Trialists, Collaborato, 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
  163. Collins R, Armitage J, Parish S, Sleigh P, Peto R, Heart Protection Study Collaborative Group. 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
  164. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002; 360:7-22
  165. Wanner C, Krane V, Marz W, Olschewski M, Mann JF, Ruf G, Ritz E, German, Diabetes

            Dialysis Study, Investigators. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 2005; 353:238-248

  1. 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. Lancet2004; 364:685-696
  2. Nissen SE, Tuzcu EM, Schoenhagen P, Brown BG, Ganz P, Vogel RA, Crowe T, Howard G, Cooper CJ, Brodie B, Grines CL, DeMaria AN. Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: a randomized controlled trial. JAMA 2004; 291:1071-1080
  3. Nissen SE, Nicholls SJ, Sipahi I, Libby P, Raichlen JS, Ballantyne CM, Davignon J, Erbel R, Fruchart JC, Tardif JC, Schoenhagen P, Crowe T, Cain V, Wolski K, Goormastic M, Tuzcu EM. Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: the ASTEROID trial. JAMA 2006; 295:1556-1565
  4. Stegman B, Puri R, Cho L, Shao M, Ballantyne CM, Barter PJ, Chapman MJ, Erbel R, Libby P, Raichlen JS, Uno K, Kataoka Y, Nissen SE, Nicholls SJ. High-intensity statin therapy alters the natural history of diabetic coronary atherosclerosis: insights from SATURN. Diabetes Care 2014; 37:3114-3120
  5. Ahmed S, Cannon CP, Murphy SA, Braunwald E. Acute coronary syndromes and diabetes: Is intensive lipid lowering beneficial? Results of the PROVE IT-TIMI 22 trial. Eur Heart J 2006; 27:2323-2329
  6. Cannon CP, Braunwald E, McCabe CH, Rader DJ, Rouleau JL, Belder R, Joyal SV, Hill KA, Pfeffer MA, Skene AM, Pravastatin or Atorvastatin E, Infection Therapy-Thrombolysis in Myocardial Infarction I. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med 2004; 350:1495-1504
  7. LaRosa JC, Grundy SM, Waters DD, Shear C, Barter P, Fruchart JC, Gotto AM, Greten H, Kastelein JJ, Shepherd J, Wenger NK, Treating to New Targets I. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med 2005; 352:1425-1435
  8. Shepherd J, Barter P, Carmena R, Deedwania P, Fruchart JC, Haffner S, Hsia J, Breazna A, LaRosa J, Grundy S, Waters D. Effect of lowering LDL cholesterol substantially below currently recommended levels in patients with coronary heart disease and diabetes: the Treating to New Targets (TNT) study. Diabetes Care 2006; 29:1220-1226
  9. Pedersen TR, Faergeman O, Kastelein JJ, Olsson AG, Tikkanen MJ, Holme I, Larsen ML, Bendiksen FS, Lindahl C, Szarek M, Tsai J, Incremental Decrease in End Points Through Aggressive Lipid Lowering Study Group. High-dose atorvastatin vs usual-dose simvastatin for secondary prevention after myocardial infarction: the IDEAL study: a randomized controlled trial. JAMA 2005; 294:2437-2445
  10. Cholesterol Treatment Trialists Colloboration, 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
  11. Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, Forder P, Pillai A, Davis T, Glasziou P, Drury P, Kesaniemi YA, Sullivan D, Hunt D, Colman P, d'Emden M, Whiting M, Ehnholm C, Laakso M. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 2005; 366:1849-1861
  12. Jun M, Foote C, Lv J, Neal B, Patel A, Nicholls SJ, Grobbee DE, Cass A, Chalmers J, Perkovic V. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet 2010; 375:1875-1884
  13. Sacks FM, Carey VJ, Fruchart JC. Combination lipid therapy in type 2 diabetes. N Engl J Med 2010; 363:692-694; author reply 694-695
  14. Robins SJ, Collins D, Wittes JT, Papademetriou V, Deedwania PC, Schaefer EJ, McNamara JR, Kashyap ML, Hershman JM, Wexler LF, Rubins HB. Relation of gemfibrozil treatment and lipid levels with major coronary events: VA-HIT: a randomized controlled trial. JAMA 2001; 285:1585-1591
  15. Rubins HB, Robins SJ, Collins D, Nelson DB, Elam MB, Schaefer EJ, Faas FH, Anderson JW. Diabetes, plasma insulin, and cardiovascular disease: subgroup analysis from the Department of Veterans Affairs high-density lipoprotein intervention trial (VA-HIT). Arch Intern Med 2002; 162:2597-2604
  16. Koskinen P, Manttari M, Manninen V, Huttunen JK, Heinonen OP, Frick MH. Coronary heart disease incidence in NIDDM patients in the Helsinki Heart Study. Diabetes Care 1992; 15:820-825
  17. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study, a randomised study. Lancet 2001; 357:905-910
  18. Elkeles RS, Diamond JR, Poulter C, Dhanjil S, Nicolaides AN, Mahmood S, Richmond W, Mather H, Sharp P, Feher MD. Cardiovascular outcomes in type 2 diabetes. A double-blind placebo-controlled study of bezafibrate: the St. Mary's, Ealing, Northwick Park Diabetes Cardiovascular Disease Prevention (SENDCAP) Study. Diabetes Care 1998; 21:641-648
  19. Clofibrate and niacin in coronary heart disease. JAMA 1975; 231:360-381
  20. Canner PL, Furberg CD, Terrin ML, McGovern ME. Benefits of niacin by glycemic status in patients with healed myocardial infarction (from the Coronary Drug Project). Am J Cardiol 2005; 95:254-257
  21. 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
  22. 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
  23. Nissen SE, Menon V, Nicholls SJ, Brennan D, Laffin L, Ridker P, Ray KK, Mason D, Kastelein JJP, Cho L, Libby P, Li N, Foody J, Louie MJ, Lincoff AM. Bempedoic Acid for Primary Prevention of Cardiovascular Events in Statin-Intolerant Patients. JAMA 2023; 330:131-140
  24. Group AS, 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
  25. ACCORD Study Group, Group AES, Chew EY, Ambrosius WT, Davis MD, Danis RP, Gangaputra S, Greven CM, Hubbard L, Esser BA, Lovato JF, Perdue LH, Goff DC, Jr., Cushman WC, Ginsberg HN, Elam MB, Genuth S, Gerstein HC, Schubart U, Fine LJ. Effects of medical therapies on retinopathy progression in type 2 diabetes. N Engl J Med 2010; 363:233-244
  26. Keech AC, Mitchell P, Summanen PA, O'Day J, Davis TM, Moffitt MS, Taskinen MR, Simes RJ, Tse D, Williamson E, Merrifield A, Laatikainen LT, d'Emden MC, Crimet DC, O'Connell RL, Colman PG. Effect of fenofibrate on the need for laser treatment for diabetic retinopathy (FIELD study): a randomised controlled trial. Lancet 2007; 370:1687-1697
  27. 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
  28. Investigators AIM-HIGH, 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
  29. Guyton JR, Slee AE, Anderson T, Fleg JL, Goldberg RB, Kashyap ML, Marcovina SM, Nash SD, O'Brien KD, Weintraub WS, Xu P, Zhao XQ, Boden WE. Relationship of lipoproteins to cardiovascular events: the AIM-HIGH Trial (Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides and Impact on Global Health Outcomes). J Am Coll Cardiol 2013; 62:1580-1584
  30. 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. The New England journal of medicine 2014; 371:203-212
  31. 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 RMI. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387-2397
  32. Bohula EA, Morrow DA, Giugliano RP, Blazing MA, He P, Park JG, Murphy SA, White JA, Kesaniemi YA, Pedersen TR, Brady AJ, Mitchel Y, Cannon CP, Braunwald E. Atherothrombotic Risk Stratification and Ezetimibe for Secondary Prevention. J Am Coll Cardiol 2017; 69:911-921
  33. 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
  34. Lee YJ, Cho JY, You SC, Lee YH, Yun KH, Cho YH, Shin WY, Im SW, Kang WC, Park Y, Lee SY, Lee SJ, Hong SJ, Ahn CM, Kim BK, Ko YG, Choi D, Hong MK, Jang Y, Kim JS. Moderate-intensity statin with ezetimibe vs. high-intensity statin in patients with diabetes and atherosclerotic cardiovascular disease in the RACING trial. Eur Heart J 2023; 44:972-983
  35. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N Engl J Med 2017; 376:1713-1722
  36. 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;
  37. 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
  38. 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
  39. 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
  40. 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
  41. 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
  42. ORIGIN Trial Investigators OT, Bosch J, Gerstein HC, Dagenais GR, Diaz R, Dyal L, Jung H, Maggiono AP, Probstfield J, Ramachandran A, Riddle MC, Ryden LE, Yusuf S. n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N Engl J Med 2012; 367:309-318
  43. Ascend Study Collaborative Group, Bowman L, Mafham M, Wallendszus K, Stevens W, Buck G, Barton J, Murphy K, Aung T, Haynes R, Cox J, Murawska A, Young A, Lay M, Chen F, Sammons E, Waters E, Adler A, Bodansky J, Farmer A, McPherson R, Neil A, Simpson D, Peto R, Baigent C, Collins R, Parish S, Armitage J. Effects of n-3 Fatty Acid Supplements in Diabetes Mellitus. N Engl J Med 2018; 379:1540-1550
  44. Feingold KR. Triglyceride Lowering Drugs. 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.
  45. 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
  46. Saito Y, Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa 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 EPA on coronary artery disease in hypercholesterolemic patients with multiple risk factors: sub-analysis of primary prevention cases from the Japan EPA Lipid Intervention Study (JELIS). Atherosclerosis 2008; 200:135-140
  47. 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 2018;
  48. 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
  49. Nicholls SJ, Lincoff AM, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Pedersen TR, Ridker PM, Ray K, Karlson BW, Lundstrom T, Wolski K, Nissen SE. Assessment of omega-3 carboxylic acids in statin-treated patients with high levels of triglycerides and low levels of high-density lipoprotein cholesterol: Rationale and design of the STRENGTH trial. Clin Cardiol 2018; 41:1281-1288
  50. 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;
  51. 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
  52. Goff ZD, Nissen SE. N-3 polyunsaturated fatty acids for cardiovascular risk. Curr Opin Cardiol 2022; 37:356-363
  53. Mason RP, Sherratt SCR, Eckel RH. Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab 2023; 37:101681
  54. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Das SR, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Kosiborod M, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S158-S190
  55. 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
  56. Jellinger PS, Handelsman Y, Rosenblit PD, Bloomgarden ZT, Fonseca VA, Garber AJ, Grunberger G, Guerin CK, Bell DSH, Mechanick JI, Pessah-Pollack R, Wyne K, Smith D, Brinton EA, Fazio S, Davidson M. American Association of Clinical Endocrinologists and American College of Endocrinology Guidelines for Management of Dyslipidemia and Prevention of Cardiovascular Disease. Endocr Pract 2017; 23:1-87
  57. Handelsman Y, Jellinger PS, Guerin CK, Bloomgarden ZT, Brinton EA, Budoff MJ, Davidson MH, Einhorn D, Fazio S, Fonseca VA, Garber AJ, Grunberger G, Krauss RM, Mechanick JI, Rosenblit PD, Smith DA, Wyne KL. Consensus Statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the Management of Dyslipidemia and Prevention of Cardiovascular Disease Algorithm - 2020 Executive Summary. Endocr Pract 2020; 26:1196-1224
  58. 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
  59. Marx N, Federici M, Schutt K, Muller-Wieland D, Ajjan RA, Antunes MJ, Christodorescu RM, Crawford C, Di Angelantonio E, Eliasson B, Espinola-Klein C, Fauchier L, Halle M, Herrington WG, Kautzky-Willer A, Lambrinou E, Lesiak M, Lettino M, McGuire DK, Mullens W, Rocca B, Sattar N. 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes. Eur Heart J 2023;
  60. SCORE2-Diabetes Working Group. SCORE2-Diabetes: 10-year cardiovascular risk estimation in type 2 diabetes in Europe. Eur Heart J 2023; 44:2544-2556
  61. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394
  62. Feingold KR, Chait A. Approach to patients with elevated low-density lipoprotein cholesterol levels. Best Pract Res Clin Endocrinol Metab 2023; 37:101658
  63. Evert AB, Dennison M, Gardner CD, Garvey WT, Lau KHK, MacLeod J, Mitri J, Pereira RF, Rawlings K, Robinson S, Saslow L, Uelmen S, Urbanski PB, Yancy WS, Jr. Nutrition Therapy for Adults With Diabetes or Prediabetes: A Consensus Report. Diabetes Care 2019; 42:731-754
  64. 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.
  65. Look ARG, Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Kitabchi AE, Knowler WC, Lewis CE, Maschak-Carey BJ, Montez MG, Murillo A, Nathan DM, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Regensteiner JG, Rickman AD, Ryan DH, Safford M, Wadden TA, Wagenknecht LE, West DS, Williamson DF, Yanovski SZ. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med 2013; 369:145-154
  66. 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 Med2013; 368:1279-1290
  67. 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
  68. 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
  69. 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
  70. 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
  71. 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
  72. Adams TD, Gress RE, Smith SC, Halverson RC, Simper SC, Rosamond WD, Lamonte MJ, Stroup AM, Hunt SC. Long-term mortality after gastric bypass surgery. N Engl J Med 2007; 357:753-761
  73. Romeo S, Maglio C, Burza MA, Pirazzi C, Sjoholm K, Jacobson P, Svensson PA, Peltonen M, Sjostrom L, Carlsson LM. Cardiovascular events after bariatric surgery in obese subjects with type 2 diabetes. Diabetes Care 2012; 35:2613-2617
  74. Sjostrom L, Peltonen M, Jacobson P, Sjostrom CD, Karason K, Wedel H, Ahlin S, Anveden A, Bengtsson C, Bergmark G, Bouchard C, Carlsson B, Dahlgren S, Karlsson J, Lindroos AK, Lonroth H, Narbro K, Naslund I, Olbers T, Svensson PA, Carlsson LM. Bariatric surgery and long-term cardiovascular events. JAMA 2012; 307:56-65
  75. Sheng B, Truong K, Spitler H, Zhang L, Tong X, Chen L. The Long-Term Effects of Bariatric Surgery on Type 2 Diabetes Remission, Microvascular and Macrovascular Complications, and Mortality: a Systematic Review and Meta-Analysis. Obes Surg 2017; 27:2724-2732
  76. Fisher DP, Johnson E, Haneuse S, Arterburn D, Coleman KJ, O'Connor PJ, O'Brien R, Bogart A, Theis MK, Anau J, Schroeder EB, Sidney S. Association Between Bariatric Surgery and Macrovascular Disease Outcomes in Patients With Type 2 Diabetes and Severe Obesity. JAMA 2018; 320:1570-1582
  77. Farmer A, Montori V, Dinneen S, Clar C. Fish oil in people with type 2 diabetes mellitus. Cochrane Database Syst Rev 2001:CD003205
  78. Preiss D, Sattar N. Statins and the risk of new-onset diabetes: a review of recent evidence. Curr Opin Lipidol2011; 22:460-466
  79. 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
  80. 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
  81. Livingstone SJ, Looker HC, Akbar T, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Fuller JH, Colhoun HM. Effect of atorvastatin on glycaemia progression in patients with diabetes: an analysis from the Collaborative Atorvastatin in Diabetes Trial (CARDS). Diabetologia 2016; 59:299-306
  82. Chiu SW, Pratt CM, Feinn R, Chatterjee S. Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors and Ezetimibe on Risk of New-Onset Diabetes: A Systematic Review and Meta-Analysis of Large, Double-Blinded Randomized Controlled Trials. J Cardiovasc Pharmacol Ther 2020; 25:409-417
  83. Shah NP, McGuire DK, Cannon CP, Giugliano RP, Lokhnygina Y, Page CB, Tershakovec AM, Braunwald E, Blazing MA. Impact of Ezetimibe on New-Onset Diabetes: A Substudy of IMPROVE-IT. J Am Heart Assoc 2023; 12:e029593
  84. Kellick KA, Bottorff M, Toth PP, The National Lipid Association's Safety Task F. A clinician's guide to statin drug-drug interactions. J Clin Lipidol 2014; 8:S30-46
  85. Linz PE, Lovato LC, Byington RP, O'Connor PJ, Leiter LA, Weiss D, Force RW, Crouse JR, Ismail-Beigi F, Simmons DL, Papademetriou V, Ginsberg HN, Elam MB. Paradoxical reduction in HDL-C with fenofibrate and thiazolidinedione therapy in type 2 diabetes: the ACCORD Lipid Trial. Diabetes Care 2014; 37:686-693
  86. Ansquer JC, Foucher C, Rattier S, Taskinen MR, Steiner G. Fenofibrate reduces progression to microalbuminuria over 3 years in a placebo-controlled study in type 2 diabetes: results from the Diabetes Atherosclerosis Intervention Study (DAIS). Am J Kidney Dis 2005; 45:485-493
  87. Davis TM, Ting R, Best JD, Donoghoe MW, Drury PL, Sullivan DR, Jenkins AJ, O'Connell RL, Whiting MJ, Glasziou PP, Simes RJ, Kesaniemi YA, Gebski VJ, Scott RS, Keech AC, Event Lowering in Diabetes Study i. Effects of fenofibrate on renal function in patients with type 2 diabetes mellitus: the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study. Diabetologia 2011; 54:280-290
  88. Rajamani K, Colman PG, Li LP, Best JD, Voysey M, D'Emden MC, Laakso M, Baker JR, Keech AC. Effect of fenofibrate on amputation events in people with type 2 diabetes mellitus (FIELD study): a prespecified analysis of a randomised controlled trial. Lancet 2009; 373:1780-1788
  89. Aldridge MA, Ito MK. Colesevelam hydrochloride: a novel bile acid-binding resin. Ann Pharmacother 2001; 35:898-907
  90. Bays HE. Colesevelam hydrochloride added to background metformin therapy in patients with type 2 diabetes mellitus: a pooled analysis from 3 clinical studies. Endocr Pract 2011; 17:933-938
  91. Song WL, FitzGerald GA. Niacin, an old drug with a new twist. J Lipid Res 2013; 54:2586-2594
  92. Elam MB, Hunninghake DB, Davis KB, Garg R, Johnson C, Egan D, Kostis JB, Sheps DS, Brinton EA. Effect of niacin on lipid and lipoprotein levels and glycemic control in patients with diabetes and peripheral arterial disease: the ADMIT study: A randomized trial. Arterial Disease Multiple Intervention Trial. JAMA 2000; 284:1263-1270
  93. Grundy SM, Vega GL, McGovern ME, Tulloch BR, Kendall DM, Fitz-Patrick D, Ganda OP, Rosenson RS, Buse JB, Robertson DD, Sheehan JP, Diabetes Multicenter Research Group. Efficacy, safety, and tolerability of once-daily niacin for the treatment of dyslipidemia associated with type 2 diabetes: results of the assessment of diabetes control and evaluation of the efficacy of niaspan trial. Archives of internal medicine 2002; 162:1568-1576
  94. Weintraub H. Update on marine omega-3 fatty acids: management of dyslipidemia and current omega-3 treatment options. Atherosclerosis 2013; 230:381-389
  95. Khan SU, Rahman H, Okunrintemi V, Riaz H, Khan MS, Sattur S, Kaluski E, Lincoff AM, Martin SS, Blaha MJ. Association of Lowering Low-Density Lipoprotein Cholesterol With Contemporary Lipid-Lowering Therapies and Risk of Diabetes Mellitus: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2019; 8:e011581
  96. Carugo S, Sirtori CR, Corsini A, Tokgozoglu L, Ruscica M. PCSK9 Inhibition and Risk of Diabetes: Should We Worry? Curr Atheroscler Rep 2022; 24:995-1004
  97. Wang X, Zhang Y, Tan H, Wang P, Zha X, Chong W, Zhou L, Fang F. Efficacy and safety of bempedoic acid for prevention of cardiovascular events and diabetes: a systematic review and meta-analysis. Cardiovasc Diabetol2020; 19:128
  98. Bays HE, Banach M, Catapano AL, Duell PB, Gotto AM, Jr., Laufs U, Leiter LA, Mancini GBJ, Ray KK, Bloedon LT, Sasiela WJ, Ye Z, Ballantyne CM. Bempedoic acid safety analysis: Pooled data from four phase 3 clinical trials. J Clin Lipidol 2020; 14:649-659 e646
  99. Jenkins DJ, Jones PJ, Lamarche B, Kendall CW, Faulkner D, Cermakova L, Gigleux I, Ramprasath V, de Souza R, Ireland C, Patel D, Srichaikul K, Abdulnour S, Bashyam B, Collier C, Hoshizaki S, Josse RG, Leiter LA, Connelly PW, Frohlich J. Effect of a dietary portfolio of cholesterol-lowering foods given at 2 levels of intensity of dietary advice on serum lipids in hyperlipidemia: a randomized controlled trial. JAMA 2011; 306:831-839
  100. Feingold KR. Utility of Advanced Lipoprotein Testing in Clinical Practice. 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)2023.
  101. Orringer CE, Jacobson TA, Maki KC. National Lipid Association Scientific Statement on the use of icosapent ethyl in statin-treated patients with elevated triglycerides and high or very-high ASCVD risk. J Clin Lipidol 2019; 13:860-872

 

 

 

Role Of Glucose And Lipids In The Atherosclerotic Cardiovascular Disease In Patients With Diabetes

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in both men and women with T1DM and T2DM. In patients with T1DM, intensive glycemic control results in a reduction in ASCVD. However, intensive glycemic control does not have a major impact in reducing ASCVD in patients with T2DM. Metformin, pioglitazone, SGLT2 inhibitors, and certain GLP-1 receptor agonists have been shown to decrease major cardiovascular events in patients with T2DM to a greater extent than other treatment modalities. In patients with T2DM other risk factors including, hypertension and dyslipidemia, play a major role in inducing ASCVD and control of these risk factors is paramount. In patients with T1DM in good glycemic control, the lipid profile is very similar to the general population. In contrast, in patients with T2DM, even with good glycemic control, there are frequently lipid abnormalities (elevated TG and non-HDL-C, decreased HDL-C, and an increase in small dense LDL). In both T1DM and T2DM, poor glycemic control increases TG levels and decreases HDL-C levels with modest effects on LDL-C levels.  Extensive studies have demonstrated that statins decrease ASCVD in patients with diabetes. Treatment with high doses of potent statins reduces ASCVD events to a greater extent than low dose statin therapy. Adding fibrates or niacin to statin therapy has not been shown to further decrease ASCVD events. In contrast, studies have shown that the combination of a statin and ezetimibe or a statin and a PCSK9 inhibitor result in a greater decrease in ASCVD events than statins alone. Studies have suggested that EPA, an omega-3-fatty acid, when added to statins also reduces ASCVD events but this result is controversial. In statin intolerant patients with T2DM bempedoic acid decreases ASCVD events. Current recommendations state that most patients with diabetes should be on statin therapy. In certain patients with diabetes ezetimibe, PCSK9 inhibitors, and bempedoic acid can play a role in reducing ASCVD.  

 

INTRODUCTION

 

Atherosclerotic cardiovascular disease (ASCVD) is a major cause of morbidity and mortality in both men and women with diabetes (1-5). In addition to coronary disease, ASCVD includes stroke and peripheral arterial disease (PAD).  PAD is common in diabetes, may be the first presentation of ASCVD and should be recognized as needing aggressive treatment of risk factors. The risk of ASCVD is increased approximately 2-fold in men and 3-4-fold in women (2-4,6,7). In the Framingham study, the annual rate of ASCVD was similar in men and women with diabetes, emphasizing that woman with diabetes need as aggressive preventive treatment as men with diabetes (2,6). In addition, several but not all studies, have shown that patients with diabetes with no history of ASCVD have a similar risk of having a myocardial infarction as non-diabetic patients who have a history of ASCVD, i.e., diabetes is an equivalent risk factor as a history of a previous cardiovascular event (8,9). The duration of diabetes and the presence of other risk factors or complications of diabetes likely determine whether a patient with diabetes has a risk equivalent to patients with a history of previous ASCVD events (10,11). In one study patients with T2DM who had the following risk factors within the target range, HbA1c, LDL-C, albuminuria, smoking, and blood pressure, the risk of an acute myocardial infarction or stroke was similar to individuals without diabetics (12). Moreover, numerous studies have shown that patients with diabetes who have ASCVD are at a very high risk of having another event, indicating that this population of patient’s needs especially aggressive preventive measures (1,8). This increased risk for the development of ASCVD in patients with diabetes is seen both in populations where the prevalence of ASCVD is high (Western societies) and low (for example, Japan) (2). However, in societies where the prevalence of ASCVD is low, the contribution of ASCVD as a cause of morbidity and mortality in patients with diabetes is relatively low compared to Western societies (2).

 

While the database is not as robust, the evidence indicates that patients with T1DM are also at high risk for the development of ASCVD (1,13-15). Interestingly, women with T1DM have twice the excess risk of fatal and nonfatal vascular events compared to men with T1DM (16,17). Additionally, developing T1DM at a young age increases the risk of ASCVD to a greater degree than late onset T1DM (17). Approximately 50% of patients with T1DM are obese or overweight and between 8% and 40% meet the criteria for the metabolic syndrome, which increases their risk of developing ASCVD (18).

 

While the development of diabetes at a young age increases the risk of ASCVD in patients with both T1DM and T2DM the deleterious impact is greater in patients with T2DM (19). Lastly, in patients with both T1DM and T2DM the presence of renal disease increases the risk of ASCVD (4,14). Of note is that the risk of developing ASCVD events in patients with diabetes has decreased recently, most likely due to better lipid and blood pressure control, which again reinforces the need to aggressively treat these risk factors in patients with diabetes (5,7,20). 

 

ROLE OF GLYCEMIC CONTROL

 

Epidemiological studies have shown an association between the level of glycemic control and the development of ASCVD in both T1DM and T2DM (1,4,5,21,22). However, the association of glycemic control with ASCVD is considerably weaker than the association of glycemic control with the microvascular complications of diabetes, such as retinopathy and nephropathy (4). It must be recognized that epidemiological studies can only demonstrate associations and that confounding variables could account for the association between poor glycemic control and ASCVD. For example, patients with poor glycemic control may not undertake other preventive measures that could reduce ASCVD such as exercise, healthy diet, etc. Furthermore, the patients with poor glycemic control may have less compliance with therapies that reduce lipids and blood pressure. Therefore, randomized studies are essential in determining the role of glycemic control on ASCVD. 

 

Early randomized studies, such as the University Group Diabetes Project (UGDP) and VA cooperative study, did not demonstrate a reduction in cardiovascular events in patients who were aggressively treated for glucose control (23-25). In fact, the data from these early studies suggested that improvements in glycemic control (VA cooperative study) or the use of certain drugs to treat diabetes (oral sulfonylureas in UGDP) may actually increase the risk of ASCVD.

 

Diabetes Control and Complications Trial (DCCT) and Kumamoto Studies

 

Latter studies, the DCCT in patients with T1DM and the Kumamoto study in patients with T2DM, while finding a decrease in ASCVD events (DCCT 41% decrease) in the subjects randomized to improved glycemic control did not have enough ASCVD events to demonstrate a statistically significant reduction (DCCT studied a population at low risk for ASCVD and the Kumamoto study had a very small number of subjects) (26-28). In contrast, both the DCCT and the Kumamoto study clearly demonstrated that improvements in glycemic control resulted in a reduction in microvascular disease (26-28). However, the long- term follow-up of the DCCT has demonstrated that those in the intensive glycemic control group had a decrease in ASCVD in subsequent years (29,30). The initial DCCT compared intensive vs. conventional therapy for a mean of 6.5 years. At the end of the study, a very large proportion of subjects agreed to participate in a follow-up observational study (Epidemiology of Diabetes Interventions and Complications- EDIC). During this follow-up period, glycemic control was relatively similar between the intensive therapy and conventional therapy group (glycosylated hemoglobin 7.9% vs. 7.8%) but during the actual trial there was a large difference in glycosylated hemoglobin levels (7.4% vs. 9.1%). After a mean 17 years of observation, the risk of any cardiovascular event was reduced by 42% and the risk of nonfatal myocardial infarction (MI), stroke, or death from ASCVD was reduced by 57% in the intensive control group. This study demonstrates that being in the intensive glycemic control group (for 6.5 of the 17 years of observation) is sufficient to have long-term beneficial effects on the risk of developing ASCVD in patients with T1DM. This beneficial effect was not entirely due to the prevention of microvascular complications as the differences between the intensive and conventional treatment groups for ASCVD persisted after adjusting for microalbuminuria and albuminuria. When an outcome of improved glycemic control is seen, or persists for years after the trial is over the phenomenon is called a “metabolic memory” effect.

 

UK Prospective Diabetes Study (UKPDS)

 

A similar finding has been reported with regard to T2DM. The UKPDS studied a large number of newly diagnosed patients with T2DM at risk for ASCVD. In this study improved glycemic control, with either insulin or sulfonylureas, reduced ASCVD by 16%, which just missed being statistically significant (p=0.052) (31). In the UKPDS, the improvement in glycemic control was modest (HbA1c reduced by approximately 0.9%) and the 16% reduction in ASCVD was in the range predicted based on epidemiological studies. The results of a 10-year follow-up of the UKPDS study have been reported (total duration of observation 25 years) (32). After termination of the study, glycosylated hemoglobin levels became very similar between the control and treatment groups. Nevertheless, risk reductions for MI became statistically significant for the insulin and the sulfonylurea group compared to controls (15% decrease, p=0.01).

 

DiGami Studies

 

Similarly, the DiGami study, which used insulin infusion during the peri-MI period to improve glycemic control followed by long-term glycemic control, demonstrated that survival post MI was significantly improved by good glycemic control (33). While this study focused on a highly-selected population and time period (patients undergoing a MI), the results are consistent with the hypothesis that improvements in glycemic control will reduce ASCVD. However, the DiGami 2 study did not confirm the benefits of tight glucose control beginning in the peri-MI period on outcomes (34). It must be noted though that the differences in glucose control achieved in DiGami 2 were much smaller than planned and the number of patients recruited was less than anticipated. Together these deficiencies could account for the failure to demonstrate significant differences in ASCVD events in this study.

 

ACCORD Study

 

Because of the need for more definitive data on the effect of glycemic control on ASCVD in T2DM, three large randomized trials, the ACCORD, ADVANCE, and VA Diabetes Trial, have been carried out. Much to everyone’s surprise and disappointment, improvement in glycemic control did not clearly result in a significant reduction in ASCVD in these trials.

 

The ACCORD study randomized 10,251 subjects with T2DM in the US and Canada with either a history of ASCVD or at increased risk for the development of ASCVD (35). Multiple different treatment protocols were used with the goal of achieving an A1c level < 6% in the intensive group and between 7-7.9% in the standard glycemic control group. During the trial the A1c levels were 6.4% in the intensive group and 7.5% in the standard group. As expected, the use of insulin therapy was much greater in the intensive group, as was the occurrence of hypoglycemia and weight gain. After a mean duration of 3.5 years this study was stopped early by the data safety monitoring board due to an increased all-cause mortality in the intensive treatment group (1.41% vs. 1.14% per year; hazard ratio 1.22 CI 1.01- 1.46). The primary outcome (MI, stroke, ASCVD death) was reduced by 10% in the intensive control group but this was not statistically significant (p=0.16). Of note, intensive glycemic control reduced the incidence of any MI (i.e. fatal or non-fatal) by 16%, nonfatal MI by 19%, coronary revascularization by 16%, and unstable angina by 19% (36).The explanation for the increased death rate in the intensive treatment arm remains unknown, but it has been speculated that the increased deaths might have been due to hypoglycemia, weight gain, too rapidly lowering A1c levels, or unrecognized drug toxicity. Long term follow-up of the ACCORD study did not reveal any beneficial effects on the primary outcome (nonfatal MI, nonfatal stroke, or cardiovascular death), death from any cause, and an expanded composite outcome that included all-cause death in the intensive glycemic control group (37).

 

ADVANCE Study

 

The ADVANCE study randomized 11,140 subjects with T2DM in Europe, Asia, Australia/New Zealand, and Canada who either had ASCVD or at least one other risk factor for ASCVD (38). In the intensive group the goal A1c was <6.5%. The achieved A1c levels during the trial were 6.3% in the intensive group and 7.3% in the standard treatment group. Of note is that compared to the ACCORD study, less insulin use was required to achieve these A1c levels. With regard to macrovascular disease (MI, stroke, and cardiovascular death), no significant differences were observed between the intensive and standard treatment groups (HR 0.94, CI 0.84-1.06, p=0.32). In contrast to the ACCORD trial, no increase in overall or cardiovascular mortality in the intensive treatment group was observed in the ADVANCE study. Long term follow-up did not demonstrate a decrease in the risk of death from any cause or major macrovascular events between the intensive-glucose-control group and the standard-glucose-control group (39).

 

VA Diabetes Trial

 

The VA Diabetes Trial randomized 1,791 subjects with poor glycemic control on maximal oral agent therapy or insulin (entry A1c 9.4%) (40). In the intensive group, the goal A1c was <6.0%. The achieved A1c levels during the trial were 6.9% in the intensive group and 8.5% in the standard treatment group. Similar to the other trials, a significant reduction in ASCVD was not observed in the intensive glycemic control group (HR 0.88, CI 0.74-1.05, p=0.12). Notably there were more ASCVD deaths and sudden deaths in the intensive treatment group, but this increase was not statistically significant. With long-term follow-up (approximately 10 years), the intensive-therapy group had a significantly lower risk of MI, stroke, congestive heart failure, amputation for ischemic gangrene, or cardiovascular-related death than did the standard-therapy group (hazard ratio, 0.83; P=0.04 (41). However, there was no reduction in cardiovascular or total mortality. Furthermore, with a longer period of follow-up (15 years) the risks of major cardiovascular events or death were not lower in the intensive-therapy group than in the standard-therapy group (42). In a careful analysis it was noted that that the risk of cardiovascular events was 17% lower in the intensive treatment group than in the standard control group during the approximate 10-year period when there was a separation of the glycated hemoglobin curves between the two groups, suggesting that glycemic control was reducing the risk of cardiovascular events (42).

 

Meta-analyses

 

In a meta-analysis of 6 randomized studies (UKPDS, Kumamoto, VA Feasibility study, ACCORD, ADVANCE, and VA Diabetes Trial) of intensive vs. conventional glycemic control in patients with T2DM (27,654 patients) there was no significant effect of tight blood glucose control on all-cause mortality (RR 1.03; 95% CI 0.90-1.17), cardiovascular mortality (RR 1.04; 95% CI 0.83-1.29), or nonfatal stroke (RR 1.02; 95% CI 0.88-1.17) but tight glucose control reduced the risk for nonfatal MI (RR 0.85; 95% CI 0.76-0.95) (43). In a meta-analysis of 4 studies (UKPDS, ACCORD, ADVANCE, and VA Diabetes Trial) the primary outcome was the composite of death from cardiovascular causes (including sudden death), non-fatal MI and non-fatal stroke, which was decreased by 9% (HR 0.91, 95% CI 0.84–0.99) in the intensive control group (44). Of note the risk of non-fatal/fatal MI was reduced by 15% (HR 0.85, 95% CI 0.76–0.94) in the intensive group without significant reductions in the risk of non-fatal/fatal stroke, fatal heart failure, all-cause mortality, or cardiovascular death.  

 

Limitations of Cardiovascular Outcome Studies

 

Thus, while the epidemiological data strongly suggests that glycemic control would favorably impact ASCVD the recent randomized trials that were designed specifically to prove this hypothesis have failed to definitively demonstrate a clear link. There are several explanations for why these trials may not have worked as planned.

 

First, in the ACCORD, ADVANCE, and VA Diabetes Trial, other ASCVD risk factors were aggressively treated (lipid and BP lowering, ASA therapy). As a result of these treatments, the actual number of ASCVD events was considerably less than expected in these trials. The lower event rate may have reduced the ability to see a beneficial effect of glucose control. Additionally, the beneficial effects of glucose control maybe more robust if other risk factors are not aggressively controlled. In this regard, it is worth noting that in the earlier UKPDS, which showed that improved glycemic control reduced ASCVD events, both BP and lipids were not aggressively treated by current standards (systolic BP 135-140mm Hg, LDL-C 135-142mg/dL), which could be why this older trial demonstrated a benefit of improving glycemic control on ASCVD.

 

Second, these three recent trials were comparing relatively low A1c levels in both the intensive and usual control groups (A1c in intensive from ~6.4-6.9% and usual control group from ~7.0-8.4%). It is likely that both levels are on the “flatter” portion of the glycemic control-cardiovascular risk curve and that if one compared patients with intensive glycemic control with a control group with higher A1c values one would see more impressive results. If the difference in A1c levels were greater in the intensive and control groups the likelihood of seeing a reduction in cardiovascular events in the intensive group would be enhanced.

 

Third, all three trials were carried out by initiating tight control in patients with long standing diabetes who either had pre-existing ASCVD or were at high risk for ASCVD. It is possible that patients with a different clinical profile would be more likely to benefit from intensive glucose control. Subgroup analysis from these trials have suggested that patients with a shorter duration of diabetes, less severe diabetes, or the absence of pre-existing ASCVD actually benefited from intensive control. It may be that glycemic control is most important prior to the development of significant atherosclerosis. Clearly additional studies on different types of patients (i.e., newly diagnosed without evidence of ASCVD) will be necessary to definitively determine the role of glycemic control in different diabetic populations.

 

Fourth, the duration of these studies was relatively short and it is possible that a much longer duration of glycemic control is required to show benefits on ASCVD. In the UKPDS study the beneficial effects of intensive glucose control was not statistically significant at the end of the study but with an extended duration of follow-up (15-25 years) became statistically significant.

 

Fifth, it may be that glycemic control will be more important in patients with T1DM where abnormalities in glucose metabolism are a major reason for the increased risk of ASCVD. In contrast, patients with T2DM have multiple risk factors for ASCVD (dyslipidemia, hypertension, inflammation, insulin resistance, coagulation disorders, etc.) and glucose may play only a minor role in the increased risk. The differences in other cardiovascular risk factors could account for why intensive glycemic control produced a marked reduction in ASCVD in the DCCT (T1DM trial) and had only minimal effects in the trials carried out in patients with T2DM.

 

Finally, it is possible that certain treatments have side effects that mask the beneficial effects of glucose control. For example, hypoglycemia and weight gain could counterbalance the beneficial effects of improvements in glycemic control. It is possible that different treatment strategies could lead to more profound benefits (see below). 

 

Summary

 

Thus, the currently available data do not definitively indicate that glycemic control will have major effects on reducing ASCVD in patients with T2DM. Furthermore, there are concerns that too tight control in patients with advanced disease could be harmful. In contrast, in patients with T1DM intensive glucose control appears to have a major beneficial effect on ASCVD based on the results of the DCCT.

 

THE EFFECT OF GLUCOSE LOWERING DRUGS ON ASCVD

 

Metformin

 

In the UKPDS, metformin, while producing a similar improvement in glycemic control as insulin or sulfonylureas, markedly reduced ASCVD by approximately 40% (45). In the ten-year follow-up the patients randomized to metformin in the UKPDS continued to show a reduction in MI and all-cause mortality (32). Two other randomized controlled trials have also demonstrated cardiovascular benefits with metformin therapy.

 

A study by Kooy et al compared the effect of adding metformin or placebo in overweight or obese patients already on insulin therapy (46). After a mean follow-up of 4.3 years this study observed a reduction in macrovascular events (HR 0.61 CI- 0.40-0.94, p=0.02), which was partially accounted for by metformin’s beneficial effects on weight. In this study the difference in A1c between the metformin and placebo group was only 0.3%.

 

Hong et al randomized non-obese patients with coronary artery disease to glipizide vs. metformin therapy for three years (47). A1c levels were similar, but there was a marked reduction in cardiovascular events in the metformin treated group (HR 0.54 CI 0.30- 0.90, p=0.026).

 

In contrast, long term follow-up (21 years) of individuals in the Diabetes Prevention Program with “pre-diabetes” did not demonstrate a reduction in cardiovascular event in individuals treated with metformin (48). A reduction in the use of metformin when the formal study ended after 3 years coupled with out-of-study metformin use over time may have diluted the potential effects of metformin therapy.

 

Support for the beneficial effects of metformin on atherosclerosis comes from long term follow-up of the Diabetes Prevention Program, which compared the effect of lifestyle changes or metformin in patients at high risk of developing diabetes (49). Coronary artery calcium scores were measured on average 13-14 years after randomization (49). There were no differences in coronary artery calcium scores between the lifestyle and placebo groups. However, in males, coronary artery calcium scores were significantly lower in the metformin group vs. the placebo group. In females treated with metformin coronary artery calcium scores were similar to the placebo group. The absence of a beneficial effect of metformin in women could be due to the lower baseline coronary artery calcium scores making it more difficult to demonstrate a beneficial effect. In HIV-infected patients with the metabolic syndrome metformin similarly reduced the progression of coronary artery calcium scores (50).

 

Thus, while there are no large cardiovascular outcome trials with metformin, together, the above results suggest that metformin may reduce ASCVD and that this effect is not due to improving glucose control. Metformin decreases weight or prevents weight gain and lowers lipid levels and these or other non-glucose effects may account for the beneficial effects on ASCVD.

 

Sulfonylureas

 

Based on the University Group Diabetes Project (UGDP) sulfonylureas carry a warning regarding an increased risk of ASCVD (24,25). However, the UKPDS studied a large number of newly diagnosed patients with T2DM at risk for ASCVD and in this study improved glycemic control with sulfonylureas reduced ASCVD by approximately 16%, which just missed being statistically significant (p=0.052) (31). In the UKPDS, A1c was reduced by approximately 0.9% and the 16% reduction in ASCVD was in the range predicted based on epidemiological studies. Thus, the reduction in cardiovascular events was likely due to improvements in glycemic control and not a direct benefit of sulfonylurea treatment. In support of this conjecture is that in the UKPDS, insulin treatment resulted in a similar decrease in A1c levels and reduction in cardiovascular events (31). Additionally, a large randomized cardiovascular outcome study (Carolina Study) reported that linagliptin, a DPP-4 inhibitor, and glimepiride, a sulfonylurea, had similar effects on cardiovascular events (hazard ratio 0.98) (51). Similarly, in the ADVANCE trial patients in the intensive therapy group were randomized to gliclazide and as noted above the occurrence of cardiovascular events was similar to the control group (38).Taken together these results suggest that sulfonylureas have a neutral effect on ASCVD.

 

Meglitinides

 

The Navigator study was a double-blind, randomized clinical trial in 9,306 individuals with impaired glucose tolerance and either ASCVD or cardiovascular risk factors who received nateglinide (up to 60 mg three times daily) or placebo (52). After 5 years, nateglinide administration did not alter the incidence of cardiovascular outcomes suggesting that meglitinides do not have an adverse or beneficial effect on cardiovascular events.

 

Thiazolidinediones

 

Studies with pioglitazone have suggested a beneficial effect on ASCVD. The PROactive study was a randomized controlled trial that examined the effect of pioglitazone vs. placebo over a 3-year period in T2DM with pre-existing macrovascular disease (53). With regard to the primary endpoint (a composite of all-cause mortality, non-fatal MI including silent MI, stroke, acute coronary syndrome, endovascular or surgical intervention in the coronary or leg arteries, and amputation above the ankle), there was a 10% reduction in events in the pioglitazone group but this difference was not statistically significant (p=0.095). It should be noted that both leg revascularization and leg amputations are not typical primary end points in ASCVD trials and these could be affected by pioglitazone induced edema. When one focuses on standard ASCVD endpoints, the pioglitazone treated group did demonstrate a 16% reduction in the main secondary endpoint (composite of all-cause mortality, non-fatal MI, and stroke) that was statistically significant (p=0.027). In the pioglitazone treated group, blood pressure, A1c, triglyceride, and HDL-C levels were all improved compared to the placebo group making it very likely that the mechanism by which pioglitazone decreased vascular events was multifactorial.

 

A multicenter, double-blind trial (IRIS Trial), randomly assigned 3,876 patients with insulin resistance (defined as score of more than 3.0 on the homeostasis model assessment  of insulin resistance [HOMA-IR] index) but without diabetes and a recent ischemic stroke or TIA to treatment with either pioglitazone (target dose, 45 mg daily) or placebo (54). After 4.8 years, the primary outcome of fatal or nonfatal stroke or MI occurred in 9.0% of the pioglitazone group and 11.8% of the placebo group (hazard ratio 0.76; P=0.007). All components of the primary outcome were reduced in the pioglitazone treated group. Fasting glucose, fasting TG, and systolic and diastolic blood pressure were lower while HDL-C and LDL-C levels were higher in the pioglitazone group than in the placebo group. Although this study excluded patients with diabetes the results are consistent with and support the results of a protective effect of pioglitazone observed in the PROactive study.

 

In contrast, the TOSCA.IT study compared the effect of pioglitazone vs. sulfonylurea on ASCVD and did not observe a reduction in events with pioglitazone treatment (55). Patients with T2DM (n= 3028), inadequately controlled with metformin monotherapy (2-3 g per day), were randomized to pioglitazone or sulfonylurea and followed for a median of 57 months. Only 11% of the participants had a previous cardiovascular event. The primary outcome, was a composite of first occurrence of all-cause death, non-fatal MI, non-fatal stroke, or urgent coronary revascularization and occurred in 6.8% of the patients treated with pioglitazone and 7.2% of the patients treated with a sulfonylurea (HR 0.96; NS) (55). Limitations of this study are the small number of events due to the low-risk population studied and the relatively small number of participants. Additionally, 28% of the subjects randomized to pioglitazone prematurely discontinued the medication. Furthermore, it should be noted that when patients in this study were analyzed based on the risk of developing ASCVD those at high risk had a marked reduction in events when treated with pioglitazone compared to the sulfonylurea (56).Thus, the results of this study should be interpreted with caution.

 

Further support for the beneficial effects of pioglitazone on atherosclerosis is provided by studies that have examined the effect of pioglitazone on carotid intima-medial thickness. Both the Chicago and Pioneer studies demonstrated favorable effects on carotid intima-medial thickness in patients treated with pioglitazone compared to patients treated with sulfonylureas (57,58). Similarly, Periscope, a study that measured atheroma volume in the coronary arteries by intravascular ultrasonography, also demonstrated less atherosclerosis in the pioglitazone treated group compared to patients treated with sulfonylureas (59).

 

While the data from a variety of different types of studies strongly suggests that pioglitazone is anti-atherogenic, the results with rosiglitazone are different. Several meta-analyses of small and short-duration rosiglitazone trials suggested that rosiglitazone was associated with an increased risk of adverse cardiovascular outcomes (60,61). However, the final results of the RECORD study, a randomized trial that was specifically designed to compare the effect of rosiglitazone vs. either metformin or sulfonylurea therapy as a second oral drug in those receiving either metformin or a sulfonylurea on ASCVD events, have been published and did not reveal a difference in ASCVD death, MI, or stroke (62-64). Similarly, an analysis of patients on rosiglitazone in the BARI 2D trial also did not suggest an increase or decrease in ASCVD events in the patients treated with rosiglitazone (65). Thus, while the available data suggests that pioglitazone is anti-atherogenic, the data for rosiglitazone suggests a neutral effect. Whether these differences between pioglitazone and rosiglitazone are accounted for by their differential effects on lipid levels are unknown (see below for information on the effects of these drugs on lipid levels).

 

Numerous studies have shown that both pioglitazone and rosiglitazone increase the risk of heart failure (66).

 

DPP4 Inhibitors

 

Because of the importance of ASCVD in patients with diabetes the FDA is requiring manufacturers of new drugs to treat diabetes to carry out studies addressing ASCVD endpoints. The effect of the DPP4 inhibitors saxagliptin, alogliptin, sitagliptin, and linagliptin on ASCVD endpoints has been reported. In the saxagliptin study (SAVOR‐TIMI 53 trial), 16,492 patients with T2DM who had a history of cardiovascular events or who were at high risk were randomized to saxagliptin or placebo for 2.1 years (67). Saxagliptin did not increase or decrease cardiovascular death, MI, or ischemic stroke. Interestingly more patients treated with saxagliptin were admitted to the hospital for heart failure. The risk of heart failure with saxagliptin was greatest in patients at a high overall risk of heart failure (i.e., history of heart failure, impaired renal function, or elevated baseline levels of NT-proBNP) (68). Additionally, in the patients treated with saxagliptin the increase in heart failure was an early event with a 6-month rate of 1.1% vs. 0.6% in the placebo group (HR 1.80, p=0·001) and a 12-month rate of 1·9% vs. 1·3% (1.46; p=0.002) (68). In contrast, after 12 months no difference in the rate of heart failure was observed in the saxagliptin and placebo groups indicating that the development of heart failure is an early event (68).

 

In the alogliptin trial (EXAMINE), 5,380 patients with either an acute MI or unstable angina within the previous 15-90 days were randomized to alogliptin or placebo and followed for a median of 18 months (69). As seen in the saxagliptin study the rates of ASCVD events were similar in the alogliptin and placebo groups. The risk of hospitalization for heart failure was not statistically increased in the entire subset of patients treated with alogliptin (70). However, the hazard ratio for the subgroup of patients without heart failure at baseline was 1.76, p=0.026) (70). 

 

In the sitagliptin trial (TECOS), 14,671 patients with established ASCVD were randomized to sitagliptin or placebo for 3 years (71). Sitagliptin did not decrease the risk of major adverse cardiovascular events or increase hospitalization for heart failure. Finally, in the linagliptin trial (CARMELINA), 6,979 patients at high risk for ASCVD were randomized to linagliptin or placebo for a median follow-up of 2.2 years (72). As in the other DPP4 inhibitor studies, linagliptin did not have a beneficial effect on ASCVD events. Additionally, linagliptin did not increase the risk of hospitalization for heart failure. Thus, these results indicate that DPP4 inhibitors do not increase or decrease ASCVD. The extent to which specific DPP4 inhibitors affect heart failure needs further investigation.

 

SGLT2 Inhibitors

 

EMPA-REG OUTCOME TRIAL   

 

The effects of empagliflozin on cardiovascular morbidity and mortality in patients with T2DM has been reported (73). In this study, 7,020 patients at high risk for ASCVD were randomly assigned to receive 10 mg or 25 mg of empagliflozin or placebo once daily and were followed for 3.1 years. In the combined empagliflozin treated groups there was a statistically significant 14% reduction in the primary outcome (death from cardiovascular causes, nonfatal MI, or nonfatal stroke). As compared with placebo, empagliflozin treatment did not result in a significant difference in the occurrence of non-fatal MI or strokes. However, empagliflozin resulted in a significantly lower risk of death from cardiovascular causes (hazard ratio, 0.62), death from any cause (hazard ratio, 0.68), and hospitalization for heart failure (hazard ratio, 0.65). The beneficial effects of empagliflozin were noted to occur very rapidly and the beneficial effects on heart failure appeared to be the dominant effect compared to effects on ASCVD events. Decreases in cardiovascular outcomes and mortality with empagliflozin occurred across the range of cardiovascular risk (74). Additionally, the reduction in hospitalizations for heart failure and cardiovascular death were observed both in patients with and without heart failure at baseline (75).

 

CANVAS TRIAL

 

The effects of placebo vs. canagliflozin were determined in two combined trials involving a total of 10,142 participants with T2DM and high cardiovascular risk (76). The primary outcome was a composite of death from cardiovascular causes, nonfatal MI, or nonfatal stroke and the mean follow-up was 188 weeks. The primary outcome was reduced in the canagliflozin group (hazard ratio, 0.86; P=0.02). Death from any cause (hazard ratio 0.87; 95% CI 0.74-1.01) and death from ASCVD (hazard ratio 0.87; 95% CI 0.72-1.06) were reduced but were not statistically significant. Similarly, canagliflozin treatment did not result in a significant difference in non-fatal strokes or non-fatal MI (hazard ratio 0.90 for stroke and 0.85 for myocardial infarction). As seen with empagliflozin, hospitalization for heart failure was markedly reduced (hazard ratio 0.67; 95% CI 0.52-0.87) and this beneficial effect occurred rapidly. Notably, there was an increased risk of amputation (hazard ratio, 1.97; 95% CI, 1.41 to 2.75), which were primarily at the level of the toe or metatarsal. The basis for the increase in amputations is unknown. In other SGLT2 inhibitor studies an increase in amputations was not noted.

 

CREDENCE TRIAL

 

In a second canagliflozin trial that focused on kidney disease, a decrease in cardiovascular events was also observed (77). In this double-blind trial 4,401 patients with chronic kidney disease and T2DM were randomized to canagliflozin 100mg per day or placebo and followed for a median of 2.62 years. All the patients had an eGFR of 30 to <90 ml per minute per 1.73 m2 and albuminuria (ratio of albumin [mg] to creatinine [g], >300 to 5000). In this trial hospitalization for heart failure was reduced by 39%. The relative benefits of canagliflozin for cardiovascular outcomes was similar in individuals across the spectrum of eGFR levels (78). In contrast to the CANVAS trial, an increased risk of amputations was not observed.

 

DECLARE–TIMI 58 TRIAL

 

The effect of dapagliflozin on cardiovascular events has also been reported (79). 17,160 patients, including 10,186 without ASCVD were randomized to dapagliflozin or placebo and followed for a median of 4.2 years. The primary outcome was a composite of major adverse cardiovascular events (MACE), defined as cardiovascular death, MI, or ischemic stroke. The primary efficacy outcomes were MACE and a composite of cardiovascular death or hospitalization for heart failure. Dapagliflozin did not result in a lower rate of major adverse cardiovascular events (8.8% in the dapagliflozin group and 9.4% in the placebo group; hazard ratio, 0.93; P=0.17) but did result in a lower rate of cardiovascular death or hospitalization for heart failure (4.9% vs. 5.8%; hazard ratio, 0.83; P=0.005), which reflected a lower rate of hospitalization for heart failure (hazard ratio, 0.73; 95% CI, 0.61 to 0.88). Interestingly, in the patients with a history of a previous MI, dapagliflozin reduced the risk of a MACE (HR 0.84; P=0.039), whereas there was no effect in patients without a previous MI (80). Additionally, there was no increase in lower extremities amputations in the dapagliflozin treated group.

 

VERTIS CV

 

Patients with ASCVD and T2DM were randomized to ertugliflozin 5mg (n=2752), 15mg (2747), or placebo (n=2747) and the primary composite outcome of cardiovascular death and non-fatal MI or stroke was determined after a mean duration of follow-up of 3.5 years (81). This trial did not demonstrate a significant difference in the primary endpoint (MACE) nor any components of the primary endpoint. However, heart failure hospitalizations were significantly reduced by 30% in the patients treated with ertugliflozin (HR 0.70; CI 0.54–0.90). The benefits on heart failure were observed in both patients with a history of heart failure (decreased 37%) and patients without a history of heart failure (decreased 21%) (82).

 

SUMMARY

 

Thus, all four SGLT2 inhibitor studies demonstrated a decrease in heart failure with SGLT2 inhibitor therapy without consistent effects on ASCVD events. For additional information on the beneficial effects of SGLT2 inhibitors and SGLT1/SGLT2 inhibitors on ASCVD and heart failure see the Endotext chapter entitled “Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes” (83).

 

GLP-1 Receptor Agonists

 

The effect of six GLP-1 receptor agonists on ASCVD has been reported.

 

ELIXA

 

In the ELIXA trial 6,068 patients with T2DM who recently had a MI or been hospitalized for unstable angina were randomized to placebo or lixisenatide and followed for a median of 25 months (84). The primary end point of cardiovascular death, MI, stroke, or hospitalization for unstable angina was similar in the placebo or lixisenatide groups.

 

LEADER TRIAL

 

In contrast, the LEADER trial has shown that liraglutide decreased cardiovascular events (85). In this trial 9,340 patients at high cardiovascular risk were randomly assigned to receive liraglutide or placebo. After a median time of 3.5 years, the primary outcome of death from cardiovascular causes, nonfatal MI, or nonfatal stroke occurred in significantly fewer patients in the liraglutide group (13.0%) than in the placebo group (14.9%) (hazard ratio, 0.87, P=0.01). Additionally, deaths from cardiovascular causes (hazard ratio 0.78, P=0.007) or any cause was lower in the liraglutide group than in the placebo group (hazard ratio, 0.85; P=0.02). Interestingly patients with established ASCVD or decreased renal function (eGFR < 60) appeared to derive the greatest benefit of liraglutide treatment (86,87). As expected, weight and blood pressure were decreased in the liraglutide treated group and A1c levels were also decreased by 0.4%.

 

SUSTAIN 6 TRIAL

 

In support of the beneficial effects of GLP1 receptor agonists to reduce cardiovascular events, semaglutide, a long acting GLP-1 receptor agonist, has been shown to also reduce cardiovascular events (88). In this trial, 3,297 patients with T2DM with established ASCVD, chronic heart failure, chronic kidney disease, or age >60 with at least one cardiovascular risk factor were randomized to receive once-weekly semaglutide (0.5 mg or 1.0 mg) or placebo for 104 weeks. The primary outcome of cardiovascular death, nonfatal MI, or nonfatal stroke occurred in 6.6% of the semaglutide group and 8.9% of the placebo group (hazard ratio, 0.74; P = 0.02). In this study, both body weight and A1c levels were decreased in the patients treated with semaglutide.

 

PIONEER 6

 

In the PIONEER 6 study 3,183 patients with T2DM at high cardiovascular risk (age ≥50 years with established cardiovascular or chronic kidney disease, or age ≥60 years with cardiovascular risk factors) were randomly assigned to receive oral semaglutide or placebo (89). After a median time of 15.9 months, major adverse cardiovascular events, the primary outcome, occurred in 3.8% of the subjects treated with oral semaglutide and 4.8% of the placebo group (HR 0.79; 95% CI 0.57 to 1.11). Deaths from cardiovascular causes were 0.9% in the oral semaglutide group and 1.9% in the placebo group (HR 0.49; 95% CI, 0.27 to 0.92) while death from any cause occurred in 1.4% in the oral semaglutide group and 2.8% in the placebo group (HR 0.51; 95% CI, 0.31 to 0.84). It should be noted that the primary outcome was not statistically decreased in this study, which may be due to the relatively small number of subjects studied and the short duration of the study that together resulted in a small number of events. Additionally, more patients in the placebo group received treatment with an SGLT2 inhibitor than in the oral semaglutide group and SGLT2 inhibitors are well recognized to reduce cardiovascular events, which could also have diminished the ability to observe a decrease in events in the oral semaglutide group. Because the direction of change in cardiovascular events in PIONEER 6 and glucose lowering, weight loss, and many other effects of oral semaglutide are very similar to injected semaglutide many experts consider the effects on cardiovascular to also be similar. 

 

EXSCEL TRIAL

 

The effect of once weekly exenatide vs. placebo on cardiovascular outcomes was tested in 14,752 patients, 73% who had ASCVD (90). The primary outcome was the occurrence of death from cardiovascular causes, nonfatal MI, or nonfatal stroke. After a median follow-up of 3.2 years (duration of drug exposure 2.4 years) the primary outcome was reduced in the exenatide treated group but this difference just missed achieving statistical significance (hazard ratio 0.91; 95% CI 0.83-1.00; p=0.06). While not statistically significant these results are consistent with the results observed with liraglutide and semaglutide treatment. It should be recognized that a high percentage of patients discontinued exenatide therapy in this trial (>40%) and this could have adversely affected the ability of exenatide treatment to favorably effect ASCD outcomes.

 

HARMONY OUTCOMES TRIAL

 

The effect of once weekly albiglutide vs. placebo was tested in 9,463 patients with ASCVD (91). The primary outcome was first occurrence of cardiovascular death, MI, or stroke. After a median follow-up of 1.6 years a 22% decrease in the primary endpoint was observed in the albiglutide group (hazard ratio 0·78, p<0·0001). It should be noted that albiglutide is no longer available as it was removed from the market due to commercial considerations by Glaxo.

 

REWIND TRIAL  

 

REWIND was a randomized study of weekly subcutaneous injection of dulaglutide (1.5 mg) or placebo in 9,901 patients with T2DM who had either a previous cardiovascular event or cardiovascular risk factors (approximately 70% of patients did not have prior ASCVD) (92).  During a median follow-up of 5.4 years the primary outcome of non-fatal MI, non-fatal stroke, or death from cardiovascular causes was decreased by 12% in the dulaglutide treated group (HR 0.88, p=0.026). The decrease in events was similar in participants with and without previous ASCVD. In an analysis that focused on stroke it was noted that dulaglutide reduced ischemic stroke by 25% compared to placebo but had no effect on hemorrhagic stroke (93).

 

SUMMARY

 

Thus, four studies have clearly demonstrated that treatment with GLP-1 receptor agonists reduces cardiovascular events, two studies has provided data consistent with these results, and one study failed to demonstrate benefit (Table 1). In a meta-analysis of these seven trials it was observed that cardiovascular death, stroke, or MI was decreased by 12% (HR 0.88, p<0.0001), death from cardiovascular causes by 12% (HR 0.88, p=0.003), fatal or non-fatal stroke by 16% (HR 0.84, p<0.0001) and fatal and non-fatal MI by 9% (HR 0.91, p=0.043) (Table 1) (94). Why there are differences in results between these studies is unknown but could be due to differential effects of the GLP-1 receptor agonists, differences in the patient populations studied, or other unrecognized variables. For additional information on the beneficial effects of GLP-1 receptor agonists on ASCVD see the Endotext chapter entitled “Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes” (83).

 

Table 1. Summary of GLP-1 Receptor Agonist Cardiovascular Outcome Trials

 

Number

Prior CVD

HbA1c

Mean Follow-up (years)

Hazard Ratio* (95% CI)

P value

ELIXA
Lixisenatide

6068

100%

7.7%

2.1

1.02
(0.89-1.17)

0.78

LEADER
Liraglutide

9340

81%

8.7%

3.8

0.87
(0.78-0.97)

0.015

SUSTAIN 6
Semaglutide

3297

83%

8.7%

2.1

0.74
(0.58-0.95)

0.016

EXSCEL
Exenatide

14,752

73%

8.0%

3.2

0.91
(0.83-1.00)

0.061

HARMONY
Albiglutide

9463

100%

8.7%

1.6

0.78
(0.68-0.90)

<0.001

REWIND
Dulaglutide

9901

31%

7.3%

5.4

0>88
(0.79-0.99)

0.026

PIONEER 6**
Semaglutide oral

3183

85%

8.2%

1.3

0.79
(0.57-1.11)

0.17

Overall (94)

       

0.88
(0.82-0.94)

<0.001

*CVD death, MI, Stroke.

 

The mechanism accounting for this decrease in ASCVD is uncertain but could be related to reductions in HbA1c, body weight, systolic blood pressure, postprandial triglyceride levels, or the direct effect of activation of GLP-1 receptors on the atherosclerotic process such as improving endothelial function (95).

 

Tirzepatide

 

Tirzepatide activates both GLP-1 and GIP receptors. Long term cardiovascular trials with tirzepatide are underway. In a meta-analysis of seven randomized controlled trials with a duration of at least 26 weeks with 4,887 participants treated with tirzepatide and 2,328 control participants a 20% decrease in cardiovascular events was observed in the tirzepatide group (HR 0.80; 95% CI 0.57-1.11) suggesting that the effect of tirzepatide will be similar to the GLP-1 receptor agonists (96).   

 

Acarbose

 

In the STOP-NIDDM trial 1,429 subjects with impaired glucose tolerance were randomized to placebo vs. acarbose and followed for 3.3 years (97). In the acarbose group a 49% relative risk reduction in the development of ASCVD events (hazard ratio 0.51; P =0.03) was observed. Among cardiovascular events, the major reduction was in the risk of MI (HR, 0.09; P =.02). In a smaller trial, 135 patients hospitalized for the acute coronary syndrome who were newly diagnosed with IGT were randomly assigned to acarbose or placebo (98). During a mean follow-up of 2.3 years the risk of recurrent major adverse cardiovascular event was decreased significantly in the acarbose group compared with that in control group (26.7% versus 46.9%, P < 0.05).

 

Despite these favorable observations a large trial failed to demonstrate a beneficial effect of acarbose in Chinese patients with impaired glucose tolerance (99). In a randomized trial acarbose vs. placebo was compared in 6,522 patients with coronary heart disease and impaired glucose tolerance. The primary outcome was cardiovascular death, non-fatal MI, non-fatal stroke, hospital admission for unstable angina, and hospital admission for heart failure and patients were followed for a median of 5 years. The primary outcome was similar in the acarbose and placebo groups (hazard ratio 0.98; 95% CI 0.86-1.11, p=0·73). No significant differences were seen for death from any cause, cardiovascular death, fatal or non-fatal MI, fatal or non-fatal stroke, hospital admission for unstable angina, hospital admission for heart failure, or impaired renal function.

 

Thus, whether acarbose favorably affects ASCVD in patients at high risk for developing diabetes is uncertain. Moreover, the effect of acarbose on ASCVD in patients with diabetes is unknown. 

 

Cycloset

 

Cycloset is a quick-release bromocriptine formulation (bromocriptine-QR) that activates the D2 dopamine receptor and is approved for the treatment of diabetes. A 52 week, randomized, double-blind, multicenter trial evaluated cardiovascular safety in 3,095 patients with T2DM treated with bromocriptine-QR or placebo (100). The composite end point of first MI, stroke, coronary revascularization, or hospitalization for angina or congestive heart failure occurred in 1.8% of the bromocriptine-QR treated vs. 3.2% of the placebo-treated patients resulting in a 40% decrease in cardiovascular events (HR 0.60; CI 0.37– 0.96). Clearly further studies to confirm this finding and to elucidate the mechanism of this beneficial effect are required.

 

Bile Acid Sequestrants

 

Colesevelam is a non-absorbed, polymeric, LDL-C lowering and glucose lowering agent that is a high-capacity bile acid-binding molecule. This drug was developed primarily to lower LDL-C levels and was later noted to have favorable effects on blood glucose levels and was approved for improving glycemic control in patients with T2DM (101).

 

There have been no randomized studies that have examined the effect of bile acid sequestrants on cardiovascular end points in subjects with diabetes. In non-diabetic-subjects bile acid sequestrants have reduced cardiovascular events(102,103). Since bile acid sequestrants have a similar beneficial impact on LDL-C levels in diabetic and non-diabetic subjects one would anticipate that these drugs would also result in a reduction in events in the diabetic population.

 

Insulin

 

As described above in patients with T1DM the DCCT trial and in T2DM in the UKPDS trial demonstrated that insulin therapy reduced cardiovascular events by improving glycemic control (29-32). In the Origin Trial 12,537 people with cardiovascular risk factors plus impaired fasting glucose, impaired glucose tolerance, or T2DM were randomized to receive insulin glargine or standard care (104). The cardiovascular outcomes, which included nonfatal MI, nonfatal stroke, death from cardiovascular causes, revascularization, or hospitalization for heart failure, were similar in the glargine and placebo groups. Extended follow-up also did not demonstrate favorable effects on cardiovascular events in the glargine treated patients (105). Additionally, in patients with T2DM at high risk for cardiovascular events the occurrence of major cardiovascular events was similar in patients treated with degludec insulin or glargine insulin (106). These studies demonstrate that insulin does not accelerate atherosclerosis and by lowering glucose levels may decrease atherosclerosis, although the protective effects are mainly observed in patients with T1DM over a protracted period of time.

 

Other Studies

 

Finally, the Bari 2D study compared the effect of insulin sensitizers (metformin/TZD- mostly rosiglitazone) vs. insulin provision therapy (sulfonylureas/insulin) on cardiovascular outcomes in patients with T2DM and coronary artery disease (> 50% stenosis and positive stress test or > 70% stenosis and classic angina) (107,108). In this study, no differences in survival or cardiovascular endpoints were observed between metformin/TZD therapy vs. sulfonylurea/insulin therapy for the entire study. However, in the group with more severe coronary artery disease who were selected for coronary artery bypass surgery, the combination of coronary artery bypass and treatment with insulin sensitizers was associated with a lower rate of cardiovascular events. Why the metformin/TZD group only derived an enhanced benefit in the coronary artery bypass patients in this study is unknown. It should be noted that the vast majority of patients on TZD therapy were treated with rosiglitazone and, as discussed above, the effects of rosiglitazone on ASCVD do not appear to be as beneficial as pioglitazone. 

 

Summary

 

These studies clearly demonstrate that the method by which one improves glycemic control may be very important with different drugs having effects in addition to glucose lowering that reduce cardiovascular events (table 2). While previous treatment algorithms have primarily focused on the effect of drugs on glycemic control, current treatment recommendations for patients with diabetes are using the results of these ASCVD trials to decide which drugs should be employed. For example, the ADA is recommending that in patients with high risk or established ASCVD an SGLT inhibitor or GLP1 receptor agonist with proven cardiovascular benefit should be part of the initial treatment regimen independent of A1c levels (109).

 

Table 2. Effect of Glucose Lowering Drugs on Atherosclerotic ASCVD

Metformin

Studies suggest benefit

Sulfonylureas

No effect

Meglitinides

No effect

Thiazolidinediones

Rosiglitazone no effect; Pioglitazone- studies suggest benefit

DPP4 Inhibitors

No effect on atherosclerosis.

SGLT2 Inhibitors

Marginal effect on ASCVD, Large effect on heart failure

GLP-1 Receptor Agonists

Decrease events

Tirzepatide

Study ongoing

Acarbose

No effect

Cycloset

Further studies required

Bile Acid Sequestrants

Decrease events, further studies required

Insulin

No effect

Thiazolidinediones clearly increase the risk of heart failure while saxagliptin and alogliptin may increase risk of heart failure. SGLT2 inhibitors decrease the risk of heart failure.

 

ROLE OF OTHER RISK FACTORS IN ASCVD

 

Numerous studies have demonstrated that the traditional risk factors for ASCVD play an important role in patients with diabetes (2,4,5,110). Patients with diabetes without other risk factors have a relatively low risk of ASCVD (in most studies higher than similar non-diabetic patients), whereas the increasing prevalence of other risk factors markedly increases the risk of developing ASCVD (2). The major reversible traditional risk factors are hypertension, cigarette smoking, and lipid abnormalities (2,4,5,14,111). Other risk factors include obesity (particularly visceral obesity), insulin resistance, small dense LDL, elevated TG, low HDL-C, procoagulant state (increased PAI-1, fibrinogen), family history of early ASCVD, homocysteine, Lp (a), renal disease, albuminuria, and inflammation (C-reactive protein, SAA, cytokines) (2,4,5,110,111). In the last decade, it has become clear that to reduce the risk of ASCVD in patients with diabetes, one will not only need to improve glycemic control but also address these other cardiovascular risk factors. In the remainder of this chapter, I will focus on the dyslipidemia that occurs in patients with diabetes.

  

ROLE OF LIPIDS IN ASCVD

 

As in non-diabetic populations, epidemiological studies have shown that increased LDL-C and non-HDL-C levels and decreased HDL-C levels are associated with an increased risk of ASCVD in patients with diabetes (2,4,110,111). In the UKPDS cohort LDL-C levels were the strongest predictor of coronary artery disease (112). While it is universally accepted that elevated levels of LDL-C and non-HDL-C cause atherosclerosis and ASCVD the role of HDL-C is uncertain. Genetic studies and studies of drugs that raise HDL-C have not supported low HDL-C levels as a causative factor for atherosclerosis (113). Rather it is currently thought that HDL function is associated with atherosclerosis risk and that this does not precisely correlate with HDL-C levels (113). In patients with diabetes, elevations in serum triglyceride (TG) levels also are associated with an increased risk of ASCVD (4,111,114). With regard to TG, it is not clear whether they are a causative factor for ASCVD or whether the elevation in TG is a marker for other abnormalities (4,111,114,115). Recent Mendelian randomization studies have provided support for the hypothesis that elevated TG levels play a causal role in atherosclerosis (115,116). Unfortunately, as will be discussed later in this chapter lowering TG levels in patients on statin therapy has not decreased cardiovascular events.

 

LIPID ABNORMALITIES IN PATIENTS WITH DIABETES

 

In patients with T1DM in good glycemic control, the lipid profile is very similar to lipid profiles in the general population (110). In some studies HDL-C levels are modestly increased in patients with T1DM (117). In contrast, in patients with T2DM, even when in good glycemic control, there are abnormalities in lipid levels (118-121). It is estimated that 30-60% of patients with T2DM have dyslipidemia (5,122). Specifically, patients with T2DM often have an increase in serum TG levels, increased VLDL and IDL, and decreased HDL-C levels. Non-HDL-C levels are increased due to the increase in VLDL and IDL. LDL-C levels are typically not markedly different than in normal subjects but there is an increase in small dense LDL, a lipoprotein particle that may be particularly pro-atherogenic (123). As a consequence, there are more LDL particles, which coupled with the increases in VLDL and IDL, leads to an increase in apolipoprotein B levels (118-121). Additionally, the postprandial increase in serum TG is accentuated and elevations in postprandial lipids may increase the risk of ASCVD (118-121).

 

It should be recognized that the lipid changes in patients with T2D are characteristic of the alterations in lipid profile seen in obesity and the metabolic syndrome (insulin resistance syndrome) (124). Since a high percentage of patients with T2DM are obese, insulin resistant, and have the metabolic syndrome, it is not surprising that the prevalence of increased TG and small dense LDL and decreased HDL-C is common in patients with T2DM even when these patients are in good glycemic control. Obesity is also accompanied by increased systemic inflammation. The increasing prevalence of obesity/overweight in patients with T1D will likely result in an increased prevalence of dyslipidemia in this population.

 

Studies have shown that the anti-oxidant and anti-inflammatory functions of HDL isolated from patients with T1DM and T2DM are reduced (117,125). Additionally, the ability of HDL to facilitate cholesterol efflux is reduced in patients with T1DM and T2DM (126,127). Together these findings indicate that HDL-C levels per se may not fully reflect risk of ASCVD in patients with diabetes and that HDL function is perturbed in patients with diabetes.

 

In both T1DM and T2DM, poor glycemic control increases serum TG levels, VLDL, and IDL, and decreases HDL-C levels (119). Poor glycemic control can also result in a modest increase in LDL-C, which because of the elevation in TG is often in the small dense LDL subfraction. It is therefore important to optimize glycemic control in patients with diabetes because this will have secondary beneficial effects on lipid levels.

 

Lp(a) levels are usually within the normal range in patients with T1DM and T2DM (128). Some studies have observed no impact of diabetes mellitus on Lp(a) concentrations while other studies reported an elevation or a decrease in Lp(a) concentrations (128). The development of microalbuminuria and the onset of renal disease are associated with an increase in Lp (a) levels (129). Of note low Lp(a) levels are associated with an increased risk of developing T2DM (128). A recent very large case control study found that an Lp(a) concentration in the bottom 10% increases T2DM risk (130).

 

Table 3. Lipid Abnormalities in Patients with Diabetes

T1DM

Lipid profile is similar to controls if glycemic control is good

T2DM

Increased TG, VLDL, IDL, and non-HDL-C. Decreased HDL-C. Normal LDL-C but increase in small dense LDL, LDL particle number, and apolipoprotein B.

Poor glycemic control

Increased TG, VLDL, IDL, and non-HDL-C.  Decreased HDL-C. Modest increase in LDL-C with increase in small dense LDL, LDL particle number, and apolipoprotein B.

 

EFFECT OF GLUCOSE LOWERING DRUGS ON LIPIDS

 

Some therapies used to improve glycemic control may have an impact on lipid levels above and beyond their effects on glucose metabolism. In reviewing the literature, it is often very difficult to separate improvements in glycemic control vs. direct effects of drugs. Additionally, many of the changes induced by drug therapy result in only small changes in LDL-C, HDL-C, and TG levels, are variable from study to study, and are of questionable clinical significance. Insulin, sulfonylureas, meglinitides, DPP4 inhibitors, and alpha-glucosidase inhibitors do not appear to markedly alter fasting lipid profiles other than by improving glucose control (there are data indicating that DPP4 inhibitors and acarbose decrease postprandial triglyceride excursions, but they do not markedly alter fasting lipid levels) (131). In contrast, metformin, thiazolidinediones, GLP1 receptor agonists, bromocriptine-QR, and SGLT2 inhibitors have effects independent of glycemic control on serum lipid levels.

 

Metformin may decrease serum TG levels and LDL-C levels without altering HDL-C levels (131). In a meta-analysis of 37 trials with 2,891 patients, metformin decreased TG by 11.4mg/dL when compared with control treatment (p=0.003) (132). In an analysis of 24 trials with 1,867 patients, metformin decreased LDL-C by 8.4mg/dL compared to control treatment (p<0.001) (132). In contrast, metformin did not significantly alter HDL-C levels (132). It should be noted that in the Diabetes Prevention Program 3,234 individuals with impaired glucose metabolism were randomized to placebo, intensive lifestyle, or metformin therapy. In the metformin therapy group no significant changes were noted in TG, LDL-C, or HDL-C levels compared to the placebo group (133). Thus, metformin may have small effects on lipid levels.    

 

The effect of thiazolidinediones depends on which agent is used. Rosiglitazone increases serum LDL-C levels, increases HDL-C levels, and only decreases serum TG if the baseline TG levels are high (131). In contrast, pioglitazone has less impact on LDL-C levels, but increases HDL-C levels, and decreases TG (131). In the PROactive study, a large randomized cardiovascular outcome study, pioglitazone decreased TG levels by approximately 10%, increased HDL-C levels by approximately 10%, and increased LDL-C by 1-4% (134). It should be noted that reductions in the small dense LDL subfraction and an increase in the large buoyant LDL subfraction are seen with both thiazolidinediones (131). In a randomized head-to-head trial, it was shown that pioglitazone decreased TG levels and increased serum HDL-C levels to a greater degree than rosiglitazone treatment (135,136). Additionally, pioglitazone increased LDL-C levels less than rosiglitazone. In contrast to the differences in lipid parameters, both rosiglitazone and pioglitazone decreased A1c and C-reactive protein to a similar extent. The mechanism by which pioglitazone induces more favorable changes in lipid levels than rosiglitazone despite similar changes in glucose levels is unclear, but differential actions of ligands for nuclear hormone receptors are well described.

 

Treatment with SGLT2 inhibitors results in a small increase in LDL-C and HDL-C levels (131). In a meta-analysis of 48 randomized controlled trials SGLT2 inhibitors significantly increased LDL-C (3.8mg/dL, p < 0.00001), HDL-C (2.3mg/dL, p < 0.00001), and decreased TG levels (8.8mg/dL, p < 0.00001) (137). The mechanism for these increases in LDL and HDL cholesterol is unknown but could be due to a decrease in plasma volume. The decrease in TG levels could be secondary to weight loss.

 

Bromocriptine-QR (Cycloset) treatment decreases TG levels but has no significant effect on LDL-C or HDL-C levels (138,139). The decrease in TG levels is thought to be due to a decrease in hepatic TG synthesis, likely due to a decrease in adipose tissue lipolysis resulting in decreased blood free fatty acid levels and reduced delivery of fatty acids to the liver for TG synthesis (140).

 

Colesevelam, a bile acid sequestrant that is approved for glucose lowering, lowers LDL-C levels by 15-20% and has only a modest effect on HDL-C levels (101,141). The effect of bile acid sequestrants on TG levels varies (141). In patients with normal TG levels, bile acid sequestrants increase TG levels by a small amount. However, as baseline TG levels increase, the effect of bile acid sequestrants on TG levels becomes greater, and can result in substantial increases in TG levels (141). In patients with TG > 500mg/dL the use of bile acid sequestrants is contraindicated (141).

 

Finally, GLP-1 receptor agonists can favorably affect the lipid profile by inducing weight loss (decreasing TG and very modestly decreasing LDL-C levels) (131). In a review by Nauck and colleagues it was noted that GLP-1 receptor agonists lowered TG levels by 18 to 62mg/dL depending upon the specific GLP-1 receptor agonist while decreasing LDL-C by 3-8mg/dL and increasing HDL-C by less than 1mg/dL (142). Additionally, GLP-1 receptor agonists reduce postprandial TG by reducing circulating chylomicrons by decreasing intestinal lipoprotein production (131,142). DPP4 inhibitors have a similar effect on postprandial TG levels as GLP-1 receptor agonists while having minimal effects on fasting lipid levels (142).

 

In the SURPASS trials, tirzepatide studies TG levels were consistently decreased by 13-25% (83,143). In most studies with the exception of SURPASS 5, HDL cholesterol levels increased by 3-11% (83,143). Total cholesterol and LDL cholesterol levels were modestly decreased in most studies (83,143). Not unexpectedly given the decrease in TG levels small LDL particles were decreased. For details see the Endotext chapter Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 Diabetes (83).

 

Table 4. Effect of Glucose Lowering Drugs on Lipid Levels

Metformin

Modestly decrease TG and LDL-C

Sulfonylureas

No effect

DPP4 inhibitors

Decrease postprandial TG

GLP1 analogues

Decrease fasting and postprandial TG, modestly decrease LDL-C

Tirzepatide

Decrease TG, modestly decrease LDL-C, increase HDL-C

Acarbose

Decrease postprandial TG

Pioglitazone

Rosiglitazone

Decrease TG and increase HDL-C. Small increase LDL-C but a decrease in small dense LDL

SGLT2 inhibitors

Small increase in LDL-C and HDL-C

Colesevelam

Decrease LDL-C. May increase TG

Bromocriptine-QR

Decrease TG

Insulin

No effect

 

PATHOPHYSIOLOGY OF THE DYSLIPIDEMIA OF DIABETES

Figure 1. Pathophysiology of the Dyslipidemia of Diabetes

 

Multiple mechanisms account for the dyslipidemia seen in patients with T2DM, which are affected both by the level of glucose control and by factors such as obesity and inflammation that also contribute to dyslipidemia.

 

Increase in TG

 

There are a number of different abnormalities that contribute to the dyslipidemia seen in patients with T2DM and obesity (figure 1) (119-122,144-146).

 

OVERPRODUCTION OF VLDL BY THE LIVER

 

A key abnormality is the overproduction of VLDL by the liver, which is a major contributor to the elevations in serum TG levels. The rate of secretion of VLDL is highly dependent on TG availability, which is determined by the levels of fatty acids available for the synthesis of TG in the liver. An abundance of TG prevents the intra-hepatic degradation of Apo B-100 allowing for increased VLDL formation and secretion. There are three major sources of fatty acids in the liver all of which may be altered in patients with T2DM. First, the flux of fatty acids from adipose tissue to the liver is increased. An increased mass of adipose tissue, particularly visceral stores, results in increased fatty acid delivery to the liver. Additionally, insulin suppresses the lipolysis of TG to free fatty acids in adipose tissue; thus, in patients with either poorly controlled diabetes due to a decrease in insulin or a decrease in insulin activity due to insulin resistance, the inhibition of TG lipolysis is blunted and there is increased TG breakdown leading to increased fatty acid deliver to the liver. A second source of fatty acids in the liver is de novo fatty acid synthesis. Numerous studies have shown that fatty acid synthesis is increased in the liver in patients with T2DM. This increase may be mediated by the hyperinsulinemia seen in patients with insulin resistance. While the liver is resistant to the effects of insulin on carbohydrate metabolism, the liver remains sensitive to the effects of insulin stimulating lipid synthesis. Specifically, insulin stimulates the activity of SREBP-1c, a transcription factor that increases the expression of the enzymes required for the synthesis of fatty acids. Thus, while the liver is resistant to the effects of insulin on carbohydrate metabolism the liver remains sensitive to the effects of insulin stimulating lipid synthesis. Additionally, in the presence of hyperglycemia, glucose can induce another transcription factor, carbohydrate responsive element binding protein (ChREBP), which also stimulates the transcription of the enzymes required for fatty acid synthesis. The third source of fatty acids is the uptake of TG rich lipoproteins by the liver. Studies have shown an increase in intestinal fatty acid synthesis and the enhanced secretion of chylomicrons in animal models of T2DM. This increase in chylomicrons leads to the increased delivery of fatty acids to the liver. The increase in hepatic fatty acids produced by these three pathways results in an increase in the synthesis of TG in the liver and the protection of Apo B-100 from degradation resulting in the increased formation and secretion of VLDL. Finally, insulin stimulates the post translational degradation of Apo B-100 in the liver and a decrease in insulin activity in patients with T2DM also allows for the enhanced survival of Apo B-100 promoting increased VLDL formation.

 

DECREASED DEGRADATION OF TRIGLYCERIDE RICH LIPOPROTEINS

 

While the overproduction of triglyceride rich lipoproteins by the liver and intestine are important contributors to the elevations in serum TG levels in patients with T2DM, there are also abnormalities in the metabolism of these TG rich lipoproteins. First, there is a modest decrease in lipoprotein lipase activity, the key enzyme that metabolizes TG rich lipoproteins. The expression of lipoprotein lipase is stimulated by insulin and decreased insulin activity in patients with T2DM results in a decrease in lipoprotein lipase, which plays a key role in the hydrolysis of the TG carried in chylomicrons and VLDL. Additionally, patients with T2DM have an increase in Apo C-III levels, a key regulator of TG rich lipoprotein clearance. Glucose stimulates and insulin suppresses Apo C-III expression; thus, diabetes with hyperglycemia and either insulin deficiency or insulin resistance contribute to an increase in Apo C-III. Apo C-III is an inhibitor of lipoprotein lipase activity and thereby reduces the clearance of TG rich lipoproteins. In addition, Apo C-III also inhibits the cellular uptake of lipoproteins. Studies have shown that loss of function mutations in Apo C-III lead to lower serum TG levels and a reduced risk of ASCVD (147,148). Interestingly, inhibition of Apo C-III expression results in a decrease in serum TG levels even in patients deficient in lipoprotein lipase, indicating that the ability of Apo C-III to modulate serum TG levels is not dependent solely on regulating lipoprotein lipase activity (149). Lastly, insulin resistance is associated with an increase in Angptl3, an inhibitor of LPL (150). Thus, in patients with diabetes, a decrease in clearance of TG rich lipoproteins also contributes to the elevation in serum triglyceride levels.  

 

Mechanism for the Increase in Small Dense LDL and Decrease in HDL

 

The elevation in TG rich lipoproteins in turn has effects on other lipoproteins. Specifically, cholesterol ester transfer protein (CETP) mediates the exchange of TG from TG rich VLDL and chylomicrons to LDL and HDL. The increase in TG rich lipoproteins per se leads to an increase in CETP mediated exchange, increasing the TG content of both LDL and HDL. The TG on LDL and HDL is then hydrolyzed by hepatic lipase and lipoprotein lipase leading to the production of small dense LDL and small HDL. Notably hepatic lipase activity is increased in patients with T2DM, which will also facilitate the removal of TG from LDL and HDL resulting in small lipoprotein particles. The affinity of Apo A-I for small HDL particles is reduced, leading to the disassociation of Apo A-I, which in turn leads to the accelerated clearance and breakdown of Apo A-I by the kidneys. Additionally, the production of Apo A-I may be reduced in patients with diabetes. High glucose levels can activate ChREBP and this transcription factor inhibits Apo A-I expression. Furthermore, insulin stimulates Apo A-I expression and a reduction in insulin activity due to insulin resistance or decreased insulin levels may also lead to a decrease in Apo A-I expression. The net result is lower levels of Apo A-I and HDL-C levels in patients with T2DM.

 

Role of Poor Glycemic Control

 

The above-described changes lead to the typical dyslipidemia observed in patients with T2DM (increased TG, decreased HDL-C, and an abundance of small dense LDL and small HDL). In patients with both Type 1 and T2DM, poor glycemic control can further adversely affect lipid and lipoprotein metabolism. As noted above the expression of lipoprotein lipase is stimulated by insulin. If insulin activity is very low the expression of lipoprotein lipase is severely suppressed and the metabolism of TG rich lipoproteins is markedly impaired. This leads to the delayed clearance of both chylomicrons and VLDL and elevations of TG rich lipoproteins. Additionally, insulinopenia results in a marked increase in lipolysis in adipose tissue, leading to the release of free fatty acids into the circulation. This increase in serum fatty acids results in the increased delivery of fatty acids to the liver, enhanced TG synthesis in the liver, and the increased production and secretion of VLDL. Whereas patients with T1DM who are well controlled and not obese or overweight typically have normal serum lipid profiles, if their control deteriorates, they will develop hypertriglyceridemia. In patients with T2DM deterioration of glycemic control will further exacerbate their underlying dyslipidemia resulting in greater increases in TG levels. If the synthesis of new VLDL is increased sufficiently this can result in an increase in LDL-C levels. HDL-C levels may decrease due to the formation of small HDL that are more susceptible to accelerated clearance. Improvements in glycemic control can markedly lower TG levels and may increase serum HDL-C levels. In patients with poorly controlled diabetes improvements in glycemic control may also lower LDL-C levels.

 

Role of Obesity and Inflammation

  

Most patients with T2DM and many patients with T1D are obese or overweight. Obesity is a pro-inflammatory state due to the macrophages that infiltrate adipose tissue. The cytokines produced by these macrophages and the adipokines that are produced by fat cells also alter lipid metabolism (151,152). The pro-inflammatory cytokines, TNF and IL-1, decrease the expression of lipoprotein lipase and increase the expression of angiopoietin like protein 4, an inhibitor of lipoprotein lipase. Together these changes decrease lipoprotein lipase activity, thereby delaying the clearance of TG rich lipoproteins. In addition, pro-inflammatory cytokines stimulate lipolysis in adipocytes increasing circulating free fatty acid levels, which will provide substrate for hepatic TG synthesis. In the liver, pro-inflammatory cytokines stimulate de novo fatty acid and TG synthesis. These alterations will lead to the increased production and secretion of VLDL. Thus, increases in the levels of pro-inflammatory cytokines will stimulate the production of TG rich lipoproteins and delay the clearance of TG rich lipoproteins, which together will contribute to the increase in serum TG that occurs in obese patients.

 

Obesity and the increase in pro-inflammatory cytokines may also affect HDL-C levels (153-155). First, pro-inflammatory cytokines inhibit the production of Apo A-I, the main protein constituent of HDL. Second, in many tissues pro-inflammatory cytokines decrease the expression of ABCA1 and ABCG1, which will lead to a decrease in the efflux of phospholipids and cholesterol from the cell to HDL decreasing the formation of mature HDL. Third, pro-inflammatory cytokines inhibit the production and activity of LCAT, which will limit the conversion of cholesterol to cholesterol esters in HDL. This conversion step is required for the formation of a normal spherical HDL particle and is crucial for the ability of HDL to increase the efflux of cholesterol from cells (including macrophages). Together these effects may lead to a decrease in HDL-C levels and a decrease in reverse cholesterol transport. Reverse cholesterol transport plays an important role in preventing cholesterol accumulation in macrophages and thereby reduces atherosclerosis.

 

Inflammation also decreases other important functions of HDL, such as its ability to prevent LDL oxidation (156). This reduction in the ability of HDL to protect from oxidation may be mediated in part by inflammation inducing lower levels of the enzyme paraoxonase, which is commonly seen in patients with diabetes (151,157). In parallel inflammation increases the oxidation of LDL and the amount of small dense LDL that is more susceptible to oxidation.

 

Role of Adipokines

 

Adipokines, such as leptin, adiponectin, and resistin, regulate lipid metabolism and the levels are altered in obese patients. Obesity increases serum leptin levels and leptin stimulates lipolysis in adipocytes which will increase serum free fatty acid levels (158). The circulating levels of adiponectin are decreased in subjects who are obese (159). Decreased adiponectin levels are associated with elevations in serum TG levels and decreases in HDL-C levels (159). This association is thought to be causal as studies in mice have shown that overexpressing adiponectin (transgenic mice) decreases TG and increases HDL-C levels while conversely, adiponectin knock-out mice have increased TG and decreased HDL-C levels (159). The adiponectin induced decrease in TG levels is mediated by an increased catabolism of TG rich lipoproteins due to an increase in lipoprotein lipase activity and a decrease Apo C-III, an inhibitor of lipoprotein lipase (159). The increase in HDL-C levels induced by adiponectin is mediated by an increase in hepatic Apo A-I and ABCA1, which results in the increased production of HDL particles (159).

 

Resistin is increased in subjects who are obese and the levels of resistin directly correlate with plasma TG levels (160). Moreover, resistin has been shown to stimulate hepatic VLDL production and secretion due to an increase in the synthesis of Apo B, TG, and cholesterol (160,161). Finally, resistin is associated with a decrease in HDL-C and Apo A-I levels (160).

 

EFFECT OF LIPID LOWERING ON ASCVD EVENTS IN PATIENTS WITH DIABETES

 

Monotherapy Studies

 

STATINS

 

The Cholesterol Treatment Trialists analyzed data from 18,686 subjects with diabetes (mostly T2DM) from 14 randomized trials (162). 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 39mg/dL (1mmol/L) 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. Thus, these studies indicate that statins are beneficial in reducing ASCVD in patients with diabetes. Because of the large number of patients with diabetes included in the Heart Protection Study (HPS) and CARDS these two studies will be discussed in greater depth.

 

The HPS was a double-blind randomized trial that focused on patients at high risk for the development of cardiovascular events, including patients with a history of MIs, other atherosclerotic lesions, diabetes, and/or hypertension (163,164). Patients were between 40 and 80 years of age and had to have total serum cholesterol levels greater than 135mg/dL (thus very few patients were excluded because they did not have a high enough cholesterol level). The major strength of this trial was the large number of patients studied (>20,000). The diabetes subgroup included 5,963 subjects and thus was as large as many other prevention trials. The study was a 2x2 study design comparing simvastatin 40mg a day vs. placebo and anti-oxidant vitamins (vitamin E 600mg, vitamin C 250mg, and beta-carotene 20mg) vs. placebo and lasted approximately 5 years. Analysis of the group randomized to the anti-oxidant vitamins revealed no beneficial or harmful effects. In contrast, simvastatin therapy (40mg per day) reduced cardiovascular events, including MIs and strokes, by approximately 25% in all participants and to a similar degree in the diabetic subjects (total ASCVD reduced 27%, coronary mortality 20%, MI 37%, stroke 24%). Further analysis of the subjects with diabetes revealed that the reduction in cardiovascular events with statin therapy was similar in individuals with diabetes diagnosed for a short duration (<6 years) and for a long duration (>13 years). Similarly, subjects with diabetes in good control (HbA1c <7%) and those not in ideal control (HbA1c >7%) also benefited to a similar degree with statin therapy. Moreover, both T1DM and T2DM patients had a comparable reduction in ASCVD with simvastatin therapy. The decrease in cardiovascular events in patients with T1DM was not statistically significant because of the small number of subjects. Nevertheless, this is the only trial that included patients with T1DM and suggests that patients with T1DM will benefit from statin therapy similar to T2DM. In general, statin therapy reduced ASCVD in all subgroups of subjects with diabetes (females, males, older age, renal disease, hypertension, high TG, low HDL, ASA therapy, etc.) i.e., statin therapy benefits all patients with diabetes (note this study did not include patients with end stage renal disease but other studies have failed to show benefits of statin therapy in patients with diabetes and end stage renal disease (165)).

 

The CARDS trial specifically focused on subjects with diabetes (166). The subjects in this trial were males and females with T2DM between the ages of 40 to 75 years of age who were at high risk of developing ASCVD based on the presence of hypertension, retinopathy, renal disease, or current smoking. Of particular note, the subjects did not have any evidence of clinical atherosclerosis (myocardial disease, stroke, peripheral vascular disease) at entry and hence this study is a primary prevention trial. Inclusion criteria included LDL-C levels less than 160mg/dL and TG levels less than 600mg/dL. It is important to recognize that the average LDL-C in this trial was approximately 118mg/dL, indicating relatively low LDL-C levels. A total of 2,838 T2DM subjects were randomized to either placebo or atorvastatin 10mg a day. Atorvastatin therapy resulted in a 40% decrease in LDL-C levels with over 80% of patients achieving LDL-C levels less than 100mg/dL. Most importantly, atorvastatin therapy resulted in a 37% reduction in cardiovascular events. In addition, strokes were reduced by 48% and coronary revascularization by 31%. As seen in the HPS, subjects with relatively low LDL-C levels (LDL <120mg/dL) benefited to a similar extent as subjects with higher LDL-C levels (>120mg/dL).

 

HPS and CARDS, in combination with the other statin trials, provide conclusive evidence that statin therapy will reduce cardiovascular events in patients with diabetes. Importantly, the benefits of statin therapy are seen in patients with diabetes in both primary and secondary prevention trials. 

 

Effect of Aggressive LDL-C Lowering with Statins

 

Studies have compared reductions of LDL-C to approximately 100mg/dL to more aggressive reductions in LDL-C on atheroma volume. The Reversal Trial studied 502 symptomatic coronary artery disease patients with an average LDL-C of 150mg/dL (167). Approximately 19% of the patients in this trial had diabetes. Patients were randomized to moderate LDL lowering therapy with pravastatin 40mg per day or to aggressive lipid lowering with atorvastatin 80mg per day. As expected, LDL-C levels were considerably lower in the atorvastatin treated group (pravastatin LDL= 110mg/dL vs. atorvastatin LDL= 79mg/dL). Most importantly, when one analyzed the change in atheroma volume determined after 18 months of therapy using intravascular ultrasound, the group treated aggressively with atorvastatin had a much lower progression rate than the group treated with pravastatin. Compared with baseline values, patients treated with atorvastatin had no change in atheroma burden (there was a very slight regression of lesions), whereas patients treated with pravastatin showed progression of lesions. When one compares the extent of the reduction in LDL-C to the change in atheroma volume, a 50% reduction in LDL (LDL-C levels of approximately 75mg/dL) resulted in the absence of lesion progression. This study suggests that lowering the LDL-C to levels well below 100mg/dL is required to prevent disease progression as measured by intravascular ultrasound. Other studies, such as Asteroid, have shown that marked reductions in LDL-C (in Asteroid the mean LDL-C levels were 61mg/dL) can also result in the regression of coronary artery atherosclerosis determined by intravascular ultrasound measurements (168). Additionally, the Saturn trial demonstrated that aggressive lipid lowering with either atorvastatin 80mg or rosuvastatin 40mg would induce regression of coronary artery atherosclerosis to a similar degree in patients with and without diabetes if the LDL-C levels were reduced to less than 70mg/dL (169). Together these trials indicate that aggressive lowering of LDL-C levels to below 70mg/dL can induce regression of atherosclerotic lesions.

 

The Prove-It trial determined in patients recently hospitalized for an acute coronary syndrome whether aggressively lowering of LDL-C with atorvastatin 80mg per day vs. moderate LDL-C lowering with pravastatin 40mg per day would have a similar effect on cardiovascular end points such as death, MI, documented unstable angina requiring hospitalization, revascularization, or stroke (170,171). In this trial, approximately 18% of the patients were diabetic. As expected, the on-treatment LDL-C levels were significantly lower in patients aggressively treated with atorvastatin compared to the moderate treated pravastatin group (atorvastatin LDL-C = approximately 62 vs. pravastatin LDL-C = approximately 95mg/dL). Of great significance, death or major cardiovascular events was reduced by 16% over the two years of the study in the group aggressively treated with atorvastatin. Moreover, the risk reduction in the patients with diabetes in the aggressive treatment group was similar to that observed in non-diabetics.

 

In the treating to new targets trial (TNT) patients with stable coronary heart disease and LDL-C levels less than 130mg/dL were randomized to either 10mg or 80mg atorvastatin and followed for an average of 4.9 years (172,173). Approximately 15% of the patients had diabetes. As expected, LDL-C levels were lowered to a greater extent in the patients treated with 80mg atorvastatin than with 10mg atorvastatin (77mg/dL vs. 101mg/dL). Impressively, the occurrence of major cardiovascular events was reduced by 22% in the group treated with atorvastatin 80mg (p<0.001). In the patients with diabetes events were reduced by 25% in the high dose statin group.

 

Finally, the IDEAL trial was a randomized study that compared atorvastatin 80mg vs. simvastatin 20-40mg in 8,888 patients with a history of ASCVD (174). Approximately 12% of the patients had diabetes. As expected, LDL-C levels were reduced to a greater extent in the atorvastatin treated group than the simvastatin treated group (approximately 81mg/dL vs. 104mg/dL). Once again, the greater reduction in LDL-C levels was associated with a greater reduction in cardiovascular events. Specifically, major coronary events defined as coronary death, nonfatal MI, or cardiac arrest was reduced by 11% (p=0.07), while nonfatal acute MI were reduced by 17% (p=0.02).

 

Combining the results of the Heart Protection Study, CARDS, Reversal, Saturn, Asteroid, Prove-It, TNT, and IDEAL leads one to the conclusion that aggressive lowering of LDL-C with statin therapy will be beneficial and suggests that in high-risk patients lowering the LDL to levels well below 100mg/dL is desirable. Moreover, the Cholesterol Treatment Trialists reviewed five trials with 39,612 subjects that were designed to determine the effect of usual vs. aggressive reductions in LDL-C (175). They reported that intensive control (approximately a 19mg/dL difference in LDL-C) resulted in a 15% decrease in major vascular events, a 13% reduction in coronary death or non-fatal MI, a 19% decrease in coronary revascularization, and a 16% decrease in strokes. As will be discussed below treatment guidelines reflect the results of these studies. Additionally, as described in detail below, studies of the addition of either ezetimibe or PCSK9 inhibitors to statins further demonstrates that aggressive lowering of LDL-C levels further reduces cardiovascular events

 

FIBRATES

 

The beneficial effect of monotherapy with fibrates (e.g., gemfibrozil, fenofibrate) on ASCVD in patients with diabetes is shown in Table 5. The results of these randomized trials suggest that monotherapy with this class of drug might reduce cardiovascular events in patients with diabetes, but the data is not very robust. The largest trial was the Field Trial (176). In this trial, 9,795 patients with T2DM between the ages of 50 and 75 not taking statin therapy were randomized to fenofibrate or placebo and followed for approximately 5 years. Fenofibrate therapy resulted in a 12% decrease in LDL-C, a 29% decrease in TG, and a 5% increase in HDL-C levels. The primary outcome was coronary events (coronary heart disease death and non-fatal MI), which were reduced by 11% in the fenofibrate group but did not reach statistical significance (p= 0.16). However, there was a 24% decrease in non-fatal MI in the fenofibrate treated group (p=0.01) and a non-significant increase in coronary heart disease mortality. Total ASCVD events (coronary events plus stroke and coronary or carotid revascularization) were reduced 11% (p=0.035). These beneficial effects of fenofibrate therapy on ASCVD were observed in patients without a previous history of ASCVD. In patients with a previous history of ASCVD no benefits were observed. Additionally, the beneficial effect of fenofibrate therapy was seen only in those subjects less than 65 years of age. The beneficial effects of fenofibrate in this study may have been muted by the increased use of statins in the placebo group, which reduced the differences in lipid levels between the placebo and fenofibrate groups. If one adjusted for the addition of lipid-lowering therapy, fenofibrate reduced the risk of coronary heart disease events by 19% (p=0.01) and of total ASCVD events by 15% (p=0.004). Thus, while the results of this large trial are intriguing they do not clearly show a benefit of fibrate therapy reducing ASCVD events. The number of patients with diabetes in the other fibrate trials are relatively small (table 4).

 

While the results of the monotherapy fibrate trials have been very heterogeneous it should be noted that fibrate trials in patients with elevated TG levels have reported a greater reduction of cardiovascular events (177). Additionally, subgroup analysis of several fibrate trials has also suggested that the benefit of fibrates was greatest in patients with elevated TG levels (177,178).

 

The mechanism by which fibrates may reduce cardiovascular events is unclear. These drugs lower serum TG levels and increase HDL-C, but it should be recognized that the beneficial effects of fibrates could be due to other actions of these drugs. Specifically, these drugs activate the nuclear hormone receptor PPAR alpha, which is present in the cells that comprise the atherosclerotic lesions, and it is possible that these compounds directly affect lesion formation and development. In addition, fibrates are anti-inflammatory. In fact, analysis of the VA-HIT study suggested that much of the benefit of fibrate therapy was not due to changes in serum lipoprotein levels (179,180).

 

To summarize, while in general the studies suggest that monotherapy with fibrates may reduce ASCVD in patients with diabetes, the results are not very robust or consistent as seen in the statin trials. Of note fibrate therapy appeared to be most effective in patients with increased TG levels and decreased HDL levels, a lipid profile typically seen in patients with T2DM. However, as will be presented in detail below (combination therapy section) the addition of fibrates to statins does not reduce ASCVD.

 

Table 5. Effect of Fibrate Monotherapy on Cardiovascular Outcomes

Study

Drug

#Diabetic subjects

%Decrease controls

% Decrease diabetics

Helsinki Heart Study (181)

Gemfibrozil

135

34

60*

VA-HIT (180)

Gemfibrozil

620

24

24

DIAS (182)

Fenofibrate

418

-

23*

Sendcap (183)

Bezafibrate

164

-

70

Field (176)

Fenofibrate

9795

-

11*

* Not statistically significant

 

NIACIN

 

A single randomized trial, the Coronary Drug Project, has examined the effect of niacin monotherapy on cardiovascular outcomes (184). This trial was carried out from 1966 to 1974 (before the introduction of statin therapy) in men with a history of a prior MI and demonstrated that niacin therapy reduced cardiovascular events. The results of this study were re-analyzed to determine the effect of niacin therapy in subjects with varying baseline fasting and 1-hour post meal glucose levels (185). It was noted that 6 years of niacin therapy reduced the risk of coronary heart disease death or nonfatal MI by approximately 15-25% regardless of baseline fasting or 1-hour post glucose challenge glucose levels. Particularly notable is that reductions in events were seen in the subjects who had a fasting glucose level >126mg/dL or 1-hour glucose levels >220mg/dL (i.e., patients with diabetes). Thus, based on this single study, niacin monotherapy reduces cardiovascular events both in normal subjects and patients with diabetes. However, as will be presented in detail below (combination therapy section) the addition of niacin to statins does not reduce ASCVD.

 

EZETIMIBE

 

A multicenter, randomized trial in Japan examined the efficacy of ezetimibe in patients aged ≥75 years with elevated LDL-C (≥140 mg/dL) without a history of coronary artery disease who were not taking lipid lowering drugs (186). Patients were randomized to ezetimibe (n=1716) or usual care (n=1695) and followed for 4.1 years. The primary outcome was a composite of sudden cardiac death, MI, 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. Approximately 25% of the patients in this trial had diabetes and the beneficial effects were similar in the diabetic and non-diabetic subjects. 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 suggestive evidence that ezetimibe monotherapy may reduce cardiovascular events in patients with diabetes.

 

BEMPEDOIC ACID

 

A multicenter study of bempedoic acid in statin intolerant patients with ASCVD or at high risk for ASCVD was recently reported (187). Patients were randomized to bempedoic acid, 180 mg daily (n=6992), or placebo (n=6978) and the primary end point was death from cardiovascular causes, nonfatal MI, nonfatal stroke, or coronary revascularization. Bempedoic acid therapy reduced LDL-C and hsCRP levels by approximately 22% compared to the placebo group. The primary composite endpoint was reduced by 13% in the bempedoic acid group (HR 0.87; 95% CI, 0.79 to 0.96; P = 0.004). The four individual components of the primary endpoint were also significantly reduced in the bempedoic acid treatment group. In this trial approximately 45% of the patients had diabetes. In an analysis of the patients without clinical ASCVD, (i.e., primary prevention) (bempedoic acid n = 2100 or placebo n = 2106), there was a 30% decrease in cardiovascular events (HR 0.70: 95% CI, 0.55-0.89; P = .002) (188). In this subgroup analysis 66% of the patients had diabetes. This study clearly indicates that monotherapy with bempedoic acid will reduce cardiovascular events.

 

OTHER DRUGS

 

With regard to PCSK9 inhibitors and bile acid sequestrants there have been no randomized monotherapy studies that have examined the effect of these drugs on cardiovascular end points in subjects with diabetes. In non-diabetic subjects, monotherapy with bile acid sequestrants have reduced cardiovascular events (102,103). Since bile acid sequestrants have a similar beneficial impact on serum lipid levels in diabetic and non-diabetic subjects one would anticipate that these drugs would also result in a reduction in events in the diabetic population. Additionally, bile acid sequestrants improve glycemic control (101). However, bile acid sequestrants can raise TG levels and therefore must be used with caution in hypertriglyceridemic patients. There are no outcome studies with PCSK9 inhibitor monotherapy in patients with diabetes but given that these drugs reduce LDL-C levels and in combination with statins reduce cardiovascular events one would anticipate that PCSK9 inhibitor monotherapy will also reduce cardiovascular events.

 

Combination Therapy

 

The studies with statins have been so impressive that most patients with diabetes over the age of 40 are routinely treated with statin therapy and younger patients with diabetes at high risk for ASCVD are also typically on statin therapy (see Current Guidelines Section). Therefore, a key issue is whether the addition of other lipid lowering drugs to statins will result in a further reduction in cardiovascular events. A difficulty with such studies is that the reduction in cardiovascular events induced by statin therapy is so robust that very large trials may be required to see additional benefit.

 

STATINS + FIBRATES

 

The ACCORD-LIPID trial was designed to determine if the addition of fenofibrate to aggressive statin therapy would result in a further reduction in ASCVD in patients with T2DM (189). In this trial, 5,518 patients on statin therapy were randomized to placebo or fenofibrate therapy. The patients had diabetes for approximately 10 years and either had pre-existing ASCVD or were at high risk for developing ASCVD. During the trial, LDL-C levels were approximately 80mg/dL in both groups. There was only a small difference in HDL-C with the fenofibrate groups having a mean HDL-C of 41.2mg/dL while the control group had an HDL-C of 40.5mg/dL. Differences in TG levels were somewhat more impressive with the fenofibrate group having a mean TG level of 122mg/dL while the control group had a TG level of 144mg/dL. First occurrence of nonfatal MI, nonfatal stroke, or death from cardiovascular causes was the primary outcome and there was no statistical difference between the fenofibrate treated group and the placebo group. Additionally, there were also no statistically significant differences between the groups with regards to any of the secondary outcome measures of ASCVD. Of note, the addition of fenofibrate to statin therapy did not result in an increase in either muscle or liver side effects. On further analysis, there was a possible benefit of fenofibrate therapy in the patients in whom the baseline TG levels were elevated (>204mg/dL) and HDL-C levels decreased (<34mg/dL). Finally, similar to what has been reported in other trials, fenofibrate had beneficial effects on the progression of microvascular disease (190,191).

 

The PROMINENT trial studied the effect of pemafibrate, a new selective PPAR-alpha activator, in reducing cardiovascular outcomes in 10,497 patients (66.9% with previous ASCVD) with diabetes (192). This was a double-blind, randomized, controlled trial, in patients with T2DM, with mild-to-moderate hypertriglyceridemia (TG level, 200 to 499 mg/dL), LDL-C < 100mg/dL, and HDL-C levels < 40 mg/dL) who received either pemafibrate (0.2-mg tablets twice daily) or placebo in addition to statin therapy (96% on statins). The primary end point was a composite of nonfatal MI, ischemic stroke, coronary revascularization, or death from cardiovascular causes. Baseline fasting TG was 271 mg/dL, HDL-C 33 mg/dL, and LDL-C 78 mg/dL. Compared with placebo, pemafibrate decreased TG by 26.2%, while HDL-C increased 5.1% and LDL-C increased 12.3%. Notably non-HDL-C levels were unchanged and Apo B levels increased 4.8%. The primary endpoint was similar in the pemafibrate and placebo group (HR 1.03; 95% CI 0.91 to 1.15). The increase in LDL-C and Apo B levels likely account for the failure to reduce cardiovascular events.

 

Taken together the ACCORD study and the PROINENT trial indicate that the addition of fibrate therapy to statin therapy will not result in a reduction in cardiovascular events in patients with diabetes.

 

STATIN + NIACIN

 

The AIM-HIGH trial was designed to determine if the addition of Niaspan to aggressive statin therapy would result in a further reduction in cardiovascular events in patients with pre-existing ASCVD (193). In this trial 3,314 patients were randomized to Niaspan vs. placebo. Approximately 33% of the patients had diabetes. On trial, LDL-C levels were in the 60-70mg/dL range in both groups. As expected, HDL-C levels were increased in the Niaspan treated group (approximately 44mg/dL vs. 38mg/dL), while TG were decreased (approximately 121mg/dL vs. 155mg/dL). However, there were no differences in the primary endpoint between the control and Niaspan treated groups (Primary endpoint consisted of the first event of death from coronary heart disease, nonfatal MI, ischemic stroke, hospitalization for an acute coronary syndrome, or symptom-driven coronary or cerebral revascularization). There were also no differences in secondary endpoints except for a possible increase in strokes in the Niaspan treated group. The addition of Niaspan to statin therapy did not result in a significant increase in either muscle or liver toxicity. Thus, this study does not provide support for the addition of niacin to statins. However, it should be recognized that this was a relatively small study and a considerable number of patients stopped taking the Niaspan during the course of the study (25.4% of patients discontinued Niaspan therapy). In addition, most of the patients included in this study did not have a lipid profile that one would typically consider treating with niacin therapy. In the subset of patients with TG > 198mg/dL and HDL-C < 33mg/dL niacin showed a trend towards benefit (hazard ratio 0.74; p=0.073), suggesting that if the appropriate patient population was studied the results may have been positive (194).

 

HPS 2 Thrive also studied the effect of niacin added to statin therapy (195). This trial utilized extended-release niacin combined with laropiprant, a prostaglandin D2 receptor antagonist that reduces the flushing side effect of niacin treatment. HPS 2 Thrive was a very large trial with over 25,000 patients randomized to either niacin therapy or placebo. Approximately 32% of the patients in this trial had diabetes. The LDL-C level was 63mg/dL, the HDL-C 44mg/dL, and the TG 125mg/dL at baseline. As expected, niacin therapy resulted in a modest reduction in LDL-C (10mg/dL), a modest increase in HDL-C (6mg/dL), and a larger reduction in TG (33mg/dL). However, despite these lipid changes there were no significant differences in major cardiovascular events between the niacin and control group (risk ratio 0.96 CI 0.90- 1.03). It is unknown whether laropiprant, the prostaglandin D2 receptor antagonist, might have effects that worsen atherosclerosis and increase event rates. Similar to the AIM-HIGH study, the group of patients included in the HPS 2 Thrive trial were not the ideal patient population to test for the beneficial effects of niacin treatment added to statin therapy. Ideally, patients with high TG and high non-HDL-C levels coupled with low HDL-C levels should be studied. Nevertheless, the results of the AIM-HIGH and HPS 2 Thrive trials do not provide support for the addition of niacin to statin therapy in patients with diabetes.

 

STATIN + EZETIMIBE

 

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 (196). This was a large trial with over 18,000 patients randomized to statin therapy vs. statin therapy + ezetimibe. Approximately 27% of the patients in this trial had diabetes. On treatment LDL-C levels were 70mg/dL in the statin alone group vs. 53mg/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 re-hospitalization, 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). These beneficial effects were particularly pronounced in the patients with diabetes (Primary endpoint hazard ratio, 0.85; 95% confidence interval, 0.78-0.94) (197,198). This trial provides evidence that the combination of a statin + ezetimibe that results in a greater reduction in LDL-C levels will lead to a larger decrease in cardiovascular events than statin alone. It should be noted that the observed reduction in events was in the range expected based on the decrease in LDL-C levels.

 

The RACING trial compared rosuvastatin 10 mg plus ezetimibe 10 mg (combination therapy) vs. rosuvastatin 20mg in 3,780 patients (1,398 patients (37.0%) with diabetes) at 26 centers in South Korea (199). In the patients with diabetes the baseline LDL-C levels was 74mg/dL and during the study the median LDL-C was 53mg/dL in the combination therapy group and 61mg/dL in the high-intensity statin group (P < 0.001). After a median follow-up of 3 years the rate of cardiovascular events in patients with diabetes was 10.0% in the combination therapy group and 11.3% in the high-intensity statin group (HR: 0.89; 95% CI: 0.64–1.22; P = 0.460). Interestingly the rate of discontinuation or dose reduction of the study drug due to intolerance was lower in the combination therapy group than in the high-intensity statin group (5.2 vs. 8.7%; P = 0.014). This study demonstrates that cardiovascular outcomes were comparable between those receiving combination therapy vs. high-intensity statin monotherapy and that combination therapy significantly reduced the rate of drug discontinuation or dose reduction due to intolerance.

 

STATIN + PCSK9 INHIBITORS

 

The FOURIER trial was a randomized, double-blind, placebo-controlled trial of evolocumab vs. placebo in 27,564 patients with atherosclerotic ASCVD and an LDL-C level of 70 mg/dL or higher who were on statin therapy (200). Approximately 40% of the patients had diabetes (201). The primary end point was cardiovascular death, MI, stroke, hospitalization for unstable angina, or coronary revascularization and the key secondary end point was cardiovascular death, MI, 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-C levels (LDL-C level on treatment approximately 30mg/dL). Evolocumab treatment significantly reduced the risk of the primary end point (HR 0.85; 95% CI 0.79 to 0.92; P<0.001) and the key secondary end point (HR 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 (202). 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 (203). Finally, 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 ASCVD (204).

 

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 Apo B level of at least 80 mg/dL while on high intensity statin therapy or the maximum tolerated statin dose (205). Approximately 29% of the patients had diabetes. 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 MI, 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 > 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 similar to the FOURIER trial 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).

 

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 takes time with only modest effects observed during the first year of treatment. In the FOURIER trial the reduction of cardiovascular death, MI, 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.

 

Additional support for the benefits of further lowering of LDL-C levels with a PCSK9 inhibitor added to statin therapy is seen in the GLAGOV trial (206). This trial was a double-blind, placebo-controlled, randomized trial of evolocumab vs. placebo in 968 patients presenting for coronary angiography. Approximately 20-21% of the patients had diabetes. 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). The results in the patients with diabetes were similar to the non-diabetic patients.

 

Taken together these trials demonstrate that further lowering LDL-C levels with PCSK9 monoclonal antibodies in patients taking statins will have beneficial effects on ASCVD outcomes. A study of the effect of inclisiran on ASCVD endpoints is currently in progress.

 

The results of the ezetimibe and PCSK9 trials have several important implications. First, it demonstrates that combination therapy may have benefits above and beyond statin therapy alone. Second, it provides further support for the hypothesis that lowering LDL per se will reduce cardiovascular events. Third, it suggests that lowering LDL levels to much lower levels than usual will have significant benefits. These results have implications for determining goals of therapy.

 

STATINS + LOW DOSE OMEGA-3-FATTY ACIDS

 

ORIGIN was a double-blind study in 12,536 patients at high risk for ASCVD who had impaired fasting glucose, impaired glucose tolerance, or diabetes (207).  Patients were randomized to receive a 1-gram capsule containing at least 900mg of ethyl esters of omega-3 fatty acids (EPA 465mg and DHA 375mg) or placebo for approximately 6 years. Greater than 50% of the patients were on statin therapy. The primary outcome was death from cardiovascular causes. TG levels were reduced by 14.5mg/dL in the group receiving omega-3-fatty acids compared to the placebo group (P<0.001), without a significant effect on other lipids. The incidence of the primary outcome was not significantly decreased among patients receiving omega-3-fatty acids as compared with those receiving placebo. The use of omega-3-fatty acids also had no significant effect on the rates of major vascular events, death from any cause, or death from arrhythmia.

 

A Study of Cardiovascular Events in Diabetes (ASCEND) was a randomized, placebo controlled, double blind trial of 1-gram omega-3-fattys acids (400mg EPA and 300mg DHA ethyl esters) vs. olive oil placebo in 15,480 patients with diabetes without a history of ASCVD (primary prevention trial) (208). Approximately 75% of patients were on statin therapy. The primary end point was serious vascular events (non-fatal MI, non-fatal stroke, transient ischemic attack, or vascular death). Total cholesterol, HDL-C, and non-HDL-C levels were not significantly altered by omega-3-fatty acid treatment (changes in TG levels were not reported). After a mean follow-up of 7.4 years the composite outcome of a serious vascular event or revascularization occurred in 882 patients (11.4%) on omega-3-fatty acids and 887 patients (11.5%) on placebo (rate ratio, 1.00; 95% CI, 0.91 to 1.09). Serious adverse events were similar in placebo and omega-3-fatty acid treated groups.

 

Taken together these studies indicate that low dose omega-3-fatty acids do not reduce cardiovascular events in patients with diabetes. Studies in non-diabetics have also found little effect of low dose omega-3-fatty acids on ASCVD (209).

 

STATINS + HIGH DOSE OMEGA-3-FATTY ACIDS

 

The Japan EPA Lipid Intervention Study (JELIS) was an open label non-placebo controlled study in patients on statin therapy with total cholesterol levels > 254mg/dL with (n= 3664) or without ASCVD (n=14,981) who were randomly assigned to be treated with 1800 mg of EPA (Vascepa) + statin (n=9326) or statin alone (n= 9319) with a 5 year follow-up (210). Approximately 16% of the patients had diabetes. The mean baseline TG level was 153mg/dL. The primary endpoint was any major coronary event, including sudden cardiac death, fatal and non-fatal MI, and other non-fatal events including unstable angina pectoris, angioplasty, stenting, or coronary artery bypass grafting. On treatment total cholesterol, LDL-C, and HDL-C levels were similar in the two groups but plasma TG were modestly decreased in the EPA treated group (5% decrease in EPA group compared to controls; p = 0.0001). In the EPA + 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). Unstable angina and non-fatal coronary events were also significantly reduced in the EPA 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 MI, stroke, or cardiovascular death. A subjective endpoint has the potential to be an unreliable endpoint in an open label study and is a major limitation of the JELIS Study. The reduction in events was similar in the subgroup of patients with diabetes. In patients with TG levels >150mg/dL and HDL-C levels < 40mg/dL there was a 53% decrease in events (211). In the EPA group, small increases in the occurrence of bleeding (1.1% vs. 0.6%, p=0.0006), gastrointestinal disturbance (3.8%% vs. 1.7%, p<0.0001) and skin abnormalities (1.7 vs. 0.7%, p<0.0001) were seen. 

 

The Reduction of Cardiovascular Events with EPA – Intervention Trial (REDUCE-IT) was a randomized, double blind trial of 2 grams twice per day of EPA ethyl ester (icosapent ethyl) (Vascepa) vs. placebo (mineral oil) in 8,179 patients with hypertriglyceridemia (135mg/dL to 499mg/dL) and established ASCVD or high ASCVD risk (diabetes plus one risk factor) who were on stable statin therapy (212). Approximately 60% of the patients in this trial had diabetes. The primary end point was a composite of cardiovascular death, nonfatal MI, 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; 95% CI 0.68 to 0.83; P<0.001), indicating a 25% decrease in events. The beneficial effects were similar in patients with and without diabetes. The number needed to treat to avoid one primary end-point event was 21. The reduction in cardiovascular events was noted after approximately 2 years of EPA treatment. Additionally, the risk of cardiovascular death was decreased by 20% in the EPA group (HR 0.80; 95% CI, 0.66 to 0.98; P=0.03). The cardiovascular benefits of EPA were similar across baseline levels of TG (<150, ≥150 to <200, and ≥200 mg/dL). 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 cardiovascular risk reduction was not associated with attainment of a normal TG levels. An increase in hospitalization for atrial fibrillation or flutter (3.1% vs. 2.1%, P=0.004) occurred in the EPA group. In addition, serious bleeding events occurred in 2.7% of the patients in the EPA group and in 2.1% in the placebo group (P=0.06). 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.

 

These results demonstrate that EPA treatment reduces ASCVD events. Of note the reduction in TG levels is relatively modest and would not be expected to result in the magnitude of the decrease in ASCVD observed in the JELIS and REDUCE-IT trials. 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 cardiovascular events (213). It is likely that the beneficial effects of EPA seen in the JELIS and REDUCE-IT trials are multifactorial.

 

The Statin Residual Risk Reduction with Epanova in High Risk Patients with Hypertriglyceridemia (STRENGTH) trial was a randomized, placebo controlled, double blind trial of 4 grams per day of omega-3-fatty acids (Epanova) (carboxylic acid formulation of EPA and DHA) vs. placebo (corn oil) in 13,000 patients on statins with hypertriglyceridemia (180-500mg/dL), optimal LDL-C levels (< 100mg/dL or on maximal statin therapy), low HDL-C (<42mg/dL in men and < 47mg/dL in women), and either ASCVD or high risk for ASCVD (214). The primary outcome was major atherosclerotic cardiovascular events (cardiovascular death, MI, stroke, coronary revascularization or hospitalization for unstable angina). The primary end point occurred in 785 patients (12.0%) treated with omega-3 CA vs 795 (12.2%) treated with corn oil (HR, 0.99: [95% CI, 0.90-1.09]; P = .84) (215). Thus, in contrast to EPA alone this omega-3-fatty acid formulation failed to show benefits despite reducing TG levels (18% decrease) to a similar degree as in the REDUCE-IT trial.

 

Whether EPA has special properties that resulted in the reduction in cardiovascular events in the REDUCE-IT trial or there were flaws in the trial design (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% (LDL-C by approximately 9mg/dL and non-HDL-C by approximately 10mg/dL) in the mineral oil placebo group (212). Additionally, Apo B levels were increased by 7% (6mg/dL) by mineral oil (212). 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 (212,216). 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 (215). 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 (217,218). Ideally, another large randomized cardiovascular trial with EPA ethyl ester (icosapent ethyl) (Vascepa) using a placebo other than mineral oil would resolve this controversy.

 

CURRENT GUIDELINES FOR SERUM LIPIDS

 

There are several different guidelines for treating lipids in patients with diabetes. While they all focus on lowering LDL-C there are differences between the various guidelines.

 

American Diabetes Association Guidelines

 

The 2023 American Diabetes Association (ADA) recommends that adult patients with diabetes have their lipid profile determined at the time of diabetes diagnosis and at least every 5 years thereafter or more frequently if indicated (219). This profile includes total cholesterol, HDL-C, TG, and calculated LDL-C. A lipid panel should be obtained immediately prior to initiating statin therapy. Once a patient is on statin therapy testing should be carried out 4-12 weeks after initiating therapy and annually thereafter to monitor adherence and efficacy. Lifestyle modifications including a reduction in saturated fat, trans fat, and cholesterol intake, weight loss if indicated, an increase in omega-3-fatty acids, viscous fiber, and plant stanols /sterol intake, and increased physical activity is indicated in all patients with diabetes. A focus on a Mediterranean style diet or Dietary Approaches to Stop Hypertension (DASH) diet should be encouraged. In patients with elevated TG levels glycemic control is beneficial and dietary changes and lifestyle changes including weight loss and abstinence from alcohol should be undertaken. Secondary disorders and medications that raise TG levels should be evaluated. Optimize glycemic control to improve TG and HDL-C levels. The recommendations for lipid lowering therapy are shown in table 6. If one follows these recommendations almost all patients with diabetes over the age of 40 will be on statin therapy and many under the age of 40 will also be treated with statins. The addition of ezetimibe should be considered to further lower LDL-C levels in high-risk primary prevention patients. In very high-risk patients with ASCVD if the LDL-C level on statin therapy is greater than 70mg/dL the use of ezetimibe or a PCSK9 inhibitor should be considered. The use of fibrates or niacin with statins were generally not recommended as there is no evidence of benefit. However, in patients with ASCVD or other cardiovascular risk factors on a statin with controlled LDL-C but elevated TG levels (135-499mg/dL) the addition of icosapent ethyl can be considered. Finally, in patients with fasting TG levels greater than 500mg/dL an evaluation for secondary causes of hypertriglyceridemia should be initiated and consideration of drug therapy to reduce the risk of pancreatitis.

 

Table 6. ADA Recommendations for Lipid Lowering Therapy

Primary Prevention

Age 20-39: With additional risk factors may be reasonable to initiate statin therapy

Age 40-75: Use moderate-intensity statin therapy* in addition to lifestyle therapy

Age 40-75: If at higher cardiovascular risk, including those with one or more ASCVD risk factors, it is recommended to use high intensity statin therapy to reduce LDL cholesterol by >50% and to target an LDL-C <70 mg/dL

Age 40-75: If at higher cardiovascular risk, especially those with multiple ASCVD risk factors and an LDL-C >70 mg/dL, it may be reasonable to add ezetimibe or a PCSK9 inhibitor to maximum tolerated statin therapy

Age > 75: Initiating moderate intensity statin therapy is reasonable after discussion and in patient already on statin therapy it is reasonable to continue statin therapy

Secondary Prevention

All ages: High intensity statin therapy**/maximally tolerated stain

For people with diabetes and ASCVD, treatment with high intensity statin therapy is recommended to target an LDL-C reduction of >50% and an LDL-C l goal of <55 mg/dL. Addition of ezetimibe or a PCSK9 inhibitor is recommended if this goal is not achieved

*Moderate intensity statin- atorvastatin 10-20mg, rosuvastatin 5-10mg, simvastatin 20-40mg, pravastatin 40-80mg, lovastatin 40mg, Fluvastatin XL 80mg, pitavastatin 3-4mg.

**High Intensity statin- atorvastatin 40-80mg, rosuvastatin 20-40mg.

 

American College of Cardiology and American Heart Association Guidelines

 

The 2018 American College of Cardiology and American Heart Association (ACC/AHA) guidelines recommend the following (220). “In patients 40 to 75 years of age with diabetes mellitus and LDL-C ≥70 mg/dL (≥1.8 mmol/L), start moderate-intensity statin therapy without calculating 10-year ASCVD risk. In patients with diabetes mellitus at higher risk, especially those with multiple risk factors or those 50 to 75 years of age, it is reasonable to use a high-intensity statin to reduce the LDL-C level by ≥50%.” In patients with diabetes and ASCVD they recommend “In patients with clinical ASCVD, reduce LDL-C with high-intensity statin therapy or maximally tolerated statin therapy. The more LDL-C is reduced on statin therapy, the greater will be subsequent risk reduction. Use a maximally tolerated statin to lower LDLC levels by ≥50%. In very high-risk ASCVD, use an LDL-C threshold of 70 mg/dL (1.8 mmol/L) to consider addition of non-statins to statin therapy. Very high-risk includes a history of multiple major ASCVD events or 1 major ASCVD event and multiple high-risk conditions. In very high-risk ASCVD patients, it is reasonable to add ezetimibe to maximally tolerated statin therapy when the LDL-C level remains ≥70 mg/dL (≥1.8 mmol/L). In patients at very high risk whose LDL-C level remains ≥70 mg/dL (≥1.8 mmol/L) on maximally tolerated statin and ezetimibe therapy, adding a PCSK9 inhibitor is reasonable, although the long-term safety (>3 years) is uncertain and cost effectiveness is low at mid-2018 list prices.” With regards to testing they recommend “Assess adherence and percentage response to LDL-C–lowering medications and lifestyle changes with repeat lipid measurement 4 to 12 weeks after statin initiation or dose adjustment, repeated every 3 to 12 months as needed”. Finally, there are several diabetes specific risk enhancers that are independent of other risk factors that should be considered in deciding the risk of cardiovascular events in a patient with diabetes (Table 7).

 

Table 7. Diabetes Specific Risk Enhancers That are Independent of Other Risk Factors in Diabetes

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

Albuminuria ≥30 mcg of albumin/mg creatinine

eGFR <60 mL/min/1.73 m2

Retinopathy

Neuropathy

ABI <0.9

ABI indicates ankle-brachial index

 

American Association of Clinical Endocrinologists/American College of Endocrinology Guidelines

 

The American Association of Clinical Endocrinologists and American College of Endocrinology guidelines consider individuals with T2DM to be at high, very high, or extreme risk for ASCVD (221,222). Patients with T1DM and a duration of diabetes of more than 15 years or two or more risk factors, poorly controlled A1c, or insulin resistance with metabolic syndrome should be considered to have an equivalent risk to patients with T2DM (221). The recommended treatment goals are shown in Table 8.

 

Table 8. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dL

Non-HDL-C mg/dL

Apo B mg/dL

TG

mg/dL

Extreme Risk

Diabetes and clinical ASCVD

<55

<80

<70

<150

Very High Risk

Diabetes with one or more risk factors

<70

<100

<80

<150

High Risk

Diabetes and no other risk factors

<100

<130

<90

<150

 

European Society of Cardiology and European Atherosclerosis Society Guidelines

 

The European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS) has 2019 guidelines for the treatment of lipids in patients with diabetes (223). These guidelines classify patients with diabetes as very high risk, high risk, or moderate risk (table 9). The recommended goals of therapy based on risk classification are shown in table 10. As with other guidelines intensification of statin therapy should be considered before the introduction of combination therapy. If the goal is not reached, statin combination with ezetimibe should be considered next.

 

Table 9. ESC/EAS Classification of Risk in Patients with Diabetes

Very High Risk- target organ damage, or at least three major risk factors, or early onset of T1DM of long duration (>20 years)

High Risk- without target organ damage, with DM duration >10 years or another additional risk factor

Moderate Risk- Young patients (T1DM <35 years; T2DM <50 years) with DM duration <10 years, without other risk factors. Calculated SCORE >1 % and <5% for 10-year risk of fatal CVD

 

Table 10. ESC/EAS Goals of Therapy in Patients with Diabetes

 

LDL-C

Non-HDL-C

Apo B

Very High Risk

>50% reduction and <55mg/dL (<1.4mmol/L)

<85mg/d;

<65mg/dL

High Risk

>50% reduction and <70mg/dL (<1.8mmol/L)

<100mg/dL

<80mg/dL

Moderate Risk

<100mg/dL

<130mg/dL

<100mg/dL

 

European Society of Cardiology

 

The ESC has updated their guidelines in 2023 (224). An important new recommendation is that in patients with T2DM without symptomatic ASCVD or severe target organ damage, it is recommended to estimate 10-year CVD risk using SCORE2-Diabetes (225). This has resulted in a new classification of risk in patients with T2DM (table 9). The LDL-C goals for each category are shown in table 11.

 

Table 11. Cardiovascular Risk Categories and Goals in Patients with Type 2 Diabetes

Very high risk

Clinically established ASCVD or

Severe target organ damage or

10-year risk of CVD> 20% using SCORE2-Diabetes

LDL-C < 55mg/dl

Non-HDL-C <85mg/dL

High risk*

10-year risk of CVD 10% to < 20% using SCORE2-Diabetes

LDL-C < 70mg/dL

Non-HDL-C <100mg/dL

Moderate risk*

10-year risk of CVD 5% to <10% using SCORE2-Diabetes

LDL-C < 100mg/dL

Low Risk*

10-year risk of CVD <5% using SCORE2-Diabetes

no recommendations

*Patients not meeting the very high-risk category.

Severe target organ damage is defined as eGFR <45 mL/min/1.73 m2 irrespective of albuminuria; or eGFR 45–59 mL/min/1.73 m2 and microalbuminuria (UACR 30–300 mg/g; stage A2); or proteinuria (UACR>300 mg/g; stage A3), or presence of microvascular disease in at least three different sites [e.g., microalbuminuria (stage A2) plus retinopathy plus neuropathy].

 

My Guideline Recommendations

 

Thus, different organizations have proposed somewhat different recommendations for the treatment of lipids in patients with diabetes. Despite these differences it is clear that the vast majority of patients with diabetes will need to be treated with statins regardless of which guidelines one elects to follow.

 

The approach I use is to combine these recommendations (Tables 12 and 13). In patients with diabetes who have pre-existing ASCVD I initiate intensive statin therapy. I prefer LDL or non-HDL-C goals over percent reduction goals. Given the extensive data showing that the lower the LDL-C the greater the reduction in cardiovascular events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering without markedly increasing costs (226). In patients with diabetes 40-75 years of age without pre-existing ASCVD I calculate the 10-year risk of developing ASCVD (http://www.cvriskcalculator.com/) and identify risk enhancing factors (Table 7) and other factors that increase risk that are not included in the calculator (for example family history, inflammatory disorders, etc.). I initiate intensive statin therapy if the 10-year risk is > 7.5% or if there are multiple risk factors/risk enhancers. I initiate moderate statin therapy if the risk is < 7.5% without multiple risk factors/enhancers. Four to twelve weeks after initiating statin therapy I obtain a lipid panel to determine if the LDL and non-HDL-C levels are at goal. In patients with pre-existing ASCVD or multiple risk factors/risk enhancers (i.e., very high-risk patients) my goal is an LDL-C < 55mg/dL and a non-HDL-C < 80mg/dL. In patients that are at high-risk the goal my goal is an LDL-C < 70mg/dL and a non-HDL-C < 100mg/dL. In patients with moderate risk an LDL-C goal of < 100mg/dL and a non-HDL c < 130mg/dL is appropriate. If the levels are not at goal, I first adjust the statin dose until the patient is taking the maximally tolerated statin dose and then consider adding additional medications.  In patients with diabetes who are less than 40 years of age I initiate statin therapy if the patient has overt ASCVD, long standing diabetes, or risk factors/risk enhancers for ASCVD and the LDL and non-HDL-C levels are not at goal. In these younger patients I also calculate the life time risk of ASCVD events to start a discussion of beginning early therapy given the abundance of data indicating that initiating LDL-C lowering therapy early has great potential in markedly lowering ASCVD risk (226,227). In patients over 75 years of age with a reasonable life expectancy I begin moderate statin therapy and adjust based on response. When there is difficulty classifying a patient’s risk, I will obtain a coronary calcium score and use the score to help stratify the patient’s risk. In all cases the benefits and risks of lipid lowering therapy needs to be discussed with patients and the patient’s personnel preferences taken into account.

 

Table 12. ASCVD Risk Categories and Treatment Goals

Risk Category

Risk Factors/10-year risk

LDL-C mg/dL

Non-HDL-C mg/dL

Very High Risk

Diabetes and clinical ASCVD, multiple risk factors, 10-year risk > 20%

<55

<85

High Risk

Diabetes with one or more risk factors, 10-year risk >7.5% to <20%

<70

<100

Moderate Risk

Diabetes and no other risk factors. 10-year risk <7.5%

<100

<130

 

Table 13. Drug Therapy According to Risk Category that is Typically Required

Very High Risk

Intensive statin therapy + ezetimibe. Add PCSK9 if not close to goal

High Risk

Intensive statin therapy. Add ezetimibe if not at goal

Moderate Risk

Moderate statin therapy. Increase to intensive statin therapy if not at goal

 

TREATMENT OF LIPID ABNORMALITIES IN PATIENT WITH DIABETES

 

Life Style Changes and Weight Loss

 

Initial treatment of lipid disorders should focus on lifestyle changes. There is little debate that exercise is beneficial and that all patients with diabetes should, if possible, exercise for at least 150 minutes per week (for example 30 minutes 5 times per week). Exercise will decrease serum TG levels and increase HDL-C levels (an increase in HDL-C requires vigorous exercise) (124). Exercise increases fitness and improves insulin resistance even with limited weight loss; reductions in obesity are even more beneficial. It should be noted that many patients with diabetes may have substantial barriers to participating in exercise programs, such as comorbidities that limit exercise tolerance, risk of hypoglycemia, and presence of microvascular complications (visual impairment, neuropathy) that make exercise difficult.

 

Diet is debated to a greater extent and for detailed information on nutrition therapy for adults with diabetes see the consensus report by the American Diabetes Association (228). Everyone agrees that weight loss in obese patients is essential (124). But how this can be achieved is hotly debated with many different "experts" advocating different dietary approaches. The wide diversity of approach is likely due to the failure of any approach to be effective in the long term for the majority of obese patients with diabetes. If successful, weight loss will decrease serum TG levels, increase HDL-C levels, and modestly reduce LDL-C (124,229). To reduce LDL-C levels, it is important that the diet decrease saturated fat, trans fatty acids, and cholesterol intake. Increasing soluble fiber is also helpful.

 

It is debated whether a low fat, high complex carbohydrate diets vs. a high monounsaturated fat  diet is ideal for obese patients with diabetes (124). One can find "experts" in favor of either of these approaches and there are pros and cons to each approach. It is essential to recognize that both approaches reduce simple sugars, saturated fat, trans fatty acids, and cholesterol intake. The high complex carbohydrate diet will increase serum TG levels in some patients and if the amount of fat in the diet is markedly reduced serum HDL-C levels may decrease. In obese patients, it has been postulated that a diet high in monounsaturated fats, because of the increase in caloric density, will lead to an increase in weight gain. Both diets reduce saturated fat and cholesterol intake that will result in reductions in LDL-C levels. Additionally, both diets also reduce trans-fatty acid intake, which will have a beneficial effect on LDL and HDL-C levels and simple sugars, which will have a beneficial effect on TG levels. Very high levels of TG (>1000mg/dL), require diets that are very low in fat.

 

The available data do not indicate that any particular diet is best for inducing weight loss and it is essential to adapt the diet to fit the food preferences of the patient. Ultimately no weight loss diet will be successful if the patient cannot follow the diet for the long term and therefore the diet needs to be tailored to the specific preferences of the patient. For more detailed information on the effect of diet on lipid and lipoprotein levels and cardiovascular disease see the Endotext chapter “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” (229).

 

While it is widely accepted that lifestyle changes will decrease ASCVD events it should be recognized that the Look Ahead trial failed to demonstrate a reduction in ASCVD events (230). In this trial, over 5,000 overweight or obese patients with T2DM were randomized to either an intensive lifestyle intervention group that promoted weight loss through decreased caloric intake and increased physical activity or to a group that received diabetes support and education (control group). After a median follow-up of 9.6 years there was no difference in cardiovascular events (hazard ratio in the intervention group, 0.95; 95% CI 0.83 to 1.09; P=0.51). A major limitation of this study was that while the weight difference between groups was impressive during the first year of the trial, over time the differences greatly narrowed such that at the end of the trial the intensive group had a 6.0% weight loss while the control group had a 3.5% weight loss. This very modest weight difference demonstrates the difficulty in sustaining long term lifestyle changes. Thus, while weight loss and diet therapy are likely to be beneficial in reducing cardiovascular events, in clinical practice they are seldom sufficient because long-term life style changes are very difficult for most patients to maintain.

 

In contrast to the failure of lifestyle therapy in the Look Ahead trial to reduce cardiovascular events, the PREDIMED trial employing a Mediterranean diet (increased monounsaturated fats) did reduce the incidence of major ASCVD events (231,232). In this multicenter trial center 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. Approximately 50% of the patients in this trial had T2DM. 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 with and without diabetes. 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 (233).

 

The CORDIOPREV study was a single center randomized trial that compared a Mediterranean diet to a low-fat diet in 1,002 patients with ASCVD (234). Approximately 50% of the patients had diabetes. 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 is unknown.

 

Finally, another secondary prevention trial of a Mediterranean diet has also demonstrated a reduction in cardiovascular events. The Lyon Diet Heart Study randomized 584 patients who had a MI within 6 months to a Mediterranean type diet vs usual diet (235,236). There was a marked reduction in events in the group of patients randomized to the Mediterranean diet (cardiac death and nonfatal MI rate was 4.07 per 100 patient years in the control diet vs. 1.24 in the Mediterranean diet; p<0.0001). Unfortunately, there is no indication of the number of patients with diabetes in the Lyon Diet Heart Study or whether patients with diabetes responded similar to the entire group. Lipid levels were similar in both groups in this trial (235).

 

The results of these three randomized trials indicate that we should be encouraging our patients to follow a Mediterranean type diet. 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.

 

For additional information on the effect of diets on lipid levels and ASCVD see the chapter entitled “The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels” in the Lipids and Lipoproteins section of Endotext (229).

 

Bariatric surgery can have profound effects on weight and can result in marked improvements in lipid profiles with a decrease in TG and LDL-C and an increase in HDL-C (124,229) Additionally, observational studies have shown a decrease in cardiovascular events following bariatric surgery in patients with and without diabetes (237-241). For additional information see the chapter entitled “Obesity and Dyslipidemia” (124).

 

Ethanol and simple sugars, in particular fructose, increase serum TG levels in susceptible patients. In patients with hypertriglyceridemia efforts should be made to reduce the intake of ethanol, simple sugars, and fructose (229).

 

Lastly, in the past some "experts" advocated the addition of fish oil supplements to reduce cardiovascular events. However, both the Origin Trial and the ASCEND Trial did not demonstrate that fish oil supplements were beneficial in patients with T2DM or patients at high risk for the development of T2DM (207,208). It should be recognized that higher doses of fish oil are required to lower serum triglyceride levels (~ 3-4 grams of DHA/EPA per day) and are useful in treating patients with high TG levels (242). Most studies of fish oil supplements in patients with diabetes have demonstrated that this is a safe approach and that worsening of glycemic control does not occur in patients with diabetes treated with fish oil supplements (242). Additionally, in some patient's high dose fish oil increases LDL-C levels, particularly when serum TG levels are very high (242). For additional information on fish oil see the chapter on Triglyceride Lowering Drugs (209).

 

Drug Therapy

 

The effect of statins, fibrates, niacin, ezetimibe, omega-3-fatty acids, bile acid sequestrants, bempedoic acid, and PCSK9 inhibitors on lipid levels in patients with diabetes is virtually identical to that seen in non-diabetic patients (Table 14). Below we will highlight issues particularly relevant to the use of these drugs in patients with diabetes. For detailed information on lipid lowering drugs see the chapters on Triglyceride Lowering Drugs and Cholesterol Lowering Drugs (141,209).

 

STATINS

 

Statins are easy to use and generally well tolerated by patients with diabetes. However, statins can adversely affect glucose homeostasis. In patients without diabetes the risk of developing diabetes is increased by approximately 10% with higher doses of statin causing a greater risk than more moderate doses (243,244). The mechanism for this adverse effect is unknown but older, obese patients with higher baseline glucose levels are at greatest risk. 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 HbA1c than the placebo group indicating that statin therapy is associated with only a very small increase in HbA1c levels in patients with diabetes, which is unlikely to be clinically significant (245). Individual studies such as CARDS and the Heart Protection Study have also shown only a very modest effect of statins on HbA1c levels in patients with diabetes (163,166,246). Muscle symptoms occur in patients with diabetes similar to what is observed in patients without diabetes.

 

EZETIMIBE

 

Ezetimibe is easy to use and generally well tolerated by patients with diabetes. Ezetimibe does not appear to increase the risk of new onset diabetes (199,247,248).

 

FIBRATES

 

Fibrates are easy to use and generally well tolerated by patients with diabetes. When combining fibrates with statin therapy it is best to use fenofibrate as the risk of inducing myositis is much less than when statins are used in combination with gemfibrozil, which can inhibit statin metabolism (249). In the ACCORD-LIPID Trial the incidence of muscle disorders was not increased in the statin + fenofibrate group compared to statin alone (189). The dose of fenofibrate needs to be adjusted in patients with renal disease and fenofibrate itself can induce a reversible increase in serum creatinine levels. It should be noted that marked reductions in HDL-C levels can occur in some patients treated with both fenofibrate and a TZD (250).

 

Diabetic Retinopathy

 

Fenofibrate has been shown to have beneficial effects on diabetic eye disease. The FIELD study, described earlier, was a randomized trial of fenofibrate vs. placebo in patients with T2DM. Laser treatment for retinopathy was significantly lower in the fenofibrate group than in the placebo group (3.4% patients on fenofibrate vs 4.9% on placebo; p=0.0002) (191). Fenofibrate therapy reduced the need for laser therapy to a similar extent for maculopathy (31% decrease) and for proliferative retinopathy (30% decrease). In the ophthalmology sub-study (n=1012), the primary endpoint of 2-step progression of retinopathy grade did not differ significantly between the fenofibrate and control groups (9.6% patients on fenofibrate vs 12.3% on placebo; p=0.19). In patients without pre-existing retinopathy there was no difference in progression (11.4% vs 11.7%; p=0.87). However, in patients with pre-existing retinopathy, significantly fewer patients on fenofibrate had a 2-step progression than did those on placebo (3.1% patients vs 14.6%; p=0.004). A composite endpoint of 2-step progression of retinopathy grade, macular edema, or laser treatments was significantly reduced in the fenofibrate group (HR 0.66, 95% CI 0.47-0.94; p=0.022).

 

In the ACCORD Study a subgroup of participants was evaluated for the progression of diabetic retinopathy by 3 or more steps on the Early Treatment Diabetic Retinopathy Study Severity Scale or the development of diabetic retinopathy necessitating laser photocoagulation or vitrectomy over a four-year period (190). At 4 years, the rates of progression of diabetic retinopathy were 6.5% with fenofibrate therapy (n=806) vs. 10.2% with placebo (n=787) (adjusted odds ratio, 0.60; 95% CI, 0.42 to 0.87; P = 0.006). Of note, this reduction in the progression of diabetic retinopathy was of a similar magnitude as intensive glycemic treatment vs. standard therapy.

 

Taken together these results indicate that fibrates have beneficial effects on the progression of diabetic retinopathy. The mechanisms by which fibrates decrease diabetic retinopathy are unknown.

 

Diabetic Nephropathy

 

The Diabetes Atherosclerosis Intervention Study (DAIS) evaluated the effect of fenofibrate therapy (n= 155) vs. placebo (n=159) on changes in urinary albumin excretion in patients with T2DM (251). Fenofibrate significantly reduced the worsening of albumin excretion (fenofibrate 8% vs. placebo 18%; P < 0.05). This effect was primarily due to reduced progression from normal albumin excretion to microalbuminuria (fenofibrate 3% vs. 18% placebo; P < 0.001).

 

In the FIELD trial, fenofibrate reduced urine albumin/creatinine ratio by 24% vs 11% in placebo group (p < 0.001), with 14% less progression and 18% more albuminuria regression (p < 0.001) in the fenofibrate group than in participants on placebo (252). As expected, fenofibrate therapy acutely increased plasma creatinine levels and decreased eGFR but over the long term, the increase in plasma creatinine was decreased in the fenofibrate group compared to the placebo group (14% decrease; p=0.01). Similarly, there was a slower annual decrease in eGFR in the fenofibrate group (1.19 vs 2.03 mL/min/1.73m2   annually, p < 0.001). The effect of fenofibrate on kidney function was greater in those with higher TG and lower HDL levels. End-stage renal disease, dialysis, renal transplant, and renal death were similar in the fenofibrate and placebo groups, but the incidence was low.

 

In the ACCORD-LIPID trial the post-randomization incidence of microalbuminuria was 38.2% in the fenofibrate group and 41.6% in the placebo group (p=0.01) and post-randomization incidence of macroalbumuria was 10.5% in the fibrate group and 12.3% in the placebo group (p=0.04) indicating a modest reduction in the development of proteinuria in patients treated with fenofibrate (189). There was no significant difference in the incidence of end-stage renal disease or need for dialysis between the fenofibrate group and the placebo group.

 

These studies suggest that fibrates may have a beneficial effect on diabetic kidney disease. One should recognize that reducing proteinuria is a surrogate marker and may not indicate a reduction in the development of end stage renal disease. The mechanisms accounting for decreased in proteinuria are unknown.

 

Amputations

 

In the FIELD study the risks of first amputation were decreased by 36% (p=0.02) and minor amputation events without known large-vessel disease by 47% (p=0.027) in the fenofibrate treated group (253). The reduction in amputations was independent of glucose control or dyslipidemia. No difference between the risks of major amputations was seen in the placebo and fenofibrate groups. The basis for this reduction in amputations is unknown.

 

Do Fibrates have an Independent Effect on Microvascular Disease?

 

Multiple studies cited above have now shown that fenofibrate decreases retinopathy, nephropathy, and amputation in the absence of large vessel disease. The effects are independent of blood glucose control. Given that there also was no effect of fenofibrate on cardiovascular (macrovascular) disease, these results may suggest that fenofibrate has an independent effect on microvascular disease.  Further studies are warranted, but these results should be taken into account when considering treatment of marked hypertriglyceridemia in patients with diabetes. 

 

BILE ACID SEQUESTRANTS

 

Bile acid sequestrants are relatively difficult to take due to GI toxicity (mainly constipation) (141). Diabetic subjects have an increased prevalence of constipation, which may be exacerbated by the use of bile acid sequestrants. On the other hand, in diabetic patients with diarrhea, the use of bile acid sequestrants may be advantageous. Bile acid sequestrants may also increase serum TG levels, which can be a problem in patients with diabetes who are already hypertriglyceridemic (141). An additional difficulty in using bile acid sequestrants is their potential for binding other drugs (141). Many drugs should be taken either two hours before or four hours after taking bile acid sequestrants to avoid the potential of decreased drug absorption. Patients with diabetes are frequently on multiple drugs for glycemic control, hypertension, etc., and it can sometimes be difficult to time the ingestion of bile resin sequestrants to avoid these other drugs. Colesevelam (Welchol) is a bile acid sequestrant that comes in pill, powder, or chewable bars and causes fewer side effects and has fewer interactions with other drugs than other preparations (254). The usual dose 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). Of particular note is that a number of studies have shown that colesevelam improves glycemic control in patients with diabetes resulting in an approximately 0.5% decrease in A1c levels (255).

 

NIACIN

 

Niacin is well known to cause skin flushing and itching and GI upset (256). Additionally, niacin reduces insulin sensitivity (i.e., causes insulin resistance), which can worsen glycemic control (256). Studies have shown that niacin is usually well tolerated in diabetic subjects who are in good glycemic control (257,258). In patients with poor glycemic control, niacin is more likely to adversely impact glucose levels. In the HPS2-Thrive trial, niacin therapy significantly worsened glycemic control in patients with diabetes and induced new onset diabetes in 1.3% of subjects that were non-diabetic (195). High doses of niacin are more likely to adversely affect glycemic control. Niacin can also increase serum uric acid levels and induce gout, both of which are already common in obese patients with T2DM (256). Additionally recent trials have reported an increased incidence of infection and bleeding with niacin therapy (256). However, niacin is the most effective drug in increasing HDL-C levels, which are frequently low in patients with diabetes. 

 

OMEGA-3-FATTY ACIDS

 

A Cochrane review of fish oil in patients with diabetes have demonstrated that this is a safe approach and does not result in worsening of glycemic control in patients with diabetes (242). Fish oil effectively lowers TG levels but, in some patients, particularly those with significant hypertriglyceridemia, high dose fish oil increases LDL-C levels (242). It should be noted that fish oil products that contain just EPA (Vascepa) do not adversely affect LDL-C levels (259). When using fish oil to lower serum TG levels it is important to recognize that one is aiming to provide 3-4 grams of DHA/EPA per day. The quantity of these active omega-3-fatty acids can vary greatly from product to product. Prescription fish oil products contain large amounts of these active ingredients whereas the amount of DHA/EPA in food supplements can vary greatly and in some products levels are very low. Additionally, while prescription omega-3-fatty acid preparations have high levels of quality control, omega-3-fish oil food supplements may have contaminants and the dosage may not be precisely controlled.

 

PCSK9 INHIBITORS

 

The beneficial effects of PCSK9 inhibitors in patients with diabetes is similar to what is observed in non-diabetic patients. Additionally, except for local reactions at the injection sites PCSK9 inhibitors do not seem to cause major side effects. PCSK9 inhibitors do not appear to increase the risk of developing new-onset diabetes (260,261).

 

BEMPEDOIC ACID

 

The effect of bempedoic acid on LDL-C levels in patients with diabetes are similar to the decreases seen on non-diabetics. Patients with T2DM often have elevated uric acid levels and an increased risk of gouty attacks and a major side effect of bempedoic acid is elevating uric acid levels (141). 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. Elevations in blood uric acid levels may lead to the development of gout and 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). 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.

 

In a meta-analysis, bempedoic acid therapy was associated with a decrease in the onset of diabetes and worsening of diabetes  (RR 0.65, p = 0.03) (7/100 vs 6.4/100 patient years) (262). In a study focusing solely on the development of new onset diabetes it was reported that new-onset diabetes/hyperglycemia occurred less frequently with bempedoic acid vs placebo (263). In the bempedoic acid cardiovascular outcome trial (CLEAR Outcomes) the development of diabetes and worsening of pre-existing diabetes was similar in the bempedoic acid and placebo groups (187).  

 

Table 14. Effect of Lipid Lowering Drugs

 

LDL-C

HDL-C

TG

 

Statins

↓ 20-60%

↑ 5-15%

↓ 0-35%*

Bile acid sequestrants

↓ 10-30%

↑ 0-10%

↑ 0-10%**

Fibrates

↓ 0-15%***

↑ 5-15%

↓ 20-50%

Niacin

↓ 10-25%

↑ 10-30%

↓ 20-50%

Ezetimibe

↓ 15-25%

↑ 1-3%

↓ 10-20%

PCSK9 Inhibitors

↓ 50-60%

↑ 5-15%

↓ 5-20%

Bempedoic Acid

↓ 15-25%

↓ 5-6%

No change

High Dose Fish Oil

↑ 0- 50%***

↑ 4- 9%

↓ 20- 50%*

 *Patients with elevated TG have largest decrease

** In patients with high TG may cause marked increase

*** In patients with high TG may increase LDL

 

Therapeutic Approach

 

FIRST PRIORITY- LDL-C

 

The first priority in treating lipid disorders in patients with diabetes is to lower the LDL-C levels to goal, unless TG are markedly elevated (> 500- 1000mg/dL), which increases the risk of pancreatitis. LDL-C is the first priority because the database linking lowering LDL-C with reducing ASCVD is extremely strong and we now have the ability to markedly decrease LDL-C levels. Dietary therapy is the initial step but, in almost all patients, will not be sufficient to achieve the LDL-C goals. If patients are willing and able to make major changes in their diet it is possible to achieve significant reductions in LDL-C levels but this seldom occurs in clinical practice (264).

 

Statins are the first-choice drugs to lower LDL-C levels and the vast majority of diabetic patients will require statin therapy. There are several statins currently available in the US and they are available as generic drugs and therefore relatively inexpensive. The particular statin used may be driven by price, ability to lower LDL-C levels, and potential drug interactions. Patients with ASCVD (secondary prevention patients) should be started on intensive statin therapy (atorvastatin 40-80mg per day or rosuvastatin 20-40mg per day). Given the extensive data showing that the lower the LDL-C the greater the reduction in ASCVD events most secondary prevention patients would benefit from the addition of ezetimibe to maximize LDL-C lowering. Ezetimibe is now a generic drug and therefore this strategy will not markedly increase costs. Similarly, primary prevention patients who are at high risk for cardiovascular events will also benefit from the use of high intensity statin therapy in combination with ezetimibe. Primary prevention patients at moderate risk can be started on moderate intensity statin therapy.

 

If a patient is unable to tolerate statins or statins as monotherapy are not sufficient to lower LDL-C to goal the second-choice drug is either ezetimibe or a PCSK9 inhibitor. Ezetimibe can be added to any statin. PCSK9 inhibitors can also be added to any statin and are the drug of choice if a large decrease in LDL-C is required to reach goal (PCSK9 inhibitors will lower LDL-C levels by 50-60% when added to a statin, whereas ezetimibe will only lower LDL-C by approximately 20%).  Bile acid sequestrants and bempedoic acid are alternatives with the use of a bile acid sequestrant particularly useful if a reduction in A1c level is also needed. It should be noted that in statin intolerant patients with ASCVD or at high risk (approximately 45% with diabetes), bempedoic acid has been shown to reduce cardiovascular events by 13% (HR 0.87; 95% CI 0.79 to 0.96; P = 0.004) (187). Ezetimibe, PCSK9 inhibitors, bempedoic acid, and bile acid sequestrants additively lower LDL-C levels when used in combination with a statin, because these drugs increase hepatic LDL receptor levels by different mechanisms, thereby resulting in a reduction in serum LDL-C levels (141). Niacin and the fibrates also lower LDL-C levels but are not usually employed to lower LDL-C levels. 

 

SECOND PRIORITY- NON-HDL-C

 

The second priority should be non-HDL-C (non-HDL-C = total cholesterol – HDL-C), which is particularly important in patients with elevated TG levels (>150mg/dL). Non-HDL-C is a measure of all the pro-atherogenic apolipoprotein B containing particles. Numerous studies have shown that non-HDL-C is a strong risk factor for the development of ASCVD (265). The non-HDL-C goals are approximately 30mg/dL greater than the LDL-C goals. For example, if the LDL goal is <100mg/dL then the non-HDL-C goal would be <130mg/dL. Drugs that reduce either LDL-C or TG levels will reduce non-HDL-C levels. To lower TG levels initial therapy should focus on glycemic control and lifestyle changes including weight loss if indicated and a decrease in simple sugars and ethanol intake. Additionally, if possible, discontinue medications that increase triglyceride levels. As discussed above, studies with the omega-3-fatty acid icosapent ethyl (EPA; Vascepa) added to statin therapy have reduced the risk of cardiovascular events. The National Lipid Association has recommended “that for patients aged ≥45 years with clinical ASCVD, or aged ≥50 years with diabetes mellitus requiring medication plus ≥1 additional risk factor, with fasting TGs 135 to 499 mg/dL on high-intensity or maximally tolerated statin therapy (±ezetimibe), treatment with icosapent ethyl is recommended for ASCVD risk reduction” (266). As noted earlier in this chapter there is controversy regarding the benefits of icosapent ethyl on cardiovascular outcomes with some experts interpreting the beneficial results of the REDUCE-IT trial as being due to the adverse effects of the mineral oil placebo (217).

 

VERY HIGH TG

 

Patients with very high TG levels (> 500-1000 mg/dL) are at risk of pancreatitis and therefore lifestyle interventions including diet, exercise, and weight loss if indicated should be initiated early. Treatment is a low-fat diet and glycemic control. Additionally, decreasing simple sugars and avoiding alcohol is beneficial. When the TG levels are very elevated (> 1000mg/dL) a very low-fat diet (5-20% of calories as fat) should be the primary treatment until the TG levels decrease to < 1000mg/dL. Treating secondary disorders that raise TG levels and when possible, stopping drugs that increase TG levels is essential. If the TG levels remain above 500mg/dL the addition of fenofibrate or omega-3-fatty acids is indicated.

 

LOW HDL-C

 

While there is strong epidemiologic data linking low HDL-C levels with ASCVD there is no clinical trials demonstrating that increasing HDL-C levels reduce ASCVD. Thus, the use of drugs such as niacin to raise HDL-C levels is not recommended.

 

CONCLUSION

 

Patients with diabetes, particularly T2DM, often have dyslipidemia. Modern therapy of patients with diabetes demands that we aggressively treat lipids to reduce the high risk of ASCVD in this susceptible population and in those with very high TG to reduce the risk of pancreatitis.

 

REFERENCES

 

  1. Milicevic Z, Raz I, Beattie SD, Campaigne BN, Sarwat S, Gromniak E, Kowalska I, Galic E, Tan M, Hanefeld M. Natural history of cardiovascular disease in patients with diabetes: role of hyperglycemia. Diabetes Care 2008; 31 Suppl 2:S155-160
  2. Feingold KR, Siperstein MD. Diabetic vascular disease. Adv Intern Med 1986; 31:309-340
  3. Regensteiner JG, Golden S, Huebschmann AG, Barrett-Connor E, Chang AY, Chyun D, Fox CS, Kim C, Mehta N, Reckelhoff JF, Reusch JE, Rexrode KM, Sumner AE, Welty FK, Wenger NK, Anton B. Sex Differences in the Cardiovascular Consequences of Diabetes Mellitus: A Scientific Statement From the American Heart Association. Circulation 2015; 132:2424-2447
  4. Fox CS, Golden SH, Anderson C, Bray GA, Burke LE, de Boer IH, Deedwania P, Eckel RH, Ershow AG, Fradkin J, Inzucchi SE, Kosiborod M, Nelson RG, Patel MJ, Pignone M, Quinn L, Schauer PR, Selvin E, Vafiadis DK. Update on Prevention of Cardiovascular Disease in Adults With Type 2 Diabetes Mellitus in Light of Recent Evidence: A Scientific Statement From the American Heart Association and the American Diabetes Association. Diabetes Care 2015; 38:1777-1803
  5. Low Wang CC, Hess CN, Hiatt WR, Goldfine AB. Clinical Update: Cardiovascular Disease in Diabetes Mellitus: Atherosclerotic Cardiovascular Disease and Heart Failure in Type 2 Diabetes Mellitus - Mechanisms, Management, and Clinical Considerations. Circulation 2016; 133:2459-2502
  6. Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA 1979; 241:2035-2038
  7. Members Writing Group, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB. Executive Summary: Heart Disease and Stroke Statistics--2016 Update: A Report From the American Heart Association. Circulation 2016; 133:447-454
  8. Haffner SM, Lehto S, Ronnemaa T, Pyorala K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229-234
  9. Evans JM, Wang J, Morris AD. Comparison of cardiovascular risk between patients with type 2 diabetes and those who had had a myocardial infarction: cross sectional and cohort studies. BMJ 2002; 324:939-942
  10. Wannamethee SG, Shaper AG, Whincup PH, Lennon L, Sattar N. Impact of diabetes on cardiovascular disease risk and all-cause mortality in older men: influence of age at onset, diabetes duration, and established and novel risk factors. Arch Intern Med 2011; 171:404-410
  11. Howard BV, Best LG, Galloway JM, Howard WJ, Jones K, Lee ET, Ratner RE, Resnick HE, Devereux RB. Coronary heart disease risk equivalence in diabetes depends on concomitant risk factors. Diabetes Care 2006; 29:391-397
  12. Rawshani A, Rawshani A, Franzen S, Sattar N, Eliasson B, Svensson AM, Zethelius B, Miftaraj M, McGuire DK, Rosengren A, Gudbjornsdottir S. Risk Factors, Mortality, and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2018; 379:633-644
  13. Lind M, Svensson AM, Kosiborod M, Gudbjornsdottir S, Pivodic A, Wedel H, Dahlqvist S, Clements M, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med 2014; 371:1972-1982
  14. de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Diabetes Care 2014; 37:2843-2863
  15. Maahs DM, Daniels SR, de Ferranti SD, Dichek HL, Flynn J, Goldstein BI, Kelly AS, Nadeau KJ, Martyn-Nemeth P, Osganian SK, Quinn L, Shah AS, Urbina E. Cardiovascular disease risk factors in youth with diabetes mellitus: a scientific statement from the American Heart Association. Circulation 2014; 130:1532-1558
  16. Huxley RR, Peters SA, Mishra GD, Woodward M. Risk of all-cause mortality and vascular events in women versus men with type 1 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol 2015; 3:198-206
  17. Rawshani A, Sattar N, Franzen S, Rawshani A, Hattersley AT, Svensson AM, Eliasson B, Gudbjornsdottir S. Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 2018; 392:477-486
  18. Chillaron JJ, Flores Le-Roux JA, Benaiges D, Pedro-Botet J. Type 1 diabetes, metabolic syndrome and cardiovascular risk. Metabolism 2014; 63:181-187
  19. Constantino MI, Molyneaux L, Limacher-Gisler F, Al-Saeed A, Luo C, Wu T, Twigg SM, Yue DK, Wong J. Long-term complications and mortality in young-onset diabetes: type 2 diabetes is more hazardous and lethal than type 1 diabetes. Diabetes Care 2013; 36:3863-3869
  20. Preis SR, Hwang SJ, Coady S, Pencina MJ, D'Agostino RB, Sr., Savage PJ, Levy D, Fox CS. Trends in all-cause and cardiovascular disease mortality among women and men with and without diabetes mellitus in the Framingham Heart Study, 1950 to 2005. Circulation 2009; 119:1728-1735
  21. Matuleviciene-Anangen V, Rosengren A, Svensson AM, Pivodic A, Gudbjornsdottir S, Wedel H, Kosiborod M, Haraldsson B, Lind M. Glycaemic control and excess risk of major coronary events in persons with type 1 diabetes. Heart 2017; 103:1687-1695
  22. Miller RG, Orchard TJ, Costacou T. 30-Year Cardiovascular Disease in Type 1 Diabetes: Risk and Risk Factors Differ by Long-term Patterns of Glycemic Control. Diabetes Care 2022; 45:142-150
  23. Abraira C, Colwell JA, Nuttall FQ, Sawin CT, Nagel NJ, Comstock JP, Emanuele NV, Levin SR, Henderson W, Lee HS. Veterans Affairs Cooperative Study on glycemic control and complications in type II diabetes (VA CSDM). Results of the feasibility trial. Veterans Affairs Cooperative Study in Type II Diabetes. Diabetes Care1995; 18:1113-1123
  24. Goldner MG, Knatterud GL, Prout TE. Effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. 3. Clinical implications of UGDP results. JAMA 1971; 218:1400-1410
  25. Meinert CL, Knatterud GL, Prout TE, Klimt CR. A study of the effects of hypoglycemic agents on vascular complications in patients with adult-onset diabetes. II. Mortality results. Diabetes 1970; 19:Suppl:789-830
  26. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 1993; 329:977-986
  27. Ohkubo Y, Kishikawa H, Araki E, Miyata T, Isami S, Motoyoshi S, Kojima Y, Furuyoshi N, Shichiri M. Intensive insulin therapy prevents the progression of diabetic microvascular complications in Japanese patients with non-insulin-dependent diabetes mellitus: a randomized prospective 6-year study. Diabetes Res Clin Pract 1995; 28:103-117
  28. Shichiri M, Kishikawa H, Ohkubo Y, Wake N. Long-term results of the Kumamoto Study on optimal diabetes control in type 2 diabetic patients. Diabetes Care 2000; 23 Suppl 2:B21-29
  29. Lachin JM, Orchard TJ, Nathan DM, Group DER. Update on cardiovascular outcomes at 30 years of the diabetes control and complications trial/epidemiology of diabetes interventions and complications study. Diabetes Care2014; 37:39-43
  30. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, Raskin P, Zinman B, Diabetes C, Complications Trial/Epidemiology of Diabetes I, Complications Study Research G. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 2005; 353:2643-2653
  31. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:837-853
  32. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359:1577-1589
  33. Malmberg K. Prospective randomised study of intensive insulin treatment on long term survival after acute myocardial infarction in patients with diabetes mellitus. DIGAMI (Diabetes Mellitus, Insulin Glucose Infusion in Acute Myocardial Infarction) Study Group. BMJ 1997; 314:1512-1515
  34. Mellbin LG, Malmberg K, Norhammar A, Wedel H, Ryden L, Investigators D. The impact of glucose lowering treatment on long-term prognosis in patients with type 2 diabetes and myocardial infarction: a report from the DIGAMI 2 trial. Eur Heart J 2008; 29:166-176
  35. Action to Control Cardiovascular Risk in Diabetes Study Group, Gerstein HC, Miller ME, Byington RP, Goff DC, Jr., Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail-Beigi F, Grimm RH, Jr., Probstfield JL, Simons-Morton DG, Friedewald WT. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545-2559
  36. Gerstein HC, Miller ME, Ismail-Beigi F, Largay J, McDonald C, Lochnan HA, Booth GL, ACCORD STUDY Group. Effects of intensive glycaemic control on ischaemic heart disease: analysis of data from the randomised, controlled ACCORD trial. Lancet 2014; 384:1936-1941
  37. ACCORD STUDY Group. Nine-Year Effects of 3.7 Years of Intensive Glycemic Control on Cardiovascular Outcomes. Diabetes Care 2016; 39:701-708
  38. Advance Collaborative Group, Patel A, MacMahon S, Chalmers J, Neal B, Billot L, Woodward M, Marre M, Cooper M, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Liu L, Mancia G, Mogensen CE, Pan C, Poulter N, Rodgers A, Williams B, Bompoint S, de Galan BE, Joshi R, Travert F. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560-2572
  39. Zoungas S, Chalmers J, Neal B, Billot L, Li Q, Hirakawa Y, Arima H, Monaghan H, Joshi R, Colagiuri S, Cooper ME, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Lisheng L, Mancia G, Marre M, Matthews DR, Mogensen CE, Perkovic V, Poulter N, Rodgers A, Williams B, MacMahon S, Patel A, Woodward M, Group A-OC. Follow-up of blood-pressure lowering and glucose control in type 2 diabetes. N Engl J Med 2014; 371:1392-1406
  40. Duckworth W, Abraira C, Moritz T, Reda D, Emanuele N, Reaven PD, Zieve FJ, Marks J, Davis SN, Hayward R, Warren SR, Goldman S, McCarren M, Vitek ME, Henderson WG, Huang GD, Investigators V. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129-139
  41. Hayward RA, Reaven PD, Wiitala WL, Bahn GD, Reda DJ, Ge L, McCarren M, Duckworth WC, Emanuele NV, Investigators V. Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. N Engl J Med2015; 372:2197-2206
  42. Reaven PD, Emanuele NV, Wiitala WL, Bahn GD, Reda DJ, McCarren M, Duckworth WC, Hayward RA, Investigators V. Intensive Glucose Control in Patients with Type 2 Diabetes - 15-Year Follow-up. N Engl J Med2019; 380:2215-2224
  43. Buehler AM, Cavalcanti AB, Berwanger O, Figueiro M, Laranjeira LN, Zazula AD, Kioshi B, Bugano DG, Santucci E, Sbruzzi G, Guimaraes HP, Carvalho VO, Bordin SA. Effect of tight blood glucose control versus conventional control in patients with type 2 diabetes mellitus: a systematic review with meta-analysis of randomized controlled trials. Cardiovasc Ther 2013; 31:147-160
  44. Control Group, Turnbull FM, Abraira C, Anderson RJ, Byington RP, Chalmers JP, Duckworth WC, Evans GW, Gerstein HC, Holman RR, Moritz TE, Neal BC, Ninomiya T, Patel AA, Paul SK, Travert F, Woodward M. Intensive glucose control and macrovascular outcomes in type 2 diabetes. Diabetologia 2009; 52:2288-2298
  45. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998; 352:854-865
  46. Kooy A, de Jager J, Lehert P, Bets D, Wulffele MG, Donker AJ, Stehouwer CD. Long-term effects of metformin on metabolism and microvascular and macrovascular disease in patients with type 2 diabetes mellitus. Arch Intern Med 2009; 169:616-625
  47. Hong J, Zhang Y, Lai S, Lv A, Su Q, Dong Y, Zhou Z, Tang W, Zhao J, Cui L, Zou D, Wang D, Li H, Liu C, Wu G, Shen J, Zhu D, Wang W, Shen W, Ning G. Effects of metformin versus glipizide on cardiovascular outcomes in patients with type 2 diabetes and coronary artery disease. Diabetes Care 2013; 36:1304-1311
  48. Goldberg RB, Orchard TJ, Crandall JP, Boyko EJ, Budoff M, Dabelea D, Gadde KM, Knowler WC, Lee CG, Nathan DM, Watson K, Temprosa M. Effects of Long-term Metformin and Lifestyle Interventions on Cardiovascular Events in the Diabetes Prevention Program and Its Outcome Study. Circulation 2022; 145:1632-1641
  49. Goldberg RB, Aroda VR, Bluemke DA, Barrett-Connor E, Budoff M, Crandall JP, Dabelea D, Horton ES, Mather KJ, Orchard TJ, Schade D, Watson K, Temprosa M. Effect of Long-Term Metformin and Lifestyle in the Diabetes Prevention Program and Its Outcome Study on Coronary Artery Calcium. Circulation 2017; 136:52-64
  50. Fitch K, Abbara S, Lee H, Stavrou E, Sacks R, Michel T, Hemphill L, Torriani M, Grinspoon S. Effects of lifestyle modification and metformin on atherosclerotic indices among HIV-infected patients with the metabolic syndrome. AIDS 2012; 26:587-597
  51. Rosenstock J, Kahn SE, Johansen OE, Zinman B, Espeland MA, Woerle HJ, Pfarr E, Keller A, Mattheus M, Baanstra D, Meinicke T, George JT, von Eynatten M, McGuire DK, Marx N. Effect of Linagliptin vs Glimepiride on Major Adverse Cardiovascular Outcomes in Patients With Type 2 Diabetes: The CAROLINA Randomized Clinical Trial. JAMA 2019;
  52. Navigator Study Group, Holman RR, Haffner SM, McMurray JJ, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, Buckley BM, Chacra AR, Chiang FT, Charbonnel B, Chow CC, Davies MJ, Deedwania P, Diem P, Einhorn D, Fonseca V, Fulcher GR, Gaciong Z, Gaztambide S, Giles T, Horton E, Ilkova H, Jenssen T, Kahn SE, Krum H, Laakso M, Leiter LA, Levitt NS, Mareev V, Martinez F, Masson C, Mazzone T, Meaney E, Nesto R, Pan C, Prager R, Raptis SA, Rutten GE, Sandstroem H, Schaper F, Scheen A, Schmitz O, Sinay I, Soska V, Stender S, Tamas G, Tognoni G, Tuomilehto J, Villamil AS, Vozar J, Califf RM. Effect of nateglinide on the incidence of diabetes and cardiovascular events. N Engl J Med 2010; 362:1463-1476
  53. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefebvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L, Laakso M, Mokan M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schernthaner G, Schmitz O, Skrha J, Smith U, Taton J. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet 2005; 366:1279-1289
  54. Kernan WN, Viscoli CM, Furie KL, Young LH, Inzucchi SE, Gorman M, Guarino PD, Lovejoy AM, Peduzzi PN, Conwit R, Brass LM, Schwartz GG, Adams HP, Jr., Berger L, Carolei A, Clark W, Coull B, Ford GA, Kleindorfer D, O'Leary JR, Parsons MW, Ringleb P, Sen S, Spence JD, Tanne D, Wang D, Winder TR. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N Engl J Med 2016; 374:1321-1331
  55. Vaccaro O, Masulli M, Nicolucci A, Bonora E, Del Prato S, Maggioni AP, Rivellese AA, Squatrito S, Giorda CB, Sesti G, Mocarelli P, Lucisano G, Sacco M, Signorini S, Cappellini F, Perriello G, Babini AC, Lapolla A, Gregori G, Giordano C, Corsi L, Buzzetti R, Clemente G, Di Cianni G, Iannarelli R, Cordera R, La Macchia O, Zamboni C, Scaranna C, Boemi M, Iovine C, Lauro D, Leotta S, Dall'Aglio E, Cannarsa E, Tonutti L, Pugliese G, Bossi AC, Anichini R, Dotta F, Di Benedetto A, Citro G, Antenucci D, Ricci L, Giorgino F, Santini C, Gnasso A, De Cosmo S, Zavaroni D, Vedovato M, Consoli A, Calabrese M, di Bartolo P, Fornengo P, Riccardi G. Effects on the incidence of cardiovascular events of the addition of pioglitazone versus sulfonylureas in patients with type 2 diabetes inadequately controlled with metformin (TOSCA.IT): a randomised, multicentre trial. Lancet Diabetes Endocrinol2017; 5:887-897
  56. Vaccaro O, Lucisano G, Masulli M, Bonora E, Del Prato S, Rivellese AA, Giorda CB, Mocarelli P, Squatrito S, Maggioni AP, Riccardi G, Nicolucci A. Cardiovascular Effects of Pioglitazone or Sulfonylureas According to Pretreatment Risk: Moving Toward Personalized Care. J Clin Endocrinol Metab 2019; 104:3296-3302
  57. Mazzone T, Meyer PM, Feinstein SB, Davidson MH, Kondos GT, D'Agostino RB, Sr., Perez A, Provost JC, Haffner SM. Effect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes: a randomized trial. JAMA 2006; 296:2572-2581
  58. Pfutzner A, Marx N, Lubben G, Langenfeld M, Walcher D, Konrad T, Forst T. Improvement of cardiovascular risk markers by pioglitazone is independent from glycemic control: results from the pioneer study. J Am Coll Cardiol2005; 45:1925-1931
  59. Nissen SE, Nicholls SJ, Wolski K, Nesto R, Kupfer S, Perez A, Jure H, De Larochelliere R, Staniloae CS, Mavromatis K, Saw J, Hu B, Lincoff AM, Tuzcu EM. Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial. JAMA2008; 299:1561-1573
  60. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 2007; 356:2457-2471
  61. Singh S, Loke YK, Furberg CD. Long-term risk of cardiovascular events with rosiglitazone: a meta-analysis. JAMA 2007; 298:1189-1195
  62. Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet 2009; 373:2125-2135
  63. Home PD, Pocock SJ, Beck-Nielsen H, Gomis R, Hanefeld M, Jones NP, Komajda M, McMurray JJ. Rosiglitazone evaluated for cardiovascular outcomes--an interim analysis. N Engl J Med 2007; 357:28-38
  64. Mahaffey KW, Hafley G, Dickerson S, Burns S, Tourt-Uhlig S, White J, Newby LK, Komajda M, McMurray J, Bigelow R, Home PD, Lopes RD. Results of a reevaluation of cardiovascular outcomes in the RECORD trial. Am Heart J 2013; 166:240-249 e241
  65. Bach RG, Brooks MM, Lombardero M, Genuth S, Donner TW, Garber A, Kennedy L, Monrad ES, Pop-Busui R, Kelsey SF, Frye RL. Rosiglitazone and outcomes for patients with diabetes mellitus and coronary artery disease in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial. Circulation 2013; 128:785-794
  66. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for Type 2 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)2022.
  67. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, Ohman P, Frederich R, Wiviott SD, Hoffman EB, Cavender MA, Udell JA, Desai NR, Mosenzon O, McGuire DK, Ray KK, Leiter LA, Raz I. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 2013; 369:1317-1326
  68. Scirica BM, Braunwald E, Raz I, Cavender MA, Morrow DA, Jarolim P, Udell JA, Mosenzon O, Im K, Umez-Eronini AA, Pollack PS, Hirshberg B, Frederich R, Lewis BS, McGuire DK, Davidson J, Steg PG, Bhatt DL. Heart failure, saxagliptin, and diabetes mellitus: observations from the SAVOR-TIMI 53 randomized trial. Circulation2014; 130:1579-1588
  69. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Cushman WC, Zannad F. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med 2013; 369:1327-1335
  70. Zannad F, Cannon CP, Cushman WC, Bakris GL, Menon V, Perez AT, Fleck PR, Mehta CR, Kupfer S, Wilson C, Lam H, White WB. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet 2015; 385:2067-2076
  71. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, Josse R, Kaufman KD, Koglin J, Korn S, Lachin JM, McGuire DK, Pencina MJ, Standl E, Stein PP, Suryawanshi S, Van de Werf F, Peterson ED, Holman RR. Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2015; 373:232-242
  72. Rosenstock J, Perkovic V, Johansen OE, Cooper ME, Kahn SE, Marx N, Alexander JH, Pencina M, Toto RD, Wanner C, Zinman B, Woerle HJ, Baanstra D, Pfarr E, Schnaidt S, Meinicke T, George JT, von Eynatten M, McGuire DK. Effect of Linagliptin vs Placebo on Major Cardiovascular Events in Adults With Type 2 Diabetes and High Cardiovascular and Renal Risk: The CARMELINA Randomized Clinical Trial. JAMA 2018;
  73. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med 2015; 373:2117-2128
  74. Fitchett D, Inzucchi SE, Cannon CP, McGuire DK, Scirica BM, Johansen OE, Sambevski S, Kaspers S, Pfarr E, George JT, Zinman B. Empagliflozin Reduced Mortality and Hospitalization for Heart Failure Across the Spectrum of Cardiovascular Risk in the EMPA-REG OUTCOME Trial. Circulation 2019; 139:1384-1395
  75. Fitchett D, Butler J, van de Borne P, Zinman B, Lachin JM, Wanner C, Woerle HJ, Hantel S, George JT, Johansen OE, Inzucchi SE. Effects of empagliflozin on risk for cardiovascular death and heart failure hospitalization across the spectrum of heart failure risk in the EMPA-REG OUTCOME(R) trial. Eur Heart J 2018; 39:363-370
  76. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med 2017; 377:644-657
  77. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, de Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med 2019; 380:2295-2306
  78. Jardine MJ, Zhou Z, Mahaffey KW, Oshima M, Agarwal R, Bakris G, Bajaj HS, Bull S, Cannon CP, Charytan DM, de Zeeuw D, Di Tanna GL, Greene T, Heerspink HJL, Levin A, Neal B, Pollock C, Qiu R, Sun T, Wheeler DC, Zhang H, Zinman B, Rosenthal N, Perkovic V. Renal, Cardiovascular, and Safety Outcomes of Canagliflozin by Baseline Kidney Function: A Secondary Analysis of the CREDENCE Randomized Trial. J Am Soc Nephrol 2020; 31:1128-1139
  79. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med2018;
  80. Furtado RHM, Bonaca MP, Raz I, Zelniker TA, Mosenzon O, Cahn A, Kuder J, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Nicolau JC, Gause-Nilsson IAM, Fredriksson M, Langkilde AM, Sabatine MS, Wiviott SD. Dapagliflozin and Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus and Previous Myocardial Infarction. Circulation 2019; 139:2516-2527
  81. Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B, Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N Engl J Med 2020; 383:1425-1435
  82. Cosentino F, Cannon CP, Cherney DZI, Masiukiewicz U, Pratley R, Dagogo-Jack S, Frederich R, Charbonnel B, Mancuso J, Shih WJ, Terra SG, Cater NB, Gantz I, McGuire DK. Efficacy of Ertugliflozin on Heart Failure-Related Events in Patients With Type 2 Diabetes Mellitus and Established Atherosclerotic Cardiovascular Disease: Results of the VERTIS CV Trial. Circulation 2020; 142:2205-2215
  83. Feingold KR. Oral and Injectable (Non-Insulin) Pharmacological Agents for the Treatment of Type 2 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)2022.
  84. Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Kober LV, Lawson FC, Ping L, Wei X, Lewis EF, Maggioni AP, McMurray JJ, Probstfield JL, Riddle MC, Solomon SD, Tardif JC. Lixisenatide in Patients with Type 2 Diabetes and Acute Coronary Syndrome. N Engl J Med 2015; 373:2247-2257
  85. 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. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2016; 375:311-322
  86. Mann JFE, Fonseca V, Mosenzon O, Raz I, Goldman B, Idorn T, von Scholten BJ, Poulter NR. Effects of Liraglutide Versus Placebo on Cardiovascular Events in Patients With Type 2 Diabetes Mellitus and Chronic Kidney Disease. Circulation 2018; 138:2908-2918
  87. Verma S, Poulter NR, Bhatt DL, Bain SC, Buse JB, Leiter LA, Nauck MA, Pratley RE, Zinman B, Orsted DD, Monk Fries T, Rasmussen S, Marso SP. Effects of Liraglutide on Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus With or Without History of Myocardial Infarction or Stroke. Circulation 2018; 138:2884-2894
  88. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsboll T. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2016;
  89. Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, Jeppesen OK, Lingvay I, Mosenzon O, Pedersen SD, Tack CJ, Thomsen M, Vilsboll T, Warren ML, Bain SC. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med 2019; 381:841-851
  90. Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB, Chan JC, Choi J, Gustavson SM, Iqbal N, Maggioni AP, Marso SP, Ohman P, Pagidipati NJ, Poulter N, Ramachandran A, Zinman B, Hernandez AF. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med 2017; 377:1228-1239
  91. Hernandez AF, Green JB, Janmohamed S, D'Agostino RB, Sr., Granger CB, Jones NP, Leiter LA, Rosenberg AE, Sigmon KN, Somerville MC, Thorpe KM, McMurray JJV, Del Prato S. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial. Lancet 2018; 392:1519-1529
  92. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Riesmeyer JS, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019; 394:121-130
  93. Gerstein HC, Hart R, Colhoun HM, Diaz R, Lakshmanan M, Botros FT, Probstfield J, Riddle MC, Ryden L, Atisso CM, Dyal L, Hall S, Avezum A, Basile J, Conget I, Cushman WC, Hancu N, Hanefeld M, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Munoz EGC, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T. The effect of dulaglutide on stroke: an exploratory analysis of the REWIND trial. Lancet Diabetes Endocrinol 2020; 8:106-114
  94. Kristensen SL, Rorth R, Jhund PS, Docherty KF, Sattar N, Preiss D, Kober L, Petrie MC, McMurray JJV. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol 2019; 7:776-785
  95. Sposito AC, Berwanger O, de Carvalho LSF, Saraiva JFK. GLP-1RAs in type 2 diabetes: mechanisms that underlie cardiovascular effects and overview of cardiovascular outcome data. Cardiovasc Diabetol 2018; 17:157
  96. Sattar N, McGuire DK, Pavo I, Weerakkody GJ, Nishiyama H, Wiese RJ, Zoungas S. Tirzepatide cardiovascular event risk assessment: a pre-specified meta-analysis. Nat Med 2022; 28:591-598
  97. Chiasson JL, Josse RG, Gomis R, Hanefeld M, Karasik A, Laakso M. Acarbose treatment and the risk of cardiovascular disease and hypertension in patients with impaired glucose tolerance: the STOP-NIDDM trial. JAMA 2003; 290:486-494
  98. Yun P, Du AM, Chen XJ, Liu JC, Xiao H. Effect of Acarbose on Long-Term Prognosis in Acute Coronary Syndromes Patients with Newly Diagnosed Impaired Glucose Tolerance. J Diabetes Res 2016; 2016:1602083
  99. Holman RR, Coleman RL, Chan JCN, Chiasson JL, Feng H, Ge J, Gerstein HC, Gray R, Huo Y, Lang Z, McMurray JJ, Ryden L, Schroder S, Sun Y, Theodorakis MJ, Tendera M, Tucker L, Tuomilehto J, Wei Y, Yang W, Wang D, Hu D, Pan C. Effects of acarbose on cardiovascular and diabetes outcomes in patients with coronary heart disease and impaired glucose tolerance (ACE): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2017; 5:877-886
  100. Gaziano JM, Cincotta AH, O'Connor CM, Ezrokhi M, Rutty D, Ma ZJ, Scranton RE. Randomized clinical trial of quick-release bromocriptine among patients with type 2 diabetes on overall safety and cardiovascular outcomes. Diabetes Care 2010; 33:1503-1508
  101. Younk LM, Davis SN. Evaluation of colesevelam hydrochloride for the treatment of type 2 diabetes. Expert Opin Drug Metab Toxicol 2012; 8:515-525
  102. The Lipid Research Clinics Coronary Primary Prevention Trial results. I. Reduction in incidence of coronary heart disease. JAMA 1984; 251:351-364
  103. 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
  104. Investigators OT, Gerstein HC, Bosch J, Dagenais GR, Diaz R, Jung H, Maggioni AP, Pogue J, Probstfield J, Ramachandran A, Riddle MC, Ryden LE, Yusuf S. Basal insulin and cardiovascular and other outcomes in dysglycemia. N Engl J Med 2012; 367:319-328
  105. Origin Trial Investigators. Cardiovascular and Other Outcomes Postintervention With Insulin Glargine and Omega-3 Fatty Acids (ORIGINALE). Diabetes Care 2016; 39:709-716
  106. Marso SP, McGuire DK, Zinman B, Poulter NR, Emerson SS, Pieber TR, Pratley RE, Haahr PM, Lange M, Brown-Frandsen K, Moses A, Skibsted S, Kvist K, Buse JB. Efficacy and Safety of Degludec versus Glargine in Type 2 Diabetes. N Engl J Med 2017; 377:723-732
  107. Chaitman BR, Hardison RM, Adler D, Gebhart S, Grogan M, Ocampo S, Sopko G, Ramires JA, Schneider D, Frye RL. The Bypass Angioplasty Revascularization Investigation 2 Diabetes randomized trial of different treatment strategies in type 2 diabetes mellitus with stable ischemic heart disease: impact of treatment strategy on cardiac mortality and myocardial infarction. Circulation 2009; 120:2529-2540
  108. Bari D. Study Group, Frye RL, August P, Brooks MM, Hardison RM, Kelsey SF, MacGregor JM, Orchard TJ, Chaitman BR, Genuth SM, Goldberg SH, Hlatky MA, Jones TL, Molitch ME, Nesto RW, Sako EY, Sobel BE. A randomized trial of therapies for type 2 diabetes and coronary artery disease. N Engl J Med 2009; 360:2503-2515
  109. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S140-S157
  110. de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Circulation 2014; 130:1110-1130
  111. Martin-Timon I, Sevillano-Collantes C, Segura-Galindo A, Del Canizo-Gomez FJ. Type 2 diabetes and cardiovascular disease: Have all risk factors the same strength? World J Diabetes 2014; 5:444-470
  112. Turner RC, Millns H, Neil HA, Stratton IM, Manley SE, Matthews DR, Holman RR. Risk factors for coronary artery disease in non-insulin dependent diabetes mellitus: United Kingdom Prospective Diabetes Study (UKPDS: 23). BMJ 1998; 316:823-828
  113. Hovingh GK, Rader DJ, Hegele RA. HDL re-examined. Curr Opin Lipidol 2015; 26:127-132
  114. Brunzell JD, Davidson M, Furberg CD, Goldberg RB, Howard BV, Stein JH, Witztum JL. Lipoprotein management in patients with cardiometabolic risk: consensus statement from the American Diabetes Association and the American College of Cardiology Foundation. Diabetes Care 2008; 31:811-822
  115. 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
  116. Nordestgaard BG. Triglyceride-Rich Lipoproteins and Atherosclerotic Cardiovascular Disease: New Insights From Epidemiology, Genetics, and Biology. Circ Res 2016; 118:547-563
  117. Ganjali S, Dallinga-Thie GM, Simental-Mendia LE, Banach M, Pirro M, Sahebkar A. HDL functionality in type 1 diabetes. Atherosclerosis 2017; 267:99-109
  118. Ginsberg HN, MacCallum PR. The obesity, metabolic syndrome, and type 2 diabetes mellitus pandemic: Part I. Increased cardiovascular disease risk and the importance of atherogenic dyslipidemia in persons with the metabolic syndrome and type 2 diabetes mellitus. J Cardiometab Syndr 2009; 4:113-119
  119. Goldberg IJ. Clinical review 124: Diabetic dyslipidemia: causes and consequences. J Clin Endocrinol Metab2001; 86:965-971
  120. Krauss RM. Lipids and lipoproteins in patients with type 2 diabetes. Diabetes Care 2004; 27:1496-1504
  121. Wu L, Parhofer KG. Diabetic dyslipidemia. Metabolism 2014; 63:1469-1479
  122. Taskinen MR, Boren J. New insights into the pathophysiology of dyslipidemia in type 2 diabetes. Atherosclerosis2015; 239:483-495
  123. Feingold KR, Grunfeld C, Pang M, Doerrler W, Krauss RM. LDL subclass phenotypes and triglyceride metabolism in non-insulin-dependent diabetes. Arterioscler Thromb 1992; 12:1496-1502
  124. Feingold KR. Obesity and Dyslipidemia. 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)2023.
  125. Morgantini C, Natali A, Boldrini B, Imaizumi S, Navab M, Fogelman AM, Ferrannini E, Reddy ST. Anti-inflammatory and antioxidant properties of HDLs are impaired in type 2 diabetes. Diabetes 2011; 60:2617-2623
  126. Apro J, Tietge UJ, Dikkers A, Parini P, Angelin B, Rudling M. Impaired Cholesterol Efflux Capacity of High-Density Lipoprotein Isolated From Interstitial Fluid in Type 2 Diabetes Mellitus-Brief Report. Arterioscler Thromb Vasc Biol 2016; 36:787-791
  127. Manjunatha S, Distelmaier K, Dasari S, Carter RE, Kudva YC, Nair KS. Functional and proteomic alterations of plasma high density lipoproteins in type 1 diabetes mellitus. Metabolism 2016; 65:1421-1431
  128. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res 2016; 57:1111-1125
  129. Durrington PN, Schofield JD, Siahmansur T, Soran H. Lipoprotein (a): gene genie. Curr Opin Lipidol 2014; 25:289-296
  130. Gudbjartsson DF, Thorgeirsson G, Sulem P, Helgadottir A, Gylfason A, Saemundsdottir J, Bjornsson E, Norddahl GL, Jonasdottir A, Jonasdottir A, Eggertsson HP, Gretarsdottir S, Thorleifsson G, Indridason OS, Palsson R, Jonasson F, Jonsdottir I, Eyjolfsson GI, Sigurdardottir O, Olafsson I, Danielsen R, Matthiasson SE, Kristmundsdottir S, Halldorsson BV, Hreidarsson AB, Valdimarsson EM, Gudnason T, Benediktsson R, Steinthorsdottir V, Thorsteinsdottir U, Holm H, Stefansson K. Lipoprotein(a) Concentration and Risks of Cardiovascular Disease and Diabetes. J Am Coll Cardiol 2019; 74:2982-2994
  131. Ferrannini E, DeFronzo RA. Impact of glucose-lowering drugs on cardiovascular disease in type 2 diabetes. Eur Heart J 2015; 36:2288-2296
  132. Wulffele MG, Kooy A, de Zeeuw D, Stehouwer CD, Gansevoort RT. The effect of metformin on blood pressure, plasma cholesterol and triglycerides in type 2 diabetes mellitus: a systematic review. J Intern Med 2004; 256:1-14
  133. Ratner R, Goldberg R, Haffner S, Marcovina S, Orchard T, Fowler S, Temprosa M, Diabetes Prevention Program Research Group. Impact of intensive lifestyle and metformin therapy on cardiovascular disease risk factors in the diabetes prevention program. Diabetes Care 2005; 28:888-894
  134. Spanheimer R, Betteridge DJ, Tan MH, Ferrannini E, Charbonnel B. Long-term lipid effects of pioglitazone by baseline anti-hyperglycemia medication therapy and statin use from the PROactive experience (PROactive 14). Am J Cardiol 2009; 104:234-239
  135. Deeg MA, Buse JB, Goldberg RB, Kendall DM, Zagar AJ, Jacober SJ, Khan MA, Perez AT, Tan MH. Pioglitazone and rosiglitazone have different effects on serum lipoprotein particle concentrations and sizes in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2007; 30:2458-2464
  136. Goldberg RB, Kendall DM, Deeg MA, Buse JB, Zagar AJ, Pinaire JA, Tan MH, Khan MA, Perez AT, Jacober SJ. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diabetes Care 2005; 28:1547-1554
  137. Sanchez-Garcia A, Simental-Mendia M, Millan-Alanis JM, Simental-Mendia LE. Effect of sodium-glucose co-transporter 2 inhibitors on lipid profile: A systematic review and meta-analysis of 48 randomized controlled trials. Pharmacol Res 2020; 160:105068
  138. Lamos EM, Levitt DL, Munir KM. A review of dopamine agonist therapy in type 2 diabetes and effects on cardio-metabolic parameters. Prim Care Diabetes 2016; 10:60-65
  139. Holt RI, Barnett AH, Bailey CJ. Bromocriptine: old drug, new formulation and new indication. Diabetes Obes Metab 2010; 12:1048-1057
  140. Raskin P, Cincotta AH. Bromocriptine-QR therapy for the management of type 2 diabetes mellitus: developmental basis and therapeutic profile summary. Expert Rev Endocrinol Metab 2016; 11:113-148
  141. Feingold KR. Cholesterol Lowering Drugs. 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.
  142. Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M, Drucker DJ. Cardiovascular Actions and Clinical Outcomes With Glucagon-Like Peptide-1 Receptor Agonists and Dipeptidyl Peptidase-4 Inhibitors. Circulation 2017; 136:849-870
  143. Kanbay M, Copur S, Siriopol D, Yildiz AB, Gaipov A, van Raalte DH, Tuttle KR. Effect of tirzepatide on blood pressure and lipids: A meta-analysis of randomized controlled trials. Diabetes Obes Metab 2023;
  144. Ginsberg HN. Diabetic dyslipidemia: basic mechanisms underlying the common hypertriglyceridemia and low HDL cholesterol levels. Diabetes 1996; 45 Suppl 3:S27-30
  145. Ginsberg HN, Zhang YL, Hernandez-Ono A. Metabolic syndrome: focus on dyslipidemia. Obesity (Silver Spring)2006; 14 Suppl 1:41S-49S
  146. Klop B, Elte JW, Cabezas MC. Dyslipidemia in obesity: mechanisms and potential targets. Nutrients 2013; 5:1218-1240
  147. Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med 2014; 371:32-41
  148. Tg,Hdl Working Group of the Exome Sequencing Project, National Heart LungBlood, Institute, Crosby J, Peloso GM, Auer PL, Crosslin DR, Stitziel NO, Lange LA, Lu Y, Tang ZZ, Zhang H, Hindy G, Masca N, Stirrups K, Kanoni S, Do R, Jun G, Hu Y, Kang HM, Xue C, Goel A, Farrall M, Duga S, Merlini PA, Asselta R, Girelli D, Olivieri O, Martinelli N, Yin W, Reilly D, Speliotes E, Fox CS, Hveem K, Holmen OL, Nikpay M, Farlow DN, Assimes TL, Franceschini N, Robinson J, North KE, Martin LW, DePristo M, Gupta N, Escher SA, Jansson JH, Van Zuydam N, Palmer CN, Wareham N, Koch W, Meitinger T, Peters A, Lieb W, Erbel R, Konig IR, Kruppa J, Degenhardt F, Gottesman O, Bottinger EP, O'Donnell CJ, Psaty BM, Ballantyne CM, Abecasis G, Ordovas JM, Melander O, Watkins H, Orho-Melander M, Ardissino D, Loos RJ, McPherson R, Willer CJ, Erdmann J, Hall AS, Samani NJ, Deloukas P, Schunkert H, Wilson JG, Kooperberg C, Rich SS, Tracy RP, Lin DY, Altshuler D, Gabriel S, Nickerson DA, Jarvik GP, Cupples LA, Reiner AP, Boerwinkle E, Kathiresan S. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med 2014; 371:22-31
  149. Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, Geary RS, Baker BF, Graham MJ, Crooke RM, Witztum JL. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med 2014; 371:2200-2206
  150. Arca M, D'Erasmo L, Minicocci I. Familial combined hypolipidemia: angiopoietin-like protein-3 deficiency. Curr Opin Lipidol 2020; 31:41-48
  151. Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, Grunfeld C. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res2004; 45:1169-1196
  152. Lara-Castro C, Fu Y, Chung BH, Garvey WT. Adiponectin and the metabolic syndrome: mechanisms mediating risk for metabolic and cardiovascular disease. Curr Opin Lipidol 2007; 18:263-270
  153. Feingold KR, Grunfeld C. The acute phase response inhibits reverse cholesterol transport. J Lipid Res 2010; 51:682-684
  154. Feingold KR, Grunfeld C. Effect of inflammation on HDL structure and function. Curr Opin Lipidol 2022; 27:521-530
  155. Lu B, Moser A, Shigenaga JK, Grunfeld C, Feingold KR. The acute phase response stimulates the expression of angiopoietin like protein 4. Biochem Biophys Res Commun 2010; 391:1737-1741
  156. Feingold KR, Grunfeld C. The Effect of Inflammation and Infection on Lipids and Lipoproteins. 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.
  157. Rosenblat M, Karry R, Aviram M. Paraoxonase 1 (PON1) is a more potent antioxidant and stimulant of macrophage cholesterol efflux, when present in HDL than in lipoprotein-deficient serum: relevance to diabetes. Atherosclerosis 2006; 187:74-81
  158. Ahima RS. Adipose tissue as an endocrine organ. Obesity (Silver Spring) 2006; 14 Suppl 5:242S-249S
  159. Christou GA, Kiortsis DN. Adiponectin and lipoprotein metabolism. Obes Rev 2013; 14:939-949
  160. Rashid S, Kastelein JJ. PCSK9 and resistin at the crossroads of the atherogenic dyslipidemia. Expert Rev Cardiovasc Ther 2013; 11:1567-1577
  161. Yu YH, Ginsberg HN. Adipocyte signaling and lipid homeostasis: sequelae of insulin-resistant adipose tissue. Circ Res 2005; 96:1042-1052
  162. Cholesterol Treatment Trialists, Collaborato, 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
  163. Collins R, Armitage J, Parish S, Sleigh P, Peto R, Heart Protection Study Collaborative Group. 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
  164. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002; 360:7-22
  165. Wanner C, Krane V, Marz W, Olschewski M, Mann JF, Ruf G, Ritz E,German, Diabetes

            Dialysis Study, Investigators. Atorvastatin in patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl J Med 2005; 353:238-248

  1. 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. Lancet2004; 364:685-696
  2. Nissen SE, Tuzcu EM, Schoenhagen P, Brown BG, Ganz P, Vogel RA, Crowe T, Howard G, Cooper CJ, Brodie B, Grines CL, DeMaria AN. Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: a randomized controlled trial. JAMA 2004; 291:1071-1080
  3. Nissen SE, Nicholls SJ, Sipahi I, Libby P, Raichlen JS, Ballantyne CM, Davignon J, Erbel R, Fruchart JC, Tardif JC, Schoenhagen P, Crowe T, Cain V, Wolski K, Goormastic M, Tuzcu EM. Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: the ASTEROID trial. JAMA 2006; 295:1556-1565
  4. Stegman B, Puri R, Cho L, Shao M, Ballantyne CM, Barter PJ, Chapman MJ, Erbel R, Libby P, Raichlen JS, Uno K, Kataoka Y, Nissen SE, Nicholls SJ. High-intensity statin therapy alters the natural history of diabetic coronary atherosclerosis: insights from SATURN. Diabetes Care 2014; 37:3114-3120
  5. Ahmed S, Cannon CP, Murphy SA, Braunwald E. Acute coronary syndromes and diabetes: Is intensive lipid lowering beneficial? Results of the PROVE IT-TIMI 22 trial. Eur Heart J 2006; 27:2323-2329
  6. Cannon CP, Braunwald E, McCabe CH, Rader DJ, Rouleau JL, Belder R, Joyal SV, Hill KA, Pfeffer MA, Skene AM, Pravastatin or Atorvastatin E, Infection Therapy-Thrombolysis in Myocardial Infarction I. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med 2004; 350:1495-1504
  7. LaRosa JC, Grundy SM, Waters DD, Shear C, Barter P, Fruchart JC, Gotto AM, Greten H, Kastelein JJ, Shepherd J, Wenger NK, Treating to New Targets I. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med 2005; 352:1425-1435
  8. Shepherd J, Barter P, Carmena R, Deedwania P, Fruchart JC, Haffner S, Hsia J, Breazna A, LaRosa J, Grundy S, Waters D. Effect of lowering LDL cholesterol substantially below currently recommended levels in patients with coronary heart disease and diabetes: the Treating to New Targets (TNT) study. Diabetes Care 2006; 29:1220-1226
  9. Pedersen TR, Faergeman O, Kastelein JJ, Olsson AG, Tikkanen MJ, Holme I, Larsen ML, Bendiksen FS, Lindahl C, Szarek M, Tsai J, Incremental Decrease in End Points Through Aggressive Lipid Lowering Study Group. High-dose atorvastatin vs usual-dose simvastatin for secondary prevention after myocardial infarction: the IDEAL study: a randomized controlled trial. JAMA 2005; 294:2437-2445
  10. Cholesterol Treatment Trialists Colloboration, 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
  11. Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, Forder P, Pillai A, Davis T, Glasziou P, Drury P, Kesaniemi YA, Sullivan D, Hunt D, Colman P, d'Emden M, Whiting M, Ehnholm C, Laakso M. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 2005; 366:1849-1861
  12. Jun M, Foote C, Lv J, Neal B, Patel A, Nicholls SJ, Grobbee DE, Cass A, Chalmers J, Perkovic V. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet 2010; 375:1875-1884
  13. Sacks FM, Carey VJ, Fruchart JC. Combination lipid therapy in type 2 diabetes. N Engl J Med 2010; 363:692-694; author reply 694-695
  14. Robins SJ, Collins D, Wittes JT, Papademetriou V, Deedwania PC, Schaefer EJ, McNamara JR, Kashyap ML, Hershman JM, Wexler LF, Rubins HB. Relation of gemfibrozil treatment and lipid levels with major coronary events: VA-HIT: a randomized controlled trial. JAMA 2001; 285:1585-1591
  15. Rubins HB, Robins SJ, Collins D, Nelson DB, Elam MB, Schaefer EJ, Faas FH, Anderson JW. Diabetes, plasma insulin, and cardiovascular disease: subgroup analysis from the Department of Veterans Affairs high-density lipoprotein intervention trial (VA-HIT). Arch Intern Med 2002; 162:2597-2604
  16. Koskinen P, Manttari M, Manninen V, Huttunen JK, Heinonen OP, Frick MH. Coronary heart disease incidence in NIDDM patients in the Helsinki Heart Study. Diabetes Care 1992; 15:820-825
  17. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study, a randomised study. Lancet 2001; 357:905-910
  18. Elkeles RS, Diamond JR, Poulter C, Dhanjil S, Nicolaides AN, Mahmood S, Richmond W, Mather H, Sharp P, Feher MD. Cardiovascular outcomes in type 2 diabetes. A double-blind placebo-controlled study of bezafibrate: the St. Mary's, Ealing, Northwick Park Diabetes Cardiovascular Disease Prevention (SENDCAP) Study. Diabetes Care 1998; 21:641-648
  19. Clofibrate and niacin in coronary heart disease. JAMA 1975; 231:360-381
  20. Canner PL, Furberg CD, Terrin ML, McGovern ME. Benefits of niacin by glycemic status in patients with healed myocardial infarction (from the Coronary Drug Project). Am J Cardiol 2005; 95:254-257
  21. 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
  22. 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
  23. Nissen SE, Menon V, Nicholls SJ, Brennan D, Laffin L, Ridker P, Ray KK, Mason D, Kastelein JJP, Cho L, Libby P, Li N, Foody J, Louie MJ, Lincoff AM. Bempedoic Acid for Primary Prevention of Cardiovascular Events in Statin-Intolerant Patients. JAMA 2023; 330:131-140
  24. Group AS, 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
  25. ACCORD Study Group, Group AES, Chew EY, Ambrosius WT, Davis MD, Danis RP, Gangaputra S, Greven CM, Hubbard L, Esser BA, Lovato JF, Perdue LH, Goff DC, Jr., Cushman WC, Ginsberg HN, Elam MB, Genuth S, Gerstein HC, Schubart U, Fine LJ. Effects of medical therapies on retinopathy progression in type 2 diabetes. N Engl J Med 2010; 363:233-244
  26. Keech AC, Mitchell P, Summanen PA, O'Day J, Davis TM, Moffitt MS, Taskinen MR, Simes RJ, Tse D, Williamson E, Merrifield A, Laatikainen LT, d'Emden MC, Crimet DC, O'Connell RL, Colman PG. Effect of fenofibrate on the need for laser treatment for diabetic retinopathy (FIELD study): a randomised controlled trial. Lancet 2007; 370:1687-1697
  27. 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
  28. Investigators AIM-HIGH, 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
  29. Guyton JR, Slee AE, Anderson T, Fleg JL, Goldberg RB, Kashyap ML, Marcovina SM, Nash SD, O'Brien KD, Weintraub WS, Xu P, Zhao XQ, Boden WE. Relationship of lipoproteins to cardiovascular events: the AIM-HIGH Trial (Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides and Impact on Global Health Outcomes). J Am Coll Cardiol 2013; 62:1580-1584
  30. 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. The New England journal of medicine 2014; 371:203-212
  31. 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 RMI. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med 2015; 372:2387-2397
  32. Bohula EA, Morrow DA, Giugliano RP, Blazing MA, He P, Park JG, Murphy SA, White JA, Kesaniemi YA, Pedersen TR, Brady AJ, Mitchel Y, Cannon CP, Braunwald E. Atherothrombotic Risk Stratification and Ezetimibe for Secondary Prevention. J Am Coll Cardiol 2017; 69:911-921
  33. 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
  34. Lee YJ, Cho JY, You SC, Lee YH, Yun KH, Cho YH, Shin WY, Im SW, Kang WC, Park Y, Lee SY, Lee SJ, Hong SJ, Ahn CM, Kim BK, Ko YG, Choi D, Hong MK, Jang Y, Kim JS. Moderate-intensity statin with ezetimibe vs. high-intensity statin in patients with diabetes and atherosclerotic cardiovascular disease in the RACING trial. Eur Heart J 2023; 44:972-983
  35. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N Engl J Med 2017; 376:1713-1722
  36. 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;
  37. 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
  38. 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
  39. 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
  40. 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
  41. 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
  42. ORIGIN Trial Investigators OT, Bosch J, Gerstein HC, Dagenais GR, Diaz R, Dyal L, Jung H, Maggiono AP, Probstfield J, Ramachandran A, Riddle MC, Ryden LE, Yusuf S. n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N Engl J Med 2012; 367:309-318
  43. Ascend Study Collaborative Group, Bowman L, Mafham M, Wallendszus K, Stevens W, Buck G, Barton J, Murphy K, Aung T, Haynes R, Cox J, Murawska A, Young A, Lay M, Chen F, Sammons E, Waters E, Adler A, Bodansky J, Farmer A, McPherson R, Neil A, Simpson D, Peto R, Baigent C, Collins R, Parish S, Armitage J. Effects of n-3 Fatty Acid Supplements in Diabetes Mellitus. N Engl J Med 2018; 379:1540-1550
  44. Feingold KR. Triglyceride Lowering Drugs. 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.
  45. 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
  46. Saito Y, Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa 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 EPA on coronary artery disease in hypercholesterolemic patients with multiple risk factors: sub-analysis of primary prevention cases from the Japan EPA Lipid Intervention Study (JELIS). Atherosclerosis 2008; 200:135-140
  47. 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 2018;
  48. 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
  49. Nicholls SJ, Lincoff AM, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Pedersen TR, Ridker PM, Ray K, Karlson BW, Lundstrom T, Wolski K, Nissen SE. Assessment of omega-3 carboxylic acids in statin-treated patients with high levels of triglycerides and low levels of high-density lipoprotein cholesterol: Rationale and design of the STRENGTH trial. Clin Cardiol 2018; 41:1281-1288
  50. 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;
  51. 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
  52. Goff ZD, Nissen SE. N-3 polyunsaturated fatty acids for cardiovascular risk. Curr Opin Cardiol 2022; 37:356-363
  53. Mason RP, Sherratt SCR, Eckel RH. Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab 2023; 37:101681
  54. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Das SR, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Kosiborod M, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA, on behalf of the American Diabetes A. 10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes-2023. Diabetes Care 2023; 46:S158-S190
  55. 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
  56. Jellinger PS, Handelsman Y, Rosenblit PD, Bloomgarden ZT, Fonseca VA, Garber AJ, Grunberger G, Guerin CK, Bell DSH, Mechanick JI, Pessah-Pollack R, Wyne K, Smith D, Brinton EA, Fazio S, Davidson M. American Association of Clinical Endocrinologists and American College of Endocrinology Guidelines for Management of Dyslipidemia and Prevention of Cardiovascular Disease. Endocr Pract 2017; 23:1-87
  57. Handelsman Y, Jellinger PS, Guerin CK, Bloomgarden ZT, Brinton EA, Budoff MJ, Davidson MH, Einhorn D, Fazio S, Fonseca VA, Garber AJ, Grunberger G, Krauss RM, Mechanick JI, Rosenblit PD, Smith DA, Wyne KL. Consensus Statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the Management of Dyslipidemia and Prevention of Cardiovascular Disease Algorithm - 2020 Executive Summary. Endocr Pract 2020; 26:1196-1224
  58. 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
  59. Marx N, Federici M, Schutt K, Muller-Wieland D, Ajjan RA, Antunes MJ, Christodorescu RM, Crawford C, Di Angelantonio E, Eliasson B, Espinola-Klein C, Fauchier L, Halle M, Herrington WG, Kautzky-Willer A, Lambrinou E, Lesiak M, Lettino M, McGuire DK, Mullens W, Rocca B, Sattar N. 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes. Eur Heart J 2023;
  60. SCORE2-Diabetes Working Group. SCORE2-Diabetes: 10-year cardiovascular risk estimation in type 2 diabetes in Europe. Eur Heart J 2023; 44:2544-2556
  61. Feingold KR. Maximizing the benefits of cholesterol-lowering drugs. Curr Opin Lipidol 2019; 30:388-394
  62. Feingold KR, Chait A. Approach to patients with elevated low-density lipoprotein cholesterol levels. Best Pract Res Clin Endocrinol Metab 2023; 37:101658
  63. Evert AB, Dennison M, Gardner CD, Garvey WT, Lau KHK, MacLeod J, Mitri J, Pereira RF, Rawlings K, Robinson S, Saslow L, Uelmen S, Urbanski PB, Yancy WS, Jr. Nutrition Therapy for Adults With Diabetes or Prediabetes: A Consensus Report. Diabetes Care 2019; 42:731-754
  64. 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.
  65. Look ARG, Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Kitabchi AE, Knowler WC, Lewis CE, Maschak-Carey BJ, Montez MG, Murillo A, Nathan DM, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Regensteiner JG, Rickman AD, Ryan DH, Safford M, Wadden TA, Wagenknecht LE, West DS, Williamson DF, Yanovski SZ. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med 2013; 369:145-154
  66. 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 Med2013; 368:1279-1290
  67. 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
  68. 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
  69. 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
  70. 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
  71. 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
  72. Adams TD, Gress RE, Smith SC, Halverson RC, Simper SC, Rosamond WD, Lamonte MJ, Stroup AM, Hunt SC. Long-term mortality after gastric bypass surgery. N Engl J Med 2007; 357:753-761
  73. Romeo S, Maglio C, Burza MA, Pirazzi C, Sjoholm K, Jacobson P, Svensson PA, Peltonen M, Sjostrom L, Carlsson LM. Cardiovascular events after bariatric surgery in obese subjects with type 2 diabetes. Diabetes Care2012; 35:2613-2617
  74. Sjostrom L, Peltonen M, Jacobson P, Sjostrom CD, Karason K, Wedel H, Ahlin S, Anveden A, Bengtsson C, Bergmark G, Bouchard C, Carlsson B, Dahlgren S, Karlsson J, Lindroos AK, Lonroth H, Narbro K, Naslund I, Olbers T, Svensson PA, Carlsson LM. Bariatric surgery and long-term cardiovascular events. JAMA 2012; 307:56-65
  75. Sheng B, Truong K, Spitler H, Zhang L, Tong X, Chen L. The Long-Term Effects of Bariatric Surgery on Type 2 Diabetes Remission, Microvascular and Macrovascular Complications, and Mortality: a Systematic Review and Meta-Analysis. Obes Surg 2017; 27:2724-2732
  76. Fisher DP, Johnson E, Haneuse S, Arterburn D, Coleman KJ, O'Connor PJ, O'Brien R, Bogart A, Theis MK, Anau J, Schroeder EB, Sidney S. Association Between Bariatric Surgery and Macrovascular Disease Outcomes in Patients With Type 2 Diabetes and Severe Obesity. JAMA 2018; 320:1570-1582
  77. Farmer A, Montori V, Dinneen S, Clar C. Fish oil in people with type 2 diabetes mellitus. Cochrane Database Syst Rev 2001:CD003205
  78. Preiss D, Sattar N. Statins and the risk of new-onset diabetes: a review of recent evidence. Curr Opin Lipidol2011; 22:460-466
  79. 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
  80. 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
  81. Livingstone SJ, Looker HC, Akbar T, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Fuller JH, Colhoun HM. Effect of atorvastatin on glycaemia progression in patients with diabetes: an analysis from the Collaborative Atorvastatin in Diabetes Trial (CARDS). Diabetologia 2016; 59:299-306
  82. Chiu SW, Pratt CM, Feinn R, Chatterjee S. Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors and Ezetimibe on Risk of New-Onset Diabetes: A Systematic Review and Meta-Analysis of Large, Double-Blinded Randomized Controlled Trials. J Cardiovasc Pharmacol Ther 2020; 25:409-417
  83. Shah NP, McGuire DK, Cannon CP, Giugliano RP, Lokhnygina Y, Page CB, Tershakovec AM, Braunwald E, Blazing MA. Impact of Ezetimibe on New-Onset Diabetes: A Substudy of IMPROVE-IT. J Am Heart Assoc 2023; 12:e029593
  84. Kellick KA, Bottorff M, Toth PP, The National Lipid Association's Safety Task F. A clinician's guide to statin drug-drug interactions. J Clin Lipidol 2014; 8:S30-46
  85. Linz PE, Lovato LC, Byington RP, O'Connor PJ, Leiter LA, Weiss D, Force RW, Crouse JR, Ismail-Beigi F, Simmons DL, Papademetriou V, Ginsberg HN, Elam MB. Paradoxical reduction in HDL-C with fenofibrate and thiazolidinedione therapy in type 2 diabetes: the ACCORD Lipid Trial. Diabetes Care 2014; 37:686-693
  86. Ansquer JC, Foucher C, Rattier S, Taskinen MR, Steiner G. Fenofibrate reduces progression to microalbuminuria over 3 years in a placebo-controlled study in type 2 diabetes: results from the Diabetes Atherosclerosis Intervention Study (DAIS). Am J Kidney Dis 2005; 45:485-493
  87. Davis TM, Ting R, Best JD, Donoghoe MW, Drury PL, Sullivan DR, Jenkins AJ, O'Connell RL, Whiting MJ, Glasziou PP, Simes RJ, Kesaniemi YA, Gebski VJ, Scott RS, Keech AC, Event Lowering in Diabetes Study i. Effects of fenofibrate on renal function in patients with type 2 diabetes mellitus: the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study. Diabetologia 2011; 54:280-290
  88. Rajamani K, Colman PG, Li LP, Best JD, Voysey M, D'Emden MC, Laakso M, Baker JR, Keech AC. Effect of fenofibrate on amputation events in people with type 2 diabetes mellitus (FIELD study): a prespecified analysis of a randomised controlled trial. Lancet 2009; 373:1780-1788
  89. Aldridge MA, Ito MK. Colesevelam hydrochloride: a novel bile acid-binding resin. Ann Pharmacother 2001; 35:898-907
  90. Bays HE. Colesevelam hydrochloride added to background metformin therapy in patients with type 2 diabetes mellitus: a pooled analysis from 3 clinical studies. Endocr Pract 2011; 17:933-938
  91. Song WL, FitzGerald GA. Niacin, an old drug with a new twist. J Lipid Res 2013; 54:2586-2594
  92. Elam MB, Hunninghake DB, Davis KB, Garg R, Johnson C, Egan D, Kostis JB, Sheps DS, Brinton EA. Effect of niacin on lipid and lipoprotein levels and glycemic control in patients with diabetes and peripheral arterial disease: the ADMIT study: A randomized trial. Arterial Disease Multiple Intervention Trial. JAMA 2000; 284:1263-1270
  93. Grundy SM, Vega GL, McGovern ME, Tulloch BR, Kendall DM, Fitz-Patrick D, Ganda OP, Rosenson RS, Buse JB, Robertson DD, Sheehan JP, Diabetes Multicenter Research Group. Efficacy, safety, and tolerability of once-daily niacin for the treatment of dyslipidemia associated with type 2 diabetes: results of the assessment of diabetes control and evaluation of the efficacy of niaspan trial. Archives of internal medicine 2002; 162:1568-1576
  94. Weintraub H. Update on marine omega-3 fatty acids: management of dyslipidemia and current omega-3 treatment options. Atherosclerosis 2013; 230:381-389
  95. Khan SU, Rahman H, Okunrintemi V, Riaz H, Khan MS, Sattur S, Kaluski E, Lincoff AM, Martin SS, Blaha MJ. Association of Lowering Low-Density Lipoprotein Cholesterol With Contemporary Lipid-Lowering Therapies and Risk of Diabetes Mellitus: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2019; 8:e011581
  96. Carugo S, Sirtori CR, Corsini A, Tokgozoglu L, Ruscica M. PCSK9 Inhibition and Risk of Diabetes: Should We Worry? Curr Atheroscler Rep 2022; 24:995-1004
  97. Wang X, Zhang Y, Tan H, Wang P, Zha X, Chong W, Zhou L, Fang F. Efficacy and safety of bempedoic acid for prevention of cardiovascular events and diabetes: a systematic review and meta-analysis. Cardiovasc Diabetol2020; 19:128
  98. Bays HE, Banach M, Catapano AL, Duell PB, Gotto AM, Jr., Laufs U, Leiter LA, Mancini GBJ, Ray KK, Bloedon LT, Sasiela WJ, Ye Z, Ballantyne CM. Bempedoic acid safety analysis: Pooled data from four phase 3 clinical trials. J Clin Lipidol 2020; 14:649-659 e646
  99. Jenkins DJ, Jones PJ, Lamarche B, Kendall CW, Faulkner D, Cermakova L, Gigleux I, Ramprasath V, de Souza R, Ireland C, Patel D, Srichaikul K, Abdulnour S, Bashyam B, Collier C, Hoshizaki S, Josse RG, Leiter LA, Connelly PW, Frohlich J. Effect of a dietary portfolio of cholesterol-lowering foods given at 2 levels of intensity of dietary advice on serum lipids in hyperlipidemia: a randomized controlled trial. JAMA 2011; 306:831-839
  100. Feingold KR. Utility of Advanced Lipoprotein Testing in Clinical Practice. 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)2023.
  101. Orringer CE, Jacobson TA, Maki KC. National Lipid Association Scientific Statement on the use of icosapent ethyl in statin-treated patients with elevated triglycerides and high or very-high ASCVD risk. J Clin Lipidol 2019; 13:860-872

 

 

 

Idiopathic Short Stature and Growth Failure of Unknown Etiology

ABSTRACT

 

Idiopathic short stature (ISS) is defined by a height standard deviation score (SDS) ≤ -2.25 (≤1.2nd percentile) in pediatric patients for whom diagnostic evaluation has excluded other causes of short stature, and growth hormone levels are above 10 nanograms per milliliter in response to physiologic/pharmacologic stimulation. This definition, while comprehensive, is static and does not consider the importance of longitudinal follow up of growth, and consideration of evolving causes of growth hormone deficiency in these children. Using three illustrative cases, we highlight the importance of longitudinal evaluation of children with short stature to establish a growth pattern, identifying evolving causes of GH deficiency which may not be apparent on the initial work up, as well as exclusion of non-growth hormone related factors that affect growth including malabsorptive diseases, familial short stature, constitutional delay in puberty, Turner's syndrome, and SHOX Gene haploinsufficiency to accurately make a diagnosis of growth failure of unknown etiology (GFUE). 

 

INTRODUCTION 

 

In the 1980s, data on growth outcomes on patients who received growth hormone treatment were reported in an observational study (1); this included children who had a normal peak growth hormone response to stimulation, a group that was termed “constitutional short stature”.  The term, idiopathic short stature (ISS), was later adopted to describe such patients (2).   Guidelines for the use of growth hormone in children published by the Pediatric Endocrine Society defined ISS by a height standard deviation score (SDS) ≤-2.25 (≤1.2nd percentile) in pediatric patients for whom diagnostic evaluation excludes other causes of short stature, and with growth hormone levels above 10 nanograms per milliliter in response to physiologic/pharmacologic stimulation (3). The evaluation of children with short stature includes observation over a prolonged period to establish a growth pattern as well as the exclusion of non-growth hormone related factors that affect growth (4, 5).  To monitor children's growth patterns in the United States, the Center for Disease Control (CDC) chart utilizes percentiles, with the 3rd and 95th percentiles serving as the defined limits. Across the globe, the World Health Organization (WHO) charts are used to track a child's growth progression, and the limits  are set at 2.3rd and 97.7th percentiles (5). There are wide variations in normal growth patterns and an even greater variety of conditions that manifest with growth abnormalities.  Various factors contribute to the etiology of short stature, and this includes normal variants of growth such as familial short stature, constitutional delay in growth and puberty, as well as systemic conditions such as malabsorptive diseases, Turner's syndrome, and SHOX Gene haploinsufficiency, among others (Table 1).  Diagnostic tests at initial presentation by pediatric endocrinologists focus on elucidating the underlying cause of poor growth (Table 2).  Globally, malnutrition remains the main cause of poor growth (5).

 

In 2003, the Food and Drug Administration (FDA) approved the use of growth hormone (GH) for children with ISS. Two decades later, growth hormone therapy for idiopathic short stature remains controversial. ISS is not universally accepted as an indication for treatment with GH. This medication was approved for ISS in US, Canada, and Latin America, but not in the European Union or Japan (6). Along with other investigators, we have expressed concerns about the definition of ISS (7). The Pediatric Endocrine Society consensus statement did not specifically exclude patients with normal variants of growth such as constitutional growth delay and familial short stature (3). The definition focuses on stature, rather than growth and growth failure, therefore making GH treatment of such patients open to criticism for its use as only a height enhancer. In addition, as we have previously argued, a child whose height decelerates from the 75 to the 25th percentile without any known cause would also be of concern even though the height would not fit the ISS definition (7).  If no cause is found after clinical, appropriate biochemical and radiologic evaluation, such a patient may have what we would prefer to term idiopathic growth failure (IGF). In as much as those initials are already in use for Insulin Like Growth Factor, we prefer the term GFUE, or growth failure of unknown etiology, thereby emphasizing the concern about growth failure, not just stature.

 

To illustrate the potential dilemmas in diagnosis, we present three patients with similar patterns of growth and a normal response to initial growth hormone stimulation testing with 2 provocative agents; each of these children may have been called ISS but demonstrate radically different outcomes. 

Table 1. Common Causes of Short Stature and Growth Failure

Normal variants of growth

 
 

Familial short stature

 

Constitutional delay in growth and puberty (CDGP)

Systemic disorders

 

Endocrine

Growth hormone deficiency

 

Hypothyroidism

Cortisol excess (endogenous or exogenous)

Non-endocrine

Malnutrition

 

Malabsorptive diseases

 

Genetic syndromes: Turner's syndrome, Noonan’s syndrome, Achondroplasia, SHOX gene haploinsufficiency

 

Chronic inflammatory conditions (e.g.: Inflammatory bowel diseases)

 

Chronic medical conditions (e.g.: asthma on inhaled steroids)

 

Table 2. Diagnostic Tests at Initial Investigation of Idiopathic Short Stature and Growth Failure of Unknown Etiology

CBC, ESR

Urinalysis

Basic metabolic panel- BUN, creatinine, electrolytes

Celiac screen

Karyotype

TSH, Free T4

IGF1, IGFBP3

Bone age x ray

 

PATIENT 1

 

A boy aged 12 years and 5 months was evaluated for poor growth; both the father and paternal uncle were “late bloomers”. The height was at the 6th percentile, weight at 9th percentile, and Body Mass Index or BMI (weight in Kg divided by the square of height in meters) was at 32nd percentile, and his growth velocity was 3.8 cm per year. Clinical examination indicated Tanner stage 1 pubertal development with testes volume of 2-3mL bilaterally. Laboratory tests performed by the referring pediatrician before endocrine consultation showed normal thyroid function tests, sedimentation rate, C-Reactive protein, and no thyroid autoantibodies. A radiological bone age study confirmed delayed skeletal maturation, with a bone age of 10 years.  No additional tests were recommended at the Pediatric endocrinology clinic, and he was observed clinically.

 

At 14 years of age, the patient showed no signs of puberty. Figure 1 shows his longitudinal growth during follow up.  His height was the 2nd percentile, weight at the 6th percentile, BMI at the 31st percentile, and growth velocity remained at 3.8 cm per year. Delayed puberty in males is defined as the absence of testicular growth at an age that is 2 to 2.5 SD later than the reference population, which usually is around 14 years of age. (7) Laboratory results revealed low IGF-1 levels (-1.8 SDS for age and gender), while LH levels were close to pubertal levels, and testosterone levels remained prepubertal. A growth hormone (GH) stimulation test with the provocative agents, arginine and clonidine, was conducted due to the declining growth velocity and the low IGF-1 levels. Upon stimulation, peak GH level was 15 ng/ml.  At 14 years and 6 months old, the patient's height was at the1st percentile and growth velocity had decreased to 1.9 cm per year. The laboratory tests at that visit showed that IGF-1 levels had increased to 226 ng/mL and testosterone was now 45.7 ng/dL, consistent with Tanner stage 2 pubic hair, and 4 ml testicular volume noted on examination.  A bone age study confirmed delayed skeletal maturation, with a bone age of 10 years and 6 months. Since the patient had not progressed in puberty at the age of 14 years and 6 months, testosterone priming at a dose of 100 mg intramuscularly, once in 4 weeks for 3 months was initiated. At 15 years of age, his growth velocity had accelerated to 16.4 cm per year and his height was at the 5th percentile. Laboratory tests performed at 15.5 years showed that testosterone level was 301 ng/dL with a LH of 0.57 mIU/ml and IGF-1 levels had increased to 340 ng/ml. A bone age study showed the bone age to be consistent with 14 years and 6 months old. At 16 years and 11 months of age, the patient's height had reached the 36th percentile. Pubertal development had advanced with Tanner stage 3-4 for pubic hair development and testicular volume of 15mL bilaterally.

 

Figure 1. Patient 1 growth chart: Longitudinal growth chart in patient 1 before and after sex hormone priming.

 

DISCUSSION PATIENT 1

 

This patient’s height at presentation met the criteria for the diagnosis of Idiopathic Short Stature (ISS) since the patient's height was below -2.25 SD (8). However, with longitudinal follow-up, patient 1 ultimately proved to have constitutional growth delay (Figure 1). The diagnostic approach employed for this patient centered on monitoring. Once testosterone priming was employed, puberty progressed, and he reached his expected adult height. Through a targeted endocrine evaluation and longitudinal monitoring, the patient's condition was successfully managed without growth hormone treatment. This case underscores the importance of judicious use of testing.

 

The onset of puberty differs between boys and girls. In girls, 95% manifest at least one sign of puberty by 13 years of age whereas boys should begin signs of puberty by 14 years of age. In patients with delayed puberty and slow growth, a careful family history may be consistent with constitutional delay of growth and puberty. There often may be a history of a family member who also did not begin puberty until late into the teenage years, as in the case with the proband’s father and uncle. These patients can be difficult to diagnose because they may appear to be slowing in growth as they cross growth percentiles around the time of the anticipated pubertal growth spurt. Patients with constitutional growth delay fall behind their peers initially but have increased growth velocity later when puberty progresses and most reach adequate adult heights. Patients with constitutional growth delay have delayed bone ages and do not require treatment with growth hormone.

 

PATIENT 2

 

A boy presents at 12 years 9 months of age with growth failure and short stature. The patient's height had declined from 25th percentile to 10th percentile over 1.5 years, BMI was 40th percentile, growth velocity was 3.8 cm per year, and he was prepubertal. The parental target height was 176.7 cm (50th percentile). Laboratory tests showed that the testosterone level was 12 ng/dL, IGF-1 level was 125ng/mL (-2.06 SD), and bone age study was consistent with 12 years of age. Because of the low growth velocity and IGF1 level, the patient had a growth hormone (GH) stimulation test with provocative agents, arginine, and clonidine. The results showed a peak GH level of 16.9 ng/ml. At 13.5 years old, the patient’s height further declined to be at -2.25 SD and growth velocity was 2.84 cm per year.  Figure 2 shows patient 2’s growth trajectory during follow-up.  His testicular volume was 6mL bilaterally while tests showed IGF-1 to be114 ng/mL (-2.21 SD), thyroid function tests were normal, early morning cortisol level was 15.2 ug/dL, prolactin was 10 ng/ml, testosterone level 79 ng/dL, the luteinizing hormone was 3.2 mIU/mL, and the follicle-stimulating hormone was 2.1 mIU/mL, all consistent with pubertal progression. As puberty progressed, the growth velocity did not increase leading to a decline in height standard deviation.  In the setting of this growth deceleration despite pubertal progression, a second growth hormone stimulation test with arginine and L-dopa was performed. The GH stimulation test revealed a peak GH level of 5.4 ng/ml. This low peak GH level is consistent with growth hormone deficiency (GHD).  On pituitary magnetic resonance imaging, the pituitary gland appeared normal but small. The patient was treated with daily recombinant human growth hormone for 4.8 years. He reached an adult height at the 90th percentile (1.3 SDS), slightly above mid parental height (Figure 2).

 

Figure 2. Patient 2 growth chart: Longitudinal growth chart in patient 2 before and after growth hormone therapy. GH denotes growth hormone. MPTH denotes mid-parental target height.

 

DISCUSSION PATIENT 2

 

This patient's referral to a pediatric endocrinologist stemmed from decline in height, subpar growth velocity considering stage of puberty, and low levels of insulin-like growth factor 1 (IGF-1). Based on his normal response to the first growth hormone (GH) stimulation test, the patient’s diagnosis was consistent with Idiopathic Short Stature (ISS). With continued monitoring, it was apparent that further growth deceleration occurred despite advent and progression of puberty. With an additional growth hormone (GH) stimulation test, the patient was ultimately diagnosed to have evolving growth hormone deficiency. GHD can be congenital or acquired, developing over time. The mechanism of idiopathic GHD might be due to a functional, transient decrease in somatomedin secretion insufficient to maintain growth as puberty sets in. Isolated GHD can be  diagnosed using a combination of measuring growth factors, bone age X-rays, and growth hormone stimulation testing (9). Long-term monitoring as children progress into puberty is essential to uncover these instances of evolving GHD (9). It is imperative to emphasize the significance of prolonged follow-up and repeated testing to ensure the accurate diagnosis of GHD, rather than classifying the patient under the category of ISS or constitutional delay in growth and puberty (CDGP). In this case, physicians employed a meticulous approach of closely observing the decline in height and height velocity as puberty advanced, alongside screening tests such as IGF-1 measurement and multiple growth hormone stimulation tests. This comprehensive methodology allowed for the accurate diagnosis of GHD.

 

Despite the recognition of their many flaws, including poor reproducibility, non-physiologic assessment, and practical considerations, provocative tests remain part of the comprehensive evaluation of growth and are essential for the diagnosis of GHD (10).  For GH stimulation testing, two agents that provoke GH secretion from the pituitary (L-dopa, clonidine, arginine, glucagon) are administered following an overnight fast. These provocative agents are not physiological and do not replicate normal secretory dynamics. In this patient with short stature, the clinical picture of low growth velocity as puberty progressed plus low IGF-1 levels are indicative of late, or evolving GHD. 

 

PATIENT 3

 

A girl aged 13 years and 6 months was referred to a pediatric endocrinology clinic to evaluate her short stature. The patient had a medical history of ADHD diagnosed at the age of 7 years, and multiple surgeries for strabismus in both eyes. Her mid-parental height was at the 5th percentile. Axillary hair developed at 11 years of age and breast development started around 10-11 years. At age 13.5 years, the height was at the 1st percentile (-2.27 SD), weight was at <1 percentile, and BMI was at the 6th percentile. Tanner stage of breast development was 3 and pubic hair had been shaved. The patient had small hands and feet, bilateral fifth finger clinodactyly, brachydactyly, and a slightly increased carrying angle. The bone age was12.5 years old with shortening and broadening of the metacarpals which raised concerns for hypochondroplasia. Chromosomal analysis showed a normal female karyotype, 46XX. SHOX gene analysis did not reveal mutations or deletions. This patient had a normal response to growth hormone stimulation test with provocative agents, arginine, and clonidine; peak growth hormone level was 18 ng/ml. The patient was referred to a medical geneticist to undergo further genetic testing to explain her short stature. Repeated chromosome analysis counting 50 cells, had normal female karyotype of 46XX excluding mosaic Turner Syndrome. The microarray results found gain in a 1.5 Mb copy at 10p15.3. The Fragile X test was negative, it did not reveal FMR1 CGG repeat expansions. A genetic testing panel for skeletal dysplasia investigated twenty-nine genes associated with skeletal dysplasia and the results showed no mutations or deletions. Whole exome sequencing did not reveal any variant in disease genes possibly associated with a short stature phenotype. In this patient with height -3 SDS and low mid parental height, treatment with growth hormone resulted in modest gains in height SDS. Figure 3 shows this patient’s growth before and after growth hormone therapy. 

 

Figure 3. Patient 3 growth chart: Longitudinal growth chart in patient 2 before and after growth hormone therapy. GH denotes growth hormone.

 

DISCUSSION PATIENT 3

 

The cause of this patient’s short stature was not apparent despite extensive testing; thus, the diagnosis remains Growth Failure of Unknown Etiology, (GFUE) or Idiopathic short stature (ISS). After repeated genetic testing and thorough whole exome sequencing the etiology of the patient’s growth failure remained elusive. Case three presents the roadmap for a proper diagnosis of GFUE/ ISS using diagnoses of exclusion. The patient's height declined below >2.25 SD score below the mean height for a given age and sex, and through extensive genetic testing, there were no abnormalities found. Through a rigorous process of extensive evaluations and the systematic elimination of alternative causes for short stature, including malabsorptive diseases, familial short stature, constitutional delay in puberty, Turner syndrome, and SHOX Gene haploinsufficiency, the patient ultimately received a diagnosis of GFUE or idiopathic short stature.

 

ISS or GFUE may exist as a primary diagnosis alone or may be subcategorized to include ISS with familial short stature or constitutional delay of growth and puberty. Patients with ISS are often predicted to have an adult height below that expected based on mid parental height. It is thought that GH may increase the adult height of patients with ISS by 3.5 to 7.5 cm when compared with controls (11).  GH-treated children with ISS have also been reported to achieve height gain similar to patients with isolated GHD (6). Additional considerations for treatment in conjunction with GH include gonadotropin- releasing hormone (GnRH) analogs and aromatase inhibitors. Both classes of medications seek to decrease bone age maturation while allowing the child to continue to grow. These agents are still considered experimental when used for improving growth and more studies are needed (12, 13).  IGF-1 has received some attention recently as a potential treatment for ISS. Currently, IGF-1 is only approved for those patients with proven IGF-1 deficiency (IGF-1 levels –3 SD from the mean) with heights below –3 SD from the mean for age and normal stimulated GH levels. There is a lack of prospective studies that show clear benefit of IGF-1 for ISS. IGF-1 has also been associated with potential side effects, including hypoglycemia, headaches, lymphoid tissue hypertrophy, and coarsening of facial features (4).  Further studies must be performed before IGF-1 can be considered a treatment option for GFUE/ISS.

 

The concept that short stature alone is a problem that must be treated often leads to patients seeking treatment for cosmetic, rather than medical reasons to attain an adult height the patient or family feel is appropriate. It is important to stress that short stature itself is not necessarily a medical problem. However, a pattern of growth consistent with growth failure (decreasing growth velocity, growth velocity consistently <50 percentile) regardless of height percentile should be evaluated further. If an underlying cause of growth failure cannot be found, we propose that it is appropriate to use a term such as “idiopathic growth failure” or “growth failure of unknown etiology” (GFUE) rather than “idiopathic short stature.” These terms take the focus off stature as the overriding problem, placing it into an element that all can agree needs investigation and, if persistent, needs intervention for its reversal. Height predictions always, but especially in the face of subnormal growth rates, are likely to be inaccurate and should be provided with a great deal of caution (5).  We believe that familial short stature and constitutional delay of growth and puberty should be removed from the current overall category of “ISS” and placed under the category “normal variant.”

 

A child should be labeled as GFUE/ISS only after an in depth evaluation for other etiologies has been performed (7). As Next Generation Sequencing (NGS) and other diagnostic techniques become established, the number of patients with unidentified causes for growth failure is likely to diminish.

 

CONCLUSION

 

The three illustrative cases above help to show the significance of comprehensive testing, thorough evaluations, and rigorous longitudinal monitoring, to exclude currently known causes of short stature. Every effort should be made to avoid any bias in referral and evaluation of short stature based on gender, ethnic or racial differences (14).  With these guidelines, two of the three patients initially presented with features aligned with the diagnostic criteria for growth failure of unknown etiology (GFUE)/ISS. Through subsequent testing and prolonged monitoring, alternative underlying diagnoses were identified: constitutional growth delay, and acquired, idiopathic, isolated growth hormone deficiency. In the third case, we present the roadmap for a proper diagnosis of GFUE/ ISS that is based on specific phenotypic characteristics. The diagnosis of ISS/GFUE should only be assigned after an exhaustive exploration of all diagnostic possibilities, ensuring that patients are provided with precise diagnoses and subsequently, if indicated, appropriate medication-based treatments to achieve height gain.

 

REFERENCES

 

  1. Gertner, J.M., et al., Prospective clinical trial of human growth hormone in short children without growth hormone deficiency. J Pediatr, 1984. 104(2): p. 172-6.
  2. Hintz, R.L., Growth hormone treatment of idiopathic short stature. Horm Res, 1996. 46(4-5): p. 208-14.
  3. Wilson, T.A., et al., Update of guidelines for the use of growth hormone in children: the Lawson Wilkins Pediatric Endocrinology Society Drug and Therapeutics Committee. J Pediatr, 2003. 143(4): p. 415-21.
  4. Grimberg, A., et al., Guidelines for Growth Hormone and Insulin-Like Growth Factor-I Treatment in Children and Adolescents: Growth Hormone Deficiency, Idiopathic Short Stature, and Primary Insulin-Like Growth Factor-I Deficiency. Horm Res Paediatr, 2016. 86(6): p. 361-397.
  5. Graber, E. and R. Rapaport, Growth and growth disorders in children and adolescents. Pediatr Ann, 2012. 41(4): p. e1-9.
  6. Child, C.J., et al., Height Gain and Safety Outcomes in Growth Hormone-Treated Children with Idiopathic Short Stature: Experience from a Prospective Observational Study. Horm Res Paediatr, 2019. 91(4): p. 241-251.
  7. Rapaport, R., J.M. Wit, and M.O. Savage, Growth failure: 'idiopathic' only after a detailed diagnostic evaluation. Endocr Connect, 2021. 10(3): p. R125-R138.
  8. Cohen, P., et al., Consensus statement on the diagnosis and treatment of children with idiopathic short stature: a summary of the Growth Hormone Research Society, the Lawson Wilkins Pediatric Endocrine Society, and the European Society for Paediatric Endocrinology Workshop. J Clin Endocrinol Metab, 2008. 93(11): p. 4210-7.
  9. Samuels, J.G., Chimatapu, S. N., Savage, M. O., Rapaport, R, Late-Onset Isolated Growth Hormone Deficiency. JCEM Case Reports, 2023. 1(2).
  10. Yau, M. and R. Rapaport, Growth Hormone Stimulation Testing: To Test or Not to Test? That Is One of the Questions. Front Endocrinol (Lausanne), 2022. 13: p. 902364.
  11. Allen DB, Cuttler L. Clinical practice. Short stature in childhood--challenges and choices. N Engl J Med. 2013 Mar 28;368(13):1220-8. doi: 10.1056/NEJMcp1213178. PMID: 23534561; PMCID: PMC5754004.
  12. Mauras, N., et al., Randomized Trial of Aromatase Inhibitors, Growth Hormone, or Combination in Pubertal Boys with Idiopathic, Short Stature. J Clin Endocrinol Metab, 2016. 101(12): p. 4984-4993.
  13. Carel, J.C., Management of short stature with GnRH agonist and co-treatment with growth hormone: a controversial issue. Mol Cell Endocrinol, 2006. 254-255: p. 226-33.
  14. Beliard, K., et al., Identifying and addressing disparities in the evaluation and treatment of children with growth hormone deficiency. Front Endocrinol (Lausanne), 2022. 13: p. 989404.

Lipoprotein (a)

ABSTRACT

 

Lipoprotein(a) is an apo B-containing lipoprotein which has a unique plasminogen-like apolipoprotein(a) attached to apo B-100. The levels of lipoprotein(a) are under strong genetic regulation and vary by several hundred-fold in the general population. Although the exact function of lipoprotein(a) is still a mystery, recent studies have shown that lipoprotein(a) has a causal role in atherosclerosis and its level has been shown to be a risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valvular disease. Measurement of lipoprotein(a) level and comparison between different assays are challenging due to differences in reporting units and the absence of a reference method. Various guidelines recommend measurement of lipoprotein(a) levels in order to define cardiovascular risk. Lifestyle modifications have a minimal effect in reducing lipoprotein(a) levels. Currently available lipid-lowering therapies also result in only modest reductions in lipoprotein(a) levels. At present, there is no approved pharmacological treatment option for lowering lipoprotein(a) levels. More potent lipoprotein(a)-lowering medications are under active investigation to prove that lowering lipoprotein(a) level reduces ASCVD events. Lipoprotein(a), therefore, remains a viable and attractive therapeutic target in ASCVD risk reduction.   

 

HISTORY

 

Lipoprotein(a) was first described by Kare Berg in 1963 (1) and later purified and characterized by Ehnholm and colleagues as part of human lipoproteins (2). Subsequent studies linked lipoprotein(a) to coronary artery disease (CAD) (3,4) and the threshold of 30 mg/dL was introduced (5). In 1987, amino acid sequences of apolipoprotein(a) [apo(a)], a major protein of lipoprotein(a), were identified and the LPA gene, encoding apo(a), was cloned (6,7). The sequence showed homology to plasminogen, a fibrinolytic proenzyme, but the protease domain was inactive (6). Apo(a) size polymorphism and its underlying molecular genetics were discovered and the inverse correlation between apo(a) isoform and lipoprotein(a) levels were demonstrated (8). Early observational and prospective studies showed higher levels of lipoprotein(a) in patients with CAD and myocardial infarction (MI) (4,5,9-12), but others did not (13,14). Lipoprotein(a) was later identified as a main carrier of oxidized phospholipids in the plasma (15). Results from large population-based studies along with Mendelian randomization studies in the past 2 decades have shown that lipoprotein(a) is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD), especially MI and atherosclerotic stenosis (16-19). A surprise finding in 2013 also identified lipoprotein(a) as a key player in calcific aortic valvular disease (CAVD) (20). Currently, lipoprotein(a) is an active therapeutic target and results from ongoing phase 3 clinical studies will soon clarify the lipoprotein(a) hypothesis in ASCVD and CAVD.           

 

STRUCTURE AND ASSEMBLY

 

Lipoprotein(a) is an apoB-containing lipoprotein resembling low-density lipoprotein (LDL), but apolipoprotein B-100 is covalently linked to a unique glycoprotein, called apolipoprotein(a) or apo(a). Apo(a), encoded by the LPA gene, is structurally similar to plasminogen, an important protein involved in fibrinolysis. In vitro, apo(a) has been shown to inhibit fibrinolysis (21)

 

Apo(a) is present only in a subset of primates, including humans, Old World monkeys, and orangutans, but not in New World monkeys or other primates. The exception is the occurrence of an apo(a)-like protein in the hedgehog (22). A characteristic feature of apo(a) is the presence of loop-like structure stabilized by 3 internal disulfide bonds, called “kringles”. These kringle domains are triple-loop structures found in other coagulation factors, including plasminogen. While plasminogen has 5 kringle domains (KI, KII, KIII, KIV and KV) and one protease domain at the end, apo(a) has only KIV, KV and an inactive protease domain (Figure 1).

 

Figure 1. Structure of lipoprotein(a) (from www.familyheart.org).

 

In apo(a), the number of the fourth kringle domain is highly variable due to expansion and differentiation of KIV into 10 different types of KIV domains, called KIV1-KIV10. While KIV1 and KIV3-KIV10 are present as single copies, kringle IV type 2 (KIV2) are further expanded, resulting in multiallelic copy number variation (1 to >40 copies). This expansion of KIV2 leads to a size polymorphism of apo(a), ranging from 300-800 kDa (23). Circulating levels of lipoprotein(a) are determined by the number of KIV2 copies. A low number of KIV2 copies results in small apo(a) isoforms, which lead to efficient secretion and contribute to higher levels of lipoprotein(a). On the other hand, the high number of KIV2 copies leads to large apo(a) isoforms and low levels of circulating lipoprotein(a).     

 

Apo(a) is mainly synthesized in the liver. Newly formed apo(a) is then bound to apo B-100 of LDL to become lipoprotein(a). The assembly of lipoprotein(a) particles is a two-step process (24). The first step is the non-covalent docking of the lysine-binding site at the KIV7-KIV8 domains of apo(a) to the lysine residues at the N-terminus of apo B-100. The second step is the covalent binding through the disulfide bond between the 2 free cysteines in the KIV-9 of apo(a) and apo B-100 (25). The site of lipoprotein(a) assembly has been controversial whether it occurs intracellularly or extracellularly (23,25,26), but a recent study using a hepatocyte cell model suggests that the first non-covalent step occurs intracellularly, whereas the second covalent bond formation occurs extracellularly (27,28).

 

FUNCTION

 

The physiological function of lipoprotein(a) remains obscure (23). Earlier, it has been suggested that lipoprotein(a) may assist in wound healing (16,29). Lipoprotein(a) can interact with fibrin and other components of the extracellular matrix (30) via the lysine-binding sites in its kringle domains and it delivers cholesterol to the site of injury. Besides carrying cholesterol esters, free cholesterol, triglycerides, and phospholipids, lipoprotein(a) is the main carrier of oxidized phospholipids in the circulation (15). Lipoprotein(a) might also act as a carrier/scavenger for oxidized lipids since oxidized phospholipids and platelet-activating factor acetylhydrolase, an enzyme involved in hydrolyzing PAF, are found on lipoprotein(a) particles. In vitro, lipoprotein(a) can interfere with many steps of blood clotting and fibrinolysis (21), but the evidence for its thrombogenic role in vivo is less convincing (16).

 

In the Finnish population, loss-of-function variants in the LPA gene have been found (31). Although these subjects have very low levels of lipoprotein(a), no identifiable clinical abnormalities have been demonstrated (31). An increased risk of type 2 diabetes has been reported in those who have very low levels of lipoprotein(a) (32-36). Whether low levels of lipoprotein(a) are causally associated with type 2 diabetes is still unclear.

 

METABOLISM

 

Circulating levels of lipoprotein(a) are primarily determined by isoform size-dependent production rate and not catabolism (37). Fractional catabolic rates for lipoprotein(a) with short and long isoforms have been shown to be relatively indifferent (37). Liver is the main site of lipoprotein(a) catabolism, with a minor contribution from the kidney (26). Apo(a), apo B-100 and oxidized phospholipids carried on lipoprotein(a) may play a role in catabolism of lipoprotein(a) particles by acting as ligands for various receptors. A number of receptors have been shown to be associated with clearance of lipoprotein(a), including lipoprotein receptors, scavenger receptors, plasminogen receptors, Toll-like receptors (TLRs), and carbohydrate receptors or lectins (38). However, the relative contribution of these different receptors in lipoprotein(a) clearance remains to be explored. The lack of suitable animal models and the conflicting data on the role of certain receptors on the catabolism of lipoprotein(a) also make it difficult to conclude the definite role of these receptors. Following uptake into the cells, it has been shown that lipoprotein(a) dissociates into 2 components, with LDL being degraded in lysosomes and apo(a) being recycled to be re-secreted (39). It has been estimated that approximately 30% of apo(a) is recycled after lipoprotein(a) internalization into the cells (39), which could potentially contribute to the circulating level of lipoprotein(a).

 

GENETICS

 

Lipoprotein(a) levels are strongly determined by the genetic locus at the LPA gene on chromosome 6, which encodes apo(a). The LPA gene is closely related to the PLG gene, which encodes plasminogen. These 2 genes diverged about 40 million years ago with the loss of KI-KIII and the expansion and differentiation of KIV domain (23). The major genetic determinant is a copy number variation of the KIV2 repeat, which explains approximately 30-70% of the variability in lipoprotein(a) levels, depending on ethnicity (40). The apo(a) size is highly variable among individuals and depends on the number of KIV2 repeats in the LPA gene. Expression of a low number (10-22) of KIV2 repeats is characterized by small apo(a) isoforms and higher lipoprotein(a) levels, whereas a higher number (>23) of KIV2 repeats results in large apo(a) isoforms and lower lipoprotein(a) levels. Smaller apo(a) isoforms are associated with higher apo(a) production rates from the liver (41), whereas larger isoforms are more susceptible to proteosomal degradation in the endoplasmic reticulum (42). Subjects who carry a low number of KIV repeats have 4-5 times higher median lipoprotein(a) levels than those with a high number of KIV repeats (43).

 

Because every individual has 2 copies of this gene, 2 different isoforms may be present in plasma. In general, plasma levels of lipoprotein(a) are determined by the net production of apo(a) in each isoform. In Caucasians, with an increasing number of KIV repeats, and hence the larger apo(a) isoform, the frequency of the non-expressed allele is also increasing (44). As a result, the major isoform in circulation is mainly driven by the smaller ones. However, in about one-quarter of Caucasians, the larger allele is reported to be the dominant one (44) so the smaller allele is not always dominant (45). The frequency of the non-expressed alleles in Caucasians was highest in the mid-range whereas in African Americans, the non-expressed alleles were fairly distributed across apo(a) sizes (44).        

 

It is of note that among those who carry the same number of KIV-2 repeats, there is a wide range of variability in the lipoprotein(a) levels. This could be explained by the presence of other genetic variants beyond the KIV repeat polymorphism that are also known to influence lipoprotein(a) levels (23,43,45) with a variable impact among various populations. A large genome-wide association meta-analysis has identified a number of independent single nucleotide polymorphisms (SNPs) around the LPA gene which are significantly associated with lipoprotein(a) levels (46). Certain SNPs are functionally lipoprotein(a)-increasing, such as rs1800769 and rs1853021, whereas other SNPS, such as rs10455872 and rs3798220, have no functional effect, but they are in linkage disequilibrium with other small apo(a) isoforms or other variants associated with elevated lipoprotein(a) levels (47).  

 

On the contrary, other genetic variants of the LPA gene are associated with low levels of lipoprotein(a), such as the common splice variants, 4733G>A (48) and 4925G>A (49) in the KIV-2 repeat, and the missense variant rs41267813 (50). Other SNPs are null alleles which decrease lipoprotein(a) levels, such as rs41272114, rs41259144 and rs139145675. Individuals who carry these variants are found to be protected against the development of ASCVD.

 

Other genes, besides the LPA gene, have also been shown to influence lipoprotein(a) levels (45) although a meta-analysis has not confirmed the findings (51), suggesting that further studies with large sample sizes are required to replicate these findings. A recent genome-wide association study from the UK Biobank in almost 300,000 individuals has identified APOE, APOH, and CETP as additional loci that affect lipoprotein(a) levels (52).

 

Although lipoprotein(a) is under a strong genetic regulation, currently, there is no recommendation or advice to perform genetic testing for the LPA gene. Measuring lipoprotein(a) level in the circulation is considered sufficient since it reflects the overall genetic interaction of all variants.

 

LIPOPROTEIN(A) LEVEL

 

The distribution of lipoprotein(a) level in plasma is highly skewed with a tail toward higher levels as shown in Figure 2 (16,17,53). The range varies widely from <0.1 mg/dL to >300 mg/dL (<0.2 – 750 nmol/L). In people of European descent, 80% of the population have a serum level of lipoprotein(a) <40 mg/dL or 90 nmol/L (18). It is well known that plasma levels of lipoprotein(a) are different among various ethnicities, which are predominantly determined by lipoprotein(a) isoform size and other genetic variants in the LPA locus (45). Data from the UK Biobank showed that the mean lipoprotein(a) levels were lowest in Chinese individuals (16 nmol/L), followed by White (19 nmol/L), South Asian (31 nmol/L), and highest in Black individuals (75 nmol/L) (17). Data from subjects with various ethnicities have demonstrated that the level of lipoprotein(a) is slightly higher in women than in men (17,53-56), although other studies have shown a lack of difference between men and women (57,58).

 

Figure 2. Distribution of lipoprotein(a) in the Danish general population (from (16)).

 

Lipoprotein(a) level is fully expressed by the age of 2 years and the adult levels are usually achieved by 5 years (59). The level of lipoprotein(a) seems to be stable throughout life because it is under a strong genetic influence (60). Except for certain medical conditions, plasma lipoprotein(a) levels are relatively stable across the lifespan independent of lifestyle. A long-term study over a period of 15 years has shown that the overall absolute change in lipoprotein(a) levels is relatively modest but may be more pronounced in subjects with very high lipoprotein(a) levels (60). The level is also not affected by fasting (61).

 

Because lipoprotein(a) levels are predominantly determined by genetics, lifestyle modifications appear to have minimal effects (45). A low carbohydrate/high saturated fat diet was shown to result in a 15% decrease in lipoprotein(a) levels (62), whereas a decrease in saturated fat was associated with a 10-15% increase in lipoprotein(a) levels (63). Dietary changes may affect lipoprotein(a) levels in the opposite direction to LDL-C. Physical activity and exercise may affect lipoprotein(a) levels but the results are inconsistent and it may depend on the host, type, intensity, and duration (64).

 

As lipoprotein(a) is produced by the liver, a reduction in lipoprotein(a) levels is observed in liver disease (65,66). Liver transplantation has been shown to change apo(a) isoform to that of the donor with corresponding changes in lipoprotein(a) levels (67). In contrast, elevations of lipoprotein(a) levels are shown in nephrotic syndrome and chronic kidney diseases, either from impaired catabolism or increased hepatic production in response to protein loss in urine or in dialysis (68,69). A large increase is found among subjects who carry large apo(a) isoform sizes (70). Initiation of dialysis has no effect on lipoprotein(a) levels (71). Kidney transplantation can rapidly normalize lipoprotein(a) levels within several weeks (72-74).

 

Infection and inflammation also affect lipoprotein(a) levels. In acute and chronic inflammatory conditions, such as autoimmune diseases, lipoprotein(a) levels were increased (10,75), but in severe life-threatening conditions, such as sepsis and burns, lipoprotein(a) levels were decreased (76). Lipoprotein(a) levels increased during COVID infections, which might be responsible for increased thromboembolic events (77,78). The LPA gene contains interleukin-6 response elements (79) and elevated lipoprotein(a) levels could be decreased using interleukin-6 receptor blockade by tocilizumab (80). Following acute myocardial infarction, earlier results on the changes in lipoprotein(a) were conflicting, which could be due to differences in assay system (81,82).

 

Changes in endogenous sex hormones, including menopause and ovariectomy, have been found to have minimal effects on lipoprotein(a) levels (83,84), although a 27% increase after menopause has also been reported (85). Castration and orchidectomy have been shown to slightly increase lipoprotein(a) levels (86,87). Hormone replacement therapy in postmenopausal women reduces lipoprotein(a) levels by approximately 12-25% (85,88).

 

Longitudinal studies during pregnancy have shown that lipoprotein(a) levels are increased during the first trimester, peak at the middle of the second trimester and return to baseline after childbirth (89-91). Other studies, however, report no changes in lipoprotein(a) during pregnancy (92,93).

 

Hyperthyroidism is associated with a reduction in lipoprotein(a) level and treatment is found to increase it (94). Hypothyroidism is also associated with an elevation in lipoprotein(a) level and treatment with thyroxine can reduce it (94). Growth hormone (GH) therapy for GH-deficient adults can markedly increase lipoprotein(a) levels (95). 

 

A decrease in lipoprotein(a) level has been reported in several genetic disorders of lipoprotein metabolism including abetalipoproteinemia, lecithin-cholesterol acyltransferase (LCAT) deficiency, and lipoprotein lipase deficiency (96). In contrast, an increase in lipoprotein(a) level is observed in familial hypercholesterolemia (FH) and familial defective apo B (96).

 

Conditions that have been reported to increase or decrease lipoprotein(a) levels are shown in Table 1.

 

Table 1. Conditions Associated with Changes in Lipoprotein(a) Levels

Increase

Decrease

No change

High carbohydrate/low fat diet (10-15%)

Low carbohydrate/high fat diet (10-15%)

Lifestyle intervention

Hypothyroidism (5-20%)

Hyperthyroidism (20-25%)

Alcohol consumption

Pregnancy (200%)

Hormone replacement therapy (25%)

Fasting

Castration/ovariectomy (small)

Severe acute-phase reactions (burn/sepsis)

Endogenous sex hormone

Growth hormone therapy (25-100%)

Tocilizumab (30-40%)

Menopause

Chronic kidney disease (200-400%)

Hepatitis/cirrhosis

 

Nephrotic syndrome (300-500%)

 

 

Severe inflammatory condition

 

 

Protease inhibitors/antiretroviral therapy

 

 

 

ISSUES IN LIPOPROTEIN(A) MEASUREMENT AND REPORTING

 

The wide variation in apo(a) isoform size among individuals poses challenges in measuring and reporting lipoprotein(a) levels. Most commercially available immunoassays use polyclonal antibodies which may cross-react with multiple KIV2 repeats (97). As a result, it may overestimate or underestimate lipoprotein(a) levels in subjects with large or small apo(a) isoforms, respectively. Currently, the latex-enhanced immunoturbidimetric method by Denka Seiken, Japan, has been shown to be less affected to apo(a) isoform size variability with high concordance of values when compared with the reference enzyme-linked immunosorbent assay (ELISA) method (98,99). Although the antibodies used in the Denka assay are still isoform dependent, the impact of apo(a) size variation is minimized by the use of five different calibrators (99). Nevertheless, results comparing six commercially available immunoassays, all of which use five-point calibrators, show a wide range of discrepancy among various assays (100). The reference ELISA method using monoclonal antibodies developed by a group at the University of Washington is considered the least apo(a) isoform size-sensitive immunoassay available (98,99). Recently, a newly developed LC-MS/MS method has been shown to be unaffected by the apo(a) isoform size polymorphism and proposed as a candidate reference method for standardization of lipoprotein(a) assay (101).

 

Besides the issues in measuring lipoprotein(a) levels, the reporting of lipoprotein(a) levels is also challenging. Currently, there are 2 types of commercially available assays to measure and report lipoprotein(a) levels. The first one reports the level in total lipoprotein(a) mass concentrations, which include the mass of apo(a), apo B-100, lipid, and carbohydrate components. The values are based on assay calibrators and are expressed in mg/dL. In the first method, there is no traceability of the various calibrators to the reference material (102). In the second method, the level is reported in lipoprotein(a) particle number and expressed in nmol/L of apo(a). The assay calibrators used in the second approach are traceable to the World Health Organization/International Federation of Clinical Chemistry and Laboratory Medicine (WHO/IFCCLM) secondary reference material (98) and the values are compared to the “gold standard” monoclonal antibody-based ELISA developed by Marcovina et al. (103). It is recommended that lipoprotein(a) levels should be measured with an assay which is least subjected to apo(a) isoform size and has been calibrated with the WHO/IFCCLM reference material. Current recommendations also encourage the reporting of lipoprotein(a) levels in molar units, but if it is not available, the units in which the assay is calibrated should be used for reporting (59,104,105).

 

It is known that different apo(a) isoform sizes give different molecular weights (106), therefore, direct conversion between molar and mass concentrations (i.e. nmol/L and mg/dL) using a single conversion factor could be misleading and is discouraged (59,99,106). Nevertheless, a factor of 2.0-2.5 is traditionally used to convert from a mass unit to a molar unit (97), although it may vary from 1.85 for a large apo(a) size to 2.85 for a small apo(a) size (99).

 

Since lipoprotein(a) and LDL have relatively similar size and density, cholesterol contained in lipoprotein(a) particles cannot be separated from that in LDL particles and is therefore collectively reported as LDL-C concentration. Previous experiments of isolated lipoprotein(a) particles showed that cholesterol accounted for approximately 30% of lipoprotein(a) mass concentration, therefore, lipoprotein(a)-cholesterol could be estimated by multiplying lipoprotein(a) mass (mg/dL) by 0.3 and used to correct LDL-C (lipoprotein(a)-cholesterol-corrected LDL-C) (59). However, a recent study directly measured lipoprotein(a)-cholesterol relative to lipoprotein(a) mass demonstrated that the percentage varied widely, ranging from 5.8% to 57.3% with a median of 17.3% (107), therefore, routine correction of LDL-C for lipoprotein(a)-cholesterol is currently not recommended in clinical practice (59). There are certain exceptions to this. First, in patients with clinically suspected familiar hypercholesterolemia (FH) and elevated lipoprotein(a) levels, measurement of lipoprotein(a)-cholesterol may be warranted since the corrected value may help exclude the diagnosis of FH and avoid unnecessary genetic testing (108). Second, correcting LDL-C for lipoprotein(a)-cholesterol may help explain the suboptimal response or resistance to statin therapy. High levels of lipoprotein(a) result in falsely elevated LDL-C. Since statins do not lower lipoprotein(a) levels, cholesterol in lipoprotein(a) is considered a statin-resistant fraction of LDL-C.

 

EFFECTS OF LIPOPROTEIN(A) ON VASCULAR DISEASE

 

Lipoprotein(a) exerts multiple effects that could be proatherogenic, proinflammatory, and prothrombotic.

 

Lipoprotein(a) could be atherogenic since lipoprotein(a) is small enough (<70 nm in diameter) to enter and become trapped in the vascular wall. Cholesterol carried on lipoprotein(a) particle could then be deposited in the arterial intima and aortic valvular leaflets. However, the number of circulating lipoprotein(a) particles is substantially lower than that of LDL particles. As a result, the amount of cholesterol deposited from lipoprotein(a) is expected to be much lower than that from LDL (109). Apo(a), the main protein of lipoprotein(a), also contains lysine-binding sites that could tightly bind to exposed surface of denuded endothelium of the vascular wall or the aortic valve leaflets. In an animal experiment of endothelial injury, it was found that lipoprotein(a) preferentially accumulated at injured sites, compared with LDL (110). These findings suggest that accumulation of lipoprotein(a) at the site of vascular injury could be the primary mechanism by which elevated lipoprotein(a) causes cardiovascular disease.   

 

There is increasing evidence that oxidized phospholipids carried on lipoprotein(a) play a major role in atherogenesis and valvular calcification. In plasma, lipoprotein(a) is the main carrier of oxidized phospholipids (15). Interaction between oxidized phospholipids and apo(a) is mediated by the histidine residues in the KIV-10 domain of apo(a) (111,112). These oxidized phospholipids are both proatherogenic and proinflammatory (113). Oxidized phospholipids can induce inflammation by binding TLR2, TLR4, CD14 and CD36 on monocytes, macrophages, and endothelial cells (79,114). The result is an activation of multiple cytokines, chemokines, and adhesion molecules that mediate monocyte activation and migration into the vascular wall, endothelial dysfunction and proliferation, proliferation and migration of vascular smooth muscle cells into the atheromatous plaques, generation of reactive oxygen species, progression of vascular wall inflammation, and cell apoptosis that could lead to plaque rupture (115,116).

 

Structurally, apo(a) is similar to plasminogen, but the protease domain of apo(a) is catalytically inactive. Therefore, apo(a) may inhibit binding of plasminogen to endothelial cell surface receptor, compete with plasminogen for fibrin affinity sites, and interfere with fibrinolytic activity of plasminogen through a molecular mimicry mechanism (21). However, lowering lipoprotein(a) levels does not affect ex vivo fibrinolysis (117), suggesting that the role of lipoprotein(a) in thrombosis may involve other mechanisms beyond fibrinolysis (118). Lipoprotein(a) has also been shown to induce tissue factor expression, inhibit tissue factor pathway inhibitor, promote platelet activation and aggregation, and alter the structure of fibrin, which ultimately leads to thrombosis (118).

 

It is of note that these proatherogenic, proinflammatory, and prothrombotic effects of lipoprotein(a) have been demonstrated mainly from in vitro studies, and the clinical significance of these findings needs to be established from human studies.  

 

LIPOPROTEIN(A) AND CARDIOVASCULAR DISEASE

 

Over the past two decades, data from epidemiological studies, meta-analyses, genome-wide association studies, and Mendelian randomization studies, have provided conclusive evidence that elevated lipoprotein(a) levels are associated with a higher risk of ASCVD (16,18,47,119-121). This is true in all ethnicities studied to date (17,55,122). The result of a genome-wide association study revealed that the most potent genetic association with coronary artery disease (CAD) was the LPA locus (121). A number of Mendelian randomization studies have shown that genetic variants associated with high lipoprotein(a) levels are more prevalent in subjects with ASCVD (18,47), whereas genetic variants associated with low lipoprotein(a) levels are protective against the development of ASCVD (31,49).

 

Several epidemiological studies, both in primary prevention and secondary prevention settings, have shown an association between lipoprotein(a) levels and ASCVD (17,19,55,120,123). The association is continuous and linear in different ethnicities without evidence of a threshold effect (17,120). Data from a large prospective cohort study from the UK Biobank showed a broadly linear relationship between lipoprotein(a) levels and ASCVD with an 11% increased risk of ASCVD with every 50 nmol/L increase in lipoprotein(a) levels (17). Another study also reported a relatively similar result, showing a 2.9% increased risk of CVD with every 18 nmol/L (10 mg/dL) increment in lipoprotein(a) levels (124).

 

Results from the clinical trials suggest that lipoprotein(a) might be a determinant of residual risk of ASCVD in patients who achieved low levels of LDL-C. For example, in the JUPITER trial using high-intensity rosuvastatin, on-statin lipoprotein(a) levels were associated with a residual risk of ASCVD even at low LDL-C levels (125). Similarly, findings from the FOURIER and ODYSSEY-OUTCOMES trials, using statins and PCSK9 inhibitors evolocumab and alirocumab, respectively, also demonstrated that patients with the higher baseline lipoprotein(a) levels were at an elevated risk of ASCVD irrespective of LDL-C levels (126,127). Lipoprotein(a) level is not associated with coronary artery calcium score, but both are independently associated with ASCVD risk (128). Elevated levels of lipoprotein(a) are associated with accelerated progression of atherosclerotic plaque (129).

 

The association between lipoprotein(a) levels and ASCVD is strongest for MI, atherosclerotic stenosis, and calcific aortic valvular disease (CAVD) (16). Additionally, very high levels of lipoprotein(a) levels are also associated with an increased risk of ischemic stroke (120,130) and heart failure (131), although the associations are weaker.  

 

Not only that lipoprotein(a) is a determinant of ASCVD in patients with lower risk of ASCVD, but in subjects who are at higher risk of ASCVD, such as those with FH, elevated lipoprotein(a) levels have also been shown to be associated with a higher risk of ASCVD (132,133). Lipoprotein(a) is also an independent predictor of MI and mortality in patients with CKD (134).

 

Regarding prothrombotic effects of lipoprotein(a), although in vitro data demonstrated that lipoprotein(a) might impair fibrinolysis, a potential role for lipoprotein(a) in prothrombotic and antifibrinolytic activity in vivo remains unproven (21). Epidemiological studies and Mendelian randomization studies have failed to establish the major role of lipoprotein(a) in venous thrombosis in adults (135,136). In children, however, there is some evidence that elevated lipoprotein(a) level is a risk factor for arterial ischemic stroke and venous thromboembolism (137).

 

LIPOPROTEIN(A) AND CALCIFIC AORTIC VALVULAR DISEASE

 

Calcific aortic valvular disease (CAVD) is the disease of the aortic valve that includes early valvular sclerosis and advanced aortic valvular stenosis. CAVD is the leading cause of aortic valve replacement in developed countries, but there is currently no available medical treatment for CAVD. CAVD is characterized by thickening of the aortic valve leaflets with progressive stenosis of the aortic valve. Although the early stages of CAVD are asymptomatic, severe aortic stenosis may lead to significant left ventricular outflow obstruction and subsequent development of syncope, angina and heart failure.

 

Early cross-sectional studies have shown a strong association between elevated lipoprotein(a) and CAVD (138,139). Such association, however, cannot be used to indicate that lipoprotein(a) is causal for CAVD. In 2013, a genome-wide association study in subjects of European ancestry reported an association between genetic variants in the LPA locus and CAVD (20), which was also confirmed across multiple ethnic groups (20,140). Subsequent cohort and case-control studies have verified this finding and further linked lipoprotein(a) levels and oxidized phospholipids to CAVD (141-144). Lipoprotein(a) is now considered as a new causal risk factor for CAVD (16). Two recent meta-analyses show that plasma lipoprotein(a) levels ³50 mg/dL are associated with a 1.76-1.79-fold increased risk of CAVD (145,146). Elevated lipoprotein(a) levels have also been associated with the faster progression of CAVD and the increased risk for aortic valve replacement and cardiovascular death (143,146-149). In contrast, genetically lowered levels of lipoprotein(a) are associated with a 37% lower risk of aortic stenosis (150).

 

The normal aortic valve consists of endothelial cells and valvular interstitial cells. In the early stage of CAVD, increased permeability of valvular endothelial cells occurs and results in endothelial barrier dysfunction. Mechanical shear stress may cause further injury to the valvular endothelial cells, leading to endothelium denudation. As a result, lipid infiltration by LDL and lipoprotein(a) follows, similar to the development of atherosclerosis. Inflammatory responses ensue and induce the valvular interstitial cells to gain a myofibroblast-like phenotype and release collagen matrix along with other bone-related proteins. The later stage of CAVD is characterized by the osteogenic differentiation of the valvular interstitial cells, resulting in progressive calcification of the aortic valve. Apoptosis of the valvular interstitial cells can lead to formation of hydroxyapatite crystals and further calcification, creating the propagation of both inflammation and calcification. Progressive fibrosis and widespread calcification ultimately result in thickening and dysfunction of the aortic valvular leaflets.  

 

The role of lipoprotein(a) in the molecular pathogenesis of CAVD is now accumulating. Apo(a) may increase the endothelial cell permeability through the Rho/RhoK signaling pathway, which is dependent on the lysine-binding site in the KIV10 domain. Since lipoprotein(a) is a major carrier of oxidized phospholipids in plasma, infiltration of lipoprotein(a) in the valvular tissue can deliver oxidized phospholipids to the valve. Oxidized phospholipids can directly attract monocytes and promote the transformation into macrophages. Oxidized phospholipids can then interact with various receptors on the macrophage, resulting in the release of several pro-inflammatory cytokines and subsequent formation of foam cells. Lipoprotein-associated phospholipase A2 (Lp-PLA2) from the macrophage carried on lipoprotein(a) can also act on oxidized phospholipids and generate lysophosphatidylcholine. Autotaxin, a lysophospholipase D enzyme secreted by the valvular interstitial cells and carried on lipoprotein(a) particles (151), can convert lysophosphatidylcholine into lysophosphatidic acid. Lysophosphatidic acid is both pro-inflammatory and pro-calcifying. By binding the lysophospholipid receptor on the valvular interstitial cells, it can activate the nuclear factor kappa B (NF-kB) and the Wnt-b-catenin pathways, leading to increased transcripts of IL-6 and various osteogenic genes, including runt-related transcription factor 2 (RUNX2) and bone morphogenetic protein 2 (BMP2). The result is the production of alkaline phosphatase, calcium deposition, inflammation, and calcification of the aortic valve (152).

 

These molecular pathways involved in the pathogenesis of CAVD provide several targets of interest, in addition to lipoprotein(a), that could be targeted and may lead to novel therapies for CAVD in the future.

 

GUIDELINE RECOMMENDATIONS FOR MEASUREMENT OF LIPOPROTEIN(A) LEVELS

 

Measurement of lipoprotein(a) levels could help refine the cardiovascular risk in certain conditions and various professional societies have recommended specific clinical conditions that warrant the measurement of lipoprotein(a) levels (59,105,153-156) as shown in Table 2.

 

Table 2. Guideline Recommendations for Lipoprotein(a) Measurement

Indications

AHA

2018

HEART UK 2019

NLA

2019

CCS 2021

BHS

2022

EAS 2022

1. Familial hypercholesterolemia

 

All
Individuals

All Adults

2. Family history (1stdegree relatives) of premature ASCVD

3. Family history (1stdegree relatives) of elevated lipoprotein(a)

 

4. Personal history of premature ASCVD

 

 

5. Recurrent CVD despite optimal statin treatment

 

 

 

6. Inadequate LDL-C reduction in response to statin

 

 

 

7. Borderline or intermediate risk of ASCVD

8. Calcific valvular aortic stenosis

 

It has been pointed out that different recommendations on measurement of lipoprotein(a) among various guidelines may be related to when the guidelines have been written (157). Most of the earlier guidelines list a number of conditions in which lipoprotein(a) should be measured, but the 2019 European Society of Cardiology/European Atherosclerosis Society (ESC/EAS) dyslipidemia guidelines have simplified this and recommended that lipoprotein(a) measurement should be considered at least once in each adult person’s lifetime (158). The rationale behind this recommendation is to identify individuals with very high levels of lipoprotein(a), i.e., >180 mg/dL or >430 nmol/L, who have a lifetime risk of ASCVD equivalent to that of heterozygous FH (158). Thus, measuring lipoprotein(a) levels in those with very high levels could make a significant contribution in the overall management to reduce cardiovascular risk. For the latest EAS lipoprotein(a) consensus statement published in 2022, it is recommended that lipoprotein(a) levels should be measured at least once in every adult (59). Incorporating lipoprotein(a) levels in the cardiovascular risk assessment could help improve risk stratification and provide comprehensive ASCVD risk evaluation. In addition, failure to consider an individual’s lipoprotein(a) level could lead to an underestimate of the absolute cardiovascular risk (59). For example, in a person with baseline risk of ASCVD events of 25%, a lipoprotein(a) level of 150 mg/dL will substantially increase the risk to 68% (59). A new risk calculator incorporating lipoprotein(a) levels and other traditional cardiovascular risk factors is now publicly available at http://www.lpaclinicalguidance.com.

 

Elevated lipoprotein(a) level is transmitted as a co-dominant trait. When an index subject is found to have high lipoprotein(a) level, screening for high lipoprotein(a) level in other family members is recommended and could help identify other affected family members (59). This approach is relatively similar to cascade screening in FH, except that genetic testing is not required. For children, there is a recommendation that children who have history of hemorrhagic or ischemic stroke, children of a parent with premature ASCVD and no other identifiable risk factors, and children of a parent with high levels of lipoprotein(a) should have lipoprotein(a) testing (59,154).

 

In general, the higher the lipoprotein(a) level, the greater risk for ASCVD. Therefore, the use of a specific threshold is biologically implausible and not appropriate. However, clinical practice guidelines prefer the use of a certain threshold for therapeutic decisions. An early study in patients who suffered acute MI proposed the lipoprotein(a) level of >30 mg/dL to reflect a higher risk of MI (5), and this cutoff values have been referred to in several guideline recommendations from various international societies (156,159,160). Another threshold level of lipoprotein(a) that has been used by other professional societies to confer an increased risk of ASCVD is 50 mg/dL or 100-125 nmol/L (153,154), which corresponds to the 80th percentile in the European descent (45). A recent 2022 consensus statement from the EAS proposed a threshold of ³50 mg/dL (125 nmol/L) to rule in and <30 mg/dL (75 nmol/L) to rule out cardiovascular risk, whereas levels between 30-50 mg/dL (75-125 nmol/L) are grey zones (59). Different thresholds according to ethnicities have been proposed (161), although a large study from the UK Biobank has shown that the ASCVD risk among various ethnicities is similar when using a uniform threshold of ³150 nmol/L (17). At present, current management should remain the same regardless of ethnicity.

 

Interestingly, an inverse relationship between lipoprotein(a) levels and the risk of type 2 diabetes has been demonstrated from various studies and subjects with very low levels of lipoprotein(a) are at increased risk of diabetes (32-36,162). Some, but not all, genetic studies also suggest that low lipoprotein(a) level is causally associated with the risk of diabetes (33-35). The exact mechanism underlying this association is currently unclear. Whether potent specific lipoprotein(a)-lowering therapy might increase the risk of diabetes remains to be determined.

 

LIPOPROTEIN(A)-LOWERING TREATMENT

 

Currently, there is no approved medication for lowering lipoprotein(a) levels. Currently available lipid-lowering medications result in modest (10-30%) reduction in lipoprotein(a) levels (table 3) (163). Data from the Mendelian randomization studies have suggested that lowering lipoprotein(a) levels by approximately 100 mg/dL (215 nmol/L) would produce a similar benefit of reducing ASCVD risk by 22% as shown by lowering LDL-C levels by 38.6 mg/dL (1 mmol/L) using statin therapy (164). A separate analysis using data from primary prevention studies suggested that this number might be overestimated and lowering lipoprotein(a) levels by 65 mg/dL would be enough to give 22% ASCVD risk reduction (165,166). Similarly, data from the Copenhagen General Population Study demonstrated that lowering lipoprotein(a) by 50 mg/dL (105 nmol/L) within a 5-year period would produce a 20% reduction in major adverse cardiovascular events for secondary prevention (167). It is of note that lowering LDL-C levels of 38.6 mg/dL to give a reduction of an adverse cardiovascular event of 22% was used only as a benchmark and it is possible that lowering lipoprotein(a) levels by smaller amount may also be beneficial as previously shown for LDL-C lowering.

 

Although early studies report an increase in lipoprotein(a) levels after statin therapy, recent meta-analyses of statin therapy show that there is heterogeneity across studies. In addition, the changes in lipoprotein(a) levels are relatively small and may not be clinically meaningful (168,169). Currently, it is widely accepted that the benefits of statin in lowering LDL-C levels and decreasing ASCVD risk outweigh the potential risk associated with a small increase in lipoprotein(a) levels. 

 

PCSK9 inhibitors, including evolocumab, alirocumab and inclisiran, can lower lipoprotein(a) levels by 10-30% and the absolute reduction is greatest in those with high lipoprotein(a) levels at baseline (126,127,170). A post-hoc analysis from the FOURIER trial using evolocumab reported a 23% decrease in adverse cardiovascular events in those with a baseline lipoprotein(a) level >37 nmol/L and a 7% reduction in those with <37 nmol/L (126). A significant relationship between a 15% lower risk per 25 nmol/L reduction of lipoprotein(a) levels was also observed. These data suggest that a small lowering in lipoprotein(a) level might have a clinical benefit. Results from the ODYSSEY Outcomes trial using alirocumab also showed a significant reduction in major cardiovascular events among those in the two highest quartiles of baseline lipoprotein(a) levels (127).    

 

Interestingly, an exploratory analysis of the FOURIER trial further showed that lipoprotein(a) level was associated with future aortic stenosis (AS) events (new or worsening CAVD or aortic valve replacement) and that evolocumab therapy beyond one year might reduce these AS events (171). However, it should be noted that this post hoc analysis was performed in only a small number of subjects and the original trial was not designed to evaluate the impact of PCSK9 inhibitors on CAVD.

 

Other lipid-lowering medications, such as fibrates, ezetimibe, bempedoic acid and bile acid sequestrants, also have modest effects on lipoprotein(a) levels (163). Niacin can lower lipoprotein(a) levels by 20% due to decreased LPA mRNA and apo(a) production rate (172). Mipomersen and CETP inhibitors also decrease lipoprotein(a) levels by 20-30% (173). Currently, the use of these medications to lower lipoprotein(a) level is not advised.

 

Lipoprotein apheresis is an option to lower lipoprotein(a) levels in clinical practice, although levels of all apoB-containing lipoproteins are reduced after treatment (174). A median reduction in lipoprotein(a) level by 70% was observed (175) but the time-averaged reduction was around 30-35% (176). Notably, a reduction in CAD events has been reported from several retrospective studies (177-180). Lipoprotein apheresis could therefore be considered in those with very high levels of lipoprotein(a) and progressive cardiovascular disease despite optimal management of other risk factors (174). Worldwide, lipoprotein apheresis is not commonly performed except in Germany where it is approved for ASCVD patients who have elevated lipoprotein(a) levels (>120 nmol/L or >60 mg/dL) and recurrent ASCVD events, irrespective of LDL-C levels (175). Recently, it is also approved in the United States for patients with lipoprotein(a) levels >60 mg/dL and LDL-C levels >100 mg/dL with documented CAD or peripheral arterial disease (163).

 

Without specific lipoprotein(a)-lowering therapy yet available, in subjects with elevated lipoprotein(a) levels, intensive risk factor management, such as healthy diet and lifestyle behavior modifications, is recommended along with the intensification of statin therapy to lower the risk of ASCVD (59). Data from the EPIC-Norfolk population-based study showed that in subjects with elevated lipoprotein(a) above 50 mg/dL, those who modified their lifestyles to maintain ideal cardiovascular health had about one third of cardiovascular risk compared to those with poor cardiovascular health (181). These modifiable cardiovascular health scores included body mass index, healthy diet, physical activity, smoking status, high blood pressure, diabetes and cholesterol concentration (181). In those whose LDL-C target levels are not achieved, ezetimibe or PCSK9 inhibitors could be considered (154).

 

Aspirin has been shown to be associated with a cardiovascular risk reduction in subjects who carried the SNP rs3798220-C with elevated lipoprotein(a) levels from the Women’s Health Study and the Aspirin in Reducing Events in the Elderly (APREE) trial (182). However, this SNP was present in only a small percentage of Caucasian subjects (approximately 3-4%) in these 2 trials. Therefore, further studies with a larger number of subjects in broader populations are needed to confirm the benefits of aspirin.

 

NEW SPECIFIC LIPOPROTEIN(A)-LOWERING TREATMENT

 

Since apo(a) is exclusively produced by the liver, novel therapies targeting hepatic apo(a) production using antisense oligonucleotide (ASO) and small interfering RNA (siRNA) technologies are under active investigation. 

 

Pelacarsen, formerly known as TQJ230, IONIS-APO(a)-LRx and AKCEA-APO(a)-LRx, is a second generation of apo(a) ASO, conjugated with N-acetylgalactosamine (GalNAc). Since GalNac is preferentially bound to asialoglycoprotein receptor on the cell surface of hepatocytes, the design of this GalNac-conjugated molecule will ensure the selective uptake by hepatocytes. Administered by subcutaneous injection every 4 weeks, it can bind apo(a) RNA in the hepatocytes, leading to an approximately 80% reduction in lipoprotein(a) levels in subjects with lipoprotein(a) level >150 nmol/L or approximately 60 mg/dL (183). Pelacarsen is generally well tolerated with the most frequently reported adverse event being injection site reactions. Currently, a phase 3 Lp(a)-HORIZON (NCT04023552) cardiovascular outcome study is underway to investigate the efficacy and safety of pelacarsen at the dose of 80 mg monthly for 4 years in subjects with ASCVD and lipoprotein(a) ³70 mg/dL.

 

Olpasiran, formerly known as AMG890 and ARO-LPA, is a GalNAc-conjugated siRNA that could be given every 3-6 months and result in a greater (>90%) reduction in lipoprotein(a) levels in subjects with lipoprotein(a) level >150 nmol/L or approximately 70 mg/dL (184). The most common adverse events were injection site reactions and hypersensitivity reactions. A cardiovascular outcome study (NCT05581303) is currently investigating the effects of olpasiran given every 12 weeks for 4 years in subjects with ASCVD and lipoprotein(a) levels ³200 nmol/L.

 

Zerlasiran, also known as SLN360, is another GalNAc-conjugated siRNA (185). A Phase 1 study investigated the safety and tolerability of zerlasiran after single ascending doses and multiple doses in healthy subjects with elevated lipoprotein(a) level ³150 nmol/L. A dose-dependent and sustained reduction in lipoprotein(a) level has been observed with a 98% reduction in the group receiving 600 mg of zerlasiran (185). A phase 2 ALPACAR-360 study (NCT05537571) to evaluate the Lipoprotein (a) lowering efficacy, safety and tolerability of zerlasiran in adult participants with elevated lipoprotein(a) (³125 nmol/L) at high risk of ASCVD is now underway.

 

Besides nucleic acid therapeutics to inhibit hepatic production of apo(a), small molecule inhibitors of lipoprotein(a) formation have been developed. Muvalaplin, an oral small molecule that blocks the initial noncovalent binding between apo(a) and apo B-100, thus disrupting lipoprotein(a) formation, has been reported to lower lipoprotein(a) level by 65% after 14 days of daily dosing in a phase 1 study (186). No serious adverse effects have been noted.

 

The effect of lipid-lowering medications on lipoprotein(a) levels is shown in Table 3.

 

Table 3. Effect of Lipid-Lowering Medications on Lipoprotein(a) Levels

Statins

No effect or slight increase

Ezetimibe

No effect or slight increase

Fibrates

No effect

Bempedoic acid

Minimal effect

Bile acid sequestrants

Minimal effect

Omega-3 fatty acids

No effect

Niacin

Decrease 15-25%

Lomitapide

Decrease 15-20%

Mipomersen*

Decrease 20-30%

CETP inhibitors*

Decrease 20-30%

Estrogen

Decrease 20-35%

PCSK9 inhibitors

Decrease 10-30%

Apo(a) ASO*

Decrease 80%

Apo(a) siRNA*

Decrease 90-98%

Apo(a) small molecule inhibitor*

Decrease 65%

*not currently available

 

The results of these ongoing and future clinical trials of lipoprotein(a) reduction are eagerly awaited and are expected to be the last piece of evidence supporting and confirming the causal relationship between lipoprotein(a) and ASCVD. Until a specific therapy for elevated lipoprotein(a) is available, intensive management of other modifiable cardiovascular risk factors is strongly recommended. Whether these lipoprotein(a)-lowering treatments would also be beneficial in CAVD remains to be further explored.

 

CONCLUSION

 

After 60 years of its discovery, several issues of lipoprotein(a) remain unresolved, including its function and metabolism. While the causality of lipoprotein(a) in ASCVD and CAVD has now been firmly established by epidemiological, genetic association, and Mendelian randomization studies, the next challenge is to prove that lowering lipoprotein(a) levels also leads to cardiovascular benefit in patients with elevated lipoprotein(a) levels.

 

REFERENCES

 

  1. Berg K. A new serum type system in man--the Lp system. Acta Pathol Microbiol Scand. 1963;59:369-382.
  2. Ehnholm C, Garoff H, Simons K, Aro H. Purification and quantitation of the human plasma lipoprotein carrying the Lp(a) antigen. Biochim Biophys Acta. 1971;236(2):431-439.
  3. Frick MH, Dahlen G, Furbery C, Ericson C, Wiljasalo M. Serum pre-beta-1 lipoprotein fraction in coronary atherosclerosis. Acta Med Scand. 1974;195(5):337-340.
  4. Berg K, Dahlen G, Frick MH. Lp(a) lipoprotein and pre-beta1-lipoprotein in patients with coronary heart disease. Clin Genet. 1974;6(3):230-235.
  5. Kostner GM, Avogaro P, Cazzolato G, Marth E, Bittolo-Bon G, Qunici GB. Lipoprotein Lp(a) and the risk for myocardial infarction. Atherosclerosis. 1981;38(1-2):51-61.
  6. Eaton DL, Fless GM, Kohr WJ, McLean JW, Xu QT, Miller CG, Lawn RM, Scanu AM. Partial amino acid sequence of apolipoprotein(a) shows that it is homologous to plasminogen. Proc Natl Acad Sci U S A. 1987;84(10):3224-3228.
  7. McLean JW, Tomlinson JE, Kuang WJ, Eaton DL, Chen EY, Fless GM, Scanu AM, Lawn RM. cDNA sequence of human apolipoprotein(a) is homologous to plasminogen. Nature. 1987;330(6144):132-137.
  8. Utermann G, Menzel HJ, Kraft HG, Duba HC, Kemmler HG, Seitz C. Lp(a) glycoprotein phenotypes. Inheritance and relation to Lp(a)-lipoprotein concentrations in plasma. J Clin Invest. 1987;80(2):458-465.
  9. Rosengren A, Wilhelmsen L, Eriksson E, Risberg B, Wedel H. Lipoprotein (a) and coronary heart disease: a prospective case-control study in a general population sample of middle aged men. BMJ. 1990;301(6763):1248-1251.
  10. Dahlen GH, Guyton JR, Attar M, Farmer JA, Kautz JA, Gotto AM, Jr. Association of levels of lipoprotein Lp(a), plasma lipids, and other lipoproteins with coronary artery disease documented by angiography. Circulation.1986;74(4):758-765.
  11. Bostom AG, Cupples LA, Jenner JL, Ordovas JM, Seman LJ, Wilson PW, Schaefer EJ, Castelli WP. Elevated plasma lipoprotein(a) and coronary heart disease in men aged 55 years and younger. A prospective study. JAMA.1996;276(7):544-548.
  12. Seman LJ, DeLuca C, Jenner JL, Cupples LA, McNamara JR, Wilson PW, Castelli WP, Ordovas JM, Schaefer EJ. Lipoprotein(a)-cholesterol and coronary heart disease in the Framingham Heart Study. Clin Chem.1999;45(7):1039-1046.
  13. Ridker PM, Hennekens CH, Stampfer MJ. A prospective study of lipoprotein(a) and the risk of myocardial infarction. JAMA. 1993;270(18):2195-2199.
  14. Ridker PM, Stampfer MJ, Hennekens CH. Plasma concentration of lipoprotein(a) and the risk of future stroke. JAMA. 1995;273(16):1269-1273.
  15. Bergmark C, Dewan A, Orsoni A, Merki E, Miller ER, Shin MJ, Binder CJ, Horkko S, Krauss RM, Chapman MJ, Witztum JL, Tsimikas S. A novel function of lipoprotein [a] as a preferential carrier of oxidized phospholipids in human plasma. J Lipid Res. 2008;49(10):2230-2239.
  16. Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J Lipid Res. 2016;57(11):1953-1975.
  17. Patel AP, Wang M, Pirruccello JP, Ellinor PT, Ng K, Kathiresan S, Khera AV. Lp(a) (Lipoprotein[a]) Concentrations and Incident Atherosclerotic Cardiovascular Disease: New Insights From a Large National Biobank. Arterioscler Thromb Vasc Biol. 2021;41(1):465-474.
  18. Kamstrup PR, Tybjaerg-Hansen A, Steffensen R, Nordestgaard BG. Genetically elevated lipoprotein(a) and increased risk of myocardial infarction. JAMA. 2009;301(22):2331-2339.
  19. Kamstrup PR, Benn M, Tybjaerg-Hansen A, Nordestgaard BG. Extreme lipoprotein(a) levels and risk of myocardial infarction in the general population: the Copenhagen City Heart Study. Circulation. 2008;117(2):176-184.
  20. Thanassoulis G, Campbell CY, Owens DS, Smith JG, Smith AV, Peloso GM, Kerr KF, Pechlivanis S, Budoff MJ, Harris TB, Malhotra R, O'Brien KD, Kamstrup PR, Nordestgaard BG, Tybjaerg-Hansen A, Allison MA, Aspelund T, Criqui MH, Heckbert SR, Hwang SJ, Liu Y, Sjogren M, van der Pals J, Kalsch H, Muhleisen TW, Nothen MM, Cupples LA, Caslake M, Di Angelantonio E, Danesh J, Rotter JI, Sigurdsson S, Wong Q, Erbel R, Kathiresan S, Melander O, Gudnason V, O'Donnell CJ, Post WS, Group CECW. Genetic associations with valvular calcification and aortic stenosis. N Engl J Med. 2013;368(6):503-512.
  21. Boffa MB, Koschinsky ML. Lipoprotein (a): truly a direct prothrombotic factor in cardiovascular disease? J Lipid Res. 2016;57(5):745-757.
  22. Lawn RM, Schwartz K, Patthy L. Convergent evolution of apolipoprotein(a) in primates and hedgehog. Proc Natl Acad Sci U S A. 1997;94(22):11992-11997.
  23. Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of lipoprotein (a). J Lipid Res.2016;57(8):1339-1359.
  24. Trieu VN, McConathy WJ. A two-step model for lipoprotein(a) formation. J Biol Chem. 1995;270(26):15471-15474.
  25. Dieplinger H, Utermann G. The seventh myth of lipoprotein(a): where and how is it assembled? Curr Opin Lipidol.1999;10(3):275-283.
  26. Kostner KM, Kostner GM. Lipoprotein (a): a historical appraisal. J Lipid Res. 2017;58(1):1-14.
  27. Youssef A, Clark JR, Marcovina SM, Boffa MB, Koschinsky ML. Apo(a) and ApoB Interact Noncovalently Within Hepatocytes: Implications for Regulation of Lp(a) Levels by Modulation of ApoB Secretion. Arterioscler Thromb Vasc Biol. 2022;42(3):289-304.
  28. Boffa MB, Koschinsky ML. Understanding the ins and outs of lipoprotein (a) metabolism. Curr Opin Lipidol.2022;33(3):185-192.
  29. Brown MS, Goldstein JL. Plasma lipoproteins: teaching old dogmas new tricks. Nature. 1987;330(6144):113-114.
  30. van der Hoek YY, Sangrar W, Cote GP, Kastelein JJ, Koschinsky ML. Binding of recombinant apolipoprotein(a) to extracellular matrix proteins. Arterioscler Thromb. 1994;14(11):1792-1798.
  31. Lim ET, Wurtz P, Havulinna AS, Palta P, Tukiainen T, Rehnstrom K, Esko T, Magi R, Inouye M, Lappalainen T, Chan Y, Salem RM, Lek M, Flannick J, Sim X, Manning A, Ladenvall C, Bumpstead S, Hamalainen E, Aalto K, Maksimow M, Salmi M, Blankenberg S, Ardissino D, Shah S, Horne B, McPherson R, Hovingh GK, Reilly MP, Watkins H, Goel A, Farrall M, Girelli D, Reiner AP, Stitziel NO, Kathiresan S, Gabriel S, Barrett JC, Lehtimaki T, Laakso M, Groop L, Kaprio J, Perola M, McCarthy MI, Boehnke M, Altshuler DM, Lindgren CM, Hirschhorn JN, Metspalu A, Freimer NB, Zeller T, Jalkanen S, Koskinen S, Raitakari O, Durbin R, MacArthur DG, Salomaa V, Ripatti S, Daly MJ, Palotie A, Sequencing Initiative Suomi P. Distribution and medical impact of loss-of-function variants in the Finnish founder population. PLoS Genet. 2014;10(7):e1004494.
  32. Mora S, Kamstrup PR, Rifai N, Nordestgaard BG, Buring JE, Ridker PM. Lipoprotein(a) and risk of type 2 diabetes. Clin Chem. 2010;56(8):1252-1260.
  33. Kamstrup PR, Nordestgaard BG. Lipoprotein(a) concentrations, isoform size, and risk of type 2 diabetes: a Mendelian randomisation study. Lancet Diabetes Endocrinol. 2013;1(3):220-227.
  34. Ye Z, Haycock PC, Gurdasani D, Pomilla C, Boekholdt SM, Tsimikas S, Khaw KT, Wareham NJ, Sandhu MS, Forouhi NG. The association between circulating lipoprotein(a) and type 2 diabetes: is it causal? Diabetes.2014;63(1):332-342.
  35. Gudbjartsson DF, Thorgeirsson G, Sulem P, Helgadottir A, Gylfason A, Saemundsdottir J, Bjornsson E, Norddahl GL, Jonasdottir A, Jonasdottir A, Eggertsson HP, Gretarsdottir S, Thorleifsson G, Indridason OS, Palsson R, Jonasson F, Jonsdottir I, Eyjolfsson GI, Sigurdardottir O, Olafsson I, Danielsen R, Matthiasson SE, Kristmundsdottir S, Halldorsson BV, Hreidarsson AB, Valdimarsson EM, Gudnason T, Benediktsson R, Steinthorsdottir V, Thorsteinsdottir U, Holm H, Stefansson K. Lipoprotein(a) Concentration and Risks of Cardiovascular Disease and Diabetes. J Am Coll Cardiol. 2019;74(24):2982-2994.
  36. Paige E, Masconi KL, Tsimikas S, Kronenberg F, Santer P, Weger S, Willeit J, Kiechl S, Willeit P. Lipoprotein(a) and incident type-2 diabetes: results from the prospective Bruneck study and a meta-analysis of published literature. Cardiovasc Diabetol. 2017;16(1):38.
  37. Rader DJ, Cain W, Ikewaki K, Talley G, Zech LA, Usher D, Brewer HB, Jr. The inverse association of plasma lipoprotein(a) concentrations with apolipoprotein(a) isoform size is not due to differences in Lp(a) catabolism but to differences in production rate. J Clin Invest. 1994;93(6):2758-2763.
  38. McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors. Pathology.2019;51(2):155-164.
  39. Sharma M, Redpath GM, Williams MJ, McCormick SP. Recycling of Apolipoprotein(a) After PlgRKT-Mediated Endocytosis of Lipoprotein(a). Circ Res. 2017;120(7):1091-1102.
  40. Scholz M, Kraft HG, Lingenhel A, Delport R, Vorster EH, Bickeboller H, Utermann G. Genetic control of lipoprotein(a) concentrations is different in Africans and Caucasians. Eur J Hum Genet. 1999;7(2):169-178.
  41. Chan DC, Watts GF, Coll B, Wasserman SM, Marcovina SM, Barrett PHR. Lipoprotein(a) Particle Production as a Determinant of Plasma Lipoprotein(a) Concentration Across Varying Apolipoprotein(a) Isoform Sizes and Background Cholesterol-Lowering Therapy. J Am Heart Assoc. 2019;8(7):e011781.
  42. White AL, Guerra B, Wang J, Lanford RE. Presecretory degradation of apolipoprotein [a] is mediated by the proteasome pathway. J Lipid Res. 1999;40(2):275-286.
  43. Coassin S, Kronenberg F. Lipoprotein(a) beyond the kringle IV repeat polymorphism: The complexity of genetic variation in the LPA gene. Atherosclerosis. 2022;349:17-35.
  44. Rubin J, Paultre F, Tuck CH, Holleran S, Reed RG, Pearson TA, Thomas CM, Ramakrishnan R, Berglund L. Apolipoprotein [a] genotype influences isoform dominance pattern differently in African Americans and Caucasians. J Lipid Res. 2002;43(2):234-244.
  45. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res. 2016;57(7):1111-1125.
  46. Mack S, Coassin S, Rueedi R, Yousri NA, Seppala I, Gieger C, Schonherr S, Forer L, Erhart G, Marques-Vidal P, Ried JS, Waeber G, Bergmann S, Dahnhardt D, Stockl A, Raitakari OT, Kahonen M, Peters A, Meitinger T, Strauch K, Kedenko L, Paulweber B, Lehtimaki T, Hunt SC, Vollenweider P, Lamina C, Kronenberg F. A genome-wide association meta-analysis on lipoprotein (a) concentrations adjusted for apolipoprotein (a) isoforms. J Lipid Res.2017;58(9):1834-1844.
  47. Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, Parish S, Barlera S, Franzosi MG, Rust S, Bennett D, Silveira A, Malarstig A, Green FR, Lathrop M, Gigante B, Leander K, de Faire U, Seedorf U, Hamsten A, Collins R, Watkins H, Farrall M, Consortium P. Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N Engl J Med. 2009;361(26):2518-2528.
  48. Schachtl-Riess JF, Kheirkhah A, Gruneis R, Di Maio S, Schoenherr S, Streiter G, Losso JL, Paulweber B, Eckardt KU, Kottgen A, Lamina C, Kronenberg F, Coassin S, Investigators G. Frequent LPA KIV-2 Variants Lower Lipoprotein(a) Concentrations and Protect Against Coronary Artery Disease. J Am Coll Cardiol. 2021;78(5):437-449.
  49. Coassin S, Erhart G, Weissensteiner H, Eca Guimaraes de Araujo M, Lamina C, Schonherr S, Forer L, Haun M, Losso JL, Kottgen A, Schmidt K, Utermann G, Peters A, Gieger C, Strauch K, Finkenstedt A, Bale R, Zoller H, Paulweber B, Eckardt KU, Huttenhofer A, Huber LA, Kronenberg F. A novel but frequent variant in LPA KIV-2 is associated with a pronounced Lp(a) and cardiovascular risk reduction. Eur Heart J. 2017;38(23):1823-1831.
  50. Said MA, Yeung MW, van de Vegte YJ, Benjamins JW, Dullaart RPF, Ruotsalainen S, Ripatti S, Natarajan P, Juarez-Orozco LE, Verweij N, van der Harst P. Genome-Wide Association Study and Identification of a Protective Missense Variant on Lipoprotein(a) Concentration: Protective Missense Variant on Lipoprotein(a) Concentration-Brief Report. Arterioscler Thromb Vasc Biol. 2021;41(5):1792-1800.
  51. Zabaneh D, Kumari M, Sandhu M, Wareham N, Wainwright N, Papamarkou T, Hopewell J, Clarke R, Li K, Palmen J, Talmud PJ, Kronenberg F, Lamina C, Summerer M, Paulweber B, Price J, Fowkes G, Stewart M, Drenos F, Shah S, Shah T, Casas JP, Kivimaki M, Whittaker J, Hingorani AD, Humphries SE. Meta analysis of candidate gene variants outside the LPA locus with Lp(a) plasma levels in 14,500 participants of six White European cohorts. Atherosclerosis. 2011;217(2):447-451.
  52. Hoekstra M, Chen HY, Rong J, Dufresne L, Yao J, Guo X, Tsai MY, Tsimikas S, Post WS, Vasan RS, Rotter JI, Larson MG, Thanassoulis G, Engert JC. Genome-Wide Association Study Highlights APOH as a Novel Locus for Lipoprotein(a) Levels-Brief Report. Arterioscler Thromb Vasc Biol. 2021;41(1):458-464.
  53. Varvel S, McConnell JP, Tsimikas S. Prevalence of Elevated Lp(a) Mass Levels and Patient Thresholds in 532 359 Patients in the United States. Arterioscler Thromb Vasc Biol. 2016;36(11):2239-2245.
  54. Nago N, Kayaba K, Hiraoka J, Matsuo H, Goto T, Kario K, Tsutsumi A, Nakamura Y, Igarashi M. Lipoprotein(a) levels in the Japanese population: influence of age and sex, and relation to atherosclerotic risk factors. The Jichi Medical School Cohort Study. Am J Epidemiol. 1995;141(9):815-821.
  55. Virani SS, Brautbar A, Davis BC, Nambi V, Hoogeveen RC, Sharrett AR, Coresh J, Mosley TH, Morrisett JD, Catellier DJ, Folsom AR, Boerwinkle E, Ballantyne CM. Associations between lipoprotein(a) levels and cardiovascular outcomes in black and white subjects: the Atherosclerosis Risk in Communities (ARIC) Study. Circulation. 2012;125(2):241-249.
  56. Gurdasani D, Sjouke B, Tsimikas S, Hovingh GK, Luben RN, Wainwright NW, Pomilla C, Wareham NJ, Khaw KT, Boekholdt SM, Sandhu MS. Lipoprotein(a) and risk of coronary, cerebrovascular, and peripheral artery disease: the EPIC-Norfolk prospective population study. Arterioscler Thromb Vasc Biol. 2012;32(12):3058-3065.
  57. Jenner JL, Ordovas JM, Lamon-Fava S, Schaefer MM, Wilson PW, Castelli WP, Schaefer EJ. Effects of age, sex, and menopausal status on plasma lipoprotein(a) levels. The Framingham Offspring Study. Circulation.1993;87(4):1135-1141.
  58. Cobbaert C, Kesteloot H. Serum lipoprotein(a) levels in racially different populations. Am J Epidemiol.1992;136(4):441-449.
  59. Kronenberg F, Mora S, Stroes ESG, Ference BA, Arsenault BJ, Berglund L, Dweck MR, Koschinsky M, Lambert G, Mach F, McNeal CJ, Moriarty PM, Natarajan P, Nordestgaard BG, Parhofer KG, Virani SS, von Eckardstein A, Watts GF, Stock JK, Ray KK, Tokgozoglu LS, Catapano AL. Lipoprotein(a) in atherosclerotic cardiovascular disease and aortic stenosis: a European Atherosclerosis Society consensus statement. Eur Heart J.2022;43(39):3925-3946.
  60. Deshotels MR, Sun C, Nambi V, Virani SS, Matsushita K, Yu B, Ballantyne CM, Hoogeveen RC. Temporal Trends in Lipoprotein(a) Concentrations: The Atherosclerosis Risk in Communities Study. J Am Heart Assoc.2022;11(21):e026762.
  61. Langsted A, Kamstrup PR, Nordestgaard BG. Lipoprotein(a): fasting and nonfasting levels, inflammation, and cardiovascular risk. Atherosclerosis. 2014;234(1):95-101.
  62. Ebbeling CB, Knapp A, Johnson A, Wong JMW, Greco KF, Ma C, Mora S, Ludwig DS. Effects of a low-carbohydrate diet on insulin-resistant dyslipoproteinemia-a randomized controlled feeding trial. Am J Clin Nutr.2022;115(1):154-162.
  63. 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(7).
  64. Enkhmaa B, Berglund L. Non-genetic influences on lipoprotein(a) concentrations. Atherosclerosis. 2022;349:53-62.
  65. Feely J, Barry M, Keeling PW, Weir DG, Cooke T. Lipoprotein(a) in cirrhosis. BMJ. 1992;304(6826):545-546.
  66. Alessandri C, Basili S, Maurelli M, Andreozzi P, Violi F, Cordova C. Relationship between lipoprotein(a) levels in serum and some indices of protein synthesis in liver cirrhosis. Clin Chim Acta. 1994;224(2):125-129.
  67. Kraft HG, Menzel HJ, Hoppichler F, Vogel W, Utermann G. Changes of genetic apolipoprotein phenotypes caused by liver transplantation. Implications for apolipoprotein synthesis. J Clin Invest. 1989;83(1):137-142.
  68. Kronenberg F. Causes and consequences of lipoprotein(a) abnormalities in kidney disease. Clin Exp Nephrol.2014;18(2):234-237.
  69. Hopewell JC, Haynes R, Baigent C. The role of lipoprotein (a) in chronic kidney disease. J Lipid Res.2018;59(4):577-585.
  70. Kronenberg F, Lingenhel A, Lhotta K, Rantner B, Kronenberg MF, Konig P, Thiery J, Koch M, von Eckardstein A, Dieplinger H. The apolipoprotein(a) size polymorphism is associated with nephrotic syndrome. Kidney Int.2004;65(2):606-612.
  71. Barbagallo CM, Averna MR, Sparacino V, Galione A, Caputo F, Scafidi V, Amato S, Mancino C, Cefalu AB, Notarbartolo A. Lipoprotein (a) levels in end-stage renal failure and renal transplantation. Nephron.1993;64(4):560-564.
  72. Kronenberg F, Konig P, Lhotta K, Ofner D, Sandholzer C, Margreiter R, Dosch E, Utermann G, Dieplinger H. Apolipoprotein(a) phenotype-associated decrease in lipoprotein(a) plasma concentrations after renal transplantation. Arterioscler Thromb. 1994;14(9):1399-1404.
  73. Black IW, Wilcken DE. Decreases in apolipoprotein(a) after renal transplantation: implications for lipoprotein(a) metabolism. Clin Chem. 1992;38(3):353-357.
  74. Segarra A, Chacon P, Vilardell M, Piera LL. Cyclosporin and serum lipids in renal transplant recipients. Lancet.1993;341(8847):766; author reply 767.
  75. Missala I, Kassner U, Steinhagen-Thiessen E. A Systematic Literature Review of the Association of Lipoprotein(a) and Autoimmune Diseases and Atherosclerosis. Int J Rheumatol. 2012;2012:480784.
  76. Mooser V, Berger MM, Tappy L, Cayeux C, Marcovina SM, Darioli R, Nicod P, Chiolero R. Major reduction in plasma Lp(a) levels during sepsis and burns. Arterioscler Thromb Vasc Biol. 2000;20(4):1137-1142.
  77. Feingold KR. Lipid and Lipoprotein Levels in Patients with COVID-19 Infections. 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)2000.
  78. Moriarty PM, Gorby LK, Stroes ES, Kastelein JP, Davidson M, Tsimikas S. Lipoprotein(a) and Its Potential Association with Thrombosis and Inflammation in COVID-19: a Testable Hypothesis. Curr Atheroscler Rep.2020;22(9):48.
  79. Dzobo KE, Kraaijenhof JM, Stroes ESG, Nurmohamed NS, Kroon J. Lipoprotein(a): An underestimated inflammatory mastermind. Atherosclerosis. 2022;349:101-109.
  80. Schultz O, Oberhauser F, Saech J, Rubbert-Roth A, Hahn M, Krone W, Laudes M. Effects of inhibition of interleukin-6 signalling on insulin sensitivity and lipoprotein (a) levels in human subjects with rheumatoid diseases. PLoS One. 2010;5(12):e14328.
  81. Maeda S, Abe A, Seishima M, Makino K, Noma A, Kawade M. Transient changes of serum lipoprotein(a) as an acute phase protein. Atherosclerosis. 1989;78(2-3):145-150.
  82. Andreassen AK, Berg K, Torsvik H. Changes in Lp(a) lipoprotein and other plasma proteins during acute myocardial infarction. Clin Genet. 1994;46(6):410-416.
  83. Derby CA, Crawford SL, Pasternak RC, Sowers M, Sternfeld B, Matthews KA. Lipid changes during the menopause transition in relation to age and weight: the Study of Women's Health Across the Nation. Am J Epidemiol. 2009;169(11):1352-1361.
  84. Anagnostis P, Antza C, Trakatelli C, Lambrinoudaki I, Goulis DG, Kotsis V. The effect of menopause on lipoprotein (a) concentrations: A systematic review and meta-analysis. Maturitas. 2023;167:39-45.
  85. Simony SB, Mortensen MB, Langsted A, Afzal S, Kamstrup PR, Nordestgaard BG. Sex differences of lipoprotein(a) levels and associated risk of morbidity and mortality by age: The Copenhagen General Population Study. Atherosclerosis. 2022;355:76-82.
  86. Denti L, Pasolini G, Cortellini P, Ferretti S, Sanfelici L, Ablondi F, Valenti G. Effects of androgen suppression by gonadotropin-releasing hormone agonist and flutamide on lipid metabolism in men with prostate cancer: focus on lipoprotein(a). Clin Chem. 1996;42(8 Pt 1):1176-1181.
  87. Henriksson P, Angelin B, Berglund L. Hormonal regulation of serum Lp (a) levels. Opposite effects after estrogen treatment and orchidectomy in males with prostatic carcinoma. J Clin Invest. 1992;89(4):1166-1171.
  88. Salpeter SR, Walsh JM, Ormiston TM, Greyber E, Buckley NS, Salpeter EE. Meta-analysis: effect of hormone-replacement therapy on components of the metabolic syndrome in postmenopausal women. Diabetes Obes Metab. 2006;8(5):538-554.
  89. Zechner R, Desoye G, Schweditsch MO, Pfeiffer KP, Kostner GM. Fluctuations of plasma lipoprotein-A concentrations during pregnancy and post partum. Metabolism. 1986;35(4):333-336.
  90. Sattar N, Clark P, Greer IA, Shepherd J, Packard CJ. Lipoprotein (a) levels in normal pregnancy and in pregnancy complicated with pre-eclampsia. Atherosclerosis. 2000;148(2):407-411.
  91. Manten GT, Franx A, van der Hoek YY, Hameeteman TM, Voorbij HA, Smolders HC, Westers P, Visser GH. Changes of plasma lipoprotein(a) during and after normal pregnancy in Caucasians. J Matern Fetal Neonatal Med.2003;14(2):91-95.
  92. Rymer J, Constable S, Lumb P, Crook M. Serum lipoprotein (A) and apolipoproteins during pregnancy and postpartum in normal women. J Obstet Gynaecol. 2002;22(3):256-259.
  93. Chiang AN, Yang ML, Hung JH, Chou P, Shyn SK, Ng HT. Alterations of serum lipid levels and their biological relevances during and after pregnancy. Life Sci. 1995;56(26):2367-2375.
  94. Kotwal A, Cortes T, Genere N, Hamidi O, Jasim S, Newman CB, Prokop LJ, Murad MH, Alahdab F. Treatment of Thyroid Dysfunction and Serum Lipids: A Systematic Review and Meta-analysis. J Clin Endocrinol Metab.2020;105(12).
  95. Eden S, Wiklund O, Oscarsson J, Rosen T, Bengtsson BA. Growth hormone treatment of growth hormone-deficient adults results in a marked increase in Lp(a) and HDL cholesterol concentrations. Arterioscler Thromb.1993;13(2):296-301.
  96. Kronenberg F, Utermann G. Lipoprotein(a): resurrected by genetics. J Intern Med. 2013;273(1):6-30.
  97. Kronenberg F. Lipoprotein(a) measurement issues: Are we making a mountain out of a molehill? Atherosclerosis.2022;349:123-135.
  98. Marcovina SM, Albers JJ, Scanu AM, Kennedy H, Giaculli F, Berg K, Couderc R, Dati F, Rifai N, Sakurabayashi I, Tate JR, Steinmetz A. Use of a reference material proposed by the International Federation of Clinical Chemistry and Laboratory Medicine to evaluate analytical methods for the determination of plasma lipoprotein(a). Clin Chem.2000;46(12):1956-1967.
  99. Marcovina SM, Albers JJ. Lipoprotein (a) measurements for clinical application. J Lipid Res. 2016;57(4):526-537.
  100. Scharnagl H, Stojakovic T, Dieplinger B, Dieplinger H, Erhart G, Kostner GM, Herrmann M, Marz W, Grammer TB. Comparison of lipoprotein (a) serum concentrations measured by six commercially available immunoassays. Atherosclerosis. 2019;289:206-213.
  101. Marcovina SM, Clouet-Foraison N, Koschinsky ML, Lowenthal MS, Orquillas A, Boffa MB, Hoofnagle AN, Vaisar T. Development of an LC-MS/MS Proposed Candidate Reference Method for the Standardization of Analytical Methods to Measure Lipoprotein(a). Clin Chem. 2021;67(3):490-499.
  102. Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM, Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S, Olive M, Liu L. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J Am Coll Cardiol. 2018;71(2):177-192.
  103. Marcovina SM, Albers JJ, Gabel B, Koschinsky ML, Gaur VP. Effect of the number of apolipoprotein(a) kringle 4 domains on immunochemical measurements of lipoprotein(a). Clin Chem. 1995;41(2):246-255.
  104. Reyes-Soffer G, Ginsberg HN, Berglund L, Duell PB, Heffron SP, Kamstrup PR, Lloyd-Jones DM, Marcovina SM, Yeang C, Koschinsky ML, American Heart Association Council on Arteriosclerosis T, Vascular B, Council on Cardiovascular R, Intervention, Council on Peripheral Vascular D. Lipoprotein(a): A Genetically Determined, Causal, and Prevalent Risk Factor for Atherosclerotic Cardiovascular Disease: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol. 2022;42(1):e48-e60.
  105. Cegla J, Neely RDG, France M, Ferns G, Byrne CD, Halcox J, Datta D, Capps N, Shoulders C, Qureshi N, Rees A, Main L, Cramb R, Viljoen A, Payne J, Soran H, Heart Uk Medical S, Research C. HEART UK consensus statement on Lipoprotein(a): A call to action. Atherosclerosis. 2019;291:62-70.
  106. Tsimikas S, Fazio S, Viney NJ, Xia S, Witztum JL, Marcovina SM. Relationship of lipoprotein(a) molar concentrations and mass according to lipoprotein(a) thresholds and apolipoprotein(a) isoform size. J Clin Lipidol.2018;12(5):1313-1323.
  107. Yeang C, Witztum JL, Tsimikas S. Novel method for quantification of lipoprotein(a)-cholesterol: implications for improving accuracy of LDL-C measurements. J Lipid Res. 2021;62:100053.
  108. Langsted A, Kamstrup PR, Benn M, Tybjaerg-Hansen A, Nordestgaard BG. High lipoprotein(a) as a possible cause of clinical familial hypercholesterolaemia: a prospective cohort study. Lancet Diabetes Endocrinol.2016;4(7):577-587.
  109. Nielsen LB, Gronholdt ML, Schroeder TV, Stender S, Nordestgaard BG. In vivo transfer of lipoprotein(a) into human atherosclerotic carotid arterial intima. Arterioscler Thromb Vasc Biol. 1997;17(5):905-911.
  110. Nielsen LB, Stender S, Kjeldsen K, Nordestgaard BG. Specific accumulation of lipoprotein(a) in balloon-injured rabbit aorta in vivo. Circ Res. 1996;78(4):615-626.
  111. Leibundgut G, Scipione C, Yin H, Schneider M, Boffa MB, Green S, Yang X, Dennis E, Witztum JL, Koschinsky ML, Tsimikas S. Determinants of binding of oxidized phospholipids on apolipoprotein (a) and lipoprotein (a). J Lipid Res. 2013;54(10):2815-2830.
  112. Koschinsky ML, Boffa MB. Oxidized phospholipid modification of lipoprotein(a): Epidemiology, biochemistry and pathophysiology. Atherosclerosis. 2022;349:92-100.
  113. van der Valk FM, Bekkering S, Kroon J, Yeang C, Van den Bossche J, van Buul JD, Ravandi A, Nederveen AJ, Verberne HJ, Scipione C, Nieuwdorp M, Joosten LA, Netea MG, Koschinsky ML, Witztum JL, Tsimikas S, Riksen NP, Stroes ES. Oxidized Phospholipids on Lipoprotein(a) Elicit Arterial Wall Inflammation and an Inflammatory Monocyte Response in Humans. Circulation. 2016;134(8):611-624.
  114. Seimon TA, Nadolski MJ, Liao X, Magallon J, Nguyen M, Feric NT, Koschinsky ML, Harkewicz R, Witztum JL, Tsimikas S, Golenbock D, Moore KJ, Tabas I. Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab. 2010;12(5):467-482.
  115. Simantiris S, Antonopoulos AS, Papastamos C, Benetos G, Koumallos N, Tsioufis K, Tousoulis D. Lipoprotein(a) and inflammation- pathophysiological links and clinical implications for cardiovascular disease. J Clin Lipidol.2023;17(1):55-63.
  116. Coassin S, Kronenberg F. Mechanistic insights into lipoprotein(a): from infamous to 'inflammous'. Eur Heart J.2020;41(24):2272-2274.
  117. Boffa MB, Marar TT, Yeang C, Viney NJ, Xia S, Witztum JL, Koschinsky ML, Tsimikas S. Potent reduction of plasma lipoprotein (a) with an antisense oligonucleotide in human subjects does not affect ex vivo fibrinolysis. J Lipid Res. 2019;60(12):2082-2089.
  118. Boffa MB. Beyond fibrinolysis: The confounding role of Lp(a) in thrombosis. Atherosclerosis. 2022;349:72-81.
  119. Tsimikas S. A Test in Context: Lipoprotein(a): Diagnosis, Prognosis, Controversies, and Emerging Therapies. J Am Coll Cardiol. 2017;69(6):692-711.
  120. Emerging Risk Factors C, Erqou S, Kaptoge S, Perry PL, Di Angelantonio E, Thompson A, White IR, Marcovina SM, Collins R, Thompson SG, Danesh J. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and nonvascular mortality. JAMA. 2009;302(4):412-423.
  121. Consortium CAD, Deloukas P, Kanoni S, Willenborg C, Farrall M, Assimes TL, Thompson JR, Ingelsson E, Saleheen D, Erdmann J, Goldstein BA, Stirrups K, Konig IR, Cazier JB, Johansson A, Hall AS, Lee JY, Willer CJ, Chambers JC, Esko T, Folkersen L, Goel A, Grundberg E, Havulinna AS, Ho WK, Hopewell JC, Eriksson N, Kleber ME, Kristiansson K, Lundmark P, Lyytikainen LP, Rafelt S, Shungin D, Strawbridge RJ, Thorleifsson G, Tikkanen E, Van Zuydam N, Voight BF, Waite LL, Zhang W, Ziegler A, Absher D, Altshuler D, Balmforth AJ, Barroso I, Braund PS, Burgdorf C, Claudi-Boehm S, Cox D, Dimitriou M, Do R, Consortium D, Consortium C, Doney AS, El Mokhtari N, Eriksson P, Fischer K, Fontanillas P, Franco-Cereceda A, Gigante B, Groop L, Gustafsson S, Hager J, Hallmans G, Han BG, Hunt SE, Kang HM, Illig T, Kessler T, Knowles JW, Kolovou G, Kuusisto J, Langenberg C, Langford C, Leander K, Lokki ML, Lundmark A, McCarthy MI, Meisinger C, Melander O, Mihailov E, Maouche S, Morris AD, Muller-Nurasyid M, Mu TC, Nikus K, Peden JF, Rayner NW, Rasheed A, Rosinger S, Rubin D, Rumpf MP, Schafer A, Sivananthan M, Song C, Stewart AF, Tan ST, Thorgeirsson G, van der Schoot CE, Wagner PJ, Wellcome Trust Case Control C, Wells GA, Wild PS, Yang TP, Amouyel P, Arveiler D, Basart H, Boehnke M, Boerwinkle E, Brambilla P, Cambien F, Cupples AL, de Faire U, Dehghan A, Diemert P, Epstein SE, Evans A, Ferrario MM, Ferrieres J, Gauguier D, Go AS, Goodall AH, Gudnason V, Hazen SL, Holm H, Iribarren C, Jang Y, Kahonen M, Kee F, Kim HS, Klopp N, Koenig W, Kratzer W, Kuulasmaa K, Laakso M, Laaksonen R, Lee JY, Lind L, Ouwehand WH, Parish S, Park JE, Pedersen NL, Peters A, Quertermous T, Rader DJ, Salomaa V, Schadt E, Shah SH, Sinisalo J, Stark K, Stefansson K, Tregouet DA, Virtamo J, Wallentin L, Wareham N, Zimmermann ME, Nieminen MS, Hengstenberg C, Sandhu MS, Pastinen T, Syvanen AC, Hovingh GK, Dedoussis G, Franks PW, Lehtimaki T, Metspalu A, Zalloua PA, Siegbahn A, Schreiber S, Ripatti S, Blankenberg SS, Perola M, Clarke R, Boehm BO, O'Donnell C, Reilly MP, Marz W, Collins R, Kathiresan S, Hamsten A, Kooner JS, Thorsteinsdottir U, Danesh J, Palmer CN, Roberts R, Watkins H, Schunkert H, Samani NJ. Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet. 2013;45(1):25-33.
  122. Cai DP, He YM, Yang XJ, Zhao X, Xu HF. Lipoprotein (a) is a risk factor for coronary artery disease in Chinese Han ethnic population modified by some traditional risk factors: A cross-sectional study of 3462 cases and 6125 controls. Clin Chim Acta. 2015;451(Pt B):278-286.
  123. Pare G, Caku A, McQueen M, Anand SS, Enas E, Clarke R, Boffa MB, Koschinsky M, Wang X, Yusuf S, Investigators I. Lipoprotein(a) Levels and the Risk of Myocardial Infarction Among 7 Ethnic Groups. Circulation.2019;139(12):1472-1482.
  124. Welsh P, Welsh C, Celis-Morales CA, Brown R, Ho FK, Ferguson LD, Mark PB, Lewsey J, Gray SR, Lyall DM, Gill JMR, Pell JP, de Lemos JA, Willeit P, Sattar N. Lipoprotein(a) and cardiovascular disease: prediction, attributable risk fraction, and estimating benefits from novel interventions. Eur J Prev Cardiol. 2021;28(18):1991-2000.
  125. Khera AV, Everett BM, Caulfield MP, Hantash FM, Wohlgemuth J, Ridker PM, Mora S. Lipoprotein(a) concentrations, rosuvastatin therapy, and residual vascular risk: an analysis from the JUPITER Trial (Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin). Circulation. 2014;129(6):635-642.
  126. 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(12):1483-1492.
  127. Bittner VA, Szarek M, Aylward PE, Bhatt DL, Diaz R, Edelberg JM, Fras Z, Goodman SG, Halvorsen S, Hanotin C, Harrington RA, Jukema JW, Loizeau V, Moriarty PM, Moryusef A, Pordy R, Roe MT, Sinnaeve P, Tsimikas S, Vogel R, White HD, Zahger D, Zeiher AM, Steg PG, Schwartz GG, Committees OO, Investigators. Effect of Alirocumab on Lipoprotein(a) and Cardiovascular Risk After Acute Coronary Syndrome. J Am Coll Cardiol. 2020;75(2):133-144.
  128. Mehta A, Vasquez N, Ayers CR, Patel J, Hooda A, Khera A, Blumenthal RS, Shapiro MD, Rodriguez CJ, Tsai MY, Sperling LS, Virani SS, Blaha MJ, Joshi PH. Independent Association of Lipoprotein(a) and Coronary Artery Calcification With Atherosclerotic Cardiovascular Risk. J Am Coll Cardiol. 2022;79(8):757-768.
  129. Kaiser Y, Daghem M, Tzolos E, Meah MN, Doris MK, Moss AJ, Kwiecinski J, Kroon J, Nurmohamed NS, van der Harst P, Adamson PD, Williams MC, Dey D, Newby DE, Stroes ESG, Zheng KH, Dweck MR. Association of Lipoprotein(a) With Atherosclerotic Plaque Progression. J Am Coll Cardiol. 2022;79(3):223-233.
  130. Langsted A, Nordestgaard BG, Kamstrup PR. Elevated Lipoprotein(a) and Risk of Ischemic Stroke. J Am Coll Cardiol. 2019;74(1):54-66.
  131. Kamstrup PR, Nordestgaard BG. Elevated Lipoprotein(a) Levels, LPA Risk Genotypes, and Increased Risk of Heart Failure in the General Population. JACC Heart Fail. 2016;4(1):78-87.
  132. Seed M, Hoppichler F, Reaveley D, McCarthy S, Thompson GR, Boerwinkle E, Utermann G. Relation of serum lipoprotein(a) concentration and apolipoprotein(a) phenotype to coronary heart disease in patients with familial hypercholesterolemia. N Engl J Med. 1990;322(21):1494-1499.
  133. Holmes DT, Schick BA, Humphries KH, Frohlich J. Lipoprotein(a) is an independent risk factor for cardiovascular disease in heterozygous familial hypercholesterolemia. Clin Chem. 2005;51(11):2067-2073.
  134. Bajaj A, Damrauer SM, Anderson AH, Xie D, Budoff MJ, Go AS, He J, Lash JP, Ojo A, Post WS, Rahman M, Reilly MP, Saleheen D, Townsend RR, Chen J, Rader DJ, Chronic Renal Insufficiency Cohort Study I. Lipoprotein(a) and risk of myocardial infarction and death in chronic kidney disease: findings from the CRIC study (Chronic Renal Insufficiency Cohort). Arterioscler Thromb Vasc Biol. 2017;37(10):1971-1978.
  135. Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Genetic evidence that lipoprotein(a) associates with atherosclerotic stenosis rather than venous thrombosis. Arterioscler Thromb Vasc Biol. 2012;32(7):1732-1741.
  136. Helgadottir A, Gretarsdottir S, Thorleifsson G, Holm H, Patel RS, Gudnason T, Jones GT, van Rij AM, Eapen DJ, Baas AF, Tregouet DA, Morange PE, Emmerich J, Lindblad B, Gottsater A, Kiemeny LA, Lindholt JS, Sakalihasan N, Ferrell RE, Carey DJ, Elmore JR, Tsao PS, Grarup N, Jorgensen T, Witte DR, Hansen T, Pedersen O, Pola R, Gaetani E, Magnadottir HB, Wijmenga C, Tromp G, Ronkainen A, Ruigrok YM, Blankensteijn JD, Mueller T, Wells PS, Corral J, Soria JM, Souto JC, Peden JF, Jalilzadeh S, Mayosi BM, Keavney B, Strawbridge RJ, Sabater-Lleal M, Gertow K, Baldassarre D, Nyyssonen K, Rauramaa R, Smit AJ, Mannarino E, Giral P, Tremoli E, de Faire U, Humphries SE, Hamsten A, Haraldsdottir V, Olafsson I, Magnusson MK, Samani NJ, Levey AI, Markus HS, Kostulas K, Dichgans M, Berger K, Kuhlenbaumer G, Ringelstein EB, Stoll M, Seedorf U, Rothwell PM, Powell JT, Kuivaniemi H, Onundarson PT, Valdimarsson E, Matthiasson SE, Gudbjartsson DF, Thorgeirsson G, Quyyumi AA, Watkins H, Farrall M, Thorsteinsdottir U, Stefansson K. Apolipoprotein(a) genetic sequence variants associated with systemic atherosclerosis and coronary atherosclerotic burden but not with venous thromboembolism. J Am Coll Cardiol. 2012;60(8):722-729.
  137. Kenet G, Lutkhoff LK, Albisetti M, Bernard T, Bonduel M, Brandao L, Chabrier S, Chan A, deVeber G, Fiedler B, Fullerton HJ, Goldenberg NA, Grabowski E, Gunther G, Heller C, Holzhauer S, Iorio A, Journeycake J, Junker R, Kirkham FJ, Kurnik K, Lynch JK, Male C, Manco-Johnson M, Mesters R, Monagle P, van Ommen CH, Raffini L, Rostasy K, Simioni P, Strater RD, Young G, Nowak-Gottl U. Impact of thrombophilia on risk of arterial ischemic stroke or cerebral sinovenous thrombosis in neonates and children: a systematic review and meta-analysis of observational studies. Circulation. 2010;121(16):1838-1847.
  138. Stewart BF, Siscovick D, Lind BK, Gardin JM, Gottdiener JS, Smith VE, Kitzman DW, Otto CM. Clinical factors associated with calcific aortic valve disease. Cardiovascular Health Study. J Am Coll Cardiol. 1997;29(3):630-634.
  139. Gotoh T, Kuroda T, Yamasawa M, Nishinaga M, Mitsuhashi T, Seino Y, Nagoh N, Kayaba K, Yamada S, Matsuo H, et al. Correlation between lipoprotein(a) and aortic valve sclerosis assessed by echocardiography (the JMS Cardiac Echo and Cohort Study). Am J Cardiol. 1995;76(12):928-932.
  140. Cairns BJ, Coffey S, Travis RC, Prendergast B, Green J, Engert JC, Lathrop M, Thanassoulis G, Clarke R. A Replicated, Genome-Wide Significant Association of Aortic Stenosis With a Genetic Variant for Lipoprotein(a): Meta-Analysis of Published and Novel Data. Circulation. 2017;135(12):1181-1183.
  141. Arsenault BJ, Boekholdt SM, Dube MP, Rheaume E, Wareham NJ, Khaw KT, Sandhu MS, Tardif JC. Lipoprotein(a) levels, genotype, and incident aortic valve stenosis: a prospective Mendelian randomization study and replication in a case-control cohort. Circ Cardiovasc Genet. 2014;7(3):304-310.
  142. Kamstrup PR, Tybjaerg-Hansen A, Nordestgaard BG. Elevated lipoprotein(a) and risk of aortic valve stenosis in the general population. J Am Coll Cardiol. 2014;63(5):470-477.
  143. Capoulade R, Chan KL, Yeang C, Mathieu P, Bosse Y, Dumesnil JG, Tam JW, Teo KK, Mahmut A, Yang X, Witztum JL, Arsenault BJ, Despres JP, Pibarot P, Tsimikas S. Oxidized Phospholipids, Lipoprotein(a), and Progression of Calcific Aortic Valve Stenosis. J Am Coll Cardiol. 2015;66(11):1236-1246.
  144. Kamstrup PR, Hung MY, Witztum JL, Tsimikas S, Nordestgaard BG. Oxidized Phospholipids and Risk of Calcific Aortic Valve Disease: The Copenhagen General Population Study. Arterioscler Thromb Vasc Biol.2017;37(8):1570-1578.
  145. Liu Q, Yu Y, Xi R, Li J, Lai R, Wang T, Fan Y, Zhang Z, Xu H, Ju J. Association Between Lipoprotein(a) and Calcific Aortic Valve Disease: A Systematic Review and Meta-Analysis. Front Cardiovasc Med. 2022;9:877140.
  146. Pantelidis P, Oikonomou E, Lampsas S, Zakynthinos GE, Lysandrou A, Kalogeras K, Katsianos E, Theofilis P, Siasos G, Vavuranakis MA, Antonopoulos AS, Tousoulis D, Vavouranakis M. Lipoprotein(a) and calcific aortic valve disease initiation and progression: a systematic review and meta-analysis. Cardiovasc Res. 2023;119(8):1641-1655.
  147. Zheng KH, Tsimikas S, Pawade T, Kroon J, Jenkins WSA, Doris MK, White AC, Timmers N, Hjortnaes J, Rogers MA, Aikawa E, Arsenault BJ, Witztum JL, Newby DE, Koschinsky ML, Fayad ZA, Stroes ESG, Boekholdt SM, Dweck MR. Lipoprotein(a) and Oxidized Phospholipids Promote Valve Calcification in Patients With Aortic Stenosis. J Am Coll Cardiol. 2019;73(17):2150-2162.
  148. Perez de Isla L, Watts GF, Alonso R, Diaz-Diaz JL, Muniz-Grijalvo O, Zambon D, Fuentes F, de Andres R, Padro T, Lopez-Miranda J, Mata P. Lipoprotein(a), LDL-cholesterol, and hypertension: predictors of the need for aortic valve replacement in familial hypercholesterolaemia. Eur Heart J. 2021;42(22):2201-2211.
  149. Capoulade R, Yeang C, Chan KL, Pibarot P, Tsimikas S. Association of Mild to Moderate Aortic Valve Stenosis Progression With Higher Lipoprotein(a) and Oxidized Phospholipid Levels: Secondary Analysis of a Randomized Clinical Trial. JAMA Cardiol. 2018;3(12):1212-1217.
  150. Emdin CA, Khera AV, Natarajan P, Klarin D, Won HH, Peloso GM, Stitziel NO, Nomura A, Zekavat SM, Bick AG, Gupta N, Asselta R, Duga S, Merlini PA, Correa A, Kessler T, Wilson JG, Bown MJ, Hall AS, Braund PS, Samani NJ, Schunkert H, Marrugat J, Elosua R, McPherson R, Farrall M, Watkins H, Willer C, Abecasis GR, Felix JF, Vasan RS, Lander E, Rader DJ, Danesh J, Ardissino D, Gabriel S, Saleheen D, Kathiresan S, Consortium CH-HF, Consortium CAE. Phenotypic Characterization of Genetically Lowered Human Lipoprotein(a) Levels. J Am Coll Cardiol. 2016;68(25):2761-2772.
  151. Bourgeois R, Devillers R, Perrot N, Despres AA, Boulanger MC, Mitchell PL, Guertin J, Couture P, Boffa MB, Scipione CA, Pibarot P, Koschinsky ML, Mathieu P, Arsenault BJ. Interaction of Autotaxin With Lipoprotein(a) in Patients With Calcific Aortic Valve Stenosis. JACC Basic Transl Sci. 2020;5(9):888-897.
  152. Mathieu P, Boulanger MC. Autotaxin and Lipoprotein Metabolism in Calcific Aortic Valve Disease. Front Cardiovasc Med. 2019;6:18.
  153. 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: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2019;73(24):3168-3209.
  154. Wilson DP, Jacobson TA, Jones PH, Koschinsky ML, McNeal CJ, Nordestgaard BG, Orringer CE. Use of Lipoprotein(a) in clinical practice: A biomarker whose time has come. A scientific statement from the National Lipid Association. J Clin Lipidol. 2019;13(3):374-392.
  155. Pearson GJ, Thanassoulis G, Anderson TJ, Barry AR, Couture P, Dayan N, Francis GA, Genest J, Gregoire J, Grover SA, Gupta M, Hegele RA, Lau D, Leiter LA, Leung AA, Lonn E, Mancini GBJ, Manjoo P, McPherson R, Ngui D, Piche ME, Poirier P, Sievenpiper J, Stone J, Ward R, Wray W. 2021 Canadian Cardiovascular Society Guidelines for the Management of Dyslipidemia for the Prevention of Cardiovascular Disease in Adults. Can J Cardiol. 2021;37(8):1129-1150.
  156. Li JJ, Ma CS, Zhao D, Yan XW, Beijing Heart S, Expert C. Lipoprotein(a) and Cardiovascular Disease in Chinese Population: A Beijing Heart Society Expert Scientific Statement. JACC Asia. 2022;2(6):653-665.
  157. Kronenberg F, Mora S, Stroes ESG. Consensus and guidelines on lipoprotein(a) - seeing the forest through the trees. Curr Opin Lipidol. 2022;33(6):342-352.
  158. 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(1):111-188.
  159. Anderson TJ, Gregoire J, Pearson GJ, Barry AR, Couture P, Dawes M, Francis GA, Genest J, Jr., Grover S, Gupta M, Hegele RA, Lau DC, Leiter LA, Lonn E, Mancini GB, McPherson R, Ngui D, Poirier P, Sievenpiper JL, Stone JA, Thanassoulis G, Ward R. 2016 Canadian Cardiovascular Society Guidelines for the Management of Dyslipidemia for the Prevention of Cardiovascular Disease in the Adult. Can J Cardiol. 2016;32(11):1263-1282.
  160. Joint committee for guideline r. 2016 Chinese guidelines for the management of dyslipidemia in adults. J Geriatr Cardiol. 2018;15(1):1-29.
  161. Guan W, Cao J, Steffen BT, Post WS, Stein JH, Tattersall MC, Kaufman JD, McConnell JP, Hoefner DM, Warnick R, Tsai MY. Race is a key variable in assigning lipoprotein(a) cutoff values for coronary heart disease risk assessment: the Multi-Ethnic Study of Atherosclerosis. Arterioscler Thromb Vasc Biol. 2015;35(4):996-1001.
  162. Ding L, Song A, Dai M, Xu M, Sun W, Xu B, Sun J, Wang T, Xu Y, Lu J, Wang W, Bi Y, Ning G. Serum lipoprotein (a) concentrations are inversely associated with T2D, prediabetes, and insulin resistance in a middle-aged and elderly Chinese population. J Lipid Res. 2015;56(4):920-926.
  163. Schwartz GG, Ballantyne CM. Existing and emerging strategies to lower Lipoprotein(a). Atherosclerosis.2022;349:110-122.
  164. Burgess S, Ference BA, Staley JR, Freitag DF, Mason AM, Nielsen SF, Willeit P, Young R, Surendran P, Karthikeyan S, Bolton TR, Peters JE, Kamstrup PR, Tybjaerg-Hansen A, Benn M, Langsted A, Schnohr P, Vedel-Krogh S, Kobylecki CJ, Ford I, Packard C, Trompet S, Jukema JW, Sattar N, Di Angelantonio E, Saleheen D, Howson JMM, Nordestgaard BG, Butterworth AS, Danesh J, European Prospective Investigation Into C, Nutrition-Cardiovascular Disease C. Association of LPA Variants With Risk of Coronary Disease and the Implications for Lipoprotein(a)-Lowering Therapies: A Mendelian Randomization Analysis. JAMA Cardiol. 2018;3(7):619-627.
  165. Lamina C, Kronenberg F, Lp GC. Estimation of the Required Lipoprotein(a)-Lowering Therapeutic Effect Size for Reduction in Coronary Heart Disease Outcomes: A Mendelian Randomization Analysis. JAMA Cardiol.2019;4(6):575-579.
  166. Kronenberg F. Therapeutic lowering of lipoprotein(a): How much is enough? Atherosclerosis. 2019;288:163-165.
  167. Madsen CM, Kamstrup PR, Langsted A, Varbo A, Nordestgaard BG. Lipoprotein(a)-Lowering by 50 mg/dL (105 nmol/L) May Be Needed to Reduce Cardiovascular Disease 20% in Secondary Prevention: A Population-Based Study. Arterioscler Thromb Vasc Biol. 2020;40(1):255-266.
  168. de Boer LM, Oorthuys AOJ, Wiegman A, Langendam MW, Kroon J, Spijker R, Zwinderman AH, Hutten BA. Statin therapy and lipoprotein(a) levels: a systematic review and meta-analysis. Eur J Prev Cardiol. 2022;29(5):779-792.
  169. Willeit P, Ridker PM, Nestel PJ, Simes J, Tonkin AM, Pedersen TR, Schwartz GG, Olsson AG, Colhoun HM, Kronenberg F, Drechsler C, Wanner C, Mora S, Lesogor A, Tsimikas S. Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: individual patient-data meta-analysis of statin outcome trials. Lancet. 2018;392(10155):1311-1320.
  170. Leiter LA, Teoh H, Kallend D, Wright RS, Landmesser U, Wijngaard PLJ, Kastelein JJP, Ray KK. Inclisiran Lowers LDL-C and PCSK9 Irrespective of Diabetes Status: The ORION-1 Randomized Clinical Trial. Diabetes Care.2019;42(1):173-176.
  171. Bergmark BA, O'Donoghue ML, Murphy SA, Kuder JF, Ezhov MV, Ceska R, Gouni-Berthold I, Jensen HK, Tokgozoglu SL, Mach F, Huber K, Gaciong Z, Lewis BS, Schiele F, Jukema JW, Pedersen TR, Giugliano RP, Sabatine MS. An exploratory analysis of proprotein convertase subtilisin/kexin type 9 inhibition and aortic stenosis in the FOURIER trial. JAMA Cardiol. 2020;5(6):709-713.
  172. Chemello K, Chan DC, Lambert G, Watts GF. Recent advances in demystifying the metabolism of lipoprotein(a). Atherosclerosis. 2022;349:82-91.
  173. Santos RD, Raal FJ, Catapano AL, Witztum JL, Steinhagen-Thiessen E, Tsimikas S. Mipomersen, an antisense oligonucleotide to apolipoprotein B-100, reduces lipoprotein(a) in various populations with hypercholesterolemia: results of 4 phase III trials. Arterioscler Thromb Vasc Biol. 2015;35(3):689-699.
  174. Waldmann E, Parhofer KG. Lipoprotein apheresis to treat elevated lipoprotein (a). J Lipid Res. 2016;57(10):1751-1757.
  175. Schettler VJJ, Neumann CL, Peter C, Zimmermann T, Julius U, Roeseler E, Heigl F, Grutzmacher P, Blume H, Vogt A, Scientific Board of GftGAWG. The German Lipoprotein Apheresis Registry (GLAR) - almost 5 years on. Clin Res Cardiol Suppl. 2017;12(Suppl 1):44-49.
  176. Julius U, Tselmin S, Schatz U, Fischer S, Birkenfeld AL, Bornstein SR. Actual situation of lipoprotein apheresis in patients with elevated lipoprotein(a) levels. Atheroscler Suppl. 2019;40:1-7.
  177. Jaeger BR, Richter Y, Nagel D, Heigl F, Vogt A, Roeseler E, Parhofer K, Ramlow W, Koch M, Utermann G, Labarrere CA, Seidel D, Group of Clinical I. Longitudinal cohort study on the effectiveness of lipid apheresis treatment to reduce high lipoprotein(a) levels and prevent major adverse coronary events. Nat Clin Pract Cardiovasc Med. 2009;6(3):229-239.
  178. Leebmann J, Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, Breitenberger P, Maerz W, Lehmacher W, Heibges A, Klingel R, ProLiFe Study G. Lipoprotein apheresis in patients with maximally tolerated lipid-lowering therapy, lipoprotein(a)-hyperlipoproteinemia, and progressive cardiovascular disease: prospective observational multicenter study. Circulation. 2013;128(24):2567-2576.
  179. Roeseler E, Julius U, Heigl F, Spitthoever R, Heutling D, Breitenberger P, Leebmann J, Lehmacher W, Kamstrup PR, Nordestgaard BG, Maerz W, Noureen A, Schmidt K, Kronenberg F, Heibges A, Klingel R, ProLiFe-Study G. Lipoprotein Apheresis for Lipoprotein(a)-Associated Cardiovascular Disease: Prospective 5 Years of Follow-Up and Apolipoprotein(a) Characterization. Arterioscler Thromb Vasc Biol. 2016;36(9):2019-2027.
  180. Moriarty PM, Gray JV, Gorby LK. Lipoprotein apheresis for lipoprotein(a) and cardiovascular disease. J Clin Lipidol. 2019;13(6):894-900.
  181. Perrot N, Verbeek R, Sandhu M, Boekholdt SM, Hovingh GK, Wareham NJ, Khaw KT, Arsenault BJ. Ideal cardiovascular health influences cardiovascular disease risk associated with high lipoprotein(a) levels and genotype: The EPIC-Norfolk prospective population study. Atherosclerosis. 2017;256:47-52.
  182. Bhatia HS. Aspirin and lipoprotein(a) in primary prevention. Curr Opin Lipidol. 2023;34(5):214-220.
  183. Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, Tardif JC, Baum SJ, Steinhagen-Thiessen E, Shapiro MD, Stroes ES, Moriarty PM, Nordestgaard BG, Xia S, Guerriero J, Viney NJ, O'Dea L, Witztum JL, Investigators AK-A-LS. Lipoprotein(a) Reduction in Persons with Cardiovascular Disease. N Engl J Med. 2020;382(3):244-255.
  184. O'Donoghue ML, Rosenson RS, Gencer B, Lopez JAG, Lepor NE, Baum SJ, Stout E, Gaudet D, Knusel B, Kuder JF, Ran X, Murphy SA, Wang H, Wu Y, Kassahun H, Sabatine MS, Investigators O-DT. Small Interfering RNA to Reduce Lipoprotein(a) in Cardiovascular Disease. N Engl J Med. 2022;387(20):1855-1864.
  185. Nissen SE, Wolski K, Balog C, Swerdlow DI, Scrimgeour AC, Rambaran C, Wilson RJ, Boyce M, Ray KK, Cho L, Watts GF, Koren M, Turner T, Stroes ES, Melgaard C, Campion GV. Single Ascending Dose Study of a Short Interfering RNA Targeting Lipoprotein(a) Production in Individuals With Elevated Plasma Lipoprotein(a) Levels. JAMA. 2022;327(17):1679-1687.
  186. Nicholls SJ, Nissen SE, Fleming C, Urva S, Suico J, Berg PH, Linnebjerg H, Ruotolo G, Turner PK, Michael LF. Muvalaplin, an Oral Small Molecule Inhibitor of Lipoprotein(a) Formation: A Randomized Clinical Trial. JAMA.2023;330(11):1042-1053.