Archives

Adrenal Androgens and Aging

ABSTRACT

                                                                                         

Dehydroepiandrosterone (DHEA) and its metabolite DHEA sulfate (DHEAS), are steroid pre-hormones synthesized and secreted primarily by the zona reticularis of the adrenal cortex in response to adrenocorticotropic hormone (ACTH). They are both precursor hormones that may be transformed into weak androgens or estrogens. During the last decades, several epidemiologic and cohort studies have shown the age-related circulating levels of DHEA/DHEAS; these first increase in childhood, a process called “adrenarche”, peak in the 3rd decade of life, and progressively decrease in midlife, a phenomenon called “adrenopause”.  Some authors have linked obesity in childhood with early adrenarche, i.e., increased circulating levels of adrenal androgens; others have associated low levels in late life with increased frailty and all-cause mortality. The potential clinical and therapeutic roles of DHEA/DHEAS have been studied extensively, but the data remain controversial and largely inconclusive. In this chapter, we provide an overview of the physiology and pathophysiology of adrenal androgen synthesis, secretion, and action and present current evidence regarding their efficacy in the management of adrenal insufficiency or aging-related disorders.

 

INTRODUCTION

 

Dehydroepiandrosterone (DHEA) and its metabolite DHEA sulfate (DHEAS), are steroid hormones synthesized and secreted primarily by the zona reticularis of the adrenal cortex in response to adrenocorticotropic hormone (ACTH). They exert weak androgenic effects and are therefore considered precursor hormones that need to be transformed to potent androgens or estrogens to exert their effects. The potential clinical roles of DHEA/DHEAS have been studied extensively, as previous epidemiologic and prospective studies associated the age-related decrease of DHEA/DHEAS levels with higher prevalence of degenerative disorders and increased frailty and mortality from all causes in the elderly, attributing to adrenal androgens anti-aging properties. But do they really suggest that they are hormones related to longevity or just another pointless alchemy against aging? This chapter summarizes the physiology and pathophysiology of adrenal androgen synthesis, secretion, and action and provides current evidence regarding their efficacy in the management of aging-related disorders.

 

THE ADRENAL ANDROGENS

 

The Adrenal Cortex; Embryology and Normal Structure

 

The adrenal cortex is derived from the mesoderm lining the posterior abdominal wall. The fetal cortex begins its development in the 5-week-old fetus. At 2 months of gestation, it is already identifiable as a separate organ and is composed of the inner fetal zone (85% of the cortex) and the outer permanent definitive zone. The anatomic relation of the fetal and definitive zones is maintained during gestation; at birth the adrenal glands are 10–20 times larger than the adult gland, relative to kilograms of body weight. After birth, the fetal zone undergoes rapid involution resulting in a rapid decrease of adrenocortical weight in the 3 months following birth. During the next 3 years, the adult adrenal cortex develops from cells of the outer layer of the cortex and differentiates into the three adult zones, the subcapsular zona glomerulosa, the zona fasciculate, which is the thickest zone (70% of the cortex), and the inner zona reticularis.

 

Biosynthesis of Adrenal Androgens

 

The adrenal cortex produces many steroid hormones among which the major ones are cortisol, aldosterone, and the adrenal androgens. The subcapsular zona glomerulosa produces aldosterone while the inner two zones fasciculata and reticularis appear to function as a unit and produce cortisol, androgens, and small amounts of estrogens under the regulatory effect of ACTH and maybe of some other factors produced within the adrenal gland, including neurotransmitters, neuropeptides, and nitric oxide. The biosynthetic pathway of the adrenal androgens is shown below (Fig. 1).

 

Figure 1. Steroid biosynthesis in the adrenal cortex.

Quantitatively, the most abundantly produced adrenal androgens are dehydroepiandrosterone (DHEA) and its sulphated form dehydroepiandrosterone sulphate (DHEAS); the latter is the most abundantly produced adrenal steroid. It also has a long half-life and provides a stable pool of circulating DHEA. The ovaries also synthesize DHEA; however, they lack the enzyme DHEA-sulphotransferase so that DHEAS is almost exclusively synthesized and secreted by the adrenals. DHEA is further metabolized to androstenedione (1,2), which may in turn be aromatized to estrone. Whether the adrenals may also produce small amounts of testosterone by further metabolism of androstenedione is controversial (3). Although DHEA and DHEAS are secreted in greater quantities, androstenedione is qualitatively more important since it is more readily converted to testosterone in peripheral tissues. Roughly, the relative androgenic potency of DHEA, androstenedione, testosterone, and dihydrotestosterone (DHT) are 5:10:100:300, respectively. As ACTH is the main regulator of adrenal androgen production in adults, both DHEA and androstenedione exhibit circadian periodicity in concert with ACTH and cortisol and their plasma concentrations increase rapidly following ACTH administration; also, they are suppressed by glucocorticoid administration. Because of its slow metabolic clearance, DHEAS does not exhibit diurnal rhythm variation.

 

Circulation of Adrenal Androgens

 

The adrenal androgens are secreted in an unbound state. Soon after their release in the circulation they bind to plasma proteins, chiefly to albumin (90%). Androstenedione, DHEA, and DHEAS circulate weakly bound to albumin, while testosterone is bound extensively to the sex hormone binding globulin (SHBG). Bound steroids are biologically inactive; the unbound steroids are free to interact with target cells either to exert their effects or to be transformed into inactive or active metabolites.

 

Metabolism of Adrenal Androgens; Gender-Dependent Synthesis Of DHEA/DHEAS

 

Due to lack or only minor inherent steroidogenic activity, adrenal androgens are precursor hormones (pro-hormones) that need to be transformed to potent androgens or estrogens to exert their effects (4,5). Their transformation into active sex steroids depends upon the level of expression of the various steroidogenic and metabolizing enzymes in each cell type which allows all androgen-sensitive and estrogen-sensitive tissues to have some control over the local levels of sex steroids according to their needs (6). Active androgens and estrogens thus synthesized exert their activity in the target cells with little diffusion, resulting in low levels in the general circulation. This intracrine mechanism serves to eliminate the exposure of other tissues to androgens or estrogens, minimizing unwanted side effects (4,7-9).

 

In males with normal gonadal function, the conversion of adrenal androgens to testosterone accounts for less than 5% of the total amount of this hormone, and thus the physiologic effect is negligible. In females of reproductive age, the adrenal contribution to total androgen production is more important; during the follicular phase, the adrenal precursors account for 2/3 of total testosterone production and 1/2 of DHT production. During midcycle, the ovarian contribution increases, and the adrenal precursors account for only 40% of testosterone production.

 

Apart from their peripheral conversion to more potent androgens, the adrenal androgens may be also aromatized to estrogens or undergo degradation and inactivation (4,5) (Fig 2). In more detail, DHEA is readily converted within the adrenal gland to DHEAS. DHEA secreted by the adrenal glands and the ovaries is also converted to DHEAS by the liver and the kidneys or it may be converted to Δ4-androstenedione. The adrenally produced DHEAS may be excreted without further metabolism or it may further undergo limited conversion to DHEA. Both DHEAS and DHEA may be metabolized to 7alpha- and 16alpha-hydroxylated derivatives and by 17β reduction to Δ5-Androstenediol and its sulfate. Androstenedione is converted either to testosterone or by reduction of its 4,5 double bonds to etiocholanolone or androsterone, which may be further converted by 17 alpha reductions to etiocholanediol and androstanediol, respectively. Testosterone is converted to DHT in androgen-sensitive tissues by 5 alpha reduction and it in turn is mainly metabolized by 3 alpha reductions to androstanediol. The metabolites of these androgens are conjugated either as glucuronides or sulfates and excreted in the urine. Regarding aromatization to estrogens, it was shown that not only androstenedione and testosterone, but also DHEA, may be converted to estrogens in peripheral tissues such as brain, bone, breast, and ovaries (6,10); this might be of importance, especially in women during the menopausal transition (see below) (11,12).

Figure 2. Metabolism of adrenal androgens; 3BHSD, 3β-hydroxysteroid dehydrogenase isozymes; 17BHSD, 17β -hydroxysteroid dehydrogenase isozymes; 5aRed, 5α -reductase isozymesP450 aromatase, steroid sulfatase, STS.

Age-Dependent Synthesis of DHEA/DHEAS

 

Fetal DHEA and DHEAS fall rapidly after birth and remain low until adrenarche; they then start rising again and peak during the third decade of life after which the serum levels of DHEA and DHEAS progressively decline with advancing age by around 2–5% per year (10,13), so that by menopause the DHEA level has decreased by 60% (14), and by 80-90% of the peak production by the eighth or ninth decade of life (15,16). This decline has been termed “adrenopause”, however, cortisol secretion does not decline with age or may even increase (16,17). Adrenopause is independent of menopause and occurs in both sexes as a gradual process at similar ages. A decrease in 17,20-lyase activity may be responsible for the progressive diminution of DHEA and DHEA-S with advancing age (18,19), although other mechanisms, such as a reduction in the mass of the zona reticularis (20) or a decrease in IGF-I and IGF-II have also been proposed (21). Recent study by Heaney et al. in accordance with previous research found that older subjects exhibited lower plasma and saliva DHEA levels overall, while with increasing age, the DHEA area under the curve was attenuated and the slope of decline became less steep (17,22).

 

Although DHEAS concentration does not vary throughout the day, DHEA secretion exhibits a diurnal rhythm like that of cortisol. Studies have indicated that DHEA secretion is reduced in the morning period resulting in a flatter diurnal rhythm among the oldest old, in contrast to cortisol which remains stable or even increases in the morning (17,23). The above diurnal rhythms of cortisol and DHEA, lead to an elevated cortisol: DHEA ratio, which is most pronounced in the morning period.

 

The age-related decline in DHEA/DHEAS levels shows high inter-individual variability (20). There is a clear sex difference in DHEA/DHEAS concentrations with lower DHEAS concentrations in adult women compared to men (24), while there is also a clear genetic component predetermining circulating DHEA/DHEAS. Notably, data from the largest population-based twin study to estimate the genetic and environmental contributions of diurnal DHEAS concentrations demonstrated that salivary DHEAS is a heritable measure, with genetic effects accounting for 37%–46% of the total variance for the late morning and afternoon age-adjusted measures (25).

 

Since DHEA is the main source of androgens in women, its age-related decline leads to a corresponding decrease in the total androgen pool. Although there is no defined level of androgen below which women can be said to be deficient, the decline of DHEA in postmenopausal women would suggest they are “deficient” in both estrogens and androgens (14). The declining circulating levels of adrenal androgens with advancing age have been related to clinical symptoms and disorders (see below).

 

In the last few years, the concept that adrenal androgen production gradually declines with advancing age has changed following the analysis of the longitudinal data collected in the Study of Women’s Health Across the Nation (SWAN) (26). When the annual serum levels of DHEAS were aligned according to ovarian status, it was recognized that despite the overall age-related decline in DHEAS, in most women (85% of those studied) the adrenal androgen production rose during the menopausal transition, starting in the early peri-menopause and continuing into the early post-menopause. The DHEAS rise was attributed to the adrenals and not the ovaries, as a similar rise was also observed in intact and ovariectomized women (27); the gender-related rise of adrenal DHEAS and the time course of that rise that returns to a progressive decline following menopause, implies ovarian influences over adrenal steroidogenesis (28). Considering previous failure to adequately attribute phenotype, symptoms, and health trajectories to the observed longitudinal changes in circulating estradiol and progesterone (29), the perimenopausal rise in adrenal androgens could potentially suggest a more important role of these hormones in the occurrence of symptoms during the menopausal transition (30). The observational, epidemiologic, and interventional studies addressing this hypothesis are analyzed below. Some conditions and diseases, like poor life quality, satisfaction and psychosocial, as well as acute stress, severe chronic systemic diseases, anorexia nervosa, Cushing syndrome and chronic administration of glucocorticoids are associated with lower levels of DHEA and DHEAS. Hyperprolactinemia is associated with elevated levels of DHEAS (31,32).

 

Biologic Effects of Adrenal Androgens; Cellular and Molecular Actions

 

ROLE AS PRO-HORMONES

 

DHEA possesses pleiotropic effects. Epidemiologic and prospective studies have associated the decline of circulating levels of androgens with the development and progression of degenerative disorders. The exact mechanism of action and clinical role of DHEA and DHEAS, if any, remain unclear. Due to lack or only minor inherent steroidogenic activity, the adrenal androgens need to be transformed to potent androgens or estrogens to exert their effects on peripheral tissues. Recent data suggest additional direct actions of the adrenal androgens further to those exerted through the androgen and estrogen receptors (see below).

 

The principal biologic effects of the adrenal androgens typically seen during adrenarche consist mainly of pubic and axillary hair growth and the change of sweat composition that produces adult body odor (33). During the reproductive years, in males with normal gonadal function, the adrenal androgens account for less than 5% of the daily production rate of testosterone and thus the physiologic effect is negligible. DHEA and DHEA-S levels have been shown to be associated with nutritional status. Obese children have higher levels of DHEA and/or DHEA-S and achieve adrenarche earlier than lean children. Indeed, a recent study showed that obese children with higher DHEAS concentrations at the age of seven years had more total and central adiposity and higher insulin than did nonobese children of the same age (34,35). Some research suggests that adrenal androgens directly or after peripheral conversion to estrogen modulate hypothalamic activity influencing the gonadarche. When produced in excess however, the adrenal androgens may have no clinical consequences in adult males or result in LH /FSH suppression and oligospermia/infertility. In boys, the adrenal androgen excess is associated with clinical manifestations including premature penile enlargement, early development of secondary sexual characteristics, premature closure of the epiphyseal growth plates and short final height. In females the excessive production of adrenal steroids as seen in Cushing syndrome, adrenal carcinoma, and congenital adrenal hyperplasia via peripheral conversion to testosterone and eventually to DHT result in acne, hirsutism, and menstrual/fertility defects or even virilization in more severe cases. 

 

MEMBRANE ASSOCIATED DHEA RECEPTORS

 

Further to their effect via the estrogen and androgen receptors, recent data support direct actions of DHEA through specific G protein-coupled membrane receptors in bovine aortic and primary human umbilical vein endothelial cells (HUVECs) (36-38) through which DHEA activates the endothelial NO synthetase (eNOS) (eNOS/cGMP pathway) [38] and increases the production of nitric oxide (NO), a key modulator of vascular function, by endothelial cells. Such receptors are also seen in the kidney, heart, and liver but at lower level than that in bovine aortic endothelial cells (39) as well as in pulmonary artery smooth muscle cells (PASMCs), where DHEA inhibits voltage-dependent T type Ca-channels (40). In systemic circulation, a plasma membrane receptor has been suggested in the anti-remodeling action of DHEA involving inhibition of the Akt/GSK-3β signaling pathway (41). Other studies have shown inhibitory effect of DHEA on proliferation and apoptosis of endothelial and vascular smooth muscle cells independently of both estrogen and androgen receptors (42,43). The above suggest the presence of a membrane-associated DHEA specific receptor; the molecular structure of this receptor remains to be elucidated.

 

CYTOSOLIC NUCLEAR RECEPTORS

 

Steroid action involves cytosolic/nuclear hormone receptors (44); thus, most of the studies looking at the mechanism(s) responsible for DHEA action focused on such receptors (45). However, since DHEA can be metabolized into androgens/estrogens, it is not always easy to determine whether DHEA exerts its effects directly through the estrogen/androgen receptors or after conversion to these hormones. There is some new evidence showing that DHEA and some of its metabolites either bind to or activate nuclear receptors such as pregnane X receptor, constitutive androstanol receptor, estrogen receptor-β, and peroxisome proliferators activated receptors (46-49). Through the activation of peroxisome proliferator-activated receptor alpha for example, DHEA inhibits the activation of nuclear factor-κB and the secretion of interleukin-6 and interleukin-12, through which DHEA exerts anti-inflammatory effects (50,51). 7α-and 7β-hydroxylated derivatives of DHEA also seem to have direct effects on nuclear receptors, but their physiological function is not clear (39). Finally, DHEA inhibits apoptosis and promotes proliferation of osteoblasts in rats through MAPK signaling pathways, independently from androgens and estrogens (52); this action could be beneficial for preservation of bone mass and reduction of fracture risk.

 

ENDOPLASMIC RETICULUM RECEPTOR SIGMA 1 RECEPTOR

 

More recently, it has been suggested that DHEA is an agonist of sigma-1 receptor (Sigma-1R) expressed in the endoplasmic reticulum of the heart, kidney, liver, and brain (53,54). Under physiological conditions, the sigma-1 receptor chaperones the functional inositol 1,4,5 trisphosphate receptor at the endoplasmic reticulum participating in the calcium signaling pathway (53,55). Animal studies have shown that via sigma-1R, but also by Akt– eNOs signaling pathway stimulation, DHEA may improve cardiac function (56) and exert vasculo-protective effects (57). There is a great volume of data suggesting antioxidant properties of DHEA; overproduction of oxygen-free radicals (oxidative stress) upregulates inflammation and cellular proliferation and is believed to play a critical role in the development of cancer, atherosclerosis, and Alzheimer's disease, as well as the basic aging process (58-60). DHEA inhibits glucose-6-phosphate dehydrogenase (G-6-PDH) (61,62) and NADPH production. The decrease in NADPH levels results in reduced oxygen-free radical production via NADPH oxidase (62). Moreover, a study found that DHEA treatment of mice increased the number of Brd U-positive neurons co-expressing β-catenin, a downstream GSK-3β target, concluding that sigma-1 receptor stimulation by DHEA led to altered OBX-induced depressive-like behaviors by increasing neurogenesis in the dentate gyrus through activation of the Akt/GSK-3β/β-catenin pathway (63). Increased plasma DHEA and DHEAS have been demonstrated in post-traumatic stress disorder (PTSD), predicting symptom improvement and coping as well as resilience adaptation. The same study suggested that decreased cortisol/DHEA ratio was associated with severe childhood trauma and current symptoms (64).

 

In summary, DHEA mediates its action via transformation into androgen and estrogen derivatives acting through their specific receptors, but also via multiple (57,65) signaling pathways involving specific membrane, cytosolic/nuclear and endoplasmic reticulum receptors.

 

POTENTIAL TREATMENT BENEFITS  

 

Data from epidemiologic and prospective studies indicate an inverse relation between low circulating levels of DHEA and DHEA-S and a host of aging-associated pathologies such as sexual dysfunction, mood defects, and poor sense of well-being (27,28), as well as higher risk of hospital admission (66), poor muscle strength (67) and mobility (66,68), and higher prevalence of frailty (69), insulin resistance, obesity, cardiovascular disease (45) and mortality from cardiovascular disease (70). At the same time, a positive relation between higher levels of DHEA-S and better health and well-being was documented (71). Furthermore, animal (primarily rodent) studies have suggested many beneficial effects of DHEA treatment, including improved immune function and prevention of atherosclerosis, cancer, diabetes, and obesity. Therefore, the therapeutic role of DHEA replacement as an anti-aging factor for the prevention and/or treatment of the above conditions was studied; recent systematic reviews of the reports do not seem promising, however (72-78).

 

Treatment Modalities

 

DHEA is considered as a hormone in Europe and thus becomes available only by prescription, while in the United States it is considered as a nutritional supplement and is sold over the counter without a prescription. This difference has no scientific foundation and is mostly a matter of declaration. Most DHEA supplements are made in laboratories from a substance called diosgenin, a plant sterol found in soy and wild yams. DHEA supplements were taken off the U.S. market in 1985 because of their unproven safety and effectiveness, but were reintroduced as a dietary supplement after the Dietary Supplement Health and Education Act was passed in 1994. At present, questionable over-the-counter DHEA preparations lacking pharmacokinetic and pharmacodynamic data are widely used in the United States. There is no standard dosage of DHEA replacement; some studies have used between 25 and 200 milligrams a day, or sometimes even higher amounts. DHEA in current preparations has a long half-life (45), which allows a single intake a day. Target levels of DHEA are around the middle of normal range for healthy young subjects, measured in a blood sample 24 hr after the last intake (79).

 

The adrenal androgens are mainly thought to act as prohormones and exert at least part of their action via conversion to androgens and/or estrogens. Previous studies have shown that the end- products of DHEA supplementation depend on the patient’s gender, with a non-symmetrical transformation of DHEA favoring androgens in women and estrogens in men (72,80,81). The above refer to oral administration of DHEA supplements; percutaneous administration of DHEA seems to provoke similar increases in both estrogens and androgens in the two genders (82).

 

Adrenal Insufficiency

 

Adrenal insufficiency, despite supplementation of glucocorticoids, has been associated with decreased quality of life when compared to a healthy population (79,83,84). DHEA supplementation has been suggested as an accessory treatment to conventional adrenal replacement therapy with glucocorticoids and mineralocorticoids. The exact physiological roles of DHEA still remain unclear and the routine therapy of individuals with adrenal insufficiency is still controversial. Some authors reported significant improvements of mood, well-being, sexual thoughts, libido, interest, and satisfaction following DHEA replacement, particularly in females (79,83-85). Other analyses of DHEA administration in women with primary and secondary adrenal insufficiency have resulted in inconsistent and unreproducible results (85). Recently, the supplementation of DHEA was suggested in women with adrenal insufficiency and low libido, depressive symptoms, or low energy levels despite optimal glucocorticoid and mineralocorticoid replacement (86).

 

LOW DHEA/DHEAS LEVELS AND ASSOCIATED COMORBIDITIES

 

DHEA And Musculoskeletal Disorders

 

The increasing incidence of fractures with advancing age has been related, among other factors, with the aging-related reduced muscle mass and strength, that increase the propensity for falling (87). A body of evidence exists on the effect of circulating DHEA/DHEAS on various markers of strength and physical function in older individuals. Studies in elderly individuals support a positive relation between DHEA blood levels and muscle mass (67), muscle strength (67,88), and mobility (68), as well as better self-reported (89) and objectively assessed physical function (90), and measured peak volume of oxygen consumed per minute (91) in elderly with higher DHEA/DHEAS concentrations. In this direction, higher DHEAS levels were associated with increased bone mass density (BMD) in both men (92) and post-menopausal women and inversely related to risk for falls (93). Finally, low DHEAS levels have been associated with a higher prevalence of frailty, a geriatric syndrome of loss of reserve characterized by weight loss, fatigue, weakness, and vulnerability to adverse events (69,94), and low back pain in both genders and slow rehabilitation of low-back pain in women (71,95,96).

 

Reports from interventional studies support a therapeutic role of DHEA replacement in aging-associated musculoskeletal defects. For example, DHEA exerted positive effects on muscle strength, body composition (97-99), and physical performance (100), as well as on bone mass density (BMD) in both lumbar spine and the hip (15,72,98,101-105) when administered to post-menopausal women and elderly people over a period of 52 weeks. The above positive effects on musculoskeletal system were attributed to the DHEA-related increase of insulin-like growth factor-1 (IGF-1) levels (97,106) and bioavailability (decrease of insulin growth factor binding protein-1 [IGFBP-1]) (106) in both men and women and/or to the increase of androgen levels mostly in women (97,106,107). Some other data also suggest aromatase activity of primary human osteoblasts converting DHEA to estrone (108), while it was shown in vitro that DHEA inhibits apoptosis and promotes proliferation of rat osteoblasts through MAPK signaling pathways, independently from androgen and estrogen effects (52). The above support a positive effect of DHEA on bone through conversion to estrogens, but also independently from its hormonal end-products. Other studies, however, failed to show a beneficial effect of DHEA supplementation on muscle function (109-111) or on BMD (99,100,112); of note all these studies were conducted over a shorter period (26 weeks only). Whether these conflicting data result from DHEA’s mild/moderate effect or from great differences between study designs, such as short duration of treatment and small number of participants, is difficult to say (73). Overall, the effect of DHEA supplementation on BMD is small in relation to other treatments for bone loss, and no fracture data are available. Therefore, its therapeutic utility in rehabilitation and/or fracture/frailty prevention and treatment protocols for older patients remains unclear.

 

A recent systematic review (73) of the literature (72,83,97,100,113-116) concluded that overall, the benefit (113,116) of DHEA on muscle strength and physical function in older adults remains inconclusive. Some measures of muscle strength may improve, although DHEA does not appear to routinely benefit measures of physical function or performance. Therefore, consensus has not been reached. Further large clinical trials are necessary to better identify the clinical role of DHEA supplementation in this population.

 

DHEA, Well-Being and Sexual Function

 

If DHEA’s effects on musculoskeletal disorders are inconclusive, its utility for the management of ageing-related poor sense of well-being and sexual dysfunction is a question for top puzzle solvers. What we know so far from epidemiologic studies is that sexual function problems are common among women and increase with increasing age (117-119). The sex steroid hormones estrogens and androgens seem to play an important role in the sexual life of women; androgens affect the reusability, pleasure, and intensity of orgasm in women and are particularly implicated in the neurovascular smooth muscle response of swelling and lubrication, whereas estrogens contribute to vulval and vaginal congestive response and affect mood and sexual responses (120). Conditions such as menopausal symptoms, loss of libido, vulvovaginal atrophy-related sexual dysfunction, and poor sense of well-being seen in menopausal and peri-menopausal women were related to the age-associated decline in sex steroids (121). Furthermore, interventional studies in postmenopausal women with estrogens have shown much improvement on vaginal atrophy and vasomotor symptoms (121-124); there is also much clinical evidence for the efficacy of testosterone treatment for low sexual function in women (119,125-129).

 

Given that a) the adrenal steroids are the most abundant sex steroids in post-menopausal women and provide a large reservoir of precursors for the intracellular production of androgens and estrogens in non-reproductive tissues, b) DHEA levels decline with age, c) pre- and post-menopausal women with lower sexual responsiveness have lower levels of serum DHEAS (130) and d) treatment of postmenopausal women with estrogen and testosterone have shown some improvement in sexual function, it was proposed that restoring the circulating levels of DHEA to those found in young women may improve sexual function and well-being in postmenopausal women (131). Some early randomized trials that suffered from methodological issues, such as small number of participants, short treatment duration, and supraphysiological doses, demonstrated positive effects of DHEA replacement on sexual function and well-being (15,130,132-134), as well as on relief of menopausal symptoms (134-136). Similarly, women with adrenal insufficiency treated with oral DHEA replacement demonstrated significant improvement in overall well-being, as well as in frequency of sexual thoughts, sexual interest, and satisfaction (80,84). Other studies, however, failed to show any benefit of DHEA replacement on sexual function, well-being, and menopausal symptoms in peri- and post-menopausal women (74,75,106,137,138) and women with adrenal insufficiency (85,139,140). A recent review of the available data concluded that current evidence does not support the routine use of DHEA in women with adrenal insufficiency (76). Furthermore, the more recent placebo-controlled randomized trials that are of superior design compared to the early trials, as they use validated measures of sexual function, have larger sample sizes, and are of longer duration, failed to document any significant benefit of oral DHEA therapy on well-being or sexual function in women (72-75,77). It has been hypothesized that the efficacy of DHEA to improve sexual function might be dependent on the route of its administration. In women, androgens and estrogens are produced from DHEA in the vagina tissue. As vaginal atrophy and dryness are common symptoms of estrogen deficiency during menopause, causing dyspareunia and sexual dysfunction (141), a possible benefit that emerged is that vaginally administered DHEA may improve the postmenopausal vaginal atrophy-related sexual dysfunction (142) without increasing the circulating levels of estrogen above the postmenopausal range (80,142-144). Despite initial promising, beneficial effects on sexual function, again, even with intravaginally administered DHEA, a study failed to show significant benefits (77).

 

In men lower circulating levels of DHEA was related to erectile dysfunction. A double-blind, placebo-controlled study that enrolled men with erectile dysfunction treated with oral DHEA 50 mg daily has shown some promise for improving sexual performance in men who had low DHEA blood levels (145). However, high-quality studies have demonstrated inconsistent results regarding DHEA supplementation for improving sexual function, libido, and erectile dysfunction. Although research in this area is promising, additional well-designed studies are required.

 

DHEA And Mood Disorders

 

The prevalence of depression increases in cohorts of the elderly and has been independently related to high morbidity and mortality (146). In the central nervous system, DHEA is considered a neurosteroid with a wide range of functions. Animal studies demonstrated several DHEA-modulated neurotransmitters, including dopamine, glutamate, and c-amino butyric acid (39), as well as DHEA-induced increased activity of 5-hydroxytryptamine (5-HT) neurons (147), providing the cellular basis for a potential antidepressant effect of DHEA. Furthermore, typical neuroleptic-like effects of DHEA were displayed in animal models of schizophrenia suggesting potential role of DHEA replacement in the treatment of schizophrenia (148).

 

Previous studies suggested a strong relation between low levels of DHEA/DHEA-S and major depression in children and adolescents (149), as well as adults and the elderly (150,151). On the contrary, higher DHEA-S levels were positively associated with depressive symptoms during the menopausal transition (152) and depression in patients with major depression (153,154); whether the elevated DHEA-S levels in the above studies represent increased adrenal activity that could explain the depressive symptoms is not clear, as cortisol was not measured. Moreover, successful treatment of depression was followed by reductions in both DHEA-S (153-155) and DHEA levels (155), making the relation between DHEA/DHEAS and depression even more confusing.

 

Several interventional studies have shown that DHEA replacement may improve negative and depressive symptoms (132,133,156-158). In women with adrenal insufficiency, oral DHEA replacement significantly improved the overall well-being, as well as scores for depression and anxiety (110); similar results were found in the management of the negative symptoms of schizophrenia (158). Recent placebo-controlled randomized trials, however, failed to demonstrate a beneficial effect of DHEA therapy on mood, quality of life, perceptions of physical and emotional health, and life satisfaction in postmenopausal women (72,74,75). However, recent data have suggested that increased circulating DHEA(S) levels may predict SSRI-associated remission in major depression (159). Thus, the therapeutic role of DHEA on mood disorders remains unclear.

 

DHEA and Psychosocial Stress

 

It has long been suggested that long-term psychosocial stress may cause or contribute to different diseases and symptoms, including atherosclerosis (160), coronary heart disease (161) and acute coronary events (162), as well as accelerated aging (163,164). Whether DHEA/DHEAS levels are related to psychological stress or not is still debatable. Exposure to prolonged psychosocial stress has been related to reduced (165-167) or elevated levels of DHEA/DHEA-S (168), while some other studies failed to show any clear association in any direction (169,170). A recent study by Lennartsson et al. demonstrated that DHEA and DHEA-S levels are markedly lower in individuals that report perceived stress at work than in individuals who report no perceived stress at work (171). Whether this is of clinical importance is not clear.

 

 

The incidence of dementia increases exponentially with increasing age in both men and women (172). The number of elderly people nowadays is the fastest growing segment of the population, which means the related personal, social, and economic burdens of dementia are extremely high and could increase dramatically over the next few decades. Therefore, effective prevention/treatment of neurodegenerative disorders is imperative. It has been proposed that DHEA and DHEAS may exert neuroprotective effects in the brain mainly through DHEA-dependent neural stem cell stimulation, genomic activity modulation, and upregulation of androgen receptor levels (173,174), as well as via the DHEA-induced inhibition of pro-inflammatory factor production, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) (39) that are involved in the pathogenesis of the amyloid plaques of Alzheimer disease (175). Higher serum levels of DHEAS have been related to more favorable cognitive function in older people, such as better concentration and working memory (176,177) and higher scores on the Mini Mental State Examination (178). In this direction, low DHEA/DHEAS levels in particular brain regions were thought to play a role in the development of Parkinson disease, which is the second most common neurodegenerative disorder, just behind Alzheimer (179), while DHEA administration showed some beneficial effect in a primate model of Parkinson disease (180,181). Inverse relations between DHEAS levels in saliva (181) and circulation (176) and some domains of memory impairment were also documented, supporting the hypothesis that DHEA supplementation may improve cognition in the elderly; yet solid evidence of associations between the endogenous levels of these steroids and measures of cognitive function is lacking.

 

No studies with DHEA replacement, either acute administration or chronic (up to 12 months) supplementation, have shown a benefit in cognitive function in healthy elderly populations (74,137,181-185). Furthermore, DHEA supplementation failed to show any benefit in patients with Alzheimer disease (186) and had only minimal beneficial effect on specific cognitive domains such as the verbal fluency in older women with mild to moderate cognitive impairment (187). Remarkably, some other studies have shown a negative effect of DHEAS replacement on cognitive performance (183,188,189). It should be noted however, that most studies included only small groups of patients and were up to a yearlong, which is probably not enough time to address the potential role of DHEA / DHEAS in neurodegenerative disorders.

 

DHEA and Metabolism

 

LIPIDS

 

In women, the effects of sex steroids on lipid profile differ according to the steroid treatment (estrogen or androgen) and to the route of administration. Thus, oral methyltestosterone lowers high-density lipoprotein (HDL)-cholesterol (190), and oral estrogen increases HDL-cholesterol and triglycerides and lowers low-density lipoprotein (LDL)-cholesterol and total cholesterol (191,192), while transdermal estradiol and transdermal testosterone have little or no effect on lipids (193). Combined oral estrogen and methyltestosterone is associated with lowering of HDL-cholesterol (194,195). Considering that DHEA can be converted intracellularly to estrogens and androgens, the effect on the lipid profile could be mixed and may vary between individuals. Most of the recent well-designed studies, addressing this issue report no association or even an adverse association (at least in women) (196,197) between plasma levels of DHEA (198,199) or DHEA administration (97,109,192,200,201) and the lipid profile.

 

BODY MASS INDEX (BMI)

 

Animal studies support a beneficial effect of DHEA administration on obesity (202-204). In humans, two sets of longitudinal analyses of studies with women in menopausal transition showed that elevated DHEAS level is negatively related to BMI (27,28). On the other hand, baseline analyses by Santoro et al [209] did not find much association between DHEAS and BMI, waist-hip ratio, or waist. Childhood obesity is associated with higher levels of DHEAS (34). Similarly, a 2-year, placebo-controlled, randomized, double-blind study involving elderly men and women with low levels of DHEAS, showed no significant effect of DHEA replacement (75 mg per day orally) on body composition measurements (72). Interestingly, a, recent meta-analysis of intervention studies showed that DHEA supplementation in elderly men can induce only a small positive effect on body composition which is strictly dependent on DHEA conversion into its bioactive metabolites such as androgens or estrogens (205). Putting together these results, current data regarding DHEA effect on BMI contradict each other, and its usage in clinical practice for body weight management is not suggested or recommended at the present.

 

INSULIN RESISTANCE

 

DHEA may at least theoretically improve endothelial function (43), and ameliorate local/systemic inflammation (50,51) and oxidative stress (58-60,206). These effects in association with DHEAS’s inverse relation with body mass index (BMI) (23,27,28,207) would most probably suggest beneficial effect of DHEA/DHEAS supplement on insulin sensitivity (207). This hypothesis was confirmed by reports from animal studies in which DHEA replacement had a beneficial effect on insulin sensitivity (202,203). In human studies, however, the results are rather inconsistent. In some studies, the lower levels of DHEA seen with aging have been associated with impaired glucose tolerance, insulin resistance, and diabetes (208-211), while in another (212) exactly the opposite relation was shown as higher levels of DHEA were associated with impaired glucose tolerance and diabetes mellitus in post-menopausal women. The truth regarding DHEA/DHEAS and insulin resistance and its associated conditions gets even more complicated considering conflicting results from interventional studies with DHEA replacement. Thus, an ameliorating effect of long-term treatment with DHEA on insulin resistance was described in a group of middle-aged hypo-adrenal women treated with DHEA (213), but also in groups of elderly men (214) and postmenopausal women (213-217) replaced with DHEA. The DHEA dose used ranged between 25 and 100 mg/day oally and the duration of treatment varied between 3 and 12 months; in one study transdermal DHEA was used (217). Most other interventional studies addressing this issue, failed to demonstrate any significant effect of DHEA on insulin resistance/sensitivity (72,97,110,112,113,139) and so did a recent review of the available data regarding use of DHEA in women with adrenal insufficiency (76). Remarkably, Mortola and Yen (84) reported worsening insulin resistance with DHEA replacement in postmenopausal women; in this study however, the number of participants was small (n=6), the duration of treatment short (28 days), and the DHEA dosage supraphysiological (1600 mg/day orally). Putting together the above, the relation between DHEA and carbohydrate metabolism is still uncertain.

 

DHEA and Cardiovascular Disease (CVD)

 

CVD represents a serious public health problem; its prevalence increases with advancing age (218). Low androgen levels have been related to atherogenic profile in men (219,220), while data from acute coronary units have shown a transient fall of the testosterone levels in the first 24 hours after myocardial infarction (MI) (221,222), which probably deprives these patients of testosterone’s pro-fibrinolytic activity (223-225) and may actually result in increased 30-day mortality rate following acute MI (226); the above findings suggest a strong relation between sex steroid hormones and CVD morbidity and mortality. Many studies have previously documented a significant inverse relation between low DHEA/DHEAS levels and key elements involved in the development of atherosclerosis and CVD, including carotid artery intima-media thickness (IMT) (227,228), oxidative stress (58,59), and endothelial dysfunction (229), independent of other coronary risk factors. Low DHEAS levels (201,230-233) were also predictive of severe coronary atherosclerosis on coronary angiography (234), but also of earlier cardiac allograft vasculopathy development in heart transplant patients (235).

 

These findings are suggestive of anti-atherogenic and cardioprotective effect of DHEA/DHEAS. Numerous epidemiological studies have, therefore, looked at the specific relation between plasma levels of adrenal androgens and CVD. Most have shown that low plasma levels of DHEA/DHEA-S were clearly associated with increased incidence of atherosclerotic vascular diseases (91,234,236-238) and cardiovascular morbidity (234,239-245), independently from classic cardiovascular risk factors, as well as with increased CVD-related mortality in elderly men but not in postmenopausal women, unless they had pre-existing coronary disease (16,70,199,241,246-248).

 

The plasma levels of DHEA were also inversely associated with the progression (249) and prognosis of heart failure (250), at least in men. The exact pathophysiologic background is still more or less unclear. Some preliminary data in patients with type 2 diabetes mellitus suggest that the adrenal androgens may increase the generation of activated protein C, an important anticoagulant protein that protects from acute coronary events (228). Furthermore, DHEA may directly stimulate eNOS phosphorylation/activation in endothelial cells and NO production (36,39,251), which in turn induces vasodilation, and preserves myocardial perfusion (252). DHEA may also exert anti-inflammatory actions (39,253), through which it may alleviate endothelial dysfunction, atherogenesis (254), and the acute thrombotic complications of atheroma (39,253,255-258) enhanced by systemic inflammation. The protective effects of DHEA on endothelium were also shown in several in vitro studies in which DHEA increased endothelial proliferation (43) and protected endothelial cells against apoptosis (59,259). Finally, DHEA can alleviate oxidative stress and inflammation in vascular smooth muscle cells (VSMCs) via ERK1/2 and NF-κB signaling pathways, although it has no effect on their phenotype transition (260).

 

Other studies, however, have failed to show a significant relation between DHEA/DHEA-S and CVD. In men for example, myocardial infarction occurrence was not altered by DHEA-S levels, and acute myocardial infarctions were seen in patients with either low or high DHEA-S levels (261-264). Similarly in women, lower DHEA-S levels in ischemic heart disease patients versus control were observed in some studies, but not in others (238,265,266). The reasons that account for the discrepancies among the above studies are not clear. It can be argued that smoking could be a possible confounding variable for both DHEA-S levels and CVD, as smoking increases DHEA-S levels but also increases the incidence of adverse cardiovascular events (267,268). The discrepancies among the above studies may also be attributed to population variability; for example, in the study by Mazat et al. the relative risk of an 8-year mortality associated with low DHEA-S was 3.4 times higher in males under 70 years compared to older men (odds ratio of 6.5 versus 1.9) (16). Finally, DHEA-S was checked just once in some retrospective studies, often several years before the adverse cardiovascular events (269).

 

Whether exogenously administered DHEA could ameliorate key elements involved in the generation and progression of the atherosclerotic process was addressed in humans with atherosclerosis and experimental animal models. The human studies have shown a beneficial effect of DHEA on angiographic evidence of atherosclerosis and improvement of vascular endothelial function (43,234,270). Several animal studies have also clearly demonstrated the inhibitory effect of orally administered DHEA on atherosclerosis and plaque progression (271,272) as well as beneficial effects on ischemia–reperfusion injury in the microcirculation (273,274) and cardiac dysfunction (56,57). Arterial stiffness, which is also considered a risk factor for CVD, significantly improved after DHEA replacement in both elderly men and women (275,276). Whether the above findings could be translated into DHEA administration in clinical practice for the reduction of CVD morbidity and/or mortality is not well documented and supported by current reports. However, since DHEA is a well-tolerated molecule and an inexpensive drug, additional large multi-centric clinical studies could address its role in the prevention and/or management of CVD.

 

DHEA and Cerebrovascular Disease

 

Stroke is the third-leading cause for disability worldwide (277); therefore, early risk stratification for an optimized allocation of health care resources is imperative. The ischemic strokes that account for the great majority of all stroke cases (87 percent) occur as a result of acute obstruction of atherosclerotic blood vessels supplying blood to the brain (278). Considering DHEAS has neuroprotective and antiatherosclerotic properties (243,279,280) and its synthesis has been documented in the brain (175,281,282), the role of DHEA/DHEAS in acute stroke incidence and outcome was investigated. Interestingly, in women from Nurses’ Health Study, lower DHEAS levels were associated with a greater risk of ischemic stroke (283). In addition, it was suggested that DHEAS levels in the blood may predict the severity and functional outcome of acute strokes (284,285). Whether the above findings suggest baseline DHEAS levels could alter stroke management in clinical practice or whether DHEA replacement has a therapeutic potential role in stroke management need to be addressed.

 

DHEA and Pulmonary Hypertension

 

The previously described vasorelaxant properties of DHEA in systemic circulation were also investigated in pulmonary hypertension in animal models and in humans. Several studies have shown that DHEA replacement could effectively prevent and reverse hypoxic pulmonary hypertension, pulmonary arterial remodeling, and right ventricular hypertrophy in rats (286-288) in a dose-dependent manner (289) and also prevent the age-related frailty induced by hypoxic pulmonary hypertension in older mice (288). The effect of DHEA is selective to the pulmonary circulation since the systemic blood pressure was not altered. It was shown that the beneficial effects of DHEA on pulmonary hypertension were at least partly independent of its conversion to estrogen/testosterone and eNOS activation. Some of the potential molecular mechanism by which DHEA promotes pulmonary artery relaxation appears to involve K+ channel activation, upregulation of soluble guanylate cyclase (287,290,291), downregulation of hypoxia inducible factor 1a (HIF-1a) (292), and by NADPH oxidation-elicited subunit dimerization of protein kinase G 1α (293).

 

As previously discussed, DHEA may inhibit and reverse chronic hypoxia-induced pulmonary hypertension in rats. Little is known, however, about the effects of DHEA on the pulmonary circulation in humans. The levels of DHEA/DHEA-S in patients with pulmonary hypertension over time have not been determined, but the recent Multi-Ethnic Study of Atherosclerosis (MESA) - Right Ventricle (RV) Study found that higher DHEA levels were associated with increased RV mass and stroke volume in women (294). Another prospective study suggested a strong inverse correlation between natural DHEA/DHEA-S blood levels and the ten-year mortality in old male smokers and ex-smokers (16). Prompted by the experimental findings in the pulmonary circulation, a recent study investigated whether DHEA can improve the clinical and hemodynamic status of patients with pulmonary hypertension associated to chronic obstructive pulmonary disease; eight patients with the disease were treated with DHEA (200mg daily orally) for 3 months. The results were very promising as DHEA treatment significantly improved the pulmonary hemodynamics and the physical performance of the patients, without worsening gas exchange, as do other pharmacological treatments of pulmonary hypertension (295).

 

Putting together the above evidence, there are experimental data to support the beneficial role of DHEA treatment in models with pulmonary hypertension, but only a few studies supporting its beneficial effect in patients with pulmonary hypertension associated with chronic obstructive pulmonary disease. Further clinical studies would probably clarify its therapeutic role in the management of pulmonary hypertension in clinical practice.

 

DHEA and Autoimmune Disorders

 

INFLAMMATORY BOWEL DISEASE (IBD)

 

DHEA has anti-inflammatory properties (50,51). Its levels appear to be low in people with ulcerative colitis and Crohn’s disease, irrespective of the patient’s age (296,297). A phase II small pilot trial in patients with active inflammatory bowel disease refractory to other drugs, treated with 200 mg dehydroepiandrosterone per day orally for 56 days (298) showed that DHEA may decrease the clinical activity of the disease and may even cause a remission. More studies are needed before saying for sure whether DHEA helps IBD or not.

 

SYSTEMIC LUPUS ERYTHEMATOSUS (SLE)

 

Several randomized controlled clinical studies have reported that regardless of the patient’s age, taking DHEA (50-200mg/day) along with other medications improves quality of life for people with mild to moderate SLE, decreases corticosteroid requirements, and reduces the frequency of flare-ups, though it probably does not change the overall course of their disease (84,299-303). A study had shown DHEA replacement may increase bone mass in women with lupus (302). A 2007 report in the Cochrane Database of Systematic Reviews (78) suggests a "modest but clinically significant impact" of DHEA replacement on health-related quality of life in the short-term for people with SLE; the impact on disease activity was inconsistent. Long- term outcomes and safety remain unstudied.

 

RHEUMATOID ARTHRITIS (RA)

 

DHEA levels have been found to be low in people with rheumatoid arthritis (304,305) and decrease further with glucocorticoid therapy (306). Considering the well-demonstrated immune-suppressive activities exerted by the adrenal androgens and their derivatives (307-309), the utility of DHEA as potential therapy for management of male and female RA patients was studied. Preliminary data from animal studies showed benefits of DHEA treatment in collagen-induced arthritis (310-312). However, in carefully controlled human clinical trials, DHEA treatment produced only modest benefits (313), probably with the exception of female-treated RA patients who benefit the most by DHEA replacement (314). The noted discrepancy in benefits from DHEA treatment between animals and humans may be related to the low endogenous DHEA in rodents relative to humans because of low levels of cytochrome P450 17α-hydroxylase (175), but also because of different DHEA metabolism between species; remarkably, in rodents DHEA has many highly oxygenated metabolites and a surprisingly complex metabolism that results in production of a multitude highly oxygenated species that may exert the beneficial effects on arthritis (315).

 

DHEA AND ADVERSE HEALTH OUTCOMES

 

DHEA supplements are generally well tolerated in studies using oral or percutaneous administration, with daily doses ranging from 25 mg to 1,600 mg. DHEA is an important precursor for estrogen and androgen production. In women DHEA when administered orally is mainly converted to androgen metabolites. As a result, some minimal androgenic adverse effects have been reported, including mild acne, seborrhea, facial hair growth, and ankle swelling (45,75,315).

 

A hormonal etiology has long been suspected for breast and endometrial cancer as several risk factors for each cancer, such as obesity, nulliparity, and early menarche are hormonally related (78,316-318). The plasma concentrations of the adrenal androgens in premenopausal women were previously associated with higher risk for development of breast cancer (319-321). Furthermore, DHEA-S levels above a cut off limit predicted disease progression in hypoestrogenized women treated for breast cancer (322). On the other hand, in vitro studies support an inhibitory effect of DHEA on the growth of human mammary cancer cells and the growth of chemically-induced mammary cancer in rats (10,62,323). It was shown that the effect of DHEA in mammary tissue depends on the level of plasma estrogens. Thus, growth inhibition occurs only in the presence of high estrogen concentrations, and growth stimulation occurs in the presence of a low-level estrogen milieu (12,324). The exact role of DHEA supplementation on breast cancer in humans has not been fully studied. A previous review of clinical, epidemiological, and experimental studies suggests late promotion of breast cancer in postmenopausal women by prolonged intake of DHEA, especially if central obesity coexists, and suggests extra caution when DHEA supplements are used by obese postmenopausal women (325). A more recent review of the medical literature investigating DHEA physiology and randomized controlled trials of the use of DHEA in postmenopausal women, however, did not find any adverse effect of DHEA supplementation (31).

 

Unopposed estrogen is also known to be associated with an increased risk of endometrial carcinoma (316). DHEA supplementation did not increase the endometrial thickness in postmenopausal women treated with 25 mg/day DHEA orally for 6 months (136) or 50 mg daily for 12 months (135,136,326). In addition, DHEA administered percutaneously for 12 months to postmenopausal women was shown to have an estrogenic effect on the vagina without affecting the endometrium that remained atrophic (105).

 

In men, DHEA supplements are mainly transformed to estrogen metabolites but also to more potent androgens. As a result, concerns regarding the effect of DHEA supplementation on prostate were raised, especially after the finding that about 15% of DHT present in the prostate comes from DHEA metabolism (327). A 2-year, placebo-controlled, randomized, double blind study involving elderly men receiving DHEA did not show any adverse effects in prostate (72).

 

As long as long-term safety data for DHEA supplementation are lacking, the American Cancer Society advices caution in its use in people who have cancer, especially types of cancer that respond to hormones, such as certain types of breast cancer, prostate cancer, and endometrial cancer (328). The authors of a Cochrane Systematic Review regarding the supplementation of DHEA in peri- and post-menopausal women, questioned the effectiveness of DHEA in women, and concluded that the overall quality of the studies analyzed was moderate to low and that the study outcomes were inconsistent and could not be pooled to obtain an overall effect due to the diversity of the measurement methods employed (329).

 

CONCLUSIONS

 

Theoretically, supplementing a pre-hormone is extremely interesting as it would provide peripheral tissues with levels of sex steroids according to local needs and would eliminate the exposure of other tissues to androgens or estrogens, minimizing unwanted side effects. Therefore, DHEA administration is closer to ‘‘hormonal optimization’’ than hormonal supplementation. In older people, lower than normal levels of DHEA/DHEAS were previously related to aging-associated degenerative disorders, including metabolic and cardiovascular diseases, poor physical performance, mood and memory defects, sexual dysfunction, and poor sense of wellbeing. Whether this is just a statistical finding with no practical clinical meaning has been investigated by many interventional studies most of which, however, were of short duration and had a small number of participants. Without exception, all recent reviews of the available data regarding DHEA replacement utility for the management of aging-related disorders do not support its use in clinical practice (72-78); no significant adverse or negative side effects of DHEA were reported in clinical studies, but also no significant evidence that low levels of DHEA cause the aging-related degenerative disorders or that taking DHEA can help prevent/treat them. Thus, current clinical modalities with DHEA supplements do not comply with evidence-based medicine. Since there are several known biochemical actions by which DHEA could ameliorate disorders affecting the elderly population and is a well-tolerated molecule and an inexpensive drug, additional large multi-center clinical studies would probably give us a better understanding of its clinical utility in the management of aging-related disorders. Till then, we should probably reconsider suggesting patients to start on a pro-hormone that would help them only as much as a placebo would help.

 

REFERENCES

 

  1. Auchus RJ. Overview of dehydroepiandrosterone biosynthesis. Seminars in reproductive medicine.2004;22(4):281-288.
  2. Payne AH, Hales DB. Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones. Endocrine reviews. 2004;25(6):947-970.
  3. Davis SR. Androgens and female sexuality. The journal of gender-specific medicine : JGSM : the official journal of the Partnership for Women's Health at Columbia. 2000;3(1):36-40.
  4. Labrie F. Extragonadal synthesis of sex steroids: intracrinology. undefined. 2003.
  5. Labrie F, Bélanger A, Cusan L, Candas B. Physiological changes in dehydroepiandrosterone are not reflected by serum levels of active androgens and estrogens but of their metabolites: intracrinology. The Journal of clinical endocrinology and metabolism. 1997;82(8):2403-2409.
  6. Labrie F. Intracrinology. Molecular and cellular endocrinology. 1991;78(3).
  7. Labrie C, Belanger A, Labrie F. Androgenic activity of dehydroepiandrosterone and androstenedione in the rat ventral prostate. Endocrinology. 1988;123(3):1412-1417.
  8. Labrie F. Adrenal androgens and intracrinology. Semin Reprod Med. 2004;22(4):299-309.
  9. Labrie F. DHEA as physiological replacement therapy at menopause. Journal of endocrinological investigation.1998;21(6):399-401.
  10. Labrie F, Luu-The V, Labrie C, Belanger A, Simard J, Lin SX, Pelletier G. Endocrine and intracrine sources of androgens in women: inhibition of breast cancer and other roles of androgens and their precursor dehydroepiandrosterone. Endocr Rev. 2003;24(2):152-182.
  11. Poulin R, Labrie F. Stimulation of cell proliferation and estrogenic response by adrenal C19-delta 5-steroids in the ZR-75-1 human breast cancer cell line. Cancer Res. 1986;46(10):4933-4937.
  12. Seymour-Munn K, Adams J. Estrogenic effects of 5-androstene-3 beta, 17 beta-diol at physiological concentrations and its possible implication in the etiology of breast cancer. Endocrinology. 1983;112(2):486-491.
  13. Labrie F, Bélanger A, Cusan L, Gomez JL, Candas B. Marked decline in serum concentrations of adrenal C19 sex steroid precursors and conjugated androgen metabolites during aging. The Journal of clinical endocrinology and metabolism. 1997;82(8):2396-2402.
  14. Labrie F, Bélanger A, Bélanger P, Bérubé R, Martel C, Cusan L, Gomez J, Candas B, Castiel I, Chaussade V, Deloche C, Leclaire J. Androgen glucuronides, instead of testosterone, as the new markers of androgenic activity in women. The Journal of steroid biochemistry and molecular biology. 2006;99(4-5):182-188.
  15. Baulieu EE, Thomas G, Legrain S, Lahlou N, Roger M, Debuire B, Faucounau V, Girard L, Hervy MP, Latour F, Leaud MC, Mokrane A, Pitti-Ferrandi H, Trivalle C, De Lacharrière O, Nouveau S, Rakoto-Arison B, Souberbielle JC, Raison J, Le Bouc Y, Raynaud A, Girerd X, Forette F. Dehydroepiandrosterone (DHEA), DHEA sulfate, and aging: contribution of the DHEAge Study to a sociobiomedical issue. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(8):4279-4284.
  16. Mazat L, Lafont S, Berr C, Debuire B, Tessier JF, Dartigues JF, Baulieu EE. Prospective measurements of dehydroepiandrosterone sulfate in a cohort of elderly subjects: relationship to gender, subjective health, smoking habits, and 10-year mortality. Proceedings of the National Academy of Sciences of the United States of America.2001;98(14):8145-8150.
  17. Heaney JL, Phillips AC, Carroll D. Ageing, physical function, and the diurnal rhythms of cortisol and dehydroepiandrosterone. Psychoneuroendocrinology. 2012;37(3):341-349.
  18. Laughlin GA, Barrett-Connor E. Sexual Dimorphism in the Influence of Advanced Aging on Adrenal Hormone Levels: The Rancho Bernardo Study*. The Journal of Clinical Endocrinology & Metabolism Printed. 2000;85(10).
  19. Baulieu EE. Androgens and aging men. Molecular and Cellular Endocrinology. 2002;198(1-2):41-49.
  20. Parker CR. Dehydroepiandrosterone and dehydroepiandrosterone sulfate production in the human adrenal during development and aging. Steroids. 1999;64(9):640-647.
  21. Yen SSC, Laughlin GA. Aging and the adrenal cortex. Experimental Gerontology. 1998;33(7-8):897-910.
  22. Ahu RS, Lee YJ, Choi JY, Kwon HB, Chun SI. Salivary cortisol and DHEA levels in the Korean population: age-related differences, diurnal rhythm, and correlations with serum levels. Yonsei medical journal. 2007;48(3):379-388.
  23. Liu CH, Laughlin GA, Fischer UG, Yen SSC. Marked attenuation of ultradian and circadian rhythms of dehydroepiandrosterone in postmenopausal women: evidence for a reduced 17,20-desmolase enzymatic activity. The Journal of clinical endocrinology and metabolism. 1990;71(4):900-906.
  24. Orentreich N, Brind JL, Rizer RL, Vogelman JH. Age changes and sex differences in serum dehydroepiandrosterone sulfate concentrations throughout adulthood. The Journal of clinical endocrinology and metabolism. 1984;59(3):551-555.
  25. Prom-Wormley EC, York TP, Jacobson KC, Eaves LJ, Mendoza SP, Hellhammer D, Maninger N, Levine S, Lupien S, Lyons MJ, Hauger R, Xian H, Franz CE, Kremen WS. Genetic and environmental effects on diurnal dehydroepiandrosterone sulfate concentrations in middle-aged men. Psychoneuroendocrinology.2011;36(10):1441-1452.
  26. Lasley BL, Santoro N, Randolf JF, Gold EB, Crawford S, Weiss G, McConnell DS, Sowers MF. The relationship of circulating dehydroepiandrosterone, testosterone, and estradiol to stages of the menopausal transition and ethnicity. The Journal of clinical endocrinology and metabolism. 2002;87(8):3760-3767.
  27. Lasley BL, Crawford SL, Laughlin GA, Santoro N, McConnell DS, Crandall C, Greendale GA, Polotsky AJ, Vuga M. Circulating dehydroepiandrosterone sulfate levels in women who underwent bilateral salpingo-oophorectomy during the menopausal transition. Menopause (New York, NY). 2011;18(5):494-498.
  28. Crawford S, Santoro N, Laughlin GA, Sowers MF, McConnell D, Sutton-Tyrrell K, Weiss G, Vuga M, Randolph J, Lasley B. Circulating dehydroepiandrosterone sulfate concentrations during the menopausal transition. J Clin Endocrinol Metab. 2009;94(8):2945-2951.
  29. Randolph JF, Sowers M, Gold EB, Mohr BA, Luborsky J, Santoro N, McConnell DS, Finkelstein JS, Korenman SG, Matthews KA, Sternfeld B, Lasley BL. Reproductive hormones in the early menopausal transition: relationship to ethnicity, body size, and menopausal status. The Journal of clinical endocrinology and metabolism. 2003;88(4):1516-1522.
  30. Lasley BL, Crawford S, McConnell DS. Adrenal androgens and the menopausal transition. Obstetrics and gynecology clinics of North America. 2011;38(3):467-475.
  31. Davis SR, Panjari M, Stanczyk FZ. Clinical review: DHEA replacement for postmenopausal women. J Clin Endocrinol Metab. 2011;96(6):1642-1653.
  32. Pluchino N, Drakopoulos P, Bianchi-Demicheli F, Wenger JM, Petignat P, Genazzani AR. Neurobiology of DHEA and effects on sexuality, mood and cognition. J Steroid Biochem Mol Biol. 2015;145:273-280.
  33. Parker LN. Adrenarche. Endocrinologist. 1993;3(6):385-391.
  34. Corvalan C, Uauy R, Mericq V. Obesity is positively associated with dehydroepiandrosterone sulfate concentrations at 7 y in Chilean children of normal birth weight. Am J Clin Nutr. 2013;97(2):318-325.
  35. Rabijewski M, Papierska L, Binkowska M, Maksym R, Jankowska K, Skrzypulec-Plinta W, Zgliczynski W. Supplementation of dehydroepiandrosterone (DHEA) in pre- and postmenopausal women - position statement of expert panel of Polish Menopause and Andropause Society. Ginekol Pol. 2020;91(9):554-562.
  36. Liu D, Dillon JS. Dehydroepiandrosterone stimulates nitric oxide release in vascular endothelial cells: Evidence for a cell surface receptor. Steroids. 2004;69(4):279-289.
  37. Liu D, Dillon JS. Dehydroepiandrosterone activates endothelial cell nitric-oxide synthase by a specific plasma membrane receptor coupled to Galpha(i2,3). The Journal of biological chemistry. 2002;277(24):21379-21388.
  38. Simoncini T, Mannella P, Fornari L, Varone G, Caruso A, Genazzani AR. Dehydroepiandrosterone modulates endothelial nitric oxide synthesis via direct genomic and nongenomic mechanisms. Endocrinology.2003;144(8):3449-3455.
  39. Traish AM, Kang HP, Saad F, Guay AT. Dehydroepiandrosterone (DHEA)-A precursor steroid or an active hormone in human physiology (CME). Journal of Sexual Medicine. 2011;8(11):2960-2982.
  40. Chevalier M, Gilbert G, Lory P, Marthan R, Quignard JF, Savineau JP. Dehydroepiandrosterone (DHEA) inhibits voltage-gated T-type calcium channels. Biochemical pharmacology. 2012;83(11):1530-1539.
  41. Bonnet S, Paulin R, Sutendra G, Dromparis P, Roy M, Watson KO, Nagendran J, Haromy A, Dyck JRB, Michelakis ED. Dehydroepiandrosterone reverses systemic vascular remodeling through the inhibition of the Akt/GSK3-{beta}/NFAT axis. Circulation. 2009;120(13):1231-1240.
  42. Williams MRI, Ling S, Dawood T, Hashimura K, Dai A, Li H, Liu JP, Funder JW, Sudhir K, Komesaroff PA. Dehydroepiandrosterone inhibits human vascular smooth muscle cell proliferation independent of ARs and ERs. The Journal of clinical endocrinology and metabolism. 2002;87(1):176-181.
  43. Williams MRI, Dawood T, Ling S, Dai A, Lew R, Myles K, Funder JW, Sudhir K, Komesaroff PA. Dehydroepiandrosterone increases endothelial cell proliferation in vitro and improves endothelial function in vivo by mechanisms independent of androgen and estrogen receptors. The Journal of clinical endocrinology and metabolism. 2004;89(9):4708-4715.
  44. Levin ER. Integration of the extranuclear and nuclear actions of estrogen. Molecular Endocrinology.2005;19(8):1951-1959.
  45. Legrain S, Massien C, Lahlou N, Roger M, Debuire B, Diquet B, Chatellier G, Azizi M, Faucounau V, Porchet H, Forette F, Baulieu EE. Dehydroepiandrosterone replacement administration: Pharmacokinetic and pharmacodynamic studies in healthy elderly subjects. Journal of Clinical Endocrinology and Metabolism.2000;85(9):3208-3217.
  46. Moore LB, Parks DJ, Jones SA, Bledsoe RK, Consler TG, Stimmel JB, Goodwin B, Liddle C, Blanchard SG, Willson TM, Collins JL, Kliewer SA. Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands. Journal of Biological Chemistry. 2000;275(20):15122-15127.
  47. Kliewer SA, Goodwin B, Willson TM. The nuclear pregnane X receptor: a key regulator of xenobiotic metabolism. Endocrine reviews. 2002;23(5):687-702.
  48. Goodwin B, Redinbo MR, Kliewer SA. Regulation of CYP3A gene transcription by the pregnane X receptor. Annual Review of Pharmacology and Toxicology. 2002;42:1-23.
  49. Ripp SL, Fitzpatrick JL, Peters JM, Prough RA. Induction of CYP3A expression by dehydroepiandrosterone: involvement of the pregnane X receptor. Drug metabolism and disposition: the biological fate of chemicals.2002;30(5):570-575.
  50. Straub RH, Konecna L, Hrach S, Rothe G, Kreutz M, Schölmerich J, Falk W, Lang B. Serum dehydroepiandrosterone (DHEA) and DHEA sulfate are negatively correlated with serum interleukin-6 (IL-6), and DHEA inhibits IL-6 secretion from mononuclear cells in man in vitro: possible link between endocrinosenescence and immunosenescence. The Journal of clinical endocrinology and metabolism. 1998;83(6):2012-2017.
  51. Poynter ME, Daynes RA. Peroxisome proliferator-activated receptor alpha activation modulates cellular redox status, represses nuclear factor-kappaB signaling, and reduces inflammatory cytokine production in aging. The Journal of biological chemistry. 1998;273(49):32833-32841.
  52. Wang L, Wang YD, Wang WJ, Zhu Y, Li DJ. Dehydroepiandrosterone improves murine osteoblast growth and bone tissue morphometry via mitogen-activated protein kinase signaling pathway independent of either androgen receptor or estrogen receptor. Journal of molecular endocrinology. 2007;38(4):467-479.
  53. Maurice T, Grégoire C, Espallergues J. Neuro(active)steroids actions at the neuromodulatory sigma1 (sigma1) receptor: biochemical and physiological evidences, consequences in neuroprotection. Pharmacology, biochemistry, and behavior. 2006;84(4):581-597.
  54. Cheng ZX, Lan DM, Wu PY, Zhu YH, Dong Y, Ma L, Zheng P. Neurosteroid dehydroepiandrosterone sulphate inhibits persistent sodium currents in rat medial prefrontal cortex via activation of sigma-1 receptors. Experimental neurology. 2008;210(1):128-136.
  55. Tsai S-Y, Hayashi T, Mori T, Su T-P. Sigma-1 receptor chaperones and diseases. Central nervous system agents in medicinal chemistry. 2009;9(3):184-189.
  56. Tagashira H, Bhuiyan S, Shioda N, Fukunaga K. Distinct cardioprotective effects of 17β-estradiol and dehydroepiandrosterone on pressure overload-induced hypertrophy in ovariectomized female rats. Menopause (New York, NY). 2011;18(12):1317-1326.
  57. Bhuiyan MS, Tagashira H, Fukunaga K. Dehydroepiandrosterone-mediated stimulation of sigma-1 receptor activates Akt-eNOS signaling in the thoracic aorta of ovariectomized rats with abdominal aortic banding. Cardiovascular therapeutics. 2011;29(4):219-230.
  58. Camporez JP, Akamine EH, Davel AP, Franci CR, Rossoni LV, Carvalho CR. Dehydroepiandrosterone protects against oxidative stress-induced endothelial dysfunction in ovariectomized rats. J Physiol. 2011;589(Pt 10):2585-2596.
  59. Nheu L, Nazareth L, Xu GY, Xiao FY, Luo RZ, Komesaroff P, Ling S. Physiological effects of androgens on human vascular endothelial and smooth muscle cells in culture. Steroids. 2011;76(14):1590-1596.
  60. Jacob MH, Janner Dda R, Bello-Klein A, Llesuy SF, Ribeiro MF. Dehydroepiandrosterone modulates antioxidant enzymes and Akt signaling in healthy Wistar rat hearts. J Steroid Biochem Mol Biol. 2008;112(1-3):138-144.
  61. Levy HR, Daouk GH. Simultaneous analysis of NAD- and NADP-linked activities of dual nucleotide-specific dehydrogenases. Application to Leuconostoc mesenteroides glucose-6-phosphate dehydrogenase. J Biol Chem. 1979;254(11):4843-4847.
  62. Schwartz AG, Pashko LL. Dehydroepiandrosterone, glucose-6-phosphate dehydrogenase, and longevity. Ageing Res Rev. 2004;3(2):171-187.
  63. Moriguchi S, Shinoda Y, Yamamoto Y, Sasaki Y, Miyajima K, Tagashira H, Fukunaga K. Stimulation of the sigma-1 receptor by DHEA enhances synaptic efficacy and neurogenesis in the hippocampal dentate gyrus of olfactory bulbectomized mice. PLoS One. 2013;8(4):e60863.
  64. Yehuda R, Brand SR, Golier JA, Yang RK. Clinical correlates of DHEA associated with post-traumatic stress disorder. Acta Psychiatr Scand. 2006;114(3):187-193.
  65. Bhuiyan MS, Fukunaga K. Stimulation of Sigma-1 receptor by dehydroepiandrosterone ameliorates hypertension-induced kidney hypertrophy in ovariectomized rats. Experimental biology and medicine (Maywood, NJ). 2010;235(3):356-364.
  66. Forti P, Maltoni B, Olivelli V, Pirazzoli GL, Ravaglia G, Zoli M. Serum dehydroepiandrosterone sulfate and adverse health outcomes in older men and women. Rejuvenation Res. 2012;15(4):349-358.
  67. Valenti G, Denti L, Maggio M, Ceda G, Volpato S, Bandinelli S, Ceresini G, Cappola A, Guralnik JM, Ferrucci L. Effect of DHEAS on skeletal muscle over the life span: the InCHIANTI study. J Gerontol A Biol Sci Med Sci.2004;59(5):466-472.
  68. Ravaglia G, Forti P, Maioli F, Boschi F, Cicognani A, Bernardi M, Pratelli L, Pizzoferrato A, Porcu S, Gasbarrini G. Determinants of functional status in healthy Italian nonagenarians and centenarians: a comprehensive functional assessment by the instruments of geriatric practice. J Am Geriatr Soc. 1997;45(10):1196-1202.
  69. Leng SX, Cappola AR, Andersen RE, Blackman MR, Koenig K, Blair M, Walston JD. Serum levels of insulin-like growth factor-I (IGF-I) and dehydroepiandrosterone sulfate (DHEA-S), and their relationships with serum interleukin-6, in the geriatric syndrome of frailty. Aging Clin Exp Res. 2004;16(2):153-157.
  70. Ohlsson C, Labrie F, Barrett-Connor E, Karlsson MK, Ljunggren O, Vandenput L, Mellstrom D, Tivesten A. Low serum levels of dehydroepiandrosterone sulfate predict all-cause and cardiovascular mortality in elderly Swedish men. J Clin Endocrinol Metab. 2010;95(9):4406-4414.
  71. Kroboth PD, Salek FS, Pittenger AL, Fabian TJ, Frye RF. DHEA and DHEA-S: a review. J Clin Pharmacol.1999;39(4):327-348.
  72. Nair KS, Rizza RA, O'Brien P, Dhatariya K, Short KR, Nehra A, Vittone JL, Klee GG, Basu A, Basu R, Cobelli C, Toffolo G, Dalla Man C, Tindall DJ, Melton LJ, 3rd, Smith GE, Khosla S, Jensen MD. DHEA in elderly women and DHEA or testosterone in elderly men. N Engl J Med. 2006;355(16):1647-1659.
  73. Baker WL, Karan S, Kenny AM. Effect of dehydroepiandrosterone on muscle strength and physical function in older adults: a systematic review. J Am Geriatr Soc. 2011;59(6):997-1002.
  74. Kritz-Silverstein D, von Muhlen D, Laughlin GA, Bettencourt R. Effects of dehydroepiandrosterone supplementation on cognitive function and quality of life: the DHEA and Well-Ness (DAWN) Trial. J Am Geriatr Soc. 2008;56(7):1292-1298.
  75. Panjari M, Bell RJ, Jane F, Wolfe R, Adams J, Morrow C, Davis SR. A randomized trial of oral DHEA treatment for sexual function, well-being, and menopausal symptoms in postmenopausal women with low libido. J Sex Med. 2009;6(9):2579-2590.
  76. Alkatib AA, Cosma M, Elamin MB, Erickson D, Swiglo BA, Erwin PJ, Montori VM. A systematic review and meta-analysis of randomized placebo-controlled trials of DHEA treatment effects on quality of life in women with adrenal insufficiency. J Clin Endocrinol Metab. 2009;94(10):3676-3681.
  77. Labrie F, Archer D, Bouchard C, Fortier M, Cusan L, Gomez JL, Girard G, Baron M, Ayotte N, Moreau M, Dube R, Cote I, Labrie C, Lavoie L, Berger L, Gilbert L, Martel C, Balser J. Effect of intravaginal dehydroepiandrosterone (Prasterone) on libido and sexual dysfunction in postmenopausal women. Menopause.2009;16(5):923-931.
  78. van den Brandt PA, Spiegelman D, Yaun SS, Adami HO, Beeson L, Folsom AR, Fraser G, Goldbohm RA, Graham S, Kushi L, Marshall JR, Miller AB, Rohan T, Smith-Warner SA, Speizer FE, Willett WC, Wolk A, Hunter DJ. Pooled analysis of prospective cohort studies on height, weight, and breast cancer risk. Am J Epidemiol.2000;152(6):514-527.
  79. Arlt W. The approach to the adult with newly diagnosed adrenal insufficiency. J Clin Endocrinol Metab.2009;94(4):1059-1067.
  80. Panjari M, Davis SR. DHEA for postmenopausal women: a review of the evidence. Maturitas. 2010;66(2):172-179.
  81. Labrie F, Belanger A, Belanger P, Berube R, Martel C, Cusan L, Gomez J, Candas B, Chaussade V, Castiel I, Deloche C, Leclaire J. Metabolism of DHEA in postmenopausal women following percutaneous administration. J Steroid Biochem Mol Biol. 2007;103(2):178-188.
  82. Labrie F, Cusan L, Gomez JL, Martel C, Berube R, Belanger P, Chaussade V, Deloche C, Leclaire J. Changes in serum DHEA and eleven of its metabolites during 12-month percutaneous administration of DHEA. J Steroid Biochem Mol Biol. 2008;110(1-2):1-9.
  83. Arlt W, Callies F, Koehler I, van Vlijmen JC, Fassnacht M, Strasburger CJ, Seibel MJ, Huebler D, Ernst M, Oettel M, Reincke M, Schulte HM, Allolio B. Dehydroepiandrosterone supplementation in healthy men with an age-related decline of dehydroepiandrosterone secretion. J Clin Endocrinol Metab. 2001;86(10):4686-4692.
  84. Arlt W, Callies F, van Vlijmen JC, Koehler I, Reincke M, Bidlingmaier M, Huebler D, Oettel M, Ernst M, Schulte HM, Allolio B. Dehydroepiandrosterone replacement in women with adrenal insufficiency. N Engl J Med.1999;341(14):1013-1020.
  85. Lovas K, Gebre-Medhin G, Trovik TS, Fougner KJ, Uhlving S, Nedrebo BG, Myking OL, Kampe O, Husebye ES. Replacement of dehydroepiandrosterone in adrenal failure: no benefit for subjective health status and sexuality in a 9-month, randomized, parallel group clinical trial. J Clin Endocrinol Metab. 2003;88(3):1112-1118.
  86. Bornstein SR, Allolio B, Arlt W, Barthel A, Don-Wauchope A, Hammer GD, Husebye ES, Merke DP, Murad MH, Stratakis CA, Torpy DJ. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016;101(2):364-389.
  87. Guideline for the prevention of falls in older persons. American Geriatrics Society, British Geriatrics Society, and American Academy of Orthopaedic Surgeons Panel on Falls Prevention. J Am Geriatr Soc. 2001;49(5):664-672.
  88. Kostka T, Arsac LM, Patricot MC, Berthouze SE, Lacour JR, Bonnefoy M. Leg extensor power and dehydroepiandrosterone sulfate, insulin-like growth factor-I and testosterone in healthy active elderly people. Eur J Appl Physiol. 2000;82(1-2):83-90.
  89. Berkman LF, Seeman TE, Albert M, Blazer D, Kahn R, Mohs R, Finch C, Schneider E, Cotman C, McClearn G, et al. High, usual and impaired functioning in community-dwelling older men and women: findings from the MacArthur Foundation Research Network on Successful Aging. J Clin Epidemiol. 1993;46(10):1129-1140.
  90. Morrison MF, Katz IR, Parmelee P, Boyce AA, TenHave T. Dehydroepiandrosterone sulfate (DHEA-S) and psychiatric and laboratory measures of frailty in a residential care population. Am J Geriatr Psychiatry.1998;6(4):277-284.
  91. Abbasi A, Duthie EH, Jr., Sheldahl L, Wilson C, Sasse E, Rudman I, Mattson DE. Association of dehydroepiandrosterone sulfate, body composition, and physical fitness in independent community-dwelling older men and women. J Am Geriatr Soc. 1998;46(3):263-273.
  92. Clarke BL, Ebeling PR, Jones JD, Wahner HW, O'Fallon WM, Riggs BL, Fitzpatrick LA. Predictors of bone mineral density in aging healthy men varies by skeletal site. Calcif Tissue Int. 2002;70(3):137-145.
  93. Bischoff-Ferrari HA, Orav EJ, Dawson-Hughes B. Additive benefit of higher testosterone levels and vitamin D plus calcium supplementation in regard to fall risk reduction among older men and women. Osteoporos Int.2008;19(9):1307-1314.
  94. Voznesensky M, Walsh S, Dauser D, Brindisi J, Kenny AM. The association between dehydroepiandosterone and frailty in older men and women. Age Ageing. 2009;38(4):401-406.
  95. Hasselhorn HM, Theorell T, Vingard E, Musculoskeletal Intervention Center -Norrtalje Study G. Endocrine and immunologic parameters indicative of 6-month prognosis after the onset of low back pain or neck/shoulder pain. Spine (Phila Pa 1976). 2001;26(3):E24-29.
  96. Schell E, Theorell T, Hasson D, Arnetz B, Saraste H. Stress biomarkers' associations to pain in the neck, shoulder and back in healthy media workers: 12-month prospective follow-up. Eur Spine J. 2008;17(3):393-405.
  97. Morales AJ, Haubrich RH, Hwang JY, Asakura H, Yen SS. The effect of six months treatment with a 100 mg daily dose of dehydroepiandrosterone (DHEA) on circulating sex steroids, body composition and muscle strength in age-advanced men and women. Clin Endocrinol (Oxf). 1998;49(4):421-432.
  98. Villareal DT, Holloszy JO, Kohrt WM. Effects of DHEA replacement on bone mineral density and body composition in elderly women and men. Clin Endocrinol (Oxf). 2000;53(5):561-568.
  99. Kaiman DS, Colker CM, Swain MA, Torina GC, Shi Q. A randomized, double-blind, placebo-controlled study of 3-acetyl-7-oxo-dehydroepiandrosterone in healthy overweight adults. Current Therapeutic Research.2000;61(7):435-442.
  100. Kenny AM, Boxer RS, Kleppinger A, Brindisi J, Feinn R, Burleson JA. Dehydroepiandrosterone combined with exercise improves muscle strength and physical function in frail older women. J Am Geriatr Soc.2010;58(9):1707-1714.
  101. Weiss EP, Shah K, Fontana L, Lambert CP, Holloszy JO, Villareal DT. Dehydroepiandrosterone replacement therapy in older adults: 1- and 2-y effects on bone. Am J Clin Nutr. 2009;89(5):1459-1467.
  102. von Muhlen D, Laughlin GA, Kritz-Silverstein D, Bergstrom J, Bettencourt R. Effect of dehydroepiandrosterone supplementation on bone mineral density, bone markers, and body composition in older adults: the DAWN trial. Osteoporos Int. 2008;19(5):699-707.
  103. Jankowski CM, Gozansky WS, Kittelson JM, Van Pelt RE, Schwartz RS, Kohrt WM. Increases in bone mineral density in response to oral dehydroepiandrosterone replacement in older adults appear to be mediated by serum estrogens. J Clin Endocrinol Metab. 2008;93(12):4767-4773.
  104. Jankowski CM, Gozansky WS, Schwartz RS, Dahl DJ, Kittelson JM, Scott SM, Van Pelt RE, Kohrt WM. Effects of dehydroepiandrosterone replacement therapy on bone mineral density in older adults: a randomized, controlled trial. J Clin Endocrinol Metab. 2006;91(8):2986-2993.
  105. Labrie F, Diamond P, Cusan L, Gomez JL, Belanger A, Candas B. Effect of 12-month dehydroepiandrosterone replacement therapy on bone, vagina, and endometrium in postmenopausal women. J Clin Endocrinol Metab.1997;82(10):3498-3505.
  106. Morales AJ, Nolan JJ, Nelson JC, Yen SS. Effects of replacement dose of dehydroepiandrosterone in men and women of advancing age. J Clin Endocrinol Metab. 1994;78(6):1360-1367.
  107. Yen SS, Morales AJ, Khorram O. Replacement of DHEA in aging men and women. Potential remedial effects. Ann N Y Acad Sci. 1995;774:128-142.
  108. Nawata H, Tanaka S, Tanaka S, Takayanagi R, Sakai Y, Yanase T, Ikuyama S, Haji M. Aromatase in bone cell: association with osteoporosis in postmenopausal women. J Steroid Biochem Mol Biol. 1995;53(1-6):165-174.
  109. Jedrzejuk D, Medras M, Milewicz A, Demissie M. Dehydroepiandrosterone replacement in healthy men with age-related decline of DHEA-S: effects on fat distribution, insulin sensitivity and lipid metabolism. Aging Male.2003;6(3):151-156.
  110. Callies F, Fassnacht M, van Vlijmen JC, Koehler I, Huebler D, Seibel MJ, Arlt W, Allolio B. Dehydroepiandrosterone replacement in women with adrenal insufficiency: effects on body composition, serum leptin, bone turnover, and exercise capacity. J Clin Endocrinol Metab. 2001;86(5):1968-1972.
  111. Flynn MA, Weaver-Osterholtz D, Sharpe-Timms KL, Allen S, Krause G. Dehydroepiandrosterone replacement in aging humans. J Clin Endocrinol Metab. 1999;84(5):1527-1533.
  112. Casson PR, Santoro N, Elkind-Hirsch K, Carson SA, Hornsby PJ, Abraham G, Buster JE. Postmenopausal dehydroepiandrosterone administration increases free insulin-like growth factor-I and decreases high-density lipoprotein: a six-month trial. Fertil Steril. 1998;70(1):107-110.
  113. Igwebuike A, Irving BA, Bigelow ML, Short KR, McConnell JP, Nair KS. Lack of dehydroepiandrosterone effect on a combined endurance and resistance exercise program in postmenopausal women. J Clin Endocrinol Metab. 2008;93(2):534-538.
  114. Muller M, van den Beld AW, van der Schouw YT, Grobbee DE, Lamberts SW. Effects of dehydroepiandrosterone and atamestane supplementation on frailty in elderly men. J Clin Endocrinol Metab.2006;91(10):3988-3991.
  115. Villareal DT, Holloszy JO. DHEA enhances effects of weight training on muscle mass and strength in elderly women and men. Am J Physiol Endocrinol Metab. 2006;291(5):E1003-1008.
  116. Percheron G, Hogrel JY, Denot-Ledunois S, Fayet G, Forette F, Baulieu EE, Fardeau M, Marini JF, Double-blind placebo-controlled t. Effect of 1-year oral administration of dehydroepiandrosterone to 60- to 80-year-old individuals on muscle function and cross-sectional area: a double-blind placebo-controlled trial. Arch Intern Med.2003;163(6):720-727.
  117. Nijland E, Davis S, Laan E, Schultz WW. Female sexual satisfaction and pharmaceutical intervention: a critical review of the drug intervention studies in female sexual dysfunction. J Sex Med. 2006;3(5):763-777.
  118. Hayes RD, Dennerstein L, Bennett CM, Koochaki PE, Leiblum SR, Graziottin A. Relationship between hypoactive sexual desire disorder and aging. Fertil Steril. 2007;87(1):107-112.
  119. Shifren JL, Braunstein GD, Simon JA, Casson PR, Buster JE, Redmond GP, Burki RE, Ginsburg ES, Rosen RC, Leiblum SR, Caramelli KE, Mazer NA. Transdermal testosterone treatment in women with impaired sexual function after oophorectomy. N Engl J Med. 2000;343(10):682-688.
  120. Genazzani AD, Lanzoni C, Genazzani AR. Might DHEA be considered a beneficial replacement therapy in the elderly? Drugs Aging. 2007;24(3):173-185.
  121. Nappi RE, Polatti F. The use of estrogen therapy in women's sexual functioning (CME). J Sex Med.2009;6(3):603-616; quiz 618-609.
  122. Bachmann GA, Leiblum SR. The impact of hormones on menopausal sexuality: a literature review. Menopause.2004;11(1):120-130.
  123. Alexander JL, Kotz K, Dennerstein L, Kutner SJ, Wallen K, Notelovitz M. The effects of postmenopausal hormone therapies on female sexual functioning: a review of double-blind, randomized controlled trials. Menopause. 2004;11(6 Pt 2):749-765.
  124. Modelska K, Cummings S. Female sexual dysfunction in postmenopausal women: systematic review of placebo-controlled trials. Am J Obstet Gynecol. 2003;188(1):286-293.
  125. Goldstat R, Briganti E, Tran J, Wolfe R, Davis SR. Transdermal testosterone therapy improves well-being, mood, and sexual function in premenopausal women. Menopause. 2003;10(5):390-398.
  126. Braunstein GD, Sundwall DA, Katz M, Shifren JL, Buster JE, Simon JA, Bachman G, Aguirre OA, Lucas JD, Rodenberg C, Buch A, Watts NB. Safety and efficacy of a testosterone patch for the treatment of hypoactive sexual desire disorder in surgically menopausal women: a randomized, placebo-controlled trial. Arch Intern Med.2005;165(14):1582-1589.
  127. Davis SR, van der Mooren MJ, van Lunsen RH, Lopes P, Ribot C, Rees M, Moufarege A, Rodenberg C, Buch A, Purdie DW. Efficacy and safety of a testosterone patch for the treatment of hypoactive sexual desire disorder in surgically menopausal women: a randomized, placebo-controlled trial. Menopause. 2006;13(3):387-396.
  128. Buster JE, Kingsberg SA, Aguirre O, Brown C, Breaux JG, Buch A, Rodenberg CA, Wekselman K, Casson P. Testosterone patch for low sexual desire in surgically menopausal women: a randomized trial. Obstet Gynecol.2005;105(5 Pt 1):944-952.
  129. Davis S, Papalia MA, Norman RJ, O'Neill S, Redelman M, Williamson M, Stuckey BG, Wlodarczyk J, Gard'ner K, Humberstone A. Safety and efficacy of a testosterone metered-dose transdermal spray for treating decreased sexual satisfaction in premenopausal women: a randomized trial. Ann Intern Med. 2008;148(8):569-577.
  130. Davis SR, Davison SL, Donath S, Bell RJ. Circulating androgen levels and self-reported sexual function in women. JAMA. 2005;294(1):91-96.
  131. Panjari M, Davis SR. DHEA therapy for women: effect on sexual function and wellbeing. Hum Reprod Update.2007;13(3):239-248.
  132. Schmidt PJ, Daly RC, Bloch M, Smith MJ, Danaceau MA, St Clair LS, Murphy JH, Haq N, Rubinow DR. Dehydroepiandrosterone monotherapy in midlife-onset major and minor depression. Arch Gen Psychiatry.2005;62(2):154-162.
  133. Bloch M, Schmidt PJ, Danaceau MA, Adams LF, Rubinow DR. Dehydroepiandrosterone treatment of midlife dysthymia. Biol Psychiatry. 1999;45(12):1533-1541.
  134. Hackbert L, Heiman JR. Acute dehydroepiandrosterone (DHEA) effects on sexual arousal in postmenopausal women. J Womens Health Gend Based Med. 2002;11(2):155-162.
  135. Genazzani AR, Stomati M, Valentino V, Pluchino N, Pot E, Casarosa E, Merlini S, Giannini A, Luisi M. Effect of 1-year, low-dose DHEA therapy on climacteric symptoms and female sexuality. Climacteric. 2011;14(6):661-668.
  136. Stomati M, Monteleone P, Casarosa E, Quirici B, Puccetti S, Bernardi F, Genazzani AD, Rovati L, Luisi M, Genazzani AR. Six-month oral dehydroepiandrosterone supplementation in early and late postmenopause. Gynecol Endocrinol. 2000;14(5):342-363.
  137. Mortola JF, Yen SS. The effects of oral dehydroepiandrosterone on endocrine-metabolic parameters in postmenopausal women. J Clin Endocrinol Metab. 1990;71(3):696-704.
  138. Wolf OT, Neumann O, Hellhammer DH, Geiben AC, Strasburger CJ, Dressendorfer RA, Pirke KM, Kirschbaum C. Effects of a two-week physiological dehydroepiandrosterone substitution on cognitive performance and well-being in healthy elderly women and men. J Clin Endocrinol Metab. 1997;82(7):2363-2367.
  139. Johannsson G, Burman P, Wiren L, Engstrom BE, Nilsson AG, Ottosson M, Jonsson B, Bengtsson BA, Karlsson FA. Low dose dehydroepiandrosterone affects behavior in hypopituitary androgen-deficient women: a placebo-controlled trial. J Clin Endocrinol Metab. 2002;87(5):2046-2052.
  140. van Thiel SW, Romijn JA, Pereira AM, Biermasz NR, Roelfsema F, van Hemert A, Ballieux B, Smit JW. Effects of dehydroepiandrostenedione, superimposed on growth hormone substitution, on quality of life and insulin-like growth factor I in patients with secondary adrenal insufficiency: a randomized, placebo-controlled, cross-over trial. J Clin Endocrinol Metab. 2005;90(6):3295-3303.
  141. Avis NE, Brockwell S, Randolph JF, Jr., Shen S, Cain VS, Ory M, Greendale GA. Longitudinal changes in sexual functioning as women transition through menopause: results from the Study of Women's Health Across the Nation. Menopause. 2009;16(3):442-452.
  142. Labrie F, Cusan L, Gomez JL, Cote I, Berube R, Belanger P, Martel C, Labrie C. Effect of intravaginal DHEA on serum DHEA and eleven of its metabolites in postmenopausal women. J Steroid Biochem Mol Biol. 2008;111(3-5):178-194.
  143. Labrie F, Archer D, Bouchard C, Fortier M, Cusan L, Gomez JL, Girard G, Baron M, Ayotte N, Moreau M, Dube R, Cote I, Labrie C, Lavoie L, Berger L, Martel C, Balser J. High internal consistency and efficacy of intravaginal DHEA for vaginal atrophy. Gynecol Endocrinol. 2010;26(7):524-532.
  144. Ibe C, Simon JA. Vulvovaginal atrophy: current and future therapies (CME). J Sex Med. 2010;7(3):1042-1050; quiz 1051.
  145. Reiter WJ, Pycha A, Schatzl G, Pokorny A, Gruber DM, Huber JC, Marberger M. Dehydroepiandrosterone in the treatment of erectile dysfunction: a prospective, double-blind, randomized, placebo-controlled study. Urology. 1999;53(3):590-594; discussion 594-595.
  146. Steffens DC. A multiplicity of approaches to characterize geriatric depression and its outcomes. Curr Opin Psychiatry. 2009;22(6):522-526.
  147. Robichaud M, Debonnel G. Modulation of the firing activity of female dorsal raphe nucleus serotonergic neurons by neuroactive steroids. J Endocrinol. 2004;182(1):11-21.
  148. Kilic FS, Kulluk D, Musmul A. Effects of dehydroepiandrosterone in amphetamine-induced schizophrenia models in mice. Neurosciences (Riyadh). 2014;19(2):100-105.
  149. Goodyer IM, Herbert J, Altham PM, Pearson J, Secher SM, Shiers HM. Adrenal secretion during major depression in 8- to 16-year-olds, I. Altered diurnal rhythms in salivary cortisol and dehydroepiandrosterone (DHEA) at presentation. Psychol Med. 1996;26(2):245-256.
  150. Barrett-Connor E, von Muhlen D, Laughlin GA, Kripke A. Endogenous levels of dehydroepiandrosterone sulfate, but not other sex hormones, are associated with depressed mood in older women: the Rancho Bernardo Study. J Am Geriatr Soc. 1999;47(6):685-691.
  151. Michael A, Jenaway A, Paykel ES, Herbert J. Altered salivary dehydroepiandrosterone levels in major depression in adults. Biol Psychiatry. 2000;48(10):989-995.
  152. Morrison MF, Freeman EW, Lin H, Sammel MD. Higher DHEA-S (dehydroepiandrosterone sulfate) levels are associated with depressive symptoms during the menopausal transition: results from the PENN Ovarian Aging Study. Arch Womens Ment Health. 2011;14(5):375-382.
  153. Takebayashi M, Kagaya A, Uchitomi Y, Kugaya A, Muraoka M, Yokota N, Horiguchi J, Yamawaki S. Plasma dehydroepiandrosterone sulfate in unipolar major depression. Short communication. J Neural Transm (Vienna).1998;105(4-5):537-542.
  154. Assies J, Visser I, Nicolson NA, Eggelte TA, Wekking EM, Huyser J, Lieverse R, Schene AH. Elevated salivary dehydroepiandrosterone-sulfate but normal cortisol levels in medicated depressed patients: preliminary findings. Psychiatry Res. 2004;128(2):117-122.
  155. Fabian TJ, Dew MA, Pollock BG, Reynolds CF, 3rd, Mulsant BH, Butters MA, Zmuda MD, Linares AM, Trottini M, Kroboth PD. Endogenous concentrations of DHEA and DHEA-S decrease with remission of depression in older adults. Biol Psychiatry. 2001;50(10):767-774.
  156. Wolkowitz OM, Reus VI, Roberts E, Manfredi F, Chan T, Raum WJ, Ormiston S, Johnson R, Canick J, Brizendine L, Weingartner H. Dehydroepiandrosterone (DHEA) treatment of depression. Biol Psychiatry.1997;41(3):311-318.
  157. Wolkowitz OM, Reus VI, Keebler A, Nelson N, Friedland M, Brizendine L, Roberts E. Double-blind treatment of major depression with dehydroepiandrosterone. Am J Psychiatry. 1999;156(4):646-649.
  158. Strous RD, Maayan R, Lapidus R, Stryjer R, Lustig M, Kotler M, Weizman A. Dehydroepiandrosterone augmentation in the management of negative, depressive, and anxiety symptoms in schizophrenia. Arch Gen Psychiatry. 2003;60(2):133-141.
  159. Hough CM, Lindqvist D, Epel ES, Denis MS, Reus VI, Bersani FS, Rosser R, Mahan L, Burke HM, Wolkowitz OM, Mellon SH. Higher serum DHEA concentrations before and after SSRI treatment are associated with remission of major depression. Psychoneuroendocrinology. 2017;77:122-130.
  160. Kaplan JR, Manuck SB, Clarkson TB, Lusso FM, Taub DM. Social status, environment, and atherosclerosis in cynomolgus monkeys. Arteriosclerosis. 1982;2(5):359-368.
  161. Kivimaki M, Nyberg ST, Batty GD, et al. Job strain as a risk factor for coronary heart disease: a collaborative meta-analysis of individual participant data. Lancet. 2012;380(9852):1491-1497.
  162. Marmot M. Psychosocial factors and cardiovascular disease: epidemiological approaches. Eur Heart J.1988;9(6):690-697.
  163. Epel ES, Blackburn EH, Lin J, Dhabhar FS, Adler NE, Morrow JD, Cawthon RM. Accelerated telomere shortening in response to life stress. Proc Natl Acad Sci U S A. 2004;101(49):17312-17315.
  164. Wolkowitz OM, Epel ES, Reus VI, Mellon SH. Depression gets old fast: do stress and depression accelerate cell aging? Depress Anxiety. 2010;27(4):327-338.
  165. Jeckel CM, Lopes RP, Berleze MC, Luz C, Feix L, Argimon, II, Stein LM, Bauer ME. Neuroendocrine and immunological correlates of chronic stress in 'strictly healthy' populations. Neuroimmunomodulation.2010;17(1):9-18.
  166. Izawa S, Saito K, Shirotsuki K, Sugaya N, Nomura S. Effects of prolonged stress on salivary cortisol and dehydroepiandrosterone: a study of a two-week teaching practice. Psychoneuroendocrinology. 2012;37(6):852-858.
  167. Brzoza Z, Kasperska-Zajac A, Badura-Brzoza K, Matysiakiewicz J, Hese RT, Rogala B. Decline in dehydroepiandrosterone sulfate observed in chronic urticaria is associated with psychological distress. Psychosom Med. 2008;70(6):723-728.
  168. Lac G, Dutheil F, Brousse G, Triboulet-Kelly C, Chamoux A. Saliva DHEAS changes in patients suffering from psychopathological disorders arising from bullying at work. Brain Cogn. 2012;80(2):277-281.
  169. Du CL, Lin MC, Lu L, Tai JJ. Correlation of Occupational Stress Index with 24-hour Urine Cortisol and Serum DHEA Sulfate among City Bus Drivers: A Cross-sectional Study. Saf Health Work. 2011;2(2):169-175.
  170. Kim MS, Lee YJ, Ahn RS. Day-to-day differences in cortisol levels and molar cortisol-to-DHEA ratios among working individuals. Yonsei Med J. 2010;51(2):212-218.
  171. Lennartsson AK, Theorell T, Rockwood AL, Kushnir MM, Jonsdottir IH. Perceived stress at work is associated with lower levels of DHEA-S. PLoS One. 2013;8(8):e72460.
  172. Jorm AF, Jolley D. The incidence of dementia: a meta-analysis. Neurology. 1998;51(3):728-733.
  173. Lu SF, Mo Q, Hu S, Garippa C, Simon NG. Dehydroepiandrosterone upregulates neural androgen receptor level and transcriptional activity. J Neurobiol. 2003;57(2):163-171.
  174. Suzuki M, Wright LS, Marwah P, Lardy HA, Svendsen CN. Mitotic and neurogenic effects of dehydroepiandrosterone (DHEA) on human neural stem cell cultures derived from the fetal cortex. Proc Natl Acad Sci U S A. 2004;101(9):3202-3207.
  175. Lang PO, Mitchell WA, Lapenna A, Pitts D, Aspinall R. Immunological pathogenesis of main age-related diseases and frailty: Role of immunosenescence. European Geriatric Medicine. 2010;1(2):112-121.
  176. Davis SR, Shah SM, McKenzie DP, Kulkarni J, Davison SL, Bell RJ. Dehydroepiandrosterone sulfate levels are associated with more favorable cognitive function in women. J Clin Endocrinol Metab. 2008;93(3):801-808.
  177. Hildreth KL, Gozansky WS, Jankowski CM, Grigsby J, Wolfe P, Kohrt WM. Association of serum dehydroepiandrosterone sulfate and cognition in older adults: sex steroid, inflammatory, and metabolic mechanisms. Neuropsychology. 2013;27(3):356-363.
  178. Valenti G, Ferrucci L, Lauretani F, Ceresini G, Bandinelli S, Luci M, Ceda G, Maggio M, Schwartz RS. Dehydroepiandrosterone sulfate and cognitive function in the elderly: The InCHIANTI Study. J Endocrinol Invest.2009;32(9):766-772.
  179. Weill-Engerer S, David JP, Sazdovitch V, Liere P, Eychenne B, Pianos A, Schumacher M, Delacourte A, Baulieu EE, Akwa Y. Neurosteroid quantification in human brain regions: comparison between Alzheimer's and nondemented patients. J Clin Endocrinol Metab. 2002;87(11):5138-5143.
  180. Belanger N, Gregoire L, Bedard PJ, Di Paolo T. DHEA improves symptomatic treatment of moderately and severely impaired MPTP monkeys. Neurobiol Aging. 2006;27(11):1684-1693.
  181. van Niekerk JK, Huppert FA, Herbert J. Salivary cortisol and DHEA: association with measures of cognition and well-being in normal older men, and effects of three months of DHEA supplementation. Psychoneuroendocrinology. 2001;26(6):591-612.
  182. Bielohuby M, Roemmler J, Manolopoulou J, Johnsen I, Sawitzky M, Schopohl J, Reincke M, Wolf E, Hoeflich A, Bidlingmaier M. Chronic growth hormone excess is associated with increased aldosterone: a study in patients with acromegaly and in growth hormone transgenic mice. Experimental biology and medicine (Maywood, NJ).2009;234(8):1002-1009.
  183. Wolf OT, Kudielka BM, Hellhammer DH, Hellhammer J, Kirschbaum C. Opposing effects of DHEA replacement in elderly subjects on declarative memory and attention after exposure to a laboratory stressor. Psychoneuroendocrinology. 1998;23(6):617-629.
  184. Wolf OT, Kirschbaum C. Actions of dehydroepiandrosterone and its sulfate in the central nervous system: effects on cognition and emotion in animals and humans. Brain Res Brain Res Rev. 1999;30(3):264-288.
  185. Grimley Evans J, Malouf R, Huppert F, van Niekerk JK. Dehydroepiandrosterone (DHEA) supplementation for cognitive function in healthy elderly people. Cochrane Database Syst Rev. 2006;2006(4):CD006221.
  186. Wolkowitz OM, Kramer JH, Reus VI, Costa MM, Yaffe K, Walton P, Raskind M, Peskind E, Newhouse P, Sack D, De Souza E, Sadowsky C, Roberts E, Research DH-AsDC. DHEA treatment of Alzheimer's disease: a randomized, double-blind, placebo-controlled study. Neurology. 2003;60(7):1071-1076.
  187. Yamada S, Akishita M, Fukai S, Ogawa S, Yamaguchi K, Matsuyama J, Kozaki K, Toba K, Ouchi Y. Effects of dehydroepiandrosterone supplementation on cognitive function and activities of daily living in older women with mild to moderate cognitive impairment. Geriatr Gerontol Int. 2010;10(4):280-287.
  188. Parsons TD, Kratz KM, Thompson E, Stanczyk FZ, Buckwalter JG. Dhea supplementation and cognition in postmenopausal women. Int J Neurosci. 2006;116(2):141-155.
  189. Morrison MF, Redei E, TenHave T, Parmelee P, Boyce AA, Sinha PS, Katz IR. Dehydroepiandrosterone sulfate and psychiatric measures in a frail, elderly residential care population. Biol Psychiatry. 2000;47(2):144-150.
  190. Somboonporn W, Davis S, Seif MW, Bell R. Testosterone for peri- and postmenopausal women. Cochrane Database Syst Rev. 2005(4):CD004509.
  191. Darling GM, Johns JA, McCloud PI, Davis SR. Estrogen and progestin compared with simvastatin for hypercholesterolemia in postmenopausal women. N Engl J Med. 1997;337(9):595-601.
  192. Barnhart KT, Freeman E, Grisso JA, Rader DJ, Sammel M, Kapoor S, Nestler JE. The effect of dehydroepiandrosterone supplementation to symptomatic perimenopausal women on serum endocrine profiles, lipid parameters, and health-related quality of life. J Clin Endocrinol Metab. 1999;84(11):3896-3902.
  193. Davis SR, Goldstat R, Newman A, Berry K, Burger HG, Meredith I, Koch K. Differing effects of low-dose estrogen-progestin therapy and pravastatin in postmenopausal hypercholesterolemic women. Climacteric.2002;5(4):341-350.
  194. Floter A, Nathorst-Boos J, Carlstrom K, von Schoultz B. Serum lipids in oophorectomized women during estrogen and testosterone replacement therapy. Maturitas. 2004;47(2):123-129.
  195. Leao LM, Duarte MP, Silva DM, Bahia PR, Coeli CM, de Farias ML. Influence of methyltestosterone postmenopausal therapy on plasma lipids, inflammatory factors, glucose metabolism and visceral fat: a randomized study. Eur J Endocrinol. 2006;154(1):131-139.
  196. Srinivasan M, Irving BA, Dhatariya K, Klaus KA, Hartman SJ, McConnell JP, Nair KS. Effect of dehydroepiandrosterone replacement on lipoprotein profile in hypoadrenal women. J Clin Endocrinol Metab.2009;94(3):761-764.
  197. Srinivasan M, Irving BA, Frye RL, O'Brien P, Hartman SJ, McConnell JP, Nair KS. Effects on lipoprotein particles of long-term dehydroepiandrosterone in elderly men and women and testosterone in elderly men. J Clin Endocrinol Metab. 2010;95(4):1617-1625.
  198. Bell RJ, Davison SL, Papalia MA, McKenzie DP, Davis SR. Endogenous androgen levels and cardiovascular risk profile in women across the adult life span. Menopause. 2007;14(4):630-638.
  199. Shufelt C, Bretsky P, Almeida CM, Johnson BD, Shaw LJ, Azziz R, Braunstein GD, Pepine CJ, Bittner V, Vido DA, Stanczyk FZ, Bairey Merz CN. DHEA-S levels and cardiovascular disease mortality in postmenopausal women: results from the National Institutes of Health--National Heart, Lung, and Blood Institute (NHLBI)-sponsored Women's Ischemia Syndrome Evaluation (WISE). J Clin Endocrinol Metab. 2010;95(11):4985-4992.
  200. Christiansen JJ, Andersen NH, Sorensen KE, Pedersen EM, Bennett P, Andersen M, Christiansen JS, Jorgensen JO, Gravholt CH. Dehydroepiandrosterone substitution in female adrenal failure: no impact on endothelial function and cardiovascular parameters despite normalization of androgen status. Clin Endocrinol (Oxf). 2007;66(3):426-433.
  201. Panjari M, Bell RJ, Jane F, Adams J, Morrow C, Davis SR. The safety of 52 weeks of oral DHEA therapy for postmenopausal women. Maturitas. 2009;63(3):240-245.
  202. Mohan PF, Ihnen JS, Levin BE, Cleary MP. Effects of dehydroepiandrosterone treatment in rats with diet-induced obesity. J Nutr. 1990;120(9):1103-1114.
  203. Cleary MP, Zisk JF. Anti-obesity effect of two different levels of dehydroepiandrosterone in lean and obese middle-aged female Zucker rats. Int J Obes. 1986;10(3):193-204.
  204. Hansen PA, Han DH, Nolte LA, Chen M, Holloszy JO. DHEA protects against visceral obesity and muscle insulin resistance in rats fed a high-fat diet. Am J Physiol. 1997;273(5):R1704-1708.
  205. Corona G, Rastrelli G, Giagulli VA, Sila A, Sforza A, Forti G, Mannucci E, Maggi M. Dehydroepiandrosterone supplementation in elderly men: a meta-analysis study of placebo-controlled trials. J Clin Endocrinol Metab.2013;98(9):3615-3626.
  206. Jia C, Chen X, Li X, Li M, Miao C, Sun B, Fan Z, Ren L. The effect of DHEA treatment on the oxidative stress and myocardial fibrosis induced by Keshan disease pathogenic factors. J Trace Elem Med Biol. 2011;25(3):154-159.
  207. Steinberg HO, Chaker H, Leaming R, Johnson A, Brechtel G, Baron AD. Obesity/insulin resistance is associated with endothelial dysfunction. Implications for the syndrome of insulin resistance. J Clin Invest.1996;97(11):2601-2610.
  208. Labrie F, Luu-The V, Belanger A, Lin SX, Simard J, Pelletier G, Labrie C. Is dehydroepiandrosterone a hormone? J Endocrinol. 2005;187(2):169-196.
  209. Wellman M, Shane-McWhorter L, Orlando, Jennings JP. The role of dehydroepiandrosterone in diabetes mellitus. Pharmacotherapy. 1999;19(5):582-591.
  210. Livingstone C, Collison M. Sex steroids and insulin resistance. Clin Sci (Lond). 2002;102(2):151-166.
  211. Golden SH, Dobs AS, Vaidya D, Szklo M, Gapstur S, Kopp P, Liu K, Ouyang P. Endogenous sex hormones and glucose tolerance status in postmenopausal women. J Clin Endocrinol Metab. 2007;92(4):1289-1295.
  212. Barrett-Connor E, Ferrara A. Dehydroepiandrosterone, dehydroepiandrosterone sulfate, obesity, waist-hip ratio, and noninsulin-dependent diabetes in postmenopausal women: the Rancho Bernardo Study. J Clin Endocrinol Metab. 1996;81(1):59-64.
  213. Dhatariya K, Bigelow ML, Nair KS. Effect of dehydroepiandrosterone replacement on insulin sensitivity and lipids in hypoadrenal women. Diabetes. 2005;54(3):765-769.
  214. Villareal DT, Holloszy JO. Effect of DHEA on abdominal fat and insulin action in elderly women and men: a randomized controlled trial. JAMA. 2004;292(18):2243-2248.
  215. Lasco A, Frisina N, Morabito N, Gaudio A, Morini E, Trifiletti A, Basile G, Nicita-Mauro V, Cucinotta D. Metabolic effects of dehydroepiandrosterone replacement therapy in postmenopausal women. Eur J Endocrinol.2001;145(4):457-461.
  216. Gomez-Santos C, Larque E, Granero E, Hernandez-Morante JJ, Garaulet M. Dehydroepiandrosterone-sulphate replacement improves the human plasma fatty acid profile in plasma of obese women. Steroids.2011;76(13):1425-1432.
  217. Diamond P, Cusan L, Gomez JL, Belanger A, Labrie F. Metabolic effects of 12-month percutaneous dehydroepiandrosterone replacement therapy in postmenopausal women. J Endocrinol. 1996;150 Suppl:S43-50.
  218. Paul S Jellinger DAS, Adi E Mehta, Om Ganda, Yehuda Handelsman, Helena W Rodbard, Mark D Shepherd, John A Seibel, AACE Task Force for Management of Dyslipidemia and Prevention of Atherosclerosis. American Association of Clinical Endocrinologists' Guidelines for Management of Dyslipidemia and Prevention of Atherosclerosis. Endocrine Practice. 2012(Mar-Apr;18):1-78.
  219. Phillips GB, Pinkernell BH, Jing TY. Are major risk factors for myocardial infarction the major predictors of degree of coronary artery disease in men? Metabolism. 2004;53(3):324-329.
  220. Freedman DS, O'Brien TR, Flanders WD, DeStefano F, Barboriak JJ. Relation of serum testosterone levels to high density lipoprotein cholesterol and other characteristics in men. Arterioscler Thromb. 1991;11(2):307-315.
  221. Tripathi Y, Hegde BM. Serum estradiol and testosterone levels following acute myocardial infarction in men. Indian J Physiol Pharmacol. 1998;42(2):291-294.
  222. Pugh PJ, Channer KS, Parry H, Downes T, Jone TH. Bio-available testosterone levels fall acutely following myocardial infarction in men: association with fibrinolytic factors. Endocr Res. 2002;28(3):161-173.
  223. Glueck CJ, Glueck HI, Stroop D, Speirs J, Hamer T, Tracy T. Endogenous testosterone, fibrinolysis, and coronary heart disease risk in hyperlipidemic men. J Lab Clin Med. 1993;122(4):412-420.
  224. Caron P, Bennet A, Camare R, Louvet JP, Boneu B, Sie P. Plasminogen activator inhibitor in plasma is related to testosterone in men. Metabolism. 1989;38(10):1010-1015.
  225. De Pergola G, De Mitrio V, Sciaraffia M, Pannacciulli N, Minenna A, Giorgino F, Petronelli M, Laudadio E, Giorgino R. Lower androgenicity is associated with higher plasma levels of prothrombotic factors irrespective of age, obesity, body fat distribution, and related metabolic parameters in men. Metabolism. 1997;46(11):1287-1293.
  226. Militaru C, Donoiu I, Dracea O, Ionescu DD. Serum testosterone and short-term mortality in men with acute myocardial infarction. Cardiol J. 2010;17(3):249-253.
  227. Fukui M, Kitagawa Y, Nakamura N, Kadono M, Yoshida M, Hirata C, Wada K, Hasegawa G, Yoshikawa T. Serum dehydroepiandrosterone sulfate concentration and carotid atherosclerosis in men with type 2 diabetes. Atherosclerosis. 2005;181(2):339-344.
  228. Suzuki T, Yano Y, Sakamoto M, Uemura M, Yasuma T, Onishi Y, Sasaki R, Matsumoto K, Hayashi T, Maruyama-Furuta N, Akatsuka H, Gabazza EC, Sumida Y, Takei Y. Correlation of circulating dehydroepiandrosterone with activated protein C generation and carotid intima-media thickness in male patients with type 2 diabetes. Diabet Med. 2012;29(7):e41-46.
  229. Akishita M, Hashimoto M, Ohike Y, Ogawa S, Iijima K, Eto M, Ouchi Y. Association of plasma dehydroepiandrosterone-sulfate levels with endothelial function in postmenopausal women with coronary risk factors. Hypertens Res. 2008;31(1):69-74.
  230. Gebre-Medhin G, Husebye ES, Mallmin H, Helstrom L, Berne C, Karlsson FA, Kampe O. Oral dehydroepiandrosterone (DHEA) replacement therapy in women with Addison's disease. Clin Endocrinol (Oxf).2000;52(6):775-780.
  231. Nestler JE, Barlascini CO, Clore JN, Blackard WG. Dehydroepiandrosterone reduces serum low density lipoprotein levels and body fat but does not alter insulin sensitivity in normal men. J Clin Endocrinol Metab.1988;66(1):57-61.
  232. Casson PR, Faquin LC, Stentz FB, Straughn AB, Andersen RN, Abraham GE, Buster JE. Replacement of dehydroepiandrosterone enhances T-lymphocyte insulin binding in postmenopausal women. Fertil Steril.1995;63(5):1027-1031.
  233. Basu R, Dalla Man C, Campioni M, Basu A, Nair KS, Jensen MD, Khosla S, Klee G, Toffolo G, Cobelli C, Rizza RA. Two years of treatment with dehydroepiandrosterone does not improve insulin secretion, insulin action, or postprandial glucose turnover in elderly men or women. Diabetes. 2007;56(3):753-766.
  234. Herrington DM. Dehydroepiandrosterone and coronary atherosclerosis. Ann N Y Acad Sci. 1995;774:271-280.
  235. Herrington DM, Nanjee N, Achuff SC, Cameron DE, Dobbs B, Baughman KL. Dehydroepiandrosterone and cardiac allograft vasculopathy. J Heart Lung Transplant. 1996;15(1 Pt 1):88-93.
  236. Shono N, Kumagai S, Higaki Y, Nishizumi M, Sasaki H. The relationships of testosterone, estradiol, dehydroepiandrosterone-sulfate and sex hormone-binding globulin to lipid and glucose metabolism in healthy men. J Atheroscler Thromb. 1996;3(1):45-51.
  237. Bernini GP, Moretti A, Sgro M, Argenio GF, Barlascini CO, Cristofani R, Salvetti A. Influence of endogenous androgens on carotid wall in postmenopausal women. Menopause. 2001;8(1):43-50.
  238. Bernini GP, Sgro M, Moretti A, Argenio GF, Barlascini CO, Cristofani R, Salvetti A. Endogenous androgens and carotid intimal-medial thickness in women. J Clin Endocrinol Metab. 1999;84(6):2008-2012.
  239. Alexandersen P, Haarbo J, Christiansen C. The relationship of natural androgens to coronary heart disease in males: a review. Atherosclerosis. 1996;125(1):1-13.
  240. Feldman HA, Johannes CB, Araujo AB, Mohr BA, Longcope C, McKinlay JB. Low dehydroepiandrosterone and ischemic heart disease in middle-aged men: prospective results from the Massachusetts Male Aging Study. Am J Epidemiol. 2001;153(1):79-89.
  241. Trivedi DP, Khaw KT. Dehydroepiandrosterone sulfate and mortality in elderly men and women. J Clin Endocrinol Metab. 2001;86(9):4171-4177.
  242. Hak AE, Witteman JC, de Jong FH, Geerlings MI, Hofman A, Pols HA. Low levels of endogenous androgens increase the risk of atherosclerosis in elderly men: the Rotterdam study. J Clin Endocrinol Metab.2002;87(8):3632-3639.
  243. Herrington DM, Gordon GB, Achuff SC, Trejo JF, Weisman HF, Kwiterovich PO, Jr., Pearson TA. Plasma dehydroepiandrosterone and dehydroepiandrosterone sulfate in patients undergoing diagnostic coronary angiography. J Am Coll Cardiol. 1990;16(6):862-870.
  244. Ishihara F, Hiramatsu K, Shigematsu S, Aizawa T, Niwa A, Takasu N, Yamada T, Matsuo K. Role of adrenal androgens in the development of arteriosclerosis as judged by pulse wave velocity and calcification of the aorta. Cardiology. 1992;80(5-6):332-338.
  245. Mitchell LE, Sprecher DL, Borecki IB, Rice T, Laskarzewski PM, Rao DC. Evidence for an association between dehydroepiandrosterone sulfate and nonfatal, premature myocardial infarction in males. Circulation.1994;89(1):89-93.
  246. Barrett-Connor E, Goodman-Gruen D. Dehydroepiandrosterone sulfate does not predict cardiovascular death in postmenopausal women. The Rancho Bernardo Study. Circulation. 1995;91(6):1757-1760.
  247. Barrett-Connor E, Goodman-Gruen D. The epidemiology of DHEAS and cardiovascular disease. Ann N Y Acad Sci. 1995;774:259-270.
  248. Berr C, Lafont S, Debuire B, Dartigues JF, Baulieu EE. Relationships of dehydroepiandrosterone sulfate in the elderly with functional, psychological, and mental status, and short-term mortality: a French community-based study. Proc Natl Acad Sci U S A. 1996;93(23):13410-13415.
  249. Nakamura S, Yoshimura M, Nakayama M, Ito T, Mizuno Y, Harada E, Sakamoto T, Saito Y, Nakao K, Yasue H, Ogawa H. Possible association of heart failure status with synthetic balance between aldosterone and dehydroepiandrosterone in human heart. Circulation. 2004;110(13):1787-1793.
  250. Jankowska EA, Biel B, Majda J, Szklarska A, Lopuszanska M, Medras M, Anker SD, Banasiak W, Poole-Wilson PA, Ponikowski P. Anabolic deficiency in men with chronic heart failure: prevalence and detrimental impact on survival. Circulation. 2006;114(17):1829-1837.
  251. Formoso G, Chen H, Kim JA, Montagnani M, Consoli A, Quon MJ. Dehydroepiandrosterone mimics acute actions of insulin to stimulate production of both nitric oxide and endothelin 1 via distinct phosphatidylinositol 3-kinase- and mitogen-activated protein kinase-dependent pathways in vascular endothelium. Mol Endocrinol.2006;20(5):1153-1163.
  252. Duncker DJ, Bache RJ. Inhibition of nitric oxide production aggravates myocardial hypoperfusion during exercise in the presence of a coronary artery stenosis. Circ Res. 1994;74(4):629-640.
  253. Wang L, Hao Q, Wang YD, Wang WJ, Li DJ. Protective effects of dehydroepiandrosterone on atherosclerosis in ovariectomized rabbits via alleviating inflammatory injury in endothelial cells. Atherosclerosis. 2011;214(1):47-57.
  254. Ross R. Atherosclerosis--an inflammatory disease. N Engl J Med. 1999;340(2):115-126.
  255. Libby P, Geng YJ, Aikawa M, Schoenbeck U, Mach F, Clinton SK, Sukhova GK, Lee RT. Macrophages and atherosclerotic plaque stability. Curr Opin Lipidol. 1996;7(5):330-335.
  256. Libby P. Current concepts of the pathogenesis of the acute coronary syndromes. Circulation. 2001;104(3):365-372.
  257. Libby P, Simon DI. Inflammation and thrombosis: the clot thickens. Circulation. 2001;103(13):1718-1720.
  258. Belosjorow S, Bolle I, Duschin A, Heusch G, Schulz R. TNF-alpha antibodies are as effective as ischemic preconditioning in reducing infarct size in rabbits. Am J Physiol Heart Circ Physiol. 2003;284(3):H927-930.
  259. Liu D, Si H, Reynolds KA, Zhen W, Jia Z, Dillon JS. Dehydroepiandrosterone protects vascular endothelial cells against apoptosis through a Galphai protein-dependent activation of phosphatidylinositol 3-kinase/Akt and regulation of antiapoptotic Bcl-2 expression. Endocrinology. 2007;148(7):3068-3076.
  260. Chen J, Xu L, Huang C. DHEA inhibits vascular remodeling following arterial injury: a possible role in suppression of inflammation and oxidative stress derived from vascular smooth muscle cells. Mol Cell Biochem.2014;388(1-2):75-84.
  261. Hautanen A, Manttari M, Manninen V, Tenkanen L, Huttunen JK, Frick MH, Adlercreutz H. Adrenal androgens and testosterone as coronary risk factors in the Helsinki Heart Study. Atherosclerosis. 1994;105(2):191-200.
  262. LaCroix AZ, Yano K, Reed DM. Dehydroepiandrosterone sulfate, incidence of myocardial infarction, and extent of atherosclerosis in men. Circulation. 1992;86(5):1529-1535.
  263. Zumoff B, Troxler RG, O'Connor J, Rosenfeld RS, Kream J, Levin J, Hickman JR, Sloan AM, Walker W, Cook RL, Fukushima DK. Abnormal hormone levels in men with coronary artery disease. Arteriosclerosis.1982;2(1):58-67.
  264. Slowinska-Srzednicka J, Zgliczynski S, Ciswicka-Sznajderman M, Srzednicki M, Soszynski P, Biernacka M, Woroszylska M, Ruzyllo W, Sadowski Z. Decreased plasma dehydroepiandrosterone sulfate and dihydrotestosterone concentrations in young men after myocardial infarction. Atherosclerosis. 1989;79(2-3):197-203.
  265. Haffner SM, Moss SE, Klein BE, Klein R. Sex hormones and DHEA-SO4 in relation to ischemic heart disease mortality in diabetic subjects. The Wisconsin Epidemiologic Study of Diabetic Retinopathy. Diabetes Care.1996;19(10):1045-1050.
  266. Golden SH, Maguire A, Ding J, Crouse JR, Cauley JA, Zacur H, Szklo M. Endogenous postmenopausal hormones and carotid atherosclerosis: a case-control study of the atherosclerosis risk in communities cohort. Am J Epidemiol. 2002;155(5):437-445.
  267. Khaw KT, Tazuke S, Barrett-Connor E. Cigarette smoking and levels of adrenal androgens in postmenopausal women. N Engl J Med. 1988;318(26):1705-1709.
  268. Salvini S, Stampfer MJ, Barbieri RL, Hennekens CH. Effects of age, smoking and vitamins on plasma DHEAS levels: a cross-sectional study in men. J Clin Endocrinol Metab. 1992;74(1):139-143.
  269. Wu FC, von Eckardstein A. Androgens and coronary artery disease. Endocr Rev. 2003;24(2):183-217.
  270. Kawano H, Yasue H, Kitagawa A, Hirai N, Yoshida T, Soejima H, Miyamoto S, Nakano M, Ogawa H. Dehydroepiandrosterone supplementation improves endothelial function and insulin sensitivity in men. J Clin Endocrinol Metab. 2003;88(7):3190-3195.
  271. Eich DM, Nestler JE, Johnson DE, Dworkin GH, Ko D, Wechsler AS, Hess ML. Inhibition of accelerated coronary atherosclerosis with dehydroepiandrosterone in the heterotopic rabbit model of cardiac transplantation. Circulation. 1993;87(1):261-269.
  272. Gordon GB, Bush DE, Weisman HF. Reduction of atherosclerosis by administration of dehydroepiandrosterone. A study in the hypercholesterolemic New Zealand white rabbit with aortic intimal injury. J Clin Invest.1988;82(2):712-720.
  273. Aragno M, Parola S, Brignardello E, Mauro A, Tamagno E, Manti R, Danni O, Boccuzzi G. Dehydroepiandrosterone prevents oxidative injury induced by transient ischemia/reperfusion in the brain of diabetic rats. Diabetes. 2000;49(11):1924-1931.
  274. Ayhan S, Tugay C, Norton S, Araneo B, Siemionow M. Dehydroepiandrosterone protects the microcirculation of muscle flaps from ischemia-reperfusion injury by reducing the expression of adhesion molecules. Plast Reconstr Surg. 2003;111(7):2286-2294.
  275. Kinlay S, Creager MA, Fukumoto M, Hikita H, Fang JC, Selwyn AP, Ganz P. Endothelium-derived nitric oxide regulates arterial elasticity in human arteries in vivo. Hypertension. 2001;38(5):1049-1053.
  276. Weiss EP, Villareal DT, Ehsani AA, Fontana L, Holloszy JO. Dehydroepiandrosterone replacement therapy in older adults improves indices of arterial stiffness. Aging Cell. 2012;11(5):876-884.
  277. Murray CJ, Vos T, Lozano R, et al. Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet.2012;380(9859):2197-2223.
  278. Donkor ES. Stroke in the 21(st) Century: A Snapshot of the Burden, Epidemiology, and Quality of Life. Stroke Res Treat. 2018;2018:3238165.
  279. Baulieu EE, Robel P. Dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS) as neuroactive neurosteroids. Proc Natl Acad Sci U S A. 1998;95(8):4089-4091.
  280. Lapchak PA, Chapman DF, Nunez SY, Zivin JA. Dehydroepiandrosterone sulfate is neuroprotective in a reversible spinal cord ischemia model: possible involvement of GABA(A) receptors. Stroke. 2000;31(8):1953-1956; discussion 1957.
  281. Corpechot C, Robel P, Axelson M, Sjovall J, Baulieu EE. Characterization and measurement of dehydroepiandrosterone sulfate in rat brain. Proc Natl Acad Sci U S A. 1981;78(8):4704-4707.
  282. Baulieu EE. Neuroactive neurosteroids: dehydroepiandrosterone (DHEA) and DHEA sulphate. Acta Paediatr Suppl. 1999;88(433):78-80.
  283. Jimenez MC, Sun Q, Schurks M, Chiuve S, Hu FB, Manson JE, Rexrode KM. Low dehydroepiandrosterone sulfate is associated with increased risk of ischemic stroke among women. Stroke. 2013;44(7):1784-1789.
  284. Pappa T, Vemmos K, Saltiki K, Mantzou E, Stamatelopoulos K, Alevizaki M. Severity and outcome of acute stroke in women: relation to adrenal sex steroid levels. Metabolism. 2012;61(1):84-91.
  285. Blum CA, Mueller C, Schuetz P, Fluri F, Trummler M, Mueller B, Katan M, Christ-Crain M. Prognostic value of dehydroepiandrosterone-sulfate and other parameters of adrenal function in acute ischemic stroke. PLoS One.2013;8(5):e63224.
  286. Hampl V, Bibova J, Povysilova V, Herget J. Dehydroepiandrosterone sulphate reduces chronic hypoxic pulmonary hypertension in rats. Eur Respir J. 2003;21(5):862-865.
  287. Bonnet S, Dumas-de-La-Roque E, Begueret H, Marthan R, Fayon M, Dos Santos P, Savineau JP, Baulieu EE. Dehydroepiandrosterone (DHEA) prevents and reverses chronic hypoxic pulmonary hypertension. Proc Natl Acad Sci U S A. 2003;100(16):9488-9493.
  288. Debonneuil EH, Quillard J, Baulieu EE. Hypoxia and dehydroepiandrosterone in old age: a mouse survival study. Respir Res. 2006;7(1):144.
  289. Oka M, Karoor V, Homma N, Nagaoka T, Sakao E, Golembeski SM, Limbird J, Imamura M, Gebb SA, Fagan KA, McMurtry IF. Dehydroepiandrosterone upregulates soluble guanylate cyclase and inhibits hypoxic pulmonary hypertension. Cardiovasc Res. 2007;74(3):377-387.
  290. Farrukh IS, Peng W, Orlinska U, Hoidal JR. Effect of dehydroepiandrosterone on hypoxic pulmonary vasoconstriction: a Ca(2+)-activated K(+)-channel opener. Am J Physiol. 1998;274(2):L186-195.
  291. Gupte SA, Li KX, Okada T, Sato K, Oka M. Inhibitors of pentose phosphate pathway cause vasodilation: involvement of voltage-gated potassium channels. J Pharmacol Exp Ther. 2002;301(1):299-305.
  292. Dessouroux A, Akwa Y, Baulieu EE. DHEA decreases HIF-1alpha accumulation under hypoxia in human pulmonary artery cells: potential role in the treatment of pulmonary arterial hypertension. J Steroid Biochem Mol Biol. 2008;109(1-2):81-89.
  293. Patel D, Kandhi S, Kelly M, Neo BH, Wolin MS. Dehydroepiandrosterone promotes pulmonary artery relaxation by NADPH oxidation-elicited subunit dimerization of protein kinase G 1alpha. Am J Physiol Lung Cell Mol Physiol. 2014;306(4):L383-391.
  294. Ventetuolo CE, Ouyang P, Bluemke DA, Tandri H, Barr RG, Bagiella E, Cappola AR, Bristow MR, Johnson C, Kronmal RA, Kizer JR, Lima JA, Kawut SM. Sex hormones are associated with right ventricular structure and function: The MESA-right ventricle study. Am J Respir Crit Care Med. 2011;183(5):659-667.
  295. Dumas de La Roque E, Savineau JP, Metivier AC, Billes MA, Kraemer JP, Doutreleau S, Jougon J, Marthan R, Moore N, Fayon M, Baulieu EE, Dromer C. Dehydroepiandrosterone (DHEA) improves pulmonary hypertension in chronic obstructive pulmonary disease (COPD): a pilot study. Ann Endocrinol (Paris). 2012;73(1):20-25.
  296. Straub RH, Vogl D, Gross V, Lang B, Scholmerich J, Andus T. Association of humoral markers of inflammation and dehydroepiandrosterone sulfate or cortisol serum levels in patients with chronic inflammatory bowel disease. Am J Gastroenterol. 1998;93(11):2197-2202.
  297. de la Torre B, Hedman M, Befrits R. Blood and tissue dehydroepiandrosterone sulphate levels and their relationship to chronic inflammatory bowel disease. Clin Exp Rheumatol. 1998;16(5):579-582.
  298. Andus T, Klebl F, Rogler G, Bregenzer N, Scholmerich J, Straub RH. Patients with refractory Crohn's disease or ulcerative colitis respond to dehydroepiandrosterone: a pilot study. Aliment Pharmacol Ther. 2003;17(3):409-414.
  299. van Vollenhoven RF, Engleman EG, McGuire JL. An open study of dehydroepiandrosterone in systemic lupus erythematosus. Arthritis Rheum. 1994;37(9):1305-1310.
  300. van Vollenhoven RF, Engleman EG, McGuire JL. Dehydroepiandrosterone in systemic lupus erythematosus. Results of a double-blind, placebo-controlled, randomized clinical trial. Arthritis Rheum. 1995;38(12):1826-1831.
  301. van Vollenhoven RF, Morabito LM, Engleman EG, McGuire JL. Treatment of systemic lupus erythematosus with dehydroepiandrosterone: 50 patients treated up to 12 months. J Rheumatol. 1998;25(2):285-289.
  302. van Vollenhoven RF, Park JL, Genovese MC, West JP, McGuire JL. A double-blind, placebo-controlled, clinical trial of dehydroepiandrosterone in severe systemic lupus erythematosus. Lupus. 1999;8(3):181-187.
  303. Chang DM, Lan JL, Lin HY, Luo SF. Dehydroepiandrosterone treatment of women with mild-to-moderate systemic lupus erythematosus: a multicenter randomized, double-blind, placebo-controlled trial. Arthritis Rheum.2002;46(11):2924-2927.
  304. Wilder RL. Adrenal and gonadal steroid hormone deficiency in the pathogenesis of rheumatoid arthritis. J Rheumatol Suppl. 1996;44:10-12.
  305. Kanik KS, Chrousos GP, Schumacher HR, Crane ML, Yarboro CH, Wilder RL. Adrenocorticotropin, glucocorticoid, and androgen secretion in patients with new onset synovitis/rheumatoid arthritis: relations with indices of inflammation. J Clin Endocrinol Metab. 2000;85(4):1461-1466.
  306. Yukioka M, Komatsubara Y, Yukioka K, Toyosaki-Maeda T, Yonenobu K, Ochi T. Adrenocorticotropic hormone and dehydroepiandrosterone sulfate levels of rheumatoid arthritis patients treated with glucocorticoids. Mod Rheumatol. 2006;16(1):30-35.
  307. Danenberg HD, Alpert G, Lustig S, Ben-Nathan D. Dehydroepiandrosterone protects mice from endotoxin toxicity and reduces tumor necrosis factor production. Antimicrob Agents Chemother. 1992;36(10):2275-2279.
  308. Daynes RA, Araneo BA, Ershler WB, Maloney C, Li GZ, Ryu SY. Altered regulation of IL-6 production with normal aging. Possible linkage to the age-associated decline in dehydroepiandrosterone and its sulfated derivative. J Immunol. 1993;150(12):5219-5230.
  309. Keller ET, Chang C, Ershler WB. Inhibition of NFkappaB activity through maintenance of IkappaBalpha levels contributes to dihydrotestosterone-mediated repression of the interleukin-6 promoter. J Biol Chem.1996;271(42):26267-26275.
  310. Williams PJ, Jones RH, Rademacher TW. Reduction in the incidence and severity of collagen-induced arthritis in DBA/1 mice, using exogenous dehydroepiandrosterone. Arthritis Rheum. 1997;40(5):907-911.
  311. Kobayashi Y, Tagawa N, Muraoka K, Okamoto Y, Nishida M. Participation of endogenous dehydroepiandrosterone and its sulfate in the pathology of collagen-induced arthritis in mice. Biol Pharm Bull.2003;26(11):1596-1599.
  312. Rontzsch A, Thoss K, Petrow PK, Henzgen S, Brauer R. Amelioration of murine antigen-induced arthritis by dehydroepiandrosterone (DHEA). Inflamm Res. 2004;53(5):189-198.
  313. Giltay EJ, van Schaardenburg D, Gooren LJ, Dijkmans BA. Dehydroepiandrosterone sulfate in patients with rheumatoid arthritis. Ann N Y Acad Sci. 1999;876:152-154.
  314. Cutolo M. Sex hormone adjuvant therapy in rheumatoid arthritis. Rheum Dis Clin North Am. 2000;26(4):881-895.
  315. Marwah A, Marwah P, Lardy H. Ergosteroids. VI. Metabolism of dehydroepiandrosterone by rat liver in vitro: a liquid chromatographic-mass spectrometric study. J Chromatogr B Analyt Technol Biomed Life Sci.2002;767(2):285-299.
  316. Gambacciani M, Monteleone P, Sacco A, Genazzani AR. Hormone replacement therapy and endometrial, ovarian and colorectal cancer. Best Pract Res Clin Endocrinol Metab. 2003;17(1):139-147.
  317. Hankinson SE, Colditz GA, Willett WC. Towards an integrated model for breast cancer etiology: the lifelong interplay of genes, lifestyle, and hormones. Breast Cancer Res. 2004;6(5):213-218.
  318. Kaaks R, Lukanova A, Kurzer MS. Obesity, endogenous hormones, and endometrial cancer risk: a synthetic review. Cancer Epidemiol Biomarkers Prev. 2002;11(12):1531-1543.
  319. Kaaks R, Berrino F, Key T, et al. Serum sex steroids in premenopausal women and breast cancer risk within the European Prospective Investigation into Cancer and Nutrition (EPIC). J Natl Cancer Inst. 2005;97(10):755-765.
  320. Tworoger SS, Missmer SA, Eliassen AH, Spiegelman D, Folkerd E, Dowsett M, Barbieri RL, Hankinson SE. The association of plasma DHEA and DHEA sulfate with breast cancer risk in predominantly premenopausal women. Cancer Epidemiol Biomarkers Prev. 2006;15(5):967-971.
  321. Key T, Appleby P, Barnes I, Reeves G, Endogenous H, Breast Cancer Collaborative G. Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. J Natl Cancer Inst. 2002;94(8):606-616.
  322. Morris KT, Toth-Fejel S, Schmidt J, Fletcher WS, Pommier RF. High dehydroepiandrosterone-sulfate predicts breast cancer progression during new aromatase inhibitor therapy and stimulates breast cancer cell growth in tissue culture: a renewed role for adrenalectomy. Surgery. 2001;130(6):947-953.
  323. Schwartz AG, Pashko LL. Cancer prevention with dehydroepiandrosterone and non-androgenic structural analogs. J Cell Biochem Suppl. 1995;22:210-217.
  324. Maggiolini M, Donze O, Jeannin E, Ando S, Picard D. Adrenal androgens stimulate the proliferation of breast cancer cells as direct activators of estrogen receptor alpha. Cancer Res. 1999;59(19):4864-4869.
  325. Stoll BA. Dietary supplements of dehydroepiandrosterone in relation to breast cancer risk. Eur J Clin Nutr.1999;53(10):771-775.
  326. Genazzani AD, Stomati M, Bernardi F, Pieri M, Rovati L, Genazzani AR. Long-term low-dose dehydroepiandrosterone oral supplementation in early and late postmenopausal women modulates endocrine parameters and synthesis of neuroactive steroids. Fertil Steril. 2003;80(6):1495-1501.
  327. Arnold JT. DHEA metabolism in prostate: For better or worse? Mol Cell Endocrinol. 2009;301(1-2):83-88.
  328. Schwartz AG. Dehydroepiandrosterone, Cancer, and Aging. Aging Dis. 2022;13(2):423-432.
  329. Scheffers CS, Armstrong S, Cantineau AEP, Farquhar C. Dehydroepiandrosterone for menopausal women. Cochrane Database of Systematic Reviews. 2014(4).

Guidelines for Screening, Prevention, Diagnosis, and Treatment of Dyslipidemia in Children and Adolescents

ABSTRACT

 

Clinical practice guidelines are developed to create a synthesis of evidence which, in turn, leads to recommendations that improve clinical decision-making. Guidelines are helpful for busy clinicians to improve outcomes and reduce unnecessary practice variation. Historically, guidelines were largely based on expert opinion. The modern approach to guideline development includes a complete review and grading of the available evidence.  The evidence is then used to construct recommendations for clinical practice with grades based on the level of evidence to support them. Pediatric lipid guidelines were first published in 1992. These guidelines included a screening approach based on family history and recommended a population approach to improve diet and physical activity in all children and adolescents, as well as a high-risk approach. This approach focused on treatment with lifestyle or with pharmacologic agents for those identified at high risk. The 2011 Integrated Guidelines provide the most comprehensive and up-to-date approach to pediatric dyslipidemia. In these guidelines, universal screening in 9-11-year-olds is recommended to identify children with genetic dyslipidemia or more lifestyle- related dyslipidemia. Pharmacologic treatment is recommended only for a small group of children and adolescents with marked elevation of LDL-C due to genetic dyslipidemias. New guidelines from the American Heart Association/American College of Cardiology largely support the Integrated Guidelines.

 

CLINICAL PRACTICE AND GUIDELINE DEVELOPMENT

 

Clinical practice guidelines are becoming an increasingly important aspect of clinical care. Guidelines are designed to create a synthesis of evidence, including expert opinion where little evidence exists, to provide a straightforward approach to clinical decision making. Guideline development recognizes that the average practicing clinician has difficulty keeping abreast of developing medical science across a wide range of areas. This is especially true for generalists in primary care who must cover a wide range of medical issues with their patients. Guidelines are particularly helpful where there may be conflicting evidence or a range of levels of quality among studies included in the evidence base. In addition, when guidelines are widely utilized, they help to diminish unnecessary practice variation, improve outcomes, and potentially can reduce costs by providing a more efficient pathway to appropriate diagnosis and treatment while eliminating unnecessary tests and procedures.

 

 

While the value of well-done clinical practice guidelines is now widely accepted, concerns have been expressed historically about their development (1). These concerns include the fact that there were no standards for the guideline development process or guideline committee composition. This sometimes resulted in concerns about the balance in expertise. In addition, relationships between committee members and pharmaceutical companies or other entities were often not disclosed, making potential conflicts of interest difficult to discern. There also have been no universal standards for the approach to reviewing and grading the evidence. This can lead to selective inclusion of research or to different approaches to weighting of the evidence. There has also been no standard approach to translating the evidence review into graded recommendations, which is the aspect of the process that is most useful, and most used by clinicians. Often, clinical recommendations were presented as unanimous when, in fact, there was substantial discussion and even disagreement on the part of the committee members. This lack of a standard accepted process has sometimes led to clinical practice guidelines from different organizations that presented very different recommendations on the same topic, which potentially increases the confusion of clinicians even more.

 

As more experience has been gained with the process of guideline development, the process has improved over time. Presently, the government and health organizations which oversee guideline construction now generally focus on more balanced committee membership and a more transparent approach to potential conflicts of interest. They require completion of a documented review of the evidence, increased transparency of committee discussion, and improved identification of expert opinion in the guideline development process. However, key elements of the process remain controversial. Good guidelines require the development of good key questions at the onset of the process. Constructing the best key questions still seems more of an art than a scientific endeavor. In addition, different organizations have different approaches to grading evidence and to constructing and grading recommendations from the evidence. For example, some organizations will essentially accept only evidence derived from randomized controlled clinical trials. While those trials do represent the strongest science, they are also the most difficult and expensive studies to perform. Clinical trials by their nature often address very narrow scientific and clinical questions. In addition, there are many areas that remain unaddressed by clinical trials for a variety of reasons, including areas where such trials are difficult to perform or even may be considered unethical, as well as areas where funding for such studies has not been available.

 

In the past, clinical practice guidelines have been viewed as static documents. This is not appropriate as the science that informs clinical decision making is always evolving. In the case where the science is rapidly evolving, a guideline may be out of date shortly after it is published. Thus, guideline creation should best be viewed as a continuous improvement process with new studies reviewed and graded as they become available. Newer electronic data bases and electronic health records make this approach to ongoing refinement of guidelines more feasible.

 

Unfortunately, guidelines are often not implemented in practice. Research has demonstrated that there is often a lag, which can be as long as a decade or more between development and routine implementation of guidelines (2). This suggests that clinicians may be implementing treatment that is not supported by the best evidence. This is an area where more research is needed to determine best practices to encourage and enhance utilization of guidelines once they are developed.

 

GUIDELINES FOR PEDIATRIC DYSLIPIDEMIA

 

National Cholesterol Education Program (NCEP) 1992

 

The first guidelines on pediatric lipid management were developed by the National Cholesterol Education Program (NCEP) of the National Heart Lung and Blood Institute (NHLBI) and were published in 1992 (3). The guidelines were developed by a pediatric committee that worked in parallel with the NCEP adult panel of experts. The guideline construction did not involve a complete, formal evidence review with grading of the evidence.  Much of the report was based on expert opinion and extrapolation of data collected in adults to create an approach to pediatric patients. The report presented two approaches to pediatric dyslipidemia. The first was a population-based approach, which focused on diet and lifestyle issues for the entire population. The second was focused on identification and treatment of higher risk children and adolescents. The goal of the population approach was to prevent dyslipidemia from developing in the first place. This has come to be called primordial prevention. The population approach encourages healthy diet and physical activity for all children and adolescents. This approach includes all family members, as well.

 

The individualized approach aimed to identify and treat children and adolescents who are at greatest risk for having high blood cholesterol as adults and who had an increased lifetime risk of atherosclerotic cardiovascular disease. In the individual approach, the committee recommended selective screening of children who have a family history of premature cardiovascular disease or at least one parent with elevated serum cholesterol. This approach assumed that all adults would have their lipid levels tested as part of routine care. The committee considered universal screening, but decided that the selective screening approach would recognize the influence of genes and environment and would be more efficient. This selective screening approach, sometimes referred to as cascade screening, is used in many European countries to identify children and adolescents with familial hypercholesterolemia (FH). The committee also presented cut points for acceptable, borderline and high elevated LDL-C based on percentiles from the Lipid Research Clinical Prevalence Study (4). The panel then used these cut points to establish an approach to initiation of and goals for diet therapy. The panel developed separate cut points derived from studies of adults for initiation of drug therapy. They developed a two-step approach to diet therapy with Step 2 having greater restriction of saturated fat and cholesterol in the diet. For drug therapy, the panel recommended the use of bile acid sequestering agents for routine use. This report did not provide a focus on triglycerides or HDL-C and did not recommend the use of HMG CoA reductase inhibitors for pharmacologic therapy.

 

These 1992 Guidelines served as the approach to screening, diagnosis, and treatment for many years. They also served as the basis for research with investigators studying the effectiveness of a selective approach to screening and other aspects of the guidelines (5). In addition, clinical trials were launched to study the effect of dietary and pharmacologic intervention in children and adolescents with dyslipidemia (6,7).

 

As new evidence became available, some of which supported the 1992 Guidelines and others which suggested alternative approaches, organizations such as the American Academy of Pediatrics (8,9) and the American Heart Association (10,11) empaneled committees to produce guidelines and recommendations, which were refinements of the original 1992 guidelines. None of these efforts included a formal, complete review and grading of the evidence or grading of the recommendations.

 

United States Preventive Services Task Force 2016

 

In a parallel process, the United States Preventive Services Task Force (USPSTF) initiated a review of the evidence regarding cholesterol screening in children and adolescents (12). This review was updated in 2016. The USPSTF uses a formal evidence review and grading based on a series of key questions. The USPSTF has reported an “I” recommendation on lipid screening. This means that they found insufficient evidence for or against lipid screening in children and adolescents. This is a call for more research in this area.

 

There are several reasons why an “I” recommendation resulted from the USPSTF review of the evidence. The first has to do with the key questions asked as the framework for the review. A close inspection of the key questions demonstrates that several of the key questions are probably not answerable because the types of studies needed to answer the questions cannot reasonably be done. The USPSTF also requires a very high standard for research, including randomized clinical trials of screening, which are much less likely to be done in children than in adults.

 

The 2016 USPSTF review of cholesterol screening was improved in several ways compared to previous reviews (13-15). First, there was a separate analysis of the evidence to support screening for individuals with familial hypercholesterolemia. In previous USPSTF reviews, these individuals had been excluded from consideration. The 2016 USPSTF review also included a review of the evidence to screen for multifactorial dyslipidemia. The key questions were also modified somewhat from previous reviews. However, the answer for key questions, such as:

 

1)   Does screening for dyslipidemia in asymptomatic children and adolescents delay or reduce the incidence of myocardial infarction or stroke in adulthood, or

 

2)   Does treatment of dyslipidemia with lifestyle modification or lipid lowering medications in children and adolescents delay or reduce the incidence of adult myocardial infarction and stroke events?

 

These questions still require studies that are virtually impossible to do. Such studies would require randomization of young individuals and following them for decades to observe the outcomes. Utilization of these key questions make it quite difficult for the USPSTF to achieve anything other than an “I” statement for pediatric lipid screening.

 

There were several commentaries of the 2016 USPSF reviews that serve to put the results in broader context (16, 17). These commentaries pointed out that a statement of insufficient evidence for or against lipid screening was not particularly helpful for the clinician on the front line and that other health organizations, such as the American Heart Association and the American Academy of Pediatricians have recommended lipid screening in children and adolescents based on separate review and grading of the evidence.

 

It is important to note that an “I” statement from the USPSTF should not be taken as a recommendation against lipid screening. The USPSTF does recommend against screening when the evidence demonstrates that screening or treatment are ineffective or harmful. In the face of an “I” statement and given the high bar for evidence required by the USPSTF, it is up to individual clinicians and health organizations to weight the available evidence and decide on a course of action.

 

National Heart Lung and Blood Institute (NHLBI) 2011

 

In 2011, the results of an NHLBI panel, which performed a complete review and grading of the evidence for screening and treatment of cardiovascular disease risk factors in children and adolescents, including dyslipidemia, were published as part of an integrated approach to CVD risk factor evaluation and management (18, 19). These Integrated Guidelines represent the most comprehensive, up-to-date approach to lipid screening, diagnosis, and treatment and are a departure from previous guidelines. First, the guidelines recommended universal screening for lipid disorders. This means that all children should have their lipids tested one time between the ages of 9-11. This can be performed with either a fasting lipid profile or a non-fasting test to evaluate non-HDL-C. This universal approach was recommended because studies showed that using only a selective screening approach based on family history would potentially miss 30-60% of children and adolescents with substantial elevations of cholesterol (5). The universal screening approach is largely designed to identify children with genetic dyslipidemia, such as familial hypercholesterolemia. However, it will also identify children with dyslipidemia, largely elevated triglycerides and low HDL-C, due to lifestyle factors and obesity.

 

The Integrated Guidelines continued to support both a population and a high-risk approach to dyslipidemia. The recommendation for diet for the general population is the Cardiovascular Health Integrated Lifestyle Diet (CHILD) 1. For higher-risk patients identified through screening, the CHILD 2-LDL diet was recommended if the LDL-C was elevated. This diet further restricts intake of saturated fat and cholesterol in the diet. For those with elevated triglycerides and low HDL-C, the CHILD 2-TG diet was recommended. This diet includes reduced intake of simple sugars in addition to reduction in saturated fat.

 

The Integrated Guidelines presented statin agents as first-line pharmacologic treatment for substantial elevation of LDL-C (>190mg/dL) with no other risk factors, or >160mg/dL with 1 high level or ≥ 2 moderate-level risk factors in children and adolescents age 10 years and older.

 

The Integrated Guidelines have not been without controversy (20-22). In addition, uptake of the Integrated Guidelines has been less than optimum (23,24). One potential reason for confusion regarding the guidelines is the potential concern about the impact of obesity on dyslipidemia. This result, in part, derives from a misunderstanding of the difference between the issues related to genetic forms of dyslipidemia, such as FH, and those that are largely due to lifestyle. It needs to be clarified that most individuals who have an LDL-C level in the range where medication would be recommended have a genetic form of dyslipidemia, usually heterozygous FH. Children and adolescents with lifestyle-based dyslipidemia rarely have LDL-C levels that would trigger the recommendation for pharmacologic treatment. Obesity results in elevated triglycerides and low HDL-C with only a modest increase in LDL-C. These children and adolescents should be treated with changes in lifestyle, including a more healthful diet and increased levels of physical activity. Estimates are that fewer than 1% of children and adolescents would qualify for statin treatment (25).

 

American Heart Association (AHA) and the National Lipid Association (NLA)

 

This potential confusion over different aspects of dyslipidemia and their consequences have led to American Heart Association (AHA) and the National Lipid Association (NLA) to sharpen the focus on familial hypercholesterolemia (25-27). While these scientific statements did not include a formal review and grading of the evidence, they provided a new focus for clinicians and may simplify the clinical approach to pediatric dyslipidemia. Clinicians should probably focus first on identification and treatment of individuals with the array of genetic defects that underlie FH and their family members who also have this genetic abnormality. Because this genetic defect occurs in approximately 1:250 individuals, it is one of the most prevalent genetic diseases. Individuals with heterozygous FH have substantial and often marked elevation of LDL-C. These individuals have been shown to be at increased lifetime risk of atherosclerotic CVD and are at risk for adverse outcomes in their 30’s, 40’s, 50’s and 60’s. Unfortunately, the first clinical sign of the disease for these patients may be a myocardial infarction or sudden cardiac death. Because this is often an asymptomatic condition, particularly in childhood, lipid testing is essentially the only way to identify affected individuals. Treatment with statins and other pharmacologic agents can be quite effective at lowering LDL-C levels and decreasing the risk for adverse cardiovascular outcomes.

 

American Heart Association/American College of Cardiology Cholesterol Clinical Practice Guidelines 2018

 

The most recent clinical practice guidelines regarding dyslipidemia are the American Heart Association/American College of Cardiology Cholesterol Clinical Practice Guidelines published in 2018 (28). These guidelines included a complete evidence review and systematic grading of the evidence and the recommendations. These guidelines largely focus on the management of blood cholesterol in adults, but also included a section on children. These guidelines indicate that, in children (age 10-19 years of age) and young adults (20-39 years of age), priority should be given to evaluation of lifetime risk of atherosclerotic cardiovascular disease and promotion of lifestyle risk reduction.

 

In the AHA/ACC 2018 guidelines, screening for dyslipidemia based on family history is given a B-nonrandomized level of evidence and a IIa strength of recommendation (28). Universal screening for dyslipidemia once between age 9-11 years and once between age 17-21 is given a B-nonrandomized level of evidence and a IIb strength of recommendation. The B-NR level of evidence indicates moderate quality of evidence from observational studies. The class IIa recommendation is a moderate recommendation, while a class IIb recommendation is considered weaker (might be reasonable).

 

For treatment of dyslipidemia in children and adolescents, lifestyle approaches receive a level A for the evidence and have a class I strength of recommendation. For children and adolescents age 10 and over with an LDL-C persistently above 190mg/dL or above 160mg/dL with a clinical presentation consistent with familial hypercholesterolemia who do not adequately respond to lifestyle change after 3-6 months, initiating statin therapy received a B-randomized level of evidence and a class IIa recommendation (22).

 

These newest guidelines are essentially in line with the 2011 Integrated Guidelines from the NHLBI. However, they also emphasize that more high-quality evidence is needed. This should drive research efforts in the areas of screening and management for pediatric dyslipidemia.

 

CONCLUSION

 

In conclusion, the evidence related to risk, identification, and effective treatment of dyslipidemia has continued to expand. This has allowed development of guidelines for management of pediatric patients with dyslipidemia. Unfortunately, uptake of these guidelines by primary care clinicians has been slow. There is a need for ongoing high-quality studies in this area so that new study results can be included in subsequent evidence reviews and clinical practice guidelines can be improved.

 

A major limiting factor in the development of Guidelines regarding the screening, identification, and treatment of dyslipidemia in children and adolescents is the lack of studies which produce the evidence to support such guidelines. There are examples of guidelines in pediatric healthcare that have been well accepted based on evidence. These include US Preventive Services Task Force recommendations for screening for obesity using Body Mass Index (22), guidelines for the diagnosis and management of asthma from the National Heart, Lung and Blood Institute (23), and for the diagnosis and management of an initial urinary tract infection in febrile infants from the American Academy of Pediatrics (24). These guidelines have generally been accepted in pediatric practice, although not always without controversy (25). As we seek to improve outcomes through better standardization of delivery of healthcare, improved evidence-based guidelines will be increasingly important.

 

REFERENCES

 

  1. Sniderman AD, Furberg Why guideline-making requires reform. JAMA. 2009;301:429- 31.
  2. Grimshaw JM, Thomas RE, MacLennan G, Fraser C, Ramsay CR, Vale L, Whitty P, Eccles MP, Matowe L, Shirran L, Wensing M, Dijkstra R, Donaldson C. Health Technol Assess. 2004;8:iii-iv, 1-72.
  3. National Cholesterol Education Program (NCEP): highlights of the report of the Expert Panel on Blood Cholesterol Levels in Children and Pediatrics. 1992;89:495- 501.
  4. Tamir I, Heiss G, Glueck CJ, Christensen B, Kwiterovich P, Rifkind BM. Lipid and lipoprotein distributions in white children ages 6-19 The Lipid Research Clinics Program Prevalence Study. J Chronic Dis. 1981;34:27-39.
  5. Ritchie SK1, Murphy EC, Ice C, Cottrell LA, Minor V, Elliott E, Neal Universal versus targeted blood cholesterol screening among youth: The CARDIAC project. Pediatrics. 2010;126:260-5.
  6. The Writing Group for the DISC Collaborative Research Group. Efficacy and safety of lowering dietary intake of fat and cholesterol in children with elevated low-density lipoprotein The Dietary Intervention Study in Children (DISC). The Writing Group for the DISC Collaborative Research Group. JAMA. 1995;273:1429-35
  7. McCrindle BW1, Ose L, Marais AD. Efficacy and safety of atorvastatin in children and adolescents with familial hypercholesterolemia or severe hyperlipidemia: a multicenter, randomized, placebo-controlled trial. J Pediatr. 2003;143:74-80.
  8. American Academy of Committee on Nutrition. American Academy of Pediatrics. Committee on Nutrition. Cholesterol in childhood. Pediatrics. 1998;101(1 Pt 1):141-7.
  9. Daniels SR, Greer FR; Committee on Lipid screening and cardiovascular health in childhood. Pediatrics. 2008;122:198-208.
  10. Kavey RE, Daniels SR, Lauer RM, Atkins DL, Hayman LL, Taubert K; American Heart Association. American Heart Association guidelines for primary prevention of atherosclerotic cardiovascular disease beginning in Circulation. 2003;107:1562- 6.
  11. McCrindle BW, Urbina EM, Dennison BA, Jacobson MS, Steinberger J, Rocchini AP, Hayman LL, Daniels SR; American Heart Association Atherosclerosis, Hypertension, and Obesity in Youth Committee; American Heart Association Council of Cardiovascular Disease in the Young; American Heart Association Council on Cardiovascular Nursing. Drug therapy of high-risk lipid abnormalities in children and adolescents: a scientific statement from the American Heart Association Atherosclerosis, Hypertension, and Obesity in Youth Committee, Council of Cardiovascular Disease in the Young, with the Council on Cardiovascular Nursing. Circulation. 2007;115:1948-67.
  12. US Preventive Services Task Screening for lipid disorders in children: US Preventive Services Task Force recommendation statement. Pediatrics. 2007;120:e215-9.
  13. US Preventive Services Task Force. Screening for lipid disorders in children and adolescents: US Preventive Services Task Force recommendation statement. JAMA. 2016;316:625-33.
  14. Lozano P, Henrikson NB, Dunn J, Morrison CC, Nguyen M, Blasi PR, Anderson L, Whitlock EP. Lipid screening in childhood and adolescence for detection of familial hypercholesterolemia: Evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2016;316(6):645-55.
  15. Lozano P, Henrikson NB, Morrison CC, Dunn J, Nguyen M, Blasi PR, Whitlock EP. Lipid screening in childhood and adolescence for detection of multifactorial dyslipidemia: Evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2016;316(6):634-44.
  16. Urbina EM, de Ferranti SD. Lipid screening in children and adolescents. JAMA. 2016;316(6):589-91.
  17. Daniels SR. On the US Preventive Services Task Force Statement on screening for lipid disorders in children and adolescents: One step forward and 2 steps sideways. JAMA Pediatrics. 2016;170(10):932-34.
  18. Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents; National Heart, Lung, and Blood Institute. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents: summary report. Pediatrics. 2011;128 Suppl 5:S213-56.
  19. Gidding SS, Daniels SR, Kavey RE; Expert Panel on Cardiovascular Health and Risk Reduction in Developing the 2011 Integrated Pediatric Guidelines for Cardiovascular Risk Reduction. Pediatrics. 2012;129:e1311-9.
  20. Newman TB, Pletcher MJ, Hulley Overly aggressive new guidelines for lipid screening in children: evidence of a broken process. Pediatrics. 2012;130:349-52.
  21. McCrindle BW1, Kwiterovich PO, McBride PE, Daniels SR, Kavey Guidelines for lipid screening in children and adolescents: bringing evidence to the debate. Pediatrics. 2012;130:353-6.
  22. Gillman MW, Daniels Is universal pediatric lipid screening justified? JAMA. 2012;307:259-60.
  23. Valle CW, Binns HJ, Quadri-Sheriff M, Benuck I, Patel Physicians' Lack of Adherence to National Heart, Lung, and Blood Institute Guidelines for Pediatric Lipid Screening. Clin Pediatr (Phila). 2015;54:1200-5.
  24. de Ferranti SD, Rodday AM, Parsons SK, Cull WL, O’Connor KG, Daniels SR, Leslie LK. Cholesterol screening and treatment practices and preferences: A survey of United States pediatricians. J Pediatr. 2017;185:99-105.
  25. McCrindle BW1, Tyrrell PN, Kavey Will obesity increase the proportion of children and adolescents recommended for a statin? Circulation. 2013;128:2162-5.
  26. Gidding SS, Champagne MA, de Ferranti SD, Defesche J, Ito MK, Knowles JW, McCrindle B, Raal F, Rader D, Santos RD, Lopes-Virella M, Watts GF, Wierzbicki AS; American Heart Association Atherosclerosis, Hypertension, and Obesity in Young Committee of Council on Cardiovascular Disease in Young, Council on Cardiovascular and Stroke Nursing, Council on Functional Genomics and Translational Biology, and Council on Lifestyle and Cardiometabolic Health. The Agenda for Familial Hypercholesterolemia: A Scientific Statement From the American Heart Association. Circulation. 2015;132:2167-92.
  27. Goldberg AC, Hopkins PN, Toth PP, Ballantyne CM, Rader DJ, Robinson JG, Daniels SR, Gidding SS, de Ferranti SD, Ito MK, McGowan MP, Moriarty PM, Cromwell WC, Ross JL, Ziajka PE. Familial hypercholesterolemia: screening, diagnosis and management of pediatric and adult patients: clinical guidance from the National Lipid Association Expert Panel on Familial J Clin Lipidol. 2011;5:133-40.
  28. Writing Committee, Cholesterol Clinical Practice Guidelines. 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. Circulation. 2019;139:e1046-e1081.

 

Gynecomastia: Etiology, Diagnosis, and Treatment

ABSTRACT

 

Gynecomastia is a relatively common disorder. Its causes range from benign physiological processes to rare neoplasms. To diagnose the etiology of the gynecomastia, the clinician must understand the hormonal factors involved in breast development. Parallel to female breast development, estrogen, growth hormone (GH), and IGF-1 are required for breast growth in males. Since a balance exists between estrogen and androgens in males, any disease state or medication that increases circulating estrogens or decreases circulating androgens, causing an elevation in the estrogen to androgen ratio, can induce gynecomastia. Due to the diversity of possible etiologies, including a neoplasm, performing a careful history and physical is imperative. Once gynecomastia has been diagnosed, treatment of the underlying cause is warranted. If no underlying cause is discovered, then close observation is appropriate. If the gynecomastia is severe and of recent onset, medical therapy can be attempted, and if ineffective, glandular tissue can be removed surgically.

 

INTRODUCTION

 

This chapter reviews the ontogeny and physiology of breast development; factors that influence breast enlargement in the male; the differential diagnosis of gynecomastia; the process of diagnostic investigation; and treatment of gynecomastia.

 

BREAST ONTOGENY AND DEVELOPMENT

 

Male breast development occurs in an analogous fashion to female breast development. At puberty in the female, a complex hormonal interplay occurs resulting in growth and maturation of the adult female breast.

 

In early fetal life, epithelial cells, derived from the epidermis of the area programmed to become the areola, proliferate into ducts, which connect to the nipple at the skin's surface. The blind ends of these ducts bud to form alveolar structures in later gestation. With the decline in fetal prolactin and placental estrogen and progesterone at birth, the infantile breast regresses until puberty (1).

 

During thelarche in females, the initial clinical appearance of the breast bud and growth and division of the ducts occur, giving rise to club-shaped terminal end buds, which then form alveolar buds. Approximately a dozen alveolar buds will cluster around a terminal duct, forming the type 1 lobule. The type 1 lobule will mature into types 2 and 3 lobules, called ductules. The number of alveolar buds increases to as many as 50 in type 2 and 80 in type 3 lobules. The entire differentiation process takes years after the onset of puberty and, if pregnancy is not achieved, may never be completed (2). On the contrary, there is usually no further development of breast because of the rising testosterone concentrations at puberty. Some peri-pubertal boys may transiently develop type 1 lobules that may undergo atrophy at a later stage.

 

HORMONAL REGULATION OF BREAST DEVELOPMENT

 

The initiation and progression of breast development involves a coordinated effort of pituitary and ovarian hormones, as well as local mediators (Figure 1).

Figure 1. Hormones affecting growth and differentiation of breast tissue.

ESTROGEN, GH AND IGF-1, PROGESTERONE, & PROLACTIN

 

Estrogen and progesterone act in an integrative fashion to stimulate normal adult female breast development. Estrogen, acting through its estrogen receptor (ER), promotes ductal growth, while progesterone, acting through its receptor (PR), promotes alveolar development (1). This is demonstrated by experiments in ER knockout mice that display grossly impaired ductal development, whereas PR knockout mice possess significant ductal development, but lack alveolar differentiation (3, 4).

 

Although estrogens and progestogens are vital to mammary growth, they are ineffective in the absence of anterior pituitary hormones (5). Thus, neither estrogen alone nor estrogen plus progesterone can sustain breast development without other mediators, such as GH and IGF-1. This was confirmed by studies involving the administration of estrogen and GH to hypophysectomized and oophorectomized female rats that resulted in breast ductal development. The GH effects on ductal growth are mediated through stimulation of IGF-1. This is demonstrated by studies of estrogen and GH administration to IGF-1 knockout rats that showed significantly decreased mammary development when compared to age-matched IGF-1- intact controls. Combined estrogen and IGF-1 treatment in these IGF-1 knockout rats restored mammary growth (6, 7). In addition, Walden et al. demonstrated that GH-stimulated production of IGF-1 mRNA in the mammary gland itself, suggesting that IGF-1 production in the stromal compartment of the mammary gland acts locally to promote breast development (8). Furthermore, other data indicates that estrogen promotes GH secretion and increases GH levels, stimulating the production of IGF-1, which synergizes with estrogen to induce ductal development. In a population-based study of healthy boys and adolescents, IGF-I levels were found to be elevated in boys with pubertal gynecomastia compared with boys without gynecomastia suggesting that the GH-IGF-I axis may be involved in the pathogenesis of pubertal gynecomastia (9). Indeed, dating back to 1950, it had already been reported that gynecomastia was found in a young patient with acromegaly (97).

 

Progesterone has minimal effects on breast development without concomitant anterior pituitary hormones, indicating that progesterone also interacts closely with pituitary hormones. For example, prolonged treatment of dogs with progestogens such as depot medroxyprogesterone acetate or with proligestone was associated with increased GH and IGF-1 levels, suggesting that progesterone may stimulate GH secretion (10). In addition, clinical studies have correlated maximal cell proliferation to specific phases in the female menstrual cycle. For example, maximal proliferation occurs not during the follicular phase when estrogens reach peak levels and progesterone is low (less than 1 ng/mL [3.1nmol/mL]), but rather, it occurs during the luteal phase when progesterone reaches concentrations of 10-20 ng/mL (31- 62 nmol/mL) and estrogen concentrations are two to three times lower than in the follicular phase (11). Furthermore, immunohistochemical studies of ER and PR showed that the highest percentage of proliferating cells, found almost exclusively in the type 1 lobules, contained the highest percentage of ER and PR positive cells (2). Similarly, there is immunocytological presence of ER, PR, and androgen receptors (AR) in gynecomastia and male breast carcinoma. ER, PR and AR expression was observed in 100% (30/30) of gynecomastia cases (12). Given these data and the fact that PR knockout mice lack alveolar development in breast tissue, it seems that progesterone, analogous to estrogen, increases GH secretion and acts through the PR on mammary cells to enhance breast development and alveolar differentiation (13, 4).

 

Prolactin is another anterior pituitary hormone integral to breast development. Prolactin is not only secreted by the pituitary gland but may be produced by normal mammary tissue epithelial cells and breast tumors (14, 15). Prolactin stimulates epithelial cell proliferation only in the presence of estrogen and enhances lobulo-alveolar differentiation only with concomitant progesterone. Gynecomastia is seen rarely in hyperprolactinemia, possibly because of the low estrogen levels due to suppression of LH secretion. Previously, receptors for LH/ human chorionic gonadotropin (hCG) have been found in male and female breast tissues, but their functional roles remain to be determined (16).

 

ANDROGEN AND AROMATASE

 

Estrogen effects on the breast is the result of circulating estradiol levels or locally produced estrogens. Aromatase P450 catalyzes the conversion of the C19 steroids-- androstenedione, testosterone, and 16- α- hydroxyandrostenedione-- to estrone, estradiol-17β and estriol. As such, an overabundance of substrate (e.g., testosterone) or an increased enzyme activity (aromatase) for estrogen production can increase serum and breast estrogen concentrations and initiate the cascade to breast development in females and males. For example, in the more complete forms of androgen insensitivity syndromes in genetically male (XY) patients, excess androgen is aromatized into estrogen that results in gynecomastia and an overall phenotypic female appearance. Furthermore, the loss of the anti-proliferative effect of androgens on breast also contributes significantly to breast development in XY individuals with complete androgen insensitivity. Likewise, the biologic effects of over-expression of the aromatase enzyme in female and male mice transgenic for the aromatase gene result in increased breast proliferation. In female transgenics, over-expression of aromatase promotes the induction of hyperplastic and dysplastic changes in breast tissue. Over-expression of aromatase in male transgenics causes increased mammary growth and histological changes similar to gynecomastia, increased estrogen and progesterone receptors, and increased downstream breast growth factors such as TGF-beta and βFGF (17). Interestingly, treatment with an aromatase inhibitor leads to involution of the mammalian gland phenotype (18). Thus, although androgens do not stimulate breast development directly, they may do so if they aromatize to estrogen. This occurs in cases of androgen excess or in patients with increased aromatase activity.

 

PHYSIOLOGIC GYNECOMASTIA

 

Gynecomastia, breast development in males, can occur normally during three phases of life. The first occurs shortly after birth in both males and females. This is partly caused by the high fetal blood levels of estradiol and progesterone (produced by the mother) that stimulate breast tissue in the newborn. Another mechanism is the increased conversion of steroid hormone precursors to sex steroids and increased aromatization of androgen as a result of neonatal surge of luteinizing hormone (LH). Neonatal gynecomastia may persist for several weeks after birth and may be associated with a milky breast discharge called "witch's milk" (2).

 

Puberty marks the second period when gynecomastia can occur physiologically. In fact, up to 60% of boys have clinically detectable gynecomastia by age 14. Although it is mostly bilateral, it is often asymmetrical and can occur unilaterally. Pubertal gynecomastia usually resolves within 3 years of onset (2). In early puberty, the pituitary gland releases gonadotropins at night and stimulates testicular production of testosterone during the very early morning hours. Serum estradiol concentration, however, remains elevated above prepubertal concentrations throughout the day. Compared with boys who do not develop gynecomastia, boys with pubertal gynecomastia have a decreased androgen to estrogen ratio (19, 100). Furthermore, another study showed increased aromatase activity in the skin fibroblasts of boys with gynecomastia (103). Thus, the mechanism by which pubertal gynecomastia occurs may be due to either decreased production of androgens or increased aromatization of circulating androgens, thus increasing the estrogen to androgen ratio (20).

 

The third age range in which gynecomastia is frequently seen is during older age (>60 years). The reported prevalence varies from 36 to 57%, possibly because of different selected populations and different diagnostic criteria (101). Although the exact mechanisms by which this occurs have not been fully elucidated, evidence suggests that it may result from increased peripheral aromatase activity secondary to increased total body fat, relatively elevated LH concentrations, and decreased serum testosterone concentrations associated with male aging. For instance, investigators have shown increased urinary estrogen concentrations in obese individuals and have demonstrated aromatase expression in adipose tissue (21). Thus, like the gynecomastia of obesity, the gynecomastia of aging may partly result from increased aromatase activity, causing increased conversion of androgens to estrogens (22). Moreover, not only does total body fat increase with age, but there may be an increase in aromatase activity in the adipose tissue already present, further increasing circulating estrogens. Serum sex hormone binding globulin (SHBG) concentrations increase with age in men. Because SHBG binds estrogen with less affinity than testosterone, the bioavailable estradiol to bioavailable testosterone ratio may increase in older men. Lastly, elderly patients may take multiple medications associated with gynecomastia. One cohort study suggests medications play a role in 80% of gynecomastia in older men (23).     

 

PATHOLOGIC GYNECOMASTIA

 

Pathologic gynecomastia is due to an increase in the circulating and/or local breast tissue ratio of estrogen to androgen.

 

Increased Estrogen

 

Breast development requires the presence of estrogen. Androgens, on the other hand, have anti-proliferative effects on breast tissue. Thus, an equilibrium exists between estrogen and androgens in the adult male to prevent growth of breast tissue, whereby either an increase in estrogen or a decrease in androgen can tip the balance toward gynecomastia. Increased estrogen levels will increase glandular proliferation by several mechanisms. These include direct stimulation of glandular tissue and by suppressing LH, therefore decreasing testosterone secretion by the testes and exaggerating the already high estrogen to androgen ratio. Since the development of breast tissue in males occurs in an analogous manner to that in females, the same hormones that affect female breast tissue can cause gynecomastia. In post-pubertal boys and adult men, the testes secrete 6-10 µg of estradiol and 2.5 µg of estrone per day. Testicular production comprises a small fraction of estrogens in circulation (i.e., 15% of estradiol and 5% of estrone), and the remainder of estrogen in men is derived from the extraglandular aromatization of testosterone and androstenedione to estradiol and estrone (24). Thus, any cause of estrogen excess from overproduction to peripheral aromatization of androgens can initiate the cascade to breast development.

 

TUMORS

 

Testicular tumors can lead to increased blood estrogen levels by the following mechanisms: estrogen overproduction, androgen overproduction with extragonadal aromatization to estrogens, and secretion of hCG that stimulates normal Leydig cells (via the LH receptor). Tumors causing an overproduction of estrogen represent an unusual but important cause of estrogen excess. Examples of estrogen-secreting tumors include Leydig cell tumors, Sertoli cell tumors, granulosa cell tumors, and adrenal tumors.

 

Leydig cell tumors constitute 1%-3% of all testicular tumors. Usually, they occur in men between the ages of 20 and 60, although up to 25% of them occur prepubertally. In prepubertal cases, isosexual precocity, rapid somatic growth, and increased bone age with elevated serum testosterone and urinary 17-ketosteroid levels are the presenting features. In adults, elevated estrogen levels coupled with a palpable testicular mass and gynecomastia suggests a testicular tumor. Of note, feminization (particularly gynecomastia) is common in adults, but it is rare in boys. Some Leydig cell tumors may only be apparent on ultrasound because of their small size; some may produce testosterone and do not cause gynecomastia. Though mostly benign, Leydig cell tumors may be malignant and metastasize to lung, liver, and retroperitoneal lymph nodes (25, 26).

 

Sertoli cell tumors comprise less than 1% of all testicular tumors and occur at all ages, but one third have occurred in patients less than 13 years, usually in boys under 6 months of age. Although they arise in young boys, they usually do not produce endocrine effects in children. Again, the majority are benign, but up to 10% are malignant. Gynecomastia occurs in one third of cases of Sertoli cell tumors, presumably due to increased estrogen production (26). Sertoli cell tumors in boys with Peutz-Jegher syndrome, an autosomal dominant disease characterized by pigmented macules on the lips, gastrointestinal polyposis, and hormonally active tumors in males and females, for instance, have aromatase overactivity, resulting in gynecomastia, rapid growth, and advanced bone age as presenting features (29, 30, 31). Feminizing Sertoli cell tumors with increased aromatase activity can also be seen in the Carney complex, an autosomal dominant disease characterized by cardiac myxomas, cutaneous pigmentation, adrenal nodules and hypercortisolism

 

Granulosa cell tumors, occurring very rarely in the testes, can also overproduce estrogen. Gynecomastia at presentation was reported in some cases (27).

 

Germ cell tumors are the most common cancer in males between the ages of 15 and 35. They are divided into seminomatous and non-seminomatous subtypes and include embryonal carcinoma, yolk sac carcinoma, choriocarcinoma, and teratoma. Elevated serum hCG or hCG subunits (e.g., may be present in both seminomatous and non-seminomatous types of germ cell tumors), whereas AFP may be elevated only in the non-seminomatous type. As a result of the increased hCG that stimulates the Leydig cell via the LH receptor, testicular testosterone and estrogen (estrogen out of proportion to testosterone) production is increased and may cause gynecomastia. Although germ cell tumors generally arise in the testes, they can also originate extragonadally, specifically in the mediastinum. These extragonadal tumors also possess the capability of producing hCG, but they must be differentiated from a multitude of other tumors such as large cell carcinomas of the lung that can synthesize hCG or hCG subunits (28).

 

Some neoplasms that overproduce estrogens also possess aromatase overactivity. Other than sex-cord tumors, fibrolamellar hepatocellular carcinoma has also been shown to possess ectopic aromatase activity, causing severe gynecomastia in two boys (32, 33).

 

Furthermore, adrenal tumors can secrete excess dehydroepiandrosterone (DHEA), DHEA-sulfate (DHEAS), and androstenedione that can then be aromatized peripherally to estradiol. Some adrenal tumors may secret estrogen directly. Typically, feminizing-adrenal tumors are large, aggressive, and malignant (90).    

 

Table 1. Tumors Causing Gynecomastia

Tumor type

Hormone produced

Aromatase overactivity

Leydig cell tumor

Testosterone, estrogen  

 

Sertoli cell tumor

Estrogen

+ (in Peutz-Jegher syndrome), + (in Carney complex)

Granulosa cell tumor

Estrogen

 

Adrenal tumor

Estrogen, dehydroepiandrosterone (DHEA), dehydroepiandrosterone-sulfate (DHEA-S), and androstenedione that are converted in the periphery to estrogens.

 

Gonadal germ cell tumor

hCG and β-hCG

 

Extragonadal germ cell tumor e. G lung, gastric, renal cell and hepatocellular carcinoma

hCG and β-hCG

 

 

NON-TUMOR CAUSES OF ESTROGEN EXCESS

 

Increased Aromatase Activity

 

Besides tumors, other conditions have also been associated with excessive aromatization of testosterone and other androgens to estrogen leading to gynecomastia. For instance, obesity is strongly associated with gynecomastia. The mechanism is thought to be related to the increased aromatase activity in adipose tissues, but most obese men do not have high estrogen concentrations (104). Leptin has also been implicated in the pathogenesis of gynecomastia because it might stimulate aromatase in adipose and breast tissue. Leptin might also directly stimulate the growth of epithelial cells in the breast or enhance the sensitivity of epithelial cells to estrogen (98). There is a very rare familial form of gynecomastia in which affected family members have an elevation of extragonadal aromatase activity (34). Novel gain-of-function mutations in chromosome 15 have been reported to cause gynecomastia, possibly by forming cryptic promoters that lead to over expression of aromatase (35). Polymorphism of the aromatase cytochrome P45019 (CYP19) has also been found to be associated with gynecomastia (36).

 

Displacement of Estrogens From SHBG   

 

Another cause of gynecomastia from estrogen excess includes steroid displacement from SHBG. SHBG binds androgens more avidly than estrogen. Thus, any condition or drug such as spironolactone that displaces steroids from SHBG more easily displace estrogen than testosterone, resulting in a higher estrogen to testosterone ratio. Drugs can cause gynecomastia by numerous mechanisms besides displacement from SHBG. These drugs and their mechanisms are discussed below.

 

Decreased Testosterone and Androgen Resistance  

 

Besides increased estrogen production, decreased testosterone levels can cause an elevation in the estrogen to androgen ratio, thereby producing gynecomastia. Primary hypogonadism, with its reduction in serum testosterone and increased serum LH levels increases aromatization of testosterone to estradiol and is associated with an increased estrogen to androgen ratio. Klinefelter syndrome occurs in 1 in 600-700 males and is caused by supernumerary X chromosomes (XXY or XXXY karyotype) and primary testicular failure and often prominent gynecomastia, due to decreased testosterone production, compensatory increased LH secretion, overstimulation of the Leydig cells, and relative estrogen excess. In addition, any acquired testicular disease resulting in primary hypogonadism such as severe, postpubertal viral and bacterial orchitis, or scrotal trauma or radiation can promote gynecomastia by the same mechanisms (24). Lastly, enzyme deficiencies in the testosterone synthesis pathway from cholesterol also result in depressed testosterone levels and hence a relative increase in estrogen. Deficiency of 17-oxosteroid reductase, the enzyme that catalyzes the conversion of androstenedione to testosterone and estrone and estrone to estradiol, for example, will cause elevation in estrone and androstenedione, which is then further aromatized to estradiol (22).

 

Secondary hypogonadism results in low serum testosterone and unopposed estrogen effect from increased conversion of adrenal precursors to estrogens (24). Thus, patients with Kallmann syndrome, a form of congenital secondary hypogonadism with anosmia, also develop gynecomastia. In fact, androgen deficiency (hypogonadism) from whatever cause constitutes most cases of gynecomastia.

 

The androgen resistance syndromes, including complete and partial testicular feminization are characterized by gynecomastia and varying degrees of pseudohermaphroditism. Kennedy disease, a neurodegenerative disease, is caused by an increased number of CAG (polyglutamine) repeats in the androgen receptor gene that leads to a decrease in sensitivity of the receptor (2). The gynecomastia is the combined result of decreased androgen responsiveness at the breast level and increased estrogen production as a result of elevated androgen precursors of estradiol and estrone. Androgen resistance at the pituitary results in elevated serum LH levels and increased circulating testosterone. The increased serum testosterone is then aromatized peripherally, promoting gynecomastia.

 

Other Diseases  

 

Men with end-stage renal disease may have reduced testosterone and elevated gonadotropins. This apparent primary testicular failure may then lead to increased breast development (13). The gynecomastia of liver disease, particularly cirrhosis, does not have a clear etiology. Cirrhosis is associated with increased SHBG that binds testosterone more avidly than estrogen. Some have speculated that the gynecomastia is the result of estrogen overproduction, possibly secondary to increased extraglandular aromatization of androstenedione, which may have decreased hepatic clearance in cirrhosis. However, testosterone administration to patient with cirrhosis causes a rise in estradiol, but decreases the prevalence of gynecomastia (5, 37, 38). Therefore, although the association of gynecomastia with liver disease is apparent, current data are conflicting and the mechanism remains unclear.

 

Thyrotoxicosis is associated with gynecomastia. Patients often have elevated estrogen that may result from a stimulatory effect of thyroid hormone on peripheral aromatase. In addition, LH is also increased in many men with thyrotoxicosis, and LH also stimulates aromatization of testosterone (13,39, 96). Furthermore, thyroxine stimulates production of SHBG in the liver. Because SHBG binds testosterone more avidly than estradiol, there is a higher ratio of free estradiol to free testosterone. Thus, with normal testosterone and increased estrogen, there is an elevated free estrogen to testosterone ratio.

 

Gynecomastia is associated with spinal cord disorders. Most patients with spinal cord disorders often have low testosterone levels and, in fact, can develop testicular atrophy with resultant hypogonadism and infertility, which may be exacerbated by increased scrotal temperature. The exact mechanism, however, remains elusive (40).

 

Refeeding gynecomastia refers to breast development in men recovering from a malnourished state (1). Although most cases regress within several months, the etiology of this phenomenon has not been fully elucidated.

 

HIV patients can also develop gynecomastia. There is a high incidence of androgen deficiency due to multifactorial causes, including primary and secondary hypogonadism and certain drugs used to treat HIV (e.g., efavirenz) (24).

 

Drugs

 

About 20% of gynecomastia is caused by medications or exogenous chemicals (41). Some drugs may increase estrogen effect by several mechanisms: 1) they possess intrinsic estrogen-like properties, 2) they increase endogenous estrogen production, or 3) they supply an excess of an estrogen precursor (e.g., testosterone or androstenedione) that can be aromatized to estrogen. Examples of drugs that cause gynecomastia are listed in Tables 2 and 3. Contact with estrogen vaginal creams, for instance, can elevate circulating estrogen levels. Since some of the creams contain synthetic estrogens, they might not be detected by standard estrogenic qualitative assays. An estrogen-containing embalming cream has been reported to cause gynecomastia in morticians (42, 43). A topical estrogen spray, used for relief of menopausal hot flushes may lead to gynecomastia in children through skin contact (44). Recreational use of marijuana, heroin, methadone, and amphetamines has also been associated with gynecomastia (45). Herbs containing phytoestrogen or ginseng with estrogen-like structure (46) may also lead to gynecomastia. It has been suggested that digitalis causes gynecomastia due to its ability to bind to estrogen receptors (13, 47). The appearance of gynecomastia has been described in body builders and athletes after the administration of aromatizable androgens. The gynecomastia was presumably caused by an excess of circulating estrogens due to the conversion of androgens to estrogen by peripheral aromatase enzymes (48).

 

Table 2. Drugs That May Induce Gynecomastia by Known or Proposed Mechanisms  

Estrogen-like, or binds to estrogen receptor

Stimulate estrogen synthesis

Supply aromatizable estrogen precursors

Direct Testicular Damage

Block testosterone synthesis

Block androgen action

Displace estrogen from SHBG

Estrogen vaginal cream

Gonadotropins

Exogenous androgen

Busulfan

Ketoconazole

Flutamide

Spironolactone

Estrogen-containing embalming cream

Growth Hormone

Androgen precursors (i.e., androstenedione and DHEA)

Nitrosurea

Spironolactone

Bicalutamide

Ethanol

Delousing powder

 

 

Vincristine

Metronidazole

Finasteride

 

Digitalis

 

 

Ethanol

Etomidate

Cyproterone

 

Clomiphene

 

 

Tyrosine kinase inhibitor

 

Zanoterone

 

Marijuana*

 

 

 

 

Cimetidine

 

 

 

 

 

 

Ranitidine*

 

 

 

 

 

 

Spironolactone

 

* Weak evidence

 

Table 3. Drugs That May Cause Gynecomastia by Uncertain Mechanisms

 

Cardiac and antihypertensive medications:

1.     Calcium channel blockers (verapamil, nifedipine, diltiazem)

2.     Angiotensin-converting enzyme Inhibitors* (captopril, enalapril)

3.     Alpha-blockers*

4.     Amiodarone

5.     Methyldopa

6.     Reserpine

7.     Nitrates

Psychoactive drugs:

1.     Neuroleptics

2.     Anxiolytic agents* (e.g., diazepam)

3.     Phenytoin

4.     Tricyclic antidepressants

5.     Haloperidol

6.     Atypical antipsychotic agents

Drugs for infectious diseases:

1.     Antiretroviral therapy for HIV/AIDS (e.g., efavirenz)

2.     Isoniazid

3.     Ethionamide

4.     Griseofulvin

5.     Minocycline

Drugs of Abuse:

1.     Amphetamines

2.     Heroin

3.     Methadone

Others:

1.     Theophylline

2.     Omeprazole

3.     Auranofin

4.     Diethylpropion

5.     Domperidone

6.     Penicillamine

7.     Sulindac

8.     Heparin

9.     Methotrexate

10.  Dipeptidyl peptidase 4 inhibitors  

11.  Statin*

* Weak evidence

 

Drugs and chemicals that cause decreased testosterone levels either by causing direct testicular damage, by blocking testosterone synthesis, or by blocking androgen action can also produce gynecomastia. For instance, phenothrin, a chemical component in delousing agents, possessing anti-androgenic activity, has been attributed as the cause of an epidemic of gynecomastia among Haitian refugees in US detention centers in 1981 and 1982 (49). Chemotherapeutic drugs, such as alkylating agents and tyrosine kinase inhibitors (102), can cause Leydig cell and germ cell damage, resulting in primary hypogonadism. Flutamide, an anti-androgen used as treatment for prostate cancer, blocks androgen action in peripheral tissues. Ketoconazole, on the other hand, can inhibit steroidogenic enzymes required for testosterone synthesis. 5α-reductase inhibitors, finasteride and dutasteride, that reduce the conversion of testosterone to dihydrotestosterone may cause gynecomastia (50). They also cause an increase in the synthesis of testosterone and, subsequently estrogen through aromatization. Spironolactone causes gynecomastia (up to 10%) by several mechanisms. Like ketoconazole, it can block androgen production by inhibiting enzymes in the testosterone synthetic pathway (i.e., 17α-hydroxylase and 17-20-desmolase), but it can also block receptor-binding of testosterone and dihydrotestosterone (51). In addition to decreasing testosterone levels and biologic effects, spironolactone also displaces estradiol from SHBG, increasing free estrogen levels. Of note, the anti-androgenic property of spironolactone has been used in gender identity disorder (from male to female) and spironolactone is considered to be a cost-saving medication (52). On the other hand, eplerenone is more specific for the mineralocorticoid receptor and less associated with ant-androgenic effects such as gynecomastia (up to 0.5%) (53). Switching from spironolactone to eplerenone may reverse painful gynecomastia induced by spironolactone in patients with cirrhosis (54). Ethanol increases the estrogen to androgen ratio and induces gynecomastia by multiple mechanisms as well. Firstly, it is associated with increased SHBG, which decreases free testosterone levels. Secondly, it increases hepatic clearance of testosterone, and thirdly, it has a direct toxic effect on the testes (24). Besides the drugs stated, a multitude of others have been associated with gynecomastia by unknown mechanisms (92) (Table 3). For many of these drugs, the causal relationship with gynecomastia has not been established or the evidence is weak.

 

MALE BREAST CANCER

 

Male breast cancer is rare and comprises only 0.2% of all male cancers. The overall prevalence of invasive carcinomas was 0.11% and of in situ carcinomas was 0.18% in in a 20-year national registry study of surgically excised breast specimens with the diagnosis of gynecomastia (55).  Although male breast cancer is rare and gynecomastia is not considered a premalignant condition (101), men with gynecomastia, especially elderly, worry about breast cancer and often seek medical advice (56) and it is important to differentiate male breast cancer from gynecomastia (Table 4). Of note, men with Klinefelter syndrome have a 20- to 50-fold increased risk of breast cancer. Other risk factors include hyperestrogenic conditions like obesity, alcohol, exogenous estrogen exposure (e.g., gender reassignment), and testicular disorders. It is unclear if these are specific risks for breast cancer are linked to the stimulatory process responsible for gynecomastia (57). Old age, working in environment with high temperature, exhaust emissions, radiation to chest, and liver damage are also risk factors for male breast cancer (58). Family history should always be explored. In particular, a family history of BRCA2 positive breast cancer significantly increases the risk of male breast cancer in carriers of mutation (59).

 

Table 4. Clinical Findings of Gynecomastia and Male Breast Cancer

Clinical findings

Gynecomastia

Male breast cancer

Unilateral/ bilateral  

Mostly bilateral, can be unilateral

Unilateral

Consistency

Rubbery or firm

Firm or hard

Location

Concentric, around the nipple

More peripheral, outside the nipple

Pain

Painful if recent onset or rapid enlargement

Usually painless

Associated features such as skin dimpling, nipple retraction, bloody discharge

No

Possible

Palpable axillary or supraclavicular lymph node(s)

No

Possible

 

PATIENT EVALUATION

 

History Taking and Physical Examination

 

At presentation, all patients require a thorough history and physical exam. Particular attention should be given to medications, drugs and alcohol abuse, as well as other chemical exposures. Symptoms of underlying systemic illness, such as hyperthyroidism, liver disease, or renal failure should be sought. Furthermore, the clinician must recall neoplasm as a possible etiology and should establish the duration and timing of breast development. Chronic gynecomastia is more reassuring because it is almost never due to malignancy. Additionally, the clinician should inquire about fertility, erectile dysfunction, and libido to rule out hypogonadism.

 

In our experience, the breast examination is best performed with the patient supine and with the examiner palpating from the periphery to the areola. When firmness is noted, the glandular mass should be measured in diameter. Clinically, gynecomastia is diagnosed by finding subareolar breast tissue of 2 cm in diameter or greater. Malignancy should be suspected if an immobile, firm mass is found on physical examination. Skin dimpling, nipple retraction or discharge, and large, firm axillary or supraclavicular lymphadenopathy further support malignancy as a possible diagnosis. Tenderness may be present in patients with gynecomastia of less than 6 months’ duration, but it is unusual in patients with breast cancer (Table 4).

 

A thorough testicular exam is essential. When clinical exam suggests a testicular mass or when serum hCG is elevated, testicular ultra-sound (USG) is warranted. Bilaterally small testes imply testicular failure, while asymmetric testes or a testicular mass suggest the possibility of neoplasm. Visual field impairment may suggest pituitary disease. Physical findings of underlying systemic conditions such as thyrotoxicosis, HIV disease, liver, or kidney failure should also be assessed. As obesity is often associated with gynecomastia, body mass index should be documented (56).

 

Laboratory Evaluation

 

All patients who present with gynecomastia should have serum testosterone, estradiol, LH, and hCG measured (93) (using an assay that detects all forms of hCG) (Fig 2). Further testing should be tailored according to the history, physical examination and the results of these initial tests. An elevated beta hCG or hCG or a markedly elevated serum estradiol suggests neoplasm and a testicular ultrasound is warranted to identify a testicular tumor. However, non-testicular tumors can also secrete beta hCG or hCG and therefore further imaging such as CT thorax and abdomen is indicated if ultrasound does not show a testicular mass. A low testosterone level, with an elevated LH indicates primary hypogonadism. If the history suggests Klinefelter syndrome, then a karyotype should be performed for definitive diagnosis. Low testosterone and low LH indicate secondary hypogonadism, and hypothalamic or pituitary causes should be sought. If testosterone and LH are elevated, then the diagnosis of androgen resistance should be considered. In case of estrogen-secreting tumor, LH is usually suppressed with low or low normal testosterone concentrations and negative pituitary imaging. Estradiol concentrations are high. Liver, kidney and thyroid function should be assessed if clinically indicated. Furthermore, if examination of breast tissue suggests malignancy, a biopsy should be performed. This is of particular importance in patients with Klinefelter syndrome, who have an increased risk of breast cancer. On the other hand, if the examination finding is compatible with breast abscess, then fine needle aspiration for microscopy and culture is warranted (60). Acid-fast bacilli and tuberculosis culture can be done if there is risk factor(s) for tuberculosis.

 

Figure 2. Algorithm for investigation of gynecomastia

TREATMENT

 

Treatment of the underlying endocrinologic or systemic disease that has caused gynecomastia is appropriate when possible. Testicular tumors, such as Leydig cell, Sertoli cell, or granulosa cell tumors should be surgically removed. In addition to surgery, germ cell tumors are further managed with chemotherapy involving cisplatin, bleomycin, and either vinblastine or etoposide (25, 26). Should underlying thyrotoxicosis, renal, or hepatic failure be discovered, appropriate therapy should be initiated. Medications that cause gynecomastia should also be discontinued whenever possible based on their role in management of the underlying condition. The improvement should be apparent within a month after discontinuation of the culprit drug (61). If the gynecomastia has been present for more than six months, regression is unlikely because of the presence of less reversible fibrotic tissues (62). Of course, if a breast biopsy indicates malignancy, then mastectomy should be performed.

 

If no pathologic etiology is detected, then appropriate treatment is close observation. A careful breast exam should be done initially every 3-6 months until the gynecomastia regresses or stabilizes, after which a breast exam can be performed yearly. It is important to remember that most cases of pubertal gynecomastia may resolve spontaneouslywithin one to two years, around 20% of patients have residual gynecomastia at the age of 20 (63). An information sheet about gynecomastia is available for those patients who are interested to know more about their conditions (64).

 

Medical Treatment  

 

If the gynecomastia is severe, does not resolve, of recent onset (less than 6 months) and does not have a treatable underlying cause, some medical therapies may be attempted. There are 3 classes of medical treatment for gynecomastia: androgens (testosterone, dihydrotestosterone, danazol), anti-estrogens (clomiphene citrate, tamoxifen), and aromatase inhibitors such as letrozole and anastrozole.

 

Once gynecomastia is established, testosterone treatment of hypogonadal men with gynecomastia often fails to produce breast regression. Testosterone treatment may theoretically produce the side effect of gynecomastia by being aromatized to estradiol, but this side effect is uncommon and transient. Having said that, there is limited data to suggest its use to specifically counteract gynecomastia in hypogonadism (65). Dihydrotestosterone, a non-aromatizable androgen, has been used in patients with prolonged pubertal gynecomastia with good response rates but it is not commercially available (66). Danazol, a weak androgen that inhibits gonadotropin secretion, resulting in decreased serum testosterone levels, has been studied in a prospective placebo-controlled trial, whereby gynecomastia resolved in 23 percent of the patients, as opposed to 12 percent of the patients on placebo (67). The dose used for gynecomastia is 200 mg orally twice daily. Unfortunately, undesirable side effects including edema, acne, and cramps have limited its use (24).

 

Investigators have reported a 64 percent response rate with 100 mg/day of clomiphene citrate, a weak estrogen and moderate anti-estrogen in a cohort study (68). Lower doses of clomiphene have shown varied results, indicating that higher doses may need to be administered, if clomiphene is to be attempted. Tamoxifen, also an anti-estrogen, has been studied in 2 randomized, double-blind studies in which a statistically significant regression in breast size was achieved, although complete regression was not documented (69). One retrospective study compared tamoxifen with danazol in the treatment of gynecomastia. It was found that patients taking tamoxifen had a greater response with complete resolution in 78 percent of patients treated with tamoxifen, as compared to only a 40 percent response in the danazol-treated group, and the relapse rate was higher for the tamoxifen group (70), though the relapse was not systemically defined and patients with chronic gynecomastia were included. Another prospective cohort study found that 90% of patients taking tamoxifen had successful resolution of their symptoms (89). Although there is a chance of recurrence with cessation of therapy, tamoxifen, due to relatively lower side effect profile and high efficacy, may be a more reasonable choice when compared to the other therapies. If used, tamoxifen should be given at a dose of 10 mg twice or 20 mg daily a day for 3-6 months (24). Responders usually improve with reduced pain within 1 month. Another anti-estrogen, raloxifene, has also been used in the treatment of pubertal gynecomastia but its efficacy needs to be evaluated in randomized prospective studies (71).

 

An aromatase inhibitor, testolactone, has also been studied in an uncontrolled trial with promising effects (72). Further studies must be performed on this drug before any recommendations can be established on its usefulness in the treatment of gynecomastia. Newer aromatase inhibitors such as anastrozole and letrozole may have therapeutic potential (73, 74), but randomized, double-blind, placebo-controlled trials have not confirmed their efficacy. In a study involving patients receiving bicalutamide therapy for prostate cancer, only tamoxifen, but not anastrozole, significantly reduced the incidence of gynecomastia/breast pain when used prophylactically and therapeutically (75, 76). In another study with pubertal gynecomastia, no significant difference was demonstrated between the anastrozole and placebo groups in patients suffering from pubertal gynecomastia (77). The use of aromatase inhibitors is notorious for accelerated bone loss in women, but it is uncertain whether the extent of bone loss is similar in adult men (91). Furthermore, men taking anastrozole results in an increase in body fat and decline in sexual function (105).

 

From various case series, many patients with idiopathic gynecomastia show no significant improvement after medical treatment. The disappointing result may be related to the stage of disease at which medical treatment is initiated. It is likely that many or all of the men who failed to respond to medical therapy had chronic gynecomastia with fibrotic breast tissue that will not change with medical therapy or over time (56, 63). Medical therapy is only used for a short time (up to 6 months) in men with idiopathic and acute (tender, breast tissue present < 6 months) gynecomastia. Tamoxifen has the best evidence for effective medical therapy of acute, idiopathic gynecomastia.    

 

Surgical Treatment  

 

When medical therapy is ineffective, particularly in cases of longstanding gynecomastia, or when the gynecomastia interferes with the patient's activities of daily living, or when there is suspicion of malignancy of breast, then surgical therapy is appropriate. On the other hand, surgical treatment should be postponed in pubertal gynecomastia, after completion of puberty, to minimize the chance of recurrent gynecomastia after surgery (62). Surgery should also be deferred until the underlying cause of gynecomastia has resolved or been treated. Surgical treatment includes removal of glandular tissue coupled with liposuction, if needed, preferably with an individualized approach (78, 79). Nowadays, minimally invasive surgery is available and it may be associated with few complications and prompt recovery (80). If malignancy is suspected, histological examination is mandatory (56). Use of delicate cosmetic surgical techniques are warranted to prevent unsightly scarring.

 

PREVENTION OF GYNECOMASTIA IN MEN WITH PROSTATE CANCER

 

Because androgen deprivation is one of the commonly used treatment modalities for advanced prostate cancer, its possible role in the development of gynecomastia is of particular concern to clinicians. Up to 80% of patients receiving non-steroidal anti-androgen therapy may develop gynecomastia, usually 6-9 months after hormonal treatment. Some patients may have painful and disfiguring gynecomastia (81). Preventive options include tamoxifen, radiation therapy, or aromatase inhibitors.

 

Tamoxifen is the most effective preventive therapy for gynecomastia due to anti-androgen therapy for treatment of prostate cancer. Tamoxifen is superior to radiotherapy in preventing gynecomastia in patients receiving bicalutamide (Casodex) for prostate cancer in a randomized controlled trial (82). Tamoxifen is superior to aromatase inhibitor to prevent gynecomastia in patients with prostate cancer. For instance, Boccardo, et al. showed that 10% patients in the tamoxifen group (20 mg daily dose) developed gynecomastia, whereas 51% in the anastrozole group and 73% in the placebo group had gynecomastia over a period of 48 weeks (74). Fradet, et al. showed tamoxifen reduced the incidence of breast events (gynecomastia and/or breast pain) in patients with prostate cancer receiving bicalutamide in a dose-dependent manner (83). Likewise, it has been shown that low dose weekly tamoxifen (20 mg/week) is inferior to the usual dose daily regimen (20mg/day) in terms of the prevention and treatment of gynecomastia (84). Current data suggests tamoxifen 10-20 mg per day is the optimum dose required for prophylaxis of gynecomastia in patients with prostate cancer receiving androgen deprivation therapy (83, 84, 85). Low dose prophylactic irradiation has been reported to reduce the rate of gynecomastia but not breast pain in men receiving estrogens or anti-androgens for prostate cancer (11, 86, 87). Compared with tamoxifen, irradiation seems to be less effective for prevention and treatment of gynecomastia but it is usually well-tolerated (94).

 

Some studies suggest that the new generation of anti-androgen drugs such as abiraterone acetate, enzalutamide, apalutamide, and darolutamide might be associated with less gynecomastia (88, 95); More recently, it has been reported that 36.6% of patients receiving enzalutamide develop gynecomastia; this incidence seems to be lower than reported in patients who were treated with older anti-androgens such as bicalutamide (99).

 

REFERENCES

 

  1. Franz A, Wilson J: Williams Textbook of Endocrinology ninth edition, 877-885, 1998.
  2. Santen R: Endocrinology fourth edition vol. 3: 2335-2341, 2001.
  3. Bocchinfuso WP, Korach KS: Mammary Gland Development and Tumorigenesis in Estrogen Receptor Knockout Mice. Journal of Mammary Gland Biology and Neoplasia 90: 323-334, 1997.
  4. Lubahn, DB, Moyer JS, Golding TS: Alteration of Reproductive Function but not Prenatal Sexual Development after Insertional Disruption of the Mouse Estrogen Receptor Gene. Proc Soc Natl Acad Sci USA 90:11162-11166, 1993
  5. Edman DC, Hemsell DL, Brenner PF: Extraglandular Estrogen Formation in Subjects with Cirrhosis. Gastroenterology 69: 819, 1975.
  6. Kleinberg DL, Feldman M, Ruan W: IGF-1: An Essential Factor in Terminal End Bud Formation and Ductal Morphogenesis. Journal of Mammary Gland Biology and Neoplasia 5(1):7-17, 2000.
  7. Ruan W, Kleinberg DL: Insulin-like Growth Factor I is Essential for Terminal End Bud Formation and Ductal Morphogenesis during Mammary Development. Endocrinology 140(11): 5075-81, 1999.
  8. Walden PD, Ruan W, Feldman M, Kleinberg DL: Evidence that the Mammary Fat Pad Mediated the Action of Growth Hormone in Mammary Gland Development, Endocrinology 139 (2): 659-62, 1998.
  9. Mieritz MG, Sorensen K, Aksglaede L et al. Elevated serum IGF-I, but unaltered sex steroid levels, in healthy boys with pubertal gynaecomastia. Clin Endocrinol (Oxf). 2014 May;80(5):691-8 
  10. Mol JA, Van Garderen E, Rutteman GR, Rijnberk A: New Insights in the Molecular Mechanism of Progestin-induced Proliferation of Mammary Epithelium: Induction of the Local Biosynthesis of Growth Hormone in the Mammary Gland of Dogs, Cats, and Humans. Journal of Steroid Biochemistry and Molecular Biology 57 (1-2): 67-71, 1996. 
  11. Ozen H, Akyol F, Toktas G, Eskicorapci S, Unluer E, Kuyumcuoglu U, Abay E, Cureklibatur I, Sengoz M, Yalcin V, Akpinar H, Zorlu F, Sengor F, Karaman I. Is prophylactic breast radiotherapy necessary in all patients with prostate cancer and gynecomastia and/or breast pain? J Urol. 2010 Aug;184(2):519-24. 
  12. Sasano H, Kimura m, Shizawa s, Kimura N, Nagua H, Aromatase and Steroid Receptors in Gynecomastia and Male Breast Carcinoma: an Immunohistochemical Study. Journal of Clinical Endocrinology and Metabolism 81 (8): 3063-7, 1996. 
  13. Glass AR: Gynecomastia. Endocrinology and Metabolism Clinics of North America. 23(4): 825-837, 1994. 
  14. LeProvost F, Leroux, C, Martin P Gaye P, Djiane, J, Prolactin Gene Expression in Ovine and Caprine Mammary Gland, Neuroendocrinology 60: 305-313, 1994. 
  15. Steinmetz R, Grant A, Malven, P: Transcription of Prolactin Gene in Milk Secretory Cells of the Rat Mammary Gland. Journal of Endocrinology 36: 305-313,1993. 
  16. Carlson HE, Kane P, Lei ZM, et al: Presence of luteinizing hormone/human chorionic gonadotropin receptors in male breast tissues. J Clin Endocrinol Metab 89(8):4119-23, 2004.  
  17. Gill K, Kirma N, Tekmal RR: Overexpression of Aromatase in Transgenic Male Mice Results in the Induction of Gynecomastia and other Biochemical Changes in Mammary Gland. Journal of Steroid Biochemistry and Molecular Biology 77(1):13-18, 2001.  
  18. Li X, Warri A, Makela S, Ahonen T, Streng T, Santti R, Poutanen M. Mammary gland development in transgenic male mice expressing human P450 aromatase. Endocrinology 143(10):4074-83, 2002. 
  19. Moore DC, Schlaepfer, LP, Sizonenko PC: Hormonal Changes During Puberty: Transient Pubertal Gynecomastia; Abnormal Androgen-Estrogen Ratios. Journal of Clinical Endocrinology and Metabolism 58:492-499, 1984.
  20. Mahoney CP: Adolescent Gynecomastia. Differential Diagnosis and Management. Pediatric Clinics of North America 37(6): 1389-1404, 1990.
  21. Niewoehner CB, Nuttall FQ: Gynecomastia in Hospitalized Male Population. American Journal of Medicine 77: 633-638, 1984. 
  22. Braunstein GD. Aromatase and Gynecomastia. Endocrine-Related Cancer 6: 315-324, 1999. 
  23. Ikard RW, Vavra D, Forbes RC, Richman JC, Roumie CL. Management of senescent gynecomastia in the Veterans Health Administration. Breast J. 2011 Mar;17(2):160-6. doi: 10.1111/j.1524-4741.2010.01050.x.  
  24. Mathur R, Braunstein: Gynecomastia: Pathomechanisms and Treatment Strategies. Hormone Research 48:95-102, 1997.  
  25. Gana BM: Leydig Cell Tumor, British Journal of Urology 75(5): 676-8, 1995. 
  26. Richie J: Campbell's Urology 7th Edition, 2439-2443, 1998. 
  27. Hanson JA, Ambaye AB. Adult testicular granulosa cell tumor: a review of the literature for clinicopathologic predictors of malignancy.Arch Pathol Lab Med. 2011 Jan;135(1):143-6.
  28. Moran CA, Suster S: Primary Mediastinal Choriocarcinoma: A Clinicopathologic and Immunohistochemical Study of Eight Cases. American Journal of Surgical Pathology 21(9): 1007-1012, 1997.  
  29. Coen P, Kulin H, Ballantine T, Zaino r, Frauenhoffer E, Boal D, Inkster S, Brodie A, Santen R: An Aromatase-Producing Sex-cord Tumor Resulting in Prepubertal Gynecomastia. New England Journal of Medicine 324 (5): 317-22, 1991. 
  30. Hertl MC, Wiebel J, Schafer H, Willig HP, Lambrecht W. Feminizing Sertoli Cell Tumors Associated with Peutz-Jeghers Syndrome: An Increasingly Recognized Cause of Prepubertal Gynecomastia. Plastic Reconstructive Surgery 102(4):1151-57, 1998.  
  31. Young S, Gooneratne S. Straus FH 2nd, Zeller WP, Bulun SE, Rosenthal IM: Feminizing Sertoli Cell Tumors in Boys with Peutz-Jehgers Syndrome. American Journal of Surgical Pathology 19 (1):50-58, 1995.   
  32. Agarwal VR, Takayama K, Van Wyk JJ, Sasano H, Simpson ER, Bulun SE: Molecular Basis of Severe Gynecomastia Associated with Aromatase Expression in a Fibrolamellar Hepatocellular Carcinoma. Journal of Clinical Endocrinology and Metab 83(5): 1797-1800, 1988.  
  33. Muramori K, Taguchi S, Taguchi T, Kohashi K, Furuya K, Tokuda K, Ishii E. High Aromatase Activity and Overexpression of Epidermal Growth Factor Receptor in Fibrolamellar Hepatocellular Carcinoma in a Child. J Pediatr Hematol Oncol. 2011. May 5.    
  34. Berkovitz GD, Guerami, Brown TR, MacDonald PC Migeon CJ: Familial Gynecomastia with Increased Extraglandular Aromatization of Plasma Carbon 19-Steroids. Journal of Clinical Investigation 75: 1763-1769, 1985.  
  35. Shozu M, Sebastian S, Takayama K, Hsu WT, Schultz RA, Neely K, Bryant M, Bulun SE. Estrogen excess associated with novel gain-of-function mutations affecting the aromatase gene. N Engl J Med 8;348(19):1855-65, May, 2003.  
  36. Czajka-Oraniec I, Zgliczynski W, Kurylowicz A, Mikula M, Ostrowski J. Association between gynecomastia and aromatase (CYP19) polymorphisms. Eur J Endocrinol. 2008 May;158(5):721-7.
  37. Bahnsen M, Gluud C, Johnsen SG: Pituitary-testicular Function in Patients with Alcoholic Cirrhosis of the Liver. European Journal of Clinical Investigation 11: 473-479, 1981.    
  38. Olivo J, Gordon GG, Raifi F: Estrogen Metabolism in Hyperthyroidism and in Cirrhosis of the Liver. Steroids 26: 47-56, 1975. 
  39. Chan WB, Yeung VT, Chow CC, So WY, Cockram CS: Gynaecomastia as a Presenting Feature of Thyrotoxicosis. Postgraduate Medical Journal 75(882): 229-231, 1999   
  40. Herito RJ, Dankner R, Berezin M, Zeilig G, Ohry A: Gynecomastia Following Spinal Cord Disorder. Archives of Physical Medicine and Rehabilitation 78(5): 534-537, 1997. 
  41. Bowman JD, Kim H, Bustamante JJ. Drug-induced gynecomastia. Pharmacotherapy. 2012 Dec;32(12):1123-40.  
  42. Bhat N, Rosato E, Gupta P: gynecomastia in a mortician: A case report. Acta Cytol 34:31, 1990.   
  43. Finkelstein J, McCully W, MacLaughlin D, et al.: The mortician's mystery: Gynecomastia and reversible hypogonadotropic hypogonadism in an embalmer. N Eng J Med 319:961, 1988. 
  44. Voelker R.Estrogen spray poses risks to children, pets through contact with treated skin. JAMA. 2010 Sep 1;304(9):953.
  45. Nordt CA, DiVasta AD. Gynecomastia in adolescents. Curr Opin Pediatr. 2008 Aug;20(4):375-82. 
  46. Kakisaka Y, Ohara T, Tozawa H. Panax ginseng: a newly identified cause of gynecomastia. Tohoku J Exp Med. 2012;228(2):143-5.  
  47. Rifka SM, Pita JC, Vigersky RA, et al. Interaction of digitalis and spironolactone with human sex steroid receptors. J Clin Endocrinol Metab 1977; 46:228-244.  
  48. Calzada L, Torres-Calleja JM, Martinez N: Measurement of Androgen and Estrogen Receptors in Breast Tissue from Subjects with Anabolic Steroid-Dependent Gynecomastia. Life Sciences 69 (2110): 1465-1479. 
  49. Brody SA, Loriaux DL. Epidemic of gynecomastia among Haitian refugees: exposure to an environmental antiandrogen. Endocr Pract 9(5):370-5, Set-Oct, 2003.  
  50. Traish AM, Hassani J, Guay AT, Zitzmann M, Hansen ML. Adverse Side Effects of 5α-Reductase Inhibitors Therapy: Persistent Diminished Libido and Erectile Dysfunction and Depression in a Subset of Patients. J Sex Med. 2010 Dec 22. doi: 10.1111/j.1743-6109.2010.02157.x  
  51. Thompson DF, Carter J: Drug-induced gynecomastia. Pharmacotherapy 13(1): 37-45. 1993 
  52. Spack NP. Management of transgenderism. JAMA. 2013 Feb 6;309(5):478-84.  
  53. Parthasarathy HK, Ménard J, White WB, Young WF Jr, Williams GH, Williams B, Ruilope LM, McInnes GT, Connell JM, Macdonald TM. A double-blind, randomized study comparing the antihypertensive effect of eplerenone and spironolactone in patients with hypertension and evidence of primary aldosteronism. J Hypertens. 2011 May;29(5):980-990.  
  54. Dimitriadis G, Papadopoulos V, Mimidis K. Eplerenone reverses spironolactone-induced painful gynaecomastia in cirrhotics. Hepatol Int. 2011 Jun;5(2):738-9.
  55. Lapid O, Jolink F, Meijer SL. Pathological findings in gynecomastia: analysis of 5113 breasts. Ann Plast Surg. 2015 Feb;74(2):163-6.  
  56. Johnson RE, Murad MH. Gynecomastia: Pathophysiology, Evaluation, and Management Mayo Clin Proc. 2009;84(11):1010-1015.   
  57. Hsing A, McLaughlin J, Cocco p, Chen H, Fraumeni JF: Risk factors for male breast cancer. Cancer Causes and Control 9; 269-275, 1998.  
  58. Fentiman IS, Fourquet A, Hortobagyi GN: Male breast cancer. Lancet 367(9510):595-604, 2006.    
  59. Evans DG, Susnerwala I, Dawson J et al. Risk of breast cancer in male BRCA2 carriers. J Med Genet. 2010 Oct;47(10):710-1.  
  60. Koh J, Tee A. Images in clinical medicine. Tuberculous abscess manifesting as unilateral gynecomastia. N Engl J Med. 2009 Dec 3;361(23):2270.    
  61. Braunstein GD. Clinical practice. Gynecomastia. N Engl J Med. 2007 Sep 20;357(12):1229-37.  
  62. Carlson HE. Approach to the patient with gynecomastia. J Clin Endocrinol Metab. 2011 Jan;96(1):15-21.   
  63. Singh Narula H, Carlson HE. Gynaecomastia. Endocrinol Metab Clin North Am2007;48:497-519.    
  64. Hormone Foundation. Patient information page. Gynecomastia. J Clin Endocrinol Metab. 2011 Jan;96(1):0   
  65. Treves N: Gynecomastia: the origins of mammary swelling in the male: and analysis of 406 patients with breast hypertrophy, 525 with testicular tumors, and 13 with adrenal neoplasms. Cancer 11: 1083-102, 1958.    
  66. Kuhn JM, Roca R, Laudat MH, et al: Studies on the treatment of idiopathic gynecomastia with percutaneous dihydrotestosterone. Clin Endo 19: 513-20, 1983.    
  67. Jones DJ, Holt SD, Surtees P, et al: A comparison of danazol and placebo in the treatment of adult idiopathic gynaecomastia: results of a prospective study in 55 patients. Ann R Coll Surg Engl, 72:296-8, 1990.   
  68. Leroith D, Sobel R, Glick SM: The effect of clomiphene citrate on pubertal gynaecomastia. Acta Endocrinol (copenh). 95:177-80, 1980. 
  69. Alagaratnam TT: Idiopathic gynecomastia treated with tamoxifen; a preliminary report. Clin Ther 9:483-7, 1987.   
  70. Ting AC, Chow LW, Leung Yf: Comparison of tamoxifen with danazol in the management of idiopathic gynecomastia. Am Surg 66(1):38-40, 2000.
  71. Lawrence SE, Faught KA, Vethamuthu J, Lawson ML Beneficial effects of raloxifene and tamoxifen in the treatment of pubertal gynecomastia. J Pediatr;145(1):71-6,2004.  
  72. Zachmann M, Eiholzer U, Muritano M, et al: Treatment of pubertal gynaecomastia with testolactone. Acta Endocrinol supple (copenh) 279:218-26, 1986.   
  73. Miller WR, Jackson J: The therapeutic potential of aromatase inhibitors. Expert Opin Investig Drugs 12(3):337-51, Mar, 2003.     
  74. Riepe FG, Baus I, Wiest S, et al: Treatment of Pubertal Gynecomastia with the Specific Aromatase Inhibitor Anastrozole. Horm Res 20;62(3):113-118, 2004.   
  75. Boccardo F, Rubagotti A, Battaglia M, et al: Evaluation of tamoxifen and anastrozole in the prevention of gynecomastia and breast pain induced by bicalutamide monotherapy of prostate cancer. J Clin Oncol 1;23(4):808-15, 2005  
  76. Saltzstein D, Sieber P, Morris T, Gallo J: Prevention and management of bicalutamide-induced gynecomastia and breast pain: randomized endocrinologic and clinical studies with tamoxifen and anastrozole. Prostate Cancer Prostatic Dis 8(1):75-83. 2005.  
  77. Plourde PV, Reiter EO, Jou HC, Desrochers PE, Rubin SD, Bercu BB, Diamond FB Jr, Backeljauw PF: Safety and efficacy of anastrozole for the treatment of pubertal gynecomastia: a randomized, double-blind, placebo-controlled trial. J Clin Endocrinol Metab 89(9):4428-33, 2004.  
  78. Fischer S, Hirsch T, Hirche C et al. Surgical treatment of primary gynecomastia in children and adolescents. Pediatr Surg Int. 2014 Jun;30(6):641-7.  
  79. Hammond DC. Surgical correction of gynecomastia. Plast Reconstr Surg. 2009 Jul;124(1 Suppl):61e-68e.   
  80. Jarrar G, Peel A, Fahmy R, Deol H, Salih V, Mostafa A. Single incision endoscopic surgery for gynaecomastia. J Plast Reconstr Aesthet Surg. 2011 May 12.     
  81. Michalopoulos NV, Keshtgar MR. Images in clinical medicine. Gynecomastia induced by prostate-cancer treatment. N Engl J Med. 2012 Oct 11;367(15):1449.  
  82. Perdonà S, Autorino R, De Placido S, D'Armiento M, Gallo A, Damiano R, Pingitore D, Gallo L, De Sio M, Bianco AR, Di Lorenzo G. Efficacy of tamoxifen and radiotherapy for prevention and treatment of gynaecomastia and breast pain caused by bicalutamide in prostate cancer: a randomised controlled trial. Lancet Oncol. 2005 May;6(5):295-300.   
  83. Fradet Y, Egerdie B, Andersen M, Tammela TL, Nachabe M, Armstrong J, Morris T, Navani S. Tamoxifen as prophylaxis for prevention of gynaecomastia and breast pain associated with bicalutamide 150 mg monotherapy in patients with prostate cancer: a randomised, placebo-controlled, dose-response study. Eur Urol. 2007 Jul;52(1):106-14    
  84. Bedognetti D, Rubagotti A, Conti G, Francesca F, De Cobelli O, Canclini L, Gallucci M, Aragona F, Di Tonno P, Cortellini P, Martorana G, Lapini A, Boccardo F. An open, randomised, multicentre, phase 3 trial comparing the efficacy of two tamoxifen schedules in preventing gynaecomastia induced by bicalutamide monotherapy in prostate cancer patients. Eur Urol. 2010 Feb;57(2):238-45.     
  85. Serretta V, Altieri V, Morgia G et al. A randomized trial comparing tamoxifen therapy vs. tamoxifen prophylaxis in bicalutamide-induced gynecomastia. Clin Genitourin Cancer. 2012 Sep;10(3):174-9.   
  86. Tyrrell CJ, Payne H, Tammela TL, Bakke A, Lodding P, Goedhals L, Van Erps P, Boon T, Van De Beek C, Andersson SO, Morris T, Carroll K. Prophylactic breast irradiation with a single dose of electron beam radiotherapy (10 Gy) significantly reduces the incidence of bicalutamide-induced gynecomastia. Int J Radiat Oncol Biol Phys. 2004;60:476–483.   
  87. Widmark A, Fossa SD, Lundmo P, Damber JE, Vaage S,Damber L, Wiklund F, Klepp O.: Does prophylactic breast irradiation prevent antiandrogen-induced gynecomastia? Evaluation of 253 patients in the randomized Scandinavian trial SPCG-7/SFUO-3. J Urol 170(1):320, 2003.  
  88. Alesini D, Iacovelli R, Palazzo A et al. Multimodality treatment of gynecomastia in patients receiving antiandrogen therapy for prostate cancer in the era of abiraterone acetate and new antiandrogen molecules. Oncology. 2013;84(2):92-9.
  89. Mannu GS, Sudul M, Bettencourt-Silva JH, Tsoti SM, Cunnick G, Ahmed SF. Role of tamoxifen in idiopathic gynecomastia: A 10-year prospective cohort study. Breast J. 2018 Nov;24(6):1043-1045.
  90. Ali SN, Jayasena CN, Sam AH. Which patients with gynaecomastia require more detailed investigation? Clin Endocrinol (Oxf). 2018 Mar;88(3):360-363.
  91. Tan RB, Guay AT, Hellstrom WJ. Clinical use of aromatase inhibitors in adult males.Sex Med Rev. 2014 Apr;2(2):79-90.
  92. He B, Carleton B, Etminan M. Risk of Gynecomastia with Users of Proton Pump Inhibitors. 2019 Mar 13. doi: 10.1002/phar.2245
  93. Gronowski AM. Clinical assays for human chorionic gonadotropin: what should we measure and how? Clin Chem. 2009 Nov;55(11):1900-4.
  94. Di Lorenzo G, Perdonà S, De Placido S, D’Armiento M, Gallo A, Damiano R, Pingitore D, Gallo L, De Sio M, Autorino R. Gynecomastia and breast pain induced by adjuvant therapy with bicalutamide after radical prostatectomy in patients with prostate cancer: the role of tamoxifen and radiotherapy. J Urol. 2005;174:2197–2203.
  95. Ghadjar P, Aebersold DM, Albrecht C, Böhmer D, Flentje M, Ganswindt U, Höcht S, Hölscher T, Müller AC, Niehoff P, Pinkawa M, Sedlmayer F, Zips D, Wiegel T; Prostate Cancer Expert Panel of the German Society of Radiation Oncology (DEGRO) and The Working Party Radiation Oncology of the German Cancer Society (DKG-ARO). Treatment strategies to prevent and reduce gynecomastia and/or breast pain caused by antiandrogen therapy for prostate cancer: Statement from the DEGRO working group prostate cancer. Strahlenther Onkol. 2020 Jul;196(7):589-597. doi: 10.1007/s00066-020-01598-9. Epub 2020 Mar 12.
  96. Mohammadnia N, Simsek S, Stam F. Gynecomastia as a presenting symptom of Graves’ disease in a 49-year-old man. Endocrinol Diabetes Metab Case Rep. 2021 Apr 1;2021:20-0181. doi: 10.1530/EDM-20-0181.
  97. SL Simpson. Acromegaly and gigantism including a new syndrome in childhood. Postgrad Med J: first published as 10.1136/pgmj.26.294.201 on 1 April 1950.
  98. Dundar B, Dundar N, Erci T, Bober E, Büyükgebiz A. Leptin levels in boys with pubertal gynecomastia. J Pediatr Endocrinol Metab. 2005 Oct;18(10):929-34. doi: 10.1515/jpem.2005.18.10.929.
  99. Shore ND, Renzulli J, Fleshner NE, Hollowell CMP, Vourganti S, Silberstein J, Siddiqui R, Hairston J, Elsouda D, Russell D, Cooperberg MR, Tomlins SA. Enzalutamide Monotherapy vs Active Surveillance in Patients With Low-risk or Intermediate-risk Localized Prostate Cancer: The ENACT Randomized Clinical Trial. JAMA Oncol. 2022 Aug 1;8(8):1128-1136. doi: 10.1001/jamaoncol.2022.1641.
  100. Reinehr T, Kulle A, Barth A, Ackermann J, Lass N, Holterhus PM. Sex hormone profile in pubertal boys with gynecomastia and pseudogynecomastia. J Clin Endocrinol Metab. 2020 Apr 1;105(4).
  101. Kanakis GA, Nordkap L, Bang AK, Calogero AE, Bártfai G, Corona G, Forti G, Toppari J, Goulis DG, Jørgensen N. EAA clinical practice guidelines-gynecomastia evaluation and management. Andrology. 2019 Nov;7(6):778-793. doi: 10.1111/andr.12636. Epub 2019 May 16.
  102. Ghalaut VS, Prakash G, Bansal P, Dahiya K, Dokwal S, Ghalaut PS, Bala M, Dhankhar R. Effect of imatinib on male reproductive hormones in BCR-ABL positive CML patients: A preliminary report. J Oncol Pharm Pract. 2014 Aug;20(4):243-8. doi: 10.1177/1078155213500686. Epub 2013 Aug 21.
  103. Bulard J, Mowszowicz I, Schaison G. Increased aromatase activity in pubic skin fibroblasts from patients with isolated gynecomastia. J Clin Endocrinol Metab. 1987;64:618–623.
  104. Stárka L, Hill M, Pospíšilová H, Dušková M. Estradiol, obesity and hypogonadism. Physiol Res. 2020 Sep 30;69(Suppl 2):S273-S278. doi: 10.33549/physiolres.934510.
  105. Finkelstein JS, Lee H, Burnett-Bowie SA, Pallais JC, Yu EW, Borges LF, Jones BF, Barry CV, Wulczyn KE, Thomas BJ, Leder BZ. Gonadal steroids and body composition, strength, and sexual function in men. N Engl J Med. 2013 Sep 12;369(11):1011-1022.

 

Utility of Advanced Lipoprotein Testing in Clinical Practice

ABSTRACT

 

A standard lipid panel includes total cholesterol, triglycerides, and HDL-C. LDL-C can then be calculated. While the Friedewald formula is the classical method to calculate LDL-C levels recently developed formulas such as Martin Hopkins formula or Sampson-NIH formula are more accurate when triglycerides are elevated and/or LCL-C levels are low. In some instances, a direct LDL-C assay is employed, particularly when the triglyceride levels are elevated (>400mg/dL). Non-HDL-C can also be calculated (non-HDL-C = total cholesterol – HDL-C). Increasing levels of LDL-C and non-HDL-C are associated with an increased risk of atherosclerotic cardiovascular disease (ASCVD). However, numerous studies have demonstrated that the association of non-HDL-C with ASCVD is more robust. It is possible to measure apolipoprotein B and A-I levels, LDL and HDL size, LDL and HDL particle number, and Lp(a). Numerous studies have documented a link between small dense LDL particles and an increased risk of ASCVD; however, the association is markedly reduced or entirely eliminated when the analyses are adjusted for other factors that affect ASCVD risk. Similarly, there is little data demonstrating that HDL subfractions are useful in risk prediction beyond HDL and other traditional risk factors. Apolipoprotein B levels and LDL particle number are more strongly associated with ASCVD than LDL-C, particularly when the levels of LDL-C and apolipoprotein B levels or LDL particle number are discordant. Similarly, while apolipoprotein B levels or LDL particle number are significantly better than non-HDL-C in predicting ASCVD risk when the levels of non-HDL-C and apolipoprotein B levels or LDL particle number are discordant whether this will alter therapy in most patients is debated. The guidelines put forth by a variety of different expert panels and organizations do not require apolipoprotein B or LDL particle number but may use them as risk enhancing factor. It is also the author’s opinion that at this time the routine measurement of apolipoprotein B and/or LDL particle number is not required. Until data demonstrate the superiority of measuring apolipoprotein B or LDL particle number on clinical outcomes it is hard to recommend the routine use of such testing. However, in situations where there is uncertainty measurement of apolipoprotein B and/or LDL particle number can be helpful. Studies have demonstrated an association of Lp(a) with ASCVD. Many experts recommend measuring Lp(a) once in all patients while other experts recommend measuring Lp(a) more selectively in a) patients with unexplained premature CHD, b) patients with a strong family history of premature CHD, c) patients with resistance to LDL lowering with statins, d) patients with rapid unexplained progression of atherosclerosis, and e) patients with familial hypercholesterolemia. Elevations in Lp(a) will stimulate more aggressive lowering of LDL and the consideration of adding drugs that lower Lp(a) such as PCSK9 inhibitors. While routine use of advanced lipoprotein testing is not routinely recommended it should be recognized that in selected patients the additional information provided can be helpful and result in changes in treatment. As additional drugs to treat lipids are developed and our understanding of lipid and lipoprotein metabolism expands in the future the use of advanced lipoprotein analysis may assume a more important role.

 

INTRODUCTION

 

A variety of specialized lipid and lipoprotein tests are available and a question that is frequently asked is whether and when to utilize these tests in evaluating and treating patients with lipid disorders. The standard lipid panel includes the measurement of total cholesterol, triglycerides, and HDL cholesterol (HDL-C). The LDL cholesterol (LDL-C) can then be calculated using the Friedewald formula (LDL-C = total cholesterol – HDL-C – TG/5). In some instances, a direct LDL-C assay is employed because as the triglyceride levels increase the accuracy of the calculated LDL-C decreases and once the triglyceride levels are greater than 400mg/dl most laboratories will no longer provide a calculated LDL-C level. In patients with normal triglyceride levels and LDL levels > 100mg/dl calculated LDL-C and directly measured LDL-C are very strongly correlated and the difference between the levels is relatively small (1-3). However, if the triglyceride levels are greater than 150-200mg/dl the calculated LDL-C will be lower than the directly measured LDL-C level (1). Additionally, if the LDL-C level is low (<100mg/dl) the calculated LDL-C also tends to underestimate the true LDL-C level (1-5). Because of the inaccuracies of LDL-C levels calculated by the Friedewald formula a new and more accurate formula (Martin Hopkins Formula) has been developed (6). Several studies have demonstrated the increased accuracy of this new formula compared to the Friedewald formula with a particular advantage in settings of low LDL-C and high triglycerides (7-12). Major laboratories such as Quest now calculate LDL-C levels using the Martin Hopkins formula. A disadvantage of the Martin Hopkins formula is that it is more complex than the Friedewald formula and the LDL-C cannot be simply calculated. However, there is free, online access that allows for the automated calculation of LDL cholesterol by the Martin Hopkins formula (www.LDL-Calculator.com/) and a smart phone application (LDL cholesterol calculator: https://www.hopkinsmedicine.org/apps/all-apps/ldl-cholesterol-calculator). In addition to the Martin Hopkins formula other formulas to more accurately calculate LDL-C levels have been developed. The Sampson-NIH equation in some studies was more accurate when the triglyceride levels were elevated than the Martin Hopkins formula (13,14) but in other studies the Martin Hopkins formula was more accurate (15). The key is that there are better methods to calculate LDL-C levels than the Friedewald formula when triglyceride levels are elevated or LDL-C levels are low.

Non-HDL cholesterol (non-HDL-C) levels can also be calculated from a routine lipid panel (non-HDL-C = total cholesterol – HDL-C). “Remnant cholesterol” can also be estimated from the routine lipid panel (Remnant cholesterol = total cholesterol – LDL-C (direct measurement) – HDL-C) (16,17). High calculated remnant cholesterol levels are associated with an increased risk of ASCVD (16). Whether remnant cholesterol levels provide information on ASCVD risk above that provided by non-HDL-C and triglyceride levels is not clear. It should be noted that there is no accepted standard for defining remnant lipoproteins or the methods used to accurately measure remnant particles (18). Of note most guidelines and risk calculators do not require lipid and lipoprotein measurements beyond a routine lipid panel. For example, the ACC/AHA (Pooled Cohort Equations), QRISK, Reynolds, SCORE, and Framingham calculators utilize total cholesterol and HDL-C levels in order to calculate the risk of atherosclerotic cardiovascular disease (ASCVD) (19-23).

 

In the past fasting lipid panels were exclusively recommended but recent guidelines recommend either fasting or non-fasting lipid panels ((24-26). Non-fasting lipid panels will increase the convenience of obtaining lipid studies. Additionally, in patients with diabetes, fasting for the lipid panel increases the risk of hypoglycemia (27). Moreover, studies have shown that the ability of fasting and non-fasting lipid panels to predict ASCVD is similar (28-32). Fasting and non-fasting total cholesterol, HDL-C, and non-HDL-C levels are virtually identical (33,34). Triglyceride levels may increase in the fed state depending upon the amount of fat consumed and the time after consumption and therefore depending upon the circumstances there may be a considerable difference between fasting and non-fasting triglyceride levels in some patients (33,34). LDL-C levels calculated by the Friedewald formula are often decreased in the fed state due to increases in triglyceride levels (33). In the non-fasting state when LDL-C levels were calculated using the Friedewald formula 30% of patients had a ≥ 10 mg/dL difference compared to direct LDL-C measurements (9). In contrast, when LDL-C was calculated using the Hopkins Martin or Sampson-NIH formula the results were very similar to direct measurements. Therefore, if one is using non-fasting LDL-C in decision making one should calculate the LDL-C level using the Hopkins Martin or Sampson-NIH formula to increase accuracy. It should be noted that in patients where a genetic disorder of lipid metabolism is suspected or with previously elevated triglyceride levels a fasting lipid panel is preferred. Similarly, if triglyceride levels are elevated (>175mg/dL) with a non-fasting lipid panel the lipid panel should be repeated while fasting.

 

LDL CHOLESTEROL VS. NON-HDL CHOLESTEROL

 

LDL-C and non-HDL-C levels are strongly correlated and increasing levels of either parameter is associated with an increased risk of ASCVD. Numerous studies have compared the ability of LDL-C and non-HDL-C to predict ASCVD events (35). In general, while both LDL-C and non-HDL-C predict an increased risk, non-HDL-C levels are a better predictor (35-42). For example, in the Women’s Health Study, a prospective cohort study of 15,632 initially healthy US women aged 45 years or older, the relative risk of a cardiovascular event in the top vs. bottom quintile was 1.62 for LDL-C and 2.51 for non-HDL-C (41). Similarly, in the Health Professionals Follow-up Study, a study of 51,529 US male health professionals between 40 to 75 years of age, the relative risk of a cardiovascular event in the highest quintile compared with the lowest quintile was 1.81 for LDL-C and 2.76 for non-HDL-C (42).

 

While LDL-C and non-HDL-C are strongly correlated there are some individuals where these measurements are discordant (i.e., a relatively low LDL-C and a relatively high non-HDL-C or conversely a relatively high LDL-C and a relatively low non-HDL-C). In discordant situations the non-HDL-C levels are a much better predictor of cardiovascular events than the LDL levels. For example, in a study by Mora of 27,533 healthy women, 11.6% had discordant levels with discordance defined as an LDL-C above the median and a non-HDL-C below the median or an LDL-C below the median with a non-HDL-C above the median (43). Most significantly, in women with a below-median LDL-C but a non-HDL-C above the median coronary risk was underestimated by almost 3-fold for women when the LDL-C was used to predict events (43). Conversely, in women with above-median LDL-C but a non-HDL-C below the median coronary risk was overestimated by almost 3-fold when their LDL-C was used to predict events (43). Thus, the risk of ASCVD tracks more closely with non-HDL-C levels and these results highlight the advantage of non-HDL-C measurements compared to LDL-C measurements in determining risk of ASCVD.

 

In addition, this discordance between calculated LDL-C (measured by the Friedewald formula) and non-HDL-C levels can result in the misclassification of patients. For example. in patients with LDL-C levels <70 mg/dl, 15% had a non-HDL-C level ≥ 100 mg/dl and if the triglyceride levels were between 150-199mg/dl 22% had a non-HDL-C ≥ 100 mg/dl (44). Thus, a significant number of patients who have reached their LDL-C goal of < 70mg/dl have not reached their non-HDL-C goal. The method used to determine LDL-C levels influences the rate of discordance between LDL-C and non-HDL-C levels. When the LDL-C levels were measured by the Friedewald formula the discordance was considerable higher than when LDL-C levels were measured using the Hopkins Martin formula (Table 1) (45).

 

Table 1. Discordance Between LDL-C and Non-HDL-C Levels

 

Percent with Non-HDL-C > 100mg/dl

LDL-C < 70mg/dL Friedewald Formula

14-15%

LDL-C < 70mg/dL Hopkins Martin Formula

~2%

 

Percent with Non-HDL-C > 130mg/dl

LDL-C < 100mg/dl Friedewald Formula

8-10%

LDL-C < 100mg/dl Hopkins Martin Formula

~ 1%

 

Finally, studies have examined the relative utility of LDL-C and non-HDL-C levels in determining the benefits of statin therapy. A meta-analysis by Boekholdt and colleagues looked at 8 statin trials with 62,154 patients (46). They found that while on treatment levels of both LDL-C and non-HDL-C were associated with the risk of future cardiovascular events the association was more robust for non-HDL-C (46).

 

Taken together these data indicate that while both LDL-C and non-HDL-C levels are predictive of ASCVD events non-HDL-C is a better predictor. The older NCEP guidelines recommended non-HDL-C as a therapeutic target if the triglyceride levels were greater than 200mg/dl and the newer National Lipid Association and American Association of Clinical Endocrinologists recommendations consider non-HDL-C as a target along with LDL-C (19,26,47). The non-HDL-C targets are 30mg/dl higher than the LDL-C targets (for example if the LDL-C target is 70mg/dl the non-HDL-C target would be 100mg/dl). It is the opinion of this author that clinicians should utilize non-HDL-C levels more frequently in the evaluation and management of patients with hyperlipidemia. Additionally, non-HDL-C levels are easily calculated when one obtains a routine lipid panel in the fed or fasted state.

 

ADVANCED LIPOPROTEIN TESTS

 

In addition to a routine lipid panel, it is possible for the clinician to measure a number of other parameters including apolipoprotein B and A-I levels, LDL and HDL size, LDL and HDL particle number, and lipoprotein (a) (Lp(a)) levels. A number of different tests are offered by large commercial laboratories. Currently, lipoprotein analysis by Nuclear Magnetic Resonance Spectroscopy (NMR) is offered by LabCorp and Ion-Mobility Analysis is offered by Quest Diagnostics. Density Gradient Ultracentrifugation (VAP) by Atherotec was discontinued (Feb 2016). Both, LabCorp and Quest provide routine lipid panel measurements plus LDL particle number, apolipoprotein B levels, indication of LDL and HDL size, and Lp(a) measurements.

 

It should be recognized that the standardization of certain of these assays is not as rigorous as the standardization of routine lipid panel assays (3). The Centers for Disease Control and Prevention (CDC) maintains a Lipid Standardization Program (LSP) that provides standards for measuring total cholesterol, triglycerides, HDL-C, apolipoprotein A-I, and apolipoprotein B. Measurements of LDL and HDL size and particle number are not as standardized and studies have shown differences in results between different methods (3,48,49). For example, Witte and colleagues compared LDL size using NMR and gradient gel electrophoresis and observed a correlation of only 0.39 between the two methods with an average difference in LDL size of 5.38nm with NMR values being lower (50). When these investigators classified patients according to whether they had small dense LDL (Pattern B) less than 50% of patients classified as pattern B using gradient gel electrophoresis were classified as pattern B using NMR (50). Similarly, Ensign et al., compared VAP, NMR, tube gel electrophoresis, and gradient gel electrophoresis to determine LDL subclasses and found a strong disagreement in patient LDL phenotyping among these four different methods (51). Measurement of LDL and HDL particle number has also shown discrepant results between different methods (52,53). These and other results highlight the lack of rigorous standardization (54).

 

LDL SIZE

 

The size of LDL particles is heterogeneous and there are a number of different methods to determine LDL size (ultracentrifugation, gradient gel electrophoresis, ion mobility, NMR) (55). As noted above, the different methods of LDL subclass analysis may produce different results and significant variations are possible even within one method (48). Studies have shown that small dense LDL is more pro-atherogenic than large LDL particles. Small dense LDL are thought to be more atherogenic because they are better able to penetrate the endothelial cell barrier and enter the intima, are more susceptible to oxidation, bind to proteoglycans in the arterial wall, and have a longer half time in the circulation than large LDL particles (56). It should be noted though that large LDL particles are also pro-atherogenic (57-61). For example, patients with familial hypercholesterolemia tend to have large LDL particles and these patients are at high risk to develop ASCVD (60). Small LDL particles are typically seen in patients with elevated triglyceride levels and decreased HDL-C levels (i.e. patients with the metabolic syndrome, obese patients, patients with diabetes) (62). Numerous studies have documented a link between small dense LDL particles and an increased risk of ASCVD (63,64). However, the association of small dense LDL with ASCVD is markedly reduced or entirely eliminated when the analyses are adjusted for other factors that affect the risk of ASCVD (63,64). The National Lipid Association expert panel was unable to identify any patient subgroups in which measuring LDL size is necessary (65). The author concurs with that viewpoint.

 

HDL SIZE

 

HDL particles are heterogeneous and vary in size (66,67). The metabolism and function of the spectrum of HDL particles is poorly understood. Additionally, there are a number of different methods of measuring HDL size and the comparability of the various methods is uncertain (54,66,67). Finally, and most importantly there is little data demonstrating that measurements of HDL subfractions are useful in risk prediction beyond measuring HDL and other traditional risk factors (64,67,68). Because of these issues the National Lipid Association Expert Panel was unable to find situations where HDL subfraction measurements would be recommended (65).

 

It should be recognized that the crucial issue with HDL may not be the HDL levels per se but rather the function of the HDL particles (54). Assays have been developed to determine the ability of HDL to facilitate cholesterol efflux from macrophages and these studies have shown that the levels of HDL-C do not necessarily indicate the ability to mediate cholesterol efflux (69). Moreover, cholesterol efflux from macrophages had a strong inverse association with both carotid intima-media thickness and the likelihood of angiographic coronary artery disease, independently of the HDL-C level (70). Additionally cholesterol efflux was also inversely associated with the incidence of cardiovascular events (71,72). These results indicate that it is the functional capability of HDL to facilitate cholesterol efflux that is important rather than simply HDL-C levels (73).

 

Assays have also been developed to measure the ability of HDL to protect LDL from oxidation (74). The ability of HDL to protect LDL from oxidation is decreased in patients with cardiovascular disease and in patients with inflammatory disorders who are at increased risk of developing cardiovascular disease (74,75). Similar to studies of cholesterol efflux these observations suggest that HDL function is a key variable. Unfortunately assays to measure cholesterol efflux or the ability of HDL to prevent oxidation are not available outside of research laboratories.

 

APOLIPOPROTEIN B

 

All of the pro-atherogenic lipoproteins (chylomicron remnants, VLDL remnants, IDL, LDL, and Lp(a)) carry one apolipoprotein B on their surface such that apolipoprotein B levels reflect the total number of atherogenic particles (76). Most of the circulating apolipoprotein B is associated with LDL particles (76). However, the contribution of very high Lp(a) levels to total Apo B levels can be substantial (Estimated Apo B in LDL/VLDL = Apo B mg/dl – (Lp(a) mg/dl x 0.16) (77). Apo B levels measured in the non-fasting state are similar to fasting values.

 

The levels of apolipoprotein B, LDL-C, and non-HDL-C are strongly correlated. Almost all studies have shown that apolipoprotein B levels are more closely associated with ASCVD than LDL-C levels and the general consensus is that apolipoprotein B levels are a more accurate predictor of ASCVD events than LDL-C (41,42,65,78-85). Apolipoprotein B levels are equivalent to non-HDL-C levels in predicting ASCVD but when these measurements are discordant apolipoprotein B levels are a more accurate predictor of ASCVD.  

 

There are two large meta-analyses that have compared the ability of non-HDL-C and apolipoprotein B to predict ASCVD. The Emerging Risks Factor Collaboration examined 22 long term perspective studies with 91,307 subjects with a large number of events (4499) (28). In this study there were no differences in the ability of non-HDL-C or apolipoprotein B to predict ASCVD. The hazard ratio was increased approximately 2-fold in the upper quantile of non-HDL-C and apolipoprotein B compared to the lowest quantile. In contrast, another meta-analysis of 12 studies (not all perspective) with 233,455 subjects and 22,950 events reported slightly different results (86). In this study the relative risk ratio for apolipoprotein B was 1.43 (1.35-1.51) vs. 1.34 (1.24-1.44) for non-HDL-C, indicating a slightly greater predictive ability of apolipoprotein B (86).

 

A recent very large study has compared the predictive ability of non-HDL-C and apolipoprotein B (87). In the UK Biobank study 346,686 individuals without baseline CVD and not taking statins were followed for a median of 8.9 years. Fatal or nonfatal CVD events occurred in 6216 participants (1656 fatal). The conclusion of this very large study was that measurement of non-HDL-C was sufficient to capture the lipid-associated risk in CVD prediction, with no meaningful improvement from addition of apolipoprotein B.

 

Studies have also examined the predictive ability of non-HDL cholesterol and apolipoprotein B during treatment of dyslipidemia. In the Heart Protection Study (placebo vs. simvastatin) with over 20,000 participants and over 5,000 events the ability of non-HDL-C and apolipoprotein B to predict cardiovascular events were virtually identical (88). A meta-analysis by Boekholdt and colleagues looked at 8 statin trials with 62,154 patients and the adjusted hazard ratios for major cardiovascular events per 1-SD increase were very similar for apolipoprotein B and non-HDL-C (46). A meta-analysis by Robinson et al of 25 trials (n = 131,134): 12 on statin, 4 on fibrate, 5 on niacin, 2 on simvastatin-ezetimibe, 1 on ileal bypass surgery, and 1 on aggressive versus standard low-density lipoprotein (LDL) cholesterol and blood pressure targets observed that decreases in non-HDL cholesterol levels modestly outperformed apolipoprotein B in predicting cardiovascular events (89). Additionally, apolipoprotein B and non-HDL-C decreases similarly predicted cardiovascular disease risk in the statin trials.

 

While apolipoprotein B and non-HDL-C are strongly correlated there are some individuals where these measurements are discordant (i.e., a relatively low apolipoprotein B and a relatively high non-HDL-C or conversely a relatively high apolipoprotein B and a relatively low non-HDL-C). An analysis of the Interheart study explored the effect of discordance of apolipoprotein B and non-HDL-C (90). The Interheart study is a case-control study of acute myocardial infarction with blood samples in 9345 cases and 12,120 controls from 52 countries. Concentrations of non-HDL-C and apolipoprotein B were expressed as percentiles within the population. Concordance was defined as percentile non-HDL-C = percentile apolipoprotein B. Discordance was defined as percentile non-HDL-C > percentile apolipoprotein B or percentile non-HDL-C < percentile apolipoprotein B by 5%. The results of this study demonstrated that when apolipoprotein B and non-HDL-C levels were discordant the apolipoprotein B measurement was a significantly better predictor of ASCVD (90). Subjects with a low apolipoprotein B and a high non-HDL-C were at low risk (Odds Ratio 0.72 (0.67-0.77 95% CI) whereas subjects with a high apolipoprotein B and a low non-HDL-C were at a high risk (Odds Ratio 1.58 (1.38-1.58 95% CI). Similar results have recently been reported from the Women’s Health Study (91). Subjects with a high apolipoprotein B level and a discordant lower non-HDL cholesterol level had an increased risk (hazard ratio 1.22 CI 1.07- 139). Of note the subjects with higher apolipoprotein B levels relative to non-HDL-C had an increased prevalence of the metabolic syndrome including higher triglyceride levels and decreased HDL-C levels. Finally, the Cardia study compared the ability of apolipoprotein B and non-HDL-C levels to predict the development of coronary artery calcium, a surrogate marker of cardiovascular events (92). In this study apolipoprotein B levels were superior to non-HDL-C in predicting the development of coronary artery calcium (Table 2) (92). It is worth noting that the number of subjects that are discordant is relatively small (430 discordant/ 2794 total; 15.4% discordant).

 

Table 2. Cardia Study

Apo B/non-HDL-C (number of subjects)               

Odds Ratio (CI)

Low/low (1184)

1.00

Low/high (213)

1.30 (0.91-1.85)

High/low (217)

1.63 (1.15-2.32)

High/high (1180

2.32 (1.91-2.83)

 

A key question is whether measuring apolipoprotein B in addition to routine risk factors will significantly affect our ability to decide on whether and how to treat patients. Using data from the Framingham Heart Study it was shown that adding apolipoprotein B to non-HDL-C and standard risk factors increased the C-statistic from 0.723 to 0.730, a very small increase suggesting that routine measurements of apolipoprotein B would not be very helpful (81,93). Similarly, the Emerging Risk Factor Collaboration group and the Women’s Health Study also examined the effect of adding apolipoprotein B results on the C-statistic and found very little change (83,94). Additionally, the Emerging Risk Factor Collaboration modelled the effect of measuring apolipoprotein B levels on patient classification using the NCEP III guidelines. In 15,436 subjects with a cardiovascular risk of 10-20% over the next 10 years the addition of apolipoprotein B measurements would result in a change in classification in only 488 subjects (3.2%) (94). Most subjects would be moved to a lower risk category (334) and a very small number would be reclassified to a higher risk category (154). These results coupled with the C-statistic results noted above suggest that the routine addition of apolipoprotein measurements in primary prevention patients would likely not have a major effect in altering patient management.

 

In patients treated with statins a meta-analysis has compared the association of apolipoprotein B and non-HDL-C levels on the risk of major cardiovascular events (46). While both on-treatment decreases in apolipoprotein B and non-HDL-C levels were associated with a decrease in cardiovascular events the strength of the association was somewhat greater for non-HDL-C than apolipoprotein B (Table 3) (46). A meta-analysis of seven randomized controlled trials comprising more than 60 000 study participants has also shown that changes in LDL-C, apoB100, and non-HDL-C all predicted similar CVD risk reduction after 1-year of statin therapy (-20, -24, and -20% risk reduction, respectively) (95). Finally, in another meta-analysis of 25 trials (12 statin, 4 fibrate, 5 niacin, 2 simvastatin-ezetimibe, 1 ileal bypass, 1 intensive vs. standard statin) the authors concluded that “across all drug classes, apo B decreases did not consistently improve risk prediction over LDL cholesterol and non-HDL cholesterol decreases” (89). Thus, in patients treated for hyperlipidemia the measurement of apolipoprotein B levels also does not appear to significantly contribute to the management of these patients.

 

Table 3.  Risk of Cardiovascular Disease in Statin Treated Patients (Hazard Ratios)

Quartiles

Non-HDL-C

Apo B

1

1 (reference)

1 (reference)

2

1.12

1.05

3

1.17

1.12

4

1.42

1.33

 

Another approach to addressing the question of the importance of routinely measuring apolipoprotein levels is to determine if measuring apolipoprotein B level will alter our therapeutic approach. While most guidelines have not included apolipoprotein B goals there are guidelines that do recommend apolipoprotein B levels. For example, the National Lipid Association recommends in very high risk patients a LDL-C < 70mg/dL, a non-HDL-C < 100mg/dL, and an apolipoprotein B level < 80mg/dL (96). In an analysis by Sathiyakumar and colleagues if the LDL-C was < 70mg/dL and the non-HDL-C was < 100mg/dL (over 9000 subjects) fewer than 2% of the patients had an apolipoprotein B level > 80mg/dL (45). These results indicate that measuring apolipoprotein B levels will not identify a large number of patients that are not meeting the proposed goals.

 

In summary while measurement of apolipoprotein B levels is an excellent and likely the best predictor of ASCVD events whether it provides a substantial amount of information above and beyond what is provided by LDL-C and non-HDL-C and standard risk factors to justify routine apolipoprotein B measurement remains to be definitively determined. Whether routinely measuring apolipoprotein B levels will alter management in a sufficient number of patients to justify the extra expense of measuring apolipoprotein B needs to be rigorously studied. As noted earlier many of the patients with elevated apolipoprotein B levels relative to non-HDL-C levels are obese, diabetic, and have the metabolic syndrome and it is likely that clinicians will recognize based on non-lipid risk factors that these individuals are at high risk for ASCVD. There will of course be individual patients where measuring apolipoprotein levels will be helpful in determining treatment. For example, in patients thought to have Familial Dysbetalipoproteinemia (Type 3 disease) the non-HDL-C/apolipoprotein B ratio is a simple test for selecting patients with mixed hyperlipidemia that may have Familial Dysbetalipoproteinemia for additional studies (97). Similarly, in patients with high cholesterol levels and biliary obstruction a low apolipoprotein B level suggests the presence of lipoprotein X, an atypical lipoprotein particle containing unesterified cholesterol and phospholipids but not apolipoprotein B (3,98).

 

LDL PARTICLE NUMBER

 

The cholesterol content of LDL is not constant and can vary greatly between individuals and can change over time in a particular individual. For example, treatments that lower serum triglyceride levels can increase the size and cholesterol content of LDL (99,100). Measuring LDL particle number is an alternative way to quantitate LDL burden. While LDL-C and LDL particle number are strongly correlated there are some individuals who are discordant (relatively high LDL-C and relatively low LDL particle number or relatively low LDL-C and relatively high particle number). In patients with elevated triglycerides and/or low HDL levels the LDL-C levels are relatively low compared to LDL particle number (101,102).  Studies have shown that LDL particle number is more strongly associated with ASCVD than LDL-C, particularly when the levels of LDL-C and LDL particle number are discordant (43,83,103-106). Whether LDL particle number is a better predictor than non-HDL-C is discussed below.

 

Several studies have compared the ability of LDL particle number and non-HDL-C to predict ASCVD. In the Framingham Offspring Study there were 3,066 subjects with 431 events and LDL particle number was measured by NMR (103). In this study LDL particle number was more strongly associated with ASCVD than non-HDL-C (Hazard ratio 1.28 (CI 1.17-1.39) for LDL particle number vs. 1.21 (CI 1.10-1.33) for non HDL-C) (103). In the Women’s Health Study there were 27,673 subjects with 1015 events and LDL particle number was also measured by NMR (83). In this study the association of LDL particle number and non-HDL-C with ASCVD was very similar with the hazard ratio of 2.51 for LDL particle number and 2.52 for non-HDL-C (83). Finally, in the Multi-Ethnic Study of Atherosclerosis subjects (n = 6693) no benefit of measuring LDL particle number compared to routine lipid measurements on predicting ASCVD could be demonstrated (107).

 

While there are several studies that have examined patients discordant for apolipoprotein B levels and non-HDL-C levels (see section on apolipoprotein B) only two studies have examined discordance between LDL particle number and non-HDL-C. In the Multi-Ethnic Study of Atherosclerosis there were 6,814 men and women and LDL particle number was measured by NMR (108). The endpoint in this study was carotid intima-media thickness (CIMT) and coronary artery calcium (CAC), surrogate markers for ASCVD events. When there was discordance between LDL particle number and non-HDL-C, LDL particle number was more closely associated with CIMT and CAC but the differences were very modest (108). In the Women’s Health Study subjects with high LDL particle number measured by NMR that was discordant with non-HDL cholesterol levels were at increased risk of CHD (hazard ratio 1.13 CI 0.99-1.29) (91).

 

In patients on-treatment there is only a single study comparing LDL particle number and non-HDL-C. In the Heart Protection study 20,536 subjects were treated with simvastatin or placebo and LDL particle number was measured by NMR (88). The predictive strength of LDL particle number and non-HDL-C was very similar in both the placebo group and the statin group indicating no advantage of measuring LDL particle number (88).

 

It should also be noted that while LDL particle number and Apo B levels are highly correlated there are circumstances when they are discordant (109). High LDL particle number relative to Apo B levels was seen with insulin resistance, smaller LDL particle size, increased systemic inflammation, and low circulating LDL-C and HDL-C levels while high Apo B levels relative to LDL particle number was seen with larger LDL particle size and elevated levels of lipoprotein(a) (109).

 

In summary, while measurement of LDL particle number is an excellent predictor of ASCVD events whether it provides a substantial amount of information beyond what is provided by non-HDL-C and standard risk factors to justify routine LDL particle measurement remains to be definitively determined.

 

Lp(a) MEASUREMENT

 

Lp(a) is an LDL particle with a single apolipoprotein B with a plasminogen like protein, apoprotein (a), attached by a disulfide bond (110-112). Apoprotein (a) is genetically very heterogeneous due to variations in molecular weight (from 300-800 kDa) due to differences in the number of Kringle repeats (110-112). The plasma levels of Lp(a) vary greatly with undetectable levels in some individuals (0.1mg/dl) and very high levels in others (>200mg/dl) (113). Individuals with genetically determined small apoprotein (a) have high plasma levels of Lp(a) whereas individuals with genetically determined large apoprotein (a) have low levels (110-112). The size of the apo(a) isoforms is inherited with an individual having two distinct apo(a) isoforms derived from apo(a) genes from their mother and father (113). This results in individuals having two different size Lp(a) particles in the serum. It is estimated that up to 90% of the variation in Lp(a) levels is determined genetically with environment having minimal effects. Lp(a) levels are very stable within an individual over their lifespan. Inflammation and renal disease increase while severe liver disease decrease Lp(a) levels (75,114).

 

Approximately 20% of subjects have Lp(a) levels greater than 50mg/dL and 30% have Lp(a) greater than 30mg/dL. Ethnicity greatly affects Lp(a) levels (114). The levels of Lp(a) in Blacks are approximately 2-3-fold higher than in Caucasians, Caucasians and Chinese have similar levels, South Asians have levels between Blacks and Caucasians, and Mexicans have levels lower than Caucasians (Blacks> South Asians > Caucasians/Chinese > Mexicans) (114). Lp(a) levels do not correlate with LDL-C, non-HDL-C, apolipoprotein B, or LDL particle number.

 

Several large meta-analyses have demonstrated an association of Lp(a) levels with ASCVD. For example, a meta-analysis by the Emerging Risk Factors Collaboration looked at the individual records of 126,634 participants in 36 prospective studies with 9,336 CHD outcomes, 1,903 ischemic strokes, and 8,114 nonvascular deaths (115). They found a continuous association of Lp(a) with the risk of ASCVD that was not greatly affected by adjustment for other lipid levels or other established risk factors. In an analysis of 31 prospective studies with 9,870 events Bennet et al reported an odds ratio of 1.45 for individuals in the top third of Lp(a) compared with those in the bottom third (116). Of note adjustment for lipid levels and other established risk factors also had little effect on this association indicating that Lp(a) is an independent risk factor (116). Additionally, in patients with familial hypercholesterolemia elevated Lp(a) levels markedly increases the risk of the development of ASCVD (117). Mendelian randomization studies and basic science studies including experiments in animals that overexpress apoprotein (a) have suggested that increases in Lp(a) are not just a risk factor for atherosclerosis but causative for atherosclerosis (111,112,118-120). Finally, elevations in Lp(a) account for a significant proportion of the increased risk of ASCVD that is related to family history (121).

 

While the above studies clearly indicate that Lp(a) levels are a risk factor for the development of ASCVD the significance of Lp(a) in secondary prevention is not clear (122). Some studies have reported that Lp(a) is a risk factor in the setting of ASCVD (123-127) while other studies have failed to demonstrate a role for Lp(a) (128-131). In a meta-analysis of 11 studies with a total of 18,978 subjects the association between Lp(a) and ASCVD  was significant in studies in which the average LDL cholesterol was ≥130 mg/dl (OR: 1.46, 95% CI: 1.23 to 1.73, p < 0.001), whereas this relationship was attenuated and did not achieve statistical significance for studies with an average LDL cholesterol <130 mg/dl (OR: 1.20, 95% CI: 0.90 to 1.60, p = 0.21) (128). This observation suggests that in individuals with elevated LDL-C levels the impact of elevated Lp(a) levels will be magnified. However, in other studies Lp(a) was a risk factor even though LDL-C levels were relatively low (123,127). Recently Williet and colleagues reported a meta-analysis of patient-level data from seven randomized, placebo-controlled, statin outcomes trials that included 29,069 patients with repeat Lp(a) measurements (132). They found that elevated baseline and on-statin lipoprotein(a) showed an independent approximately linear relation with cardiovascular disease risk. Additionally, studies have shown that genetic variations at the LPA locus (apo(a) gene that effects Lp(a) levels) are associated with ASCVD events during statin therapy in patients (133). Taken together the bulk of the data suggests that elevated Lp(a) levels increase ASCVD risk even in patients with underlying cardiovascular disease.

 

The Emerging Risk Factor Collaboration modelled the effect of measuring Lp(a) levels on patient classification using the NCEP III guidelines (94). In 15,436 subjects with a cardiovascular risk of 10-20% over the next 10 years the addition of Lp(a) measurements would result in a change in classification in 1,517 subjects (9.8%). Most subjects would be moved to a lower risk category (962) and a number of subjects would be reclassified to a higher risk category (555) (94). These results coupled with the above findings suggest that the addition of Lp(a) measurements in patients might be useful in selected patients.

 

The potential benefits of measuring Lp(a) levels will become clearer when drugs are developed that specifically lower Lp(a) levels and clinical trials determining the effect of these drugs on ASCVD outcomes are completed. Without definitive data from randomized outcome trials demonstrating that specifically lowering Lp(a) levels results in a reduction in ASCVD events the advantages of measuring and treating Lp(a) will remain uncertain. Therapy to specifically lower Lp(a) is under development and hopefully in the near future will provide a clear demonstration of the benefits of monitoring and treating Lp(a) levels (134,135).

 

In the meantime, many experts would recommend measuring Lp(a) levels once in all patients (136-138) while other experts would measure Lp(a) in selected patients (Table 4) (65,139,140).  Elevations in Lp(a) will stimulate more aggressive lowering of LDL levels and the consideration of adding drugs that lower Lp(a) such as PCSK9 inhibitors (141).

 

Table 4. WHEN TO MEASURE LP(a) LEVELS

·       Patients with unexplained premature CHD

·       Patients with a strong family history of premature CHD

·       Patients with a family history of elevated Lp(a) levels (Cascade screening)

·       Patients with resistance to LDL-C lowering with statins

·       Patients with rapid unexplained progression of atherosclerosis

·       Patients with familial hypercholesterolemia

·       Patients with aortic valvular stenosis of uncertain cause

·       Patients with intermediate risk profiles?

 

Standard measurements of LDL-C (either calculated or measured) include Lp(a) cholesterol (139,142). When Lp(a) levels are very high they can make a significant contribution to LDL-C levels. Similarly, when LDL-C levels are markedly reduced with treatment the LDL-C measured may include a significant contribution from Lp(a). The contribution of Lp(a) cholesterol to calculated LDL-C is approximately mg/dL Lp(a) x 0.3 (when both are expressed in mg/dL) (139,142). For example, if the Lp(a) level is 100mg/dL one can estimate that approximately 30mg/dL of the calculated LDL level is due to Lp(a). Note that these estimates are not precise and the percent cholesterol per mg Lp(a) particle can vary from 5.8% to 57.3% (143). Assays are underdevelopment to accurately determine the cholesterol in Lp(a) to allow for more accurate determinations of LDL-C levels (143).

 

Accurate measurement of Lp(a) represents a formidable technical challenge, unequalled in the world of biochemical diagnostics (139,144). This is due to the extreme length polymorphism of apo(a), whose size can vary over five-fold. Currently Lp(a) assays are not well standardized and there can be considerable variation between commercial assays. One study of 6 different assays found a variation from reference material of −8% to +22% (145) and another study found considerable variation in Lp(a) levels between 5 different assays (146). Hopefully more accurate assays using monoclonal antibodies will become widely available (147).  

 

Measuring Lp(a) mass (in mg/dL), as it is frequently done in commercial clinical labs, will not allow for a reliable and consistent way to convert Lp(a) concentration to nmol/l. For example, 50 mg/dL of Lp(a) with 40 kringle IV type 2 repeats is actually fewer particles than 30 mg/dL of an Lp(a) with 15 kringle IV type repeats. The solution is the adoption of an isoform-independent method that equally identifies each Lp(a) particle (139). Such a method is currently approximated by the use of a spectrum of isoform-specific calibrators, and providers should, if possible, have Lp(a) measured using this method and reported as concentration in nmol/l.

 

CONCLUSIONS 

 

While advanced lipoprotein measurements can provide additional insights and information it is not clear that for the evaluation and treatment of the vast majority of our patients that these measurements are necessary. Notably, the guidelines on the evaluation and treatment of hyperlipidemia put forth by a variety of different expert panels and organizations do not require advanced lipoprotein measurements. It is also the author’s opinion that at this time the routine use of advanced lipoprotein testing in clinical practice is not required and that LDL-C and non-HDL-C levels provide sufficient information to guide evaluation and treatment for most patients. Until clinical trial data demonstrate the superiority of utilizing advanced lipoprotein testing on clinical outcomes it is hard to recommend the routine use of such testing. However, it should be recognized that in selected patients the additional information provided can be helpful and result in changes in treatment. It is hoped that as additional drugs to treat lipids are developed and our understanding of lipid and lipoprotein metabolism expands that in the future the use of advanced lipoprotein analysis will assume a more important role in the evaluation and treatment of patients to prevent ASCVD.

 

REFERENCES

 

  1. Martin SS, Blaha MJ, Elshazly MB, Brinton EA, Toth PP, McEvoy JW, Joshi PH, Kulkarni KR, Mize PD, Kwiterovich PO, Defilippis AP, Blumenthal RS, Jones SR. Friedewald-estimated versus directly measured low-density lipoprotein cholesterol and treatment implications. J Am Coll Cardiol 2013; 62:732-739
  2. Mora S, Rifai N, Buring JE, Ridker PM. Comparison of LDL cholesterol concentrations by Friedewald calculation and direct measurement in relation to cardiovascular events in 27,331 women. Clin Chem 2009; 55:888-894
  3. Wilson PWF, Jacobson TA, Martin SS, Jackson EJ, Le NA, Davidson MH, Vesper HW, Frikke-Schmidt R, Ballantyne CM, Remaley AT. Lipid measurements in the management of cardiovascular diseases: Practical recommendations a scientific statement from the national lipid association writing group. J Clin Lipidol 2021; 15:629-648
  4. Scharnagl H, Nauck M, Wieland H, Marz W. The Friedewald formula underestimates LDL cholesterol at low concentrations. Clinical chemistry and laboratory medicine 2001; 39:426-431
  5. Meeusen JW, Snozek CL, Baumann NA, Jaffe AS, Saenger AK. Reliability of Calculated Low-Density Lipoprotein Cholesterol. Am J Cardiol 2015; 116:538-540
  6. Martin SS, Blaha MJ, Elshazly MB, Toth PP, Kwiterovich PO, Blumenthal RS, Jones SR. Comparison of a novel method vs the Friedewald equation for estimating low-density lipoprotein cholesterol levels from the standard lipid profile. JAMA 2013; 310:2061-2068
  7. Quispe R, Hendrani A, Elshazly MB, Michos ED, McEvoy JW, Blaha MJ, Banach M, Kulkarni KR, Toth PP, Coresh J, Blumenthal RS, Jones SR, Martin SS. Accuracy of low-density lipoprotein cholesterol estimation at very low levels. BMC Med 2017; 15:83
  8. Whelton SP, Meeusen JW, Donato LJ, Jaffe AS, Saenger A, Sokoll LJ, Blumenthal RS, Jones SR, Martin SS. Evaluating the atherogenic burden of individuals with a Friedewald-estimated low-density lipoprotein cholesterol <70 mg/dL compared with a novel low-density lipoprotein estimation method. J Clin Lipidol 2017; 11:1065-1072
  9. Sathiyakumar V, Park J, Golozar A, Lazo M, Quispe R, Guallar E, Blumenthal RS, Jones SR, Martin SS. Fasting Versus Nonfasting and Low-Density Lipoprotein Cholesterol Accuracy. Circulation 2018; 137:10-19
  10. Martin SS, Giugliano RP, Murphy SA, Wasserman SM, Stein EA, Ceska R, Lopez-Miranda J, Georgiev B, Lorenzatti AJ, Tikkanen MJ, Sever PS, Keech AC, Pedersen TR, Sabatine MS. Comparison of Low-Density Lipoprotein Cholesterol Assessment by Martin/Hopkins Estimation, Friedewald Estimation, and Preparative Ultracentrifugation: Insights From the FOURIER Trial. JAMA Cardiol 2018; 3:749-753
  11. Lee J, Jang S, Son H. Validation of the Martin Method for Estimating Low-Density Lipoprotein Cholesterol Levels in Korean Adults: Findings from the Korea National Health and Nutrition Examination Survey, 2009-2011. PLoS One 2016; 11:e0148147
  12. Chaen H, Kinchiku S, Miyata M, Kajiya S, Uenomachi H, Yuasa T, Takasaki K, Ohishi M. Validity of a Novel Method for Estimation of Low-Density Lipoprotein Cholesterol Levels in Diabetic Patients. J Atheroscler Thromb2016; 23:1355-1364
  13. Sampson M, Ling C, Sun Q, Harb R, Ashmaig M, Warnick R, Sethi A, Fleming JK, Otvos JD, Meeusen JW, Delaney SR, Jaffe AS, Shamburek R, Amar M, Remaley AT. A New Equation for Calculation of Low-Density Lipoprotein Cholesterol in Patients With Normolipidemia and/or Hypertriglyceridemia. JAMA Cardiol 2020; 5:540-548
  14. Ginsberg HN, Rosenson RS, Hovingh GK, Letierce A, Samuel R, Poulouin Y, Cannon CP. LDL-C calculated by Friedewald, Martin-Hopkins, or NIH equation 2 versus beta-quantification: pooled alirocumab trials. J Lipid Res2022; 63:100148
  15. Sajja A, Park J, Sathiyakumar V, Varghese B, Pallazola VA, Marvel FA, Kulkarni K, Muthukumar A, Joshi PH, Gianos E, Hirsh B, Mintz G, Goldberg A, Morris PB, Sharma G, Blumenthal RS, Michos ED, Post WS, Elshazly MB, Jones SR, Martin SS. Comparison of Methods to Estimate Low-Density Lipoprotein Cholesterol in Patients With High Triglyceride Levels. JAMA Netw Open 2021; 4:e2128817
  16. Varbo A, Nordestgaard BG. Remnant lipoproteins. Curr Opin Lipidol 2017; 28:300-307
  17. Nordestgaard BG, Langlois MR, Langsted A, Chapman MJ, Aakre KM, Baum H, Boren J, Bruckert E, Catapano A, Cobbaert C, Collinson P, Descamps OS, Duff CJ, von Eckardstein A, Hammerer-Lercher A, Kamstrup PR, Kolovou G, Kronenberg F, Mora S, Pulkki K, Remaley AT, Rifai N, Ros E, Stankovic S, Stavljenic-Rukavina A, Sypniewska G, Watts GF, Wiklund O, Laitinen P, European Atherosclerosis S, the European Federation of Clinical C, Laboratory Medicine Joint Consensus I. Quantifying atherogenic lipoproteins for lipid-lowering strategies: Consensus-based recommendations from EAS and EFLM. Atherosclerosis 2020; 294:46-61
  18. Krauss RM, King SM. Remnant lipoprotein particles and cardiovascular disease risk. Best Pract Res Clin Endocrinol Metab 2022:101682
  19. Expert Panel on Detection E, Treatment of High Blood Cholesterol in A. Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA 2001; 285:2486-2497
  20. Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, Blum CB, Eckel RH, Goldberg AC, Gordon D, Levy D, Lloyd-Jones DM, McBride P, Schwartz JS, Shero ST, Smith SC, Jr., Watson K, Wilson PW, Eddleman KM, Jarrett NM, LaBresh K, Nevo L, Wnek J, Anderson JL, Halperin JL, Albert NM, Bozkurt B, Brindis RG, Curtis LH, DeMets D, Hochman JS, Kovacs RJ, Ohman EM, Pressler SJ, Sellke FW, Shen WK, Smith SC, Jr., Tomaselli GF, American College of Cardiology/American Heart Association Task Force on Practice G. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation2014; 129:S1-45
  21. Hippisley-Cox J, Coupland C, Robson J, Brindle P. Derivation, validation, and evaluation of a new QRISK model to estimate lifetime risk of cardiovascular disease: cohort study using QResearch database. BMJ 2010; 341:c6624
  22. Conroy RM, Pyorala K, Fitzgerald AP, Sans S, Menotti A, De Backer G, De Bacquer D, Ducimetiere P, Jousilahti P, Keil U, Njolstad I, Oganov RG, Thomsen T, Tunstall-Pedoe H, Tverdal A, Wedel H, Whincup P, Wilhelmsen L, Graham IM, Score Project Group. Estimation of ten-year risk of fatal cardiovascular disease in Europe: the SCORE project. Eur Heart J 2003; 24:987-1003
  23. Ridker PM, Paynter NP, Rifai N, Gaziano JM, Cook NR. C-reactive protein and parental history improve global cardiovascular risk prediction: the Reynolds Risk Score for men. Circulation 2008; 118:2243-2251, 2244p following 2251
  24. 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. Circulation 2018:CIR0000000000000625
  25. Nordestgaard BG, Langsted A, Mora S, Kolovou G, Baum H, Bruckert E, Watts GF, Sypniewska G, Wiklund O, Boren J, Chapman MJ, Cobbaert C, Descamps OS, von Eckardstein A, Kamstrup PR, Pulkki K, Kronenberg F, Remaley AT, Rifai N, Ros E, Langlois M, European Atherosclerosis Society, the European Federation of Clinical Chemistry, Laboratory Medicine Joint Consensus, Initiative. Fasting is not routinely required for determination of a lipid profile: clinical and laboratory implications including flagging at desirable concentration cut-points-a joint consensus statement from the European Atherosclerosis Society and European Federation of Clinical Chemistry and Laboratory Medicine. Eur Heart J 2016; 37:1944-1958
  26. Jacobson TA, Ito MK, Maki KC, Orringer CE, Bays HE, Jones PH, McKenney JM, Grundy SM, Gill EA, Wild RA, Wilson DP, Brown WV. National Lipid Association recommendations for patient-centered management of dyslipidemia: part 1 - executive summary. J Clin Lipidol 2014; 8:473-488
  27. Aldasouqi S, Sheikh A, Klosterman P, Kniestedt S, Schubert L, Danker R, Hershey DS. Hypoglycemia in patients with diabetes who are fasting for laboratory blood tests: the Cape Girardeau Hypoglycemia En Route Prevention Program. Postgrad Med 2013; 125:136-143
  28. Emerging Risk Factors Collaboration, Di Angelantonio E, Sarwar N, Perry P, Kaptoge S, Ray KK, Thompson A, Wood AM, Lewington S, Sattar N, Packard CJ, Collins R, Thompson SG, Danesh J. Major lipids, apolipoproteins, and risk of vascular disease. JAMA 2009; 302:1993-2000
  29. Mora S, Rifai N, Buring JE, Ridker PM. Fasting compared with nonfasting lipids and apolipoproteins for predicting incident cardiovascular events. Circulation 2008; 118:993-1001
  30. Langsted A, Freiberg JJ, Nordestgaard BG. Fasting and nonfasting lipid levels: influence of normal food intake on lipids, lipoproteins, apolipoproteins, and cardiovascular risk prediction. Circulation 2008; 118:2047-2056
  31. Doran B, Guo Y, Xu J, Weintraub H, Mora S, Maron DJ, Bangalore S. Prognostic value of fasting versus nonfasting low-density lipoprotein cholesterol levels on long-term mortality: insight from the National Health and Nutrition Examination Survey III (NHANES-III). Circulation 2014; 130:546-553
  32. Mora S, Chang CL, Moorthy MV, Sever PS. Association of Nonfasting vs Fasting Lipid Levels With Risk of Major Coronary Events in the Anglo-Scandinavian Cardiac Outcomes Trial-Lipid Lowering Arm. JAMA Intern Med 2019; 179:898-905
  33. Rahman F, Blumenthal RS, Jones SR, Martin SS, Gluckman TJ, Whelton SP. Fasting or Non-fasting Lipids for Atherosclerotic Cardiovascular Disease Risk Assessment and Treatment? Curr Atheroscler Rep 2018; 20:14
  34. Cartier LJ, Collins C, Lagace M, Douville P. Comparison of fasting and non-fasting lipid profiles in a large cohort of patients presenting at a community hospital. Clin Biochem 2018; 52:61-66
  35. Verbeek R, Hovingh GK, Boekholdt SM. Non-high-density lipoprotein cholesterol: current status as cardiovascular marker. Curr Opin Lipidol 2015; 26:502-510
  36. Arsenault BJ, Rana JS, Stroes ES, Despres JP, Shah PK, Kastelein JJ, Wareham NJ, Boekholdt SM, Khaw KT. Beyond low-density lipoprotein cholesterol: respective contributions of non-high-density lipoprotein cholesterol levels, triglycerides, and the total cholesterol/high-density lipoprotein cholesterol ratio to coronary heart disease risk in apparently healthy men and women. J Am Coll Cardiol 2009; 55:35-41
  37. Bittner V, Hardison R, Kelsey SF, Weiner BH, Jacobs AK, Sopko G, Bypass Angioplasty Revascularization I. Non-high-density lipoprotein cholesterol levels predict five-year outcome in the Bypass Angioplasty Revascularization Investigation (BARI). Circulation 2002; 106:2537-2542
  38. Cui Y, Blumenthal RS, Flaws JA, Whiteman MK, Langenberg P, Bachorik PS, Bush TL. Non-high-density lipoprotein cholesterol level as a predictor of cardiovascular disease mortality. Arch Intern Med 2001; 161:1413-1419
  39. Kastelein JJ, van der Steeg WA, Holme I, Gaffney M, Cater NB, Barter P, Deedwania P, Olsson AG, Boekholdt SM, Demicco DA, Szarek M, LaRosa JC, Pedersen TR, Grundy SM, TNT Study Group, Ideal Study Group. Lipids, apolipoproteins, and their ratios in relation to cardiovascular events with statin treatment. Circulation2008; 117:3002-3009
  40. Lu W, Resnick HE, Jablonski KA, Jones KL, Jain AK, Howard WJ, Robbins DC, Howard BV. Non-HDL cholesterol as a predictor of cardiovascular disease in type 2 diabetes: the strong heart study. Diabetes Care2003; 26:16-23
  41. Ridker PM, Rifai N, Cook NR, Bradwin G, Buring JE. Non-HDL cholesterol, apolipoproteins A-I and B100, standard lipid measures, lipid ratios, and CRP as risk factors for cardiovascular disease in women. JAMA 2005; 294:326-333
  42. Pischon T, Girman CJ, Sacks FM, Rifai N, Stampfer MJ, Rimm EB. Non-high-density lipoprotein cholesterol and apolipoprotein B in the prediction of coronary heart disease in men. Circulation 2005; 112:3375-3383
  43. Mora S, Buring JE, Ridker PM. Discordance of low-density lipoprotein (LDL) cholesterol with alternative LDL-related measures and future coronary events. Circulation 2014; 129:553-561
  44. Elshazly MB, Martin SS, Blaha MJ, Joshi PH, Toth PP, McEvoy JW, Al-Hijji MA, Kulkarni KR, Kwiterovich PO, Blumenthal RS, Jones SR. Non-high-density lipoprotein cholesterol, guideline targets, and population percentiles for secondary prevention in 1.3 million adults: the VLDL-2 study (very large database of lipids). J Am Coll Cardiol 2013; 62:1960-1965
  45. Sathiyakumar V, Park J, Quispe R, Elshazly MB, Michos ED, Banach M, Toth PP, Whelton SP, Blumenthal RS, Jones SR, Martin SS. Impact of Novel Low-Density Lipoprotein-Cholesterol Assessment on the Utility of Secondary Non-High-Density Lipoprotein-C and Apolipoprotein B Targets in Selected Worldwide Dyslipidemia Guidelines. Circulation 2018; 138:244-254
  46. Boekholdt SM, Arsenault BJ, Mora S, Pedersen TR, LaRosa JC, Nestel PJ, Simes RJ, Durrington P, Hitman GA, Welch KM, DeMicco DA, Zwinderman AH, Clearfield MB, Downs JR, Tonkin AM, Colhoun HM, Gotto AM, Jr., Ridker PM, Kastelein JJ. Association of LDL cholesterol, non-HDL cholesterol, and apolipoprotein B levels with risk of cardiovascular events among patients treated with statins: a meta-analysis. JAMA 2012; 307:1302-1309
  47. 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
  48. Chung M, Lichtenstein AH, Ip S, Lau J, Balk EM. Comparability of methods for LDL subfraction determination: A systematic review. Atherosclerosis 2009; 205:342-348
  49. Delatour V, Clouet-Foraison N, Gaie-Levrel F, Marcovina SM, Hoofnagle AN, Kuklenyik Z, Caulfield MP, Otvos JD, Krauss RM, Kulkarni KR, Contois JH, Remaley AT, Vesper HW, Cobbaert CM, Gillery P. Comparability of Lipoprotein Particle Number Concentrations Across ES-DMA, NMR, LC-MS/MS, Immunonephelometry, and VAP: In Search of a Candidate Reference Measurement Procedure for apoB and non-HDL-P Standardization. Clin Chem 2018; 64:1485-1495
  50. Witte DR, Taskinen MR, Perttunen-Nio H, Van Tol A, Livingstone S, Colhoun HM. Study of agreement between LDL size as measured by nuclear magnetic resonance and gradient gel electrophoresis. J Lipid Res 2004; 45:1069-1076
  51. Ensign W, Hill N, Heward CB. Disparate LDL phenotypic classification among 4 different methods assessing LDL particle characteristics. Clin Chem 2006; 52:1722-1727
  52. Hopkins PN, Pottala JV, Nanjee MN. A comparative study of four independent methods to measure LDL particle concentration. Atherosclerosis 2015; 243:99-106
  53. Matera R, Horvath KV, Nair H, Schaefer EJ, Asztalos BF. HDL Particle Measurement: Comparison of 5 Methods. Clin Chem 2018; 64:492-500
  54. Ramasamy I. Update on the laboratory investigation of dyslipidemias. Clin Chim Acta 2018; 479:103-125
  55. Ivanova EA, Myasoedova VA, Melnichenko AA, Grechko AV, Orekhov AN. Small Dense Low-Density Lipoprotein as Biomarker for Atherosclerotic Diseases. Oxid Med Cell Longev 2017; 2017:1273042
  56. Berneis KK, Krauss RM. Metabolic origins and clinical significance of LDL heterogeneity. J Lipid Res 2002; 43:1363-1379
  57. Campos H, Moye LA, Glasser SP, Stampfer MJ, Sacks FM. Low-density lipoprotein size, pravastatin treatment, and coronary events. JAMA 2001; 286:1468-1474
  58. Campos H, Roederer GO, Lussier-Cacan S, Davignon J, Krauss RM. Predominance of large LDL and reduced HDL2 cholesterol in normolipidemic men with coronary artery disease. Arterioscler Thromb Vasc Biol 1995; 15:1043-1048
  59. Mora S, Szklo M, Otvos JD, Greenland P, Psaty BM, Goff DC, Jr., O'Leary DH, Saad MF, Tsai MY, Sharrett AR. LDL particle subclasses, LDL particle size, and carotid atherosclerosis in the Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis 2007; 192:211-217
  60. Patsch W, Ostlund R, Kuisk I, Levy R, Schonfeld G. Characterization of lipoprotein in a kindred with familial hypercholesterolemia. J Lipid Res 1982; 23:1196-1205
  61. Rudel LL, Parks JS, Johnson FL, Babiak J. Low density lipoproteins in atherosclerosis. J Lipid Res 1986; 27:465-474
  62. Krauss RM, Siri PW. Metabolic abnormalities: triglyceride and low-density lipoprotein. Endocrinol Metab Clin North Am 2004; 33:405-415
  63. Ip S, Lichtenstein AH, Chung M, Lau J, Balk EM. Systematic review: association of low-density lipoprotein subfractions with cardiovascular outcomes. Ann Intern Med 2009; 150:474-484
  64. Krauss RM. Lipoprotein subfractions and cardiovascular disease risk. Curr Opin Lipidol 2010; 21:305-311
  65. Davidson MH, Ballantyne CM, Jacobson TA, Bittner VA, Braun LT, Brown AS, Brown WV, Cromwell WC, Goldberg RB, McKenney JM, Remaley AT, Sniderman AD, Toth PP, Tsimikas S, Ziajka PE, Maki KC, Dicklin MR. Clinical utility of inflammatory markers and advanced lipoprotein testing: advice from an expert panel of lipid specialists. J Clin Lipidol 2011; 5:338-367
  66. Hafiane A, Genest J. High density lipoproteins: Measurement techniques and potential biomarkers of cardiovascular risk. BBA Clin 2015; 3:175-188
  67. Savolainen MJ. Epidemiology: disease associations and modulators of HDL-related biomarkers. Handb Exp Pharmacol 2015; 224:259-283
  68. Superko HR, Pendyala L, Williams PT, Momary KM, King SB, 3rd, Garrett BC. High-density lipoprotein subclasses and their relationship to cardiovascular disease. Journal of clinical lipidology 2012; 6:496-523
  69. Hovingh GK, Rader DJ, Hegele RA. HDL re-examined. Curr Opin Lipidol 2015; 26:127-132
  70. Khera AV, Cuchel M, de la Llera-Moya M, Rodrigues A, Burke MF, Jafri K, French BC, Phillips JA, Mucksavage ML, Wilensky RL, Mohler ER, Rothblat GH, Rader DJ. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N Engl J Med 2011; 364:127-135
  71. Rohatgi A, Khera A, Berry JD, Givens EG, Ayers CR, Wedin KE, Neeland IJ, Yuhanna IS, Rader DR, de Lemos JA, Shaul PW. HDL cholesterol efflux capacity and incident cardiovascular events. N Engl J Med 2014; 371:2383-2393
  72. Qiu C, Zhao X, Zhou Q, Zhang Z. High-density lipoprotein cholesterol efflux capacity is inversely associated with cardiovascular risk: a systematic review and meta-analysis. Lipids Health Dis 2017; 16:212
  73. Anastasius M, Kockx M, Jessup W, Sullivan D, Rye KA, Kritharides L. Cholesterol efflux capacity: An introduction for clinicians. Am Heart J 2016; 180:54-63
  74. Navab M, Reddy ST, Van Lenten BJ, Anantharamaiah GM, Fogelman AM. The role of dysfunctional HDL in atherosclerosis. J Lipid Res 2009; 50 Suppl:S145-149
  75. 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) 2022.
  76. Feingold KR. Introduction to 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) 2021.
  77. Enkhmaa B, Anuurad E, Zhang W, Berglund L. Significant associations between lipoprotein(a) and corrected apolipoprotein B-100 levels in African-Americans. Atherosclerosis 2014; 235:223-229
  78. Benn M, Nordestgaard BG, Jensen GB, Tybjaerg-Hansen A. Improving prediction of ischemic cardiovascular disease in the general population using apolipoprotein B: the Copenhagen City Heart Study. Arterioscler Thromb Vasc Biol 2007; 27:661-670
  79. Chien KL, Hsu HC, Su TC, Chen MF, Lee YT, Hu FB. Apolipoprotein B and non-high density lipoprotein cholesterol and the risk of coronary heart disease in Chinese. J Lipid Res 2007; 48:2499-2505
  80. Holme I, Aastveit AH, Jungner I, Walldius G. Relationships between lipoprotein components and risk of myocardial infarction: age, gender and short versus longer follow-up periods in the Apolipoprotein MOrtality RISk study (AMORIS). J Intern Med 2008; 264:30-38
  81. Ingelsson E, Schaefer EJ, Contois JH, McNamara JR, Sullivan L, Keyes MJ, Pencina MJ, Schoonmaker C, Wilson PW, D'Agostino RB, Vasan RS. Clinical utility of different lipid measures for prediction of coronary heart disease in men and women. JAMA 2007; 298:776-785
  82. McQueen MJ, Hawken S, Wang X, Ounpuu S, Sniderman A, Probstfield J, Steyn K, Sanderson JE, Hasani M, Volkova E, Kazmi K, Yusuf S, Interheart Study Investigators. Lipids, lipoproteins, and apolipoproteins as risk markers of myocardial infarction in 52 countries (the INTERHEART study): a case-control study. Lancet 2008; 372:224-233
  83. Mora S, Otvos JD, Rifai N, Rosenson RS, Buring JE, Ridker PM. Lipoprotein particle profiles by nuclear magnetic resonance compared with standard lipids and apolipoproteins in predicting incident cardiovascular disease in women. Circulation 2009; 119:931-939
  84. Parish S, Peto R, Palmer A, Clarke R, Lewington S, Offer A, Whitlock G, Clark S, Youngman L, Sleight P, Collins R, International Studies of Infarct Survival Collaboration. The joint effects of apolipoprotein B, apolipoprotein A1, LDL cholesterol, and HDL cholesterol on risk: 3510 cases of acute myocardial infarction and 9805 controls. Eur Heart J 2009; 30:2137-2146
  85. Shai I, Rimm EB, Hankinson SE, Curhan G, Manson JE, Rifai N, Stampfer MJ, Ma J. Multivariate assessment of lipid parameters as predictors of coronary heart disease among postmenopausal women: potential implications for clinical guidelines. Circulation 2004; 110:2824-2830
  86. Sniderman AD, Williams K, Contois JH, Monroe HM, McQueen MJ, de Graaf J, Furberg CD. A meta-analysis of low-density lipoprotein cholesterol, non-high-density lipoprotein cholesterol, and apolipoprotein B as markers of cardiovascular risk. Circ Cardiovasc Qual Outcomes 2011; 4:337-345
  87. Welsh C, Celis-Morales CA, Brown R, Mackay DF, Lewsey J, Mark PB, Gray SR, Ferguson LD, Anderson JJ, Lyall DM, Cleland JG, Jhund PS, Gill JMR, Pell JP, Sattar N, Welsh P. Comparison of Conventional Lipoprotein Tests and Apolipoproteins in the Prediction of Cardiovascular Disease. Circulation 2019; 140:542-552
  88. Parish S, Offer A, Clarke R, Hopewell JC, Hill MR, Otvos JD, Armitage J, Collins R, Heart Protection Study Collaborative Group. Lipids and lipoproteins and risk of different vascular events in the MRC/BHF Heart Protection Study. Circulation 2012; 125:2469-2478
  89. Robinson JG, Wang S, Jacobson TA. Meta-analysis of comparison of effectiveness of lowering apolipoprotein B versus low-density lipoprotein cholesterol and nonhigh-density lipoprotein cholesterol for cardiovascular risk reduction in randomized trials. Am J Cardiol 2012; 110:1468-1476
  90. Sniderman AD, Islam S, Yusuf S, McQueen MJ. Discordance analysis of apolipoprotein B and non-high density lipoprotein cholesterol as markers of cardiovascular risk in the INTERHEART study. Atherosclerosis 2012; 225:444-449
  91. Lawler PR, Akinkuolie AO, Ridker PM, Sniderman AD, Buring JE, Glynn RJ, Chasman DI, Mora S. Discordance between Circulating Atherogenic Cholesterol Mass and Lipoprotein Particle Concentration in Relation to Future Coronary Events in Women. Clin Chem 2017; 63:870-879
  92. Wilkins JT, Li RC, Sniderman A, Chan C, Lloyd-Jones DM. Discordance Between Apolipoprotein B and LDL-Cholesterol in Young Adults Predicts Coronary Artery Calcification: The CARDIA Study. J Am Coll Cardiol 2016; 67:193-201
  93. Pencina MJ, D'Agostino RB, Zdrojewski T, Williams K, Thanassoulis G, Furberg CD, Peterson ED, Vasan RS, Sniderman AD. Apolipoprotein B improves risk assessment of future coronary heart disease in the Framingham Heart Study beyond LDL-C and non-HDL-C. Eur J Prev Cardiol 2015; 22:1321-1327
  94. Emerging Risk Factors Collaboration, Di Angelantonio E, Gao P, Pennells L, Kaptoge S, Caslake M, Thompson A, Butterworth AS, Sarwar N, Wormser D, Saleheen D, Ballantyne CM, Psaty BM, Sundstrom J, Ridker PM, Nagel D, Gillum RF, Ford I, Ducimetiere P, Kiechl S, Koenig W, Dullaart RP, Assmann G, D'Agostino RB, Sr., Dagenais GR, Cooper JA, Kromhout D, Onat A, Tipping RW, Gomez-de-la-Camara A, Rosengren A, Sutherland SE, Gallacher J, Fowkes FG, Casiglia E, Hofman A, Salomaa V, Barrett-Connor E, Clarke R, Brunner E, Jukema JW, Simons LA, Sandhu M, Wareham NJ, Khaw KT, Kauhanen J, Salonen JT, Howard WJ, Nordestgaard BG, Wood AM, Thompson SG, Boekholdt SM, Sattar N, Packard C, Gudnason V, Danesh J. Lipid-related markers and cardiovascular disease prediction. JAMA 2012; 307:2499-2506
  95. Thanassoulis G, Williams K, Ye K, Brook R, Couture P, Lawler PR, de Graaf J, Furberg CD, Sniderman A. Relations of change in plasma levels of LDL-C, non-HDL-C and apoB with risk reduction from statin therapy: a meta-analysis of randomized trials. J Am Heart Assoc 2014; 3:e000759
  96. Jacobson TA, Ito MK, Maki KC, Orringer CE, Bays HE, Jones PH, McKenney JM, Grundy SM, Gill EA, Wild RA, Wilson DP, Brown WV. National lipid association recommendations for patient-centered management of dyslipidemia: part 1--full report. J Clin Lipidol 2015; 9:129-169
  97. Boot CS, Middling E, Allen J, Neely RDG. Evaluation of the Non-HDL Cholesterol to Apolipoprotein B Ratio as a Screening Test for Dysbetalipoproteinemia. Clin Chem 2019; 65:313-320
  98. Arvind A, Osganian SA, Cohen DE, Corey KE. Lipid and Lipoprotein Metabolism in Liver Disease. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2019.
  99. Frost RJ, Otto C, Geiss HC, Schwandt P, Parhofer KG. Effects of atorvastatin versus fenofibrate on lipoprotein profiles, low-density lipoprotein subfraction distribution, and hemorheologic parameters in type 2 diabetes mellitus with mixed hyperlipoproteinemia. Am J Cardiol 2001; 87:44-48
  100. Yuan J, Tsai MY, Hunninghake DB. Changes in composition and distribution of LDL subspecies in hypertriglyceridemic and hypercholesterolemic patients during gemfibrozil therapy. Atherosclerosis 1994; 110:1-11
  101. Cromwell WC, Otvos JD. Heterogeneity of low-density lipoprotein particle number in patients with type 2 diabetes mellitus and low-density lipoprotein cholesterol <100 mg/dl. Am J Cardiol 2006; 98:1599-1602
  102. Otvos JD, Jeyarajah EJ, Cromwell WC. Measurement issues related to lipoprotein heterogeneity. Am J Cardiol2002; 90:22i-29i
  103. Cromwell WC, Otvos JD, Keyes MJ, Pencina MJ, Sullivan L, Vasan RS, Wilson PW, D'Agostino RB. LDL Particle Number and Risk of Future Cardiovascular Disease in the Framingham Offspring Study - Implications for LDL Management. J Clin Lipidol 2007; 1:583-592
  104. Otvos JD, Mora S, Shalaurova I, Greenland P, Mackey RH, Goff DC, Jr. Clinical implications of discordance between low-density lipoprotein cholesterol and particle number. J Clin Lipidol 2011; 5:105-113
  105. Blake GJ, Otvos JD, Rifai N, Ridker PM. Low-density lipoprotein particle concentration and size as determined by nuclear magnetic resonance spectroscopy as predictors of cardiovascular disease in women. Circulation2002; 106:1930-1937
  106. El Harchaoui K, van der Steeg WA, Stroes ES, Kuivenhoven JA, Otvos JD, Wareham NJ, Hutten BA, Kastelein JJ, Khaw KT, Boekholdt SM. Value of low-density lipoprotein particle number and size as predictors of coronary artery disease in apparently healthy men and women: the EPIC-Norfolk Prospective Population Study. J Am Coll Cardiol 2007; 49:547-553
  107. Manickam P, Rathod A, Panaich S, Hari P, Veeranna V, Badheka A, Jacob S, Afonso L. Comparative prognostic utility of conventional and novel lipid parameters for cardiovascular disease risk prediction: do novel lipid parameters offer an advantage? J Clin Lipidol 2011; 5:82-90
  108. Degoma EM, Davis MD, Dunbar RL, Mohler ER, 3rd, Greenland P, French B. Discordance between non-HDL-cholesterol and LDL-particle measurements: results from the Multi-Ethnic Study of Atherosclerosis. Atherosclerosis 2013; 229:517-523
  109. Varvel SA, Dayspring TD, Edmonds Y, Thiselton DL, Ghaedi L, Voros S, McConnell JP, Sasinowski M, Dall T, Warnick GR. Discordance between apolipoprotein B and low-density lipoprotein particle number is associated with insulin resistance in clinical practice. J Clin Lipidol 2015; 9:247-255
  110. Gudnason V. Lipoprotein(a): a causal independent risk factor for coronary heart disease? Curr Opin Cardiol2009; 24:490-495
  111. Koschinsky ML, Boffa MB. Lipoprotein(a): an important cardiovascular risk factor and a clinical conundrum. Endocrinol Metab Clin North Am 2014; 43:949-962
  112. Lamon-Fava S, Diffenderfer MR, Marcovina SM. Lipoprotein(a) metabolism. Curr Opin Lipidol 2014; 25:189-193
  113. Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of lipoprotein (a). J Lipid Res 2016; 57:1339-1359
  114. Enkhmaa B, Anuurad E, Berglund L. Lipoprotein (a): impact by ethnicity and environmental and medical conditions. J Lipid Res 2016; 57:1111-1125
  115. Emerging Risk Factors Collaboration, 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; 302:412-423
  116. Bennet A, Di Angelantonio E, Erqou S, Eiriksdottir G, Sigurdsson G, Woodward M, Rumley A, Lowe GD, Danesh J, Gudnason V. Lipoprotein(a) levels and risk of future coronary heart disease: large-scale prospective data. Arch Intern Med 2008; 168:598-608
  117. Alonso R, Andres E, Mata N, Fuentes-Jimenez F, Badimon L, Lopez-Miranda J, Padro T, Muniz O, Diaz-Diaz JL, Mauri M, Ordovas JM, Mata P, Safeheart Investigators. Lipoprotein(a) levels in familial hypercholesterolemia: an important predictor of cardiovascular disease independent of the type of LDL receptor mutation. J Am Coll Cardiol 2014; 63:1982-1989
  118. 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, Procardis Consortium. Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N Engl J Med 2009; 361:2518-2528
  119. Hobbs HH, Chiesa G, Gaw A, Lawn R, Maika SD, Koschinsky M, Hammer R. Apo(a) expression in transgenic mice. Ann N Y Acad Sci 1994; 714:231-236
  120. Liu AC, Lawn RM. Vascular interactions of lipoprotein (a). Curr Opin Lipidol 1994; 5:269-273
  121. Durrington PN, Ishola M, Hunt L, Arrol S, Bhatnagar D. Apolipoproteins (a), AI, and B and parental history in men with early onset ischaemic heart disease. Lancet 1988; 1:1070-1073
  122. Boffa MB, Stranges S, Klar N, Moriarty PM, Watts GF, Koschinsky ML. Lipoprotein(a) and secondary prevention of atherothrombotic events: A critical appraisal. J Clin Lipidol 2018; 12:1358-1366
  123. Albers JJ, Slee A, O'Brien KD, Robinson JG, Kashyap ML, Kwiterovich PO, Jr., Xu P, Marcovina SM. Relationship of apolipoproteins A-1 and B, and lipoprotein(a) to cardiovascular outcomes: the AIM-HIGH trial (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglyceride and Impact on Global Health Outcomes). J Am Coll Cardiol 2013; 62:1575-1579
  124. Nestel PJ, Barnes EH, Tonkin AM, Simes J, Fournier M, White HD, Colquhoun DM, Blankenberg S, Sullivan DR. Plasma lipoprotein(a) concentration predicts future coronary and cardiovascular events in patients with stable coronary heart disease. Arterioscler Thromb Vasc Biol 2013; 33:2902-2908
  125. Arsenault BJ, Barter P, DeMicco DA, Bao W, Preston GM, LaRosa JC, Grundy SM, Deedwania P, Greten H, Wenger NK, Shepherd J, Waters DD, Kastelein JJ, Treating to New Targets Investigators. Prediction of cardiovascular events in statin-treated stable coronary patients of the treating to new targets randomized controlled trial by lipid and non-lipid biomarkers. PLoS One 2014; 9:e114519
  126. Berg K, Dahlen G, Christophersen B, Cook T, Kjekshus J, Pedersen T. Lp(a) lipoprotein level predicts survival and major coronary events in the Scandinavian Simvastatin Survival Study. Clin Genet 1997; 52:254-261
  127. 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). Circulation2014; 129:635-642
  128. O'Donoghue ML, Morrow DA, Tsimikas S, Sloan S, Ren AF, Hoffman EB, Desai NR, Solomon SD, Domanski M, Arai K, Chiuve SE, Cannon CP, Sacks FM, Sabatine MS. Lipoprotein(a) for risk assessment in patients with established coronary artery disease. J Am Coll Cardiol 2014; 63:520-527
  129. Zewinger S, Kleber ME, Tragante V, McCubrey RO, Schmidt AF, Direk K, Laufs U, Werner C, Koenig W, Rothenbacher D, Mons U, Breitling LP, Brenner H, Jennings RT, Petrakis I, Triem S, Klug M, Filips A, Blankenberg S, Waldeyer C, Sinning C, Schnabel RB, Lackner KJ, Vlachopoulou E, Nygard O, Svingen GFT, Pedersen ER, Tell GS, Sinisalo J, Nieminen MS, Laaksonen R, Trompet S, Smit RAJ, Sattar N, Jukema JW, Groesdonk HV, Delgado G, Stojakovic T, Pilbrow AP, Cameron VA, Richards AM, Doughty RN, Gong Y, Cooper-DeHoff R, Johnson J, Scholz M, Beutner F, Thiery J, Smith JG, Vilmundarson RO, McPherson R, Stewart AFR, Cresci S, Lenzini PA, Spertus JA, Olivieri O, Girelli D, Martinelli NI, Leiherer A, Saely CH, Drexel H, Mundlein A, Braund PS, Nelson CP, Samani NJ, Kofink D, Hoefer IE, Pasterkamp G, Quyyumi AA, Ko YA, Hartiala JA, Allayee H, Tang WHW, Hazen SL, Eriksson N, Held C, Hagstrom E, Wallentin L, Akerblom A, Siegbahn A, Karp I, Labos C, Pilote L, Engert JC, Brophy JM, Thanassoulis G, Bogaty P, Szczeklik W, Kaczor M, Sanak M, Virani SS, Ballantyne CM, Lee VV, Boerwinkle E, Holmes MV, Horne BD, Hingorani A, Asselbergs FW, Patel RS, consortium G-C, Kramer BK, Scharnagl H, Fliser D, Marz W, Speer T. Relations between lipoprotein(a) concentrations, LPA genetic variants, and the risk of mortality in patients with established coronary heart disease: a molecular and genetic association study. Lancet Diabetes Endocrinol 2017; 5:534-543
  130. Schwartz GG, Ballantyne CM, Barter PJ, Kallend D, Leiter LA, Leitersdorf E, McMurray JJV, Nicholls SJ, Olsson AG, Shah PK, Tardif JC, Kittelson J. Association of Lipoprotein(a) With Risk of Recurrent Ischemic Events Following Acute Coronary Syndrome: Analysis of the dal-Outcomes Randomized Clinical Trial. JAMA Cardiol2018; 3:164-168
  131. Puri R, Ballantyne CM, Hoogeveen RC, Shao M, Barter P, Libby P, Chapman MJ, Erbel R, Arsenault BJ, Raichlen JS, Nissen SE, Nicholls SJ. Lipoprotein(a) and coronary atheroma progression rates during long-term high-intensity statin therapy: Insights from SATURN. Atherosclerosis 2017; 263:137-144
  132. 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; 392:1311-1320
  133. Wei WQ, Li X, Feng Q, Kubo M, Kullo IJ, Peissig PL, Karlson EW, Jarvik GP, Lee MTM, Shang N, Larson EA, Edwards T, Shaffer CM, Mosley JD, Maeda S, Horikoshi M, Ritchie M, Williams MS, Larson EB, Crosslin DR, Bland ST, Pacheco JA, Rasmussen-Torvik LJ, Cronkite D, Hripcsak G, Cox NJ, Wilke RA, Stein CM, Rotter JI, Momozawa Y, Roden DM, Krauss RM, Denny JC. LPA Variants Are Associated With Residual Cardiovascular Risk in Patients Receiving Statins. Circulation 2018; 138:1839-1849
  134. Tsimikas S, Viney NJ, Hughes SG, Singleton W, Graham MJ, Baker BF, Burkey JL, Yang Q, Marcovina SM, Geary RS, Crooke RM, Witztum JL. Antisense therapy targeting apolipoprotein(a): a randomised, double-blind, placebo-controlled phase 1 study. Lancet 2015; 386:1472-1483
  135. Tsimikas S. RNA-targeted therapeutics for lipid disorders. Curr Opin Lipidol 2018; 29:459-466
  136. Kronenberg F, Mora S, Stroes ESG. Consensus and guidelines on lipoprotein(a) - seeing the forest through the trees. Curr Opin Lipidol 2022; 33:342-352
  137. 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; 37:1129-1150
  138. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Group ESCSD. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188
  139. 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; 71:177-192
  140. 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. Don P. Wilson, MD, on behalf of the Writing group. J Clin Lipidol 2019;
  141. van Capelleveen JC, van der Valk FM, Stroes ES. Current therapies for lowering lipoprotein (a). J Lipid Res2016; 57:1612-1618
  142. Yeang C, Witztum JL, Tsimikas S. 'LDL-C' = LDL-C + Lp(a)-C: implications of achieved ultra-low LDL-C levels in the proprotein convertase subtilisin/kexin type 9 era of potent LDL-C lowering. Curr Opin Lipidol 2015; 26:169-178
  143. 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
  144. Marcovina SM, Albers JJ. Lipoprotein (a) measurements for clinical application. J Lipid Res 2016; 57:526-537
  145. 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
  146. Wyness SP, Genzen JR. Performance evaluation of five lipoprotein(a) immunoassays on the Roche cobas c501 chemistry analyzer. Pract Lab Med 2021; 25:e00218
  147. Marcovina SM, Navabi N, Allen S, Gonen A, Witztum JL, Tsimikas S. Development and validation of an isoform-independent monoclonal antibody-based ELISA for measurement of lipoprotein(a). J Lipid Res 2022; 63:100239

Reproductive Health and Its Impact On Lipid Management in Adolescent and Young Adult Females

ABSTRACT

 

Lipids disorders are common in youth.  Adolescents and young women of childbearing age who have moderate to severe lipid disorders may benefit from treatment with lipid lowering medications (LLM). However, most of these medications are contraindicated in pregnancy.  Those who are sexually active should receive counseling on effective methods to prevent unplanned pregnancies. While contraceptives, when appropriate, are typically prescribed by primary care physicians, lipidologists are often asked to address the unique aspects related to use of long-term LLMs, such as statins, in females with hypercholesterolemia.  Appropriate counseling and management require not only knowledge of the effects of sexual maturation on lipid and lipoprotein metabolism, but a thorough understanding of current recommendations and potential harms associated with the use of some LLMs, such as statins, should pregnancy occur.  In this chapter, we review changes in lipid and lipoprotein metabolism during puberty, current guidelines for use of contraceptive methods in adolescent and young adult females and laws that pertain to this unique population.

 

CANDIDATES FOR LIPID LOWERING MEDICATIONS IN YOUTH

 

There are a number of lipid disorders, both acquired and genetic, that may warrant pharmacotherapy, beginning at an early age.  These include primary hypercholesterolemia, such as familial hypercholesterolemia (FH), and the severe hypertriglyceridemia characteristic of familial chylomicronemia syndrome (FCS).  A discussion of the specific disorders causing moderate to severe dyslipidemia in adolescents and young adults can be found in Endotext (1-3).

 

LIPID AND LIPOPROTEIN METABOLISM DURING PUBERTY

 

It should be noted that lipid levels vary throughout childhood and adolescence.  Prior studies have described these changes. At young ages, lipid levels are similar between boys and girls (3-12). Cholesterol levels at birth are typically very low.  A review of studies of serum cholesterol in the U.S. concluded that total cholesterol (TC) concentration, which is approximately 65 mg/dl (1.68 mmol/l) in umbilical cord blood, rises after birth to reach a mean level of 165 mg/dl (4.27 mmol/l) by 2 years-of-age (13). When trends are analyzed by chronological age, mean TC levels generally peak between ages 9 to 11, followed by a decline during puberty.  There are gender differences in the levels of other lipids and lipoproteins as well.  A review of data from NHANES demonstrated that the prevalence of elevated levels of non-high-density lipoprotein cholesterol (non-HDL-C) was greater in girls (9.4%) than in boys (7.5%) (14). Levels rise again after puberty to reach the adult values. 

 

Since puberty has such a remarkable effect on lipid levels, it is important to understand the stages of pubertal development in assessing and making recommendations for treatment of dyslipidemia in youth.  Puberty is defined as the age at or period during which the body of a girl or boy matures and becomes capable of reproduction. Classically, there are five well-defined stages of puberty, often referred to as Tanner or pubertal stages 1 to 5 (Figure 1) (15-17). 

 

Figure 1. Pubertal Stages

Menarche, the term used to define the first menstrual cycle or episode of vaginal bleeding in females, generally occurs at pubertal stage 4; while pubertal stage 5 is used to define adult sexual maturity in both males and females.  Despite earlier onset of secondary sexual characteristics, such as pubic hair, historically the median age at menarche has remained relatively stable - occurring at approximately 12 to 13 years-of-age in most females - across well-nourished populations in developed countries.  Over the past 30 years, data from the U.S. NHANES has found no significant change in the median age at menarche, except among the non-Hispanic black population, which has a 5.5-month earlier median age at menarche than occurred previously. 

 

Excessive weight gain during childhood is associated with earlier onset of puberty.  Environmental factors, including socioeconomic conditions, nutrition, and access to preventive health care, may also influence the timing and progression of puberty.

 

By 15 years-of-age, 98% of females will have experienced menarche (18). In addition to a benchmark marking the beginning of reproductive life in females, both premature and delayed menarche appear to be associated with increased cardiovascular disease (CVD) risk (19-21).  Women with prolonged and irregular menstrual cycles also appear to have higher risk for premature CVD and type 2 diabetes (T2D) (22,23).

 

Previous studies have taken into account pubertal changes in assessing lipid and lipoprotein levels in youth (24-30). It should be noted that youth of the same age, sex, and race show considerable variability in their degree of sexual maturation and somatic growth. Thus, at all pubertal stages, chronological age can vary widely. This suggests that the use of age alone may be misleading when assessing the levels and changes in lipid levels in this population. In a longitudinal study that assessed pubertal stage at various ages, the levels of TC, low density-lipoprotein cholesterol (LDL-C), and non-HDL-C decreased in all groups during puberty (31).  HDL-C decreased and triglyceride (TG) levels increased in males during puberty, while no changes were observed in females. For HDL-C, sex differences in the pattern of change emerged by pubertal stage 3 (31).  Prior data has also suggested that HDL-C concentrations continue to decrease in males into early adulthood while remaining constant in females (32).  Pubertal development should, therefore, be considered when determining criteria for initiation of lipid screening in youth; pre-pubertal (often before 9 years-of-age) and post-pubertal (typically after 17 years-of-age) screening might be useful despite current recommendations to screen between 9 to 11 years-of-age (31).  For females who become pregnant, it is important to be aware of the additional changes in lipids that have been well-described (33).

 

EFFECT OF PREGNANCY ON LIPID DISORDERS

 

Management of dyslipidemia in adolescent and young adult females requires a thorough knowledge of lipid metabolism and physiologic changes that occur during pregnancy. Those who are considering pregnancy in the setting of a lipid disorder may benefit from preconception consultation with an obstetrician/maternal-fetal medicine specialist with knowledge of complex pregnancies to discuss the impact of pregnancy on maternal disease. If not seen prior to becoming pregnant, referral of those with severe dyslipidemia or FH is recommended early in pregnancy. Severe dyslipidemia has been associated with a variety of obstetric and maternal complications including preterm delivery, hypertension-related disorders of pregnancy, fetal growth restriction, increased fat deposition in the fetus, and maternal pancreatitis (34).

 

Physiologic changes that occur during pregnancy lead to an increase in plasma concentrations of lipids and lipoproteins. Maternal hyperlipidemia routinely occurs in the later part of pregnancy with levels of TG, very low-density lipoproteins (VLDL-C), LDL-C, and HDL-C increased compared to non-pregnant females (35). These changes are related to increased insulin resistance during pregnancy as well as increased production of estrogen (36). Given the expected increases seen in cholesterol during pregnancy and the limited treatment options that are available, routinely following lipid levels during pregnancy is not recommended. However, patients with severe hypertriglyceridemia who are at-risk of pancreatitis and those with FH who may develop symptomatic ASCVD during pregnancy are an exception and should be followed closely (37). Following delivery, cholesterol concentrations begin to decline with levels returning to baseline in most women by 6 weeks postpartum. However, some data suggest levels may remain elevated for as long as 20 weeks postpartum. Breastfeeding leads to decreases in TG levels, however HDL-C levels increase. The effect of breastfeeding on LDL-C levels is unclear (36).

 

SAFETY AND EFFICACY OF LIPID-LOWERING THERAPY WITH A RISK OF PREGNANCY

 

In addition to heart-healthy living, use of LLMs is currently recommended as a treatment option for females with moderate-to-severe dyslipidemia (39,40).  When prescribing these medications, it is important to understand the potential risks of each drug class as well as each individual drug, and effects on reproductive health.  While rare, most adverse events are similar for male and female youth (41). 

 

Recommendations for use of LLMs, such as statins, in females of childbearing age should consider the potential ofteratogenicity (33, 39). To date there have been two systematic reviews that evaluated statins and teratogenicity.  Neither found evidence that statins cause congenital anomalies independent of concomitant medical conditions associated with their use (42,43).  Of interest, in women with a prior history of pre-eclampsia use of pravastatin during subsequent pregnancy has shown promising results for preventing recurrence (44,45). Nonetheless, caution is advised when recommending the use of statins in females, including adolescent and young adult females.

 

 

If an adolescent or young adult female becomes pregnant while taking a LLM other than a bile acid sequestrant (e.g., colesevelam), best practice has been to immediately stop the LLM and the patient should be followed closely by an obstetrician in addition to a lipid specialist. LDL apheresis can be safely used during pregnancy and may be beneficial to some women.

 

It should be stated, however, that in 2021 the FDA requested revisions to the prescribing information about statin use during pregnancy, noting that contraindication of these drugs in all pregnant women is not appropriate.  The FDA recommended removing this labeling, based upon the benefits statins may have in preventing serious or potentially fatal events in a small group of very high-risk pregnant patients. Removing the contraindication enables health care professionals and patients to make individual decisions about benefit and risk, especially for those at very high risk of heart attack or stroke, such as homozygous FH and females who have previously had a heart attack or stroke (47).

 

NON-STATIN THERAPIES

 

Lifestyle changes, including dietary modifications, are recommended for all individuals with lipid disorders and should be considered a cornerstone of lipid management in pregnancy as well. There are limited data, however, on use of non-statin medications to treat elevations in cholesterol and triglyceride during pregnancy. If any of the following medications are considered in an adolescent or woman of child-bearing age, the potential for pregnancy and relative risks must be taken into consideration.

 

Bile Acid Sequestrants

 

Despite reassurance of statin safety, only bile acid sequestrants are currently considered safe for use in treating LDL-C disorders during pregnancy and breastfeeding.

 

Ezetimibe

 

No data are available on use during pregnancy. Animal studies have found ezetimibe crosses the placenta. At levels significantly higher than those achieved with human doses, there appears to be a slightly increased risk of skeletal abnormalities in rats and rabbits. Therefore, this agent is not recommended for use during pregnancy. If used prior to pregnancy, ezetimibe should be discontinued prior to attempting to become pregnant (48).

 

PCSK9 Inhibitors (Monoclonal Evolocumab, Alirocumab and mRNA Therapy Inclisiran)

 

No data are available on use during pregnancy. An observational trial of evolocumab in pregnant women with FH was terminated after being unable to enroll sufficient subjects (4 patients in 4 years; clinical trials.gov NCT02906124). PCSK9 inhibitors are not approved for use in pregnancy nor currently recommended (48).

 

Bempedoic acid

 

This drug should be discontinued when pregnancy is recognized, unless the benefits of therapy outweigh the potential risks to the fetus (49).

 

Evinacumab

 

No data are available on use during pregnancy. Based on animal studies, exposure during pregnancy may lead to fetal harm (49).

 

Lomitapide

 

This drug is not recommended during pregnancy due to concerns for fetal harm (49).

 

Fibrates

 

Limited data are available on use during pregnancy.  Adverse outcomes from the use of fibrates during the second trimester have not been reported. However, such observations are based on case reports (50). Most reported use of fibrates (both gemfibrozil and fenofibrate) during pregnancy occurred in the second trimester, after embryogenesis occurs. Studies in animals have found no increased risk of congenital malformations (48).

 

Omega-3-Fatty Acid

 

Lifestyle modifications with increase in dietary omega-3-fatty acids appear to be safe during pregnancy. Prescription omega-3-fatty acids are not approved for use during pregnancy (48).

 

Volanesorsen

 

No data are available on use during pregnancy. If used prior to pregnancy, volanesoren should be discontinued one month before attempting conception (51). This drug is not approved for use in the U.S.

 

Plasmapheresis

 

In those at-risk of severe elevations in TG due to acquired (insulin resistance/diabetes) and genetic causes (FCS and MCS), case reports and reviews have reported use of plasmapheresis in those who develop pancreatitis. The procedure appears to be safe and has the advantage of quickly lowering TG levels (52,53).

 

LDL Apheresis

 

This procedure can be safely used during pregnancy and may be beneficial to some women with severely elevated lipids, such as FH (39).

 

SEXUAL ACTIVITY, RISK OF PREGNANCY AND SEXUAL HISTORY TAKING

 

Drugs such as statins are increasingly prescribed to females of childbearing age, including prior to the onset of sexual maturity. Since current guidelines still suggest avoidance or discontinuation of most LLMs during pregnancy, it is important for clinicians to consider the appropriate age at which a conversation regarding contraceptive options should be initiated. In addition to knowledge regarding pubertal development and reproductive ability, clinicians should have an awareness of current sexual practices amongst adolescents. According to the National Center for Health Statistics Reports, an estimated 55% of U.S. male and female teens have had sexual intercourse by 18 years-of-age; approximately 80% of teens used some form of contraception during their first episode of sex (54).

 

The proportion of youth who have had sexual intercourse increases rapidly throughout adolescence. In 2013, approximately 1% of 10-year-olds, 20% of 15-year-olds and 65% of 18-year-olds reported having had sexual intercourse (Figure 2).

 

Figure 2. Percent of individuals 10-40 years-of-age who have had sexual intercourse. Modified from J. Philbin, Guttmacher Institute, unpublished data from the National Survey of Family Growth, 2013 (93).

For youth 11 years-of-age and older, the American Academy of Pediatrics (AAP) recommends healthcare providers obtain a developmentally-appropriate sexual history, including assessing risk of sexually transmitted infections (STIs) and pregnancy, and provision of appropriate screening, counseling, and, if needed, contraceptives options for adolescents during clinic visits (55).

 

An adolescent’s sexual history should be updated regularly and conducted in a confidential and non-judgmental manner, re-addressing the needs for contraception, STI screening, and appropriate counseling regarding reduction of health risks related to sexual activity (56). Pregnancy testing should be conducted when appropriate or requested.

 

Key to effective history taking is an honest, caring, non-judgmental approach by the healthcare provider. Interviews should be conducted in a comfortable, matter-of-fact manner to encourage questions and to build trust. This can be accomplished by assessing the “5 Ps” of sexual history taking:

 

  • Partners
  • Prevention of pregnancy
  • Protection from STIs
  • Sexual Practices
  • Past history of STIs and pregnancy

 

To encourage compliance, counseling should incorporate techniques of motivational interviewing (57).

 

Although there has been a decline in recent years, the pregnancy rate amongst adolescent females in the U.S. remains substantially higher than in other Western industrialized nations and racial/ethnic and geographic disparities in teen birth rates persist (58-62).

 

EFFECT OF CONTRACEPTIVE OPTIONS ON LIPOPROTEIN METABOLISM

 

While clinicians should review the effects on lipid levels when prescribing contraception, there are limited data to aid selection of a specific contraceptive method based upon the individual’s underlying presumed or confirmed lipid disorder.

 

Review of the CDC Summary Chart of US Medical Eligibility Criteria for Contraceptive Use suggests avoidance of estrogen containing birth control as well as DMPA in individuals at increased risk of cardiovascular disease, which would include those with lipid disorders. Preferred methods of contraception for this vulnerable population would include the copper IUD, which contains no hormones, followed by a levonorgestrel containing IUD, implant, and progestin only contraceptive pills.

 

Although the impact of estrogen and progestin on lipid parameters has been well described, it is not known whether the hormone formulation or the means of administration of various contraceptive methods have any clinical significance either in women with normal baseline lipid levels or in those with lipid disorders (Table 1) (63). Furthermore, insufficient data are available in regard to the effect of various contraceptive methods when used in individuals with well-defined lipid disorders.

 

Table 1. The Effects of Contraceptive Methods on Lipids and Lipoproteins

Contraceptive Method

LDL-C

HDL-C

TG

Comments/References

Combined Oral Contraceptive Pill

·       Estrogen

Decrease

Increase

Increase

For OCPs with an identical dose of estrogen, the choice and dose of the progestin component may affect net lipid changes (63,64)

·       Progestin

Increase

Decrease

Decrease

Transdermal Patch

Decrease

Increase

Increase

(65)

Vaginal Ring

---

---

Increase

(66)

DMPA

Increase

Decrease

Neutral

(67,68)

LDL-C = low-density lipoprotein cholesterol; HDL-C = high-density lipoprotein cholesterol; TG = triglycerides; DMPA = Depot medroxyprogesterone acetate

 

In general, combined oral contraceptives (COCs) raise TGs slightly.  The effects on LDL-C and HDL-C are less predictable, but the effects are thought to be related to the dose of the ethinyl estradiol, the type of progestin, and health status of the patient (for example, obese versus not obese).  Once OCPs are discontinued, lipid and lipoprotein levels appear to return to pre-treatment levels (64). If an oral contraceptive is preferred, the use of COCs that contain 35 mcg or less of estrogen is generally recommended for most adult women with controlled dyslipidemia. 

 

Compared to COCs, transdermal and vaginal contraception have similar effects on lipid profiles. Barrier methods and IUDs are generally considered to be lipid neutral (65,66).

 

COCs have also been shown to increase plasma insulin and glucose levels and reduce insulin sensitivity in women; however, these effects are negligible for current formulations and among women of normal weight without polycystic ovary syndrome (PCOS).  For females who are overweight/obese and those with PCOS, potential adverse effects should be considered in the choice of contraceptive method (67-74).

 

SAFETY AND EFFICACY OF CONTRACEPTIVE OPTIONS IN ADOLESCENT AND YOUNG ADULT FEMALES

 

How does one determine which contraceptive is the best option for an adolescent or young adult female with dyslipidemia?  From a practical point of view, this is largely dependent upon the individual’s needs, preference, resources, and ability to adhere to the method chosen. Abstinence is 100% effective in preventing pregnancy and sexually transmitted infections, and is an important part of contraceptive counseling. Although adolescents should be encouraged to delay onset of sexual activity, adherence to abstinence in this age group is low. Therefore, healthcare providers are encouraged to discuss comprehensive sexual health and the risks/benefits of contraceptive options with all adolescents (56,75).

 

A review of the many options available for contraception is beyond the scope of this discussion. The safety and efficacy of contraceptive methods is also reviewed in the Endotext Chapter entitled “Contraception” (76). It is important to note that currently the most effective form of birth control is the contraceptive implant, followed by the IUD, and the progestin injection (Figures 3) (77).

 

Figure 3. Effectiveness of Contraceptive Options. (Adapted from World Health Organization 2011 and Trussell, 2011. (78,79). * The percentages indicate the number of women out of every 100 who experienced an unintended pregnancy within the first year of typical use of each contraceptive method.

Given their efficacy, safety, and ease of use, in coordination with the American College of Obstetricians and Gynecologists, the AAP currently recommends long-acting reversible contraception (LARC) be considered first-line contraceptive choices for adolescents (56,80).

 

Key points for healthcare providers when recommending contraceptive methods:

 

  • Depot medroxyprogesterone acetate (DMPA) and the contraceptive patch are highly effective methods of contraception that are much safer than pregnancy.
  • It is appropriate to prescribe contraceptives or refer for IUD placement without first conducting a pelvic examination. Screenings for STIs, especially chlamydia, can be performed without a pelvic examination.
  • If appropriate, consistent and correct use of condoms with every act of sexual intercourse should be encouraged.
  • Physicians should have a working knowledge of the different combined hormonal methods and regimens for contraception and medical management of common conditions, such as acne, dysmenorrhea, and heavy menstrual bleeding.
  • Adolescents with chronic illnesses and disabilities (estimated to be16 to 25% of adolescents) have similar sexual health and contraceptive needs as their healthy adolescent counterparts, although the medical illness may complicate contraceptive choices (56).

 

Healthcare providers who desire more information regarding contraception options for adolescent and young adult females, including those with medical conditions, are encouraged to consult the Centers for Disease Control and Prevention (CDC) U.S. Selected Practice Recommendations for Contraceptive Use (81,82).

 

HORMONAL CONTRACEPTIVE METHODS IN FEMALES WITH COMPLEX MEDICAL CONDITIONS

 

Aside from concerns regarding the effects of medications during unplanned pregnancy, recommendations for choice of contraceptive methods in females with primary lipid disorders should follow the same guidelines as outlined for age-appropriate females.  Those with secondary dyslipidemia related to complicated, long-term medical conditions, such as chronic inflammatory diseases (e.g., rheumatoid arthritis, systemic lupus), diabetes, and HIV may require addition considerations prior to recommending use of a specific contraceptive method. The concurrent use of medications may also affect contraceptive choices. Table 2 provides a brief summary of the current guidelines to assist in clinical decision-making. For a more detailed discussion, a review of the US Selected Practice Recommendations (US SPR) for Contraceptive Use, 2016 is recommended (74-81).

 

Table 2. Summary of Classifications for Hormonal Contraceptive Methods and Intrauterine Devices

Condition

Cu-IUD

LNG-IUD

Implants

DMPA

POP

CHCs

Obesity

a. BMI ≥30 kg/m2

1

1

1

1

1

2

b. Menarche <18 years and BMI ≥30 kg/m2

1

1

1

2

1

2

Cardiovascular Disease

a.              Multiple risk factors for ASCVDa

1

2

2*

3*

2*

3/4*

Hypertensionb

a. Adequately controlled hypertension

1*

1*

1*

2*

1*

3*

b. Elevated blood pressure levels (properly taken measurements)

i.  SBP 140–159 mm Hg or DBP 90–99 mm Hg

1*

1*

1*

2*

1*

3*

ii. SBP ≥160 mm Hg or DBP ≥100 mm Hg

1*

2*

2*

3*

2*

4*

c. Vascular disease

1*

2*

2*

3*

2*

4*

Known thrombogenic mutationsc

1*

2*

2*

2*

2*

4*

Rheumatic Diseases

Systemic lupus erythematosusd

a. Positive (or unknown) antiphospholipid antibodies

 

3*

3*

 

3*

4*

Initiation

1*

 

 

3*

 

 

Continuation

1*

 

 

3*

 

 

b. Severe thrombocytopenia

 

2*

2*

 

2*

2*

Initiation

3*

 

 

3*

 

 

Continuation

2*

 

 

2*

 

 

c. Immunosuppressive therapy

 

2*

2*

 

2*

2*

Initiation

2*

 

 

2*

 

 

Continuation

1*

 

 

2*

 

 

d. None of the above

 

2*

2*

 

2*

2*

Initiation

1*

 

 

2*

 

 

Continuation

1*

 

 

2*

 

 

Rheumatoid arthritis

a. Receiving immunosuppressive therapy

 

 

1

2/3*

1

2

Initiation

2

2

 

 

 

 

Continuation

1

1

 

 

 

 

b. Not receiving immunosuppressive therapy

1

1

1

2

1

2

Reproductive Tract Infections and Disorders

a. Irregular pattern without heavy bleeding

1

 

2

2

2

1

Initiation

 

1

 

 

 

 

Continuation

 

1

 

 

 

 

b. Heavy or prolonged bleeding (regular and irregular patterns)

2*

 

2*

2*

2*

1*

Initiation

 

1*

 

 

 

 

Continuation

 

2*

 

 

 

 

Severe dysmenorrhea

2

1

1

1

1

1

HIV

High risk for HIV

 

 

1

1*

1

1

Initiation

2

2

 

 

 

 

Continuation

2

2

 

 

 

 

HIV infectione

 

 

1*

1*

1*

1*

Initiation

 

 

 

 

Continuation

 

 

 

 

a. Clinically well receiving ARV therapy

 

 

Initiation

1

1

 

 

 

 

Continuation

1

1

 

 

 

 

b. Not clinically well or not receiving ARV therapy

 

 

Initiation

2

2

 

 

 

 

Continuation

1

1

 

 

 

 

Endocrine Conditions

Diabetes

a.     Non-insulin dependent and Insulin dependentf

1

2

2

2

2

2

b.     Nephropathy, retinopathy, or neuropathy

1

2

2

3

2

3/4*

Hypothyroid

1

1

1

1

1

1

* Consult the respective appendix for each contraceptive method in the 2016 U.S. Medical Eligibility Criteria for Contraceptive Use for clarifications to the numeric categories.

aOlder age, smoking, diabetes, hypertension, low HDL, high LDL, or high triglyceride levels; bSystolic blood pressure ≥160 mm Hg or diastolic blood pressure ≥100 mm Hg are associated with increased risk for adverse health events as a result of pregnancy; cFactor V Leiden; prothrombin mutation; and protein S, protein C, and antithrombin deficiencies are associated with increased risk for adverse health events as a result of pregnancy; dThis condition is associated with increased risk for adverse health events as a result of pregnancy; dFor women with HIV infection who are not clinically well or not receiving ARV therapy, this condition is associated with increased risk for adverse health events as a result of pregnancy; eInsulin-dependent diabetes; diabetes with nephropathy, retinopathy, neuropathy, or diabetes with other vascular disease; or diabetes of >20 years’ duration are associated with increased risk of adverse health events as a result of pregnancy; fNonvascular disease

 

Categories for classifying hormonal contraceptives and intrauterine devices

1 = A condition for which there is no restriction for the use of the contraceptive method.

2 = A condition for which the advantages of using the method generally outweigh the theoretical or proven risks.

3 = A condition for which the theoretical or proven risks usually outweigh the advantages of using the method.

4 = A condition that represents an unacceptable health risk if the contraceptive method is used.

 

Modified from Curtis, 2016 (81,82).

               

 

WHEN CAN A CONTRACEPTIVE METHOD BE INITIATED?

 

Same day initiation of a contraceptive, often referred to as a “quick start”, should be considered when appropriate, since delayed initiation may represent a barrier. All contraceptive methods can be initiated at any time, including on the day of the visit, if there is reasonable certainty that the adolescent or young adult female is not pregnant. This can be ascertained via history (no intercourse since last menstrual period or less than 7 days from the first day of last menstrual period) and a negative urine pregnancy test. In the setting of uncertainty regarding the possibility of pregnancy, initiation of COC, progestin only pills, and DMPA can proceed as the benefits are thought to outweigh the risks.  Insertion of an IUD, however, should be avoided until the absence of pregnancy can be reasonably confirmed. https://www.acog.org/Clinical-Guidance-and-Publications/Committee-Opinions/Committee-on-Adolescent-Health-Care/Adolescent-Pregnancy-Contraception-and-Sexual-Activity?IsMobileSet=false

 

Although pregnancy tests are often performed before initiating contraception, it should be noted that the accuracy of qualitative urine pregnancy tests varies. Pregnancy detection rates can vary widely because of differences in test sensitivity and the timing of testing relative to missed menses (83,84).  A history of starting a normal menstrual period within the last 7 days or a denial of sexual intercourse since the start of the last normal menstrual period may not always be reliable. In addition, a young adolescent female may not have undergone menarche or may have irregular cycles within the first several months of initiating menarche, making it difficult to use this measure to rule out pregnancy.

 

Prior to starting contraception, expectation of bleeding and possible menstrual changes associated with various methods should be reviewed.

 

For females in which there is an uncertainty about the risk of pregnancy, except for an IUD, the benefits of starting other contraceptive methods likely exceed any risk.  A pregnancy test should be repeated in 2-4 weeks. Additional information is available in the CDC’s U.S. Selected Practice Recommendations for Contraceptive Use.

 

SHOULD LABORATORY SCREENING AND PELVIC EXAMINATION BE PERFORMED PRIOR TO INITIATION OF HORMONE CONTRACEPTION?

 

As opposed to the general population, adolescent and young adult females with known dyslipidemia face a unique challenge. Machado and colleagues, noted dyslipidemia in 33% of 516 women (18-40 years-of-age), often accompanied by a history of smoking and an elevated BMI (85). Those with known medical problems or other special conditions might need additional examinations or tests before being considered appropriate candidates for a particular method of contraception.

 

All adolescent and young adult females at-risk of CVD should have their blood pressure measured before initiation of COCs to ensure there is no underlying hypertension that might be exacerbated by the medication. Measurements of weight and a calculated BMI at baseline is helpful in monitoring changes and offering timely counseling to those who might be concerned about weight change perceived to be associated with their contraceptive method.

 

For most healthy females, few examinations or tests are generally needed before initiation of most contraceptive methods. Research suggests that mandatory laboratory screening prior to initiation of contraceptive methods in this population can increase costs and may impose barriers to contraceptive access, critical in reducing unintended pregnancy (81,82,86). In general, laboratory tests, such as glucose, liver enzymes, hemoglobin and thrombogenic gene variants, pelvic examination and even screening for STD/HIV in the general population, are not recommended prior to initiation of treatment, since they do not contribute substantially to safe and effective use of the contraceptive method.

 

CURRENT RECOMMENDATIONS FOR MONITORING OF ADOLESCENT AND YOUNG ADULT WOMEN WITH DYSLIPIDEMIA DURING CONTRACEPTIVE USE

 

Guidelines for monitoring teenage girls with dyslipidemia during use of contraceptives are lacking. It seems reasonable, however, to measure fasting serum lipid levels within 3 months following initiation of a contraceptive; and less frequently once lipid parameters are stable. In those with an LDL-C 160 mg/dL or more, or multiple additional CVD risk factors (including smoking, diabetes, obesity, hypertension, TGs greater than 250 mg/dL, HDL-C less than 35 mg/dL, or a family history of premature coronary artery disease), use of alternative contraception method should be considered.  Preferred methods of contraception for this at-risk population include the copper IUD, which contains no hormones, followed by a levonorgestrel containing IUD, implant, and progestin only contraceptive pills. All are equally acceptable. No major concerns have been raised with interactions between lipid lowering medication and contraception.

 

 

One of the common legal concerns in treating youth less than 18 years-of-age is that of confidentiality for care surrounding reproductive health. The AAP supports policies of informed consent and protection of confidentiality for adolescents seeking contraception and sexual healthcare services. Confidentiality is critical in all discussions, care recommendations and documentation of sexual identity, sexual practices, sexually transmitted infections (STIs), and contraceptive choices (56,87,88). These concepts are important, since limitations of confidentiality and consent are linked to lower use of contraceptives and higher adolescent pregnancy rates (89-92).

 

Over the past 30 years, in the U.S. states have expanded minors’ authority to consent to health care, including care related to sexual activity. This trend reflects the 1977 U.S. Supreme Court ruling in Carey v. Population Services International that affirmed the constitutional right, in all states, to privacy for a minor to obtain contraceptives. The ruling also recognizes that while parental involvement is desirable, many minors will remain sexually active but may fail to seek reproductive advice or services if parental consent or acknowledged is required (93).

 

The majority of states have specific laws regarding a minor’s consent to contraception. The Guttmacher report (93) on current state laws and policies found:

 

  • 23 states and the District of Columbia explicitly allow all minors to consent to contraceptive services.
  • 24 states explicitly permit minors to consent to contraceptive services in one or more circumstances.
  • 4 states have no explicit policy on minors’ authority to consent to contraceptive services.

 

For states without specific laws, best practice guidelines, federal statutes and federal case law may support minor confidentiality and consent.  For example, family planning clinics funded by Title X of the federal Public Health Services Act (42 USC §§300–300a-6 [1970]) are required to provide confidential services to adolescents (94).

 

Even when a state has no relevant policy, case law or an explicit limitation, healthcare providers may provide medical care to a mature minor without parental consent, particularly if the state allows a minor to consent to related health services.

 

The Health Insurance Portability and Accountability Act (HIPAA) also specifically addresses minor confidentiality (95). Although HIPAA allows parents access to a minor’s medical record as personal representatives, that access is denied when the minor is provided with confidentiality under state or other laws or when the parent agrees that the minor may have confidential care (96).

 

Therefore, the AAP recommends that pediatricians have clinic policies that explicitly outline applicable confidential services and that healthcare providers discuss (and document) confidentiality policies with all parents and adolescents. HIPAA also states that if there is no applicable state law about the rights of parents to access the protected health information of their children, pediatricians (or other licensed health professionals) may exercise their professional judgment in providing or denying parental access to the medical records.  Providers are encouraged to include detailed documentation of the decision in the child's medical record (96).

 

Insurance, billing, and electronic health record systems create additional challenges, including an ability to maintain the confidentiality of visits, visit content, associated laboratory test results, and payment for the contraceptive method.  For additional discussion of electronic health records, the AAP has published a policy statement on health information technology (97). 

 

Although contraception services should be provided as a confidential service, adolescent females should be encouraged to involve parents or trusted adults whenever possible. In fact, many parents are supportive of minor consent and confidentiality for sexual health services (98,99). Adolescents who discuss sexuality and contraception with a parent or guardian are also more likely to use contraceptives consistently and are less likely to become pregnant (100,101).

 

For individuals who are sexually active, it is important to discuss and document a plan for pregnancy prevention.  Dermatologists have extensive experience with risk monitoring in adolescents. Use of isotretinoin, a medication with known teratogenic potential, requires an FDA mandated pregnancy prevention program (iPLEDGE).  In this program, females are required to undergo monthly pregnancy testing, and pharmacies, wholesalers and prescribers are all required to participate in a system of informed written consent, warning labels, database registration and monthly identification of contraceptive methods.  Despite its attempts to prevent adverse outcomes, the efficacy of this approach is debated (102).

 

SUMMARY AND ADDITIONAL RESOURCES

 

For adolescent and young adult females who may benefit from use of lipid-lowering medication, it is important to consider the individual’s stage of sexual maturation and sexual history in addition to the lipid disorder when making recommendation for contraception.  For those who are sexually active, a comprehensive, developmentally appropriate discussion and documentation of a plan for reproductive health and pregnancy prevention is recommended.  Most adolescents consider healthcare providers a highly reliable source of healthcare information.  Establishing relationships with adolescents and families allow them to inquire about sensitive topics, such as sexuality and relationships, and to promote healthy decision-making. Several organizations provide excellent resources and extensive guidance in the appropriate use of contraceptive methods. With careful attention to confidentiality and reliable implementation of the individual plan for pregnancy prevention, healthcare providers can navigate the legal and ethical concerns while providing appropriate and compassionate care.  When used cautiously in a supportive healthcare environment, lipid-lowering medications are safe and effective in treating lipid disorders in adolescent and young adult females.

 

ACKNOWLEDGEMENTS

 

The authors would like to acknowledge Luke Hamilton, Suzanne Beckett, Dena Hanson, and Ashley Brock for their assistance in preparing and editing this manuscript.

 

REFERENCES

 

  1. Levenson AE, de Ferranti SD. Familial Hypercholesterolemia. 2020 Feb 8. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 27809433.
  2. Shah AS, Wilson DP. Genetic Disorders Causing Hypertriglyceridemia in Children and Adolescents. 2020 Jan 22. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 27809432.
  3. Benuck I, Wilson DP, McNeal C. Secondary Hypertriglyceridemia. 2020 Jun 1. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 27809435.
  4. Tamir I, Heiss G, Glueck CJ, Christensen B, Kwiterovich P, Rifkind BM. Lipid and lipoprotein distributions in white children ages 6-19 yr. The Lipid Research Clinics Program Prevalence Study. J Chronic Dis. 1981;34(1):27-39. doi:10.1016/0021-9681(81)90079-5
  5. Lauer RM, Lee J, Clarke WR. Factors affecting the relationship between childhood and adult cholesterol levels: the Muscatine Study. Pediatrics. 1988;82(3):309-318.
  6. Srinivasan SR, Wattigney W, Webber LS, Berenson GS. Race and gender differences in serum lipoproteins of children, adolescents, and young adults--emergence of an adverse lipoprotein pattern in white males: the Bogalusa Heart Study. Prev Med. 1991;20(6):671-684. doi:10.1016/0091-7435(91)90063-a
  7. Twisk JW, Kemper HC, Mellenbergh GJ. Longitudinal development of lipoprotein levels in males and females aged 12-28 years: the Amsterdam Growth and Health Study. Int J Epidemiol. 1995;24(1):69-77. doi:10.1093/ije/24.1.69
  8. Hickman TB, Briefel RR, Carroll MD, et al. Distributions and trends of serum lipid levels among United States children and adolescents ages 4-19 years: data from the Third National Health and Nutrition Examination Survey. Prev Med. 1998;27(6):879-890. doi:S0091-7435(98)90376-0
  9. Ford ES, Mokdad AH, Ajani UA. Trends in risk factors for cardiovascular disease among children and adolescents in the United States. Pediatrics. 2004;114(6):1534-1544. doi:114/6/1534
  10. Dai S, Fulton JE, Harrist RB, Grunbaum JA, Steffen LM, Labarthe DR. Blood lipids in children: age-related patterns and association with body-fat indices: Project HeartBeat! Am J Prev Med. 2009;37(1 Suppl):56. doi:10.1016/j.amepre.2009.04.012.
  11. Kit BK, Carroll MD, Lacher DA, Sorlie PD, DeJesus JM, Ogden C. Trends in serum lipids among US youths aged 6 to 19 years, 1988-2010. JAMA. 2012;308(6):591-600. doi:10.1001/jama.2012.9136
  12. Mellerio H, Alberti C, Druet C, et al. Novel modeling of reference values of cardiovascular risk factors in children aged 7 to 20 years. Pediatrics. 2012;129(4):1020. doi:10.1542/peds.2011-0449
  13. Drash A, Hengstenberg F. The identification of risk factors in normal children in the development of arteriosclerosis. Ann Clin Lab Sci. 1972;2(5):348-359.
  14. Kit BK, Kuklina E, Carroll MD, Ostchega Y, Freedman DS, Ogden CL. Prevalence of and trends in dyslipidemia and blood pressure among US children and adolescents, 1999-2012. JAMA Pediatr. 2015;169(3):272-279. doi:10.1001/jamapediatrics.2014.3216
  15. Marshall WA, Tanner JM. Variations in pattern of pubertal changes in girls. Arch Dis Child. 1969;44(235):291-303. doi:10.1136/adc.44.235.291
  16. Marshall WA, Tanner JM. Variations in the pattern of pubertal changes in boys. Arch Dis Child. 1970;45(239):13-23. doi: 0.1136/adc.45.239.13
  17. Beccuti G, Ghizzoni L. Normal and abnormal puberty. In: Feingold KR, Anawalt B, Boyce A, et al, eds. Endotext.MDText.com, Inc; South Dartmouth, MA: 2015.
  18. ACOG Committee Opinion No. 651: Menstruation in girls and adolescents: using the menstrual cycle as a vital sign. Obstet Gynecol. 2015;126(6):e143-e146. doi:10.1097/AOG.0000000000001215
  19. Lakshman R, Forouhi NG, Sharp SJ, et al. Early age at menarche associated with cardiovascular disease and mortality. J Clin Endocrinol Metab. 2009;94(12):4953-4960. doi:10.1210/jc.2009-1789
  20. Rudra CL, Williams MA. BMI as a modifying factor in the relations between age at menarche, menstrual cycle characteristics, and risk of preeclampsia. Gynecol Endocrinol. 2005;21(4):200-205. doi:P2G017365M63654M
  21. Freedman DS, Khan LK, Serdula MK, et al. The relation of menarcheal age to obesity in childhood and adulthood: the Bogalusa heart study. BMC Pediatr. 2003;3:3-3. Epub 2003 Apr 30. doi:1471-2431-3-3
  22. Solomon CG, Hu FB, Dunaif A, et al. Menstrual cycle irregularity and risk for future cardiovascular disease. J Clin Endocrinol Metab. 2002;87(5):2013-2017. doi:10.1210/jcem.87.5.8471
  23. Veltman-Verhulst SM, van Rijn BB, Westerveld HE, et al. Polycystic ovary syndrome and early-onset preeclampsia: reproductive manifestations of increased cardiovascular risk. Menopause. 2010;17(5):990-996. doi:10.1097/gme.0b013e3181ddf705
  24. Porkka KV, Viikari JS, Rönnemaa T, Marniemi J, Akerblom HK. Age and gender specific serum lipid and apolipoprotein fractiles of Finnish children and young adults. The Cardiovascular Risk in Young Finns Study. Acta Paediatr. 1994;83(8):838-848. doi:10.1111/j.1651-2227.1994.tb13155.x
  25. Armstrong N, Balding J, Gentle P, Kirby B. Serum lipids and blood pressure in relation to age and sexual maturity. Ann Hum Biol. 1992;19(5):477-487. doi:10.1080/03014469200002312
  26. Morrison JA, Laskarzewski PM, Rauh JL, et al. Lipids, lipoproteins, and sexual maturation during adolescence: the Princeton maturation study. Metabolism. 1979;28(6):641-649. doi:0026-0495(79)90017-9
  27. Altwaijri YA, Day RS, Harrist RB, Dwyer JT, Ausman LM, Labarthe DR. Sexual maturation affects diet-blood total cholesterol association in children: Project HeartBeat! Am J Prev Med. 2009;37(1 Suppl):65. doi:10.1016/j.amepre.2009.04.007
  28. Srinivasan SR, Elkasabany A, Berenson GS. Distribution and correlates of serum high-density lipoprotein subclasses (LpA-I and LpA-I:A-II) in children from a biracial community. The Bogalusa Heart Study. Metabolism. 1998;47(6):757-763. doi:S0026-0495(98)90042-7
  29. Tell GS, Mittelmark MB, Vellar OD. Cholesterol, high density lipoprotein cholesterol and triglycerides during puberty: the Oslo Youth Study. Am J Epidemiol. 1985;122(5):750-761. doi:10.1093/oxfordjournals.aje.a114158
  30. Sprecher DL, Morrison JA, Simbartl LA, et al. Lipoprotein and apolipoprotein differences in black and white girls. The National Heart, Lung, and Blood Institute Growth and Health Study. Arch Pediatr Adolesc Med. 1997;151(1):84-90. doi:10.1001/archpedi.1997.02170380088014
  31. Eissa MA, Mihalopoulos NL, Holubkov R, Dai S, Labarthe DR. Changes in fasting lipids during puberty. J Pediatr. 2016;170:199-205. doi:S0022-3476(15)01371-2
  32. Kreisberg RA, Kasim S. Cholesterol metabolism and aging. Am J Med. 1987;82(1B):54-60. doi:0002-9343(87)90272-5
  33. Grimes SB, Wild R. Effect of pregnancy on lipid metabolism and lipoprotein levels. In: Feingold KR, Anawalt B, Boyce A, et al, eds. Endotext. MDText.com, Inc; South Dartmouth, MA: 2018.
  34. Mukherjee M. Dyslipidemia in pregnancy, expert analysis. American College of Cardiology web site. Updated 2014. Accessed March 8, 2022. https://www.acc.org/latest-in-cardiology/articles/2014/07/18/16/08/dyslipidemia-in-pregnancy
  35. Lippi G, Albiero A, Montagnana M, et al. Lipid and lipoprotein profile in physiological pregnancy. Clin Lab. 2007;53(3-4):173-177.
  36. Maternal physiology. In: Cunningham FG, Leveno KJ, Bloom SL, et al, eds. Williams obstetrics. 25th ed. McGraw-Hill Education Medical; 2018:49-78.
  37. Gupta M, Liti B, Barrett C, Thompson PD, Fernandez AB. Prevention and Management of Hypertriglyceridemia-Induced Acute Pancreatitis During Pregnancy: A Systematic Review. Am J Med. 2022 Jun;135(6):709-714. doi: 10.1016/j.amjmed.2021.12.006. Epub 2022 Jan 23. PMID: 35081380.
  38. Gunderson EP, Chiang V, Pletcher MJ, et al. History of gestational diabetes mellitus and future risk of atherosclerosis in mid-life: the Coronary Artery Risk Development in Young Adults study. J Am Heart Assoc. 2014;3(2):e000490. doi:10.1161/JAHA.113.000490
  39. Grundy SM, Stone NJ, Bailey AL, et al. 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 [published correction appears in J Am Coll Cardiol. 2019 Jun 25;73(24):3237-3241]. J Am Coll Cardiol. 2019;73(24):e285-e350. doi:10.1016/j.jacc.2018.11.003
  40. de Ferranti SD, Steinberger J, Ameduri R, et al. Cardiovascular risk reduction in high-risk pediatric patients: a scientific statement from the American Heart Association. Circulation. 2019;139(13):e603-e634. doi:10.1161/CIR.0000000000000618
  41. Miller, ML, Wright, CC, Rodrigues B, et al. Use of lipid lowering medications in youth. In: Feingold KR, Anawalt B, Boyce A, et al, eds. Endotext. MDText.com, Inc; South Dartmouth, MA: 2020.
  42. Karalis DG, Hill AN, Clifton S, Wild RA. The risks of statin use in pregnancy: a systematic review. J Clin Lipidol. 2016;10(5):1081-1090. doi:S1933-2874(16)30234-3
  43. Kusters DM, Hassani Lahsinoui H, van de Post, J. A., et al. Statin use during pregnancy: a systematic review and meta-analysis. Expert Rev Cardiovasc Ther. 2012;10(3):363-378. doi:10.1586/erc.11.196
  44. Costantine MM, Cleary K, Eunice Kennedy Shriver National Institute of Child Health and Human Development Obstetric--Fetal Pharmacology Research Units Network*. Pravastatin for the prevention of preeclampsia in high-risk pregnant women. Obstet Gynecol. 2013;121(2 Pt 1):349-353. doi:10.1097/AOG.0b013e31827d8ad5
  45. Costantine MM, Cleary K, Hebert MF, et al. Safety and pharmacokinetics of pravastatin used for the prevention of preeclampsia in high-risk pregnant women: a pilot randomized controlled trial. Am J Obstet Gynecol. 2016;214(6):720.e1-720.e17. doi:10.1016/j.ajog.2015.12.038
  46. Jellinger PS, Handelsman Y, Rosenblit PD, et al. American Association of Clinical Endocrinologists and American College of Endocrinology Guidelines for Management of Dyslipidemia and Prevention of Cardiovascular Disease. Endocr Pract. 2017;23(Suppl 2):1-87. doi:10.4158/EP171764.APPGL
  47. Federal Drug Administration. Statins: Drug Safety Communication - FDA requests removal of strongest warning against using cholesterol-lowering statins during pregnancy. FDA web site. Updated 2021. Accessed Mar 8, 2022. https://www.fda.gov/safety/medical-product-safety-information/statins-drug-safety-communication-fda-requests-removal-strongest-warning-against-using-cholesterol.
  48. Reprotox - online database for medication and pregnancy/lactation
  49. Writing Committee, Lloyd-Jones DM, Morris PB, Ballantyne CM, Birtcher KK, Covington AM, DePalma SM, Minissian MB, Orringer CE, Smith SC Jr, Waring AA, Wilkins JT. 2022 ACC Expert Consensus Decision Pathway on the Role of Nonstatin Therapies for LDL-Cholesterol Lowering in the Management of Atherosclerotic Cardiovascular Disease Risk: A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2022 Oct 4;80(14):1366-1418. doi: 10.1016/j.jacc.2022.07.006. Epub 2022 Aug 25. PMID: 36031461.
  50. Wong B, Ooi TC, Keely E. Severe gestational hypertriglyceridemia: A practical approach for clinicians. Obstet Med. 2015 Dec;8(4):158-67. doi: 10.1177/1753495X15594082. Epub 2015 Aug 21. PMID: 27512474; PMCID: PMC4935053.
  51. Kolovou G, Kolovou V, Katsiki N. Volanesorsen: A New Era in the Treatment of Severe Hypertriglyceridemia. J Clin Med. 2022 Feb 13;11(4):982. doi: 10.3390/jcm11040982. PMID: 35207255; PMCID: PMC8880470.
  52. Cruciat G, Nemeti G, Goidescu I, Anitan S, Florian A. Hypertriglyceridemia triggered acute pancreatitis in pregnancy - diagnostic approach, management and follow-up care. Lipids Health Dis. 2020 Jan 4;19(1):2. doi: 10.1186/s12944-019-1180-7. PMID: 31901241; PMCID: PMC6942404.
  53. Rawla P, Sunkara T, Thandra KC, Gaduputi V. Hypertriglyceridemia-induced pancreatitis: updated review of current treatment and preventive strategies. Clin J Gastroenterol. 2018 Dec;11(6):441-448. doi: 10.1007/s12328-018-0881-1. Epub 2018 Jun 19. PMID: 29923163.
  54. Abma JC, Martinez GM. Sexual activity and contraceptive use among teenagers in the United States, 2011-2015. Natl Health Stat Report. 2017;(104):1-23.
  55. Hagan JF, Shaw JS, Duncan PM. Bright futures: Guidelines for health supervision of infants, children, and adolescents. 4th ed. American Academy of Pediatrics; 2017.
  56. American Academy of Pediatrics Committee on Adolescence. Contraception for adolescents. Pediatrics. 2014;134(4):e1244-e1256.. doi:10.1542/peds.2014-2299
  57. Reno H, Park I, Workowski K, Machefsky A, Bachmann L. A guide to taking a sexual history. Centers for Disease Control and Prevention (CDC) web site.  Reviewed January 14, 2022. Accessed February 24, 2022. https://www.cdc.gov/std/treatment/sexualhistory.pdf.
  58. Sedgh G, Finer LB, Bankole A, Eilers MA, Singh S. Adolescent pregnancy, birth, and abortion rates across countries: levels and recent trends. J Adolesc Health. 2015;56(2):223-230. doi:S1054-139X(14)00387-5
  59. Romero L, Pazol K, Warner L, et al. Reduced disparities in birth rates among teens aged 15-19 years - United States, 2006-2007 and 2013-2014. MMWR Morb Mortal Wkly Rep. 2016;65(16):409-414. doi:10.15585/mmwr.mm6516a1
  60. Lindberg L, Santelli J, Desai S. Understanding the Decline in Adolescent Fertility in the United States, 2007-2012. J Adolesc Health. 2016;59(5):577-583. doi:S1054-139X(16)30172-0
  61. Martin JA, Hamilton BE, Osterman MJ, Driscoll AK, Drake P. Births: Final data for 2017. National Vital Statistics Reports. 2018;67(8):1-50. https://www.cdc.gov/nchs/data/nvsr/nvsr67/nvsr67_08-508.pdf.
  62. Santelli JS, Lindberg LD, Finer LB, Singh S. Explaining recent declines in adolescent pregnancy in the United States: the contribution of abstinence and improved contraceptive use. Am J Public Health. 2007;97(1):150-156. doi:AJPH.2006.089169
  63. ACOG Committee on Practice Bulletins-Gynecology. ACOG Practice Bulletin. No. 73: Use of hormonal contraception in women with coexisting medical conditions. Obstet Gynecol. 2006;107(6):1453-1472. doi:107/6/1453
  64. Fotherby K. Oral contraceptives and lipids. BMJ. 1989;298(6680):1049-1050. doi:10.1136/bmj.298.6680.1049
  65. Elkind-Hirsch KE, Darensbourg C, Ogden B, Ogden LF, Hindelang P. Contraceptive vaginal ring use for women has less adverse metabolic effects than an oral contraceptive. Contraception. 2007;76(5):348-356. doi:S0010-7824(07)00350-2
  66. Guazzelli CA, Barreiros FA, Barbosa R, Torloni MR, Barbieri M. Extended regimens of the contraceptive vaginal ring versus hormonal oral contraceptives: effects on lipid metabolism. Contraception. 2012;85(4):389-393. doi:10.1016/j.contraception.2011.08.014
  67. Cheang KI, Essah PA, Sharma S, Wickham EP,3rd, Nestler JE. Divergent effects of a combined hormonal oral contraceptive on insulin sensitivity in lean versus obese women. Fertil Steril. 2011;96(2):353-359.e1. doi:10.1016/j.fertnstert.2011.05.039
  68. Adeniji AA, Essah PA, Nestler JE, Cheang KI. Metabolic effects of a commonly used combined hormonal oral contraceptive in women with and without polycystic ovary syndrome. J Womens Health. 2016;25(6):638-645. doi:10.1089/jwh.2015.5418
  69. Walsh BW, Schiff I, Rosner B, Greenberg L, Ravnikar V, Sacks FM. Effects of postmenopausal estrogen replacement on the concentrations and metabolism of plasma lipoproteins. N Engl J Med. 1991;325(17):1196-1204. doi:10.1056/NEJM199110243251702
  70. Walsh BW, Sacks FM. Effects of low dose oral contraceptives on very low density and low density lipoprotein metabolism. J Clin Invest. 1993;91(5):2126-2132. doi:10.1172/JCI116437
  71. Creasy GW, Fisher AC, Hall N, Shangold GA. Transdermal contraceptive patch delivering norelgestromin and ethinyl estradiol. Effects on the lipid profile. J Reprod Med. 2003;48(3):179-186.
  72. Tuppurainen M, Klimscheffskij R, Venhola M, Dieben TO. The combined contraceptive vaginal ring (NuvaRing) and lipid metabolism: a comparative study. Contraception. 2004;69(5):389-394. doi:S0010782404000241
  73. Kongsayreepong R, Chutivongse S, George P, et al. A multicentre comparative study of serum lipids and apolipoproteins in long-term users of DMPA and a control group of IUD users. World Health Organization. Task Force on Long-Acting Systemic Agents for Fertility Regulation Special Programme of Research, Development and Research Training in Human Reproduction. Contraception. 1993;47(2):177-191. doi:0010-7824(93)90090-T
  74. Westhoff C. Depot medroxyprogesterone acetate contraception. Metabolic parameters and mood changes. J Reprod Med. 1996;41(5 Suppl):401-406.
  75. American Academy of Pediatrics. Contraception Explained: Options for Teens & Adolescents. HealthyChildren.org web site. Updated 2020. Accessed Feb 24, 2022.https://www.healthychildren.org/English/ages-stages/teen/dating-sex/Pages/Birth-Control-for-Sexually-Active-Teens.aspx.
  76. Horvath S, Schreiber CA, Sonalkar S. Contraception. 2018 Jan 17. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–. PMID: 25905371.
  77. Horvath S, Schreiber CA, Sonalkar S. Contraception. In: Feingold KR, Anawalt B, Boyce A, et al, eds. Endotext.MDText.com, Inc; South Dartmouth, MA: 2018.
  78. World Health Organization, Johns Hopkins Bloomberg School of Public Health. Family planning: a global handbook for providers (2018 update): evidence-based guidance developed through worldwide collaboration. 3rd ed. World Health Organization; 2018.
  79. Trussell J. Contraceptive failure in the United States. Contraception. 
    2011;83(5):397-404. doi:10.1016/j.contraception.2011.01.021
  80. Committee on Practice Bulletins-Gynecology, Long-Acting Reversible Contraception Work Group. Practice Bulletin No. 186: Long-acting reversible contraception: implants and intrauterine devices. Obstet Gynecol. 2017;130(5):e251-e269. doi:10.1097/AOG.0000000000002400
  81. Curtis KM, Jatlaoui TC, Tepper NK, et al. U.S. selected practice recommendations for contraceptive use, 2016. MMWR Recomm Rep. 2016;65(4):1-66. doi:10.15585/mmwr.rr6504a1
  82. Curtis KM, Tepper NK, Jatlaoui TC, et al. U.S. medical eligibility criteria for contraceptive use, 2016. MMWR Recomm Rep. 2016;65(3):1-103. doi:10.15585/mmwr.rr6503a1
  83. Cole LA. Human chorionic gonadotropin tests. Expert Rev Mol Diagn. 
    2009;9(7):721-747. doi:10.1586/erm.09.51
  84. Wilcox AJ, Baird DD, Dunson D, McChesney R, Weinberg CR. Natural limits of pregnancy testing in relation to the expected menstrual period. JAMA. 
    2001;286(14):1759-1761. doi:jbr10110
  85. Machado RB, Bernardes CR, de Souza IM, Santana N, Morimoto M. Is lipid profile determination necessary in women wishing to use oral contraceptives? Contraception. 
    2013;87(6):801-805. doi:S0010-7824(12)01034-7
  86. Tepper NK, Steenland MW, Marchbanks PA, Curtis KM. Laboratory screening prior to initiating contraception: a systematic review. Contraception. 2013;87(5):645-649. doi:S0010-7824(12)00736-6
  87. Christopher BA, Pagidipati NJ. Clinical updates in women's health care summary: evaluation and management of lipid disorders: primary and preventive care review. Obstet Gynecol. 2019;133(3):609. doi:10.1097/AOG.0000000000003139.
  88. Ford C, English A, Sigman G. Confidential health care for adolescents: position paper for the Society for Adolescent Medicine. J Adolesc Health. 2004;35(2):160-167. doi:S1054-139X(04)00086-2
  89. Reddy DM, Fleming R, Swain C. Effect of mandatory parental notification on adolescent girls' use of sexual health care services. JAMA. 2002;288(6):710-714. doi:joc11794
  90. Zabin LS, Stark HA, Emerson MR. Reasons for delay in contraceptive clinic utilization. Adolescent clinic and nonclinic populations compared. J Adolesc Health. 1991;12(3):225-232. doi:0197-0070(91)90015-E
  91. Zavodny M. Fertility and parental consent for minors to receive contraceptives. Am J Public Health. 2004;94(8):1347-1351. doi:94/8/1347
  92. Lehrer JA, Pantell R, Tebb K, Shafer MA. Forgone health care among U.S. adolescents: associations between risk characteristics and confidentiality concern. J Adolesc Health. 2007;40(3):218-226. doi:S1054-139X(06)00375-2
  93. Guttmacher Institute. An Overview of Consent to Reproductive Health Services by Young People. Guttmacher Institute web site. Updated 2022. Accessed Feb 24, 2022. https://www.guttmacher.org/state-policy/explore/overview-minors-consent-law
  94. Center for Adolescent Health & the Law. State minor consent laws: A summary. 3rd ed. Chapel Hill, NC: Center for Adolescent Health & the Law; 2010.
  95. The Health Insurance Portability and Accountability Act of 1996 (HIPA), Pub. L. No. 104-191, 110 Stat. 1936. 1996
  96. English A, Ford CA. The HIPAA privacy rule and adolescents: legal questions and clinical challenges. Perspect Sex Reprod Health. 2004;36(2):80-86. doi:368004
  97. Council on Clinical Information Technology. Health information technology and the medical home. Pediatrics. 2011;127(5):978-982. doi:10.1542/peds.2011-0454
  98. Dempsey AF, Singer DD, Clark SJ, Davis MM. Adolescent preventive health care: what do parents want? J Pediatr. 2009;155(5):689-94.e1. doi:10.1016/j.jpeds.2009.05.029
  99. Jones RK, Purcell A, Singh S, Finer LB. Adolescents' reports of parental knowledge of adolescents' use of sexual health services and their reactions to mandated parental notification for prescription contraception. JAMA. 2005;293(3):340-348. doi:293/3/340
  100. Amialchuk A, Gerhardinger L. Contraceptive use and pregnancies in adolescents' romantic relationships: role of relationship activities and parental attitudes and communication. J Dev Behav Pediatr. 2015;36(2):86-97. doi:10.1097/DBP.0000000000000125
  101. Crosby RA, DiClemente RJ, Wingood GM, et al. Low parental monitoring predicts subsequent pregnancy among African-American adolescent females. J Pediatr Adolesc Gynecol. 2002;15(1):43-46. doi:S1083318801001383
  102. Abroms L, Maibach E, Lyon-Daniel K, Feldman SR. What is the best approach to reducing birth defects associated with isotretinoin? PLoS Med. 2006;3(11):e483. doi: 10.1371/journal.pmed.0030483

Osteoporosis: Prevention and Treatment

ABSTRACT

 

Despite the health consequences of osteoporosis and the availability of effective treatments, it is under-diagnosed and under-treated. For example, although 90% of patients with hip fractures have osteoporosis, in 2007 only 20% of patients with fragility fractures were evaluated and treated. In a retrospective study of patients with hip fractures, less than 15% of subjects were diagnosed and less than 13% were treated with medications for osteoporosis, including calcium and vitamin D. Fracture patients require evaluation of secondary causes and treatment of osteoporosis to help prevent subsequent fractures. The preceding chapters summarize the pathogenesis and the clinical evaluation of osteoporosis. This chapter will review established therapeutic options and new approaches for the prevention and treatment of osteoporosis. Strategies include both lifestyle and medical approaches to enhance bone strength.

 

INTRODUCTION

 

Osteoporosis is a major growing global health problem, resulting in 200 million osteoporotic fractures worldwide each year (1,2). Characterized by reduced bone mass and architectural deterioration, it leads to an increased risk of fragility fractures often occurring with minimal trauma such as falling from a standing height. These fractures rise exponentially with advancing age and most commonly involve the spine, hip or distal forearm. An estimated 1 in 2 women and 1 in 4 men aged 50 years and older will suffer a fragility fracture in their remaining lifetime. Hip fractures are the most serious of these fractures, given the high rates of morbidity and mortality. Approximately 50% of patients who sustain a hip fracture lose the ability to walk independently and 12-24% of women suffering a hip fracture die within the 1st year, compared to 33% of men (3-5). Vertebral compression fractures are the most common osteoporotic fractures, but they are often asymptomatic and found incidentally on imaging done for other reasons. Vertebral fractures are, however, associated with high rates of morbidity involving height loss, kyphosis, restrictive lung disease, back pain, and functional impairment. Vertebral fractures are associated with a 5-fold increased risk of future vertebral fractures and a 2 to 3-fold risk of other fragility fractures. Although there are very effective treatments to reduce fracture risk, only 30% of patients with fragility fractures have a bone density test and/or are treated for their underlying osteoporosis. There are currently critically needed national and international efforts to improve fracture care and bone health in women and men. Identification of osteoporosis at the time of a hip, spine, or other fragility fracture is imperative so that patients with fragility fractures can be evaluated for secondary causes of osteoporosis and treated with osteoporosis medications for their underlying bone disease.

 

The preceding chapters summarize the pathogenesis and the clinical evaluation of osteoporosis. This chapter will focus on reviewing established therapeutic options and new approaches for the prevention and treatment of osteoporosis. Strategies include both lifestyle and medical approaches to enhance bone strength and reduce fractures.

 

PATHOPHYSIOLOGY

 

Bone is a dynamic organ with continuous remodeling occurring as osteoclasts resorb bone and osteoblasts form new bone. Among the key regulators of this process is the receptor activator of nuclear factor-kappa B (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system. Interaction between RANKL, produced by the osteoblast lineage, and RANK receptor stimulates osteoclastic differentiation and activity; OPG, made by osteoblasts, is an endogenous decoy receptor that binds with RANKL, inhibiting bone resorption. In addition, the Wnt signaling pathway is involved in activation of transcription of genes that direct the differentiation and proliferation of osteoblasts. In the skeletal life cycle, there is acquisition of peak bone mass during adolescence and young adulthood. For women, bone loss is accelerated surrounding the time of menopause with decreases in bone mineral density (BMD) of approximately 2-3%/year. With advancing age, the decline in BMD occurs at a slower rate of approximately 0.1 to 0.5% per year in women and men.

 

DIAGNOSIS

 

BMD testing is typically measured in the proximal femur and lumbar spine, though the distal radius should be measured in patients with hyperparathyroidism or in those in whom the other major sites cannot be adequately assessed. Each SD below peak bone mass represents approximately 2-fold increase in fracture risk. Osteopenia is present when the BMD is between 1.0 and 2.5 SDs below bone density of young healthy individuals. More than 50% of fragility fractures occur in these patients (6). Osteoporosis is defined as a BMD≤-2.5 SDs of young normal, healthy individuals.

 

Vertebral imaging by DXA or X-ray is useful for identification of spinal fractures that frequently are not clinically evident. The Bone Health and Osteoporosis Foundation (BHOF, previously the National Osteoporosis Foundation) currently recommends DXA for women ≥65 years and men ≥70 years, or earlier if clinical risk factors are present. Physicians should routinely perform height measurements preferably with a stadiometer as there is an association between height loss and spinal fractures. The BHOF Clinical Guide recommends vertebral imaging for spinal fractures in the presence of height loss of 1.5 inches or more and longitudinal height loss of 0.8 inches or more for postmenopausal women and men age 50-69. Vertebral imaging is also recommended in women and men age 70 and 80 years and older, respectively (7). When the diagnosis of a low bone density compared with age-adjusted controls or osteoporosis is made, a work-up to look for secondary causes of osteoporosis should be considered. See Table 1.

 

Table 1. Secondary Causes of Osteoporosis

Endocrinological Abnormalities

Glucocorticoid excess, hyperthyroidism, hypogonadism (androgen insensitivity, Turner’s and Klinefelter’s Syndrome, hyperprolactinemia, premature menopause), anorexia, athlete triad, vitamin D deficiency, hyperparathyroidism, diabetes mellitus (Types 1 and 2)

Cardiovascular, Renal, Pulmonary and Miscellaneous Disorders

Chronic kidney disease, post-transplant bone disease, congestive heart failure, chronic obstructive lung disease, AIDS/HIV

Connective Tissue Disorders

Osteogenesis imperfecta, Ehlers-Danlos syndrome, Marfan Syndrome, ankylosing spondylitis,

Gastrointestinal Diseases

Celiac sprue, Inflammatory bowel disease, post-gastrectomy, primary biliary cirrhosis, bariatric surgery

Hematological Disorders

Multiple myeloma, mastocytosis, leukemia, hemophilia, sickle cell disease, leukemia, lymphoma, thalassemia

Other Genetic Disorders

Homocystinuria, cystic fibrosis, hemochromatosis, hypophosphatasia

Rheumatological Disorders

Ankylosing spondylitis, rheumatoid arthritis

Medications

Aromatase inhibitors, heparin (long term), anticonvulsants, methotrexate, cytoxan, gonadotropin-releasing hormone (GnRH) agonists and antagonists, tamoxifen (in premenopausal women), excess thyroid hormone, lithium, cyclosporine A, tacrolimus, glucocorticoids, thiazolidinediones, depo-medroxyprogesterone (premenopausal women) proton-pump inhibitors, selective serotonin reuptake inhibitors (SSRIs), tenofovir

 

Laboratory evaluation may include the following: Calcium, phosphorus, liver function tests (including alkaline phosphatase), complete blood count, 25-hydroxyvitamin D, 24-hour urine calcium +/- PTH, TSH (if clinical evidence of hyperthyroidism or those already on thyroid hormone replacement), and serum testosterone in men. For select cases, one may consider sending specialized tests for gastrointestinal disorders (tissue transglutaminase with an IgA level for celiac sprue), infiltrative diseases (serum tryptase for mastocytosis), neoplastic (serum and urine protein electrophoresis), or excess glucocorticoid (cortisol levels, dexamethasone suppression test for Cushing’s syndrome).

 

To quantify an individual’s absolute fracture risk, the World Health Organization (WHO) developed the FRAX® calculator (http://www.shef.ac.uk/FRAX), an integrative measure of various risk factors and femoral neck bone mineral density. In addition to BMD, the following risk factors are included - ethnicity, age, BMI, prior fracture history (designated as a previous fracture in adult life that occurred spontaneously or a fracture arising from trauma, which in a healthy individual would not have resulted in a fracture), glucocorticoid use, excessive alcohol (≥3 units per day), smoking, rheumatoid arthritis, and certain secondary causes of osteoporosis. These secondary causes include Type 1 diabetes, osteogenesis imperfecta, long-standing hyperthyroidism, hypogonadism, premature menopause, malnutrition, malabsorption, or liver disease. If the 10-year absolute fracture risk is ≥3% for hip fractures or ≥20% for other major osteoporotic fractures, pharmacologic therapy should be considered (7). The FRAX® calculator should be utilized in postmenopausal woman ≥ 40 years and men ≥ 50 years with osteopenia. Although there are data analyzing the use of FRAX® in patients who have been treated with osteoporosis medications, its use is not currently validated for patients currently or formerly treated with pharmacotherapy for osteoporosis. Additionally, the FDA has approved the use of trabecular bone score (TBS), a structural measure derived from spinal bone density images that is associated with bone microarchitecture and fracture risk. Combining TBS and the FRAX score may increased the predictive value of the absolute fracture risk assessment (8).

 

Although the FRAX® calculator has greatly enhanced treatment of osteopenic women and men at risk for fractures, certain risk factors predictive of fracture risk are not accurately measured in this calculator. Patients on chronic glucocorticoids may warrant treatment earlier or at a lower threshold than determined by FRAX®; further, this tool does not include current or cumulative glucocorticoid doses or duration of treatment (9). Also, of note, spine BMD is not included in the algorithm. Once an initial bone density is measured, a follow-up BMD should be done 1-2 years after the initial screening depending on whether pharmacologic therapy was initiated. Biochemical bone turnover markers, collagen breakdown products, (e.g., N-telopeptide, C-telopeptide) may be helpful in select patients as an indicator of skeletal remodeling or to determine patient’s adherence to treatment.

 

EXERCISE

 

While pharmacological therapies are a major focus of this chapter, exercise and strategies to strengthen muscles and prevent falls are important components of osteoporosis care. Skeletal loading and mechanical loads from muscle forces have important effects on bone strength (10). Meta-analyses and clinical investigations have shown that exercise produces modest increases in BMD often ranging between 1% and 3% (11). Physical activity helps to maximize BMD during adolescence and young adulthood, diminish bone loss during aging, and improve stability and strength to minimize falls and fractures in the elderly (11-14). However, these benefits come from slow skeletal adaptations to training over time. Because it takes three to four months to complete the bone remodeling cycle of bone resorption, formation, and mineralization, a minimum of at least six to eight months of an exercise intervention is likely required to achieve a change in bone mass that is quantifiable (15,16). The benefits of exercise are lost when people stop exercising, therefore lifelong physical activity at all ages is strongly endorsed by the BHOF. Exercise recommendations generally should include weight-bearing, muscle-strengthening, and balance training exercises for 30 minutes 5 days per week or 75 minutes twice weekly, often consistent with other general health recommendations. Weight-bearing exercises are activities that make the body move against gravity such as walking, jogging, dancing, tennis, and Tai Chi. To protect the spine in patients with low spinal bone density, maintaining a straight spine and avoiding arching and twisting are generally recommended.

 

CALCIUM

 

Adequate calcium intake is essential to prevent calcium mobilization from the bone where 99% of calcium is stored. The effects of calcium supplementation on bone depend on age, menopausal status, calcium intake, and vitamin D sufficiency. Increased calcium intake is necessary during acquisition of peak bone mass and with advancing age. Calcium has modest effects on bone density (17). It is ineffective or minimally effective for prevention of bone loss in women within five years of menopause when there may be predominant effects of estrogen deficiency and other hormonal changes.

 

The Institute of Medicine's recommendations for daily calcium intake that meet the requirements of 97% of the population are shown in Table 2 (18). Unless a patient has an underlying disorder of calcium homeostasis, the upper limit of safety is considered 2,500 mg for adults aged 19 to 50 years and 2,000 mg for those >50 years (19). As maximum absorption of elemental calcium is about 500 to 600 mg at once, calcium intakes need to be divided into multiple doses throughout the day.

 

Table 2. Recommended Daily Elemental Calcium Intake (Adapted from 2011 IOM Report)

9-18 years 
Lactating Women

1,300 mg

Women 19-50 years, Men 19-70 years

1,000 mg

Women > 50 years, Men > 70 years

1,200 mg

 

Obtaining calcium through the diet is preferred. While dairy products contain the largest amount of endogenous calcium, many foods including juices, cereals, and cereal bars, may contain added calcium. An 8-ounce glass of milk or calcium-supplemented orange juice contains ~300 mg of elemental calcium, calcium-supplemented soy and almond milk contains ~450 mg, one ounce (or 1 cubic inch) of cheese contains ~200 mg, and certain cereals contain as much as 1000 mg. It is important for physicians to calculate the dietary calcium intake. Resources helpful for patients to calculate their calcium intake include the U.S. dairy council of California website, http://www.healthyeating.org/Healthy-Eating/Healthy-Eating-Tools/Calcium-Quiz.aspx?action=quiz, the International Osteoporosis Foundation website, https://www.iofbonehealth.org/calcium-calculator, and the NOF Clinical Guide also available on the website https://link.springer.com/article/10.1007/s00198-021-05900-y (7). The former allows patients to check off the type and quantity of calcium-containing foods they usually consume and then calculates total daily calcium, with suggestions on how to increase calcium intake to recommended levels. The latter provides an easy tool to calculate calcium intakes from calcium-rich, dietary sources.

 

Supplemental calcium should be used if an individual’s dietary calcium intake does not meet the recommended daily calcium intake. Calcium carbonate contains 40% of elemental calcium and is a commonly used calcium supplement (e.g., Tums™, Oscal™, Caltrate™, and generic preparations). Calcium carbonate should be taken with food because patients with achlorhydria (or those on proton pump inhibitors chronically) cannot absorb this calcium salt well on an empty stomach (20). Adverse effects of calcium carbonate may include bloating and constipation. Calcium citrate (e.g., Citracal™), which contains 24% elemental calcium, is more bioavailable than calcium carbonate, can be taken while fasting and as a result is the formulation suggested when patients are on proton pump inhibitors chronically.

 

There have been a number of concerns related to the use of supplemental calcium and the risk of kidney stones and cardiovascular disease. Data from epidemiologic research and clinical trials suggest that vitamin D reduces the incidence of fractures and may also prevent falls and declining physical function, yet the available data are not consistent (21). Data from the Women’s Health Initiative (WHI) calcium and vitamin D clinical trial (CT) of supplemental calcium (1000 mg daily) plus vitamin D (400 IU daily) versus placebo in 36,282 women showed a 17% increased risk of developing renal stones in those assigned calcium plus vitamin D. However, among those compliant with the calcium plus vitamin D regimen versus placebo, there was a 29% reduced risk of hip fracture over seven years (22). Some evidence suggests that calcium supplements but generally not dietary calcium may be associated with vascular calcifications and an increased risk for myocardial infarction (23). In a prospective study in the National Institutes of Health AARP Diet and Health Study of 388,229 women and men in whom baseline calcium intakes were ascertained after an average of 12 years of follow-up, supplemental but not dietary calcium intakes were associated with excess cardiovascular death in men but not women; adverse cardiovascular effects were only observed among smokers (23) (24). An analysis of the WHI randomized placebo-controlled calcium and vitamin D trial (CT) and the WHI prospective observational study (OS) showed that in the CT, in postmenopausal women who did not take supplemental calcium and vitamin D at baseline, supplemental calcium (1000 mg/day) and vitamin D (400 IU/D) versus placebo for ≥ 5years was associated with a 38% reduction in the risk of hip fracture. In a combined analysis of data from the CT and OS, supplemental calcium and vitamin D reduced the risk of a hip fracture by 35%. In these subset analyses of the large WHI, it is important to note that there were no adverse effects of supplemental calcium plus vitamin D on risks of myocardial infarction, stroke, or other cardiovascular disease (25). Although additional analyses are ongoing, calcium intakes within the ranges recommended by the IOM appear not to increase cardiovascular risk.

 

Recently, however, the United States Preventive Services Task Force (USPSTF) recommended against supplemental calcium (≤1000 mg/day) and low-dose vitamin D (≤400 IU/D) in healthy postmenopausal women due to lack of evidence of benefit in fracture reduction and evidence for increased risk of kidney stones. Thus, the risk of renal stones with calcium supplementation needs to be balanced with fracture reduction. These recommendations did not apply to adults with osteoporosis or vitamin D deficiency (22,26).

 

VITAMIN D

 

Vitamin D insufficiency and deficiency is a common problem in many individuals. Individuals at increased risk for low vitamin D levels include the elderly and those with low vitamin D intake, malabsorption, inadequate sunlight exposure, use of sunblock, dark skin pigment, obesity, chronic kidney disease, and use of medications that increase the metabolism of vitamin D. Vitamin D deficiency and insufficiency are common in adults with hip fractures (30,31). Vitamin D deficiency can lead to reduced calcium absorption, secondary hyperparathyroidism, and increased risk of fractures (30,32-34). Mild vitamin D insufficiency may not cause symptoms, but contributes to low bone mass. Severe vitamin D deficiency causes osteomalacia. In addition, although more data are needed, vitamin D deficiency has been associated with proximal muscle weakness, impaired physical performance, increased risk of falls, and possibly increased risks of some cancers (including colorectal, breast among others) (19,35-41). Deficient levels of vitamin D are generally defined as a 25-(OH) vitamin D <20 ng/ml, relative insufficiency as 21 to 29 ng/ml, and sufficient levels of vitamin D to prevent the rise in parathyroid hormone levels as a 25-(OH) vitamin D ≥ 30 to 32 ng/ml (42). The National Health and Nutrition Examination Survey (NHANES) report showed that 32% of Americans have vitamin D deficiency (43).

 

Sources of dietary intake of vitamin D are limited and these include vitamin D-fortified milk and some soy milks (100 IU/glass), certain cereals, egg yolk, and oily fishes (e.g., salmon, mackerel, and sardines). Multivitamins typically contain 400 IU to 1,000 IU of vitamin D3, and many calcium preparations are supplemented with vitamin D. The NOF recommends 800 to 1000 IU vitamin D daily for adults aged 50 years and older, as do the International Osteoporosis Foundation and Endocrine Society (44,45). The IOM Committee report on the Dietary Reference Intakes for 97.5% of the population in North America was 600 IU/d of vitamin D for children and adults until age 70 and 800 IU/day for adults 71 years and older (46).

 

The USPTF recommended supplemental vitamin D for reduction in fall risk in women aged 65 and older. Although a meta-analysis of 31,022 individuals indicated that the highest quartile of vitamin D intakes (median 800 IU (and range 792 to 2000 IU/d) was associated with a 30% and 14% reduction in the risks of hip fractures and non-vertebral fractures, respectively, the USPSTF reported that recommendations concerning the safety and efficacy of higher doses of vitamin D on fracture reduction await additional research (26,27).

 

In the Vitamin D and Omega-3 Fatty Acid trial, a large randomized, placebo-controlled trial in 25,874 women and men across the United States of the effects of supplemental 2000 IU/d of cholecalciferol versus placebo determined the effect on the primary prevention of cardiovascular, fractures, cancer and other health outcomes. In addition, detailed in-person visits in a sub cohort provide extensive information on effects of supplemental vitamin D and/or omega-3 fatty acids on cardiovascular outcomes, bone health and many other clinical outcomes (28,29). This study found that in general, in a healthy population not preselected for low vitamin D levels or osteoporosis, supplemental vitamin D had no effect on bone density or bone structural measures or incident falls or fractures (194,195,196).

 

Patients with vitamin D deficiency need much higher doses. The upper limit of safety for vitamin D is 4000 IU/day. There are currently differing recommendations regarding the optimal 25-hydroxyvitamin D (25-OHD) level for bone health with the IOM committee recommending a 25-OH D level ≥20-29 ng/mL while several other societies recommend a 25-OHD level ≥30 ng/mL (44,45).

 

In the presence of vitamin D deficiency, it is safe to normalize vitamin D levels to a 25-(OH)D level of 30 ng/ml to prevent the compensatory rise in parathyroid hormone (PTH) level (33,47). This may be done in a variety of ways. High doses of vitamin D may be needed [e.g., 50,000 IU of D2 (ergocalciferol) or equivalent dose of D3(cholecalciferol) weekly for 8 weeks or according to the 25-hydroxyvitamin D level] (45). Individuals with malabsorption often require very high doses of supplemental vitamin D, and may benefit from evaluation by a bone specialist.

 

TREATMENT AND/OR PREVENTION OF OSTEOPOROSIS

 

There are effective therapies for osteoporosis and promising therapeutics under development. The antiresorptive therapies that reduce bone turnover include: bisphosphonates; estrogen or hormone therapy, estrogen agonists/antagonists [selective estrogen-receptor modulators (SERMs)]; calcitonin; and denosumab, a human monoclonal antibody to RANK-ligand. At present there are two FDA-approved anabolic or bone forming osteoporosis therapy, teriparatide [PTH (1-34)] and abaloparatide. Romosozumab is a monoclonal antibody to sclerostin and stimulates bone formation and inhibits bone resorption. In selection of the optimal therapy for a given individual, it is important to consider patient preference, cost, mode of administration, duration of treatment, and the effects of a treatment on reduction of spine, hip and other non-spine fractures. Tables 4 and 5 lists the currently available osteoporosis drugs approved by the FDA, their dosage, indication, and general efficacy for fracture reduction.

 

HORMONE REPLACEMENT THERAPY

 

In postmenopausal women, it is well known that estrogen therapy (ET) and hormone therapy [estrogen plus progesterone (HT)] prevent bone loss and increase BMD through interaction with estrogen receptors on bone cells, activation of tissue-specific genes and proteins, and/or a reduction in cytokines that stimulate osteoclast function (51-54). In addition to the bone density benefit, the Women’s Health Initiative (WHI) did show that HT resulted in a 34% reduction in the risk of hip fractures and clinical spine fractures (55). However, the risks – increases in breast cancer, coronary heart disease (CHD), pulmonary embolism (PE), and stroke, outweighed the benefits. In addition, after cessation of ET or HT, the benefit of fracture reduction is not sustained (56,57). Although data from the WHI show that ET and HT reduce fractures, ET and HT are FDA-approved for the prevention of fractures but not for the treatment of osteoporosis (55,58).

Data has shown potential cardiovascular safety with use of ET in early menopause, though this remains controversial (the “critical window” hypothesis) (59-61).

 

Unlike oral estrogens, in postmenopausal women transdermal estrogens do not adversely affect clotting factors, and are therefore preferred. Transdermal estrogens prevent bone loss and are available in low doses (e.g., 0.014 to 0.0375 mg daily patch applied 2x/week). In women with premature or early menopause, hormone replacement can be considered until the natural age of menopause (51.3 years) (62). Before estrogen is prescribed, the benefits versus the risks of cardiovascular disease, stroke, and breast cancer should be reviewed. When prescribing estrogen, the FDA recommends the following: consider all non-estrogen preparations first for osteoporosis prevention; use the lowest dose of HT/ET for the shortest time interval to achieve therapeutic goals; and prescribe HT/ET when benefits outweigh risks in a given woman.

 

Estrogen Agonist/Antagonists

 

Estrogen Selective agonists/antagonists previously classified as selective estrogen receptor modulators (SERMs) are a class of drugs that bind to estrogen receptors and can selectively function as agonists or antagonists in different tissues. Raloxifene (Evista™) is Food and Drug Administration (FDA) approved for the prevention and treatment of osteoporosis. Raloxifene was also approved by the FDA in 2007 for reduction in the risk of invasive breast cancer in post-menopausal women with osteoporosis and postmenopausal women at high risk for invasive breast cancer. The Multiple Outcomes of Raloxifene Evaluation (MORE) study was a randomized clinical trial of the effects of raloxifene versus placebo on bone density and fractures in 7,705 postmenopausal women (mean age of 67 years) with osteoporosis. Compared with placebo, raloxifene treatment for three years increased BMD of the spine by 2.6% and of the femoral neck by 2.1%. Over three years, raloxifene reduced spine fractures by 55% in women without prevalent vertebral fractures and by 30% in women with more than one prevalent vertebral fracture (63). Raloxifene therapy did not lead to a reduction in hip or wrist fractures, which was further confirmed in the Continuing Outcomes Relevant to Evista (CORE) trial (64). Additional benefits of raloxifene include the reduction in invasive breast cancer risk and mild decreases in LDL-cholesterol, with no effect on the risk of cardiovascular disease.

 

The side effects of raloxifene include an increase in deep venous thrombosis similar to use of estrogen, along with a small increase in hot flashes and leg cramps, and a small increased risk of fatal stroke in the Raloxifene Use for the Heart (RUTH trial).

 

Tamoxifen, a SERM used for the prevention and treatment of estrogen receptor-positive breast cancer, has estrogen-like effects in bone. It also stimulates the endometrium and can result in uterine hyperplasia or malignancy (65). Bazedoxifene, lasofoxifene, and arzoxifene are third-generation SERMs, none of which appear to cause endometrial hyperplasia (66,67). In a study of 7492 postmenopausal women with osteoporosis, women who received bazedoxifene (20 mg or 40 mg daily) compared with placebo had a lower incidence of new vertebral fractures, but not non-spine fractures (68). In a 7-year phase III, placebo-controlled study of 7492 women with osteoporosis , bazodoxifene versus placebo resulted in a 36.5% (40 mg daily dose) and 30.4% (20 mg daily dose) reduction in morphogenic spine fractures and no effect of overall incidence of nonvertebral fractures (69). In October, 2013, a combination of conjugated estrogens plus bazedoxifene (DuaveeTM) was FDA-approved for the treatment of moderate-severe vasomotor symptoms related to menopause and to prevent osteoporosis after menopause.

 

At present raloxifene and bazodoxifene, are the only estrogen agonist/antagonist that are FDA-approved for prevention (raloxifene and bazodoxifene) and treatment (raloxifene only) of osteoporosis.

 

Table 4. Effects of FDA-Approved Hormonal Osteoporosis Therapies on Fractures

Drug

Most Common Dosage

Fracture Risk Reduction

FDA Indications*

Estrogen Therapy (ET) Hormone Therapy (HT)

Many oral and transdermal preparations

Spine, total hip

PMO-Prevention

Selective Estrogen Receptor Modulators
Raloxifene

60 mg PO once daily

Spine

PMO - Prevention & Treatment; Reduce risk of invasive breast cancer in patients with osteoporosis and increased risk of breast cancer.

Basodoxifene + conjugated estrogens

20 mg/0.45 mg PO once daily

Spine

PMO- Prevention

PMO: postmenopausal osteoporosis; GIO: Glucocorticoid-induced osteoporosis

 

CALCITONIN

 

Calcitonin is a 32-amino acid peptide produced by the parafollicular cells of the thyroid that inhibits bone resorption through direct effects on the osteoclasts. Calcitonin is a highly conserved protein, with human and salmon calcitonin differing by only one amino acid. Injectable salmon calcitonin was approved by the FDA in 1984 for the treatment of osteoporosis, although current use is limited because of the availability of other more effective medications for the treatment of osteoporosis. Calcitonin nasal spray (Miacalcin™ and Fortical™ 200 IU daily) is a form of calcitonin (70) approved by the FDA for the treatment of osteoporosis in women more than five years past menopause. Although studies have shown calcitonin nasal spray to decrease spine fractures, there is no effect on the prevention of hip and other non-spine fractures. Current and future use of calcitonin for osteoporosis has been limited, however, because of data analyses showing a potential increased risk of cancers, particularly liver cancer with calcitonin use, though this remains controversial (71). An FDA review found no causal relationship between calcitonin use and cancer but cautioned that physicians should evaluate the potential benefit to relative risk of calcitonin use in patients.

 

BISPHOSPHONATES

 

Bisphosphonates are analogs of pyrophosphate that inhibit bone turnover and because of their phosphorous-carbon-phosphorous structure are resistant to hydrolysis. They have a strong affinity for calcium crystals and bind avidly to the surface of bone. Bisphosphonates suppress bone resorption and interrupt osteoclast activity directly through several mechanisms including inhibition of acid production, lysosomal enzymes, and the mevalonate pathway (72-74) and indirectly through their effects on osteoblasts and macrophages. They also inhibit osteoclast recruitment and induce osteoclast apoptosis. Thus, through various mechanisms, bisphosphonates reduce the depth of resorption pits (thereby producing positive bone balance at individual bone remodeling units) and decrease the formation of new bone remodeling units.

 

Pharmacodynamics

 

Oral bisphosphonates are poorly absorbed. Less than 3% is absorbed in the fasting state, and absorption is significantly reduced if these drugs are taken with food, calcium, or beverages other than water. The skeleton rapidly takes up approximately half of the absorbed bisphosphonate, and the remainder is excreted unchanged by the kidney within hours. The drug remains at the bone surface for several weeks before becoming embedded in bone, where it is biologically inert. The embedded drug then remains in bone for many years and is slowly released, although the skeletal retention varies among bisphosphonates. Potency and side effects of the bisphosphonates vary according to the side chains (75,76).

 

Effective Therapies for Osteoporosis

 

Alendronate (Fosamax™), risedronate (Actonel™, Atelvia™), ibandronate (Boniva™), and zoledronic acid (Reclast™) are all FDA approved for osteoporosis prevention and/or treatment. Their indication and specific fracture benefits on fracture reduction are shown in Table 5. It is important to select an osteoporosis medication that reduces spine, hip and non-spine fractures, especially in high-risk individuals. Since around 50% of patients discontinue bisphosphonates within 1 year of treatment, it is essential to review compliance and adherence with patients. Of the approved bisphosphonates, Alendronate, Risedronate, and Zoledronic acid are now generic, making them affordable options for patients.

 

ALENDRONATE

 

Several longitudinal studies have shown that oral alendronate increases BMD and decreases the risk of osteoporotic fractures, and can be used for primary and secondary prevention

 

In a meta-analysis of randomized controlled trials published between 1966 and 2007, the efficacy of alendronate in the primary and secondary prevention of osteoporotic fractures in postmenopausal women was evaluated (77). Eleven studies were selected, including three primary prevention studies (78-80) and eight secondary prevention studies involving women with low BMD on DXA and/or high prevalence of vertebral fracture (81-88). A total of 12,068 women received at least one year of oral alendronate (6543 women) or placebo (5525 women). Three trials, including the largest secondary prevention trial, Fracture Intervention Trial (FIT), used an initial daily dose of 5 mg and then switched to 10 mg for the remaining study duration. Other studies used 5 mg, 10 mg, or 20 mg of alendronate daily. The length of follow-up ranged from one to four years, and the mean ages were 53 to 78 years. With alendronate 10 mg daily for secondary prevention, there was a significant 45% relative risk reduction (RRR) in vertebral fractures, 23% RRR in non-vertebral fractures, and 53% RRR in hip fractures. For primary prevention, the RRR was only significant for vertebral fractures (45%). No statistically significant differences in adverse events were found in any included study.

 

The prevalence of osteoporosis is lower in men than in women. It is estimated that one out of two women and one out of four men over age 50 will develop an osteoporotic fracture (89). Several longitudinal studies have evaluated the efficacy of treatment interventions on bone in osteoporotic men. Orwoll et al. enrolled 241 men with a femoral neck T score of ≤ -2 with a lumbar spine T score ≤ -1 or a history of osteoporotic fracture and a femoral neck T score ≤ -1. Compared with placebo, alendronate significantly increased BMD at each site and decreased markers of bone turnover over two years. From baseline, alendronate increased BMD by 3.1% in the total hip and by 7.1% in the lumbar spine and decreased urinary N-telopeptides by 59% and bone-specific alkaline phosphatase by 38%. The incidence of vertebral fractures was 7.1% in the placebo group versus 0.8% in the alendronate group; there was insufficient power to assess the effects of alendronate on non-vertebral fractures (90). Similar results were seen in a smaller study of hypogonadism-induced osteoporosis, indicating no difference in the skeletal response to alendronate in the presence of hypogonadism.

 

Alendronate is also effective in the treatment of glucocorticoid-induced osteoporosis. In glucocorticoid-treated men and women, alendronate resulted in increases in BMD (91,92) and decreases in incidence of radiographic vertebral fractures at two years (6.8% vs. 0.7%) (92).

Data show that weekly alendronate (70 mg) is effective and well tolerated, and this dosage has become the standard of care for use of this oral bisphosphonate. Alendronate is suitable for weekly dosing because of its long skeletal retention. It is often the first line treatment that is cost-effective as a generic preparation.

 

Long-term treatment with alendronate has beneficial effects on BMD. Bone et al. showed that spine BMD continued to rise in small increments during 10 years of treatment. Femoral neck and trochanter BMD increased during the first three years and then remained stable (93,94).

 

In an extension of FIT, the FIT Long-term Extension (FLEX) trial, 1099 women who had received alendronate (5 mg daily for two years and 10 mg daily thereafter) were again randomized to receive either 5 or 10 mg alendronate daily or placebo for five more years. With a pooled analysis of the alendronate doses, after five years, the alendronate-treated subjects had significantly better BMD changes at the total hip, femoral neck, lumbar spine, total body, and forearm. These changes included less loss of BMD at the total hip (placebo 3.38% decrease, pooled alendronate 1.02% decrease) and more gain in BMD at the lumbar spine (placebo 1.52% increase, pooled alendronate 5.26% increase). Subjects on placebo had increases in bone turnover markers compared with alendronate users. Alendronate users had lower risk of clinically recognized vertebral fractures, but the cumulative risk of nonvertebral fractures was not significantly different between the alendronate-treated women and those who received placebo. The authors concluded that for many women the discontinuation of alendronate for up to five years did not appear to significantly increase fracture risk, but women at high risk of vertebral fractures with a history of spinal fracture and a BMD T-score of -2 or less as well as those with osteoporosis according to BMD testing (T-score less than -2.5) after 5 years of treatment may benefit from continued alendronate use (95,96). This trial has limitations because patients with severe osteoporosis were excluded from enrollment, while those with osteopenia were included. There was an uncontrolled phase between FIT completion and FLEX enrollment. There was also a high dropout rate, limiting statistical power (97). As summarized below in the section on a bisphosphonate holiday, with these limitations, risk of fracture versus benefit of continuing treatment should be individualized.

 

RISEDRONATE

 

Risedronate increases BMD and decreases fracture risk among postmenopausal women with osteoporosis. Harris et al. reported data on 2,458 postmenopausal women with established osteoporosis (subjects had either two or more vertebral fractures or one vertebral fracture and lumbar spine T score of -2 or less) and who were randomized to risedronate (5 mg daily) or placebo. Over three years, risedronate increased lumbar spine BMD by 5.4% and femoral neck BMD by 1.6%. Risedronate decreased the risk of new vertebral fractures by 41% and decreased the risk of non-vertebral fractures by 39% at three years (98). Reginster et al. showed in osteoporotic women that risedronate reduced spine fractures within the first year of treatment (99).

 

Risedronate therapy also reduces fracture risk in men (100), and is effective in the prevention and treatment of glucocorticoid-induced osteoporosis in men and women (101).

 

Weekly risedronate (35 mg) preparation used clinically is effective and well tolerated (102-104). Brown et al. randomized 1,468 women to daily or weekly risedronate. The increase in lumbar spine BMD at one year was similar between groups. Weekly risedronate was well tolerated, and the occurrence of adverse events was similar in daily and weekly treatment groups (102). A weekly preparation of risedronate that can be taken after breakfast is also available for clinical use. Monthly dosing of risedronate is available (150 mg once a month). Both monthly dosing regimens were shown to be non-inferior in efficacy and safety to the 5 mg daily regimen at one year (105,106). Thus, monthly risedronate provides alternative regimen for the prevention and treatment of osteoporosis. A formulation that can be taken with food is also available.

 

ZOLEDRONIC ACID

 

Zoledronic acid, an intravenous bisphosphonate, has been FDA approved for years for the treatment of hypercalcemia of malignancy, multiple myeloma, and bone metastases from solid tumors. In August 2007, zoledronic acid (Reclast®) became the second intravenous bisphosphonate after ibandronate (Boniva®) to be FDA approved for treatment of postmenopausal osteoporosis. It is considerably more potent than other available bisphosphonates. Thus, small doses and longer dosing intervals may be used (107). Reid et al. showed that zoledronic acid (4 mg annually) increases BMD and decreases markers of bone turnover in postmenopausal women.

 

In the Health Outcomes and Reduced Incidence with Zoledronic Acid Once Yearly (HORIZON) study, a double-blind, placebo-controlled trial of 7765 post-menopausal women with osteoporosis were randomly assigned to receive a single 15-minute infusion of 5 mg of zoledronic acid or placebo at baseline, at 12 months, and at 24 months. The patients were followed over 36 months. In addition to positive effects on BMD and reduction in bone turnover biomarkers, treatment with zoledronic acid was associated with 70% RRR in morphogenic vertebral fractures and 41% RRR in hip fractures compared with placebo (108). Nonvertebral fractures, clinical fractures, and clinical vertebral fractures were reduced by 25%, 33%, and 77%, respectively. While adverse events, including change in renal function, were similar in both study groups, serious atrial fibrillation (AF) occurred slightly more frequently in the zoledronic acid group in the 3-year but not the 6-year data (108). Further analysis of the trial data and possible risk factors for rare AF are presented below under Adverse Effects (109).

 

In a study in 9355 women randomized to zoledronic acid versus placebo, zoledronic acid resulted in an early reduction in clinical fractures at one year that persisted for 3 years (110). Zoledronic acid is also effective in decreasing fracture risk in men (111).

 

In Horizon Recurrent Fracture trial, a double-blind, placebo-controlled study in adults with hip fractures, zoledronic acid versus placebo administered two weeks to 90 days post-surgical repair resulted in a 35% reduction in new clinical fractures and a 28% reduction in mortality (112). In a sub-sample analysis of this multi-national study, vitamin D deficiency was common and the median 25(OH)D level was only 14.7 ng/ml in these hip fracture study participants (113). Most study participants received 50,000 to 125,000 IU vitamin D at least two weeks prior to the zoledronic acid infusion. Once yearly infusion of zoledronic acid administered 2 weeks to 3 months after a hip fracture and after vitamin D supplementation, therefore, produced a decrease in clinical fractures and evidence of improved survival. Zoledronic acid is only FDA-approved therapy to reduce clinical fracture risk in adults with new hip fractures and provides skeletal protection for hip fracture patients as a once a year dosing. Zoledronic acid administered every 18 months for 6 years also decreased fracture incidence in women with low bone mass (197)

 

OTHER BISPHOSPHONATES

 

Ibandronate (oral and IV) is FDA-approved for the prevention and treatment of postmenopausal osteoporosis. In the larger clinical trial, it increased bone density and decreased vertebral fractures with both an oral daily regimen (2.5 mg daily) and an intermittent regimen (20 mg every other day for 12 doses every three months, 150 mg monthly) without reduction in hip fractures (114-116). Thus, unlike other bisphosphonates, ibandronate was not effective in decreasing non-spine fractures.

 

Pamidronate is not FDA approved for use in osteoporosis; however, it is occasionally used “off-label” for patients in patients with esophageal abnormalities (i.e., stricture or achalasia), organ transplants, or osteogenesis imperfecta. In adults, usually 30 to 60 mg is infused over two to four hours every three months. Pamidronate has been shown to increase BMD, but no fracture data are available (117-121).

 

Adverse Effects

 

GI EFFECTS

 

In general, the bisphosphonates are safe medications. Studies showing the long-term safety of alendronate, risedronate, and zoledronic acid are available for up to 10, 7, and 6 years respectively. Oral bisphosphonates are associated with some GI symptoms, and rare cases of severe esophagitis have been reported with alendronate, although reports are not consistent. However, Lanza et al. carried out a placebo-controlled endoscopic study in 277 subjects and found that the incidence of upper GI symptoms and endoscopic lesions was similar in the placebo and weekly alendronate groups (122). While in controlled trials the incidence of GI adverse effects did not differ in alendronate versus placebo groups, in clinical practice some patients discontinue bisphosphonates because of adverse GI experiences.

 

Because of the risk of esophagitis, alendronate is contraindicated for patients with esophageal abnormalities that delay esophageal emptying such as stricture or achalasia, and both alendronate and risedronate should not be used in patients who are unable to stand or sit upright for at least 30 minutes after drug administration because of increased risk of adverse esophageal effects.

 

ATYPICAL FEMUR FRACTURES

 

There has been concern over long-term bisphosphonate use and the reported risk of atypical femur fractures (AFF). AFF are thought to be stress or insufficiency fractures, caused by anti-resorptive-mediated suppression of intracortical remodeling, though the definite pathogenesis remains unclear. The absolute risk of AFF for patients taking bisphosphonates ranges from 3.2 to 50 per 100,000 person-years, but the risk with long-term bisphosphonate use is higher, ~100 per 100,000 person-years.

 

The Second Task Force of the American Society for Bone and Mineral Research (ASBMR) has defined AFF for case recognition. AFF must be located along the femoral diaphysis distal to the lesser trochanter and proximal to the supracondylar flare, and satisfy 4/5 major features: 1) the fracture is associated with minimal or no trauma, 2) the fracture line originates at the lateral cortex and is substantially transverse in its orientation (but can also be oblique as it progresses medially), 3) a complex fracture extends through both cortices and may have medial spike; or an incomplete fracture involves the lateral cortex, 4) the fracture is noncomminuted or minimally comminuted, and/or 5) localized periosteal or endosteal thickening of the lateral cortex is present at the fracture site (“beaking” or “flaring”). Other common features (minor features) include generalized increase in thickness of the femoral diaphyses, prodromal symptom of dull or aching pain in the groin or thigh, bilateral incomplete or complete femoral diaphysis fractures, and delayed fracture healing, though these are not required for case definition. Risk factors include use of bisphosphonates for >3-5 years, low vitamin D levels, and use of glucocorticoids (123).

 

The consensus has been that the number of fractures prevented far exceeds the number of AFF occurring as a result of bisphosphonate therapy, though further data is needed to guide decision-making around AFF risk.

 

Management of AFF recommended by the ASBMR task force includes surgical management with intramedullary fixation nailing or plating if the fracture is complete or incomplete accompanied by pain, with discontinuation of anti-resorptives, and adequate calcium and vitamin D intake. If the fracture is incomplete and pain is minimal, a trial of conservative management may be considered with use of crutches for 2-3 months, though there is a risk of progression to complete fracture with this method. In addition, obtaining X-ray imaging of the contralateral femur is recommended by the FDA, as ~28% of AFF also affect the contralateral leg. AFF noted on X-ray imaging should be followed by higher-order imaging, such as MRI or CT (123). Lastly, teriparatide may be considered in those who do not heal with other therapy (124).

 

OSTEONECROSIS OF THE JAW

 

Bisphosphonate-associated osteonecrosis of the jaws (ONJ) has also drawn attention even though this is a rare occurrence in patients treated with antiresorptive therapies. The International Task Force on Osteonecrosis of the Jaw defines ONJ as exposed bone in the maxillofacial region that does not heal within 8 weeks after identification by a health care provider, with prior exposure to an antiresorptive agent, and no history of radiation to the craniofacial region (125). It has been hypothesized that ONJ is the result of bone remodeling suppression combined with additional factors such as dental intervention or infection (126). Although very rare, it is more common after dental procedures such as tooth extraction. In 2005, the FDA requested that all oral and IV bisphosphonates include a class “precaution” labeling for ONJ. There have been no cases reported in randomized, placebo-controlled trials of alendronate, risedronate, or ibandronate. However, in a 2006 Medline review, 368 published cases were found, 94% of which involved patients receiving intravenous bisphosphonates, 85% of which involved patients with multiple myeloma or metastatic cancer. Only 4% of patients had osteoporosis and data suggests a time- and dose-dependent effect. 60% of reported cases of ONJ occurred after dentoalveolar surgery for infections (tooth extractions), and the remaining 40% were likely related to infection, denture trauma, or other oral trauma (127). Based on both published and unpublished data, the risk of ONJ associated with oral bisphosphonate treatment for osteoporosis is low, estimated between one in 10,000 and less than one in 100,000 patient-treatment years (128). Some experts have suggested stopping bisphosphonates during a time before and after-invasive dental procedures. The American Dental Association 2011 Recommendations indicate that for patients receiving bisphosphonate therapy, the risk of developing osteonecrosis of the jaw is low and that for dental care they do not currently recommend stopping bisphosphonates (129,130). The American Dental Association does recommend maintenance of good dental hygiene and routine dental care.

 

The International Task Force on Osteonecrosis of the Jaw in 2015 reported an incidence of ONJ of 0.001% to 0.01% in osteoporosis patients, which is slightly higher than the incidence in the general population (<0.001%). Risk factors for ONJ included glucocorticoid use, maxillary or mandibular bone surgery, poor oral hygiene, chronic inflammation, diabetes mellitus, ill-fitting dentures, as well as other drugs such as antiangiogenic agents. Incidence is greater in the oncology population (1-15%), who are receiving significantly more frequent and higher doses of anti-resorptives than the osteoporotic population. The task force recommended prevention of ONJ by eliminating or stabilizing oral disease prior to initiation of antiresorptive therapy, and considering the withholding of antiresorptive therapy in those at high risk for ONJ, such as cancer patients receiving bisphosphonates or denosumab and following extensive oral surgery until the surgical site heals with mature mucosal coverage (125). In a 2022 update by the American Association of Oral and Maxillofacial Surgeons’ Position Paper on Medication-Related Osteonecrosis of the Jaws (MRONJ), the risk of MRONJ in osteoporotic patients treated with bisphosphonates was 0.02 to 0.05 percent, compared to 0 to 0.02 percent with placebo (198).

 

ATRIAL FIBRILLATION

 

In the HORIZON trial, serious atrial fibrillation (AF) was seen more frequently in patients who received IV zoledronic acid (50 subjects, 1.5%) than in those who received placebo (20 subjects 0.5%) (108). Significant risk factors were active tachyarrhythmia, congestive heart failure, previous bisphosphonate use, and advanced age (109). In a review of the results from FIT, there were more serious AF cases in the alendronate group (N=47 subjects, 1.5%) than in the placebo group (N=31 subjects, 1.0%), but these differences were not significant (131). These findings raised concern about a risk of AF with bisphosphonate use. In a case-control study published in 2008, researchers found more AF subjects than controls had ever used alendronate (n=47, 6.5% versus n=40, 4.1%) (132). A review of data from multiple trials did not find an association between risedronate use and AF (133). It is unclear how bisphosphonates may increase the risk of AF. Hypotheses include the release of inflammatory cytokines when IV bisphosphonates are administered, calcium shifts that can occur with IV and potent oral bisphosphonates, and relative binding affinity of the various bisphosphonates to bone. Both cytokines and calcium shifts may increase the risk of AF. The FDA released a review of spontaneous post-marketing reports of AF associated with oral and IV bisphosphonates and did not identify a risk of AF (134,135). The FDA continues to monitor such reports.

 

Post-Hip Fracture Care

 

Given the high rates of morbidity and mortality, particularly within the first-year post-fracture, hip fractures are the most serious of the osteoporotic fractures. There is a high prevalence of low vitamin D levels among hip fracture patients that warrants correction at the time of fracture (30,31). Nationally and internationally there is a large gap in fracture care and only 20% of fracture patients are evaluated and treated for their underlying osteoporosis. A fracture liaison service that identifies patients with fractures and initiates bone density testing and treatment has been very effective in reducing costs and improving post-fracture care (136-139). At Brigham and Women’s Hospital (BWH) Endocrinologists and members of the Department of Orthopedic Surgery have worked together since 2004 to implement a hospital-based approach to advance fracture care and reverse the high prevalence of vitamin D deficiency among hip fracture patient using the electronic health record (140). This inter-disciplinary fracture pathway for hip fracture patients called the Brigham Fracture Intervention Team Initiative or “B-Fit®” includes testing of 25(OH)D, calcium, and creatinine levels on admission to the hospital, administration of one dose of 50,000 units of vitamin D, daily supplemental calcium and vitamin D, and an Endocrinology evaluation. Outpatient care coordination between endocrinologists and Orthopedic Surgeons include assessment for secondary causes of osteoporosis, bone density testing, and pharmacological intervention to reduce subsequent fractures (7, 140-142). Many national organizations are seeking to bring together stakeholders and improve patient care so patients with fragility fractures are evaluated and treated for their underlying osteoporosis (7).

 

Other Precautions

 

Bisphosphonates are excreted by the kidneys and should not be used for patients with severe renal insufficiency (creatinine clearance < 35ml/min, Creatinine clearance <30 ml/min for Ibandronate). Studies in cancer patients, in whom cumulative doses are several-fold higher than in osteoporosis patients, show that age, concomitant non-steroidal anti-inflammatory drug use, prior pamidronate use, history of hypercalcemia, renal disease, hypertension, and smoking are risk factors for renal failure (143,144).

 

Approximately 20% to 30% of subjects treated initially with intravenous administration of pamidronate or zoledronic acid (108,145) may develop an acute-phase reactions (e.g. fever, malaise, myalgia), which is typically less severe with subsequent infusions. Patients should be hydrated and often are premedicated with acetaminophen; symptoms are usually mild and transient.

 

Hypocalcemia may occur, but this is usually mild and asymptomatic. To avert marked hypocalcemia, it is important to ensure that the patient is vitamin D sufficient, which according to the authors’ practices, can best be achieved by checking a 25-hydroxy vitamin D level prior to each infusion. In addition, calcium and creatinine levels should be tested before each intravenous bisphosphonate treatment.

 

Bisphosphonate Holiday

 

Bisphosphonates have robust effects on fracture reduction when used for 3-5 years. There are concerns about the long-term use. According to the 2011 FDA review as summarized in the New England Journal of Medicine (146) there is no global regulatory restriction on duration of use. Post-hoc analyses of data from the FIT and FLEX studies for alendronate (up to 10 years of alendronate therapy) and the randomized extension to the HORIZON-Pivotal Fracture Trial (up to 6 years of zoledronic acid therapy) provide some guidance in these important clinical decisions (96,147).

 

According to the available data, alendronate and zoledronic acid may be discontinued in patients at low risk of fracture after 5 or 3 years of therapy, respectively. In the FLEX trial, continuation of alendronate to 10 years duration of therapy did reduce non-vertebral fractures in those with FN T-scores <-2.5 assessed at year 5, but not in those with T-scores >-2.0 at year 5 (96). In the HORIZON extension trial, stopping Zoledronic acid after 3 years duration of therapy did not significantly increase the risk of subsequent fracture in those with T-score >-2.5, no recent fractures, and no greater than 1 risk factor(148). The subgroups of patients who might benefit from continued therapy without holiday at 5 (oral) or 3 (IV) years of therapy include those with T-score <-2.5 at the hip, recent fracture on therapy, and prevalent spine fractures. Otherwise, annual evaluation while on holiday to assess each individuals fracture risk is recommended, in order to decide when to resume therapy (149). High risk individuals may benefit from use of an alternative treatment such as teriparatide or in some instances, raloxifene, during the time of bisphosphonate holiday. Ongoing evaluation of patients on a bisphosphonate holiday is important to reduce the risk of subsequent fractures (95,96,146,147,150).

 

The ASBMR Task Force for managing osteoporosis in patients on long-term bisphosphonate therapy included consideration of continuing therapy in any patients with history of hip, spine, or multiple other osteoporosis fractures before or during therapy, those with hip BMD T-score<=2.5 after treatment, or high fracture risk (151). However, these approaches do not replace clinical judgment.

 

Drug Administration

 

Oral bisphosphonates should be taken in the morning with water on an empty stomach. Because oral bisphosphonates are poorly absorbed, patients should wait at least 30 minutes before ingesting other beverages, food, or medications. To help patients avoid esophageal irritation, they are instructed to swallow oral bisphosphonates with six to eight ounces of water and to remain upright for at least 30 minutes and until they have had their first meal of the day (152). Intravenous preparations must be infused slowly to avoid renal toxicity.

 

When choosing an oral bisphosphonate and in the absence of contraindications, alendronate is often selected as initial therapy because of its efficacy in reduction of spine and non-spine fractures and its availability as a low cost, generic preparation. In addition to alendronate, risedronate has been on the market for more than 10 years and has favorable safety profiles when used in the indicated populations. While oral ibandronate is popular for its monthly dosing schedule, ibandronate reduces the incidence of spine but not non-spine fractures. In addition, ibandronate’s IV dosing is more expensive and requires more frequent dosing than the once-yearly, zoledronic acid. Thus, it has a limited role in osteoporosis treatment. In patients who are unable to comply with the administration requirements of the oral agents, and in those who experience intolerable GI effects, intravenous zoledronic acid is an effective therapy to reduce spine and non-spine fractures. Like alendronate and risedronate, it reduces the incidence of vertebral and nonvertebral fractures. Zoledronic acid (5 mg infusion once a year) should also be considered in patients with a recent hip fracture after two weeks to 90 days. A post-hoc analysis suggested a superior bone density response when zoledronic acid was administered 4-6 weeks after a hip fracture than at the earlier time points (153). Vitamin D deficiency should be optimally corrected prior to use of zoledronic acid.

 

DENOSUMAB

 

Denosumab is the first FDA-approved human monoclonal antibody that binds to the receptor activator of nuclear factor kappa B ligand (RANKL), an important regulator of bone remodeling. RANKL is secreted by osteoblast precursors and binds to its receptor, RANK, located on osteoclasts. Osteoprotegrin is an endogenous cytokine and decoy receptor that binds RANKL and inhibits osteoclast activation (154). The binding of RANKL to RANK promotes osteoclast proliferation, differentiation, activation, and survival. Denosumab inhibits RANKL and osteoclastogenesis and markedly reduces bone resorption.

 

Fracture Data

 

Denosumab is administered for osteoporosis treatment as a subcutaneous injection of 60 mg every 6 months. In its pivotal phase III randomized placebo-controlled study of 7868 osteoporotic women ages 60-90 years (FREEDOM), denosumab compared with placebo given twice yearly for 3 years was associated with a relative decrease in the risk of vertebral, hip, and nonvertebral fractures by 68%, 40%, and 20% respectively (155). In the extension of this trial, denosumab use for up to 10 years was associated with cumulative BMD gains of 21.7% at the lumbar spine and 9.2% at the total hip. Persistent reductions of bone turnover markers and fracture incidence was also noted, with a positive safety profile with up to 10 years of continued use (199).

 

Drug Administration

 

Denosumab may have advantages over current osteoporosis therapies: infrequent dosing (every six months), and rapid, effective, but reversible antiresorptive activity; drug adherence is, however, important to prevent the increase in bone turnover markers after 6 months of therapy.

 

Adverse Effects

 

Adverse effects of densoumab include hypocalcemia, nausea, musculoskeletal pain, serious skin infections (small risk), infections, dermatologic reactions, and cystitis. Infection risk has been a concern based on RANKL inhibition of non-skeletal immune cells causing theoretical immune suppression. The initial FREEDOM trial showed slightly higher infection rates (3 cases in densoumab arm vs. 0 cases in placebo arm of endocarditis, 0.4% risk in densoumab arm vs. <0.1% in placebo arm of severe skin events) while the extension trial showed no increased risk of infection compared to placebo. Furthermore, a meta-analysis failed to show an increased risk of serious infections with denosumab use (157). Given the unclear infection risk, its use in immunocompromised patients should be cautious. In addition, very rare osteonecrosis of the jaw and atypical femur fractures have occurred with denosumab use (similar to bisphosphonates). Stopping denosumab therapy has been shown to result in bone loss and, in some instances, spine fractures (200). Therefore, unlike bisphosphonates, a treatment holiday is not recommended. The FDA recommends initiation of antiresorptive therapy and a number of treatment regimens are undergoing evaluation in an effort to prevent this bone loss.

 

PARATHYROID HORMONE

 

Anabolic Action on Bone

 

Animal studies show that PTH is capable of both anabolic and catabolic actions on bone. PTH stimulates both bone formation and bone resorption; the net effect on BMD depends on the balance between these two processes (160). A continuous infusion of PTH increases both formation and resorption and leads to bone breakdown (160,161). However, intermittent exposure preferentially increases formation, thereby producing an anabolic effect on bone (160,162,163). Therefore, PTH can increase or decrease BMD depending on the pattern of exposure. Dosing PTH in a manner leading to stimulation of bone formation before causing bone resorption has become known as maximizing the “anabolic window” of PTH (164).

 

Cellular Mechanisms

 

PTH acts directly on osteoblasts and cells of the osteoblast lineage. PTH promotes differentiation of pre-osteoblasts to osteoblasts (161) and inhibits osteoblast apoptosis, thereby increasing the number of active osteoblasts (165). Furthermore, PTH triggers the production of several growth factors in bone cells, including insulin-like growth factor I (IGF-I) (161,166).

 

Teriparatide

 

In 2002, the FDA approved teriparatide (Forteo™), injectable recombinant human PTH (1-34), for the treatment of men and postmenopausal women with osteoporosis who are at high risk for fracture (see Table 5). The biologically active fragment PTH (1-34) has properties similar to the full-length molecule PTH (1-84), which is approved for use in Europe. Antiresorptive agents, such as bisphosphonates, increase BMD up to ~ 8%. However, many patients with osteoporosis have lost as much as 30% of their peak bone mass. Thus, agents that have an anabolic effect on bone are desirable (158). PTH directly stimulates bone formation before bone resorption, has robust effects on spinal BMD, improves bone structure, and reduces spine and non-spine fractures. The sequence of changes in bone formation and resorption leads to what is described as the anabolic window (159).

 

FRACTURE DATA

 

In a large multicenter, randomized placebo-controlled trial, Neer et al. reported the effects of PTH (1-34) on bone density and fractures in 1,637 postmenopausal women with baseline vertebral fractures randomized to 20 µg PTH daily, 40 µg PTH daily, or placebo. At a mean of 18 months’ follow-up, 20 µg PTH daily increased lumbar spine BMD by 9.7%, femoral neck BMD by 2.8%, and total hip BMD by 2.6%. There was a decrease of 0.1% at the distal radius, but this was not significantly different from the change seen in the placebo group. PTH (20 µg daily) reduced the risk of vertebral fractures by 65% and non-vertebral fragility fractures by 53% (and is the FDA-approved dose for treatment of osteoporosis). The two PTH (1-34) doses reduced fractures to a similar degree, but headache and nausea were more common in the group receiving the higher dose of 40 µg daily (167).

 

Abaloparatide

 

In 2017, an additional PTH analog was FDA approved for the treatment of post-menopausal osteoporosis. Abaloparatide (Tymlos™) is a parathyroid (1-34) hormone-related protein (PTHrp) analog drug that shares similar anabolic effects as teriparatide. 

 

FRACTURE DATA

 

In the ACTIVE trial, a double-blind, placebo-controlled trial, Miller et al (202) studied the effect of abaloparatide 80 mcg daily versus placebo in 1901 women with osteoporosis and baseline vertebral fractures over 18 months. At a mean of 18 months’ follow-up, abaloparatide increased lumbar spine BMD by 11.2%, femoral neck BMD by 3.6%, and total hip BMD by 4.18%. New vertebral fracture incidence was 0.6% with abaloparatide versus 4.2% with placebo (86% relative risk reduction, p<0.001). There was a 43% relative risk reduction of non-vertebral fracture with abaloparatide, which just met statistical significance, P=0.049.

 

Combination Therapy of Teriparatide and Bisphosphonates or Denosumab

 

The effects of concurrent or sequential therapy with PTH and antiresorptive agents have been studied. Black et al. compared the effects of PTH (1-84), alendronate, or both in combination in postmenopausal women (168). At one year, spine DXA had increased in all three groups. There was no difference in spine DXA between the PTH group and the combination group. However, the PTH group had a significantly greater increase in volumetric BMD of the spine on quantitative CT than the alendronate and combination groups. Finkelstein et al. also carried out a study in men (169). PTH (1-34) was started at 6 months, and all three groups were followed for 30 months. Spine BMD as measured by both DXA and quantitative CT increased to a greater degree in the PTH group than in the alendronate and combination groups. Thus, these studies show no evidence of synergy between PTH and alendronate. Furthermore, alendronate administered prior to teriparatide may impair the anabolic activity of PTH. It is hypothesized that PTH is less effective when bone turnover is suppressed.

 

While concurrent treatment with PTH and alendronate does not appear to be additive, bisphosphonate therapy initiated immediately upon completion of PTH course is beneficial. Rittmaster et al. demonstrated that PTH followed by alendronate produces progressive increases in BMD. In this study, 66 postmenopausal women were randomized to either 50 µg of recombinant human PTH (1-84) daily or placebo for the first year, and then all subjects were treated with alendronate on an open label extension for the second year. During the first year, the PTH group gained 4.3% BMD at the lumbar spine while the placebo group gained 1.3%. During the second year, the PTH group gained 6.3% BMD at the lumbar spine while the placebo group gained 5.7%. Thus, subjects previously treated with PTH continued to gain BMD with subsequent alendronate therapy (158). Black et al. extended their trial mentioned above (168). Post-menopausal women who had received PTH (1-84) in year one were randomly assigned to an additional year of placebo (n = 60) or alendronate (n = 59). Over two years, alendronate after PTH (1-84) led to significant increases in BMD compared to placebo after PTH (1-84), most notable at trabecular bone areas of the spine as assessed by quantitative CT [31% increase in alendronate after PTH (1-84) group versus14% increase in placebo after alendronate group]. Significant BMD loss was seen in year two in the placebo after PTH (1-84)group (170). Kurland et al. reported similar findings in men (171). Twenty-one men were followed for up to two years after discontinuing PTH (1-34). Those who were treated with a bisphosphonate immediately upon completion of the PTH gained an additional 8.9% BMD at the lumbar spine at two years, while the men who did not go on bisphosphonate therapy lost 3.7% BMD at the lumbar spine at one year. These studies support the immediate use of bisphosphonates upon completion of the recommended 24-month course of PTH therapy to consolidate the increases in bone density.

 

The Denosumab and Teriparatide Administration (DATA) trial investigated the combination of denosumab and teriparatide vs. monotherapy for 2 years. Combination therapy of daily teriparatide and denosumab every 6 months showed increases in spine and hip bone density greater than either drug alone (172).In the absence of fracture outcomes, the role of combination teriparatide and denosumab therapy in osteoporosis remains to be determined, but this regimen may be a therapeutic option in patients with severe osteoporosis or in those who have failed conventional therapy. In the DATA-Switch study, an extension of the DATA trial, subjects who were on denosumab only were switched to teriparatide, and those on teriparatide only were switch to denosumab; the former group were found to have bone loss, whereas the latter group have continued BMD increase (173). This may indicate that the choice of initial and subsequent osteoporosis treatment is an important consideration.

 

In an overlap study of teriparatide with alendronate added to teriparatide after 9 months, found a greater increase in BMD with overlap compared to teriparatide alone (174). These findings may be due to a “reopening” of the anabolic window described with teriparatide use. Of note, fracture data is not available.

 

Adverse Effects

 

In general, teriparatide and abaloparatide, injections are well tolerated and have been safely used for a decade (175). PTH is cleared from the circulation within four hours of subcutaneous administration. A daily injection is necessary and transient redness at the injection site has been noted. Headache and nausea occur in less than 10% of subjects receiving a daily dose of teriparatide 20 µg. Mild, early, transient hypercalcemia can occur, but severe hypercalcemia is rare. Prior to starting a PTH or PTH-rp analog, it is suggested to obtain serum calcium, alkaline phosphatase, parathyroid hormone, 25-hydroxyvitamin D, and creatinine levels. Routine monitoring of serum calcium levels while on PTH or PTH-rp is not recommended by the manufacturer, though may be considered. Increases in urinary calcium (by 30 mg per day) and serum uric acid concentrations (by 13%) are seen but do not appear to have clinical consequences.

 

Fisher 344 rats treated with nearly life-long daily teriparatide or abaloparatide have an increased risk of osteosarcoma. Upon approval of teriparatide in 2002, the FDA placed a black box warning about osteosarcoma in rodents treated with teriparatide and the manufacturer has warned against using teriparatide in the following settings: Paget's disease or unexplained elevations of alkaline phosphatase, open epiphyses in children or young adults, bone metastases, prior radiation therapy involving the skeleton, metabolic bone disease other than osteoporosis, and hypercalcemia. As summarized by Cipriani et al in 2013, there have been 3 reported cases of osteosarcoma in adults treated with PTH (1-34), which does not appear to be greater than the prevalence of osteosarcoma in the population (175). In the Osteosarcoma Surveillance Study, a 15-year surveillance study with 7 years of follow-up, there has not been evidence of a causal relationship between use of teriparatide and risk of osteosarcoma in humans. Among the 1448 cases of osteosarcoma, no patient in this study had been previously treated with teriparatide (176).

 

 In 2021, the FDA removed the black box warning for teriparatide based on 18 years of post-marketing surveillance using case-finding studies, which ruled out any but a small potential increase in risk of osteosarcoma in humans with the drug. The FDA no longer limits the lifetime use to a total of 2 years and longer use can be considered in patients at high fracture risk. The black box warning was also removed for abaloparatide, however, use is limited to 2 years in patient’s lifetime until more data is available. Use of teriparatide and abaloparatide, however, should be avoided in patients at risk for osteosarcoma (e.g., younger patients with open epiphyses or those with a history of skeletal malignancies, unexplained alkaline phosphatase, Paget’s disease of bone or radiation therapy to bone).

 

Off Label Uses

 

Teriparatide has been used off-label for numerous reasons, including improvement of bone healing with atypical femur fractures, and for treatment of vertebral fracture pain and fracture healing. More clinical data is needed in these areas. A systemic review of teriparatide use for healing of bisphosphonate-related AFF found anecdotal evidence of beneficial effects on fracture healing, noting the need for prospective data (124). In a small study of 34 patients with acute vertebral fractures given teriparatide vs. risedronate, those who received Teriparatide had lower rates of vertebral collapse, though had no significant difference in back pain scores (177).

 

Drug Administration

 

Teriparatide is supplied in a disposable pen device for subcutaneous injection into the thigh or abdomen. The pen requires refrigeration between uses. The recommended dosage is 20 µg once a day for two years, though its lifetime use may be extended beyond this in certain clinical situations (such as if a patient remains at or returns to a high risk of fracture). Abaloparatide is also supplied in a disposable pen device for subcutaneous injection into the thigh or abdomen, and can be stored at room temperature after first use for up to 30 days. The recommendation dosage is 80 µg once daily for no more than two years.

 

ROMOSOZUMAB

 

Romosozumab is a monoclonal antibody to sclerostin, a potent inhibitor of osteoblast differentiation and bone formation by way of Wnt signaling inhibition.  Animal studies show that blocking the effect of sclerosin was associated with large increases in bone mass. In phase II trials, romosozumab administration shows increased BMD at the spine of 11.3%, as well as increased bone formation and decreased bone resorption (193). A dual effect of transiently increasing markers of bone formation (P1NP) while simultaneously lowering marker of bone resorption (CTX) was also demonstrated in the phase II trial.

 

Fracture Data

 

In its pivotal phase III trial (203) of 7180 women with osteoporosis, romosozumab reduced incidence of vertebral fractures compared to placebo by 73% at 12 months, and 75% at 24-months after transition to denosumab at 12 months. Non-vertebral fracture reduction was not demonstrated. In the ARCH trial (204), 4093 women with severe osteoporosis were randomized to Romosozumab or alendronate for 12 months. Incidence of new vertebral fractures was 4% with Romosozumab vs. 6.3% with alendronate (risk ratio 0.63, p=0.003). Changes in bone density were greater with Romosozumab compared to alendronate, 13.7% vs. 5% increase in lumbar BMD, and 6.2% vs.  2.8% increase in total hip BMD was demonstrated, respectively. In extension data, preservation of BMD accrual was achieved with transition to alendronate for up to 36 months based on trial duration.

 

Adverse Effects

 

Romosozumab has been associated with hypersensitivity reactions such as angioedema and urticaria. The most common side effects were arthralgia and headache (>5%). Cases of ONJ and AFF have been reported. Upon approval by the FDA in 2019, a black box warning was applied regarding a potential risk of heart attack, stroke, and cardiovascular death. In the ARCH trial, there was a higher rate of major adverse cardiac events (MACE), a composite endpoint of cardiovascular death, nonfatal myocardial infarction, and nonfatal stroke. However, in post-hoc pooled analysis by the TIMI Group of both FRAME and ARCH data, a significantly high rate of cardiovascular event was not demonstrated. The Endocrine Society advises that women at high risk for cardiovascular disease or stroke should not be considered romosozumab pending further studies on its cardiovascular risk (201).

 

GLUCOCORTICOID-INDUCED OSTEOPOROSIS

 

Glucocorticoid induced osteoporosis (GIO) affects the spine greater than other sites. The 2010 American College of Rheumatology (ACR) guidelines can be used to help clinicians determine appropriate therapeutic options in those on glucocorticoid therapy (181). Epidemiological data has consistently shown that those taking glucocorticoids have fractures at higher T-scores. Glucocorticoids not only increase bone resorption, but also reduce bone formation. Thus, there are two important steps for targeted intervention—bisphosphonates and teriparatide, respectively. Rapid bone loss is prevalent in the first 6-12 months of glucocorticoid therapy; however, the increased fracture risk is already present within 3 months of initiating glucocorticoids. Thus, bone protection therapy should be started, at the onset, if the duration of glucocorticoids is anticipated to be 3 months or longer. For postmenopausal women and men over age 50, treatment for GIO is determined based on whether the patient’s risk for fracture—using FRAX® and clinical judgment—is low (<10%), moderate (10-20%), or high (>20%). For those taking prednisone dose >7.5 mg/day, the FDA has approved the following bisphosphonates—Risedronate, Alendronate, Zoledronate—and the anabolic agent, Teriparatide, for the treatment of GIO. In a 3-year randomized trial evaluating the prevention and treatment of GIO, teriparatide was statistically superior to alendronate in preventing BMD declines at the spine and hip (182).

 

Table 5. Effects of FDA-Approved Osteoporosis Therapies on Fractures

 

Most Common Dosage

Fracture Risk Reduction

FDA Indications*

Alendronate

70 mg PO weekly

Spine, non-spine, hip

PMO Treatment and Prevention in women, Treatment of osteoporosis in men, GIO treatment.

Ibandronate

150 mg PO monthly;
3 mg IV every 3 months

Spine

PMO Treatment and Prevention in women.

Risedronate

35 mg PO weekly;
150 mg PO monthly

Spine, non-spine, hip

PMO Treatment and Prevention in women, Treatment of osteoporosis in men, GIO treatment.

Zoledronic Acid (ZA)

5 mg IV / year (Treatment)
5 mg every other year (Prevention)

Spine, non-spine, hip

PMO Treatment and Prevention in women, Treatment of osteoporosis in men, GIO treatment

RANKL inhibitor
Denosumab

60 mg SC every 6 months

Spine, non-Spine, hip

PMO-Treatment in women and men at high fracture risk;

PTH - Teriparatide (PTH 1-34)

20 mcg SC daily (for maximum of 2 years)

Spine, non-Spine

PMO and GIO Treatment in women and men at high risk of fracture

PTH- Abaloparatide (PTH-rp 1-34)

80 mcg SC daily (for maximum of 2 years)

Spine, non-spine

PMO treatment in women at high risk of fracture

Anti-Sclerostin Antibody- Romosozumab

210 mcg SC monthly (for maximum of 12 months)

Spine, non-spine

PMO treatment in women at high risk of fracture

PMO: postmenopausal osteoporosis; GIO: Glucocorticoid-induced osteoporosis

 

CONSIDERATIONS REGARDING SELECTION OF ANTI-FRACTURE TREATMENT

 

When approaching a patient at high risk for fracture, several considerations may help guide the initial treatment selection. Anabolic agents (i.e., romosozumab, abaloparatide, or teriparatide) should be considered as first-line agents In patients deemed “very high risk” for fracture. This may include patients with very low T-scores <-3.0 at the lumbar spine or hip, recent fragility fracture, multiple risk factors for fractures or fractures while on approved osteoporosis therapy or intolerance to osteoporosis therapies. If anabolic treatment is contraindicated or not available for a patient, a parental anti-resorptive agent should be considered.

 

Denosumab can also increase bone density and reduce fracture risk in women and men at high risk for fracture. Denosumab is FDA approved to treat glucocorticoid-induced osteoporosis in men and women at high risk for fracture, in women at high fracture risk on adjuvant aromatase inhibitor therapy for breast cancer,  and in men treated with androgen deprivation for prostate cancer.

 

In patients with advanced chronic kidney disease, treatment options can be limited. Bisphosphonates are generally contraindicated in those with eGFR <30-35. Denosumab (Prolia) is the preferred agent for those with more advanced kidney disease, given lack of direct renal toxicity and renal metabolism compared to bisphosphonates. However, it is important to note that though denosumab has been shown to improve bone mineral density in those with advanced renal disease, there is little evidence of fracture reduction in this population. Since patients with CKD may have several different types of metabolic bone diseases including osteoporosis, use of denosumab should be approached with caution given the increased complexity of bone disease in these patients.

 

It is important to note that when selecting an anabolic agent or denosumab, a plan for the next agent in their treatment sequence should be considered at the onset. Anabolic agents are approved for 1-2 years of use, thereafter their effects wane. At present use of teriparatide can be used for more than a total of 2 years in patients at high risk of fracture. Anti-resorptive agents should ideally be given after completion of a course of anabolic in order to prevent the bone loss that occurs with discontinuation of these therapies. Regarding denosumab, this is approved for 5-10 years of continuous use, but at the point when denosumab is discontinued, it must be followed at the time of first missed dose or just after with an alternative anti-resorptive to prevent rapid rebound bone loss and spine fractures. Ongoing research is assessing different approaches to prevent the bone loss associated with the discontinuation of denosumab. In patients with intolerance or a renal contraindication to using bisphosphonates, the options for the treatment sequence must be taken into account and discussed with the patient as part of shared-decision making.

 

Zoledronic acid is an appropriate first-line option for several different patient scenarios. As mentioned in the Zoledronic acid (ZA) section above, it is the optimal choice in patients post-hip fracture given the benefit in morbidity and mortality in this setting. ZA should also be considered in patients at high fracture risk who have upper gastrointestinal/esophageal disease, or significant malabsorption (i.e. post-gastric bypass surgery), as oral bisphosphonates may be associated with increased risk of GI intolerance or poor absorption and efficacy, respectively. In patients with compliance difficulties or major transportation concerns, zoledronic acid may also be optimal given its infrequent and flexible dosing (once yearly, though less frequently may also be appropriate in select patients). This is in contrast to denosumab, which requires strict adherence to an every 6 month schedule of injections in order to avoid the consequence of rebound bone loss if doses are missed or significantly delayed.

 

Lastly, raloxifene may be considered in patients within 10 years of menopause, who are at high fracture risk at the spine, and high risk for breast cancer based on familial history. Otherwise, an oral bisphosphonate (e.g., alendronate or risedronate) or intravenous bisphosphonate or denosumab is preferred over raloxifene as these therapies have been shown to reduce spine and non-spine fractures.

 

Clinical guides from the Bone Health and Osteoporosis Foundation (7), American Association of Clinical Endocrinologists/American College of Endocrinology (205), and the Endocrine Society (201) provide more detailed information on the management of osteoporosis in high- risk patients.

 

 

TREATMENT GAP IN OSTEOPOROSIS THERAPY

 

Despite having highly effective and well-tolerated available therapeutics for the treatment and prevention of osteoporosis, the rate of treatment of at-risk patients is much lower than desired. Based on prescription databases, bisphosphonate use declined by greater than 50% between 2008 and 2012 (183). In addition, the use of bisphosphonates among those with hip fractures declined from 15% in 2004 to only 3% in 2013, which is concerning given the high risk for future fracture in the setting of hip fracture (139). This decline in use temporally coincides with FDA warnings regarding potential risks related to anti-resorptive use, such as rare atypical femur fracture and osteonecrosis of the jaw, though the FDA has not restricted their use based on these risks (184). It is clear that many patients who would benefit from osteoporosis treatment are not receiving it, and this is a major concern for those who treat osteoporosis. Providers must be able to hold thorough and honest discussions with patients regarding the benefits and risks of osteoporosis treatment options in order for patients to accept and comply with needed treatment.

 

CONCLUSION

 

Osteoporosis is a major public health problem that affects approximately 50% of women and 25% of men aged 50 years and older and fractures increase exponentially with advancing age.  At present, a number of safe and very effective osteoporosis therapies are available. Antiresorptive agents, such as the bisphosphonates, raloxifene, estrogen (not approved for treatment) and denosumab increase bone density and reduce fractures. Teriparatide, abaloparatide, and romosozumab are anabolic therapies and their treatment effects are best consolidated with an inhibitor of bone resorption such as a bisphosphonate or denosumab. A comprehensive review of the prevention and treatment of osteoporosis is summarized in the 2022 Bone Health and Osteoporosis Foundation Clinician’s Guide (7). A multifaceted approach including calcium and vitamin D, exercise, pharmacologic therapy, and fall prevention strategies can reduce the risk of fractures and promote independent healthy lives in older men and women.  

 

ACKOWLEDGEMENTS

 

We wish to acknowledge Anjali Grover, MD, Kara Mikulec, MD and Kathryn E. Ackerman, MD, MPH, and thank them for their past contributions to the Endotext chapter and Jill MacLeod for her assistance in preparation of this review.

 

REFERENCES

 

  1. Cooper C, Campion G, Melton LJ 3rd. Hip fractures in the elderly: a world-wide projection. Osteoporos Int. 1992;2:285–289. 
  2. Burge R, Dawson-Hughes B, Solomon DH, Wong JB, King A, Tosteson A. Incidence and economic burden of osteoporosis-related fractures in the United States, 2005-2025. Journal of bone and mineral research: the official journal of the American Society for Bone and Mineral Research 2007; 22:465-475.
  3. Chrischilles EA, Butler CD, Davis CS, Wallace RB. A model of lifetime osteoporosis impact. Arch Intern Med. 1991;151:2026–2032. 
  4. Magaziner J, Lydick E, Hawkes W, Fox KM, Zimmerman SI, Epstein RS, Hebel JR. Excess mortality attributable to hip fracture in white women aged 70 years and older. Am J Public Health. 1997;87:1630–1636. 
  5. Melton LJ 3rd, Thamer M, Ray NF, Chan JK, Chesnut CH 3rd, Einhorn TA, Johnston CC, Raisz LG, Silverman SL, Siris ES. Fractures attributable to osteoporosis: report from the National Osteoporosis Foundation. J Bone Miner Res. 1997;12:16–23. 
  6. Siris ES, Chen YT, Abbott TA, Barrett-Connor E, Miller PD, Wehren LE, Berger ML. Bone mineral density thresholds for pharmacological intervention to prevent fractures. Arch Intern Med. 2004;164:1108–1112. 
  7. LeBoff MS, Greenspan SL, Insogna KL, Lewiecki EM, Saag KG, Singer AJ, Siris ES. The clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2022; 33(10):2049-2102.
  8. McCloskey EV, Oden NC, Harvey WD, Leslie D, Hans H, Johansson, et al. A Meta-analysis of trabecular bone score in fracture risk prediction and its relationship to FRAX. J Bone Miner Res. 2016; 31; 940-948.
  9. Watts NB, Ettinger B, LeBoff MS. Perspective: FRAX Facts. Journal of Bone and Mineral Research. 2009;24:975–979. 
  10. Kohrt WM, Barry DW, Schwartz RS. Muscle forces or gravity: what predominates mechanical loading on bone? Med Sci Sports Exerc. 2009;41:2050–2055. 
  11. Shea B, Bonaiuti D, Iovine R, Negrini S, Robinson V, Kemper HC, Wells G, Tugwell P, Cranney A. Cochrane Review on exercise for preventing and treating osteoporosis in postmenopausal women. Eura Medicophys. 2004;40:199–209. 
  12. Kelley GA, Kelley KS, Kohrt WM. Exercise and bone mineral density in premenopausal women: a meta-analysis of randomized controlled trials. Int J Endocrinol. 2013;2013:741639. 
  13. Khosla S, Bellido TM, Drezner MK, Gordon CM, Harris TB, Kiel DP, Kream BE, LeBoff MS, Lian JB, Peterson CA, Rosen CJ, Williams JP, Winer KK, Sherman SS. Forum on aging and skeletal health: summary of the proceedings of an ASBMR workshop. J Bone Miner Res. 2011;26:2565–2578. 
  14. Kohrt WM, Bloomfield SA, Little KD, Nelson ME, Yingling VR. American College of Sports Medicine Position Stand: physical activity and bone health. Med Sci Sports Exerc. 2004;36:1985–1996. 
  15. Mundy G. Bone remodeling. In: Primer on the metabolic bone diseases and disorders of mineral metabolism, MJ Favus (ed.), Philadelphia: Lippincott Williams & Wilkins, 1999, 30–38.
  16. Watson SL, Weeks BK, Weis LJ, Horan SA, Beck BR. Heavy resistance training is safe and improves bone, function, and stature in postmenopausal women with low to very low bone mass: novel early findings from the LIFTMOR trial. Osteoporos Int. 2015;26(12):2889-2894.
  17. Dawson-Hughes B, Dallal GE, Krall EA, Sadowski L, Sahyoun N, Tannenbaum S. A controlled trial of the effect of calcium supplementation on bone density in postmenopausal women. N Engl J Med. 1990;323:878–883. 
  18. Standing Committee on the Scientific Evaluation of Dietary Reference Intakes. Dietary Reference Intakes for Calcium, Phosphorus, Magnesium, Vitamin D, and Flouride. Washington, DC, National Academy Press.
  19. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Mayne ST, Rosen CJ, Shapses SA. The 2011 Report on Dietary Reference Intakes for Calcium and Vitamin D from the Institute of Medicine: What Clinicians Need to Know. J Clin Endocrinol Metab. 2011;96:53–58. 
  20. Recker RR. Calcium absorption and achlorhydria. N Engl J Med. 1985;313:70–73. 
  21. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Mayne ST, Rosen CJ, Shapses SA. The 2011 Dietary Reference Intakes for Calcium and Vitamin D: what dietetics practitioners need to know. J Am Diet Assoc. 2011;111:524–527. 
  22. Jackson RD, LaCroix AZ, Gass M, Wallace RB, Robbins J, Lewis CE, Bassford T, Beresford SA, Black HR, Blanchette P, Bonds DE, Brunner RL, Brzyski RG, Caan B, Cauley JA, Chlebowski RT, Cummings SR, Granek I, Hays J, Heiss G, Hendrix SL, Howard BV, Hsia J, Hubbell FA, Johnson KC, Judd H, Kotchen JM, Kuller LH, Langer RD, Lasser NL, Limacher MC, Ludlam S, Manson JE, Margolis KL, McGowan J, Ockene JK, O'Sullivan MJ, Phillips L, Prentice RL, Sarto GE, Stefanick ML, Van Horn L, Wactawski-Wende J, Whitlock E, Anderson GL, Assaf AR, Barad D. Calcium plus vitamin D supplementation and the risk of fractures. N Engl J Med. 2006;354:669–683. 
  23. Bolland MJ, Avenell A, Baron JA, Grey A, MacLennan GS, Gamble GD, Reid IR. Effect of calcium supplements on risk of myocardial infarction and cardiovascular events: meta-analysis. Bmj. 2010;341:c3691. 
  24. Xiao Q, Murphy RA, Houston DK, Harris TB, Chow WH, Park Y. Dietary and Supplemental Calcium Intake and Cardiovascular Disease Mortality: The National Institutes of Health-AARP Diet and Health Study. JAMA internal medicine. 2013:1–8. 
  25. Prentice RL, Pettinger MB, Jackson RD, Wactawski-Wende J, Lacroix AZ, Anderson GL, Chlebowski RT, Manson JE, Van Horn L, Vitolins MZ, Datta M, Leblanc ES, Cauley JA, Rossouw JE. Health risks and benefits from calcium and vitamin D supplementation: Women's Health Initiative clinical trial and cohort study. Osteoporos Int. 2013;24:567–580. 
  26. Moyer VA., USPSTF. Vitamin D and Calcium Supplementation to Prevent Fractures in Adults: U.S. Preventive Services Task Force Recommendation Statement. Ann Intern Med. 2013 
  27. Bischoff-Ferrari HA, Willett WC, Orav EJ, Lips P, Meunier PJ, Lyons RA, Flicker L, Wark J, Jackson RD, Cauley JA, Meyer HE, Pfeifer M, Sanders KM, Stahelin HB, Theiler R, Dawson-Hughes B. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012;367:40–49. 
  28. Manson JE, Bassuk SS, Lee IM, Cook NR, Albert MA, Gordon D, Zaharris E, Macfadyen JG, Danielson E, Lin J, Zhang SM, Buring JE. The VITamin D and OmegA-3 TriaL (VITAL): rationale and design of a large randomized controlled trial of vitamin D and marine omega-3 fatty acid supplements for the primary prevention of cancer and cardiovascular disease. Contemp Clin Trials. 2012;33:159–171. 
  29. LeBoff MS, Yue AY, Copeland T, Cook NR, Buring JE, Manson JE. VITAL-Bone Health: rationale and design of two ancillary studies evaluating the effects of vitamin D and/or omega-3 fatty acid supplements on incident fractures and bone health outcomes in the VITamin D and OmegA-3 TriaL (VITAL). Contemp Clin Trials. 2015;41:259–268. 
  30. Leboff MS, Hawkes WG, Glowacki J, Yu-Yahiro J, Hurwitz S, Magaziner J. Vitamin D-deficiency and post-fracture changes in lower extremity function and falls in women with hip fractures. Osteoporos Int. 2008
  31. LeBoff MS, Kohlmeier L, Hurwitz S, Franklin J, Wright J, Glowacki J. Occult vitamin D deficiency in postmenopausal US women with acute hip fracture. Jama. 1999;281:1505–1511. 
  32. Heaney RP, Dowell MS, Hale CA, Bendich A. Calcium absorption varies within the reference range for serum 25-hydroxyvitamin D. J Am Coll Nutr. 2003;22:142–146. 
  33. Haden ST, Fuleihan GE, Angell JE, Cotran NM, LeBoff MS. Calcidiol and PTH levels in women attending an osteoporosis program. Calcif Tissue Int. 1999;64:275–279. 
  34. Chapuy MC, Arlot ME, Duboeuf F, Brun J, Crouzet B, Arnaud S, Delmas PD, Meunier PJ. Vitamin D3 and calcium to prevent hip fractures in the elderly women. N Engl J Med. 1992;327:1637–1642. 
  35. Bischoff-Ferrari HA, Dietrich T, Orav EJ, Hu FB, Zhang Y, Karlson EW, Dawson-Hughes B. Higher 25-hydroxyvitamin D concentrations are associated with better lower-extremity function in both active and inactive persons aged > or =60 y. Am J Clin Nutr. 2004;80:752–758. 
  36. Bischoff HA, Stahelin HB, Dick W, Akos R, Knecht M, Salis C, Nebiker M, Theiler R, Pfeifer M, Begerow B, Lew RA, Conzelmann M. Effects of vitamin D and calcium supplementation on falls: a randomized controlled trial. J Bone Miner Res. 2003;18:343–351. 
  37. Moyer VA. Prevention of Falls in Community-Dwelling Older Adults: U.S. Preventive Services Task Force Recommendation Statement. Ann Intern Med 2012;
  38. Murad MH, Elamin KB, Abu Elnour NO, Elamin MB, Alkatib AA, Fatourechi MM, Almandoz JP, Mullan RJ, Lane MA, Liu H, Erwin PJ, Hensrud DD, Montori VM. The Effect of Vitamin D on Falls: A Systematic Review and Meta-Analysis. J Clin Endocrinol Metab. 2011 
  39. John EM, Schwartz GG, Dreon DM, Koo J. Vitamin D and breast cancer risk: the NHANES I Epidemiologic follow-up study, 1971-1975 to 1992. National Health and Nutrition Examination Survey. Cancer Epidemiol Biomarkers Prev. 1999;8:399–406. 
  40. Rosen CJ, Adams JS, Bikle DD, Black DM, Demay MB, Manson JE, Murad MH, Kovacs CS. The nonskeletal effects of vitamin D: an Endocrine Society scientific statement. Endocr Rev. 2012;33:456–492. 
  41. Giovannucci E, Liu Y, Rimm EB, Hollis BW, Fuchs CS, Stampfer MJ, Willett WC. Prospective study of predictors of vitamin D status and cancer incidence and mortality in men. J Natl Cancer Inst. 2006;98:451–459. 
  42. Dawson-Hughes B, Heaney R. P., Holick M. F., Lips P., Meunier P. J. R. V. Estimates of optimal vitamin D status. Osteoporos Int. 2005;16:713–716. 
  43. Looker AC, Johnson CL, Lacher DA, Pfeiffer CM, Schleicher RL, Sempos CT. Vitamin D status: United States, 2001-2006. NCHS Data Brief. 2011:1–8. 
  44. Dawson-Hughes B, Mithal A, Bonjour JP, Boonen S, Burckhardt P, Fuleihan GE, Josse RG, Lips P, Morales-Torres J, Yoshimura N. IOF position statement: vitamin D recommendations for older adults. Osteoporosis international: a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA 2010; 21:1151-1154.
  45. Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley DA, Heaney RP, Murad MH, Weaver CM. Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. The Journal of clinical endocrinology and metabolism. 2011;96:1911–1930. 
  46. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Mayne ST, Rosen CJ, Shapses SA. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab. 2011;96:53–58. 
  47. Chapuy MC, Preziosi P, Maamer M, Arnaud S, Galan P, Hercberg S, Meunier PJ. Prevalence of vitamin D insufficiency in an adult normal population. Osteoporos Int. 1997;7:439–443. 
  48. Bischoff-Ferrari HA, Willett WC, Wong JB, Giovannucci E, Dietrich T, Dawson-Hughes B. Fracture prevention with vitamin D supplementation: a meta-analysis of randomized controlled trials. Jama. 2005;293:2257–2264. 
  49. Vieth R, Ladak Y, Walfish PG. Age-related changes in the 25-hydroxyvitamin D versus parathyroid hormone relationship suggest a different reason why older adults require more vitamin D. J Clin Endocrinol Metab. 2003;88:185–191. 
  50. Vieth R. Why the optimal requirement for Vitamin D3 is probably much higher than what is officially recommended for adults. J Steroid Biochem Mol Biol. 2004;89-90:575–579. 
  51. Eriksen EF, Colvard DS, Berg NJ, Graham ML, Mann KG, Spelsberg TC, Riggs BL. Evidence of estrogen receptors in normal human osteoblast-like cells. Science. 1988;241:84–86. 
  52. Manolagas SC, Jilka RL. Cytokines, hematopoiesis, osteoclastogenesis, and estrogens. Calcif Tissue Int. 1992;50:199–202. 
  53. Pacifici R, Rifas L, McCracken R, Vered I, McMurtry C, Avioli LV, Peck WA. Ovarian steroid treatment blocks a postmenopausal increase in blood monocyte interleukin 1 release. Proc Natl Acad Sci U S A. 1989;86:2398–2402. 
  54. Pacifici R. Is there a causal role for IL-1 in postmenopausal bone loss? Calcif Tissue Int. 1992;50:295–299. 
  55. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, Kotchen JM, Ockene J. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA. 2002;288:321–333. 
  56. Heiss G, Wallace R, Anderson GL, Aragaki A, Beresford SA, Brzyski R, Chlebowski RT, Gass M, LaCroix A, Manson JE, Prentice RL, Rossouw J, Stefanick ML, Investigators WHI. Health risks and benefits 3 years after stopping randomized treatment with estrogen and progestin. JAMA. 2008;299:1036–1045. 
  57. LaCroix AZ, Chlebowski RT, Manson JE, Aragaki AK, Johnson KC, Martin L, Margolis KL, Stefanick ML, Brzyski R, Curb JD, Howard BV, Lewis CE, Wactawski-Wende J, Investigators WHI. Health outcomes after stopping conjugated equine estrogens among postmenopausal women with prior hysterectomy: a randomized controlled trial. JAMA. 2011;305:1305–1314. 
  58. Espeland MA, Rapp SR, Shumaker SA, Brunner R, Manson JE, Sherwin BB, Hsia J, Margolis KL, Hogan PE, Wallace R, Dailey M, Freeman R, Hays J. Women's Health Initiative Memory S. Conjugated equine estrogens and global cognitive function in postmenopausal women: Women's Health Initiative Memory Study. JAMA. 2004;291:2959–2968. 
  59. Manson JE. The Kronos Early Estrogen Prevention Study by Charlotte Barker. Womens Health (Lond Engl). 2013;9:9–11. 
  60. Harman SM, Black DM, Naftolin F, Brinton EA, Budoff MJ, Cedars MI, Hopkins PN, Lobo RA, Manson JE, Merriam GR, Miller VM, Neal-Perry G, Santoro N, Taylor HS, Vittinghoff E, Yan M, Hodis HN. Arterial imaging outcomes and cardiovascular risk factors in recently menopausal women: a randomized trial. Ann Intern Med. 2014;161:249–260. 
  61. Hodis HN, Mack WJ, Henderson VW, Shoupe D, Budoff MJ, Hwang-Levine J, Li Y, Feng M, Dustin L, Kono N, Stanczyk FZ, Selzer RH, Azen SP, Group ER. Vascular Effects of Early versus Late Postmenopausal Treatment with Estradiol. N Engl J Med. 2016;374:1221–1231. 
  62. North American Menopause S. The 2012 hormone therapy position statement of: The North American Menopause Society. Menopause. 2012;19:257–271. 
  63. Ettinger B, Black DM, Mitlak BH, Knickerbocker RK, Nickelsen T, Genant HK, Christiansen C, Delmas PD, Zanchetta JR, Stakkestad J, Gluer CC, Krueger K, Cohen FJ, Eckert S, Ensrud KE, Avioli LV, Lips P, Cummings SR. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. Jama. 1999;282:637–645. 
  64. Siris ES, Harris ST, Eastell R, Zanchetta JR, Goemaere S, Diez-Perez A, Stock JL, Song J, Qu Y, Kulkarni PM, Siddhanti SR, Wong M, Cummings SR. Skeletal effects of raloxifene after 8 years: results from the continuing outcomes relevant to Evista (CORE) study. J Bone Miner Res. 2005;20:1514–1524. 
  65. Love RR, Mazess RB, Barden HS, Epstein S, Newcomb PA, Jordan VC, Carbone PP, DeMets DL. Effects of tamoxifen on bone mineral density in postmenopausal women with breast cancer. N Engl J Med. 1992;326:852–856. 
  66. Albertazzi P, Sharma S. Urogenital effects of selective estrogen receptor modulators: a systematic review. Climacteric. 2005;8:214–220. 
  67. Burke TW, Walker CL. Arzoxifene as therapy for endometrial cancer. Gynecol Oncol. 2003;90:S40–46. 
  68. Silverman SL, Christiansen C, Genant HK, Vukicevic S, Zanchetta JR, de Villiers TJ, Constantine GD, Chines AA. Efficacy of bazedoxifene in reducing new vertebral fracture risk in postmenopausal women with osteoporosis: results from a 3-year, randomized, placebo-, and active-controlled clinical trial. J Bone Miner Res. 2008;23:1923–1934. 
  69. Palacios S, Silverman SL, de Villiers TJ, Levine AB, Goemaere S, Brown JP, De Cicco Nardone F, Williams R, Hines TL, Mirkin S, Chines AA. Bazedoxifene Study G. A 7-year randomized, placebo-controlled trial assessing the long-term efficacy and safety of bazedoxifene in postmenopausal women with osteoporosis: effects on bone density and fracture. Menopause. 2015;22:806–813. 
  70. Overgaard K, Riis BJ, Christiansen C, Podenphant J, Johansen JS. Nasal calcitonin for treatment of established osteoporosis. Clin Endocrinol (Oxf). 1989;30:435–442. 
  71. Sun LM, Lin MC, Muo CH, Liang JA, Kao CH. Calcitonin nasal spray and increased cancer risk: a population-based nested case-control study. J Clin Endocrinol Metab. 2014;99:4259–4264. 
  72. Zimolo Z, Wesolowski G, Rodan GA. Acid extrusion is induced by osteoclast attachment to bone. Inhibition by alendronate and calcitonin. J Clin Invest. 1995;96:2277–2283. 
  73. Felix R, Graham R, Russell G, Fleisch H. The effect of several diphosphonates on acid phosphohydrolases and other lysosomal enzymes. Biochim Biophys Acta. 1976;429:429–438. ]
  74. Luckman SP, Hughes DE, Coxon FP, Graham R, Russell G, Rogers MJ. Nitrogen-containing bisphosphonates inhibit the mevalonate pathway and prevent post-translational prenylation of GTP-binding proteins, including Ras. Journal of bone and mineral research: the official journal of the American Society for Bone and Mineral Research 1998; 13:581-589.
  75. Fleisch H. Bisphosphonates in Bone Disease from the Laboratory to the Patient. Fourth ed: Academic Press.
  76. Watts NB. Treatment of osteoporosis with bisphosphonates. Endocrinol Metab Clin North Am. 1998;27:419–439. 
  77. Wells GA, Cranney A, Peterson J, Boucher M, Shea B, Robinson V, Coyle D, Tugwell P. Alendronate for the primary and secondary prevention of osteoporotic fractures in postmenopausal women. Cochrane Database Syst Rev. 2008:CD001155. 
  78. Ascott-Evans BH, Guanabens N, Kivinen S, Stuckey BG, Magaril CH, Vandormael K, Stych B, Melton ME. Alendronate prevents loss of bone density associated with discontinuation of hormone replacement therapy: a randomized controlled trial. Arch Intern Med. 2003;163:789–794. 
  79. Cummings SR, Black DM, Thompson DE, Applegate WB, Barrett-Connor E, Musliner TA, Palermo L, Prineas R, Rubin SM, Scott JC, Vogt T, Wallace R, Yates AJ, LaCroix AZ. Effect of alendronate on risk of fracture in women with low bone density but without vertebral fractures: results from the Fracture Intervention Trial. JAMA. 1998;280:2077–2082. 
  80. Hosking D, Chilvers CE, Christiansen C, Ravn P, Wasnich R, Ross P, McClung M, Balske A, Thompson D, Daley M, Yates AJ. Prevention of bone loss with alendronate in postmenopausal women under 60 years of age. Early Postmenopausal Intervention Cohort Study Group. N Engl J Med. 1998;338:485–492. 
  81. Black DM, Cummings SR, Karpf DB, Cauley JA, Thompson DE, Nevitt MC, Bauer DC, Genant HK, Haskell WL, Marcus R, Ott SM, Torner JC, Quandt SA, Reiss TF, Ensrud KE. Randomised trial of effect of alendronate on risk of fracture in women with existing vertebral fractures. Fracture Intervention Trial Research Group. Lancet. 1996;348:1535–1541. 
  82. Adams AL, Shi J, Takayanagi M, Dell RM, Funahashi TT, Jacobsen SJ. Ten-year hip fracture incidence rate trends in a large California population, 1997-2006. Osteoporos Int. 2013;24:373–376. 
  83. Chesnut CH 3rd, McClung MR, Ensrud KE, Bell NH, Genant HK, Harris ST, Singer FR, Stock JL, Yood RA, Delmas PD, et al. Alendronate treatment of the postmenopausal osteoporotic woman: effect of multiple dosages on bone mass and bone remodeling. Am J Med. 1995;99:144–152. 
  84. Dursun N, Dursun E, Yalcin S. Comparison of alendronate, calcitonin and calcium treatments in postmenopausal osteoporosis. Int J Clin Pract. 2001;55:505–509. 
  85. Greenspan S, Field-Munves E, Tonino R, Smith M, Petruschke R, Wang L, Yates J, de Papp AE, Palmisano J. Tolerability of once-weekly alendronate in patients with osteoporosis: a randomized, double-blind, placebo-controlled study. Mayo Clin Proc. 2002;77:1044–1052. 
  86. Greenspan SL, Parker RA, Ferguson L, Rosen HN, Maitland-Ramsey L, Karpf DB. Early changes in biochemical markers of bone turnover predict the long-term response to alendronate therapy in representative elderly women: a randomized clinical trial. Journal of bone and mineral research: the official journal of the American Society for Bone and Mineral Research 1998; 13:1431-1438.
  87. Liberman UA, Weiss SR, Broll J, Minne HW, Quan H, Bell NH, Rodriguez-Portales J, Downs RW Jr, Dequeker J, Favus M. Effect of oral alendronate on bone mineral density and the incidence of fractures in postmenopausal osteoporosis. The Alendronate Phase III Osteoporosis Treatment Study Group. N Engl J Med. 1995;333:1437–1443. 
  88. Pols HA, Felsenberg D, Hanley DA, Stepan J, Munoz-Torres M, Wilkin TJ, Qin-sheng G, Galich AM, Vandormael K, Yates AJ, Stych B. Multinational, placebo-controlled, randomized trial of the effects of alendronate on bone density and fracture risk in postmenopausal women with low bone mass: results of the FOSIT study. Fosamax International Trial Study Group. Osteoporos Int. 1999;9:461–468. 
  89. Office of the Surgeon General. Bone health and osteoporosis: a report of the Surgeon General. Rockville, Md : U.S. Dept. of Health and Human Services, Public Health Service, Office of the Surgeon General.
  90. Orwoll E, Ettinger M, Weiss S, Miller P, Kendler D, Graham J, Adami S, Weber K, Lorenc R, Pietschmann P, Vandormael K, Lombardi A. Alendronate for the treatment of osteoporosis in men. N Engl J Med. 2000;343:604–610. 
  91. Saag KG, Emkey R, Schnitzer TJ, Brown JP, Hawkins F, Goemaere S, Thamsborg G, Liberman UA, Delmas PD, Malice MP, Czachur M, Daifotis AG. Alendronate for the prevention and treatment of glucocorticoid-induced osteoporosis. Glucocorticoid-Induced Osteoporosis Intervention Study Group. N Engl J Med. 1998;339:292–299. 
  92. Adachi JD, Saag KG, Delmas PD, Liberman UA, Emkey RD, Seeman E, Lane NE, Kaufman JM, Poubelle PE, Hawkins F, Correa-Rotter R, Menkes CJ, Rodriguez-Portales JA, Schnitzer TJ, Block JA, Wing J, McIlwain HH, Westhovens R, Brown J, Melo-Gomes JA, Gruber BL, Yanover MJ, Leite MO, Siminoski KG, Nevitt MC, Sharp JT, Malice MP, Dumortier T, Czachur M, Carofano W, Daifotis A. Two-year effects of alendronate on bone mineral density and vertebral fracture in patients receiving glucocorticoids: a randomized, double-blind, placebo-controlled extension trial. Arthritis Rheum. 2001;44:202–211. 
  93. Tonino RP, Meunier PJ, Emkey R, Rodriguez-Portales JA, Menkes CJ, Wasnich RD, Bone HG, Santora AC, Wu M, Desai R, Ross PD. Skeletal benefits of alendronate: 7-year treatment of postmenopausal osteoporotic women. Phase III Osteoporosis Treatment Study Group. J Clin Endocrinol Metab. 2000;85:3109–3115. 
  94. Bone HG, Hosking D, Devogelaer JP, Tucci JR, Emkey RD, Tonino RP, Rodriguez-Portales JA, Downs RW, Gupta J, Santora AC, Liberman UA. Ten years' experience with alendronate for osteoporosis in postmenopausal women. N Engl J Med. 2004;350:1189–1199. 
  95. Black DM, Schwartz AV, Ensrud KE, Cauley JA, Levis S, Quandt SA, Satterfield S, Wallace RB, Bauer DC, Palermo L, Wehren LE, Lombardi A, Santora AC, Cummings SR, Group FR. Effects of continuing or stopping alendronate after 5 years of treatment: the Fracture Intervention Trial Long-term Extension (FLEX): a randomized trial. JAMA. 2006;296:2927–2938. 
  96. Schwartz AV, Bauer DC, Cummings SR, Cauley JA, Ensrud KE, Palermo L, Wallace RB, Hochberg MC, Feldstein AC, Lombardi A, Black DM. Efficacy of continued alendronate for fractures in women with and without prevalent vertebral fracture: the FLEX trial. Journal of bone and mineral research: the official journal of the American Society for Bone and Mineral Research 2010; 25:976-982.
  97. Briot K, Tremollieres F, Thomas T, Roux C. How long should patients take medications for postmenopausal osteoporosis? Joint Bone Spine. 2007;74:24–31. 
  98. Harris ST, Watts NB, Genant HK, McKeever CD, Hangartner T, Keller M, Chesnut CH 3rd, Brown J, Eriksen EF, Hoseyni MS, Axelrod DW, Miller PD. Effects of risedronate treatment on vertebral and nonvertebral fractures in women with postmenopausal osteoporosis: a randomized controlled trial. Vertebral Efficacy With Risedronate Therapy (VERT) Study Group. Jama. 1999;282:1344–1352. 
  99. Reginster J, Minne HW, Sorensen OH, Hooper M, Roux C, Brandi ML, Lund B, Ethgen D, Pack S, Roumagnac I, Eastell R. Randomized trial of the effects of risedronate on vertebral fractures in women with established postmenopausal osteoporosis. Vertebral Efficacy with Risedronate Therapy (VERT) Study Group. Osteoporos Int. 2000;11:83–91. 
  100. Ringe JD, Faber H, Farahmand P, Dorst A. Efficacy of risedronate in men with primary and secondary osteoporosis: results of a 1-year study. Rheumatol Int. 2006;26:427–431. ]
  101. Cohen S, Levy RM, Keller M, Boling E, Emkey RD, Greenwald M, Zizic TM, Wallach S, Sewell KL, Lukert BP, Axelrod DW, Chines AA. Risedronate therapy prevents corticosteroid-induced bone loss: a twelve-month, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Arthritis Rheum. 1999;42:2309–2318. 
  102. Brown JP, Kendler DL, McClung MR, Emkey RD, Adachi JD, Bolognese MA, Li Z, Balske A, Lindsay R. The efficacy and tolerability of risedronate once a week for the treatment of postmenopausal osteoporosis. Calcif Tissue Int. 2002;71:103–111. 
  103. Gordon MS, Gordon MB. Response of bone mineral density to once-weekly administration of risedronate. Endocr Pract. 2002;8:202–207. 
  104. Delaney MF, Hurwitz S, Shaw J, LeBoff MS. Bone density changes with once weekly risedronate in postmenopausal women. J Clin Densitom. 2003;6:45–50. 
  105. Delmas PD, Benhamou CL, Man Z, Tlustochowicz W, Matzkin E, Eusebio R, Zanchetta J, Olszynski WP, Recker RR, McClung MR. Monthly dosing of 75 mg risedronate on 2 consecutive days a month: efficacy and safety results. Osteoporos Int. 2008;19:1039–1045. 
  106. Delmas PD, McClung MR, Zanchetta JR, Racewicz A, Roux C, Benhamou CL, Man Z, Eusebio RA, Beary JF, Burgio DE, Matzkin E, Boonen S. Efficacy and safety of risedronate 150 mg once a month in the treatment of postmenopausal osteoporosis. Bone. 2008;42:36–42. 
  107. Cheer SM, Noble S. Zoledronic acid. Drugs. 2001;61:799–805. 
  108. Black DM, Delmas PD, Eastell R, Reid IR, Boonen S, Cauley JA, Cosman F, Lakatos P, Leung PC, Man Z, Mautalen C, Mesenbrink P, Hu H, Caminis J, Tong K, Rosario-Jansen T, Krasnow J, Hue TF, Sellmeyer D, Eriksen EF, Cummings SR, Trial HPF. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med. 2007;356:1809–1822. 
  109. Cummings SR MP, Eriksen EF, Eastell R, Black DM. Risk factors for serious adverse events of atrial fibrillation in the HORIZON-PFT Trial of zoledronic acid. Am Soc Mineral Bone Res 29th Annual Meeting 2007, Abstract 1056.
  110. Boonen S, Eastell R, Su G, Mesenbrink P, Cosman F, Cauley JA, Reid IR, Claessens F, Vanderschueren D, Lyles KW, Black DM. Time to onset of antifracture efficacy and year-by-year persistence of effect of zoledronic acid in women with osteoporosis. Journal of bone and mineral research: the official journal of the American Society for Bone and Mineral Research 2012; 27:1487-1493.
  111. Boonen S, Reginster JY, Kaufman JM, Lippuner K, Zanchetta J, Langdahl B, Rizzoli R, Lipschitz S, Dimai HP, Witvrouw R, Eriksen E, Brixen K, Russo L, Claessens F, Papanastasiou P, Antunez O, Su G, Bucci-Rechtweg C, Hruska J, Incera E, Vanderschueren D, Orwoll E. Fracture risk and zoledronic acid therapy in men with osteoporosis. N Engl J Med. 2012;367:1714–1723.
  112. Lyles KW, Colon-Emeric CS, Magaziner JS, Adachi JD, Pieper CF, Mautalen C, Hyldstrup L, Recknor C, Nordsletten L, Moore KA, Lavecchia C, Zhang J, Mesenbrink P, Hodgson PK, Abrams K, Orloff JJ, Horowitz Z, Eriksen EF, Boonen S. Zoledronic Acid in Reducing Clinical Fracture and Mortality after Hip Fracture. N Engl J Med 2007; 357:nihpa40967.
  113. Pieper CF, Colon-Emeric C, Caminis J, Betchyk K, Zhang J, Janning C, Shostak J, LeBoff MS, Heaney RR, Lyles KW. Distribution and correlates of serum 25-hydroxyvitamin D levels in a sample of patients with hip fracture. Am J Geriatr Pharmacother. 2007;5:335–340. 
  114. Chesnut IC, Skag A, Christiansen C, Recker R, Stakkestad JA, Hoiseth A, Felsenberg D, Huss H, Gilbride J, Schimmer RC, Delmas PD. Effects of oral ibandronate administered daily or intermittently on fracture risk in postmenopausal osteoporosis. J Bone Miner Res. 2004;19:1241–1249. 
  115. Reginster JY, Adami S, Lakatos P, Greenwald M, Stepan JJ, Silverman SL, Christiansen C, Rowell L, Mairon N, Bonvoisin B, Drezner MK, Emkey R, Felsenberg D, Cooper C, Delmas PD, Miller PD. Efficacy and tolerability of once-monthly oral ibandronate in postmenopausal osteoporosis: 2 year results from the MOBILE study. Ann Rheum Dis. 2006;65:654–661. 
  116. Cranney A, Wells GA, Yetisir E, Adami S, Cooper C, Delmas PD, Miller PD, Papapoulos S, Reginster JY, Sambrook PN, Silverman S, Siris E, Adachi JD. Ibandronate for the prevention of nonvertebral fractures: a pooled analysis of individual patient data. Osteoporos Int. 2008 
  117. Miller RG, Chretien KC, Meoni LA, Liu YP, Klag MJ, Levine MA. Comparison of intravenous pamidronate to standard therapy for osteoporosis: use in patients unable to take oral bisphosphonates. J Clin Rheumatol. 2005;11:2–7. 
  118. Heijckmann AC, Juttmann JR, Wolffenbuttel BH. Intravenous pamidronate compared with oral alendronate for the treatment of postmenopausal osteoporosis. Neth J Med. 2002;60:315–319. 
  119. Vis M, Bultink IE, Dijkmans BA, Lems WF. The effect of intravenous pamidronate versus oral alendronate on bone mineral density in patients with osteoporosis. Osteoporos Int. 2005;16:1432–1435. 
  120. Cauza E, Etemad M, Winkler F, Hanusch-Enserer U, Partsch G, Noske H, Dunky A. Pamidronate increases bone mineral density in women with postmenopausal or steroid-induced osteoporosis. J Clin Pharm Ther. 2004;29:431–436. 
  121. Chan SS, Nery LM, McElduff A, Wilmshurst EG, Fulcher GR, Robinson BG, Stiel JN, Gunton JE, Clifton-Bligh PB. Intravenous pamidronate in the treatment and prevention of osteoporosis. Intern Med J. 2004;34:162–166. 
  122. Lanza F, Sahba B, Schwartz H, Winograd S, Torosis J, Quan H, Reyes R, Musliner T, Daifotis A, Leung A. The upper GI safety and tolerability of oral alendronate at a dose of 70 milligrams once weekly: a placebo-controlled endoscopy study. Am J Gastroenterol. 2002;97:58–64. 
  123. Shane E, Burr D, Abrahamsen B, Adler RA, Brown TD, Cheung AM, Cosman F, Curtis JR, Dell R, Dempster DW, Ebeling PR, Einhorn TA, Genant HK, Geusens P, Klaushofer K, Lane JM, McKiernan F, McKinney R, Ng A, Nieves J, O'Keefe R, Papapoulos S, Howe TS, van der Meulen MC, Weinstein RS, Whyte MP. Atypical subtrochanteric and diaphyseal femoral fractures: second report of a task force of the American Society for Bone and Mineral Research. J Bone Miner Res. 2014;29:1–23. 
  124. Im GI, Lee SH. Effect of Teriparatide on Healing of Atypical Femoral Fractures: A Systemic Review. Journal of bone metabolism. 2015;22:183–189. 
  125. Khan AA, Morrison A, Hanley DA, Felsenberg D, McCauley LK, O'Ryan F, Reid IR, Ruggiero SL, Taguchi A, Tetradis S, Watts NB, Brandi ML, Peters E, Guise T, Eastell R, Cheung AM, Morin SN, Masri B, Cooper C, Morgan SL, Obermayer-Pietsch B, Langdahl BL, Al Dabagh R, Davison KS, Kendler DL, Sandor GK, Josse RG, Bhandari M, El Rabbany M, Pierroz DD, Sulimani R, Saunders DP, Brown JP, Compston J. International Task Force on Osteonecrosis of the J. Diagnosis and management of osteonecrosis of the jaw: a systematic review and international consensus. J Bone Miner Res. 2015;30:3–23. 
  126. Allen MR. Bisphosphonates and Osteonecrosis of the Jaw: Moving from the Bedside to the Bench. Cells Tissues Organs. 2008 
  127. Woo SB, Hellstein JW, Kalmar JR. Narrative [corrected] review: bisphosphonates and osteonecrosis of the jaws. Ann Intern Med. 2006;144:753–761. 
  128. Khosla S, Burr D, Cauley J, Dempster DW, Ebeling PR, Felsenberg D, Gagel RF, Gilsanz V, Guise T, Koka S, McCauley LK, McGowan J, McKee MD, Mohla S, Pendrys DG, Raisz LG, Ruggiero SL, Shafer DM, Shum L, Silverman SL, Van Poznak CH, Watts N, Woo SB, Shane E. Bisphosphonate-associated osteonecrosis of the jaw: report of a task force of the American Society for Bone and Mineral Research. J Bone Miner Res. 2007;22:1479–1491. 
  129. Edwards BJ, Gounder M, McKoy JM, Boyd I, Farrugia M, Migliorati C, Marx R, Ruggiero S, Dimopoulos M, Raisch DW, Singhal S, Carson K, Obadina E, Trifilio S, West D, Mehta J, Bennett CL. Pharmacovigilance and reporting oversight in US FDA fast-track process: bisphosphonates and osteonecrosis of the jaw. Lancet Oncol. 2008;9:1166–1172. 
  130. Hellstein JW, Adler RA, Edwards B, Jacobsen PL, Kalmar JR, Koka S, Migliorati CA, Ristic H. Managing the care of patients receiving antiresorptive therapy for prevention and treatment of osteoporosis: executive summary of recommendations from the American Dental Association Council on Scientific Affairs. J Am Dent Assoc. 2011;142:1243–1251. ]
  131. Cummings SR, Schwartz AV, Black DM. Alendronate and atrial fibrillation. N Engl J Med. 2007;356:1895–1896. 
  132. Heckbert SR, Li G, Cummings SR, Smith NL, Psaty BM. Use of alendronate and risk of incident atrial fibrillation in women. Arch Intern Med. 2008;168:826–831. 
  133. Karam R, Camm J, McClung M. Yearly zoledronic acid in postmenopausal osteoporosis. N Engl J Med. 2007;357:712–713. 
  134. Cauley JA, Ensrud KE. Considering competing risks . . . Not all black and white. Arch Intern Med. 2008;168:793–795. 
  135. Administration. UFaD. Early communication of an ongoing safety review (updated January 7, 2008). http://www.fda.gov/cder/drug/early_comm/bisphosphonates.htm.
  136. Adler RA, Bates DW, Dell RM, LeBoff MS, Majumdar SR, Saag KG, Solomon DH, Suarez-Almazor ME. Systems-based approaches to osteoporosis and fracture care: policy and research recommendations from the workgroups. Osteoporosis international: a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA 2011; 22 Suppl 3:495-500.
  137. Dell R, Greene D. Is osteoporosis disease management cost effective? Current osteoporosis reports. 2010;8:49–55. 
  138. Dell R. Fracture prevention in Kaiser Permanente Southern California. Osteoporos Int. 2011;22 Suppl 3:457–460. 
  139. Eisman JA, Bogoch ER, Dell R, Harrington JT, McKinney RE Jr, McLellan A, Mitchell PJ, Silverman S, Singleton R, Siris E. Prevention ATFoSF. Making the first fracture the last fracture: ASBMR task force report on secondary fracture prevention. J Bone Miner Res. 2012;27:2039–2046. 
  140. Glowacki J, Harris MB, Simon J, Wright J, Kolatkar NS, Thornhill TS, Leboff MS. Brigham fracture intervention team initiatives for hospital patients with hip fractures: a paradigm shift. Int J Endocrinol. 2010;2010:590751. 
  141. Glowacki J, LeBoff MS, Kolatkar NS, Thornhill TS, Harris MB. Importance of vitamin D in hospital-based fracture care pathways. J Nutr Health Aging. 2008;12:291–293. 
  142. Harris MB, LeBoff MS, Thornhill TS, Glowacki J. Evolution of comprehensive hospital care pathways to advance the treatment of fragility fractures. The Orthopaedic Journal at Harvard Medical School 2007:95-97.
  143. McDermott RS, Kloth DD, Wang H, Hudes GR, Langer CJ. Impact of zoledronic acid on renal function in patients with cancer: Clinical significance and development of a predictive model. J Support Oncol. 2006;4:524–529. 
  144. Oh WK, Proctor K, Nakabayashi M, Evan C, Tormey LK, Daskivich T, Antras L, Smith M, Neary MP, Duh MS. The risk of renal impairment in hormone-refractory prostate cancer patients with bone metastases treated with zoledronic acid. Cancer. 2007;109:1090–1096. 
  145. Reid IR, Brown JP, Burckhardt P, Horowitz Z, Richardson P, Trechsel U, Widmer A, Devogelaer JP, Kaufman JM, Jaeger P, Body JJ, Brandi ML, Broell J, Di Micco R, Genazzani AR, Felsenberg D, Happ J, Hooper MJ, Ittner J, Leb G, Mallmin H, Murray T, Ortolani S, Rubinacci A, Saaf M, Samsioe G, Verbruggen L, Meunier PJ. Intravenous zoledronic acid in postmenopausal women with low bone mineral density. N Engl J Med. 2002;346:653–661. 
  146. Whitaker M, Guo J, Kehoe T, Benson G. Bisphosphonates for Osteoporosis - Where Do We Go from Here? N Engl J Med. 2012 
  147. Black DM, Reid IR, Boonen S, Bucci-Rechtweg C, Cauley JA, Cosman F, Cummings SR, Hue TF, Lippuner K, Lakatos P, Leung PC, Man Z, Martinez RL, Tan M, Ruzycky ME, Su G, Eastell R. The effect of 3 versus 6 years of zoledronic acid treatment of osteoporosis: a randomized extension to the HORIZON-Pivotal Fracture Trial (PFT). J Bone Miner Res. 2012;27:243–254. 
  148. Cosman F, Cauley JA, Eastell R, Boonen S, Palermo L, Reid IR, Cummings SR, Black DM. Reassessment of fracture risk in women after 3 years of treatment with zoledronic acid: when is it reasonable to discontinue treatment? J Clin Endocrinol Metab. 2014;99:4546–4554. 
  149. Watts NB, Diab DL. Long-term use of bisphosphonates in osteoporosis. J Clin Endocrinol Metab. 2010;95:1555–1565. 
  150. Black DM, Bauer DC, Schwartz AV, Cummings SR, Rosen CJ. Continuing Bisphosphonate Treatment for Osteoporosis - For Whom and for How Long? N Engl J Med. 2012 
  151. Adler RA, El-Hajj Fuleihan G, Bauer DC, Camacho PM, Clarke BL, Clines GA, Compston JE, Drake MT, Edwards BJ, Favus MJ, Greenspan SL, McKinney R Jr, Pignolo RJ, Sellmeyer DE. Managing Osteoporosis in Patients on Long-Term Bisphosphonate Treatment: Report of a Task Force of the American Society for Bone and Mineral Research. J Bone Miner Res. 2016;31:16–35. 
  152. de Groen PC, Lubbe DF, Hirsch LJ, Daifotis A, Stephenson W, Freedholm D, Pryor-Tillotson S, Seleznick MJ, Pinkas H, Wang KK. Esophagitis associated with the use of alendronate. N Engl J Med. 1996;335:1016–1021. 
  153. Eriksen EF, Lyles KW, Colon-Emeric CS, Pieper CF, Magaziner JS, Adachi JD, Hyldstrup L, Recknor C, Nordsletten L, Lavecchia C, Hu H, Boonen S, Mesenbrink P. Antifracture efficacy and reduction of mortality in relation to timing of the first dose of zoledronic acid after hip fracture. J Bone Miner Res. 2009;24:1308–1313. 
  154. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Luthy R, Nguyen HQ, Wooden S, Bennett L, Boone T, Shimamoto G, DeRose M, Elliott R, Colombero A, Tan HL, Trail G, Sullivan J, Davy E, Bucay N, Renshaw-Gegg L, Hughes TM, Hill D, Pattison W, Campbell P, Sander S, Van G, Tarpley J, Derby P, Lee R, Boyle WJ. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997;89:309–319. 
  155. Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, Zoog HB, Austin M, Wang A, Kutilek S, Adami S, Zanchetta J, Libanati C, Siddhanti S, Christiansen C. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361:756–765. 
  156. von Keyserlingk C, Hopkins R, Anastasilakis A, Toulis K, Goeree R, Tarride JE, Xie F. Clinical efficacy and safety of denosumab in postmenopausal women with low bone mineral density and osteoporosis: a meta-analysis. Seminars in arthritis and rheumatism. 2011;41:178–186. 
  157. Rittmaster RS, Bolognese M, Ettinger MP, Hanley DA, Hodsman AB, Kendler DL, Rosen CJ. Enhancement of bone mass in osteoporotic women with parathyroid hormone followed by alendronate. J Clin Endocrinol Metab. 2000;85:2129–2134. 
  158. Rubin MR, Bilezikian JP. The anabolic effects of parathyroid hormone therapy. Clinics in geriatric medicine. 2003;19:415–432. 
  159. Tam CS, Heersche JN, Murray TM, Parsons JA. Parathyroid hormone stimulates the bone apposition rate independently of its resorptive action: differential effects of intermittent and continuous administration. Endocrinology. 1982;110:506–512. 
  160. Dempster DW, Cosman F, Parisien M, Shen V, Lindsay R. Anabolic actions of parathyroid hormone on bone. Endocr Rev. 1993;14:690–709. 
  161. Podbesek R, Edouard C, Meunier PJ, Parsons JA, Reeve J, Stevenson RW, Zanelli JM. Effects of two treatment regimes with synthetic human parathyroid hormone fragment on bone formation and the tissue balance of trabecular bone in greyhounds. Endocrinology. 1983;112:1000–1006. 
  162. Dobnig H, Turner RT. The effects of programmed administration of human parathyroid hormone fragment (1-34) on bone histomorphometry and serum chemistry in rats. Endocrinology. 1997;138:4607–4612. 
  163. Bilezikian JP. Combination anabolic and antiresorptive therapy for osteoporosis: opening the anabolic window. Current osteoporosis reports. 2008;6:24–30. 
  164. Jilka RL, Weinstein RS, Bellido T, Roberson P, Parfitt AM, Manolagas SC. Increased bone formation by prevention of osteoblast apoptosis with parathyroid hormone. J Clin Invest. 1999;104:439–446. 
  165. Rosen CJ, Bilezikian JP. Clinical review 123: Anabolic therapy for osteoporosis. J Clin Endocrinol Metab. 2001;86:957–964. 
  166. Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, Hodsman AB, Eriksen EF, Ish-Shalom S, Genant HK, Wang O, Mitlak BH. Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med. 2001;344:1434–1441. 
  167. Black DM, Greenspan SL, Ensrud KE, Palermo L, McGowan JA, Lang TF, Garnero P, Bouxsein ML, Bilezikian JP, Rosen CJ. The effects of parathyroid hormone and alendronate alone or in combination in postmenopausal osteoporosis. N Engl J Med. 2003;349:1207–1215. 
  168. Finkelstein JS, Hayes A, Hunzelman JL, Wyland JJ, Lee H, Neer RM. The effects of parathyroid hormone, alendronate, or both in men with osteoporosis. N Engl J Med. 2003;349:1216–1226. 
  169. Black DM, Bilezikian JP, Ensrud KE, Greenspan SL, Palermo L, Hue T, Lang TF, McGowan JA, Rosen CJ, Pa THSI. One year of alendronate after one year of parathyroid hormone (1-84) for osteoporosis. N Engl J Med. 2005;353:555–565. 
  170. Kurland ES, Heller SL, Diamond B, McMahon DJ, Cosman F, Bilezikian JP. The importance of bisphosphonate therapy in maintaining bone mass in men after therapy with teriparatide. Osteoporos Int. 2004;15:992–997. [human parathyroid hormone(1-34)]
  171. Tsai J, Uihlein A, Lee H, et al. Teriparatide and denosumab, alone or combined, in women with postmenopausal osteoporosis: the DATA study randomised trial. Lancet 2013 6;382(9886):50-6.
  172. Leder BZ, Tsai JN, Uihlein AV, Wallace PM, Lee H, Neer RM, Burnett-Bowie SA. Denosumab and teriparatide transitions in postmenopausal osteoporosis (the DATA-Switch study): extension of a randomised controlled trial. Lancet. 2015;386:1147–1155. 
  173. Muschitz C, Kocijan R, Fahrleitner-Pammer A, Lung S, Resch H. Antiresorptives overlapping ongoing teriparatide treatment result in additional increases in bone mineral density. J Bone Miner Res. 2013;28:196–205. 
  174. Cipriani C, Irani D, Bilezikian JP. Safety of osteoanabolic therapy: a decade of experience. J Bone Miner Res. 2012;27:2419–2428. 
  175. Andrews EB, Gilsenan AW, Midkiff K, Sherrill B, Wu Y, Mann BH, Masica D. The US postmarketing surveillance study of adult osteosarcoma and teriparatide: study design and findings from the first 7 years. J Bone Miner Res. 2012;27:2429–2437. 
  176. Tsuchie H, Miyakoshi N, Kasukawa Y, Nishi T, Abe H, Segawa T, Shimada Y. The effect of teriparatide to alleviate pain and to prevent vertebral collapse after fresh osteoporotic vertebral fracture. Journal of bone and mineral metabolism. 2016;34:86–91. 
  177. Michalska D, Luchavova M, Zikan V, Raska I Jr, Kubena AA, Stepan JJ. Effects of morning vs. evening teriparatide injection on bone mineral density and bone turnover markers in postmenopausal osteoporosis. Osteoporos Int. 2012;23:2885–2891. 
  178. Matsumoto T, Shiraki M, Hagino H, Iinuma H, Nakamura T. Daily nasal spray of hPTH(1-34) for 3 months increases bone mass in osteoporotic subjects: a pilot study. Osteoporos Int. 2006;17:1532–1538. 
  179. Levin G MC. Transdermaly-delivered PTH(1-34), a new treatment for osteoporotic patients: results of phase I studies. Journal of Bone and Mineral Research. 2007;22 Suppl 1:S324.
  180. Grossman JM, Gordon R, Ranganath VK, Deal C, Caplan L, Chen W, Curtis JR, Furst DE, McMahon M, Patkar NM, Volkmann E, Saag KG. American College of Rheumatology 2010 recommendations for the prevention and treatment of glucocorticoid-induced osteoporosis. Arthritis Care Res (Hoboken). 2010;62:1515–1526. 
  181. Saag KG, Shane E, Boonen S, Marin F, Donley DW, Taylor KA, Dalsky GP, Marcus R. Teriparatide or alendronate in glucocorticoid-induced osteoporosis. N Engl J Med. 2007;357:2028–2039. 
  182. Jha S, Wang Z, Laucis N, Bhattacharyya T. Trends in Media Reports, Oral Bisphosphonate Prescriptions, and Hip Fractures 1996-2012: An Ecological Analysis. J Bone Miner Res. 2015;30:2179–2187. 
  183. Khosla S, Shane E. A Crisis in the Treatment of Osteoporosis. J Bone Miner Res. 2016 
  184. Leder BZ, O'Dea LS, Zanchetta JR, Kumar P, Banks K, McKay K, Lyttle CR, Hattersley G. Effects of abaloparatide, a human parathyroid hormone-related peptide analog, on bone mineral density in postmenopausal women with osteoporosis. J Clin Endocrinol Metab. 2015;100:697–706. 
  185. Miller P, Leder BZ, Hattersley G, Lau E, Alexandersen P, Hala T, Mustatea S, Storgaard Nedergaard B, Krogsaa A, Slesinger J, Zerbini CA, Valter I, Visockiene Z, Jendrych B, Kulak CA, Marquez F, Harris AG, Williams GC, Hu M, Riis BJ, Russo L, Christiansen C. Effects of Abaloparatide on Vertebral and Non-Vertebral Fracture Incidence in Postmenopausal Women with Osteoporosis- Results of the Phase 3 Active Trial. Endocr Rev. 2015;(Apr):36.
  186. Jee WSS, Li X, Tian XY, Paszty C, Ke HZ. Anti-Sclerostin Antibody Increases Bone Mass by Stimulating Bone Formation and Inhibiting Bone Resorption in a Hindlimb-Immobilization Rat Model. Abstracts of the 30th Annual Meeting of the American Society for Bone and Mineral Research 2008:S 1138.
  187. Li X, Warmington K, Niu QT, Grisanti M, Tan H, Dwyer D, Stolina M, Simonet WS, Kostenuik PJ, Paszty C, Ke HZ. Increases in BMD Observed with Anti-Sclerostin Antibody Treatment Are Reversible: A Longitudinal Ovariectomized Rat Study. Abstracts of the 30th Annual Meeting of the American Society for Bone and Mineral Research 2008:S 1211.
  188. Recker RR, Benson CT, Matsumoto T, Bolognese MA, Robins DA, Alam J, Chiang AY, Hu L, Krege JH, Sowa H, Mitlak BH, Myers SL. A randomized, double-blind phase 2 clinical trial of blosozumab, a sclerostin antibody, in postmenopausal women with low bone mineral density. J Bone Miner Res. 2015;30:216–224. 
  189. McClung MR, Grauer A, Boonen S, Bolognese MA, Brown JP, Diez-Perez A, Langdahl BL, Reginster JY, Zanchetta JR, Wasserman SM, Katz L, Maddox J, Yang YC, Libanati C, Bone HG. Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med. 2014;370:412–420. 
  190. LeBoff MS, Chou SH, Ratliff KA, Cook NR, Khurana B, Kim E, Cawthon PM, Bauer DC, Black D, Gallagher JC, Lee I, Buring JE, Manson JE. Supplemental vitamin D and incident fractures in midlife and older adults. NEJM. 2022;387(4):299-309.
  191. LeBoff MS, Murata EM, Cook NR, Cawthon P, Chou SH, Kotler G, Bubes V, Buring JE, Manson JE. VITamin D and OmegA-3 TriaL (VITAL): Effects of Vitamin D Supplements on Risk of Falls in the US Population. J Clin Endocrinol Metab. 2020 Jun 3;. doi: 10.1210/clinem/dgaa311. [Epub ahead of print] PubMed PMID: 32492153.
  192. LeBoff MS, Chou SH, Murata EM, Donlon CM, Cook N, Mora S, Lee I, Kotler G, Bubes V, Buring JE, Manson JE. Effects of Supplemental Vitamin D on Bone Health Outcomes in Women and Men in the VITamin D and OmegA-3 TriaL (VITAL). J Bone Miner Res. 2020. PMID: 31923341.
  193. Reid IR, Horne AM, Mihov B, Stewart A, Garratt E, Wong S, Wiessing KR, Bolland MJ, Bastin S, Gamble GD. Fracture Prevention with Zoledronate in Older Women with Osteopenia. N Engl J Med. 2018;20:379(25):2407-2416. PMID: 30575489.
  194. Ruggiero SL, Dodson TB, Aghaloo T, Carlson ER, Ward BB, Kademani D. American Association of Oral and Maxillofacial Surgeons' Position Paper on Medication-Related Osteonecrosis of the Jaws-2022 Update. J Oral Maxillofac Surg. 2022;80(5):920-943.
  195. Bone HG, Wagman RB, Brandi ML, Brown JP, Chapurlat R, Cummings SR, Czerwiński E, Fahrleitner-Pammer A, Kendler DL, Lippuner K, Reginster JY, Roux C, Malouf J, Bradley MN, Daizadeh NS, Wang A, Dakin P, Pannacciulli N, Dempster DW, Papapoulos S. 10 years of denosumab treatment in postmenopausal women with osteoporosis: results from the phase 3 randomised FREEDOM trial and open-label extension. Lancet Diabetes Endocrinol. 2017 Jul;5(7):513-523. doi: 10.1016/S2213-8587(17)30138-9. Epub 2017 May 22. PMID: 28546097.
  196. Cummings SR, Ferrari S, Eastell R, Gilchrist N, Jensen JB, McClung M, Roux C, Törring O, Valter I, Wang AT, Brown JP. Vertebral Fractures After Discontinuation of Denosumab: A Post Hoc Analysis of the Randomized Placebo-Controlled FREEDOM Trial and Its Extension. J Bone Miner Res. 2018;33(2):190-198.
  197. Shoback D, Rosen CJ, Black DM, Cheung AM, Murad MH, Eastell R. Pharmacological Management of Osteoporosis in Postmenopausal Women: An Endocrine Society Guideline Update. J Clin Endocrinol Metab. 2020;105(3)

202   Miller PD, Hattersley G, Riis BJ, Williams GC, Lau E, Russo LA, Alexandersen P, Zerbini CA, Hu MY, Harris AG, Fitzpatrick LA, Cosman F, Christiansen C; ACTIVE Study Investigators. Effect of Abaloparatide vs Placebo on New Vertebral Fractures in Postmenopausal Women With Osteoporosis: A Randomized Clinical Trial. JAMA. 2016 Aug 16;316(7):722-33. doi: 10.1001/jama.2016.11136. Erratum in: JAMA. 2017 Jan 24;317(4):442. PMID: 27533157.

  1. Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, Hofbauer LC, Lau E, Lewiecki EM, Miyauchi A, Zerbini CA, Milmont CE, Chen L, Maddox J, Meisner PD, Libanati C, Grauer A. Romosozumab Treatment in Postmenopausal Women with Osteoporosis. N Engl J Med. 2016 Oct 20;375(16):1532-1543. doi: 10.1056/NEJMoa1607948. Epub 2016 Sep 18. PMID: 27641143.
  2. Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T, Maddox J, Fan M, Meisner PD, Grauer A. Romosozumab or Alendronate for Fracture Prevention in Women with Osteoporosis. N Engl J Med. 2017 Oct 12;377(15):1417-1427. doi: 10.1056/NEJMoa1708322. Epub 2017 Sep 11. PMID: 28892457.
  3. Camacho PM, Petak SM, Binkley N, Diab DL, Eldeiry LS, Farooki A, Harris ST, Hurley DL, Kelly J, Lewiecki EM, Pessah-Pollack R, McClung M, Wimalawansa SJ, Watts NB.. American Association of Clinical Endocrinologists/American College of Endocrinology clinical practice guidelines for the diagnosis and treatment of postmenopausal osteoporosis—2020 update. Endocr Pract. 2020;26(suppl 1):1-46

 

 

The Role of Exercise in Diabetes

ABSTRACT

 

Exercise is a key component to lifestyle therapy for prevention and treatment of type 2 diabetes (T2D). These recommendations are based on positive associations between physical activity and T2D prevention, treatment, and disease-associated morbidity and mortality. For type 1 diabetes (T1D), we have evidence to support that exercise can reduce diabetes associated complications. However, there are physiological and behavioral barriers to exercise that people with both T2D and T1D must overcome to achieve these benefits. Physiological barriers include diabetes-mediated impairment in functional exercise capacity, increased rates of perceived exertion with lower workloads, and decision making regarding glycemic management. There are additional social and psychological stressors including depression and reduced self-efficacy. Interestingly, there is variability in the response to exercise by sex, genetics, and environment, further complicating the expectations for individual benefit from physical activity. Defining optimal dose, duration, timing, and type of exercise is still uncertain for individual health benefits of physical activity. In this review, we will discuss the preventative value of exercise for T2D development, the therapeutic impact of exercise on diabetes metabolic and cardiovascular outcomes, the barriers to exercise including hypoglycemia, and the impact of sex and gender on cardiorespiratory fitness and adaptive training response in people with and without diabetes. There are still many unknowns regarding the diabetes-mediated impairment in cardiorespiratory fitness, the variability and individual response to exercise training, and the impact of sex and gender. However, there is no debate that exercise provides a health benefit for people with and at risk for diabetes.

 

INTRODUCTION

 

Exercise, together with medical nutrition therapy, forms the cornerstone of diabetes therapy. In their 2022 Standards of Medical Care in Diabetes, The American Diabetes Association (ADA) recommends that adults with diabetes participate in both aerobic activity and resistance training. They specify that this should entail at least 150 minutes of moderate-to-vigorous aerobic activity per week, spread over at least three days per week to minimize consecutive days without activity, and two to three sessions of resistance exercise per week on nonconsecutive days (1). Regular exercise is associated with prevention and minimization of weight gain, reduction in blood pressure, improvement in insulin sensitivity and glucose control, and optimization of lipoprotein profile, all of which are independent risk factors for the development of T2D (2,3). Meeting physical activity guidelines has been associated with a 40% decrease in cardiovascular mortality with an even greater impact on all-cause mortality (3,4). This association is especially significant given that people with T1D and T2D have a two to six-fold increase in morbidity and premature mortality from clinical cardiovascular disease (CVD) (5).

 

Despite these positive links, 34.3% of Americans diagnosed with diabetes are categorized as physically inactive (<10 minutes per week of moderate or vigorous physical activity) and 23.8% are meeting the 150-minute segment of physical activity guidelines (6). A worldwide pooled analysis of data from 358 surveys across 168 countries showed that the global age standardized prevalence of insufficient physical activity was 27.5% in 2016. The highest levels of insufficient activity were in women in Latin America and the Caribbean (43.7%), South Asia (43.0%) and high-income Western countries (42.3%) and the lowest levels were in men in Oceania (12.3%) (7). It is important for health care providers to understand that diabetes can lead to significant physiological barriers to exercise. These barriers include impaired maximal and submaximal exercise capacity (8,9), social and psychological barriers to exercise in T2D (10,11), the direct stress on the cardiovascular system caused by exercise, and the risk of hypoglycemia (12). Additionally, exercise studies have shown individual variation in response to physical activity, suggesting that there may be some individuals who are “non-responders” to exercise, in that they do not reap the specific anticipated benefits of exercise therapy such as improved glucose, blood pressure, or lipid profiles. This variation in “response” may be due to the modality employed (aerobic vs resistance exercise), the adaptive response to timing of intervention, and the endpoint examined (13). For example, someone with diabetes may respond with increased fitness but experience no change in glucose. There are also sex differences in cardiorespiratory fitness (CRF), discussed in more detail below (14). These findings speak to the complexity of the pathophysiology involved in exercise and the impact that diabetes has on these processes (Figure 1).

Figure 1. Cardiorespiratory fitness and Premature Mortality. CRF is a systems biology measure of the physiological response to a workload. Exercise requires cardiac, vascular, and skeletal muscle integration. Impairment is this integration is a risk for cardiovascular and all-cause mortality. Evidence supports a model wherein multiple modest functional derangements contribute to impaired CRF in uncomplicated type 2 diabetes.

 

In this chapter, we will discuss the relationship between exercise physiology and diabetes pathophysiology via an overview of the literature demonstrating the associations between exercise and preventative effects for diabetes, therapeutic value for established diabetes, and prognostic value for development of diabetic complications. We will discuss physiological and behavioral barriers that contribute to lack of achievement of physical activity guidelines including hypoglycemia and the impaired exercise capacity that diabetes itself can cause. We will conclude with a discussion on sex differences in exercise in diabetes.

 

THE VALUE OF EXERCISE IN DIABETES PREVENTION

 

Exercise is an established strategy for T2D prevention (3). The incidence of T2D is inversely proportional to participation in physical activity. In a systematic review by Warburton et al that analyzed 20 cohort studies, all were noted to show this inverse relationship with T2D incidence; additionally, when comparing the most active participants to the least active participants, they calculated the average risk reduction of the exercise intervention to be 42%. Within these studies, 84% showed a dose-response relationship to suggest that even small changes in physical activity level led to great reductions in T2D incidence (15).  Manson et al demonstrated that women who reported at least weekly vigorous exercise had a 16% reduced risk of developing T2D, when controlled for age and body mass index (16). In Hu et al’s analysis of the nurses’ Health Study, there was a 34% reduction in diabetes incidence for each hour per day of brisk walking (17). Furthermore, among high-risk women with a history of gestational diabetes, physical activity has been shown to be inversely associated with the incidence of type 2 diabetes in a dose-dependent manner (18).

 

Physical activity is also a modifiable risk factor that influences CRF; there is a strong association between CRF and incidence of T2D. In the Henry Ford Exercise Testing Project, people who achieved >= 12 metabolic equivalents (METs) had a 54% lower risk of incident diabetes compared to people achieving <6 METs (controlled for age, sex, race, obesity, hypertension, and hyperlipidemia) (19). In a study of middle-aged men by Lynch et al, men with CRF levels greater than 31.0 mL of oxygen per kilogram per minute who exercised at moderate intensity (>5.5 METs) for >40 minutes per week had a decreased incidence of diabetes. This effect was seen even within a subgroup of men at high risk for diabetes (overweight or hypertensive with positive parental history); engagement in this level of moderate intensity exercise in this group reduced their risk of diabetes by 64% compared to men who did not engage in physical activity (20). For reference, 1 MET is equivalent to the amount of oxygen consumed while sitting at rest, which is 3.5 ml/O2/kg/min (21) and expending 2 METs means that an individual is exerting 2 times the energy than they would be while sitting still. Examples of common activities and their associated energy costs in METs are shown in Table 1 (21, 22).

 

Table 1. Metabolic Equivalents (METs) Expended for Common Activities

Activity

METs

Slow Walking (3 kilometers/hour)

3

Walking up stairs

4.7

Brisk Walking (6 kilometers/hour)

5.4

Bicycling (20 kilometers/hour)

7.1

Running (8 kilometers/hour)

8.2

Hockey

12.9

Boxing

13.4

 

 

CRF can be measured in a few different ways. The gold standard includes gas analysis and is reported as maximal oxygen uptake (VO2max) or peak oxygen uptake (VO2peak) (23). This can be impractical in a clinical setting, so several walk tests have been developed to estimate CRF that either measures how much distance a person can cover within the designated time frame or how long it takes them to cover a designated distance. The 6-minute walk test is used in at-risk populations (23) and the 400-meter walk test is often used in older adults (24). 

 

At a practical level, it is useful to ask individuals a few questions about their ability to climb stairs, any changes in their ability to walk a given distance, and if they’ve experienced any changes in perceived exertion or shortness of breath with activity.

 

Weight loss is important for prevention of T2D (25). Analysis of people in the intensive lifestyle intervention arm of the Diabetes Prevention Program (DPP) Intensive Lifestyle indicated that there was a 16% reduction in diabetes risk per kilogram of weight loss (26). Theoretically, an increase in physical activity can lead to negative energy balance, which may result in weight loss if diet is unchanged. A study by Ross et al analyzed the effect of exercise-induced weight loss via a 500-700 kcal/day deficit during a 12-week intervention and showed an average weight loss of 7.6kg (8% initial body weight). Their findings also showed that exercise-induced weight loss decreases total fat percentage with increases in cardiovascular fitness to a greater degree than similar diet-induced weight loss (27). This degree of weight loss is uncommon in exercise interventional studies without simultaneous calorie restriction, so diet and exercise interventions should be administered simultaneously for maximal benefit (25). At the same time, there is a dynamic relationship between exercise dose, weight status, and diabetes incidence, wherein each of these components affects the other (3). To assess the complex association between obesity and physical inactivity for interaction, Quin et al conducted a systematic review that showed positive biological interaction on an additive scale (28). This interaction was further shown in a meta-analysis of 9 prospective cohort studies by Cloostermans et al, where there was a 7.4-fold increased risk of T2D in those who were obese and with a low physical activity level when compared to normal weight, highly active individuals (29).

 

Exercise aids with diabetes prevention even if weight loss is not achieved. There is a strong association between increased physical activity and prevention of weight gain (3). In DPP, those who achieved 150 minutes of moderate intensity activity per week had a 46% reduction in diabetes incidence, despite not always meeting weight loss goals (21). This effect was similarly seen in other international studies (Sweden (30), Finland (31), China (32), Japan (33), India (34)) when intensive lifestyle intervention was used for prevention of diabetes. The effect of exercise alone was specifically evaluated in the Chinese study where there was a reduction in incidence of diabetes by 33% in the diet-only group, 47% in the exercise-only group, and 38% in the diet-plus-exercise group; this effect was seen even when adjusting for interaction of BMI (31%, 46%, and 42% for diet, exercise, and diet-plus-exercise groups, respectively) (32). Additionally, Dai et al looked further into the efficacy of the type of exercise on prevention of diabetes. They randomized patients with prediabetes into 3 intervention groups of aerobic training (AT), resistance training (RT), and combined training (AT + RT). After 2 years of intervention, the T2D incidence was reduced by 74% in the AT + RT group, 65% in the RT alone group, and 72% in the AT alone group compared to controls. There was no significant difference in 2-hour glucose tolerance tests between intervention groups, providing support for both AT and RT, alone or in combination, benefiting T2D prevention (35).

 

Physical activity can also lead to improvement in cardiovascular risk factors. With regards to hypertension, there is an inverse relationship between blood pressure and physical activity level, with greater responses noted in those with hypertension/pre-hypertension compared to individuals with normal blood pressure (3). In the DPP, participants who received intensive lifestyle intervention had improved cardiovascular disease risk factor profiles (decreased blood pressure, LDL cholesterol, and triglyceride levels) compared to the metformin treated and placebo groups after 5 years; this improvement was achieved with a decreased need for lipid and blood pressure medication initiation (36). Additionally, while the LOOK AHEAD trial in overweight or obese adults with T2D was negative for its primary cardiovascular outcome (37), further analysis showed that increasing fitness had a beneficial effect on fasting blood glucose, HbA1c, and other cardiovascular risk factors (HDL, triglycerides, and diastolic blood pressure), and cognition beyond the effect of weight change (38).

 

There is significant variability in changes to CRF with exercise therapy; not all individuals respond positively to exercise intervention. CRF is not always related to physical activity and is determined by genetics and other factors. In the HERITAGE Family Study, maximal oxygen uptake (VO2max, a measurement of CRF) response to exercise therapy varied significantly with some participants showing no improvement with exercise training and others exhibiting maximal improvement (>1L/min). Interestingly, there was 2.5 times more variance between families than within families, suggestive of a possible genetic component to exercise response (39). These individuals with little to no improvement with exercise are termed “non-responders.” In cross-over interventional studies that assessed poor responsiveness to aerobic exercise and resistance training, it was found that those who did not benefit from aerobic training, improved their CRF with resistance training. Alternatively, not all individuals who improved CRF with aerobic training had improvements with resistance training. This finding suggests that “non-responsiveness” may be related to exercise modality and that incidence of non-responsiveness to exercise for the endpoint of CRF may be resolved by changing the mode of training (40,41). All in all, to achieve the desired benefits of exercise (improvement in weight, glucose control, endurance, etc.), an individualized approach is key. One gap in practice is a lack of a commonly employed clinical measure of response to an exercise intervention. There is a need for exercise physiology expertise or provider comfort with exercise as a therapeutic tool to tailor and adjust sustained exercise interventions and employ exercise as medicine.

 

THERAPEUTIC VALUE OF EXERCISE IN DIABETES MANAGEMENT

 

Diet and exercise (lifestyle modification) are considered by all diabetes clinical guidelines to be the foundation for diabetes management. Exercise can augment glucose disposal and improve insulin action, and thus can be a tool to aid in glucose regulation. Muscle contraction and contraction-mediated skeletal muscle blood flow leads to glucose uptake via insulin-dependent and independent mechanisms. Exercise-mediated glucose disposal can decrease circulating blood glucose but may be affected by other determinants of systemic glucose metabolism. The components of glucose disposal need to be considered to better understand the impact of exercise on glucose clearance. Glucose transporter 4 (GLUT4) translocation is acutely stimulated by muscle contraction, increasing facilitated transport of glucose into the muscle. In addition, contraction augments skeletal muscle blood flow and thereby increases the rate of glucose dispersion into the muscle interstitial space (42). Insulin also recruits GLUT4 to the muscle surface. Muscle glycogen stores and exogenous glucose are consumed during exercise leading to a glucose/glucose-6-phosphate gradient that favors additional glucose entry into the skeletal muscle. Based on these factors and other molecular changes in skeletal muscle signaling, exercise can impact glucose homeostasis for up to 48 hours (43).

 

Exercise training increases skeletal muscle GLUT4 expression and augments insulin receptor signaling and oxidative capacity which optimizes insulin action and glucose oxidation and storage (44). Therefore, routine moderate exercise usually improves sensitivity to insulin in individuals with T2D (45). This exercise effect is impacted by exercise type (aerobic versus resistance), dose, duration, and intensity of activity. For example, the energy expended per week, is a product of frequency, intensity, and duration of exercise and correlates with changes in insulin sensitivity (46,47). There is also an impact of each bout of exercise. Newsom et al found that low intensity activity (50% VO2peak) improved insulin sensitivity for ~19 hours after exercise in obese adults (48). These findings support the recommendation that people with T2D should engage in daily exercise, with no more than 2 days elapsing between episodes of physical activity; consistency is key and even small amounts of exercise are beneficial (49).

 

The modality of exercise to induce maximal intended benefit in individuals with T2D is not as clear. Physical activity guidelines for Americans suggest a mixture of resistance and aerobic activity based on limited prospective studies in this population (50,51). Studies vary by intervention structure and duration and in most cases specific exercise interventions have not been compared head-to-head. In one randomized control trial of sedentary individuals with T2D, a combination of aerobic and resistance training for 9 months significantly lowered HbA1c levels compared to a non-exercise control group (50). Similarly, high intensity interval training (HIIT) session (10 minutes of intense exercise) reduces postprandial hyperglycemia in patients with T2D, suggesting that it can be a time efficient way to achieve benefits of exercise training (52). At the same time, any type of exercise is beneficial. Individuals with T2D who engage in exercise have a decrease in HbA1c by 0.67%, regardless of type of exercise (structured aerobic, resistance, or combined exercise training) (53). Therefore, the best therapy is one that an individual can and will maintain.

 

In patients with T1D, available evidence is mixed for whether exercise improves overall glycemic control, but it has been shown to have multiple benefits (54). Supervised exercise programs increase fitness in patients with T1D and inone study, VO2max increased by 27% after 4 months of participation in a bicycle exercise training program (55,56). Insulin requirements have also been shown to be reduced with exercise training in patients with T1D, with anywhere from a 6% to 18% daily insulin dose reduction across multiple studies (56–58). In the Pittsburgh Insulin-dependent Diabetes Mellitus Morbidity and Mortality Study, activity level was inversely related to mortality risk and men who were sedentary were 3 times more likely to die than active males. A similar but nonsignificant trend was seen in women (59).

 

Regular exercise provides a physiological stress to the body and can generate adaptations such as induction of antioxidant defense mechanisms. Low exposure to a toxic or stress environment leads to positive biological responses, hormesis, whereas high exposure leads to negative responses (U-shaped dose response effect). Exercise induces low amounts of reactive oxygen species (ROS) acutely, which positively stimulates oxidative damage-repairing enzyme activity and results in improved biological fitness (60). For example, in the context of exercise, ROS formation can stimulate nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor that is dormant in the cytoplasm. Low levels of oxidative stress stimulate Nrf2 translocation to the nucleus to stimulate expression of antioxidant enzymes; when Nrf2 activity is diminished, as in endothelial dysfunction, insulin resistance and abnormal angiogenesis is seen, such as in individuals with T2D (61). This is one example of the molecular response to exercise. Many such examples exist and demonstrate similarly positive profiles: reduction in inflammatory markers (c-reactive protein, interleukin-6, and tumor necrosis factor-α) and upregulation of anti-inflammatory substances (interleukin-4 and interleukin 10) (62). Ristow et al showed that exercise mediated ROS are integral to the process by which exercise improves insulin sensitivity (as measured by glucose infusion rates during a hyperinsulinemic, euglycemic clamp and plasma adiponectin) (63). In their study, exercised muscles of previously untrained individuals showed a two-fold increase in oxidative stress (as measured by thiobarbituric acid-reactive substances [TBARS]). However, daily intake of antioxidant dietary supplementation (vitamin C and E) blunted this affect by blocking this initial step of transient increase of oxidative stress. Exercise mediated ROS induced expression of molecular regulators (PPARgand its coactivators PGC1a and PGC1b,) that coordinate insulin-sensitizing gene expression. Those treated with vitamin C and E had decreased expression of these molecular regulators. Consequently, non-supplemented individuals without diabetes had significant improvement in insulin sensitivity while those on antioxidant supplements had no change in insulin sensitivity. The NIH Molecular Transducers of Exercise (MoTrPAC) program will examine the molecular response to exercise in healthy people and rodent models to set the stage for more detailed assessments of these endpoints in disease states such as diabetes (64).

 

While lifestyle intervention through diet and exercise are the initial step in T2D treatment, pharmacologic therapy may also be needed to achieve glycemic targets for a person with T2D. Regardless, at each step of intensification of medical therapy for glucose or blood pressure lowering, exercise should be reinforced as an important part of treatment. Combination therapy with metformin monotherapy plus post-meal exercise, led to a 21% reduction in postprandial hyperglycemia, a comparable effect to that of sulfonylureas (-14%), thiazolidinediones (-20%), and dipeptidyl peptidase-4inhibitors (-23%) (65). At the same time, there is some evidence to suggest that metformin may attenuate the positive effects of exercise on insulin sensitivity and inflammation (66,67). Of note, these studies were performed in people with insulin resistance or increased risk of T2D and not in people with diabetes. Incorporation of exercise and diet into all diabetes management strategies is crucial for cardiometabolic health.

 

IMPACT OF EXERCISE ON DIABETES OUTCOMES

 

Beyond the therapeutic and preventative benefits of exercise discussed in previous sections, exercise also holds great prognostic value for people with diabetes. Observational studies have shown an inverse linear dose-response relationship between physical activity amount and mortality (68). Exercise capacity has been shown to be predictive of mortality in people with diabetes (69), echoing findings in the general population (70). Furthermore, decreased exercise capacity in people with T2D is associated with development of future cardiovascular events (71).

 

Additionally, associations between higher levels of physical activity and reduced complications in diabetes have been noted. Gulsin et al were able to show that exercise improved diastolic function in adults with T2D whereas weight loss via a low-energy diet alone did not improve diastolic function despite the diet leading to weight loss, improved glycemic control, and improved aortic stiffness and concentric LV remodeling (72). A meta-analysis on 18 studies of patients T1D and T2D showed that physical activity also increased glomerular filtration rate and decreased the urinary albumin creatinine ratio (73). In the Finish Diabetic Nephropathy (FinnDiane) Study, low levels of self-reported leisure-time physical activity in people with T1D was associated with a greater degree of renal dysfunction, proteinuria, CVD, and retinopathy (74) and Kriska et al found that men with insulin-dependent diabetes who reported higher levels of physical activity in their past had lower prevalence of nephropathy and neuropathy (75). Bohn et al also found an inverse relationship between physical activity level and both retinopathy and microalbuminuria in people with T1D in the Diabetes-Patienten-Verlaufsdokumentation (DPV) database (76). Interestingly, a large cohort study of adults with T1D and T2D in Australia found that physical activity was protective against developing advanced diabetic retinopathy requiring retinal photocoagulation (however this finding was only significant for men) (77).

 

EXERCISE INTOLERANCE AS A BARRIER TO EXERCISE ADHERENCE IN DIABETES

 

Exercise holds great promise as a preventative and therapeutic intervention for people with diabetes. Yet, diabetes presents significant physiological, psychological, and socioeconomic barriers to physical activity. Despite these barriers, exercise remains a cornerstone of treatment for diabetes, and as such, it is useful to understand the barriers to exercise in diabetes and consider strategies for overcoming them (Table 2).

 

People with T2D are disproportionately sedentary and overweight (78) and report more physical discomfort during exercise (10). Excess weight itself can be a physical barrier to increased activity; in a study of obese subjects with diabetes, those who reported physical discomfort as a barrier to exercise had a significantly higher body mass index compared to those individuals who did not report it (36 vs 34, respectively, p=0.021) (79). A decreased level of fitness also contributes to this barrier of discomfort with physical activity. Functional exercise capacity (FEC), measured by VO2max, is impaired in both youth and adults with uncomplicated T1D and T2D (8,69). Insulin sensitivity has a direct association with VO2peak (80,81). Studies by Reusch, Regensteiner, and colleagues have demonstrated that adolescents and adults with uncomplicated T2D have reduced CRF compared to those without T2D. These findings persist in the absence of clinical cardiovascular disease and when matched by baseline exercise status and weight (82-84).

 

CRF is an outcome determined by various measures of cardiac and skeletal muscle function. Reductions in CRF are associated with reduced cardiac performance (85,86). Women recently diagnosed with T2D have been shown to have significantly increased pulmonary capillary wedge pressure and abnormal diastolic parameters during exercise compared to healthy control subjects, a finding concerning for subclinical diastolic dysfunction (14,87). Additionally, adolescents with T2D have been shown to have abnormal cardiac circumferential strain (CS), increased indexed LV mass, and decreased CRF compared to obese and lean healthy controls. In this study of youth with T2D, fat mass and low adiponectin correlated with CS and CRF. These associations suggest a role for obesity in cardiac impairment and CRF in T2D (88). In skeletal muscle, Reusch, Regensteiner and colleagues have reported a mismatch between skeletal muscle oxygen extraction, oxidative flux, and VO2peak in individuals with T2D (89,90). Additionally, studies have shown evidence of degradation of the vascular endothelial glycocalyx in individuals with T2D (91). These changes at the muscular level are thought to cause impaired microvascular perfusion, which likely ultimately contributes to decreased CRF in these individuals (92,93). Consistent with a relationship between microvascular dysfunction and fitness, people with diabetes who have developed microvascular complications (retinopathy, neuropathy, nephropathy with microalbuminuria) have decreased CRF compared to those without these complications (94). Fortunately, certain types of exercise can resolve the T2D associated impairment of skeletal muscle in vivomitochondrial oxidative flux. Scalzo et al showed that single-leg exercise training for 2 weeks increased in vivooxidative flux in participants with T2D but not in matched controls without T2D (95).

 

In addition to these cardiovascular contributions to impaired exercise function in diabetes, mitochondrial capacity is impaired (96), and mitochondrial content is reduced (97). Observations of an association between insulin sensitivity and exercise capacity (81) may also reflect additional metabolic determinants of exercise impairment beyond impaired muscle perfusion and reduced mitochondrial function. As a proof of concept, the PPARg  insulin sensitizer rosiglitazone has been shown to improve exercise capacity and insulin sensitivity in T2D in a three-month intervention (despite weight gain) (98,99). Improved CRF correlated with an improvement in endothelial function and blood flow (98). In contrast, in men with established coronary artery disease and T2D, a year-long-treatment with rosiglitazone lead to a decrease in CRF related to increased weight and subcutaneous fat mass expansion. Our current interpretation is that insulin action is a modifiable target for augmenting CRF but that currently available insulin sensitizers are not a durable intervention (100).

 

Exercise can be a cardiovascular stressor, and while chronic exercise is associated with a reduction in cardiovascular risk (101), acute exercise may precipitate events in susceptible individuals (102). Thus, in people at high risk for acute cardiovascular events, some caution is warranted in initiating a new exercise regimen. Low intensity exercise with high consistency may be a safer and more effective strategy than more sporadic, high intensity exercise. A cardiac rehabilitation approach is a great consideration, but not often covered by insurance. Discussion with a provider for people with diabetes prior to initiating an exercise program is recommended by the American College of Sports Medicine, especially if they are currently sedentary or have chronic complications from their diabetes (103). This recommendation is echoed but less formal in the ADA guidelines. In the opinion of these authors, people with diabetes should be encouraged to exercise and to build up to an exercise program. Providers should discuss anginal equivalents, and significant changes in exercise tolerance (for example, change in the distance a person can walk, or fewer flights of stairs) or shortness of breath with exercise as an indication for concern. Since exercise should be a vital sign, these discussions should happen with each clinical encounter. 

 

Additionally, presence of diabetes complications can be a barrier to exercise (74). There is a high association between diabetes complications and depression (104), which can reduce the desire to perform any activity. Decreased kidney function, such as that seen in diabetic nephropathy, is associated with a higher prevalence of anemia (105) which can make it difficult to exercise due to decreased oxygen delivery. Additionally, diabetic retinopathy with decreased vision, diabetic neuropathy with loss of balance, and diabetic foot ulcers can all pose physical limitations to exercise (106). Weight bearing exercise can increase foot trauma. Therefore, it is important for people with T2D to conduct frequent foot examinations when participating in physical activity. Contact footwear use can reduce rate of foot-related injury (107,108). However, these special considerations can lead to decreased incentive and increased distress when engaging in physical activity.

 

As may be expected, motivating people with diabetes to exercise regularly is often a considerable challenge in both T1D and T2D. Engaging people with diabetes to exercise generally requires changing ingrained lifestyle habits. Habitual and social barriers to exercise also add to the motivational difficulties of lifestyle-based interventions. Finally, barriers to exercise in T2D may be confounded by socioeconomic class. People with T2D tend to have lower socioeconomic status (109), which is itself associated with less physical activity (110). There is also increased concern for safety in low socioeconomic neighborhoods. Overcoming this array of physiological, psychological, and socioeconomic barriers to regular exercise in people with diabetes requires a nuanced, patient-specific approach. Strategies for motivating patients to engage in regular physical exercise include motivational interviewing (111), community-based interventions (112), group exercise, and surveillance using activity-tracking devices such as pedometers (113). Each of these strategies has been shown to achieve at least modest success, but the increasing prevalence and costs of T2D (114,115) indicate that more work is needed. 

EXERCISE INDUCED HYPOGLYCEMIA

 

Exercise can be acutely dangerous for people with diabetes who are on certain glucose lowering medications, such as insulin and sulfonylureas medications, as exercise can increase the risk of hypoglycemia in these patients. Hypoglycemia and fear of hypoglycemia with exercise represent real and major considerations for people with diabetes. These considerations are especially relevant to people with T1D, as episodes of severe (and particularly nocturnal) hypoglycemia are associated with large increases in mortality (116), and exercise can precipitate nocturnal hypoglycemia and impaired counterregulatory responses in people with T1D (117,118). This is also a risk, albeit to a lesser extent, for people with T2D on insulin or sulfonylureas (119). Exercise increases both the translocation and expression of GLUT4 (120), thus potentiating the effects of insulin, and greatly increases the metabolic demand for glucose (121). These factors predispose towards hypoglycemia. Exercise can impact glucose homeostasis for up to 48 hours (43). Fear of hypoglycemia is the primary barrier to exercise in people with T1D (12).

 

Different exercise modalities can cause varied effects on blood glucose in the acute setting. We will discuss simplified differences during a bout of moderate vs vigorous physical activity in the setting of a healthy individual (Figure 2) to contextualize the discussion that follows. The uptake of blood glucose by skeletal muscle increases with increasing intensity and duration of physical activity. With moderate activity, the fall in plasma glucose from muscle glucose uptake is coordinated with a fall in plasma insulin and increase in counterregulatory hormones, particularly glucagon, that help mobilize glucose (122). With vigorous activity, the distinction is that there is an exercise stimulated surge of counterregulatory hormones, independent of plasma glucose level, and this can stimulate an acute increase in plasma glucose (123). People with diabetes who are treated with insulin lose the ability to physiologically decrease circulating insulin with exercise and can have an impaired ability to augment secretion of glucagon, cortisol, growth hormone and catecholamines with exercise; factors that particularly predispose them to hypoglycemia. Post bout, muscle glycogen depletion from physical activity will lead to increased skeletal muscle glucose uptake for glycogen repletion and this increased insulin-independent glucose clearance contributes to a decrease in plasma glucose (124) (Figure 3).

Figure 2. Glucose homeostasis during a bout of moderate vs. vigorous physical activity.

Figure 3. Glucose homeostasis following a bout of physical activity.

 

In the literature, aerobic and resistance exercise are often compared as activities that have differing effects on hypoglycemia. The aerobic exercise regimens specified in the studies presented here are of moderate intensity and can be conceptualized as a moderate bout of physical activity and the resistance exercise regimens can be conceptualized as a vigorous bout. Yardley et al showed that resistance exercise tends to cause an acute increase in blood glucose superimposed with a subsequent increase in insulin sensitivity, whereas aerobic exercise causes a larger initial decrease in blood glucose but somewhat less sustained hypoglycemic effect. However, resistance exercise was associated with overall less blood glucose variability post-exercise (125). Additionally, a HIIT session is less likely to cause hypoglycemia compared to moderate-intensity aerobic exercise (126). There is also evidence that performing resistance exercise prior to aerobic exercise can also lead to decreased glucose variability during exercise and attenuate post-exercise hypoglycemia (127). The optimal duration, intensity, and order of specific types of physical activities to prevent hypoglycemia in patients with T1D is the subject of continued research. Steineck et al found that the time patients with T1D spent in hypoglycemia over a 5-day period was similar if they exercised 5 consecutive days, consisting of 4 minutes of resistance training followed by 30 minutes of aerobic training per session, or if they exercised 2 days in this 5-day period and performed 10 minutes of resistance training followed by 75 minutes of aerobic exercise each session (128). Much like all aspects of diabetes management, the way an individual responds to exercise can be anticipated based on the literature, however, each individual will need to measure their blood glucose pre- and post- exercise for 4-24 hours post bout to understand their needs. Other factors such as sleep, stress, general physical fitness, and prior exercise training can all impact the glucose response to an exercise bout.

 

Beyond the features of a session of exercise, the cornerstone of mitigating the risk of exercise induced hypoglycemia in patients who are on multiple daily injections of insulin or insulin pumps without hybrid closed loop features, includes insulin dose reduction and consumption of carbohydrates. Consensus recommendations consist of complex and personalized algorithms, but some generalizations are to reduce pre-exercise meal bolus within 90 minutes before aerobic exercise by 30-50% and to consume 30-60gm of high glycemic index carbohydrates per hour of sport. Post-exercise recommendations are especially important for afternoon and evening exercise as nocturnal hypoglycemia occurs commonly in individuals with T1D and this risk is increased with exercise that is done later in the day. Some recommendations are to decrease the bolus for the meal after exercise by 50% and reduce basal rate by 20% for 6 hours at bedtime if exercise occurred in the afternoon (129).

 

Hybrid closed loop (HCL) systems are becoming more widely available and used in practice. They require clinicians to modify recommendations for exercise to account for the principles that affect a specific system’s automated insulin delivery algorithms. One clear advantage of HCL systems in this context is that they have a predictive low glucose suspend feature that suspends insulin delivery when a low glucose is predicted in the next 30 minutes (130). An adage that does need to be re-examined for HCL is one described in the previous paragraph wherein patients may eat uncovered carbohydrate snacks or partially covered meals prior to exercise. In HCL systems, the rise in glucose from eating uncovered carbohydrates prior to exercise can lead to an increase in automated insulin delivery (130) and in our clinical experience, extra insulin on board can then sometimes precipitate hypoglycemia with exercise. More research is needed in this arena. One main strategy that is agreed upon to use for hypoglycemia prevention with HCL is to increase the target glucose for a session of exercise. Some systems call this a “temporary target” while in others, an increased target is embedded into their “exercise mode”. Based upon personalized factors, the increased target should be set anywhere from 30 minutes to 2 hours prior to initiating physical activity and it should remain on for the duration of the activity and in some situations, up to a few hours afterwards (130). In a study of patients with T1D placed on HCL, their target was increased from 2 hours prior to exercise initiation to 15 minutes after. They engaged in either HIIT or moderate intensity exercise in a cross-over study design and only 1 of 12 participants experienced hypoglycemia and it was during their session of moderate intensity exercise. Time spent in hypoglycemia for 24 hours afterwards measured by continuous glucose monitors was minimal in both groups (0 and 0.4% respectively for HIIT and moderate intensity) (131). Tagougui et al studied adults with T1D using a HCL system during 60 minutes of 60% VO2 peak exercise who were randomized to either 1) increase target glucose level and reduce their meal bolus by 33% 90 minutes before exercise 2) increase target glucose but take a full meal bolus 90 minutes before exercise or 3) not change target glucose and take a full meal bolus. The increased target was maintained until 1 hour after exercise. During exercise and the 1-hour recovery period, time spent in hypoglycemia was significantly reduced in both groups 1 and 2 compared to 3 and there was a trend towards less time in hypoglycemia in group 1 vs group 2 (p=0.06) but at the expense of 24.6% more time in hyperglycemia (132).

 

SEX DIFFERENCES WITHIN DIABETES AND EXERCISE

 

According to the IDF Diabetes Atlas, the prevalence of diabetes in adult women in 2021 was 10.2%, compared to 10.8% of men worldwide (133). When adjusted for associated risk factors, women with diabetes have a higher incidence of CVD death and congestive heart failure compared to men (134). Excess CVD in women with T2D correlates with increased adiposity and CVD risk factor burden present in T2D women (135,136).

 

Additionally, based on National Health and Nutrition Examination Surveys between 2007 and 2016, girls and women with T2D have lower physical activity levels than men across all age groups and settings (137). This observation may be due to barriers to exercise, as mentioned above. Of importance, there are sex differences in barriers to exercise as well (138). Women are more likely than men to consider activities of daily living as exercise when referring to physical activity behavior. They are also more likely to report decreased knowledge or skills associated with physical activity (139). Additional barriers for exercise specific to women include decreased perceived neighborhood safety and decreased perceived easy access to locations for physical activity (140). Women also had less self-efficacy, i.e. successful execution of a physical activity behavioral change, than men for participating in physical activity when other common barriers emerged (e.g. time constraints, bad weather) (139). In a meta-analysis of T2D across the lifespan it was shown that across all ages, males participated in more moderate and vigorous activity than females and in adulthood and late adulthood, men were more likely to achieve physical activity recommendations than women (141).

 

Furthermore, women with T2D have a more pronounced exercise impairment in cardiorespiratory fitness then men with T2D (84,87). Interestingly, while obese women with T2D have reduced VO2 kinetics when compared with controls, there is no difference in impairments based on menopausal status (142). The mechanism behind these differences and how it relates to insulin-mediated cardiac and skeletal muscle perfusion impairments is currently being studied.

 

CONCLUSIONS AND FUTURE DIRECTIONS

 

Exercise is an important therapy in prevention and treatment of diabetes. At the same time, this is easier said than done, especially given the barriers to exercise that individuals with diabetes must overcome. These barriers are further complicated by sex differences, with sex also affecting prognosis with a diabetes diagnosis. The etiology of diabetes-related decreases in cardiorespiratory fitness is not yet fully understood; further research is being undertaken in this area to address potential therapeutic targets. Given the discussed correlation between CRF and morbidity and mortality, such an approach could aid in reduction of disability and mortality associated with diabetes. Additionally, a better strategy is needed to measure response to exercise therapy to aid in modification of a regimen to ensure continuous benefit. Given the high heterogeneity in response to exercise, other genetic and environmental components may be responsible. Further research on genetic contributions to exercise response is needed. Ultimately, future therapy will need to be more personalized such that every individual with diabetes receives a specific prescription for exercise based on factors such as sex, diabetes type and duration, comorbidities, genetic background and exercise phenotype, and environment.

 

REFERENCES

 

  1. American Diabetes Association. Facilitating Behavior Change and Well-being to Improve Health Outcomes: Standards of Medical Care in Diabetes—2022. Diabetes Care 2022;45(Suppl. 1):S60–S82.
  2. Piercy KL, Troiano RP. Physical Activity Guidelines for Americans From the US Department of Health and Human Services. Circ Cardiovasc Qual Outcomes. 2018;11(11):e005263.
  3. Committee PAGA. 2018 Physical Activity Guidelines Advisory Committee Scientific Report. Washington, DC: U.S. Department of Health and Human Services; 2018.
  4. Sundquist K, Qvist J, Sundquist J, Johansson SE. Frequent and occasional physical activity in the elderly: a 12-year follow-up study of mortality. Am J Prev Med. 2004;27(1):22-27.
  5. Morrish NJ, Wang SL, Stevens LK, Fuller JH, Keen H. Mortality and causes of death in the WHO Multinational Study of Vascular Disease in Diabetes. Diabetologia. 2001;44 Suppl 2:S14-21.
  6. Centers for Disease Control and Prevention. National Diabetes Statistics Report Website. https://www.cdc.gov/diabetes/data/statistics-report/index.html. Accessed [9/22/2022].
  7. Guthold R, Stevens GA, Riley LM, Bull FC. Worldwide trends in insufficient physical activity from 2001 to 2016: a pooled analysis of 358 population-based surveys with 1·9 million participants. Lancet Glob Health. 2018;6(10):e1077-e1086.
  8. Awotidebe TO, Adedoyin RA, Yusuf AO, Mbada CE, Opiyo R, Maseko FC. Comparative functional exercise capacity of patients with type 2-diabetes and healthy controls: a case control study. Pan Afr Med J. 2014;19:257.
  9. Komatsu WR, Gabbay MA, Castro ML, Saraiva GL, Chacra AR, de Barros Neto TL, Dib SA. Aerobic exercise capacity in normal adolescents and those with type 1 diabetes mellitus. Pediatr Diabetes. 2005;6(3):145-149.
  10. Huebschmann AG, Reis EN, Emsermann C, Dickinson LM, Reusch JE, Bauer TA, Regensteiner JG. Women with type 2 diabetes perceive harder effort during exercise than nondiabetic women. Applied physiology, nutrition, and metabolism = Physiologie appliquee, nutrition et metabolisme. 2009;34(5):851-857.
  11. Korkiakangas EE, Alahuhta MA, Laitinen JH. Barriers to regular exercise among adults at high risk or diagnosed with type 2 diabetes: a systematic review. Health Promot Int. 2009;24(4):416-427.
  12. Brazeau AS, Rabasa-Lhoret R, Strychar I, Mircescu H. Barriers to physical activity among patients with type 1 diabetes. Diabetes Care. 2008;31(11):2108-2109.
  13. Pickering C, Kiely J. Do Non-Responders to Exercise Exist-and If So, What Should We Do About Them? Sports Med. 2019;49(1):1-7.
  14. Regensteiner JG, Bauer TA, Huebschmann AG, Herlache L, Weinberger HD, Wolfel EE, Reusch JE. Sex differences in the effects of type 2 diabetes on exercise performance. Med Sci Sports Exerc. 2015;47(1):58-65.
  15. Warburton DE, Charlesworth S, Ivey A, Nettlefold L, Bredin SS. A systematic review of the evidence for Canada's Physical Activity Guidelines for Adults. Int J Behav Nutr Phys Act. 2010;7:39.
  16. Manson JE, Rimm EB, Stampfer MJ, Colditz GA, Willett WC, Krolewski AS, Rosner B, Hennekens CH, Speizer FE. Physical activity and incidence of non-insulin-dependent diabetes mellitus in women. Lancet. 1991;338(8770):774-778.
  17. Hu FB, Li TY, Colditz GA, Willett WC, Manson JE. Television watching and other sedentary behaviors in relation to risk of obesity and type 2 diabetes mellitus in women. JAMA. 2003;289(14):1785-1791.
  18. Yang J, Qian F, Chavarro JE, et al. Modifiable risk factors and long term risk of type 2 diabetes among individuals with a history of gestational diabetes mellitus: prospective cohort study. BMJ. 2022;378:e070312.
  19. Juraschek SP, Blaha MJ, Blumenthal RS, Brawner C, Qureshi W, Keteyian SJ, Schairer J, Ehrman JK, Al-Mallah MH. Cardiorespiratory fitness and incident diabetes: the FIT (Henry Ford ExercIse Testing) project. Diabetes Care. 2015;38(6):1075-1081.
  20. Lynch J, Helmrich SP, Lakka TA, Kaplan GA, Cohen RD, Salonen R, Salonen JT. Moderately intense physical activities and high levels of cardiorespiratory fitness reduce the risk of non-insulin-dependent diabetes mellitus in middle-aged men. Arch Intern Med. 1996;156(12):1307-1314.
  21. Jetté M, Sidney K, Blümchen G. Metabolic equivalents (METS) in exercise testing, exercise prescription, and evaluation of functional capacity. Clin Cardiol. 1990;13(8):555-565.
  22. Mendes MA, da Silva I, Ramires V, et al. Metabolic equivalent of task (METs) thresholds as an indicator of physical activity intensity. PLoS One. 2018;13(7):e0200701.
  23. Lang JJ, Wolfe Phillips E, Orpana HM, et al. Field-based measurement of cardiorespiratory fitness to evaluate physical activity interventions. Bull World Health Organ. 2018;96(11):794-796.
  24. Patrizio E, Calvani R, Marzetti E, Cesari M. Physical Functional Assessment in Older Adults. J Frailty Aging. 2021;10(2):141-149.
  25. Balk EM, Earley A, Raman G, Avendano EA, Pittas AG, Remington PL. Combined Diet and Physical Activity Promotion Programs to Prevent Type 2 Diabetes Among Persons at Increased Risk: A Systematic Review for the Community Preventive Services Task Force. Ann Intern Med. 2015;163(6):437-451.
  26. Hamman RF, Wing RR, Edelstein SL, Lachin JM, Bray GA, Delahanty L, Hoskin M, Kriska AM, Mayer-Davis EJ, Pi-Sunyer X, Regensteiner J, Venditti B, Wylie-Rosett J. Effect of weight loss with lifestyle intervention on risk of diabetes. Diabetes care. 2006;29(9):2102-2107.
  27. Ross R, Dagnone D, Jones PJ, Smith H, Paddags A, Hudson R, Janssen I. Reduction in obesity and related comorbid conditions after diet-induced weight loss or exercise-induced weight loss in men. A randomized, controlled trial. Ann Intern Med. 2000;133(2):92-103.
  28. Qin L, Knol MJ, Corpeleijn E, Stolk RP. Does physical activity modify the risk of obesity for type 2 diabetes: a review of epidemiological data. Eur J Epidemiol. 2010;25(1):5-12.
  29. Cloostermans L, Wendel-Vos W, Doornbos G, Howard B, Craig CL, Kivimaki M, Tabak AG, Jefferis BJ, Ronkainen K, Brown WJ, Picavet SH, Ben-Shlomo Y, Laukkanen JA, Kauhanen J, Bemelmans WJ. Independent and combined effects of physical activity and body mass index on the development of Type 2 Diabetes - a meta-analysis of 9 prospective cohort studies. Int J Behav Nutr Phys Act. 2015;12:147.
  30. Eriksson KF, Lindgarde F. Prevention of type 2 (non-insulin-dependent) diabetes mellitus by diet and physical exercise. The 6-year Malmo feasibility study. Diabetologia. 1991;34(12):891-898.
  31. Tuomilehto J, Lindstrom J, Eriksson JG, Valle TT, Hamalainen H, Ilanne-Parikka P, Keinanen-Kiukaanniemi S, Laakso M, Louheranta A, Rastas M, Salminen V, Uusitupa M, Finnish Diabetes Prevention Study G. Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N Engl J Med. 2001;344(18):1343-1350.
  32. Pan XR, Li GW, Hu YH, Wang JX, Yang WY, An ZX, Hu ZX, Lin J, Xiao JZ, Cao HB, Liu PA, Jiang XG, Jiang YY, Wang JP, Zheng H, Zhang H, Bennett PH, Howard BV. Effects of diet and exercise in preventing NIDDM in people with impaired glucose tolerance. The Da Qing IGT and Diabetes Study. Diabetes Care. 1997;20(4):537-544.
  33. Kosaka K, Noda M, Kuzuya T. Prevention of type 2 diabetes by lifestyle intervention: a Japanese trial in IGT males. Diabetes Res Clin Pract. 2005;67(2):152-162.
  34. Ramachandran A, Snehalatha C, Mary S, Mukesh B, Bhaskar AD, Vijay V, Indian Diabetes Prevention P. The Indian Diabetes Prevention Programme shows that lifestyle modification and metformin prevent type 2 diabetes in Asian Indian subjects with impaired glucose tolerance (IDPP-1). Diabetologia. 2006;49(2):289-297.
  35. Dai X, Zhai L, Chen Q, Miller JD, Lu L, Hsue C, Liu L, Yuan X, Wei W, Ma X, Fang Z, Zhao W, Liu Y, Huang F, Lou Q. Two-year-supervised resistance training prevented diabetes incidence in people with prediabetes: A randomised control trial. Diabetes Metab Res Rev. 2019 Jul;35(5):e3143.
  36. Diabetes Prevention Program Outcomes Study Research G, Orchard TJ, Temprosa M, Barrett-Connor E, Fowler SE, Goldberg RB, Mather KJ, Marcovina SM, Montez M, Ratner RE, Saudek CD, Sherif H, Watson KE. Long-term effects of the Diabetes Prevention Program interventions on cardiovascular risk factors: a report from the DPP Outcomes Study. Diabet Med. 2013;30(1):46-55.
  37. 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(2):145-154.
  38. Gibbs BB, Brancati FL, Chen H, Coday M, Jakicic JM, Lewis CE, Stewart KJ, Clark JM. Effect of improved fitness beyond weight loss on cardiovascular risk factors in individuals with type 2 diabetes in the Look AHEAD study. Eur J Prev Cardiol. 2014;21(5):608-617.
  39. Bouchard C, An P, Rice T, Skinner JS, Wilmore JH, Gagnon J, Perusse L, Leon AS, Rao DC. Familial aggregation of VO(2max) response to exercise training: results from the HERITAGE Family Study. J Appl Physiol (1985). 1999;87(3):1003-1008.
  40. Hautala AJ, Kiviniemi AM, Makikallio TH, Kinnunen H, Nissila S, Huikuri HV, Tulppo MP. Individual differences in the responses to endurance and resistance training. Eur J Appl Physiol. 2006;96(5):535-542.
  41. Bonafiglia JT, Rotundo MP, Whittall JP, Scribbans TD, Graham RB, Gurd BJ. Inter-Individual Variability in the Adaptive Responses to Endurance and Sprint Interval Training: A Randomized Crossover Study. PLoS One. 2016;11(12):e0167790.
  42. McClatchey PM, Williams IM, Xu Z, Mignemi NA, Hughey CC, McGuinness OP, Beckman JA, Wasserman DH. Perfusion Controls Muscle Glucose Uptake by Altering the Rate of Glucose Dispersion In Vivo. Am J Physiol Endocrinol Metab. 2019.
  43. Cartee GD. Mechanisms for greater insulin-stimulated glucose uptake in normal and insulin-resistant skeletal muscle after acute exercise. Am J Physiol Endocrinol Metab. 2015;309(12):E949-959.
  44. McGarrah RW, Slentz CA, Kraus WE. The Effect of Vigorous- Versus Moderate-Intensity Aerobic Exercise on Insulin Action. Curr Cardiol Rep. 2016;18(12):117.
  45. Way KL, Hackett DA, Baker MK, Johnson NA. The Effect of Regular Exercise on Insulin Sensitivity in Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Diabetes Metab J. 2016;40(4):253-271.
  46. Magkos F, Tsekouras Y, Kavouras SA, Mittendorfer B, Sidossis LS. Improved insulin sensitivity after a single bout of exercise is curvilinearly related to exercise energy expenditure. Clin Sci (Lond). 2008;114(1):59-64.
  47. Dube JJ, Allison KF, Rousson V, Goodpaster BH, Amati F. Exercise dose and insulin sensitivity: relevance for diabetes prevention. Med Sci Sports Exerc. 2012;44(5):793-799.
  48. Newsom SA, Everett AC, Hinko A, Horowitz JF. A single session of low-intensity exercise is sufficient to enhance insulin sensitivity into the next day in obese adults. Diabetes Care. 2013;36(9):2516-2522.
  49. Colberg SR, Sigal RJ, Yardley JE, Riddell MC, Dunstan DW, Dempsey PC, Horton ES, Castorino K, Tate DF. Physical Activity/Exercise and Diabetes: A Position Statement of the American Diabetes Association. Diabetes Care. 2016;39(11):2065-2079.
  50. Church TS, Blair SN, Cocreham S, Johannsen N, Johnson W, Kramer K, Mikus CR, Myers V, Nauta M, Rodarte RQ, Sparks L, Thompson A, Earnest CP. Effects of aerobic and resistance training on hemoglobin A1c levels in patients with type 2 diabetes: a randomized controlled trial. JAMA. 2010;304(20):2253-2262.
  51. Sigal RJ, Kenny GP, Boule NG, Wells GA, Prud'homme D, Fortier M, Reid RD, Tulloch H, Coyle D, Phillips P, Jennings A, Jaffey J. Effects of aerobic training, resistance training, or both on glycemic control in type 2 diabetes: a randomized trial. Ann Intern Med. 2007;147(6):357-369.
  52. Gillen JB, Little JP, Punthakee Z, Tarnopolsky MA, Riddell MC, Gibala MJ. Acute high-intensity interval exercise reduces the postprandial glucose response and prevalence of hyperglycaemia in patients with type 2 diabetes. Diabetes Obes Metab. 2012;14(6):575-577.
  53. Umpierre D, Ribeiro PA, Kramer CK, Leitao CB, Zucatti AT, Azevedo MJ, Gross JL, Ribeiro JP, Schaan BD. Physical activity advice only or structured exercise training and association with HbA1c levels in type 2 diabetes: a systematic review and meta-analysis. JAMA. 2011;305(17):1790-1799.
  54. Chimen M, Kennedy A, Nirantharakumar K, Pang TT, Andrews R, Narendran P. What are the health benefits of physical activity in type 1 diabetes mellitus? A literature review. Diabetologia. 2012;55(3):542-551.
  55. Laaksonen DE, Atalay M, Niskanen LK, et al. Aerobic exercise and the lipid profile in type 1 diabetic men: a randomized controlled trial. Med Sci Sports Exerc. 2000;32(9):1541-1548.
  56. Fuchsjäger-Mayrl G, Pleiner J, Wiesinger GF, et al. Exercise training improves vascular endothelial function in patients with type 1 diabetes. Diabetes Care. 2002;25(10):1795-1801.
  57. Yki-Järvinen H, DeFronzo RA, Koivisto VA. Normalization of insulin sensitivity in type I diabetic subjects by physical training during insulin pump therapy. Diabetes Care. 1984;7(6):520-527.
  58. Ramalho AC, de Lourdes Lima M, Nunes F, et al. The effect of resistance versus aerobic training on metabolic control in patients with type-1 diabetes mellitus. Diabetes Res Clin Pract. 2006;72(3):271-276.
  59. Moy CS, Songer TJ, LaPorte RE, et al. Insulin-dependent diabetes mellitus, physical activity, and death. Am J Epidemiol. 1993;137(1):74-81.
  60. Golbidi S, Badran M, Laher I. Antioxidant and anti-inflammatory effects of exercise in diabetic patients. Exp Diabetes Res. 2012;2012:941868.
  61. Cheng X, Siow RC, Mann GE. Impaired redox signaling and antioxidant gene expression in endothelial cells in diabetes: a role for mitochondria and the nuclear factor-E2-related factor 2-Kelch-like ECH-associated protein 1 defense pathway. Antioxid Redox Signal. 2011;14(3):469-487.
  62. Kasapis C, Thompson PD. The effects of physical activity on serum C-reactive protein and inflammatory markers: a systematic review. J Am Coll Cardiol. 2005;45(10):1563-1569.
  63. Ristow M, Zarse K, Oberbach A, Kloting N, Birringer M, Kiehntopf M, Stumvoll M, Kahn CR, Bluher M. Antioxidants prevent health-promoting effects of physical exercise in humans. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(21):8665-8670.
  64. Sanford JA, Nogiec CD, Lindholm ME, et al. Molecular Transducers of Physical Activity Consortium (MoTrPAC): Mapping the Dynamic Responses to Exercise. Cell. 2020;181(7):1464-1474.
  65. Erickson ML, Little JP, Gay JL, McCully KK, Jenkins NT. Postmeal exercise blunts postprandial glucose excursions in people on metformin monotherapy. J Appl Physiol (1985). 2017;123(2):444-450.
  66. Malin SK, Braun B. Impact of Metformin on Exercise-Induced Metabolic Adaptations to Lower Type 2 Diabetes Risk. Exerc Sport Sci Rev. 2016;44(1):4-11.
  67. Konopka AR, Laurin JL, Schoenberg HM, Reid JJ, Castor WM, Wolff CA, Musci RV, Safairad OD, Linden MA, Biela LM, Bailey SM, Hamilton KL, Miller BF. Metformin inhibits mitochondrial adaptations to aerobic exercise training in older adults. Aging Cell. 2019;18(1):e12880.
  68. Lee IM, Skerrett PJ. Physical activity and all-cause mortality: what is the dose-response relation? Med Sci Sports Exerc. 2001;33(6 Suppl):S459-471; discussion S493-454.
  69. Church TS, Cheng YJ, Earnest CP, Barlow CE, Gibbons LW, Priest EL, Blair SN. Exercise capacity and body composition as predictors of mortality among men with diabetes. Diabetes Care. 2004;27(1):83-88.
  70. Myers J, Prakash M, Froelicher V, Do D, Partington S, Atwood JE. Exercise capacity and mortality among men referred for exercise testing. N Engl J Med. 2002;346(11):793-801.
  71. Seyoum B, Estacio RO, Berhanu P, Schrier RW. Exercise capacity is a predictor of cardiovascular events in patients with type 2 diabetes mellitus. Diab Vasc Dis Res. 2006;3(3):197-201.
  72. Gulsin GS, Swarbrick DJ, Athithan L, et al. Effects of Low-Energy Diet or Exercise on Cardiovascular Function in Working-Age Adults With Type 2 Diabetes: A Prospective, Randomized, Open-Label, Blinded End Point Trial. Diabetes Care. 2020;43(6):1300-1310.
  73. Cai Z, Yang Y, Zhang J. Effects of physical activity on the progression of diabetic nephropathy: a meta-analysis. Biosci Rep. 2021;41(1):BSR20203624.
  74. Waden J, Forsblom C, Thorn LM, Saraheimo M, Rosengard-Barlund M, Heikkila O, Lakka TA, Tikkanen H, Groop PH, FinnDiane Study G. Physical activity and diabetes complications in patients with type 1 diabetes: the Finnish Diabetic Nephropathy (FinnDiane) Study. Diabetes Care. 2008;31(2):230-232.
  75. Kriska AM, LaPorte RE, Patrick SL, Kuller LH, Orchard TJ. The association of physical activity and diabetic complications in individuals with insulin-dependent diabetes mellitus: the Epidemiology of Diabetes Complications Study--VII. J Clin Epidemiol. 1991;44(11):1207-1214.
  76. Bohn B, Herbst A, Pfeifer M, Krakow D, Zimny S, Kopp F, Melmer A, Steinacker JM, Holl RW, Initiative DPV. Impact of Physical Activity on Glycemic Control and Prevalence of Cardiovascular Risk Factors in Adults With Type 1 Diabetes: A Cross-sectional Multicenter Study of 18,028 Patients. Diabetes Care. 2015;38(8):1536-1543.
  77. Yan X, Han X, Wu C, Shang X, Zhang L, He M. Effect of physical activity on reducing the risk of diabetic retinopathy progression: 10-year prospective findings from the 45 and Up Study. PLoS One. 2021;16(1):e0239214.
  78. Kriska AM, Saremi A, Hanson RL, Bennett PH, Kobes S, Williams DE, Knowler WC. Physical activity, obesity, and the incidence of type 2 diabetes in a high-risk population. Am J Epidemiol. 2003;158(7):669-675.
  79. Egan AM, Mahmood WA, Fenton R, Redziniak N, Kyaw Tun T, Sreenan S, McDermott JH. Barriers to exercise in obese patients with type 2 diabetes. QJM. 2013;106(7):635-638.
  80. Reusch JE, Bridenstine M, Regensteiner JG. Type 2 diabetes mellitus and exercise impairment. Rev Endocr Metab Disord. 2013;14(1):77-86.
  81. Nadeau KJ, Zeitler PS, Bauer TA, Brown MS, Dorosz JL, Draznin B, Reusch JE, Regensteiner JG. Insulin resistance in adolescents with type 2 diabetes is associated with impaired exercise capacity. J Clin Endocrinol Metab. 2009;94(10):3687-3695.
  82. Nadeau KJ, Regensteiner JG, Bauer TA, Brown MS, Dorosz JL, Hull A, Zeitler P, Draznin B, Reusch JE. Insulin resistance in adolescents with type 1 diabetes and its relationship to cardiovascular function. J Clin Endocrinol Metab. 2010;95(2):513-521.
  83. Regensteiner JG, Bauer TA, Reusch JE, Brandenburg SL, Sippel JM, Vogelsong AM, Smith S, Wolfel EE, Eckel RH, Hiatt WR. Abnormal oxygen uptake kinetic responses in women with type II diabetes mellitus. J Appl Physiol (1985). 1998;85(1):310-317.
  84. Regensteiner JG, Sippel J, McFarling ET, Wolfel EE, Hiatt WR. Effects of non-insulin-dependent diabetes on oxygen consumption during treadmill exercise. Med Sci Sports Exerc. 1995;27(5):661-667.
  85. Wong CY, O'Moore-Sullivan T, Fang ZY, Haluska B, Leano R, Marwick TH. Myocardial and vascular dysfunction and exercise capacity in the metabolic syndrome. The American journal of cardiology. 2005;96(12):1686-1691.
  86. Moir S, Hanekom L, Fang ZY, Haluska B, Wong C, Burgess M, Marwick TH. Relationship between myocardial perfusion and dysfunction in diabetic cardiomyopathy: a study of quantitative contrast echocardiography and strain rate imaging. Heart. 2006;92(10):1414-1419.
  87. Regensteiner JG, Bauer TA, Reusch JE, Quaife RA, Chen MY, Smith SC, Miller TM, Groves BM, Wolfel EE. Cardiac dysfunction during exercise in uncomplicated type 2 diabetes. Med Sci Sports Exerc. 2009;41(5):977-984.
  88. Bjornstad P, Truong U, Dorosz JL, Cree-Green M, Baumgartner A, Coe G, Pyle L, Regensteiner JG, Reusch JE, Nadeau KJ. Cardiopulmonary Dysfunction and Adiponectin in Adolescents With Type 2 Diabetes. J Am Heart Assoc. 2016;5(3):e002804.
  89. Mason McClatchey P, Bauer TA, Regensteiner JG, Schauer IE, Huebschmann AG, Reusch JEB. Dissociation of local and global skeletal muscle oxygen transport metrics in type 2 diabetes. J Diabetes Complications. 2017;31(8):1311-1317.
  90. Cree-Green M, Scalzo RL, Harrall K, Newcomer BR, Schauer IE, Huebschmann AG, McMillin S, Brown MS, Orlicky D, Knaub L, Nadeau KJ, McClatchey PM, Bauer TA, Regensteiner JG, Reusch JEB. Supplemental Oxygen Improves In Vivo Mitochondrial Oxidative Phosphorylation Flux in Sedentary Obese Adults With Type 2 Diabetes. Diabetes. 2018;67(7):1369-1379.
  91. Broekhuizen L, Lemkes B, Mooij HL, Meuwese MC, Verberne H, Holleman F, Schlingemann RO, Nieuwdorp M, Stroes ES, Vink H. Effect of sulodexide on endothelial glycocalyx and vascular permeability in patients with type 2 diabetes mellitus. Diabetologia. 2010;53(12):2646-2655.
  92. Cabrales P, Vazquez BY, Tsai AG, Intaglietta M. Microvascular and capillary perfusion following glycocalyx degradation. J Appl Physiol (1985). 2007;102(6):2251-2259.
  93. McClatchey PM, Schafer M, Hunter KS, Reusch JE. The endothelial glycocalyx promotes homogenous blood flow distribution within the microvasculature. Am J Physiol Heart Circ Physiol. 2016;311(1):H168-176.
  94. Estacio RO, Regensteiner JG, Wolfel EE, Jeffers B, Dickenson M, Schrier RW. The association between diabetic complications and exercise capacity in NIDDM patients. Diabetes care. 1998;21(2):291-295.
  95. Scalzo RL, Schauer IE, Rafferty D, et al. Single-leg exercise training augments in vivo skeletal muscle oxidative flux and vascular content and function in adults with type 2 diabetes. J Physiol. 2022;600(4):963-978.
  96. Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes. 2002;51(10):2944-2950.
  97. Ritov VB, Menshikova EV, He J, Ferrell RE, Goodpaster BH, Kelley DE. Deficiency of subsarcolemmal mitochondria in obesity and type 2 diabetes. Diabetes. 2005;54(1):8-14.
  98. Regensteiner JG, Bauer TA, Reusch JE. Rosiglitazone improves exercise capacity in individuals with type 2 diabetes. Diabetes Care. 2005;28(12):2877-2883.
  99. Kadoglou NP, Iliadis F, Angelopoulou N, Perrea D, Liapis CD, Alevizos M. Beneficial effects of rosiglitazone on novel cardiovascular risk factors in patients with Type 2 diabetes mellitus. Diabet Med. 2008;25(3):333-340.
  100. Bastien M, Poirier P, Brassard P, Arsenault BJ, Bertrand OF, Despres JP, Costerousse O, Piche ME. Effect of PPARgamma agonist on aerobic exercise capacity in relation to body fat distribution in men with type 2 diabetes mellitus and coronary artery disease: a 1-yr randomized study. Am J Physiol Endocrinol Metab. 2019;317(1):E65-E73.
  101. Bassuk SS, Manson JE. Epidemiological evidence for the role of physical activity in reducing risk of type 2 diabetes and cardiovascular disease. J Appl Physiol (1985). 2005;99(3):1193-1204.
  102. Thompson PD, Franklin BA, Balady GJ, Blair SN, Corrado D, Estes NA, Fulton JE, Gordon NF, Haskell WL, Link MS, Maron BJ, Mittleman MA, Pelliccia A, Wenger NK, Willich SN, Costa F, American Heart Association Council on Nutrition PyA, and Metabolism, Cardiology AHACoC, Medicine ACoS. Exercise and acute cardiovascular events placing the risks into perspective: a scientific statement from the American Heart Association Council on Nutrition, Physical Activity, and Metabolism and the Council on Clinical Cardiology. Circulation. 2007;115(17):2358-2368.
  103. Kanaley JA, Colberg SR, Corcoran MH, et al. Exercise/Physical Activity in Individuals with Type 2 Diabetes: A Consensus Statement from the American College of Sports Medicine. Med Sci Sports Exerc. 2022;54(2):353-368.
  104. de Groot M, Anderson R, Freedland KE, Clouse RE, Lustman PJ. Association of depression and diabetes complications: a meta-analysis. Psychosom Med. 2001;63(4):619-630.
  105. Astor BC, Muntner P, Levin A, Eustace JA, Coresh J. Association of kidney function with anemia: the Third National Health and Nutrition Examination Survey (1988-1994). Arch Intern Med. 2002;162(12):1401-1408.
  106. Crews RT, Schneider KL, Yalla SV, Reeves ND, Vileikyte L. Physiological and psychological challenges of increasing physical activity and exercise in patients at risk of diabetic foot ulcers: a critical review. Diabetes Metab Res Rev. 2016;32(8):791-804.
  107. Bonner T, Foster M, Spears-Lanoix E. Type 2 diabetes-related foot care knowledge and foot self-care practice interventions in the United States: a systematic review of the literature. Diabet Foot Ankle. 2016;7:29758.
  108. Boulton AJ, Armstrong DG, Albert SF, Frykberg RG, Hellman R, Kirkman MS, Lavery LA, Lemaster JW, Mills JL, Sr., Mueller MJ, Sheehan P, Wukich DK, American Diabetes A, American Association of Clinical E. Comprehensive foot examination and risk assessment: a report of the task force of the foot care interest group of the American Diabetes Association, with endorsement by the American Association of Clinical Endocrinologists. Diabetes Care. 2008;31(8):1679-1685.
  109. Connolly V, Unwin N, Sherriff P, Bilous R, Kelly W. Diabetes prevalence and socioeconomic status: a population based study showing increased prevalence of type 2 diabetes mellitus in deprived areas. J Epidemiol Community Health. 2000;54(3):173-177.
  110. Giles-Corti B, Donovan RJ. Socioeconomic status differences in recreational physical activity levels and real and perceived access to a supportive physical environment. Prev Med. 2002;35(6):601-611.
  111. Miller ST, Marolen KN, Beech BM. Perceptions of physical activity and motivational interviewing among rural African-American women with type 2 diabetes. Womens Health Issues. 2010;20(1):43-49.
  112. Goldhaber-Fiebert JD, Goldhaber-Fiebert SN, Tristán ML, Nathan DM. Randomized controlled community-based nutrition and exercise intervention improves glycemia and cardiovascular risk factors in type 2 diabetic patients in rural Costa Rica. Diabetes Care. 2003;26(1):24-29.
  113. Araiza P, Hewes H, Gashetewa C, Vella CA, Burge MR. Efficacy of a pedometer-based physical activity program on parameters of diabetes control in type 2 diabetes mellitus. Metabolism. 2006;55(10):1382-1387.
  114. Association AD. Economic Costs of Diabetes in the U.S. in 2017. Diabetes Care. 2018;41(5):917-928.
  115. Hogan P, Dall T, Nikolov P, Association AD. Economic costs of diabetes in the US in 2002. Diabetes Care. 2003;26(3):917-932.
  116. McCoy RG, Van Houten HK, Ziegenfuss JY, Shah ND, Wermers RA, Smith SA. Increased mortality of patients with diabetes reporting severe hypoglycemia. Diabetes Care. 2012;35(9):1897-1901.
  117. Maran A, Pavan P, Bonsembiante B, Brugin E, Ermolao A, Avogaro A, Zaccaria M. Continuous glucose monitoring reveals delayed nocturnal hypoglycemia after intermittent high-intensity exercise in nontrained patients with type 1 diabetes. Diabetes Technol Ther. 2010;12(10):763-768.
  118. Ertl AC, Davis SN. Evidence for a vicious cycle of exercise and hypoglycemia in type 1 diabetes mellitus. Diabetes Metab Res Rev. 2004;20(2):124-130.
  119. Shahar J, Hamdy O. Medication and exercise interactions: considering and managing hypoglycemia risk. Diabetes Spectr. 2015;28(1):64-67.
  120. Richter EA, Hargreaves M. Exercise, GLUT4, and skeletal muscle glucose uptake. Physiol Rev. 2013;93(3):993-1017.
  121. Katz A, Broberg S, Sahlin K, Wahren J. Leg glucose uptake during maximal dynamic exercise in humans. Am J Physiol. 1986;251(1 Pt 1):E65-70.
  122. Cockcroft EJ, Narendran P, Andrews RC. Exercise-induced hypoglycaemia in type 1 diabetes. Exp Physiol. 2020;105(4):590-599.
  123. Marliss EB, Vranic M. Intense exercise has unique effects on both insulin release and its roles in glucoregulation: implications for diabetes. Diabetes. 2002;51 Suppl 1:S271-S283.
  124. Richter, E. A., Derave, W., & Wojtaszewski, J. F. (2001). Glucose, exercise and insulin: emerging concepts. The Journal of physiology535(Pt 2), 313–322.
  125. Yardley JE, Kenny GP, Perkins BA, Riddell MC, Balaa N, Malcolm J, Boulay P, Khandwala F, Sigal RJ. Resistance versus aerobic exercise: acute effects on glycemia in type 1 diabetes. Diabetes Care. 2013;36(3):537-542.
  126. Guelfi KJ, Jones TW, Fournier PA. The decline in blood glucose levels is less with intermittent high-intensity compared with moderate exercise in individuals with type 1 diabetes. Diabetes Care. 2005;28(6):1289-1294.
  127. Yardley JE, Kenny GP, Perkins BA, Riddell MC, Malcolm J, Boulay P, Khandwala F, Sigal RJ. Effects of performing resistance exercise before versus after aerobic exercise on glycemia in type 1 diabetes. Diabetes Care. 2012;35(4):669-675.
  128. Steineck IIK, Ranjan AG, Schmidt S, Norgaard K. Time spent in hypoglycemia is comparable when the same amount of exercise is performed 5 or 2 days weekly: a randomized crossover study in people with type 1 diabetes. BMJ Open Diabetes Res Care. 2021;9(1):e001919.
  129. Riddell MC, Gallen IW, Smart CE, Taplin CE, Adolfsson P, Lumb AN, Kowalski A, Rabasa-Lhoret R, McCrimmon RJ, Hume C, Annan F, Fournier PA, Graham C, Bode B, Galassetti P, Jones TW, Millan IS, Heise T, Peters AL, Petz A, Laffel LM. Exercise management in type 1 diabetes: a consensus statement. Lancet Diabetes Endocrinol. 2017;5(5):377-390.
  130. Zaharieva DP, Messer LH, Paldus B, O'Neal DN, Maahs DM, Riddell MC. Glucose Control During Physical Activity and Exercise Using Closed Loop Technology in Adults and Adolescents with Type 1 Diabetes. Can J Diabetes. 2020 Dec;44(8):740-749.
  131. Lee MH, Vogrin S, Paldus B, Jayawardene D, Jones HM, McAuley SA, Obeyesekere V, Gooley J, La Gerche A, MacIsaac RJ, Sundararajan V, Jenkins AJ, Ward GM, O'Neal DN. Glucose and Counterregulatory Responses to Exercise in Adults With Type 1 Diabetes and Impaired Awareness of Hypoglycemia Using Closed-Loop Insulin Delivery: A Randomized Crossover Study. Diabetes Care. 2020 Feb;43(2):480-483.
  132. Tagougui S, Taleb N, Legault L, et al. A single-blind, randomised, crossover study to reduce hypoglycaemia risk during postprandial exercise with closed-loop insulin delivery in adults with type 1 diabetes: announced (with or without bolus reduction) vs unannounced exercise strategies. Diabetologia. 2020;63(11):2282-2291.
  133. Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JCN, Mbanya JC, Pavkov ME, Ramachandaran A, Wild SH, James S, Herman WH, Zhang P, Bommer C, Kuo S, Boyko EJ, Magliano DJ. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022 Jan;183:109119.
  134. Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA. 1979;241(19):2035-2038.
  135. Sattar N. Gender aspects in type 2 diabetes mellitus and cardiometabolic risk. Best Pract Res Clin Endocrinol Metab. 2013;27(4):501-507.
  136. Huebschmann AG, Huxley RR, Kohrt WM, Zeitler P, Regensteiner JG, Reusch JEB. Sex differences in the burden of type 2 diabetes and cardiovascular risk across the life course. Diabetologia. 2019.
  137. Scholes S, Bann D. Education-related disparities in reported physical activity during leisure-time, active transportation, and work among US adults: repeated cross-sectional analysis from the National Health and Nutrition Examination Surveys, 2007 to 2016. BMC Public Health. 2018;18(1):926.
  138. Miller TM, Gilligan S, Herlache LL, Regensteiner JG. Sex differences in cardiovascular disease risk and exercise in type 2 diabetes. J Investig Med. 2012;60(4):664-670.
  139. Barrett JE, Plotnikoff RC, Courneya KS, Raine KD. Physical activity and type 2 diabetes: exploring the role of gender and income. Diabetes Educ. 2007;33(1):128-143.
  140. Bengoechea E, Spence JC, McGannon KR. International Journal of Behavioral Nutrition and Physical Activity. 2005;2(1):12.
  141. Whipple MO, Pinto AJ, Abushamat LA, Bergouignan A, Chapman K, Huebschmann AG, Masters KS, Nadeau KJ, Scalzo RL, Schauer IE, Rafferty D, Reusch JEB, Regensteiner JG. Sex Differences in Physical Activity Among Individuals With Type 2 Diabetes Across the Life Span: A Systematic Review and Meta-analysis. Diabetes Care. 2022 Sep 1;45(9):2163-2177.
  142. Kiely C, Rocha J, O'Connor E, O'Shea D, Green S, Egana M. Influence of menopause and Type 2 diabetes on pulmonary oxygen uptake kinetics and peak exercise performance during cycling. Am J Physiol Regul Integr Comp Physiol. 2015;309(8):R875-883.

 

Endocrine testing for the Syndrome of Inappropriate Antidiuretic Hormone Secretion (SIADH)

ABSTRACT

 

The diagnosis of Syndrome of Inappropriate Antidiuretic Hormone Secretion (SIADH) relies on an adequate assessment of a hyponatremic state (that is a serum sodium level <136 mmol/l) and on the exclusion of other causative conditions leading to an appropriate secretion of antidiuretic hormone (ADH). The understanding of mechanisms involved in pathological ADH secretion is essential for diagnosis and therapy. Although some forms are due to dysregulation in central nervous system regulation, other forms are dependent on diseases in peripheral organs and structures including ADH-producing/secreting neuroendocrine tumors, while others are induced by drugs. ADH regulation is closely linked to other systems such as the sympathetic nervous system via and baroreflex regulation. Patients with hyponatremia should be assessed carefully whether or not neurological symptoms exist. Further, assessment of volume status is needed. Based on symptoms and volume status, the need for intensive-care monitoring is determined. In parallel, laboratory findings of blood and urine must be analyzed appropriately. It is important to demonstrate true hyponatremia, which is paralleled by a decrease in serum osmolality. Mandatory laboratory diagnostic steps comprise the determination of blood and urine electrolytes and serum and urine osmolality, analysis of thyroid, adrenocortical, and kidney function as well as uric acid. Different test results such as a high fractional uric acid excretion may hint to an existing SIADH. Assessment of urine osmolality and urine sodium concentration and intravascular volume level may allow for further discrimination and may be indicative for a specific underlying disorder, causing SIADH. Brain volume changes (“hydrocephalus ex vacuo”) may depend on age rendering the elderly more tolerant to acute or chronic serum sodium changes. The course of incident hyponatremia, if documented, may affect therapy. A serum sodium drop within less than 48 hours is considered acute hyponatremia. A rapid bolus of 100 to 150 ml of intravenous 3% hypertonic saline is appropriate to avoid catastrophic outcomes in severe cases of acute symptomatic hyponatremia.

 

INTRODUCTION

 

“Antidiuretic hormone” or “arginine vasopressin” (AVP) is physiologically released into the blood stream upon increases of plasma osmolality. AVP, a nine-amino-acid peptide, originates in the supraophthalmic nucleus (SON) of the hypothalamus and is directly regulated by plasma osmolality detected via a splice variant of the capsaicin receptor, the transient receptor potential vanilloid type-1 (Trpv1) receptor (1). AVP is axonally transported to the posterior pituitary and released into the blood upon respective stimulation. AVP regulation is, however, more complex than a mere response to changes of plasma osmolality. In fact, hypovolemia enhances AVP release upon increases of plasma osmolality. Conversely, hypervolemia attenuates AVP release for given increases of plasma osmolality.

In patients with “Syndrome of Inappropriate Antidiuretic Hormone Secretion” or SIADH, the cornerstone of diagnosis is hyponatremia (Na<136 mmol/l) (2) in a state of euvolemia, i.e. absence of either over- or dehydration. SIADH-related hyponatremia is caused by excess water reabsorption due to inappropriately high levels of AVP. Specifically, AVP binds to stimulatory G-protein-coupled vasopressin V2-receptors of the basolateral membrane of collecting-duct cells, thereby increasing the intracellular cAMP level, which, in turn, activates Aquaporin-2 channels of the brush-border membrane or urine side of the collecting-duct principal cells. AVP action results in a reduced free-water clearance resulting in a more concentrated urine and total-body water (TBW) expansion. AVP-mediated TBW expansion mediates sympathoinhibition (Figure 1).

 

Figure 1. Role of AVP in regulation of sympathetic tone. AVP translates into sympathoinhibition (Figure amended from (3) ).

 

In addition, AVP-induced TBW expansion translates into plasma-solute dilution leading to hyponatremia. Thereby, plasma osmolality (OSM) is decreased given the fact that sodium strongly determines OSM according to the following equation:  

 

OSM (calculated) = 2 x Na (mmol/l) + Glucose (mmol/l) + Urea (mmol/l) 

 

Besides AVP actions on OSM, AVP also enhances endothelial-cell synthesis and the release of von-Willebrand Factor, thereby affecting hemostasis (4). This AVP effect on hemostasis is therapeutically used in bleeding disorders involving Factor VIII or von-Willebrand factor deficiency (5, 6).

 

SIADH may be viewed as a primary central-nervous system dysregulation of OSM and/or thirst. The etiology still is incompletely understood. Alternatively, SIADH may relate to baroreceptor unloading due to clinically inapparent hypovolemia or, hypothetically, to carotid-artery atherosclerosis affecting baroreflex regulation (7). This alternative route of increased AVP release may ultimately translate into the clinical picture of SIADH. Generally, less wall distension of the carotid arterial walls and/or the aortic arch may lead to a decrease of arterial baroreceptor-related afferent autonomic nerve traffic to the rostral ventrolateral medulla and nucleus tractus solitarii (NTS) translating into less sympathoinhibition (sympathoexcitation) and to an increased release of AVP (8) (Figure 2).

 

Figure 2. Baroreflex regulation and SIADH: arterial hypotension lowers baroreflex-mediated afferent nerve traffic to the nucleus tractus solitarii leading to an elevated efferent sympathetic nerve activity and increased AVP release.

 

Lastly, hypovolemia-related cardiopulmonary (CP) reflex deactivation mediated by less wall distension of the right atrial wall and pulmonary veins may increase plasma AVP leading to SIADH (9, 10). Conversely, CP reflex activation mediated by more right-atrial wall distension e.g. after body immersion in water is able to decrease plasma AVP (11).

 

The term “primary SIADH” is used for all above-mentioned causes involving a known or suspected dysregulation of OSM and/or circulating-blood volume. The term “secondary SIADH” is attributed to pituitary-independent causes of AVP increases, e.g., in hormone-active neoplasms such as small-cell lung cancer. In addition, a drug-induced type of SIADH is detailed here.

 

CLINICAL PRESENTATION OF SIADH

 

Both moderate and especially severe hyponatremia (Na < 125 mmol/l) found in newly admitted hospital patients is linked with a significantly elevated in-hospital mortality of 28% compared to 9% in-hospital mortality in normonatremic, matched control patients (12). Mortality, in fact, increases when serum Na levels are below 137 mmol/L (13). While (neurological) symptoms of hyponatremia such as gait disturbances, cognitive dysfunction and dizziness may lead to falls leading to subsequent injuries requiring medical care, either preceding symptom may be subtle and difficult to diagnose. Therefore, hyponatremia often is overseen or not given full attention. Furthermore, if hyponatremia is diagnosed, it is regularly classified to be asymptomatic. Diagnostic differentiation remains absent or incomplete. Thus, underlying reasons often remain obscure (14). However, cognitive and/or geriatric functional tests regularly reveal a significant impairment in states of hyponatremia.

 

Clearly, symptoms of hyponatremia depend on the time elapsed since the start of hyponatremia development. Hyponatremia developing in less than 48 hours may already present with severe symptoms which are mainly caused by cerebral edema and a high intracranial pressure. These include epileptic convulsions, a pronounced somnolence or coma, vomiting and/or a compromised respiratory regulation. Symptoms like headache or modest nausea generally reflect a rather moderate severity.

 

In patients with acute hyponatremia, a brief patient history and a physical examination should be performed (Table 1). In cases of a rather slowly developing or chronic hyponatremia, intracellular regulations such as decreased uptake of taurin aim to adapt to the decreased extracellular osmolality. Therefore, those patients may show very subtle or even no clinical alterations. Taurin is an endogenous amino acid that mediates cellular adaptation to hyperosmotic stress (15).

Table 1. Anamnestic Factors and Conditions Responsible for the Occurrence of an Acute Hyponatremic State (<48h)

Medical interventions:

General post-operative phase

Resection of the prostate

Exercise (e.g., long distance run) with increased and rapid fluid (water) intake

Extended sauna visit

Polydipsia (transient)

Severe pain attacks (including concomitant pharmacotherapy)

Initiation of new drugs

e.g., thiazides, terlipressin, psychiatric medication

For the diagnosis of SIADH, the first diagnostic step, hyponatremia needs to be ascertained. False laboratory “measurements” of serum Na+ comprise primarily a hyperglycemic state. According to Hillier et al (16) an estimation of the true sodium concentration in serum can be drawn from the formula:

 

Corrected (Na+) = Measured (Na+) + 2.4 x (glucose (mg/dl) - 100 mg/dl)/100mg/dl

 

For example, a serum glucose level of 400 mg/dl with a measured serum sodium of 120 mmol/l corresponds to a true sodium value of 127 mmol/l (16).

 

Likewise, pseudohyponatremia may occur in patients with paraproteinemia, e.g. multiple-myeloma patients (17).

 

Another pitfall with serum sodium is the mode of its determination: the usual method involves ion-selective electrodes, not flame-photometric determination and may yield occasionally “diluted” Na+ levels (elevated triglyceride levels, paraproteinemia). Measured, not calculated serum osmolality may help discriminate true from pseudohyponatremia: true hyponatremia associates with a decreased serum osmolality.

 

True-hyponatremic patients are regularly identified at hospital admission, e.g., in the emergency room. However, a large number of hospitalized patients have or develop either mild (Na 131 – 136 mmol/l), moderate (Na 126 – 130 mmol/l) or severe (Na <125 mmol/l) hyponatremia after admission during the hospital stay. Although many hyponatremic patients will present with chronic hyponatremia, however, some patients present with a proven acute hyponatremia. Since acute hyponatremia means an incomplete adjustment of the difference in osmolality between plasma (extracellular space) and intracellular cell space of tissues and organs such as the brain, a 48-hours threshold to discern acute from chronic hyponatremia appears reasonable. However, in everyday practice, this discrimination might not be feasible due to a lack of documentation of serum sodium levels in newly hospitalized patients.

 

As an important step in the approach to the hyponatremic patient is to determine volume status. Accordingly, a state of overhydration characterized by the presence of peripheral edema needs to be ruled out. Hyponatremia accompanied by peripheral edema, anasarca, jugular vein distension in absence of a significant tricuspid-valve regurgitation, dyspnea, and/or signs of a lung fluid or pulmonary edema on the chest radiograph are not consistent with the diagnosis of SIADH. Here, underlying diseases such as chronic heart failure should be diagnosed and addressed. Likewise, hyponatremia in a state of hypovolemia needs to be excluded. In exsiccosis, e.g., due to diuretics, both water and solutes may be lost. Hypovolemia triggers sympathoactivation via CP and baroreflex leading to an appropriate ADH release.

 

In patients presenting with hyponatremia in a state of euvolemia, i.e., absence of overhydration or exsiccosis, the diagnosis of SIADH should be considered, after excluding conditions such as chronic heart failure. A known and medically treated chronic heart failure condition may present as a hypervolemic, euvolemic or – when vigorously treated – hypovolemic state. Both, chronic heart failure and liver cirrhosis are characterized by arterial underfilling and, hence, activation of both the renin-angiotensin-aldosterone system and antidiuretic hormone leading to both sodium chloride retention and water retention. The net effect may be hyponatremia, if AVP stimulation dominates. Thus, urine-sodium measurements in 24-hours urine collection may prove the presence of a sodium-sparing disorder, thereby rendering SIADH unlikely.

 

A useful algorithm to approach hyponatremia is depicted in Figure 3.

 

Figure 3. Approach to the patient with hyponatremia (adapted and modified from (18), with permission).

 

In the clinical assessment, euvolemic patients suspected to have primary SIADH often show a slight weight gain by 5 – 10 % of body weight and/or a worsened condition of arterial hypertension, and urine output is normal or slightly reduced. In secondary, neoplasm-associated SIADH, however, an unexplained weight loss and/or state of cachexia may prompt further diagnostics including chest radiograph and thoracic computed tomography scan, if deemed necessary.

 

It is crucial to assess and to treat infections effectively, since they may establish or worsen a tendency to hyponatremia.

                           

To correctly evaluate laboratory results besides aspects of methodology, diuretics,

especially thiazides, should be discontinued, and nutritional sodium chloride intake should not exceed 5 – 6 g per day.

 

HYPONATREMIA IN EUVOLEMIA: ASSESSMENT FOR SIADH

 

The diagnostic, step-wise approach for evaluating hyponatremia in euvolemic patients is detailed below:

 

1st Step

 

Here, a laboratory work-up (Table 2) is proposed to establish a preliminary diagnosis being consistent with SIADH.

 

Table 2. Laboratory Work-Up for SIADH

 

Parameter

SIADH Diagnosis

Serum

Sodium

Potassium

Glucose

Urea

Creatinine

Uric acid

Thyroid hormones

Cortisol

Aldosterone

Copeptin

<136 mmol/l

Normal

Normal

Normal

Normal

Normal

Normal

Normal

Normal

Elevated

Urine

Osmolality

Sodium

Uric acid

Creatinine

Osmolality

>100 mosm/kg

> 30 mmol/l

Normal or Low

Normal

>12%

 

As a diagnostic cornerstone of SIADH diagnosis, determination of fractional uric-acid excretion has recently emerged (SIADH increased). Fenske et al. (19) confirmed earlier reports demonstrating fractional uric-acid excretion (cut-off greater than 12 %) to rule out hyponatremic states with reduced extracellular fluid volume, e.g. due to diuretics (Figure 4).

 

Figure 4. Calculation of fractional uric acid excretion.

There is sometimes debate on whether measurements of AVP in the plasma are helpful or not for diagnosis of SIADH or other hyponatremic circumstances. There are, however; multiple obstacles rendering AVP determination and interpretation difficult. Only a few laboratories provide tests with good sensitivity, since AVP is very unstable when isolated from plasma and binds to other structures. A potential alternative is the more stable copeptin, also called C-terminal proarginine vasopressin, which is generated by enzymatic cleavage of the vasopressin prohormone. Strikingly, SIADH patients were shown to have an elevated plasma copeptin (20). As an additional way to diagnose SIADH, correlation of plasma copeptin with changes in plasma osmolality (step 2, step 3) can be used (21). Furthermore, a hypertonic (3%) saline test has been proposed for SIADH (20). However, this test strategy still needs to be validated.

 

2nd Step

 

Administration of 500 ml saline (NaCl 0.9 %) is regarded as an empiric, first-line therapeutic and diagnostic measure. Especially when arterial underfilling and baroreflex and/or CP reflex deactivation are thought to be relevant, i.v. saline corrects the initial disturbance that led to hyponatremia/SIADH. This relates, strictly speaking, to volume regulation and respective reflex stimulation. Isotonic saline may not resolve hyponatremia in cases of ongoing urine concentration with urine osmolality exceeding 400 mosm/kg.

 

In addition, a water-load test as described in www.endotext.org, NEUROENDOCRINOLOGY, HYPOTHALAMUS, AND PITUITARY may be performed the next morning under utmost scrutiny and precaution, in the hospital. The water-load test relates to the fact that normal subjects excrete 78-82% of the ingested water load within 4 hours due to AVP suppression. In patients with SIADH, the expected urine amount within 4 hours is reduced to 30-40%. However, the test involves a massive water intake in a short period of time and, therefore, is not considered a safe procedure for the majority of patients for the following three reasons:

  • In SIADH patients, a relative intravascular overhydration will be enhanced.
  • An underlying cardiac co-morbidity may be adversely affected.
  • The water challenge may worsen the hyponatremia increasing the 

 risk of symptoms such as epileptic convulsions.

 

In the majority of patients with hyponatremia suspected to have SIADH, this test can be replaced by the following 3rdand 4th diagnostic step (below). The water-load test only adds information in hospitalized individuals free of cardiac conditions presenting with rather mild hyponatremia in whom the 4th step usually will not be performed. Again, the physician has to weigh risks and benefits of this water challenge versus alternate diagnostic steps (step 3, step 4 below).

 

3rd Step

 

A balanced fluid-intake restriction (500 ml/day) is able to correct hyponatremia over the next 3 to 4 days with an aimed plasma-sodium increase of 0.5 mmol/l/h or less than 10 mmol/l/day. Most patient do not tolerate a very strict fluid intake reduction.

 

4th Step

 

If step 1 – step 3 did not lead to an improvement of hyponatremia, therapy with an antagonist of the vasopressin V2-receptors, e.g. tolvaptan, for four days should be instituted (22) with an aimed plasma-sodium increase of 0.5 mmol/l/h or less than 10 mmol/l/day.

 

INTERPRETATION OF CLINICAL AND LABORATORY RESULTS

 

SIADH leads to an increase of free-water reabsorption, thereby increasing the circulating blood volume. By virtue of dilution mediated by AVP, both hematocrit and plasma sodium are decreased. Likewise, a decrease of urine output can be found.

 

In cases of a prolonged, subclinical hypovolemia, baroreflex- and/or CP-reflex unloading stimulates AVP secretion leading to the clinical picture of SIADH. There, discontinuation of diuretics and/or the empirical infusion of 500 ml saline (0.9 %) as outlined above in step 2 may correct such a state of subclinical hypovolemia and lead to an improvement in hyponatremia driven by SIADH.

 

In assessing key laboratory results including plasma and urine sodium concentration and -osmolality, both the theoretical or calculated OSM should be compared to the actually measured OSM. That way, states of hyponatremia due to uremia or hyperglycemia can be ruled out. In such cases of hyponatremia, high plasma urea or high plasma glucose lead to a rise in OSM prompting a physiologic release of AVP, which, in turn, leads to a plasma-sodium dilution in order to maintain a normal OSM.

 

Urine sodium within normal range rules out a dietary sodium deficiency or states of increased tubular sodium reabsorption such as in chronic heart failure or liver cirrhosis.

 

After fulfilling the above-mentioned steps to diagnose SIADH, the following conditions should be separately considered as a possible differential diagnosis:

 

DIFFERENTIAL DIAGNOSIS OF HYPONATEMIA OTHER THAN SIADH

 

  • Sodium chloride depletion, low dietary sodium intake regularly is accompanied by hypovolemia, low urine sodium, elevated serum uric acid and serum urea.

 

  • Anterior-lobe pituitary gland insufficiency often is accompanied with signs and symptoms, and respective laboratory findings indicating hormone deficiencies such as hypothyroidism, hypocortisolism or hypogonadism. In addition, bitemporal hemianopsia and hyperprolactinemia are found in cases of anterior-lobe pituitary tumors as a cause of anterior-lobe pituitary gland insufficiency.

 

  • Adrenal-gland insufficiency including iatrogenic mineralocorticoid-receptor antagonism (spironolactone/eplerenone) regularly is accompanied by hyperkalemia and hypovolemia.

 

  • Thiazide diuretics can induce hyponatremia by an AVP-dependent mechanism and by a thiazide-induced increase of water permeability in the medullary collecting duct.

 

  • Severe hypothyroidism regularly is accompanied by dilutional hyponatremia due to a reduced free-water clearance.

 

  • Chronic kidney disease: In salt losing nephropathy, a condition that occurs in advanced kidney failure with a GFR below 15 ml/min, hyponatremia is paralleled by hypovolemia. This is a feature classically seen in interstitial kidney disease. On the other hand, many patients with near end-stage renal failure show increased Na excretion to balance body sodium content, but, due to (continuous) reduction in urine production, a diluted urine cannot be achieved, leading to hyponatremia.

 

  • Acute kidney (transplant) failure without signs of uremia, a water-excretion dysfunction may lead to dilutional hyponatremia.

 

  • Hyperglycemia or poor diabetes mellitus control may lead to a so-called translational hyponatremia due to intra- to extracellular water shift and consequent plasma sodium dilution (see above).

 

  • Cerebral salt wasting is an important differential diagnosis to SIADH occurring in cases of aneurysmal subarachnoidal hemorrhage and in other intracranial pathologies. Cerebral salt wasting still is not completely characterized and most likely involves a putative central nervous system-derived factor and/or a sudden decrease of renal sympathetic nerve activity favoring a urinary loss of sodium chloride. Cerebral-salt wasting - associated urinary sodium-chloride loss improves after successful neurosurgical care of the initial intracranial disease condition and may require temporary high amounts of sodium chloride replacement.

 

  • Overdose of antidiuretic-hormone analogs in cases of known central diabetes insipidus. If the primary physician is unaware of the underlying medical condition, SIADH may be suspected based on laboratory results described above. Patient history including the medication list clarifies the diagnosis.

 

THERAPY OF SIADH

If left untreated, SIADH may lead to a severe, life-threatening hyponatremia with or without clinically apparent overhydration. Acute complications of SIADH include cerebral edema and epileptic convulsions. SIADH therapy clearly depends on the specific etiology and the severity of symptoms.

 

Once therapy is initiated, repeat measurements of plasma sodium are mandatory to gauge the therapeutic response, and, most importantly, to ascertain a slow plasma-sodium normalization with a recommended maximum rate of 0.5 mmol/l/h plasma-sodium increase. Again, the delivery of higher concentrated sodium chloride solution is allowed strictly for symptomatic patients. A significant proportion of in-hospital mortality relating to hyponatremia likely is due to a too rapid sodium normalization in long-standing hyponatremia. The consequence of too rapid sodium normalization is the osmotic demyelination syndrome due to a rapid intra- to-extracellular water transfer and subsequent brain swelling that exceeds the percentage of cerebrospinal fluid volume capacity (usually around 8% but higher in elderly with a hydrocephalus ex vacuo).

 

Besides the therapeutic goal to avoid rapid changes in plasma osmolality, the underlying reason of hyponatremia in SIADH, excess total body water, should be addressed by balanced fluid-intake reduction. All therapeutic interventions discussed here target the consequences of exaggerated AVP secretion rather than sodium-chloride supplementation.

 

In primary SIADH, plasma-sodium dilution can be addressed by an ongoing fluid-intake restriction of 500-800 ml/day which many patients do not tolerate well.

 

However, subclinical hypovolemia and ensuing baroreflex and CP reflex suppression leading to AVP stimulation should be kept in mind. Addressing arterial hypotension and/or central-venous hypotension is effective in lowering AVP in plasma. At the same time, fluid-intake restriction may appear contradictory. Even though fluid-intake restriction appears to be a proven measure in terms of attenuation of AVP consequences, it is the clinician`s judgement to test both interventions and compare the best results in terms of a slow plasma-sodium increase.

 

In secondary SIADH, identification of the neoplasm is the goal. A thorough tumor search using positron-emission tomography and computed tomography is warranted to further determine the underlying pathology and, if applicable, consider all options of curative therapy. Chemotherapy, surgical and/or radiation therapy of malignancies with AVP activity represent definitive therapeutic approaches. On clinical grounds, neoplasm-associated, secondary SIADH often requires V2-receptor antagonism therapy until a specific oncologic care plan is employed. This might be especially true while performing cytostatic therapy cycles with increased intravenously administered fluid volumes. However, to date, no survival benefit has been demonstrated in favor of V2-receptor antagonism in oncologic care of patients with secondary, neoplasm-associated SIADH.

 

Besides malignancies, infections such as tuberculosis have been associated with occurrence of SIADH (24).

 

 

Only if there are severe symptoms related hyponatremia, is it advised to administer a small amount of a hypertonic NaCl-solution for a very short period of time. In practice, this must be accomplished with closely monitored sodium concentration measurements which can be accomplished on a regular ward rather than in the intensive care unit, especially when considering the urgency. 150 ml of a 3% saline solution can be infused over 20 minutes. If the symptoms do not ameliorate with this management, the infusion with 100 ml of 3% hypertonic saline can be repeated every 30 minutes until the target serum Na is reached (usually 5-8 mmol increase from baseline). Above all, a 5 mmol/l increase in serum sodium concentration should not be exceeded within the first hour. These recommendations have also been summarized in recent guideline reports (18).

 

The management after relief of the symptoms should be focused on a careful administration of 0.9% (only) sodium chloride solution. Independently of the initial rise in serum sodium concentration by the above measures, the maximal rise within the first 24 hours should not exceed 10 mmol/l. In some cases of overcorrection, desmopressin can be administered (13, 23). In terms of reaching a sodium goal within a short time, a rapid intermittent bolus infusion of hypertonic solution is preferred to a slow continuous infusion (25).

 

Drug-Induced SIADH

 

Drugs (see Table in Ref. 18) such as vincristine, vinblastine, cyclophosphamide, carbamazepine, tricyclic antidepressants, selective serotonin reuptake inhibitors (e.g., citalopram), oxytocin, opiates, barbiturates, and nicotine may cause SIADH. Either they enhance ADH release, are analogues of ADH, or they amplify the renal effects of ADH. However, for some drugs, the mechanism remains unclear. If applicable, a suspected drug should be discontinued with close supervision of plasma sodium levels. Once hyponatremia improves after cessation of a specific drug, drug-induced SIADH is likely. However, unless re-exposure takes place, drug- induced SIADH is not proven.

 

Primary and Secondary SIADH

 

In both primary and secondary SIADH or in euvolemic, hyponatremic patients with suspected SIADH without a therapeutic effect of fluid-intake restriction, the lowest recommended standard dose of a vasopressin V2-receptor blocker, e.g., Tolvaptan, should be administered orally. Besides the diagnostic approach outlined above as step 4, empiric V2-receptor antagonism represents a rescue therapy and is suitable to gain time needed to perform further diagnostics and therapies in (suspected) SIADH.

 

Low-dose Tolvaptan therapy was shown to significantly improve hyponatremia (by 3 – 4 mmol/l) within 4 days when compared to placebo (26, 27). Alternatively, conivaptan (approved for SIADH in the United States) has a broader target than its competitors tolvaptan or mozavaptan. Conivaptan selectively targets the V1a and V2 receptors. Conivaptan can be administered intravenously in patients who are unable to take drugs orally. In addition, conivaptan has a longer bioavailability than newer vaptans including tolvaptan (28). In a large proportion of SIADH patients, whether or not the underlying circumstances leading to SIADH are known, fluid-intake restriction controls hyponatremia attributed to SIADH. It is important to note, that with the use of higher daily  tolvaptan dosages, the risk of liver injury may increase, as revealed in a recent study in which vaptans were tested to treat autosomal-polycystic kidney disease (ADPKD) (29).

 

In essence, V2-receptor antagonism remains both a diagnostic and a therapeutic tool for SIADH when applied under scrutiny and for a limited period of time. To date, data on long-term use of V2-receptor blockers have not been published, especially for the use of a combined V1a-V2 receptor blockade. V2 antagonists that have been used for the treatment of SIADH are listed in Table 3. The fact that hyponatremia may reoccur shortly after V2-receptor-blocker discontinuation emphasizes the need to identify the underlying cause of SIADH in order to devise a definitive therapy.

 

Table 3. V2 Receptor Antagonists

Drug

Route of administration

Receptor affinity

Literature

Tolvaptan

 

 

oral

 

          V2

(26, 27)

(Lixivaptan)

(30)

(Mozavaptan)

(only approved in Japan)

(31)

(Satavaptan)

(32)

(Conivaptan)

(only approved in U.S.A.)

intravenous

      V2  /V1A

(28, 33)

In parenthesis: limited or no availability.

 

REFERENCES

 

1)      Naeini RS, Witty MF, Seguela P, Bourque CW. An N-terminal variant of Trpv1 channel is required for osmosensory transduction. Nat Neurosci 2006; 9(1):93-8.

2)      Hoorn EJ, Zietse R. Diagnosis and Treatment of Hyponatremia: Compilation of the Guidelines. J Am Soc Nephrol 2017;28(5):1340-1349 .

3)      Zucker IH, Pliquett RU. Novel mechanisms of sympatho-excitation in chronic heart failure. Heart Fail Monit 2002; 3(1):2-7.

4)      Kaufmann JE, Oksche A, Wollheim CB, Gunther G, Rosenthal W, Vischer UM. Vasopressin-induced von Willebrand factor secretion from endothelial cells involves V2 receptors and cAMP. J Clin Invest 2000 Jul; 106(1):107-16.

5)      Castaman G, Tosetto A, Federici AB, Rodeghiero F. Bleeding tendency and efficacy of anti-haemorrhagic treatments in patients with type 1 von Willebrand disease and increased von Willebrand factor clearance. Thromb Haemost 2011 Apr; 105(4):647-54.

6)      Chistolini A, Dragoni F, Ferrari A, et al. Intranasal DDAVP: biological and clinical evaluation in mild factor VIII deficiency. Haemostasis 1991; 21(5):273-7.

7)      Magovern JA, Sieber PR, Thiele BL. The syndrome of inappropriate secretion of antidiuretic hormone following carotid endarterectomy. A case report and review of the literature. J Cardiovasc Surg (Torino) 1989 Jul; 30(4):544-6.

8)      Leimbach WN, Jr., Schmid PG, Mark AL. Baroreflex control of plasma arginine vasopressin in humans. Am J Physiol 1984 Oct; 247(4 Pt 2):H638-H644.

9)      Egan B, Grekin R, Ibsen H, Osterziel K, Julius S. Role of cardiopulmonary mechanoreceptors in ADH release in normal humans. Hypertension 1984 Nov; 6(6 Pt 1):832-6.

10)    Rogge JD, Moore WW. Influence of lower body negative pressure on peripheral venous ADH levels in man. J Appl Physiol 1968 Aug; 25(2):134-8.

(11)   Epstein M, Preston S, Weitzman RE. Isoosmotic central blood volume expansion suppresses plasma arginine vasopressin in normal man. J Clin Endocrinol Metab 1981 Feb; 52(2):256-62.

12)    Hoorn EJ, van der Lubbe N, Zietse R. SIADH and hyponatraemia: why does it matter? NDT Plus 2009 Nov 1; 2(suppl 3):iii5-iii11.

13)    Verbalis JG, Goldsmith SR, Greenberg A, et al. Diagnosis, evaluation, and treatment of hyponatremia: expert panel recommendations. Am J Med 2013 Oct; 126(10 Suppl 1):S1-42.

14)    Clayton JA, Le J, I, Hall IP. Severe hyponatraemia in medical in-patients: aetiology, assessment and outcome. QJM 2006 Aug; 99(8):505-11.

15)    Ito T, Fujio Y, Hirata M, et al. Expression of taurine transporter is regulated through the TonE (tonicity-responsive element)/TonEBP (TonE-binding protein) pathway and contributes to cytoprotection in HepG2 cells. Biochem J 2004 Aug 15; 382(Pt 1):177-82.

16)    Hillier TA, Abbott RD, Barrett EJ. Hyponatremia: evaluating the correction factor for hyperglycemia. The American Journal of Medicine 1999 Apr; 106(4):399-403.

17)    Overlack A, Niederle N, Klautke G, Stumpe KO, Kruck F. (Pseudohyponatremia in multiple myeloma (author's transl)). Klin Wochenschr 1980 Sep 1; 58(17):875-80.

18)    Spasovski G, Vanholder R, Allolio B, et al. Clinical practice guideline on diagnosis and treatment of hyponatraemia. Eur J Endocrinol 2014 Mar; 170(3):G1-47.

19)    Fenske W, Stork S, Koschker AC, et al. Value of fractional uric acid excretion in differential diagnosis of hyponatremic patients on diuretics. J Clin Endocrinol Metab 2008 Aug; 93(8):2991-7.

20)    Fenske WK, Christ-Crain M, Hörning A, Simet J, Szinnai G,‖ Fassnacht M, Rutishauser J, Bichet DG, Störk S, Allolio B. A Copeptin-Based Classification of the Osmoregulatory Defects in the Syndrome of Inappropriate Antidiuresis. Am Soc Nephrol. 2014 Oct; 25(10): 2376–2383.

21)    Wuttke A, Dixit KC, Szinnai G, Werth SC, Haagen U, Christ-Crain M, Morgenthaler N, Brabant G. Copeptin as a marker for arginine-vasopressin/antidiuretic hormone secretion in the diagnosis of paraneoplastic syndrome of inappropriate ADH secretion. Endocrine. 2013 Dec;44(3):744-9.

22)    Schrier RW, Gross P, Gheorghiade M, et al. Tolvaptan, a Selective Oral Vasopressin V2-Receptor Antagonist, for Hyponatremia. New England Journal of Medicine 2006 Nov 16; 355(20):2099-112.

23)    Gharaibeh KA, Craig MJ, Koch CA, Lerant AA, Fulop T, Csongradi E. Desmopressin is an effective adjunct treatment for reversing excessive hyponatremia overcorrection. World J Clin Cases 2013 Aug 16; 1(5):155-8.

24)    Knoop H, Knoop U, Behr J, et al. Syndrome of inadequate antidiuretic hormone secretion in pulmonary tuberculosis - a therapeutic challenge. Pneumologie 2013 Apr; 67(4):219-22.

25)    Baek SH, Jo YH, Ahn S, Medina-Liabres K,, Oh YK, Lee JB, Kim S. Risk of Overcorrection in Rapid Intermittent Bolus vs Slow Continuous Infusion Therapies of Hypertonic Saline for Patients With Symptomatic Hyponatremia: The SALSA Randomized Clinical Trial. JAMA Intern Med 2020 26;e205519

26)    Verbalis JG, Adler S, Schrier RW, Berl T, Zhao Q, Czerwiec FS. Efficacy and safety of oral tolvaptan therapy in patients with the syndrome of inappropriate antidiuretic hormone secretion. Eur J Endocrinol 2011 May; 164(5):725-32.

27)    Schrier RW, Gross P, Gheorghiade M, et al. Tolvaptan, a selective oral vasopressin V2-receptor antagonist, for hyponatremia. N Engl J Med 2006 Nov 16; 355(20):2099-112.

28)    Li-Ng M, Verbalis JG. Conivaptan: Evidence supporting its therapeutic use in hyponatremia. Core Evid 2010 Jun 15; 4:83-92.

29)    Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in patients with autosomal dominant polycystic kidney disease. N Engl J Med 2012 Dec 20; 367(25):2407-18.

30)    Abraham WT, Shamshirsaz AA, McFann K, Oren RM, Schrier RW. Aquaretic effect of lixivaptan, an oral, non-peptide, selective V2 receptor vasopressin antagonist, in New York Heart Association functional class II and III chronic heart failure patients. J Am Coll Cardiol 2006 Apr 18; 47(8):1615-21.

31)    Yamaguchi K, Shijubo N, Kodama T, et al. Clinical implication of the antidiuretic hormone (ADH) receptor antagonist mozavaptan hydrochloride in patients with ectopic ADH syndrome. Jpn J Clin Oncol 2011 Jan; 41(1):148-52.

32)    Soupart A, Gross P, Legros JJ, et al. Successful long-term treatment of hyponatremia in syndrome of inappropriate antidiuretic hormone secretion with satavaptan (SR121463B), an orally active nonpeptide vasopressin V2-receptor antagonist. Clin J Am Soc Nephrol 2006 Nov; 1(6):1154-60.

33)    Annane D, Decaux G, Smith N. Efficacy and safety of oral conivaptan, a vasopressin-receptor antagonist, evaluated in a randomized, controlled trial in patients with euvolemic or hypervolemic hyponatremia. Am J Med Sci 2009 Jan; 337(1):28-36.

Cryptorchidism and Hypospadias

ABSTRACT

 

Undescended testis (UDT) is a common abnormality, affecting about 1/20 males at birth. Half of these have delayed testicular descent, with the testis in the scrotum by 10-12 weeks after term. Beyond this spontaneous descent is rare. Current treatment recommendations are that UDT beyond 3 months of age need surgery before 12 months of age. Some children have scrotal testes in infancy but develop UDT later in childhood because the spermatic cord does not elongate with age, leaving the testes behind as the scrotum moves further from the groin with growth of the pelvis. This is now known as ascending/acquired cryptorchidism, and orchidopexy is controversial. Many authors recommend surgery once the testes no longer reside spontaneously in the scrotum, but some groups recommend conservative treatment. The fetal testis descends in 2 separate hormonal and anatomical steps, with the first step occurring between 8-15 weeks’ gestation. Insulin-like hormone 3 (INSL3) from developing Leydig cells stimulates the genito-inquinal ligament, or gubernaculum, to swell where it ends in the inguinal area of the abdominal wall. This holds the testis near the future inguinal canal as the fetal abdomen enlarges. By contrast, in female fetuses, lack of INSL3 allows the gubernaculum to elongate into a round ligament and lets the ovary move away from the groin. The second or inguinoscrotal phase is controlled by androgen and occurs between 25-35 weeks’ gestation, where the gubernaculum and testis migrate together to the scrotum. Androgens guide this complex process, both directly and indirectly via a neurotransmitter, calcitonin gene-related peptide (CGRP), released from the genitofemoral nerve. After migration is complete the proximal processus vaginalis closes (preventing inguinal hernia) and then the fibrous remnant disappears completely, allowing the spermatic cord to elongate with age, to keep the testis scrotal. The transabdominal phase is a simple mechanical process, and abnormalities are uncommon, with intra-abdominal testes found in 5-10% of boys with UDT. Anomalies of the complex inguinoscrotal phase account for most UDT seen clinically. The undescended testis suffers heat stress when not at the lower scrotal temperature (33 degrees Celsius), interfering with testicular physiology and development of germ cells into spermatogonia. UDT interrupts transformation of neonatal gonocytes into type-A spermatogonia, the putative spermatogenic stem cells at 3-9 months of age. Recent evidence suggests orchidopexy between 6-12 months improves germ cell development, with early reports of improved fertility, but little evidence yet for changes in malignancy prognosis. Hypospadias is also a common abnormality in newborn males, affecting about 1/150 boys. Androgens control masculinization of the genital tubercle into penis between 8-12 weeks’ gestation, with tubularization of the urethra from the perineum to the tip of the glans. If this process is disrupted hypospadias occurs, with a variable proximal urethral meatus, failed ventral preputial development producing a dorsal hood, and discrepancy in the ventral versus dorsal penile length, causing a ventral bend in the penis, known as chordee. Surgery to correct hypospadias is recommended between 6-18 months, as technical advances now allow operation to be done before the infant acquires long-term memory of the surgery. Severe hypospadias overlaps with disorders of sex development (DSD), so that babies without a fused scrotum containing 2 testes and who present with ‘hypospadias’ need full DSD investigations at birth.

 

INTRODUCTION

 

Undescended testis, or "cryptorchidism", is a very common anomaly in male infants and pre-adolescent boys, with about 1 in 20 boys undergoing treatment by the time they reach puberty. Not only is it prevalent, but also there remain unresolved questions about prognosis in adult life. It is not known yet whether the dramatic changes in the recommended age for surgery (from 15 years of age in the 1950’s, to six months old now) (1-4) will decrease the risk of infertility or testicular cancer. However, current treatment is based on the assumption that early surgery will prevent germ cell degeneration during childhood, leading to improved fertility and fewer tumors (5, 6).

 

Our understanding of the embryology has advanced rapidly in recent years, with new theories and experimental evidence supporting a complex anatomical process controlled directly and indirectly by hormones (7, 8). The classification of cryptorchidism also is changing, with the recent recognition of acquired anomalies (9, 10). With so much change in the way we view and treat cryptorchidism, endocrinologists will need to keep checking on the evolving controversies described in this chapter.

 

EMBRYOLOGY

 

The testes descend prenatally from their initial intra-abdominal location on the urogenital ridge into the low-temperature environment of the scrotum via a complex multi-stage mechanism (11).  Prior to 7-8 weeks of development, the gonadal position is similar in both sexes.  With the onset of sexual differentiation, the fetal testis begins producing anti-Müllerian hormone (AMH; also called Müllerian inhibiting substance (MIS)) from the Sertoli cells, as well as androgen and insulin-like hormone 3 (INSL3) from the Leydig cells. These hormones are involved in controlling descent of the male gonad, which is held by two thickenings in the mesentery, the cranial suspensory ligament at the upper pole, and the genito-inguinal ligament, or “gubernaculum”, at the lower pole (12).

 

During the initial (transabdominal) phase of descent, regression of the cranial ligament and thickening of the gubernaculum allows the testis to be held near the inguinal region (13).  By contrast, in the female the cranial ligament persists while the gubernaculum remains thin and elongates, which together hold the ovary higher on the posterior abdominal wall as the fetal abdomen enlarges. The inguinal canal forms by the abdominal wall muscles developing around the caudal, gelatinous end of the gubernaculum, which initially ends at the future external inguinal ring.  By 15 weeks the testis is attached by a short, stout and gelatinous gubernaculum to the future internal inguinal ring, while the ovary is higher in the pelvis (14).

 

In mid gestation a diverticulum of the peritoneal membrane, known as the processus vaginalis, begins to elongate within the gubernaculum, which retains a central connection (known as the gubernacular cord) with the epididymal tail and the lower pole of the testis. The caudal end of the gubernaculum grows out of the abdominal wall and elongates towards the scrotum, extending the processus vaginalis eventually to the scrotum. Between 25-30 weeks’ gestation the testis descends rapidly through the inguinal canal, and then more slowly across the pubic region and into the scrotum, with descent within this peritoneal diverticulum complete by 35 weeks. After the testis reaches the scrotum, two further anatomical events complete the inguinoscrotal phase, the first of which is obliteration of the proximal processus vaginalis (15). The second event is involution of the gelatinous gubernacular bulb and its anchoring to the inside of the scrotum.  The former process prevents inguinal hernia or hydrocele and the latter process prevents extravaginal or perinatal torsion of the testis (6).

 

Figure 1. The embryological stages of testicular descent and the postnatal growth required to keep the testis in the scrotum.

 

The two main phases of descent appear to be controlled independently by hormones.  (Fig. 1). INSL3 is the major factor controlling gubernacular enlargement (16-18) and androgen, particularly DHT, and AMH appear to play minor roles in this "swelling reaction" of the gubernaculum (19-22).  Under the influence of the hormones mentioned above, the caudal end of the gubernaculum, where it attaches to the inguinal abdominal wall, enlarges by proliferation of the embryonic mesenchyme and deposition of extracellular matrix.  Androgens also are responsible for regression of the cranial suspensory ligament, but they are not sufficient alone for transabdominal descent. The phase of gubernacular migration is controlled both directly and indirectly by androgens, with the aid of the genitofemoral nerve (GFN) releasing calcitonin gene-related peptide (CGRP) (23, 24).  Androgens act during a critical time window to regulate gubernacular development (25).  Recent evidence suggests that the androgen receptors controlling this masculinization of the GFN may not be in the nerve itself, but in the target organ, the inguinoscrotal fat pad in the  mammary line (26) .  The number of sensory neurons and the amount of CGRP in the genitofemoral nerve of rats are significantly less after exposure to the anti-androgen, flutamide, consistent with androgens stimulating structural and functional changes in the nerve. The nerve is proposed to orient the direction of gubernacular migration, while the physical force needed for elongation of the processus vaginalis is probably provided by intra-abdominal pressure (27). CGRP released from the nerve stimulates mitosis and cremaster muscle development in the gubernacular tip, enabling elongation to the scrotum (16). Estrogens have a minor inhibitory role in normal gubernacular development, but estrogenic endocrine disruptors may be responsible in larger doses for cryptorchidism secondary to suppression of the "swelling reaction" by inhibition of INSL3.

 

The trigger that initiates active migration of the caudal tip of the gubernaculum may come from the inguinoscrotal fat pad in the mammary line (28), as the androgen receptors are present in the mammary line mesenchyme but not in the adjacent gubernaculum during the critical window of androgenic programming in rodents (25, 29).  The gubernaculum has a surprisingly close link with the embryonic breast in normal marsupials as well as in eutherian animal models, such as the rat and mouse, especially after they have been exposed to the antiandrogen, flutamide (30, 31).

 

The primitive mammary line is in continuity with the apical ectodermal ridges of the upper and lower limb buds, and hence is likely to contain similar activated signaling systems as seen in limb bud development (32).  These signals are likely to initiate outgrowth of the gubernaculum from the abdominal wall, so that it can migrate to the scrotum.

 

ETIOLOGY

 

Any anomaly in either the hormonal control or the anatomical processes in normal testicular descent will cause cryptorchidism (33). Hormonal defects in INSL3, AMH or androgenic action are identified only rarely, suggesting that mechanical anomalies may be more common.  Those patients with hormonal defects may present with rare disorders of sexual development (DSD) with cryptorchidism as part of the complex genital anomaly. The first or transabdominal phase involves little movement of the testis and this may explain the low frequency (5-10%) of intra-abdominal testes.  As the gubernacular swelling reaction holds the testis close to the inguinal canal while other structures grow further away, the transabdominal phase is only relative movement of the testis and hence less likely to be abnormal.  By contrast, the inguinoscrotal migration phase requires very significant mechanical and anatomical re-arrangements, and consequently, anomalies are common: over 60% of testes are found just outside the external inguinal ring, consistent with anomalous or arrested gubernacular migration. Transient deficiency of androgen production in utero,perhaps related to deficiency of gonadotropin production by the fetal pituitary or the placenta (34), may account for some, particularly where there is intra-uterine growth retardation. Anomalies of the genitofemoral nerve also may cause undescended testes. For example, perineal testes may be caused by an anomalous location of the genitofemoral nerve (35).

 

Inherited syndromes frequently are associated with cryptorchidism. Hypothalamic dysfunction, connective tissue disorders, neurogenic (e.g., spina bifida), and mechanical anomalies (e.g., arthrogryposis multiplex congenita) may all cause disruption in testicular descent (36-38).  Cryptorchidism is also common in infants with abdominal wall defects, such as exomphalos or omphalocele, gastroschisis and exstrophy of the bladder (39).

 

There is much current interest in the potential adverse effects of environmental estrogenic endocrine disruptors on the incidence of both cryptorchidism and hypospadias (40). In addition, there are data on the effect of diethylstilbestrol (DES) on cryptorchidism in male offspring of exposed mothers (41). In the latter case there is supporting evidence from animal models (42), although in the former, the cause-and-effect relationship is more tenuous, because the level of exposure is less clear, and the epidemiology may not have allowed for changes in diagnostic criteria over recent decades. More work is needed before we can ascertain a proven cause-and-effect link with synthetic molecules in the environment.

 

The body of the epididymis is hypoplastic and frequently is not tightly adherent to the cryptorchid testis (43). This is more common in high intra-abdominal testes and probably indicates significantly decreased androgen production. Whether epididymal-testicular separation is the cause or the result of cryptorchidism is not known (44). In addition, its effect on fertility is uncertain, even though the rete testis is nearly always still connected to the head of the epididymis.  Recent studies show a strong link between maternal smoking and cryptorchidism in male offspring (45, 46).

 

CLINICAL PRESENTATION

 

Up to 4-5% of newborn males show cryptorchidism, but this falls to 1-2% by 12 weeks after term, following normal (but postnatal) descent in premature infants, and delayed postnatal descent in some term babies. Beyond 12 weeks, spontaneous testicular descent is rare (47). Geographic differences in prevalence of cryptorchidism have been reported, with 9% of Danish boys with undescended testes at birth, compared with only 2% of males from Finland.  Some of these apparent differences, however, may be related to the definitions used for ‘cryptorchidism’ in these studies.  An undescended testis is best defined as a testis that cannot be manipulated into the bottom of the scrotum (without excess tension on the spermatic cord) by 12 weeks of age. Most testes (about 85%) are near the neck of the scrotum, or just lateral to the external inguinal ring, described by Denis Browne as the "superficial inguinal pouch”(48).

 

A few cryptorchid testes are within the inguinal canal, making them unpalpable unless they can be squeezed out of the external inguinal ring by compression. Ten percent of testes are intra-abdominal or absent (presumed to be secondary to prenatal torsion). Ectopic cryptorchid testes are rare (< 5%), and occur in the perineum, prepubic region, thigh, or the contralateral inguinal canal (transverse testicular ectopia) (49).

 

ENDOCRINE EFFECTS OF CRYPTORCHIDISM

 

In infants with undescended testes, the testosterone and gonadotropin levels are diminished compared with normal infants between one and four months of age (50, 51), which is during the normal, transient hormonal surge, known as ‘minipuberty’ (52).  Whether this is a sign of primary endocrinopathy or secondary dysfunction of the testis, caused by heat stress when the gonad is not in the low temperature environment of the scrotum, is unknown. Postnatal increase in testosterone production is also diminished in premature infants, perhaps secondary to inadequate stimulation by chorionic gonadotropin in utero (53).  HCG is low compared with early pregnancy and may be of functional significance. Despite lower than normal androgen levels between 1 and 4 months of age, there is no apparent anomaly in androgen receptors from gonadal or skin biopsies collected at orchidopexy (54).

 

The postnatal secretion of both AMH and inhibin-B in cryptorchid infants is also deranged. Production of AMH from Sertoli cells normally increases between 4-12 months, but this surge is blunted in undescended testes (55, 56).  Inhibin-B normally increases at minipuberty and remains elevated into the second year of life , but levels in infants with cryptorchidism are lower (57).

 

GERM CELL MATURATION IN CRYPTORCHIDISM

 

Germ cells mature postnatally from a primitive gonocyte through a series of steps to primary spermatocytes by 3-4 years. This process is perturbed in cryptorchid testes, with failure of transformation of gonocytes into type-A spermatogonia between 4-12 months (58-60).  These observations suggest that germ cell deficiency may be at least partly secondary to early postnatal dysfunction, rather than being congenital, as proposed by some authors (61, 62).

 

Lack of germ cell transformation has been proposed to be secondary to postnatal androgen deficiency (60, 63) or low AMH levels (63).   Recent studies, however, suggest that transformation is normal in both infants and mice with complete androgen insensitivity syndrome (CAIS), and may be mediated by activin or another TGF-family factor (64).  Abnormal postnatal maturation of gonocytes could lead to both infertility and malignancy (65)), although some authors propose that there may be congenital carcinoma in-situ-cells in the cryptorchid testis (61, 66, 67).

 

There is now a consensus that type-A spermatogonia are likely to be the stem cells for future spermatogenesis, and that their appearance between 3 and 12 months of age, as neonatal gonocytes transform, is the key step in postnatal germ cell development  (68, 69).  Should this be confirmed, it implies that early surgical intervention should lead to an excellent prognosis, as long as the subsequent germ cell deficiency is secondary to postnatal heat stress of the maldescended testis, and therefore reversible.  Failure of the totipotential gonocytes to transform into unipotential spermatogenic stem cells may leave some persisting gonocytes in the undescended testis, which is speculated to be the origin of subsequent tumors.

 

DIAGNOSIS

 

The aim of clinical examination is to locate the gonad, if palpable, and determine its lowest position without causing painful traction on the spermatic cord (which probably corresponds to the caudal limit of the tunica vaginalis) (70).  In infants, the diagnosis is straightforward because the scrotum is thin and pendulous.  Hypoplasia of the hemiscrotum indicates it does not contain a testis. The inguinal testis is within its tunica vaginalis which gives it significant mobility. Ultrasonography has become more frequently used for diagnosis of the impalpable testis, but generally is not contributory for true intra-abdominal testes. This is because absence of the testis (secondary to possible perinatal torsion) is common, and also because intra-abdominal testes are often concealed by the bowel and other viscera (71). In addition, the mobility of the undescended testis within its tunica vaginalis may make location by ultrasonography difficult.  An ultrasound scan can be justified in bilateral impalpable testes, to confirm the presence of a testis.  In addition AMH and inhibin-B should be measured to confirm the presence of functioning Sertoli cells (57).  A simple and reliable approach is to use laparoscopy, which readily locates the testis itself (or blind-ending gonadal vessels), and allows orchidopexy in experienced hands (72).

 

TREATMENT

 

Newborn and Infant

 

Hormone therapy has become extremely controversial (73, 74) as it was based on the two assumptions that cryptorchidism is not only secondary to a deficiency of the hypothalamic-pituitary-gonadal axis, but also the mechanical processes were simple. Both hCG and GnRH therapy have been tried, with success rates ranging from 10-50%. Randomized, double-blind, placebo-controlled studies have not shown more than marginal benefit with either hCG or GnRH (75-77). Despite proven endocrine control of descent, the mechanical factors appear to be too complex for this simple approach to be successful except for acquired undescended testes (76).  Because of its poor efficacy and possible side effects, a consensus meeting in Scandinavia several years ago recommended that hormone treatment be abandoned completely (73, 74).

 

Surgical treatment is based on the premise that early intervention will prevent secondary testicular degeneration caused by high temperature (35-37oC) as the lower temperature of the scrotum (33oC) is essential for normal postnatal germ cell maturation (78). Evidence of progressive germ cell loss in the cryptorchid testis after six months of age has accumulated over the last 50 years and now suggests that orchidopexy should be considered between 6 and 12 months of age (1-4).  The first signs of abnormal germ cell development can be seen between 4-12 months of age (60), and intervention is based on the premise that these changes are secondary to high temperature and should be reversible. Certainly in animal models, early intervention prevents germ cell loss (79).  A prospective study of children randomized to early (9 months) or late (3 years) surgery is showing improved testicular development with early intervention, as measured by ultrasonography at 4 years of age (80, 81).  Surgery at this very early age ideally needs a trained pediatric surgeon, as the technique is quite different from that for a 5-10 year-old boy (82, 83)

 

All baby boys need examination at birth to document gonadal position. Those infants without two fully descended testes should be re-examined at 12 weeks of age and, if a testis is still undescended, the child should be referred to a pediatric surgeon for possible surgical treatment. Orchidopexy is done as an ambulatory procedure, with discharge home a few hours after operation. General anesthesia is supplemented with local/regional analgesia, which will provide pain relief for the first few hours postoperatively.

 

Prognosis

 

The complication rate after orchidopexy is less than 5% in experienced hands (82, 83).  Wound infection is common in infants secondary to external contamination of the wound, although there is a low risk of atrophy of the testis which is greatest when intra-abdominal testes are pulled down under tension. Laparoscopy, with or without ligation of the testicular vessels (Fowler-Stephens procedure) (84), shows increasing success for high intra-abdominal gonads (72, 85, 86).  The prognosis for fertility, the primary aim of orchidopexy, remains uncertain (61),(87-89). However, extensive review of the recent literature suggests improved outcomes with very early surgery (89, 90).  Now that early germ cell maturation in the first year is known to be deranged, improved fertility is to be expected with very early orchidopexy (88-90). Unfortunately, it will be a few more years before the long-term outcome of this new consensus policy is known.

 

The risk of malignancy was previously calculated to be 5 -10 times greater than normal for a man with a history of unilateral cryptorchidism (91-95) when surgery was performed in mid-childhood.  The risk in a future generation for men who underwent orchidopexy in infancy is unknown at present, but is anticipated to be much lower than in the past, as supported by preliminary evidence (90) . 

 

Some clinical features are associated with statistically better outcomes, and include testes near the neck of the scrotum, and ascending or retractile testes (see below), where malignancy risk is now thought to be similar to men without cryptorchidism in childhood (61, 89). Poor prognostic factors are primary testicular or epididymal dysplasia, intra-abdominal or intra-canalicular position, associated strangulated inguinal hernia and (possibly) surgery late in childhood or adolescence (96).

 

ACQUIRED CRYPTORCHIDISM

 

Retractile Testes

 

Retraction of the testis out of the scrotum secondary to reflex contraction of the cremaster muscle is both normal and common and is involved in temperature control and protecting the testis from trauma. The reflex is absent or weak at birth and becomes more active after one year, reaching a peak in 5-10 year-old boys (97).

 

Many testes are erroneously described as "retractile" when they can be pulled down into the scrotum during the physical examination but retract back out of the scrotum on release. This retractability is assumed to be secondary to cremasteric activity, but an alternative explanation has been proposed recently, which is that the malposition may be caused by failure of the spermatic cord to elongate with age (98). Since the distance from external inguinal ring to the bottom of the scrotum increases from 5 cm at birth to 8-10 cm at 10 years of age, the spermatic cord must double in length to keep the testis in the scrotum during the first decade. Preliminary evidence suggests that failure of complete obliteration of the processus vaginalis may prevent normal postnatal elongation of the vas and vessels (99) (Fig. 2).

 

Figure 2. Acquired cryptorchidism occurs when the spermatic cord fails to elongate in proportion to growth between birth and late childhood. This figure shows what happens between birth and 5-10 years of age when the spermatic cord does not elongate with age.

 

Ascending Testes

 

The ascending testis is a special variant of acquired maldescent, in which there is delayed postnatal descent of the testis in the first three months after birth (100), (101).  Follow-up studies suggest that subsequent "ascent" of the testis is common later in childhood (102-104)). The cause for ascending testes is not resolved, with the only well-documented cause being neuronal dysfunction as seen in children with cerebral palsy and spastic diplegia (105). In normal children, the explanation is likely to be persistence of the processus vaginalis, either patent or as a fibrous remnant (106).

 

Management of Acquired Cryptorchidism

 

Both “retractile” and ascending testes are likely to be different names for what is, in effect, acquired cryptorchidism caused by persistence of the processus vaginalis (107-110). The normal spermatic cord elongates gradually with growth, and hence acquired cryptorchidism develops insidiously, presenting mostly between 5 and 10 years of age (111).  Orchidopexy is recommended by some authors once the testis can no longer reside spontaneously in the scrotum, and can be performed in the standard manner or by a scrotal approach (112). Once the fibrous remnant of the processus vaginalis is divided, the testis can reach the scrotum easily.  In The Netherlands recently there has been a consensus to treat acquired cryptorchidism conservatively (113), with follow-up suggesting a poor outcome for fertility (114).  However, whether early treatment by orchidopexy will improve the prognosis for fertility is not yet known.

 

The prognosis for this special group is probably much better than for congenital cryptorchidism, as the testis is normally located in the scrotum during infancy (89), (115, 116), when germ cell maturation is occurring. Unfortunately, previous studies of outcome for fertility and malignancy have not discriminated between congenital and acquired cryptorchidism, but recent studies suggest a mild suppression of fertility and little risk of malignancy (61),(89). The frequency of acquired cryptorchidism, may account for up to half of all children coming to orchidopexy (111),(117).

 

HYPOSPADIAS

 

The primitive phallus begins to enlarge at 8 weeks of development in the male, in response to fetal androgens. The inner genital folds fuse in the midline in association with elongation and canalization of the endodermal urethral plate on the penile shaft, to create the anterior urethra up to the coronal groove by about 12 weeks’ gestation, while the urethra within the glans forms in mid-gestation by canalization of the endoderm forming the distal urethral plate (118, 119). The preputial skin forms from low folds on the dorsum of the shaft at the corona, eventually covering the entire glans (118).  Recent evidence suggests that some of the effects of androgen in penile development may be mediated by aromatization to estrogen, and estrogen receptors (ERα and ERβ) are located in the developing prepuce, glans and urethral plate (120).

 

Failure of urethral canalization and fusion leads to hypospadias (Greek for "hole underneath"), with secondary deficiency of the ventral prepuce ("dorsal hood") and relative deficiency in growth of the peri-urethral tissues compared with the corpora cavernosa, leading to "chordee", or ventral curvature of the penis (121), (Fig. 3).

 

Figure 3. Hypospadias is associated with a) failure of the urethral meatus to be located on the tip of the glans, and failed ventral fusion of the prepuce, causing a ‘dorsal hood’; and b) inadequate growth of the ventral shaft around the urethra, leading to bend, known as chordee.

 

Hypospadias occurs in one in every 100-300 boys, depending on the criteria used for diagnosis (122). About 10% of patients with hypospadias have a sibling or father with the anomaly, suggesting a polygenic inheritance pattern (123). The severity of the anomaly varies widely, from a perineal opening to an opening on the proximal glans, or even chordee with a normal urethral meatus.

 

Care is needed in diagnosis, as some infants with a disorder of sex development (DSD) and ambiguous genitalia may be diagnosed as "simple hypospadias” (124). Since hypospadias is an anatomical anomaly of anterior urethral development, the rest of the external (and also internal) genitalia should be normal. Patients with DSD, by contrast, have a more extensive genital anomaly, reflecting the failure of all androgen-dependent development.

 

A useful rule-of-thumb is to assume that any baby with "hypospadias", as well as an undescended testis and/or bifid scrotum, should be investigated for DSD, with immediate hormonal, chromosomal and anatomical studies. Immediate gender assignment as male is only safe when the scrotum is fused and both testes are descended fully (i.e., androgen-dependent genital development is normal).

 

Surgical treatment is required to reconstruct the penis in hypospadias (125-127). Despite numerous different operative techniques available, there are a few principles of management: a). Create an extension to the urethra to bring it to the tip of the glans, allowing normal micturition; b) Correct the chordee to create a straight shaft for normal sexual function; c) Finally, repair the dorsal hood for cosmetic reasons. In severe cases the skin is moved ventrally to create the urethra and elongate the ventral surface; in mild cases the dorsal hood can be repaired to restore the normal appearance of the foreskin. Surgery is best between 6-18 months, and this is the recommended age, as this avoids much psychological stress (128) (129) but the operation should be completed at the latest in infants or young children prior to school entry.  The operation may be done as day surgery, but may need admission with urinary diversion, depending on the severity of the anomaly and the surgeon’s preference.

 

The prognosis for micturition and sexual function is good, with improving cosmetic appearance with newer procedures (130). However, wound infection, hematoma, urethral breakdown to create a fistula, and stricture, continue to be serious problems, as the surgery requires significant skill (131, 132).


REFERENCES

 

  1. Lee P.A. and Houk C.P., Cryptorchidism. Current opinion in endocrinology, diabetes, and obesity, 2013. 20(3): p. 210-16.
  2. Kolon T.F., Herndon C.D., Baker L.A., et al., Evaluation and treatment of cryptorchidism: AUA guideline. J Urol, 2014. 192(2): p. 337-45.
  3. Pastuszak A.W. and Lipshultz L.I., AUA guideline on the diagnosis and treatment of cryptorchidism. J Urol, 2014. 192(2): p. 346-9.
  4. Steinbrecher H., The undescended testis: working towards a unified care pathway for 2014. Archives of Disease in Childhood, 2014. 99(5): p. 397-8.
  5. Mouriquand P.D., Undescended testes in children: the paediatric urologist's point of view. Eur J Endocrinol, 2008. 159 Suppl 1: p. S83-6.
  6. Hutson J.M. and Thorup J., Evaluation and management of the infant with cryptorchidism. Curr Opin Pediatr, 2015. 26(4): p. 520-4.
  7. Foresta C., Zuccarello D., Garolla A., et al., Role of hormones, genes, and environment in human cryptorchidism. Endocr Rev, 2008. 29(5): p. 560-80.
  8. Hughes I.A. and Acerini C.L., Factors controlling testis descent. Eur J Endocrinol, 2008. 159 Suppl 1: p. S75-82.
  9. Guven A. and BA K., Undescended testis in older boys: further evidence that ascending testes are common. J Pediatr Surg, 2008. 43(9): p. 1700-4.
  10. van der Plas E.M., Zijp G.W., Froeling F.M., et al., Long-term testicular volume after orchiopexy at diagnosis of acquired undescended testis. The Journal of Urology, 2013. 190(1): p. 257-62.
  11. Hutson J.M., Southwell B.R., Li R., et al., The Regulation of Testicular Descent and the Effects of Cryptorchidism. Endocr Rev, 2013. 134(5): p. 725-52.
  12. Backhouse K.M., The natural history of testicular descent and maldescent. Proc R Soc Med, 1966. 59(4): p. 357-60.
  13. van der Schoot P., The name cranial ovarian suspensory ligaments in mammalian anatomy should only be used to indicate the structures derived from the foetal cranial mesonephric and gonadal ligaments. Anat. Rec., 1993. 237: p. 434-8.
  14. England M., A colour atlas of life before birth, in Normal fetal development, W. Medical, Editor. 1983: The Netherlands.
  15. Heyns C.F., The gubernaculum during testicular descent in the human fetus. Journal of Anatomy., 1987. 153: p. 93-112.
  16. Hutson J. and Hasthorpe S., Testicular descent and cryptorchidism: the state of the art in 2004. J Pediatr Surg, 2005. 40: p. 297-302.
  17. Park J.I., Semyonov J., Chang C.L., et al., Origin of INSL3-mediated testicular descent in therian mammals. Genome Res, 2008. 18(6): p. 974-85.
  18. Feng S., Ferlin A., Truong A., et al., INSL3/RXFP2 signaling in testicular descent. Ann N Y Acad Sci, 2009. 1160: p. 197-204.
  19. Zimmermann S., Steding G., Emmen J.M., et al., Targeted disruption of the Insl3 gene causes bilateral cryptorchidism. Mol Endocrinol, 1999. 13(5): p. 681-91.
  20. Nef S. and Parada L.F., Cryptorchidism in mice mutant for Insl3. Nat Genet, 1999. 22(3): p. 295-9.
  21. Emmen J.M., McLuskey A., Adham I.M., et al., Hormonal control of gubernaculum development during testis descent: gubernaculum outgrowth in vitro requires both insulin-like factor and androgen. Endocrinology, 2000. 141(12): p. 4720-7.
  22. Kubota Y., Temelcos C., Bathgate R.A., et al., The role of insulin 3, testosterone, Mullerian inhibiting substance and relaxin in rat gubernacular growth. Mol Hum Reprod, 2002. 8(10): p. 900-5.
  23. Yamanaka J., Metcalfe S.A., Hutson J.M., et al., Testicular descent. II. Ontogeny and response to denervation of calcitonin gene-related peptide receptors in neonatal rat gubernaculum. Endocrinology, 1993. 132(1): p. 280-4.
  24. Momose Y., Griffiths A.L. and Hutson J.M., Testicular descent. III. The neonatal mouse gubernaculum shows rhythmic contraction in organ culture in response to calcitonin gene-related peptide. Endocrinology, 1992. 131(6): p. 2881-4.
  25. Welsh M., Saunders P.T., Fisken M., et al., Identification in rats of a programming window for reproductive tract masculinization, disruption of which leads to hypospadias and cryptorchidism. J Clin Invest, 2008. 118(4): p. 1479-1490.
  26. Nation T., Balic A., Buraundi S., et al., The antiandrogen flutamide perturbs inguinoscrotal testicular descent in the rat and suggests a link with mammary development. Journal of Pediatric Surgery, 2009. 44(12): p. 2330-2334.
  27. Tomiyama H., Sasaki Y., Huynh J., et al., Testicular descent, cryptorchidism and inguinal hernia: the Melbourne perspective. J Pediatr Urol, 2005. 1(1): p. 11-25.
  28. Balic A., Nation T., Buraundi S., et al., Hidden in plain sight: the mammary line in males may be the missing link regulating inguinoscrotal testicular descent. J Pediatr Surg. 45(2): p. 414-8; discussion 418.
  29. Nation T.R., Buraundi S., Balic A., et al., The effect of flutamide on expression of androgen and estrogen receptors in the gubernaculum and surrounding structures during testicular descent. Journal of Pediatric Surgery, 2011. 46(12): p. 2358-62.
  30. Balic A., Hidden in plain sight:the mammary line in males may be the missing link regulating inguinoscrotal testicular descent. J Pediatr Surg, 2009. in press.
  31. Nightingale S.S., Western P. and Hutson J.M., The migrating gubernaculum grows like a "limb bud". J Pediatr Surg, 2008. 43(2): p. 387-90.
  32. Tickle C., Patterning systems--from one end of the limb to the other. Dev Cell, 2003. 4(4): p. 449-58.
  33. Hutson J. and Beasley S., Descent of the Testis. Edward Arnold, London, 1992: p. 1-87.
  34. Husmann D.A. and Levy J.B., Current concepts in the pathophysiology of testicular undescent. Urology, 1995. 46(2): p. 267-76.
  35. Hutson J.M., Hasthorpe S. and Heyns C.F., Anatomical and functional aspects of testicular descent and cryptorchidism. Endocr Rev, 1997. 18(2): p. 259-80.
  36. Hadziselimovic F., Duckett J.W., Snyder H.M., 3rd, et al., Omphalocele, cryptorchidism, and brain malformations. J Pediatr Surg, 1987. 22(9): p. 854-6.
  37. Fallat M., Hersh J. and JM H., Theories on the relationship between cryptorchidism and arthrogryposos. Pediatr Surg Int, 1992. 7(271).
  38. Hutson J., Cryptorchidism in spina bifida andspinal cord transection: a clue to the mechanism of transinguinal descent of the testis. J Pediatr Surg, 1988. 23: p. 275-277.
  39. Kaplan L.M., Koyle M.A., Kaplan G.W., et al., Association between abdominal wall defects and cryptorchidism. J Urol, 1986. 136(3): p. 645-7.
  40. Sharpe R.M. and Skakkebaek N.E., Are oestrogens involved in falling sperm counts and disorders of the male reproductive tract? Lancet, 1993. 341(8857): p. 1392-5.
  41. Block K., Kardana A., Igarashi P., et al., In utero diethylstilbestrol (DES) exposure alters Hox gene expression in the developing mullerian system. Faseb J, 2000. 14(9): p. 1101-8.
  42. Bergh A. and Soder O., Studies of cryptorchidism in experimental animal models. Acta Paediatr, 2007. 96(5): p. 617-21.
  43. Johansen T., Non-union of testis and epididymis. Scand J Urol Nephrol., 1988. 22: p. 165-70.
  44. Gill B., Kogan S., Starr S., et al., Significance of epididymal and ductal anomalies associated with testicular maldescent. J Urol, 1989. 142(2 Pt 2): p. 556-8; discussion 572.
  45. Hakonsen L.B., Ernst A. and Ramlau-Hansen C.H., Maternal cigarette smoking during pregnancy and reproductive health in children: a review of epidemiological studies. Asian Journal of Andrology, 2014. 16(1): p. 39-49.
  46. Hackshaw A., Rodeck C. and Boniface S., Maternal smoking in pregnancy and birth defects: a systematic review based on 173 687 malformed cases and 11.7 million controls. Hum Reprod Update, 2011. 17(5): p. 589-604.
  47. Virtanen H.E. and Toppari J., Embryology and physiology of testicular development and descent. Pediatric endocrinology reviews : PER, 2014. 11 Suppl 2: p. 206-13.
  48. Browne D., The diagnosis of undescended testicle. British Medical Journal, 1938. 11: p. 92-7.
  49. Dogruyol H. and Balkan E., Perineal ectopic testis. Br J Urol, 1992. 69(5): p. 547.
  50. Gendrel D., Roger M. and Job J.C., Plasma gonadotropin and testosterone values in infants with cryptorchidism. J Pediatr, 1980. 97(2): p. 217-20.
  51. Job J.C., Toublanc J.E., Chaussain J.L., et al., Endocrine and immunological findings in cryptorchid infants. Horm Res, 1988. 30(4-5): p. 167-72.
  52. Hadziselimovic F. and Hoecht B., Testicular histology related to fertility outcome and postpubertal hormone status in cryptorchidism. Klinische Pädiatrie, 2008. 220(5): p. 302-307.
  53. Baker B.A., Morley R. and Lucas A., Plasma testosterone in preterm infants with cryptorchidism. Arch Dis Child, 1988. 63(10): p. 1198-200.
  54. Brown T., Berkovits G. and Gearheart J., Androgen receptors in boys with isolated bilateral cryptorchidism. Am J Dis Child, 1988. 142: p. 933.
  55. Baker M., Metcalfe S. and Hutson J., Serum levels of Mullerian inhibiting substance in boys from birth to 18 years, as determined by enzyme immunoassay. Clin Endocr Metab 1990. 70: p. 11-15.
  56. Yamanaka J., Baker M., Metcalfe S., et al., Serum levels of Mullerian inhibiting substance in boys with cryptorchidism. J Pediatr Surg, 1991. 26(5): p. 621-3.
  57. Thorup J., Clasen-Linde E., Thorup S.C., et al., Pre- and postoperative status of gonadotropins (FSH and LH) and inhibin-B in relation to testicular histopathology at orchiopexy in infant boys with unilateral undescended testes. J Pediatr Urol, 2015. 11(1): p. 25 e1-5.
  58. Huff D.S., Hadziselimovic F., Snyder H.M., 3rd, et al., Postnatal testicular maldevelopment in unilateral cryptorchidism. J Urol, 1989. 142(2 Pt 2): p. 546-8; discussion 572.
  59. Huff D.S., Hadziselimovic F., Snyder H.M., 3rd, et al., Early postnatal testicular maldevelopment in cryptorchidism. J Urol, 1991. 146(2 ( Pt 2)): p. 624-6.
  60. Huff D.S., Hadziselimovic F., Snyder H.M., 3rd, et al., Histologic maldevelopment of unilaterally cryptorchid testes and their descended partners. Eur J Pediatr, 1993. 152 Suppl 2: p. S11-4.
  61. Virtanen H.E., Bjerknes R., Cortes D., et al., Cryptorchidism: classification, prevalence and long-term consequences. Acta Paediatr, 2007. 96(5): p. 611-6.
  62. Sharpe R.M., Perinatal determinants of adult testis size and function. J Clin Endocrinol Metab, 2006. 91(7): p. 2503-5.
  63. Zhou B., Watts L.M. and Hutson J.M., Germ cell development in neonatal mouse testes in vitro requires mullerian inhibiting substance. J Urol, 1993. 150(2 Pt 2): p. 613-6.
  64. Su S., Szarek M., Vooght A., et al., Gonocyte transformation to spermatogonial stem cells occurs earlier in patients with undervirilisation syndromes. Journal of Pediatric Surgery, 2014. 49(2): p. 323-7.
  65. Hutson J., Undescended testes, in Pediatric Surgery and Urology:Longterm outcomes, M.P. Oldham K, Editor. 2006, University Press: Cambridge. p. 652-663.
  66. Giwercman A., Bruun E., Frimodt-Moller C., et al., Prevalence of carcinoma in situ and other histopathological abnormalities in testes of men with a history of cryptorchidism. J Urol, 1989. 142(4): p. 998-1001: discussion 1001-2.
  67. Giwercman A., Muller J. and Skakkebaek N.E., Carcinoma in situ of the testis: possible origin, clinical significance, and diagnostic methods. Recent Results Cancer Res, 1991. 123: p. 21-36.
  68. Hutson J.M., Li R., Southwell B.R., et al., Germ cell development in the postnatal testis: the key to prevent malignancy in cryptorchidism? Frontiers in endocrinology, 2012. 3: p. 176.
  69. Vikraman J., Hutson J.M., Li R., et al., The undescended testis:clinical management and scientific advances. unpublished, 2015.
  70. Beltran-Brown F. and Villegas-Alvarez F., Clinical classification for undescended testes: experience in 1,010 orchidopexies. J Pediatr Surg, 1988. 23(5): p. 444-7.
  71. Hrebinko R.L. and Bellinger M.F., The limited role of imaging techniques in managing children with undescended testes. J Urol, 1993. 150(2 Pt 1): p. 458-60.
  72. Cortes D., Thorup J.M., Lenz K., et al., Laparoscopy in 100 consecutive patients with 128 impalpable testes. Br J Urol, 1995. 75(3): p. 281-7.
  73. Thorsson A.V., Christiansen P. and Ritzen M., Efficacy and safety of hormonal treatment of cryptorchidism: current state of the art. Acta Paediatr, 2007. 96(5): p. 628-30.
  74. Ritzen E.M., Bergh A., Bjerknes R., et al., Nordic consensus on treatment of undescended testes. Acta Paediatr, 2007. 96(5): p. 638-43.
  75. Rajfer J., Handelsman D.J., Swerdloff R.S., et al., Hormonal therapy of cryptorchidism. A randomized, double-blind study comparing human chorionic gonadotropin and gonadotropin-releasing hormone. N Engl J Med, 1986. 314(8): p. 466-70.
  76. deMuinck Keizer-Schrama S., Hazebroek F.W., Matroos A.W., et al., Double-blind, placebo-controlled study of luteinising-hormone-releasing-hormone nasal spray in treatment of undescended testes. Lancet, 1986. 1(8486): p. 876-80.
  77. Bica D.T. and Hadziselimovic F., Buserelin treatment of cryptorchidism: a randomized, double-blind, placebo-controlled study. J Urol, 1992. 148(2 Pt 2): p. 617-21.
  78. Mieusset R., Fonda P., Vaysse P., et al., Increase in testicular temperature in case of cryptorchidism in boys. Fertility and Sterility, 1993. 59: p. 1319-1321.
  79. Zhou B., Hutson J.M. and Hasthorpe S., Efficacy of orchidopexy on spermatogenesis in the immature mutant 'trans-scrotal' rat as a cryptorchid model by quantitative cytological analysis. Br J Urol, 1998. 81(2): p. 290-4.
  80. Kollin C., Hesser U., Ritzen E.M., et al., Testicular growth from birth to two years of age, and the effect of orchidopexy at age nine months: a randomized, controlled study. Acta Paediatr, 2006. 95(3): p. 318-24.
  81. Kollin C. and Ritzen E.M., Cryptorchidism: a clinical perspective. Pediatric endocrinology reviews : PER, 2014. 11 Suppl 2: p. 240-50.
  82. Wilson-Storey D., McGenity K. and Dickson J.A., Orchidopexy: the younger the better? J R Coll Surg Edinb, 1990. 35(6): p. 362-4.
  83. Ein S.H., Nasr A., Wales P.W., et al., Testicular atrophy after attempted pediatric orchidopexy for true undescended testis. J Pediatr Surg, 2014. 49(2): p. 317-22.
  84. Elder J.S., Two-stage Fowler-Stephens orchiopexy in the management of intra-abdominal testes. J Urol, 1992. 148(4): p. 1239-41.
  85. Pintus C., Coppola R., Talamo M., et al., Laparoscopic approach for nonpalpable abdominal testis. Surg Laparosc Endosc, 1997. 7(2): p. 156-8.
  86. Samadi A.A., Palmer L.S. and Franco I., Laparoscopic orchiopexy: report of 203 cases with review of diagnosis, operative technique, and lessons learned. J Endourol, 2003. 17(6): p. 365-8.
  87. Lee P.A., O'Leary L.A., Songer N.J., et al., Paternity after bilateral cryptorchidism. A controlled study. Arch Pediatr Adolesc Med, 1997. 151(3): p. 260-3.
  88. Lee P.A., O'Leary L.A., Songer N.J., et al., Paternity after unilateral cryptorchidism: a controlled study. Pediatrics, 1996. 98(4 Pt 1): p. 676-9.
  89. Thorup J., Haugen S., Kollin C., et al., Surgical treatment of undescended testes. Acta Paediatr, 2007. 96(5): p. 631-7.
  90. Feyles F., Peiretti V., Mussa A., et al., Improved sperm count and motility in young men surgically treated for cryptorchidism in the first year of life. Eur J Pediatr Surg, 2014. 24(5): p. 376-80.
  91. Chilvers C., Dudley N.E., Gough M.H., et al., Undescended testis: the effect of treatment on subsequent risk of subfertility and malignancy. J Pediatr Surg, 1986. 21(8): p. 691-6.
  92. Moller H. and Skakkebaek N.E., Risks of testicular cancer and cryptorchidism in relation to socio-economic status and related factors: case-control studies in Denmark. Int J Cancer, 1996. 66(3): p. 287-93.
  93. Moller H., Prener A. and Skakkebaek N.E., Testicular cancer, cryptorchidism, inguinal hernia, testicular atrophy, and genital malformations: case-control studies in Denmark. Cancer Causes Control, 1996. 7(2): p. 264-74.
  94. Muller J. and NE S., Cryptorchidism. Current Therapy in Endocrinology and Metabolism, 1997. 6: p. 363-6.
  95. Swerdlow A.J., Higgins C.D. and Pike M.C., Risk of testicular cancer in cohort of boys with cryptorchidism. Bmj, 1997. 314(7093): p. 1507-11.
  96. Stone J.M., Cruickshank D.G., Sandeman T.F., et al., Laterality, maldescent, trauma and other clinical factors in the epidemiology of testis cancer in Victoria, Australia. Br J Cancer, 1991. 64(1): p. 132-8.
  97. Farrington G., The position and retractibility of the normal testis in childhood with reference to the diagnosis and treatment of cryptorchidism. J.Pediatr.Surg, 1968. 3: p. 53.
  98. Hutson J.M. and Goh D.W., Can undescended testes be acquired? Lancet, 1993. 341(8843): p. 504.
  99. Clarnette T.D., Rowe D., Hasthorpe S., et al., Incomplete disappearance of the processus vaginalis as a cause of ascending testis. J Urol, 1997. 157(5): p. 1889-91.
  100. Mayr J., Rune G.M., Holas A., et al., Ascent of the testis in children. Eur J Pediatr, 1995. 154(11): p. 893-5.
  101. John Radcliffe Hospital Cryptorchidism Study Group, Boys with late ascending testes:The source of patients with "rectractile" testes undergoing orchidopexy. BMJ, 1986. 293: p. 789.
  102. Hutson J.M., Treatment of undescended testes--time for a change in European traditions. Acta Paediatr, 2007. 96(5): p. 608-10.
  103. Fenton E., Woodward A. and Hutson J., The ascending testis. Pediatr Surg Int, 1990. 5: p. 6-9.
  104. Davey R.B., Undescended testes: early versus late maldescent. Pediatr Surg Int, 1997. 12(2/3): p. 165-7.
  105. Smith A., Hutson J., Beasley S., et al., The relationship between cerebral palsy and cryptorchidism. J. Pediatr. Surg., 1989. 24: p. 1303-1305.
  106. Clarnette T.D. and Hutson J.M., Is the ascending testis actually 'stationary'? Normal elongation of the spermatic cord is prevented by a fibrous remnant of the processus vaginalis. Pediatr Surg Int, 1997. 12(2/3): p. 155-7.
  107. Myers N. and CB O., Undescended testes: congenital or acquired? Aust.Paediatr.J, 1975. 11: p. 76-80.
  108. Schiffer K.A., Kogan S.J., Reda E.F., et al., Acquired undescended testes. Am J Dis Child, 1987. 141(1): p. 106-7.
  109. Atwell J.D., Ascent of the testis: fact or fiction. Br J Urol, 1985. 57(4): p. 474-7.
  110. Robertson J.F., Azmy A.F. and Cochran W., Assent to ascent of the testis. Br J Urol, 1988. 61(2): p. 146-7.
  111. Donaldson K.M., Tong S.Y. and Hutson J.M., Prevalence of late orchidopexy is consistent with some undescended testes being acquired. Indian J Pediatr, 1996. 63(6): p. 725-9.
  112. Bianchi A. and Squire B., Trans-scrotal orchidopexy:Orchidopexy revisited. Pediatr. Surg. Int, 1989. 4: p. 189.
  113. Hack W.W., Goede J., van der Voort-Doedens L.M., et al., Acquired undescended testis: putting the pieces together. Int J Androl, 2011. 35(1): p. 41-5.
  114. van Brakel J., Kranse R., de Muinck Keizer-Schrama S.M., et al., Fertility potential in a cohort of 65 men with previously acquired undescended testes. J Pediatr Surg, 2014. 49(4): p. 599-605.
  115. Puri P. and O'Donnell B., Semen analysis of patients who had orchidopexy at or after seven years of age. Lancet, 1988. 2(8619): p. 1051-2.
  116. Nistal M. and Paniagua R., Infertility in adult males with retractile testes. Fertil Steril, 1984. 41(3): p. 395-403.
  117. Pike M.C., Boys with late descending testes. Br Med J (Clin Res Ed), 1986. 293(6557): p. 1307-8.
  118. Penington E.C. and Hutson J.M., The urethral plate--does it grow into the genital tubercle or within it? BJU Int, 2002. 89(7): p. 733-9.
  119. Baskin L., Hypospadias and urethral development. The Journal of Urology, 2000. 163(3): p. 951-960.
  120. Rodriguez E., Jr., Weiss D.A., Ferretti M., et al., Specific morphogenetic events in mouse external genitalia sex differentiation are responsive/dependent upon androgens and/or estrogens. Differentiation, 2012. 84(3): p. 269-79.
  121. Stephens F.D., Smith E. and Hutson J., Congenital anomalies of the kidney, urinary and genitral tracts. 2nd ed. 2002, London: Martin Dunitz. 410-429.
  122. Baskins L., Hypospadias, in Pediatric Surgery 6th ed., O.N.J. Grosfeld JL, Fonkalsrud EW, Coran AG, Editor. 2006, Mosby Elsevier: Philadelphia. p. 1870-1898.
  123. Bauer S.B., Retik A.B. and Colodny A.H., Genetic aspects of hypospadias. Urol Clin North Am, 1981. 8(3): p. 559-64.
  124. Tong S.Y., Donaldson K. and Hutson J.M., When is hypospadias not hypospadias? Med J Aust, 1996. 164(3): p. 153-4.
  125. Belman A.B., De-epithelialized skin flap coverage in hypospadias repair. J Urol, 1988. 140(5 Pt 2): p. 1273-6.
  126. Keating M.A., Cartwright P.C. and Duckett J.W., Bladder mucosa in urethral reconstructions. J Urol, 1990. 144(4): p. 827-34.
  127. Mollard P., Mouriquand P. and Felfela T., Application of the onlay island flap urethroplasty to penile hypospadias with severe chordee. Br J Urol, 1991. 68(3): p. 317-9.
  128. Jones B.C., O'Brien M., Chase J., et al., Early hypospadias surgery may lead to a better long-term psychosexual outcome. J Urol, 2009. 182(4 Suppl): p. 1744-9.
  129. Schultz J.R., Klykylo W.M. and Wacksman J., Timing of elective hypospadias repair in children. Pediatrics, 1983. 71(3): p. 342-51.
  130. Baskins L., Hypospadias, in Pediatric Surgery and Urology:Long term outcomes, O.K. Stringer MD, Mouriquand PDE., Editor. 2006, University Press: Cambridge. p. 611-620.
  131. Secrest C.L., Jordan G.H., Winslow B.H., et al., Repair of the complications of hypospadias surgery. J Urol, 1993. 150(5 Pt 1): p. 1415-8.
  132. Scherz H.C., Kaplan G.W., Packer M.G., et al., Post-hypospadias repair urethral strictures: a review of 30 cases. J Urol, 1988. 140(5 Pt 2): p. 1253-5.

 

Androgens and Cardiovascular Disease in Men

ABSTRACT

 

Testosterone is the principal male sex hormone or androgen, which regulates sexual characteristics and body composition. Testosterone is converted to bioactive metabolites dihydrotestosterone and estradiol. Circulating testosterone peaks in early adulthood and declines gradually across middle and older age: older men exhibit lower testosterone and dihydrotestosterone concentrations compared to younger men. In older men, lower testosterone concentrations are associated with higher incidence of cardiovascular events. Lower testosterone and dihydrotestosterone concentrations have also been associated with higher cardiovascular mortality in older men. However, causation is unproven as a randomized placebo-controlled trial of testosterone treatment sufficiently powered to examine outcomes of cardiovascular events or mortality has yet to be reported. Potential mechanisms by which testosterone could exert beneficial actions in the vasculature include reduction in cholesterol accumulation and modulation of inflammation. Smaller randomized trials of testosterone therapy have shown improvements in surrogate endpoints related to cardiovascular risk. However, other trials of testosterone have not shown improvements in carotid atherosclerosis, as assessed by carotid intima-media thickness. One study reported an increase in coronary atheroma assessed using coronary computed tomography angiography in older men receiving testosterone therapy over 12 months. Although one randomized trial of testosterone therapy in older men with mobility limitations reported an excess of adverse events in the treatment arm, larger recent trials in middle-aged to older men did not find any excess of cardiovascular adverse events with testosterone treatment. Meta-analyses of testosterone trials generally have not shown an increase in cardiovascular adverse events. Retrospective case-control studies of health insurance databases have major methodological limitations. The results from such studies are inconsistent, associating testosterone prescriptions with either increased or decreased risk of cardiovascular events, and with lower mortality. While androgen deprivation in men with prostate cancer results in adverse metabolic effects, abuse of high dosages of androgenic steroids is associated with harm. Thus, while some epidemiological studies associate higher circulating concentrations (but within the normal range) of endogenous androgens with lower risk of cardiovascular events and mortality, the effects of exogenous androgens in the form of testosterone therapy seeking to maintain physiological circulating androgen concentrations on the cardiovascular system remain uncertain. This evidence gap has to be accommodated in the current clinical management of hypogonadal men and should be addressed by further randomized interventional studies to clarify whether testosterone treatment has beneficial, neutral or adverse effects on the cardiovascular system.

 

INTRODUCTION

 

Androgen Physiology

 

THE HYPOTHALAMIC-PITUITARY AXIS

 

Testosterone (T) is the principal male sex hormone or androgen that regulates sexual maturation and secondary sexual characteristics and body composition in adult men (1). T undergoes conversion into two major bioactive metabolites, dihydrotestosterone (DHT) a more potent ligand for the androgen receptor, and estradiol (E2), a ligand for estrogen receptors (2). T is produced primarily by the testis, under stimulation of luteinizing hormone (LH) from the pituitary gland, itself under regulation of gonadotrophin releasing hormone (GnRH) from the hypothalamus. The hypothalamic-pituitary-testicular (HPT) axis is under negative feedback regulation via T and E2, acting on the central components of the HPT axis (1,3-5).

 

CONSEQUENCES OF ANDROGEN DEFICIENCY    

 

Androgens play diverse roles in the body, and androgen deficiency in men results in multiple symptoms and signs extending from loss of libido, lethargy, fatigue, poor concentration to gynecomastia, accumulation of fat, loss of muscle mass, and osteopenia or osteoporosis (6,7). Actions of T are amplified by local conversion to DHT in tissues such as the prostate and skin by the enzyme 5α-reductase (8). Of note, some of actions of T, such as in bone and adipose tissue, are mediated via conversion to E2 by the enzyme aromatase (9-11).

 

MEASUREMENT OF CIRCULATING ANDROGEN CONCENTRATIONS

 

Immunoassays have been the standard method for measurement of circulating sex hormones: however, these can exhibit non-specificity and method-dependent bias, particularly at lower hormone concentrations (12,13). Mass spectrometry is regarded as the gold standard for assay of T concentrations (14). Although mass spectrometry is preferred, it is not widely available and validated immunoassays can be informative (6,7). In the circulation, T (and DHT and E2) are bound with high affinity to sex hormone-binding globulin (SHBG), T is also bound with lower affinity to albumin with a small fraction unbound or free (15). However, whether unbound or “free” T represents a more biologically active form of the hormone in vivo is controversial (16). A major difficulty is that measurement of free T using equilibrium dialysis is technically demanding and thus rarely performed. Instead, free T is typically calculated, and this result may vary according to the equations used (15-18). Furthermore, established reference ranges for free T are lacking (7,19). Thus, analysis of actions of T in the male body and cardiovascular system involves consideration of not only its bioactive metabolites, but also the accuracy of assays used and the limitations of calculated free T as a biomarker.

 

Androgens and Male Ageing

 

DECLINE IN CIRCULATING ANDROGENS WITH MALE AGING

 

Circulating T peaks in early adulthood and declines gradually across middle and older age, thus older men exhibit lower T and DHT concentrations compared to younger men (20-22). Observational studies have shown longitudinal declines in T and DHT in ageing men, with parallel increases in LH and SHBG (23-25). This phenomenon suggests that in some men, there is progressive impairment of testicular endocrine function with ageing (25). However, whether older men with symptoms consistent with androgen deficiency and lower circulating concentrations of T compared to younger men, have an androgen deficiency state remains unclear (26,27). The bioactive metabolites of T, DHT and E2 measured with mass spectrometry have been associated with longer leucocyte telomere length, a measure of slower biological ageing, in middle-aged and older men (28,29). However, a study in mostly middle-aged men showed no association between T measured with immunoassay and leucocyte telomere length (30). Thus, there is considerable interest in the question of whether lower T concentrations might contribute to various manifestations of ill health in ageing men.

 

CONTROVERSIES OVER FUNCTIONAL HYPOGONADISM

 

Men with disorders of the hypothalamus, pituitary, or testes resulting in hypogonadism have symptoms and signs of androgen deficiency, and low circulating T concentrations (6). These men are classified as having pathological (or classic or organic) hypogonadism, and T treatment is routinely offered to improve symptoms and restore body composition (6,7). However, low circulating T concentrations are found in many conditions where the HPT axis is intact, including ageing, obesity, and systemic illnesses (7,31). In this context, low T concentrations result from reduction or suppression of HPT axis function, rather than an intrinsic disorder of the HPT axis, with the label of “functional hypogonadism” applied (7,31). Obesity is closely associated with reduced circulating T and SHBG, and loss of excess weight restores endogenous production of T (32-35). Thus, low T might be a biomarker for the presence of systemic illnesses, rather than a contributing or causal factor. Whether or not, and if so which, men with functional hypogonadism should receive T treatment, remains the subject of debate (7,19,20).

 

Androgens and Risks of Cardiovascular Disease

 

AGE AND OBESITY AS COMMON RISK FACTORS

 

Advancing age is an established risk factor for chronic diseases including cardiovascular disease (CVD) and mortality (36,37). Age is a component of all major cardiovascular risk calculators (38). Similarly, obesity, with its close associations with insulin resistance and diabetes risk, is a robust cardiovascular risk factor, and - unlike age - is a potentially modifiable one, whether via bariatric surgery or incretin-based medical therapy (41-44). This is illustrated by observational studies which show a reduction in cardiovascular events and mortality following bariatric surgery in patients who are obese (mean age 48 years) (41,42). Thus, advancing age and obesity are both associated with low circulating T concentrations, and are also risk factors for CVD. 

 

CONFLUENCE OF AGE, OBESITY, LOW TESTOTERONE AND CARDIOVASCULAR DISEASE

 

Given that age and obesity are associated with both low circulating T concentrations and increased risk of CVD, understanding the relationship between these factors becomes vitally important (45). Demographic change will result in increasing numbers of older men in communities worldwide, who will have lower circulating T concentrations and who are at risk of ill health including from CVD (36). If low T contributes to CVD risk, either directly or via its association with obesity, then this represents a potential pathway for intervention to preserve health in ageing men. Conversely, if T is a biomarker for CVD, it may still have a role in risk stratification and identification of men at risk who may benefit from non-hormonal interventions directed at conventional risk factors for CVD.

 

EPIDEMIOLOGICAL STUDIES

 

Associations of Androgens with Cardiovascular Events

 

PROSPECTIVE COHORT STUDIES: IMMUNOASSAY RESULTS

 

Prospective cohort studies report the association of endogenous sex hormones with incidence of cardiovascular events (Table 1). Analyses invariably adjust for age, and typically also include adjustment for body mass index (or waist circumference) and other conventional cardiovascular risk factors. From studies reporting sex hormone results based on immunoassays, some longitudinal analyses have shown no association of total T concentrations with incidence of myocardial infarction (MI) or ischemic heart disease (IHD) events (46,49,53,63,64). In an analysis of 3,443 men aged ≥70 years from the Western Australian Health In Men Study (HIMS), low total T concentrations were associated with an increased incidence of stroke (50). This finding was confirmed by later studies (57,59). In another analysis from HIMS, higher LH was associated with incident IHD (52). One smaller study reported a U-shaped association of total T with incidence of cardiovascular events (54). There were conflicting associations of total E2 with stroke (47,48), and there was no association of total E2 with the incidence of MI (49). In one study, men with lower total T had a higher risk of heart failure, but this was not confirmed in another study (65,68).

 

Table 1. Cohort Studies Examining Associations Between Sex Hormones with Cardiovascular Events in Middle-Aged and Older Men

Study author and year

Size (n of men)

Follow-up (yr)

Age (yr)

Summary of results

Smith GD, 2005 (46)

2,512

16.5

45-59

No association of total T with IHD events or deaths.

Arnlov J, 2006 (47)

2,084

10

56

Higher total E2 at baseline associated with lower incidence of CVD events, total T was not associated.

Abbott RD, 2007 (48)

2,197

≤7

71-93

Baseline total E2 in top quintile (≥125 pmol/L) associated with higher risk of stroke, total T was not associated.

Vikan T, 2009 (49)

1,568

≤13

59.6

No association of total T or E2 with incident MI, or with CVD or IHD mortality.

Yeap BB, 2009 (50)

3,443

3.5

≥70

Total and free T in the lowest quartiles (<11.7 nmol/liter and <222 pmol/liter) predicted increased incidence of stroke or TIA.

* Ohlsson C, 2011 (51)

2,416

5

69-81

Men with total Ta in highest quartile (≥19 mol/L) had lower risk of CVD events. E2 was not associated.

Hyde Z, 2011 (52) 

3,637

5.1

70-88

Higher LH was associated with incident IHD.

Haring R, 2013 (53)

254

5, 10

75.5

No associations of baseline total T or total E2 with incident CVD events.

Soisson V, 2013 (54)

495; 146

4

>65

Total T in lowest and highest quintiles associated with CHD or stroke.

* Shores MM, 2014 (55)

1,032

9

76

DHTb <1.7 or >2.6 nmol/L associated with cardiovascular events. Total T was not associated.

* Shores MM, 2014 (56)

1,032

10

76

Non-linear association of DHTb with stroke with lowest risk in men with DHT 1.7-2.6 nmol/L. Total Tb was not associated with stroke. DHT <0.86 nmol/L associated with CVD mortality.

* Yeap BB, 2014 (57)

3,690

6.6

70-89

Higher total Tc (>12.6 nmol/L) or DHT (>1.34 nmol/L) associated with lower incidence of stroke. Tc, DHT and E2 were not associated with MI.

* Srinath R, 2015 (58)

1,558

12.8

63.1

Td was not associated with incidence of CHD events, or cardiac-related mortality.

Holmegard HN, 2016 (59)

4,602

20

57

Total T in lowest decile (0-10thpercentile) associated with stroke.

* Chan YX, 2016 (60)

1,804

14.9

50.3

Total Tc, DHT and E2 were not associated with CVD events.

* Srinath R, 2016 (61)

1,558

14.1

63.1

Td was not associated with stroke.

Wang A, 2019 (62)

5,553

6

63.5

Neither total T nor free T were associated with CVD events.

* Gyawali P, 2019 (63)

1,492

4.9

54.2

Higher total Tc associated with lower risk of incident CVD events, but not with CVD mortality. E2 not associated.

Hatami H, 2020 (64)

816

12

46.1

Total T was not associated with risk of CVD events.

Zhao D, 2020 (65)

4,107

19.2

63.2

Lower total T associated with increased risk of incident heart failure.

* Collett T-H, 2020 (66)

552

7.4

72.4

Total Td not associated with CVD events.

* Boden WE, 2020 (67)

2,118

3

≥40

643 men with total Tb <10.4 nmol/L had higher risk of combined endpoint of CHD death, MI or stroke, compared with 1,475 men with total T ≥10.4 nmol/L.

Shafer S, 2021 (68)

3,865

13.8

48.2

Lower total T not associated with incident heart failure.

Yeap BB, 2022 (69)

210,700

9

58

Total T not associated with incident MI, stroke, heart failure or MACE. Calculated free T not associated with incident MI, stroke or heart failure, but associated with incidence of MACE.

IHD=ischemic heart disease, CVD=cardiovascular disease, MI=myocardial infarction, TIA=transient ischemic attack, CHD=coronary heart disease, MACE=major cardiovascular adverse event. * denotes studies where total T, DHT and/or E2 were measured using mass spectrometry. aT and E2 assayed using gas chromatography-mass spectrometry (GC-MS), bT and DHT assayed using liquid chromatography-tandem mass spectrometry (LC-MS), cT, DHT and E2 assayed by LC-MS, dT assayed using LC-MS

 

Recently the association of testosterone with CVD events was examined in the largest prospective cohort study to date, the United Kingdom (UK) Biobank (69). In this study of 210,700 men aged 40-69 years at baseline, with 9 years follow-up, 8,790 had an incident CVD event. Total T was not associated with risk of incident MI, ischemic stroke, hemorrhagic stroke, heart failure, nor major cardiovascular adverse events (MACE) defined as the composite endpoint of non-fatal MI, non-fatal ischemic stroke, and CVD death. The large size of the UK Biobank, and accumulation of outcome events over the period of follow-up, provided power to examine these associations in a robust fashion. UK Biobank used an immunoassay for measurement of serum T which may underestimate results compared to mass spectrometry (70), and UK Biobank men were generally healthier than the UK male population as a whole (71). Therefore, while these results are convincing, their generalizability to different populations in other regions needs to be established.

 

PROSPECTIVE COHORT STUDIES: MASS SPECTROMETRY RESULTS

 

Prospective cohort studies where sex hormones were measured using mass spectrometry are shown (Table 1, marked with *). In the Osteoporotic Fractures in Men Study in Sweden (MrOS), a large prospective cohort study of 2,416 men aged 69-81 years, men with higher total T had a lower incidence of CVD events (51). In HIMS, a later analysis of 3,690 men aged 70-89 years with sex hormones measured using mass spectrometry confirmed the association of low total T with higher incidence of stroke (57). In these analyses, total E2 was not associated with these outcomes (51,57). Analyses from the Cardiovascular Health Study (CHS) of 1,032 men aged 76 years suggested a U-shaped association of DHT with CVD events and stroke risk (55,56). Of note, an analysis from the Busselton Health Study (BHS) of 1,804 predominantly middle-aged men found no association of sex hormones with incidence of CVD events (60). Analyses from the Atherosclerosis Risk in Communities Study of 1,558 men aged 63 years found no association of sex hormones with CVD events (58,61), similar to the findings from a small subset of the MrOS USA study (66). By contrast, in the Men Androgen Inflammation Lifestyle Environment and Stress (MAILES) cohort of 1,492 men followed for 4.9 years, higher total T was associated with a lower risk of CVD events (63). Furthermore, in a post-hoc analysis of 2,118 men with metabolic syndrome participating in a trial of niacin or placebo plus simvastatin, men who had a baseline total T <10.4 nmol/L had higher risk of the combined endpoint of coronary heart disease death, MI or stroke, compared with men with higher total T concentrations (65).

 

SUMMARY: ENDOGENOUS SEX HORMONES VS. CARDIOVASCULAR DISEASE

 

Taken together, these epidemiological studies suggest that there may be an association of lower endogenous T concentrations with increased risk of CVD events in middle-aged and older men. However, the studies provide a mix of positive, equivocal and negative results. Major cohort studies using mass spectrometry for assay of sex hormones associated higher total T concentrations with lower risk of CVD events in older (51,57) and middle-aged to older men (63,67). There may be a predilection for lower testosterone, measured by mass spectrometry, to be associated with stroke risk (57). In one study lower DHT was associated with CVD events including stroke, with a non-linear association (55,56). Inconsistent results in other studies may have been due to smaller cohort sizes and fewer outcome events reducing the power available to detect underlying associations. However, in the largest ever prospective cohort study, the UK Biobank, there was no association of testosterone with a range of CVD events (69). Overall, epidemiological studies would suggest a possible protective effect of endogenous androgens against CVD events in the older population of men, rather than in relatively healthy middle-aged men.

 

Associations of Androgens with Cardiovascular Mortality

 

PROSPECTIVE COHORT STUDIES: IMMUNOASSAY RESULTS

 

Several of the studies in Table 1 reported CVD-related mortality in addition to events. Other studies where the outcome was based on CVD-related mortality are summarized in Table 2. Studies almost invariably adjusted for age, and typically adjusted for BMI and other cardiovascular risk factors. In two studies using immunoassay for assay of sex hormones, T was not associated with CVD or ischemic heart disease (IHD) mortality (46,49). However, several other studies using immunoassay for sex hormones did find associations of lower endogenous total T concentrations with increased risk of CVD-related death (72,74,77,81). Lower calculated free T was also associated with increased CVD-related mortality in some studies (76,78,81), but was associated with lower IHD mortality in one study (73). Lower total E2 was associated with CVD mortality in one study (76). In an analysis from the UK Biobank of 149,436 men followed for 11.3 years, there was no association of either total or calculated free T with risk of CVD mortality (84).

 

Table 2. Cohort Studies Examining Associations Between Sex Hormones and CVD-Related Mortality in Middle-Aged and Older Men

Study author and year

Size (n of men)

Follow-up (yr)

Age (yr)

Summary of results

Khaw K-T, 2007 (72)

825 and 1489

≤10

40-79

Total T inversely related to mortality from all causes, CVD and cancer.

Araujo AB, 2007 (73)

1,686

15.3

40-70

Lower free T associated with lower IHD mortality. Equivocal association of lower DHT with IHD mortality.

Laughlin GA, 2008 (74)

794

11.8

50-91

Total T in the lowest quartile (<8.4 nmol/L) predicted increased mortality from all causes and from CVD and respiratory causes.

* Tivesten A, 2009 (75)

3,014

4.5

75

Total Ta and E2 levels in the lowest quartiles predicted all-cause and non-CVD mortality. T and E2 were not associated with CVD mortality.

Menke A, 2010 (76)

1,114

18

≥20

Lower free T associated with overall and CVD mortality in first 9 years of follow-up. Lower total E2 associated with CVD mortality. (Difference between 90th and 10thpercentiles for free T and total E2)

Haring R, 2010 (77)

1,954

7.2

20-79

Total T <8.7 nmol/L associated with increased all-cause, CVD and cancer mortality.

Hyde Z, 2012 (78)

3,637

5.1

70-88

Lower free T (100 vs 280 pmol/L) predicted all-cause and CVD mortality.

* Yeap BB, 2014 (79)

3,690

7.1

70-89

Optimal total Tb (9.8-15.8 nmol/L) predicted lower all-cause mortality. Higher DHT (>1.3 nmol/L) predicted lower IHD mortality. E2 was not associated with mortality.

* Pye SR, 2014 (80)

2,599

4.3

40-79

Presence of sexual symptoms and total Tc <8 nmol/L associated with all-cause and CVD mortality, total or free T not associated.

Holmboe SA, 2015 (81)

5,323

18.5

30-70

Higher T or free T (highest vs lowest quartile) associated with lower CVD mortality.

* Hsu B, 2016 (82)

1,705, 1,367 and 958

0, 2 and 5

≥70

Decrease in total Td over time associated with all-cause but not CVD mortality. Decrease in total E2d was associated with all-cause and CVD mortality.

* Chasland L, 2017 (83)

1,649

20

49.8

Higher physical activity and total Tb, DHT and E2 were not associated with CVD events. Men with higher physical activity and DHT had the lowest risk of CVD death. Men with lower physical activity and higher E2 had greater risk of CVD death.

Yeap BB, 2021 (84)

149,436

11.3

58.0

Men with lower total T had higher all-cause and cancer-related mortality, no association with CVD deaths.

IHD=ischemic heart disease, CVD=cardiovascular disease, CHD=coronary heart disease. * denotes studies where total T, DHT and/or E2 were measured by mass spectrometry; free T was calculated. aT and E2 measured using gas chromatography-mass spectrometry (GC-MS). bT, DHT and E2 measured using liquid chromatography-tandem mass spectrometry (LC-MS), cT measured using GC-MS, dT and E2 measured using LC-MS.

 

PROSPECTIVE COHORT STUDIES: MASS SPECTROMETRY RESULTS

 

Prospective cohort studies using mass spectrometry for assay of sex hormones are of interest. In an analysis from MrOS in Sweden, lower endogenous total T and E2 concentrations were associated with all-cause and non-CVD mortality, but not with CVD mortality (75). Interestingly, in an analysis from HIMS of 3,690 men aged 70-89 years at baseline, optimal endogenous total T concentrations were associated with survival, and higher DHT predicted lower IHD mortality (79). The CHS study reported consistent findings with lower DHT concentrations being associated with CVD mortality (56). An analysis from the European Male Ageing Study found that the combination of sexual symptoms and lower total T was associated with all-cause and CVD mortality, rather than total T or free T on their own (80). In an analysis from the Concord Health and Ageing in Men Project (CHAMP), longitudinal decreases in total T, DHT or E2 were associated with all-cause mortality, but only the longitudinal decrease in total E2 was predictive of CVD mortality (82). Finally, in an analysis from BHS in which physical activity and sex hormones concentrations were analyzed, men with higher levels of physical activity and higher DHT concentrations had the lowest risk of CVD death (83).

 

SUMMARY: ENDOGENOUS SEX HORMONES VS. CARDIOVASCULAR MORTALITY

 

Several cohort studies have reported an association between lower endogenous T concentrations and increased mortality related to CVD, after adjusting for age and other cardiovascular risk factors. Of large studies using mass spectrometry for assay of sex steroids, MrOS in Sweden found an association of lower total T and E2 with all-cause rather than CVD mortality, while HIMS found an optimal total T to be associated with survival (75,79). HIMS found higher DHT was associated with lower IHD mortality (79), consistent with results from CHS (56), and in BHS the combination of higher DHT and higher levels of physical activity was associated with lower risk of death from CVD (83). Declining E2 may also have a role, being associated with CVD mortality in CHAMP (82). However, in relatively healthy middle-aged men (UK Biobank), there was no evidence of an association between total T and CVD mortality risk (84).

 

Therefore, allowing for some heterogeneity in cohort characteristics and results, lower endogenous T concentrations, measured using mass spectrometry, may be predictors of CVD related deaths in older men, as might lower or declining concentrations of its bioactive metabolites DHT and E2. However, this may not be the case in generally healthy middle-aged men. Whether lower concentrations of endogenous sex hormones are biomarkers or possibly contributing factors to these outcomes remains unclear from these observational studies, as proof of causality ultimately requires interventional studies and randomized controlled trials (RCTs).

 

MECHANISTIC STUDIES

 

Potential Mechanisms

 

Knowledge of potential mechanisms by which androgens might exert protective effects against atherosclerosis and reduce the risk of cardiovascular events would bridge the findings from epidemiological studies and clinical investigation. There are a substantial number of such studies with diverse models and results, a comprehensive discussion of each being beyond the scope of this chapter (for reviews, see (85,86)). Selected studies are discussed briefly in this context.

 

Cholesterol Accumulation in Animal Models

 

Experimental studies in castrated male rabbits fed a high cholesterol diet reported effects of testosterone treatment to reduce accumulation of cholesterol in the aortic wall and to reduce atheromatous plaque area and aortic intimal thickness (87-90). Similar results have also been reported in miniature pigs (91). Castration of low-density lipoprotein receptor (LDLR)-deficient male mice results in increased fatty steak lesion formation in the aorta compared to non-castrated controls, which is attenuated with testosterone supplementation (92). At least part of this effect may be mediated via conversion of T to E2. Of note, T and DHT increased calcification of plaque in apolipoprotein E (ApoE)-null mice, even as T had a neutral effect on plaque volume and DHT decreased plaque volume (93). In testicular feminized mice with a non-functional androgen receptor (AR) and low circulating T concentrations, T supplementation to physiological levels reduced fatty streak formation (94). Similarly, AR knockout mice (ARKO) showed increased aortic atherosclerosis, and atherosclerotic lesion area that was reduced with T treatment (95). In wild-type mice, T treatment reduced the presence of necrotic cores within plaque compared with placebo. Therefore, these animal studies suggest an effect of T treatment in reducing cholesterol accumulation and the development of atheromatous plaque, while increasing calcification. However, the actions of sex hormones are complex, being mediated partly via aromatization of T to E2, and occurring at least to an extent via AR-independent mechanisms.

 

Neointimal Formation and Vascular Smooth Muscle Proliferation

 

NEOINTIMAL RESPONSES TO INJURY

 

In a male rabbit aorta model of neointimal plaque formation induced by endothelial denudation, T treatment in vitroinhibited plaque development (96). In a male porcine model of coronary neointimal plaque formation following moderate angioplasty-induced arterial injury, castrated males exhibited greater intimal area compared to intact males and castrated males treated with T (97). T inhibited proliferation and increased expression of the cell-cycle regulator p27kip1 during neointimal formation. However, despite castration of wild-type mice resulting in increased neointimal formation following wire injury, selective deletion of AR from endothelial cells or smooth muscle cells did not affect lesion size (98). Therefore, effects of T on neointimal formation may be indirect, or mediated by AR-independent mechanisms.

 

VASCULAR SMOOTH MUSCLE 

 

Vascular smooth muscle cells contribute to progression of atherosclerotic lesions and formation of the fibrous cap (99). T was shown to regulate expression of proliferation-associated genes in skeletal myocytes and in myofibers in different muscles (100,101). Its role in smooth muscle cells in the vasculature is not well defined. In one study, T exerted a pro-proliferative effect on vascular smooth muscle cells in vitro, with increased DNA synthesis assessed using a thymidine incorporation assay (102). In that study the effect of T was blocked by the AR antagonist flutamide. In another study, T induced apoptosis in cultured vascular smooth muscle cells, in an AR-dependent manner (103). In one study, deletion of the AR in vascular smooth muscle cells did not change atherosclerotic plaque size in LDLR knockout mice (104). However, another study demonstrated that T, acting via the AR in vascular smooth muscle cells, might be involved in promoting vascular calcification (105). Therefore, T seems to exert indirect effects on neointimal proliferation in response to injury and may play a secondary role in the development and calcification of atheromatous plaque via complex actions in vascular smooth muscle.

 

Inflammation

 

INFLAMMATION AND ATHEROTHROMBOSIS

 

A mechanistic link likely exists between inflammation and atherothrombosis (for reviews, see (106,107)). Statin therapy lowered both LDL cholesterol and C-reactive protein (CRP) concentrations, reducing the risk of cardiovascular events in a primary prevention setting in adults with LDL <3.4 mmol/L and high-sensitivity CRP ≥2.0 mg/L (108). Recently, anti-inflammatory intervention utilizing canakinumab, a monoclonal antibody targeting interleukin-1β, in a secondary prevention setting in adults with high-sensitivity CRP ≥2.0 mg/L demonstrated a modest reduction in major cardiovascular events (109). However, a major trial using low-dose methotrexate showed no benefit in adults with previous MI or multivessel coronary artery disease and either type 2 diabetes or metabolic syndrome (110). By contrast, colchicine has shown promise in major RCTs as an anti-inflammatory agent to reduce cardiovascular risk in both acute and chronic secondary prevention settings (111,112). These results underscore the relationship between inflammation and atherosclerosis.

 

TESTOSTERONE EFFECTS ON IMMUNE CELLS

 

Of note, in vitro studies have shown effects of T to reduce production of inflammatory cytokines from monocytes, macrophages and endothelial cells (113-115). Deletion of monocyte-macrophage AR in LDLR knockout mice resulted in reduced atherosclerosis compared to LDLR knockout mice, suggesting a role for AR-mediated actions in inflammatory cells (104). In an elegant study, pre-pubertal castration of male ApoE knockout mice increased atherosclerotic lesion area, which was abolished by an anti-CD3 antibody targeting T cells, linking hormonal and immunologic regulation of atherosclerosis (116). In that study, both castration and depletion of AR in epithelial cells resulted in increased thymus weight, and mice with depletion of AR in epithelial cells showed increased atherosclerosis and increased infiltration of T cells in the vascular adventitia (116). These findings support a mechanism by which deficiency of androgen action modulates immune/inflammatory responses to promote atherosclerosis.

 

TESTOSTERONE EFFECTS ON CYTOKINES

 

Older men treated with gonadotrophin-releasing hormone agonists to suppress HPT axis function showed increased concentrations of circulating tumor necrosis factor-α and interleukin-6 (117). In a randomized cross-over trial of 27 men ranging in age from 36-78 years, T treatment given via intramuscular injections over one month reduced circulating concentrations of tumor necrosis factor-α and interleukin-1β, and increased concentrations of the anti-inflammatory cytokine interleukin-10 (118). In another study of 20 men with type 2 diabetes, there was an inverse correlation of baseline T and interleukin-6, but T treatment over three months, while reducing waist circumference, did not alter tumor necrosis factor-α, interleukin-6, or C-reactive protein (CRP) concentrations (119). In the Testosterone Trials (T-Trials), T treatment via transdermal gel over 12 months in men aged ≥65 years did not change concentrations of high-sensitivity CRP or interleukin-6 (120). In a study of men treated with DHT or with recombinant human chorionic gonadotrophin over three months, neither intervention affected markers of endothelial cell activation or inflammation (121). By contrast, in a trial of men with metabolic syndrome, men in the T treatment arm showed a reduction in high sensitivity CRP after 12 months of treatment (122). In a trial of 76 men with newly diagnosed type 2 diabetes, T treatment over 9 months reduced markers of endothelial cell activation and inflammation, namely circulating concentrations of intracellular adhesion molecule type 1, p-selectin, and CRP (123). Therefore, the results of clinical studies are not wholly consistent. In summary, although the concept that T might exert anti-inflammatory actions protective against atherosclerosis is plausible, more evidence is needed using a direct measure of atherosclerosis.

 

CLINICAL TRIALS WITH SURROGATE ENDPOINTS

 

Testosterone Effects on Angina and Vascular Function

 

EFFECTS OF EXOGENOUS TESTOSTERONE ON ANGINA

 

Mechanistic studies in cell and animal models provide a plausible rationale for the epidemiological findings associating lower endogenous T concentrations with higher risk of CVD. However, clinical studies are necessary to clarify whether administration of T modulates clinical manifestations of CVD in vivo. Case series from the 1940s reported a beneficial effect of T therapy using intramuscular testosterone propionate to decrease the frequency and severity of angina attacks in an era where nitrate therapy was the mainstay of therapy (124-126). These early reports in men (and a small number of women) describe gradual improvements in symptoms over periods ranging from weeks to months. Conversely, a study in the 1960s found that administration of oral conjugated estrogen (that would suppress the HPT axis and serum androgen concentrations) to men resulted in adverse cardiovascular effects (127). In any case, as T is the native hormone which is metabolized in vivo to DHT and E2 (2), it is the preferred treatment for hypogonadal men (128), and represents the logical candidate for interventional studies.

 

More recent RCTs have revisited the issue of T treatment in men with CAD (Table 3A). In studies lasting from eight weeks to 12 months, T supplementation in men with CAD increased post-exercise ST segment depression (129), time to ischemia on exercise testing (130,132) and in a study in older men with diabetes, reduced the frequency of angina and silent myocardial ischemia during ECG Holter monitoring (133). These findings suggest either a protective effect of T on the myocardium, or an improvement in exercise capacity. A cross-over study found increased perfusion of myocardium supplied by unobstructed arteries, consistent with a vasodilatory action (131). Therefore, while existing data are limited, contemporary RCTs support historical observations suggesting a potentially beneficial effect of T supplementation in men with CAD.

 

Table 3. Selected Randomized Controlled Trials (RCTs) of Testosterone Supplementation in Middle-Aged and Older Men Reporting Outcomes Related to Angina (A), Artery Health (B), and Cardiovascular Adverse Events (C)

Study author and year

Population (men)

Formulation of T

N

active

N placebo

Duration

Result

A

 

 

 

 

 

 

Jaffe MD, 1977 (129)

Men with ST segment depression on exercise (mean age 58 years)

T cypionate 200 mg weekly

25

25

8 weeks

Decreased postexercise ST segment depression in T-treated but not placebo group

English KM, 2000 (130)

Men with coronary artery disease (mean age 62 years)

Transdermal patch 5 mg

22

24

12 weeks

Increased time to 1-mm ST- segment depression during treadmill exercise

Webb CM, 2008 (131)

Men with angiographically proven coronary artery disease, 40-75 years

Oral T undecanoate 80 mg bd

22

8 weeks, cross-over

No difference in angina or endothelial function, decreased arterial stiffness, increased perfusion of myocardium

Mathur A, 2009 (132)

Men with chronic stable angina (men age 65 years)

Depot intramuscular T undecanoate

7

6

12 months

Increased time to ischemia, non-significant trend for decreased CIMT

Cornoldi A, 2009 (133)

Men with proven coronary artery disease and type 2 diabetes (mean age 74 years)

Oral T undecanoate 40 mg tds

45

44

12 weeks

Reduced number of angina attacks and silent ischemic episodes on ECG Holter monitoring

B

 

 

 

 

 

 

Aversa A, 2010 (122)

Men with metabolic syndrome, T ≤11 nmol/L or free T ≤250 pmol/L (mean age 57 years)

Depot intramuscular T undecanoate 1,000 mg every 12 weeks

40

 

10

12 months*

Decreased high sensitivity CRP, improvement in CIMT

Basaria S, 2015 (164)

≥60 years, T 3.5-13.9 nmol/L or free T <173 pmol/L

Transdermal T gel 75 mg daily

156

152

3 years

No difference in rates of change in CIMT or coronary artery calcium

Budoff MJ, 2017 (168)

Men aged ≥65 years with T <9.5 nmol/L

Transdermal T gel 50 mg daily

73

65

12 months

Greater increase in non-calcified coronary plaque volume

Hildreth KL, 2018 (141)

Mean age 66 years, T 6.9-12.1 nmol/L

Transdermal gel, titrated to 13.9-19.1 or 20.8-34.7 nmol/L

41, 43

40

12 months

No effect of T on endothelial function or on CIMT

C

 

 

 

 

 

 

Basaria S, 2010 (172)

≥65 years, T 3.5-12.1 nmol/L or free T <173 pmol/L, mobility limitation

Transdermal gel 100 mg daily

106

103

6 months

Trial stopped prematurely due to excess cardiovascular events in T arm

Srinivas-Shankar U, 2010 (173)

≥65 years, T ≤12 nmol/L or free T ≤250 pmol/L, frail or intermediate frail

Transdermal gel 50 mg daily

138

136

6 months

T improved muscle strength and physical function, no signal for cardiovascular adverse events

Snyder P, 2016 (174)

≥65 years, T <9.5 nmol/L, sexual dysfunction (A), diminished vitality (B) and/or mobility limitations (C)

Transdermal gel 50 mg daily

395

(A 230,

B 236,

C 193)

395

(A 229,

B 238,

C 197)

12 months

Modest benefit of T on sexual function, no signal for cardiovascular adverse events

Wittert GA, 2021 (175)

50-74 years, waist ≥95 cm, T ≤14 nmol/L, and impaired glucose tolerance or newly diagnosed type 2 diabetes 

Depot intramuscular testosterone undecanoate 1000 mg every 3 months

504

503

24 months

All men received background lifestyle intervention (Weight Watchers). T reduced risk of type 2 diabetes at 2 years by 40%.

T=testosterone, CIMT=carotid intima media thickness, CRP=C-reactive protein. * men in placebo group switched to T after 12 months, extension study to 24 months no longer randomized.

 

EFFECTS OF EXOGENOUS TESTOSTERONE ON VASCULAR FUNCTION

 

Brachial artery endothelial function is an established measure of cardiovascular health examining both endothelial and vascular smooth muscle function, which mirrors responses in the coronary arteries (134). Assessment of arterial stiffness provides complementary insights into vascular health (135,136). In uncontrolled open-label studies in men with low baseline T concentrations, T supplementation improved both endothelial function and arterial stiffness (137,138). A study in hypogonadal older men found an improvement in arterial stiffness with transdermal T therapy (139). In a RCT of 55 obese men with type 2 diabetes, one year’s treatment with T undecanoate given as a depot intramuscular injection every 10 weeks improved endothelial function compared to placebo (140). However, other studies in middle-aged and older men did not show any effect of transdermal T treatment on endothelial function (141,142). There is a pathway by which T treatment is expected to improve endothelial function as in vitro studies demonstrate stimulation of nitric oxide synthesis in human aortic endothelial cells exposed to T (143). T might exert beneficial effects in the vasculature via actions to improve endothelial function and arterial stiffness, but additional studies are needed before a definitive conclusion can be made.

 

Testosterone and Atherosclerosis

 

CIRCULATING CHOLESTEROL CONCENTRATION

 

Clinical studies of T have shown consistent results for T treatment to reduce circulating concentrations of total cholesterol to a modest degree (144-146). A trend for T to lower LDL cholesterol has been noted (144). T treatment appears to lower high density lipoprotein (HDL) cholesterol again to a small degree (147). HDL cholesterol is involved in reverse cholesterol transport thus exerting anti-atherogenic activity, such that HDL function is an independent predictor of cardiovascular events (148,149). However, T may modulate HDL concentrations without a corresponding effect on HDL function (150). Of note, in observational studies, endogenous T concentrations correlated with circulating HDL and were inversely associated with total cholesterol (151,152). Thus, the prognostic significance of T-induced changes in lipid profiles, and the effect of T treatment on HDL-mediated anti-atherogenic action in men at risk for CVD remains unclear.

 

CAROTID ATHEROSCLEROSIS

 

Carotid intima-media thickness (CIMT) and the presence of carotid plaque are measures of preclinical carotid atherosclerosis, which can be assessed non-invasively using ultrasound (153). While low endogenous T concentrations are associated with increased CIMT in observational studies (154,155), it is less clear whether low endogenous T (or E2) predicts progression of CIMT (156-158). One study implicated low-grade inflammation in this process, finding an association of low non-SHBG-bound T with CIMT in older men with CRP ≥2 mg/L, but not in those with CRP <2 mg/L (159). Two cohort studies observed cross-sectional associations of low T concentrations with greater carotid plaque area (158,160). However, no association was found between baseline sex hormone concentrations and change in plaque area during follow-up, possibly due to increased use of anti-hypertensive and lipid-lowering therapy (158). One study reported an association of higher T concentrations with reduced CIMT and lower prevalence of carotid plaque in a cohort of community-dwelling men, but not in a cohort of men with angiographically proven CAD (161). In men with proven CAD, higher DHT was associated with less carotid plaque. Of note, E2 was associated with increased CIMT in community-dwelling men, but with less carotid plaque in men with CAD (161). One study found that higher E2 was associated with the presence of lipid core in carotid plaques in men, with no association of T concentrations (162). In a study of men who underwent carotid endarterectomy, the ratio of circulating T/E2 was inversely associated with plaque calcifications, macrophage staining and plaque neutrophil content, as well as plaque IL-6 protein (163). These findings associate sex hormone concentrations with CIMT and carotid plaque in men.

 

Several interventional studies reporting CIMT as an outcome are summarized (Table 3B). Of note, in a RCT of intramuscular T undecanoate 1,000 mg given every 12 weeks to men with metabolic syndrome, there was improvement in CIMT after 12 months of treatment (122). However, a larger RCT, Testosterone Effects on Atherosclerosis Progression in Aging Men (TEAAM), conducted over three years showed no effect of transdermal T treatment on rates of progression of CIMT (164). A smaller study, which tested both exercise training and transdermal T treatment over a period of 12 months, found no effect of either intervention on CIMT (141). Thus, while the RCT data are limited, the effects of T treatment on preclinical carotid atherosclerosis may be beneficial or neutral, but are unlikely to be adverse. See table 3, part B.

 

CORONARY ATHEROSCLEROSIS

 

Coronary computed tomography angiography (CCTA) has emerged as a non-invasive method for imaging coronary atherosclerosis (165-167). Normal findings on CCTA are associated with a low risk of cardiovascular events, while the presence and extent of CAD demonstrated on CCTA are risk predictors for future cardiovascular events in large epidemiological studies (165-167). Therefore, there is considerable interest in the Cardiovascular sub-study of the T-Trials which reported CCTA outcomes for 73 men treated with transdermal T and 65 men receiving placebo over a 12-month period (168). In this study, men in the T-treated group experienced a greater increase in non-calcified and total coronary artery plaque volume, compared to men in the placebo group (168). However, the groups were unbalanced with men in the T-treated group having considerably lower non-calcified and total plaque volumes at baseline and at the end of the study compared with placebo-treated men. The fact that the two groups of men differed substantively in a key baseline characteristic makes the result of the study challenging to interpret (169). There was no difference in the rate of change of coronary calcium score between groups, albeit men in the T-treated group had lower coronary calcium scores at baseline and at the end of the study compared with men in the placebo group (168). The results for coronary calcium scores are concordant with the TEAAM trial that also showed no difference in coronary calcium scores with T treatment (164). Men in the T-Trials Cardiovascular sub-study overall had a high burden of plaque at baseline with 32% of T-treated men and 38% of placebo recipients having baseline Agatston scores ≥300 (168).

 

Of note, in the T-Trials Cardiovascular sub-study, data on plaque volume were not analyzed relative to vessel lumen to address the issue of whether vascular remodeling was occurring (170). Since then, CCTA technology has progressed to allow more detailed and sophisticated analysis of plaque characteristics associated with higher risk of coronary events, which were not applied in that study (171). While these findings are important and noteworthy, a larger RCT with balanced groups using current CCTA methodology would be needed to clarify the effect of T on coronary plaque characteristics. The findings are a timely reminder that until definitive RCTs are available, the effects of T on the cardiovascular system remain uncertain and may be beneficial, neutral or adverse.

 

TRIALS REPORTING ADVERSE EVENTS

 

Testosterone RCTs and Cardiovascular Adverse Events

 

Selected T RCTs, which have influenced this field, are summarized (Table 3, part C). These studies were underpowered for cardiovascular events and the possibility of type 1 and type 2 errors should be considered. A key RCT randomized 209 men aged 65 years and older, with mobility limitations and low or low-normal baseline T or free T, to transdermal T gel vs placebo for six months (172). Of note the starting dose of transdermal T (100 mg daily) was greater than the usual recommended starting dose (50 mg daily). The study was discontinued after an excess of adverse events was noted in the T arm (172). A contemporaneous RCT in a comparable population of 274 older men who were frail or intermediate frail, using a 50 mg daily dose of transdermal T over 6 months, was successfully completed showing improved muscle strength and physical function, with no signal for cardiovascular adverse events (173). The Testosterone Trials (T-Trials) has reported results from the main study and component sub-studies (174,176). These have been extensively reviewed (169,176). In T-Trials, 790 men aged 65 years and older, with symptoms of sexual dysfunction, diminished vitality or mobility limitations and baseline T <9.5 nmol/L (<275 ng/dL), were randomized to transdermal T gel at a starting dose of 50 mg daily vs placebo for 12 months (174). In T-Trials, T treatment improved sexual function to a moderate degree, while the primary outcomes for physical function and vitality were not met (174). T treatment improved anemia and volumetric bone density, with a neutral effect on cognition (176). The effects on coronary artery plaque volume have been discussed (see Section 4.2.3). T-Trials had a low rate of major cardiovascular adverse events (7 in each of the T and placebo arms).

 

Recently, a larger Australian RCT, Testosterone for the Prevention of Type 2 Diabetes Mellitus (T4DM) has been reported (175). T4DM was a randomized, double-blind, placebo-controlled, 2-year, phase 3b trial done at six Australian tertiary care centers, which randomized 1,007 men to depot intramuscular testosterone undecanoate injections given every three months for two years, vs placebo, on a background of a lifestyle intervention (Weight Watchers) given to all participants. Inclusion criteria were age 50-74 years, waist circumference ≥95 cm and baseline T ≤14.0 nmol/L (≤403.8 ng/dL), and the presence of either impaired glucose tolerance or newly diagnosed type 2 diabetes based on oral glucose tolerance testing (175). In T4DM, testosterone treatment reduced the risk of type 2 diabetes at two years by 40% beyond the effect of the lifestyle intervention (relative risk 0·59, 95% confidence interval 0·43 to 0·80; p=0·0007). T4DM is the largest T RCT reported to date, and the incidence of cardiovascular adverse effects were similar in both T and placebo arms. In T4DM, 17 men in the placebo group and 12 in the T group had a major cardiovascular adverse event (13 men in the placebo arm and 7 in the testosterone arm had an ischemic heart disease event, 3 and 4 had cerebrovascular disease events respectively, and one in each group died from a cardiovascular-related cause) (175). Therefore, in keeping with T-Trials, the rates of major cardiovascular adverse events in T4DM were low, and comparable in testosterone and placebo-treated men.

 

At this point, it is worth commenting on the outcome of cardiorespiratory fitness. Low cardiorespiratory fitness, assessed as maximal oxygen consumption during exercise testing (VO2peak), is a strong independent predictor of all-cause and CVD mortality in apparently healthy men and in men with established CVD (177-179). Cardiorespiratory fitness has been recommended as a vital sign for use in clinical assessment (180). An earlier study over 12 months did not show an effect of T treatment on fitness (141), nor did a more recent study with a 12-week intervention (181). That study used a 2x2 factorial design, while exercise training resulted in improved VO2 peak within the 12-week period of intervention, T treatment did not, and there was no evidence within this relatively short timeframe of additive benefit (181). In the TEAAM study conducted over 3 years, placebo-treated men showed a decline in VO2peak over time, but the decline was attenuated in T-treated men (182). In TEAAM, there was no signal for cardiovascular adverse events with T (164). Part of the beneficial effect of T treatment on VO2peak might be mediated via its effect to raise hemoglobin concentrations (147,169,176), or its action on skeletal muscle (183,184). The net effect might be to preserve (or at least attenuate the loss of) cardiorespiratory fitness in ageing men, that could translate into a reduction in cardiovascular risk. However, in keeping with the results of T4DM, an extended duration of T intervention may be required to realize these benefits.

 

Meta-analyses of Cardiovascular Adverse Events in Testosterone RCTs

 

To date, no T RCT large and long enough to be powered for the outcome of cardiovascular events has been reported. However, meta-analyses of reported T RCTs have been performed to determine whether T is associated with a difference in the rate of cardiovascular adverse events (Table 4). Earlier meta-analyses done in 2007 and 2010 had found no significant difference in risk for cardiovascular adverse events (147,185). One analysis done in 2013 claimed an association of T treatment with increased risk of cardiovascular-related adverse events (186). However, subsequent meta-analyses done from 2013 to 2020 have not supported this finding (187-193). Instead, these have found no association of T treatment with risk of cardiovascular adverse events (Table 4).

 

Table 4. Meta-Analyses of Cardiovascular Adverse Events in Randomized Controlled Trials (RCTs) of T Supplementation in Men

Study characteristics

Results

Study author and year

N of RCTs

N

active

N placebo

Adverse signal

No adverse signal

Haddad RM, 2007 (185)

30

808

834

 

No significant difference in odds ratio for any cardiovascular adverse event or MI.

Fernandez-Balsells MM, 2010 (147)

51

2,716

 

No significant difference for all-cause mortality, coronary bypass surgery or MI.

Xu L, 2013 (186)

27

2,994

T associated with increased risk cardiovascular-related event (OR 1.54, 95% CI=1.09-2.18)*.

 

Ruige JB, 2013 (187)

10 (>100 participants)

1,289

848

 

No significant difference in cardiovascular adverse events.

Corona G, 2014 (188)

75

 

3,016

2,448

 

No association of T supplementation with cardiovascular risk. For MACE OR=1.01 (95% CI 0.57-1.77).

Borst SE, 2015 (189)

35

3,703

 

No significant risk for cardiovascular-related adverse events.

Alexander GC, 2017 (190)

39, cut-off for T 10.4-16.7 nmol/L

3,230

2,221

 

No significant increase in risk of MI OR=0.87 (95% CI 0.39-1.93)*, stroke or mortality.

Elliott J, 2017 (191)

87, cut-off for T 12 nmol/L or cFT 225 pmol/L

1,462-2088

1,372-1,851

 

Improved QoL, libido, depression and erectile function. No increase in risk of adverse events.

Corona G, 2018 (192)

93

4,653

3,826

 

No clear effect of T on incidence of CVD events. For MACE OR=0.97 (95% CI 0.64-1.46).

Diem SJ, 2020 (193)

38

N/A

N/A

 

Small improvement in sexual function and quality of life. Pooled risk for adverse cardiovascular outcomes did not differ between groups (OR=1.22, 95% CI 0.66-2.23).*

Hudson J, 2022 (194)

35 RCTs: 17 included in IPD meta-analysis

1,750 (IPD)

1,681 (IPD)

 

No significant difference between groups. For cardiovascular or cerebrovascular events OR=1·07 (95% CI 0·81–1·42).

MI=myocardial infarction, MACE=major adverse cardiovascular events, OR=odds ratio, 95% CI=confidence interval. QoL=quality of life, IPD=individual participant data. Unless otherwise specified, meta-analyses were conducted using random effects models. *fixed effects model

 

It is worth commenting on more recent meta-analyses that have included the results of the T-Trials. In the meta-analysis by Alexander et al. (2017), 39 RCTs were included. The meta-analysis found no significant increase in risk of MI (data from 30 RCTs utilized), stroke (9 RCTs) or mortality (20 RCTs) (190). However, caveats were noted with respect to the quality of the available evidence. In a network meta-analysis, Elliott et al. (2017) included RCTs that enrolled men with baseline T ≤12 nmol/L (≤346 ng/dl) or free T ≤225 pmol/L, including 87 RCTs overall (191). T treatment was associated with improved quality of life and libido, improvement in depression and in erectile function. There was no increase in risk of adverse events such as cardiovascular death, MI or stroke (191). Corona et al. (2018) studied 93 RCTs and found no clear effect of T on incidence of CVD events, with an odds ratio of 0.97 (95% confidence interval 0.64-1.46) for major cardiovascular adverse events (192).

 

Diem et al. (2020) examined 38 RCTs of at least six months duration, noting that few exceeded a 1-year duration, there was a lack of power to assess important harms, and limited data for men aged 18-50 years (193). They concluded that in older men with lower testosterone concentrations, in the absence of organic hypogonadism, T treatment resulted in small improvements in in sexual functioning and quality of life, with long-term safety and efficacy still uncertain. Recently Hudson et al. (2022) analyzed RCTs involving men with T ≤12 nmol/L and minimum duration of three months, identifying 35 studies and obtaining individual participant data from 17 of these with 3,431 participants (including T-Trials but not T4DM) (194). There was no significant difference in cardiovascular adverse events in testosterone vs placebo-treated men. The authors concluded that their results provide some reassurance about the short- to medium-term safety of T treatment for male hypogonadism (194). Clearly, the results of the TRAVERSE study, a testosterone cardiovascular safety trial, will be of considerable interest (195). For now, bearing in mind the limitations of meta-analyses of RCTs using reported adverse events as the endpoint, the weight of the currently available evidence from these sources indicates that T treatment is not associated with risk of cardiovascular adverse events.

 

RETROSPECTIVE CASE-CONTROL STUDIES

 

Retrospective Studies of Testosterone Prescriptions 

 

Pending an adequately powered T RCT to clarify its effect on the risk of cardiovascular events, retrospective case-control studies have sought to fill this gap (Table 5). These studies are typically based on health insurance databases recording prescriptions for T and subsequent outcomes in men prescribed or not prescribed T. Limitations of these studies include lack of clinical data such as indications for prescribing, the absence of randomization and the possibility of recall and misclassification bias (196). Initial studies in male veterans and in men with type 2 diabetes associated T prescriptions with lower mortality (197,198). By contrast, two studies associated T prescriptions with increased risk of major cardiovascular events (199,200). Both have been subject of criticisms: the first over confusing statistical methodology and data inaccuracies (resulting in publication of an erratum), the second over the lack of an appropriate comparison group (209,210). Subsequent studies have associated T prescriptions with no increase in risk of MI (201), and with lower risk of death, MI and stroke (202,203). An interesting distinction was made in the studies by Sharma et al. (2015) and Andersen et al. (2016) in that men who were prescribed T who then had “normal” T concentrations, did better than men who had persistently low T concentrations or who did not receive T (202,203). However, it is important to note that these studies did not systematically assess testosterone concentrations at multiple times or multiple days. A single measurement of testosterone on a particular day, may not be an accurate reflection of testosterone concentrations achieved over sustained periods of time with different testosterone formulations (211).

 

Table 5. Retrospective Case-Control Studies of Men Prescribed T that Examined Associations of T Prescriptions with Cardiovascular Events and Mortality in Middle-Aged and Older Men

Study characteristics

Results

Study author and year

Size (n of men)

Follow-up (yr)

Age (yr)

Favors no T

Favors T

Shores MM, 2012 (197)

1,031

3.4

62.1

 

Male veterans with total T ≤8.7 nmol/L, T prescribed in 398. T supplementation associated with lower mortality.

Muraleedharan V, 2013 (198)

581

5.8

59.5

 

Men with Type 2 diabetes, 238 with total T ≤10.4 nmol/L. T supplementation associated with lower mortality.

Vigen R, 2013 (199)

8,709

2.3

63.4

Male veterans who had coronary angiography and total T ≤10.4 nmol/L. T prescription associated with increased risk of death, MI or stroke.

 

Finkle WD, 2014 (200)

55,593

90 days

54.4

Men prescribed T. Higher rate of non-fatal MI in 90 days following prescription compared to preceding 1 year.

 

Baillargeon J, 2014 (201)

6,355; 19,065

4.1; 3.3

≥66

 

6,355 men prescribed T vs 19,065 matched non-users. T prescription not associated with increased risk of MI. For men with worse prognostic scores, T associated with reduced risk of MI.

Sharma R, 2015 (202)

83,010

6.2; 4.6; 4.7

66

 

Male veterans with low T. TRT resulting in normalization of circulating T (n=43,931) was associated with lower risk of death, MI and stroke, compared to TRT without normalization of T (n=25,701) or no TRT (n=13,378).

Anderson J, 2016 (203)

4,736

≥3

61.2

 

Men with T <7.4 nmol/L. T therapy achieving normal T (n=2,241) was associated with reduced risk of MACE compared to persistent low T (n=801). T therapy achieving either normal T or high T (n=1,694) associated with lower all-cause mortality compared to persistent low T.

Wallis CJD, 2016 (204)

10,311: 28,029

5.3

≥66

 

Men treated with T. T treatment associated with lower mortality HR=0.88 (95% CI=0.84-0.93) and prostate cancer risk HR=0.86 (95% CI=0.75-0.99). Shorter exposure (2 months) associated with increased risk of cardiovascular events and mortality, longer exposure (35 months) with reduced risk.

Cheetham TC, 2017 (205)

8,808: 35,527

3.2

58.4

 

Men ≥40 years, diagnosis or T <10.4 nmol/L. T associated with reduced risk of outcome of MACE, unstable angina, coronary revascularization, TIA. HR=0.67 (95% CI=0.62-0.73).

Loo SY, 2019 (206)

15,401

4.7

≥45

Men with low T and no evidence of HPT axis disease. T associated with increased risk of composite outcome of stroke/TIA/MI (HR=1.21, 95% CI 1.00-1.46), with risk highest in first 6 months to 2 years of T use (HR 1.35, 95% CI, 1.01-1.79). Risk of all-cause mortality lower with current T use (HR=0.64, 95% CI 0.52-0.78) and higher with past T use (HR=1.72, 95% CI 1.21-2.45), compared with non-use.

 

Oni OA, 2019 (207)

1,470

3.2-4.0

≥50

 

Male veterans with low total T and history of MI. All-cause mortality lower in men treated with T who normalized total T (N=755), vs men treated with T who did not normalize total T (N=542, HR=0.76, 95% CI 0.64-0.90), or men not treated with T (N=173, HR=0.76, 95% CI 0.60-0.98). No significant difference in the risk of recurrent MI between groups.

Shores MM, 2021 (208)

204,857

4.3

60.9

 

Male veterans with low T. Current transdermal T use not associated with risk for incident MI/ischemic stroke/venous thromboembolism (HR=0.89, 95% CI 0.76-1.05) in men without prevalent CVD, and in those with prevalent CVD was associated with lower risk (HR=0.80; 95% CI, 0.70-0.91). Current intramuscular T use not associated with risk for composite endpoint in men without or with prevalent CVD (HR=0.91, 95% CI 0.80-1.04; HR=0.98, 95% CI 0.89-1.09, respectively).

MI=myocardial infarction, TRT=testosterone replacement therapy, MACE=major cardiovascular adverse event comprising death, non-fatal MI and non-fatal stroke, TIA=transient ischemic attack.

 

A study by Wallis et al. (2016) found that T treatment was associated with lower mortality overall, but men who had T for a relatively shorter duration of exposure had increased risk, while men with longer duration of exposure had reduced risk (204). In a study by Cheetham et al. (2017) of men aged ≥40 years diagnosed with low T or with T <10.4 nmol/L, T treatment was associated with a reduced risk of major cardiovascular adverse events (205). Loo et al. (2019) reported contrary findings: analyzing a cohort of men with no evidence of HPT axis disease via the UK Clinical Practice Research Datalink, they associated use of testosterone with higher risk of a composite outcome of stroke, transient ischemic attack or MI, with the risk highest in the first six to 24 months of T use (206). In that study all-cause mortality risk was lower with current T use, and higher with past T use. Those findings are not supported by two more recent analyses, involving men with prior heart disease or with multiple comorbidities (207,208). In a study of male veterans with low T concentrations and a history of MI, men receiving T treatment who had a subsequent normal testosterone concentration had a lower risk of death from any cause compared to men receiving T treatment who had a subsequent low T concentration (207). Those men also had a lower risk of death compared with men who did not receive T treatment. Finally, in a cohort of 204,857 male veterans with a mean age of 60.9 years and 4.7 chronic medical conditions who were followed for 4.3 years, current transdermal T use was not associated with risk for the composite outcome of incident MI, ischemic stroke or venous thromboembolism in men without prevalent CVD (208). On the other hand, it was associated with lower risk in men with prevalent CVD. In that study, current intramuscular T use not associated with risk for the composite endpoint in men without or with prevalent CVD (208).

 

In an earlier retrospective cohort study that compared the use of T gel with T injections, T injections were associated with greater risk of cardiovascular adverse events (212). Bearing in mind the limitations of non-randomized studies and the possibility of bias, and the absence of a control group not receiving T, an additional factor is that more than 90% of the T injections were T cypionate, enanthate or propionate (212), which are short acting formulations typically requiring fortnightly administration with marked fluctuations in blood concentrations. The analysis would not apply to long-acting depot injections of T undecanoate typically administered every 12 weeks, which provide more stable pharmacokinetics (128). It is worth noting in this context that a population-based case-control study (involving 19,215 patients with confirmed venous thromboembolism and 909,530 age-matched controls) found an increased risk of venous thromboembolism within the first six months of T treatment but not thereafter (213). By contrast a systematic review and meta-analysis including six RCTs (2,236 participants) and five observational studies (1,249,640 participants) found no evidence of an association between T treatment and venous thromboembolism (214). However, the authors of that study noted that the available RCT data might have had inadequate power to detect an increased risk.

 

In summary, earlier findings associating T prescriptions with adverse cardiovascular outcomes were echoed in a more recent study. However, most studies do not show such adverse signals, and associated T use with lower risk of adverse cardiovascular events or mortality, including several recent large studies. There is a suggestion that T treatment which achieves normal T concentrations may relate to lower risk of cardiovascular events and mortality. Bearing in mind the limitations of these retrospective, observational and non-randomized studies, which cannot prove causality, the available data provide some reassurance but are far from definitive.

 

Abuse of Androgenic Steroids

 

Androgenic steroids can serve as appearance and performance-enhancing drugs and are abused by some competitive athletes, recreational sportspersons and body builders (1,215,216). The use/abuse of androgenic steroids occurs in contravention of medical advice and applicable sporting regulations, typically without medical supervision using unapproved formulations often in excessive doses (216). The use/abuse can be interspersed with periods of non-use. Adverse effects include suppression of the endogenous HPT axis, reduced spermatogenesis and impaired fertility, decreased testicular volume, hair loss and gynecomastia and are well-recognized (1,215). There is also an appreciation that long-term abuse results in cardiovascular toxicity in the form of myocardial dysfunction and accelerated coronary atherosclerosis (217). However, this study could be confounded as men who abuse androgenic steroids may also consume many other substances and the androgen preparations might have harmful contaminants. Abuse using pharmacological dosing of various products is very distinct from medically supervised T therapy aiming to achieve physiological circulating concentrations of T (128). Nevertheless, this is a reminder that excessive exposure to androgens carries the risk of harm (1).

 

DISCUSSION

 

Lessons from the Available Evidence

 

Epidemiological data are consistent with a protective role for endogenous androgens against CVD. In some studies of middle-aged and older men, lower circulating concentrations of endogenous T are associated with higher incidence of cardiovascular events, particularly stroke. Lower circulating T and DHT concentrations have also been associated with higher cardiovascular mortality (discussed in sections 2.1-2.2). Potential mechanisms by which T could exert beneficial actions in the vasculature have been explored in experimental models. These include reduced cholesterol accumulation and modulation of inflammation (sections 3.1-3.4). Clinical studies have reported favorable effects of T treatment on angina symptoms and exercise tolerance, but its effect on subclinical atherosclerosis remains uncertain (sections 4.1-4.2). The T-Trials, T4DM and meta-analyses of existing T RCTs in general do not show any signal for cardiovascular adverse events (sections 5.1-5.2). Retrospective case-control studies have reported contrasting results but in general, men receiving T prescriptions appear to have lower risk of major cardiovascular events and lower mortality compared to men who did not receive T, particularly if T treatment was associated with subsequent normal concentrations of circulating T (section 6.1). It is important to bear in mind that there are contrasting findings, and beneficial associations of T with cardiovascular outcomes may be less evident in healthier middle-aged men. Therefore, epidemiological evidence and mechanistic data could be used to argue for an anti-atherogenic or a protective effect of T on the cardiovascular system, as could the majority of retrospective case-control studies. However, this remains to be proven in the context of prospective RCTs of T intervention.

 

Gaps in the Current Evidence Base

 

RCT data are lacking as to whether treatment of middle-aged and older men with T would reduce the risk of cardiovascular events. The T-Trials which used transdermal T gel over a 12-month intervention offer important evidence as to benefits of T treatment for sexual function, anemia and bone density in older men without apparent diseases of the HPT axis, who had lower circulating T concentrations compared with younger men and symptoms suggestive of (but not diagnostic for) androgen deficiency (174,176). T4DM demonstrated the benefit of T treatment to reduce the risk of type 2 diabetes in men at high risk, beyond the effects of a lifestyle intervention (175). T4DM also showed a beneficial effect of T treatment on sexual function, and on bone microarchitecture and density (175,218). The T-Trials and T4DM are also noteworthy for the absence of any adverse cardiovascular safety signal for T treatment in these populations of men (174,175). However, the findings of the T-Trials Cardiovascular sub-study regarding an increase in total coronary atheroma plaque volume, in men with substantial baseline atheromatous disease, require clarification (168).

 

Major evidence gaps pertain to the effects of T on the cardiovascular system, as to whether T acts to slow development or progression of coronary or carotid atheromatous plaque in middle-aged and older men, in the differing contexts of either primary or secondary prevention for CVD. If the action of T is to reduce cholesterol accumulation, and to reduce inflammation and neointimal response to injury (sections 3.1-3.4) then these actions may have more impact to prevent or reduce progression of early atherosclerosis, rather than to reverse established disease. The related questions are whether T intervention in a primary prevention setting will reduce growth of coronary or carotid atheromatous plaque, or whether in a secondary prevention setting T intervention would influence the incidence of cardiovascular events. Another important question relates whether transdermal vs depot intramuscular (T undecanoate) formulations of T have similar or differing effects on the cardiovascular system.

 

Neither T-Trials nor T4DM had any cardiovascular endpoints and will not answer the question as to whether T exerts beneficial, neutral or adverse effects on the cardiovascular system. The US multicenter RCT “A study to evaluate the effect of testosterone replacement therapy (TRT) on the incidence of major adverse cardiovascular events (MACE) and efficacy measures in hypogonadal men (TRAVERSE)” commenced recruitment in 2018 of men aged 45-80 years with T <10.4 nmol/L (<300 ng/dl) with evidence of CVD or at increased risk for CVD (195). TRAVERSE was designed as a cardiovascular safety study with the endpoint of myocardial infarction, stroke or death due to cardiovascular causes, aimed to enroll 6,000 men randomized to transdermal T gel or placebo, and is planned to complete in 2022. TRAVERSE will also examine outcomes of prostate cancer, sexual function, bone fractures, depression, anemia and diabetes (195). TRAVERSE will address the issue of the cardiovascular safety of T treatment in what would largely be a secondary prevention setting. This leaves unanswered the question of whether T intervention in a primary prevention setting would reduce development or progression of coronary or carotid atheroma.

 

Application to Clinical Practice

 

Current clinical practice recommendations prioritize the identification of men with classical or pathological hypogonadism who are androgen deficient due to diseases of the hypothalamus, pituitary or testes (6,7). In such men, T treatment consistently resolves symptoms and signs of androgen deficiency (6,7,19). In men with classical or pathological hypogonadism the benefits of T treatment likely outweigh possible cardiovascular risks. In any case, individualized assessment and management of cardiovascular risk factors and disease should be part of routine clinical care. Of note, the US regulatory agency required labelling to warn of a possible increased risk of cardiovascular events with T, but the European regulatory agency concluded there was no consistent evidence of increased risk of coronary heart disease with T therapy (19). Until more evidence is available, it may be prudent to adopt a degree of caution in older men who are frail or who have known CVD, and to optimize management of cardiovascular risk factors and disease before starting T treatment. Treatment should aim for physiological replacement of T using approved formulations and avoid excessive doses (128).

 

It is beyond the scope of this chapter to discuss controversies regarding the management of men with low endogenous T concentrations due to obesity or presence of systemic illnesses where the HPT axis is intact, but its activity may be suppressed (19,31). However, it is worth noting that in men where a clear indication for T treatment is lacking, the risks and benefits of an intervention need to be considered with special care. Further research is needed to determine whether and how T treatment might impact on the risk of CVD in men.

 

Conclusions

 

Some epidemiological studies have associated higher circulating concentrations (but within the normal range) of endogenous androgens with lower risk of cardiovascular events and mortality. In men with pathological hypogonadism, the benefits of T treatment likely outweigh putative risks of cardiovascular adverse events. However, the effects of exogenous androgens in the form of T therapy seeking to maintain physiological circulating androgen concentrations on the cardiovascular system remain uncertain. Additional information will be forthcoming once the results of the TRAVERSE trial are known. Current clinical care of hypogonadal men should recognize this evidence gap and allow for individualized assessment and management of pre-existing cardiovascular risk factors and disease in men requiring T therapy. Well-designed and adequately powered RCTs are needed to clarify whether T treatment has beneficial, neutral or adverse effects on the cardiovascular system in the general population of middle-aged and older men.

 

REFERENCES

 

  1. Handelsman DJ. Androgen physiology, pharmacology, use and misuse. In endotext.org, version of October 5, 2020, published by MDTEXT.COM, Inc., South Dartmouth, MA 02748.
  2. Lakshman KM, Kaplan B, Travison TG, et al. The effects of injected testosterone dose and age on the conversion of testosterone to estradiol and dihydrotestosterone in young and older men. J Clin Endocrinol Metab 2010; 95: 3955-3964.
  3. Finkelstein JS, Whitcomb RW, O’Dea LStL, Longcope C, Schoenfeld DA, Crowley WF. Sex steroid control of gonadotrophin secretion in the human male. I. Effects of testosterone administration in normal and gonadotrophin-releasing hormone-deficient men. J Clin Endocrinol Metab 1991; 73: 609-620.
  4. Finkelstein JS, O’Dea LStL, Whitcomb RW, Crowley WF. Sex steroid control of gonadotropin secretion in the human male. II. Effects of estradiol administration in normal and gonadotropin-releasing hormone-deficient men. J Clin Endocrinol Metab 1991; 73: 621-628.
  5. Bagatell CJ, Dahl KD, Bremner WJ. The direct pituitary effect of testosterone to inhibit gonadotropin secretion in men is partially mediated by aromatization to estradiol. J Androl 1994; 15: 15-21.
  6. Yeap BB, Grossmann M, McLachlan RI, et al. Endocrine Society of Australia position statement on male hypogonadism (part 1): assessment and indications for testosterone therapy. Med J Aust 2016; 205: 173-178.
  7. Bhasin S, Brito JP, Cunningham GR, et al. Testosterone therapy in men with hypogonadism: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2018; 103: 1715-1744.
  8. Ryl A, Rotter I, Grzywacz A, et al. Molecular analysis of the SRD5A1 and SRD5A2 genes in patients with benign prostatic hyperplasia with regard to metabolic parameters and selected hormone levels. Int J Environ Res Public Health 2017; 14: 1318.
  9. Mellstrom D, Vandenput L, Mallmin H, et al. Older men with low serum estradiol and high serum SHBG have an increased risk of fractures. J Bone Mineral Res 2008; 23: 1552-1560.
  10. LeBlanc ES, Nielson CM, Marshall LM, et al. The effects of serum testosterone, estradiol, and sex hormone binding globulin levels on fracture risk in older men. J Clin Endocrinol Metab 2009; 94: 3337-3346.
  11. Finkelstein JS, Lee H, Burnett-Bowie S-A, et al. Gonadal steroids and body composition, strength, and sexual function in men. N Engl J Med 2013; 369: 1011-1022.
  12. Sikaris K, McLachlan RI, Kazlauskas R, et al. Reproductive hormone reference intervals for healthy fertile young men: evaluation of automated platform assays. J Clin Endocrinol Metab. 2005; 90:5928-5936.
  13. Jasuja GK, Travison TG, Davda M, et al. Age trends in estradiol and estrone levels measured using liquid chromatography tandem mass spectrometry in community-dwelling men of the Framingham Heart Study. J Gerontol Med Sci 2013; 68:733-740.
  14. Handelsman DJ, Wartofsky L. Requirement for mass spectrometry sex steroid assays in the Journal of Clinical Endocrinology and Metabolism. J Clin Endocrinol Metab. 2013; 98:3971-3973. Subsequent comment in Wierman ME, et al. J Clin Endocrinol Metab. 2014; 99:4375.
  15. Goldman AL, Bhasin S, Wu FCW, et al. A reappraisal of testosterone’s binding in circulation: physiological and clinical implications. Endocr Rev 2017; 38: 302-324.
  16. Handelsman DJ. Free testosterone: pumping up the tires or ending the free ride? Endocr Rev 2017; 38: 297-301.
  17. Ly LP, Sartorius G, Hull L, et al. Accuracy of calculated free testosterone formulae in men. Clin Endocrinol 2010; 73: 382-388.
  18. Fiers T, Wu F, Moghetti P, et al. Reassessing free-testosterone calculation by liquid chromatography-tandem mass spectrometry direct equilibrium dialysis. J Clin Endocrinol Metab 2018; 103: 2167-2174.
  19. Yeap BB, Wu FCW. Clinical practice update on testosterone therapy for male hypogonadism: contrasting perspectives to optimise care. Clin Endocrinol 2019; 90: 56-65.
  20. Bhasin S, Valderrabano RJ, Gagliano-Juca T. Age-related changes in the male reproductive system. In endotext.org, version of February 10, 2022, published by MDTEXT.COM, Inc., South Dartmouth, MA 02748.
  21. Handelsman DJ, Yeap BB, Flicker L, et al. Age-specific population centiles for androgen status in men. Eur J Endocrinol 2015; 173: 809-817.
  22. Yeap BB, Alfonso H, Chubb SAP, et al. Reference ranges and determinants of testosterone, dihydrotestosterone and estradiol levels measured using liquid chromatography-tandem mass spectrometry in a population-based cohort of older men. J Clin Endocrinol Metab. 2012; 97:4030-4039.
  23. Hsu B, Cumming RG, Hirani V, et al. Temporal trend in androgen status and androgen-sensitive outcomes in older men. J Clin Endocrinol Metab 2016; 101: 1836-1846.
  24. Ahern T, Swiecicka A, Eendebak RJAH, et al. Natural history, risk factors and clinical features of primary hypogonadism in ageing men: longitudinal data from the European Male Ageing Study. Clin Endocrinol 2016; 85: 891-901.
  25. Yeap BB, Manning L, Chubb SAP, et al. Progressive impairment of testicular endocrine function in ageing men: testosterone and dihydrotestosterone decrease, and luteinising hormone increases, in men transitioning from the 8th to 9th decades of life. Clin Endocrinol 2018; 88: 88-95.
  26. Wu FCW, Tajar A, Beynon JM, et al. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med 2010 363: 123-135.
  27. Tajar A, Huhtaniemi IT, O’Neill TW, et al. Characteristics of androgen deficiency in late-onset hypogonadism: results from the European Male Aging Study (EMAS). J Clin Endocrinol Metab 2012; 97: 1508-1516.
  28. Yeap BB, Knuiman MW, Divitini ML, et al. Epidemiological and Mendelian randomisation studies of dihydrotestosterone and estradiol, and leucocyte telomere length in men. J Clin Endocrinol Metab 2016; 101: 1299-1306.
  29. Yeap BB, Hui J, Knuiman MW, et al. Cross-sectional associations of sex hormones with leucocyte telomere length, a marker of biological age, in a community-based cohort of older men. Clin Endocrinol 2019; 90: 562-569.
  30. Coburn SB, Graubard BI, Trabert B, McGlynn KA, Cook MB. Associations between circulating sex steroid hormones and leucocyte telomere length in men in the National Health and Nutrition examination Survey. Andrology 2018; 6: 542-546.
  31. Wittert GA, Yeap BB, Grossmann M. Hypothalamo-pituitary-testicular axis function in systemic diseases and effects of medications. In: Oxford textbook of Endocrinology and Diabetes, 3rd Edition, Section 10, Chapter 6, pages 1597-1604. Oxford University Press, Oxford, United Kingdom, 2022.
  32. Yeap BB, Marriott RJ, Antonio L, et al. Sociodemographic, lifestyle and medical influences on serum testosterone and sex hormone-binding globulin in men from UK Biobank. Clin Endocrinol 2021; 94: 290-302.
  33. Grossmann M. Low testosterone in men with type 2 diabetes: significance and treatment. J Clin Endocrinol Metab 2011; 96: 2341-2353.
  34. Ng MTF, Dupuis P, Grossmann M. Lowered testosterone in male obesity: mechanisms, morbidity and management. Asian J Androl 2014; 16: 223-231.
  35. Grossmann M. Hypogonadism and male obesity: Focus on unresolved questions. Clin Endocrinol 2018; 89: 11-21.
  36. Global Burden of Disease Study 2013 Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990-2013: a systematic analysis for the Global Burden of disease Study 2013. Lancet 2015; 386: 743-800.
  37. Roth GA, Nyugen G, Forouzanfar MH, et al. Estimates of global and regional premature cardiovascular mortality in 2015. Circulation 2015; 132: 1270-1282.
  38. Allan GM, Nouri F, Korownyk C, et al. Agreement among cardiovascular disease risk calculators. Circulation 2013; 127: 1948-1956.
  39. Haslam DW, James WP. Obesity. Lancet 2005; 366: 1197-1209.
  40. Fan J, Song Y, Chen Y, et al. Combined effect of obesity and cardio-metabolic abnormality on the risk of cardiovascular disease: a meta-analysis of prospective cohort studies. Int J Cardiol 2013; 168: 4761-4768.
  41. Kwok CS, Pradhan A, Khan MA, et al. Bariatric surgery and its impact on cardiovascular disease and mortality: a systematic review and meta-analysis. Int J Cardiol 2014; 173: 20-28.
  42. Zhou X, Yu J, Li L, et al. Effects of bariatric surgery on mortality, cardiovascular events, and cancer outcomes in obese patients: a systematic review and meta-analysis. Obes Surg 2016; 26: 2590-2611.
  43. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. N Engl J Med 2021; 384: 989-1002.
  44. Jastreboff AM, Aronne LJ, Ahmad NN, et al. Tirzepatide once weekly for the treatment of obesity. N Engl J Med 2022; 387: 205-216.
  45. Yeap BB, Araujo AB, Wittert GA. Do low testosterone levels contribute to ill-health during male ageing? Crit Rev Clin Lab Sci. 2012; 49:168-182.
  46. Smith GD, Ben-Shlomo Y, Beswick A, et al. Cortisol, testosterone, and coronary heart disease. Prospective evidence from the Caerphilly Study. Circulation 2005; 112: 332-340.
  47. Arnlov J, Pencina MJ, Amin S, et al. Endogenous sex hormones and cardiovascular disease incidence in men. Ann Intern Med 2006; 145: 176-184.
  48. Abbott RD, Launer LJ, Rodriguez BL, et al. Serum estradiol and risk of stroke in elderly men. Neurology 2007; 68: 563-568.
  49. Vikan T, Schirmer H, Njolstad I, Svartberg J. Endogenous sex hormones and the prospective association with cardiovascular disease and mortality in men: the Tromso study. Eur J Endocrinol 2009; 161: 435-442.
  50. Yeap BB, Hyde Z, Almeida OP, et al. Lower testosterone levels predict incident stroke and transient ischemic attack in older men. J Clin Endocrinol Metab 2009; 94: 2353-2359.
  51. Ohlsson C, Barrett-Connor E, Bhasin S, et al. High serum testosterone is associated with reduced risk of cardiovascular events in elderly men. J Am Coll Cardiol 2011; 58: 1674-1681.
  52. Hyde Z, Norman PE, Flicker L, et al. Elevated luteinizing hormone predicts ischaemic heart disease events in older men. The Health In Men Study. Eur J Endocrinol 2011; 164: 569-577.
  53. Haring R, Teng Z, Xanthakis V, et al. Associations of sex steroids, gonadotrophins, and their trajectories with clinical cardiovascular disease and all-cause mortality in elderly men from the Framingham Heart Study. Clin Endocrinol 2013; 78: 629-634.
  54. Soisson V, Brailly-Tabard S, Helmer C, et al. A J-shaped association between plasma testosterone and risk of ischemic arterial event in elderly men: The French 3C Cohort Study. Maturitas 2013; 75: 282-288.
  55. Shores MM, Biggs ML, Arnold AM, et al. Testosterone, dihydrotestosterone, and incident cardiovascular disease and mortality in the cardiovascular health study. J Clin Endocrinol Metab 2014; 99: 2061-2068.
  56. Shores MM, Arnold AM, Biggs ML, et al. Testosterone and dihydrotestosterone and incident ischaemic stroke in men in the Cardiovascular Health Study. Clin Endocrinol 2014; 81: 746-753.
  57. Yeap BB, Alfonso H, Chubb SAP, et al. In older men, higher plasma testosterone or dihydrotestosterone is an independent predictor for reduced incidence of stroke but not myocardial infarction. J Clin Endocrinol Metab 2014; 99: 4565-4573.
  58. Srinath R, Golden SH, Carson KA, Dobs A. Endogenous testosterone and its relationship to preclinical and clinical measures of cardiovascular disease in the Atherosclerosis Risk in Communities Study. J Clin Endocrinol Metab 2015; 100: 1602-1608.
  59. Holmegard HN, Nordestgaard BG, Jensen GB, et al. Sex hormones and ischemic stroke: a prospective cohort study and meta-analyses. J Clin Endocrinol Metab 2016; 101: 69-78.
  60. Chan YX, Knuiman MW, Hung J, et al. Neutral associations of testosterone, dihydrotestosterone and estradiol with fatal and non-fatal cardiovascular events, and mortality in men aged 17-97 years. Clin Endocrinol 2016; 85: 575-582.
  61. Srinath R, Gottesman RF, Golden SH, et al. Association between endogenous testosterone and cerebrovascular disease in the ARIC Study (Atherosclerosis Risk in Communities). Stroke 2016; 47: 2682-2688.
  62. Wang A, Arver S, Boman K, et al. Testosterone, sex hormone-binding globulin and risk of cardiovascular events: a report from the Outcome Reduction with an Initial Glargine Intervention trial. Eur J Prev Cardiol. 2019; 26:847-854.
  63. Gyawali P, Martin SA, Heilbronn LK, et al. Higher serum sex hormone-binding globulin levels are associated with incident cardiovascular disease in men. J Clin Endocrinol Metab 2019; 104: 6301-6315.
  64. Hatami H, Parizadeh D, Yarandi RB, et al. Endogenous testosterone does not improve prediction of incident cardiovascular disease in a community-based cohort of adult men: results from the Tehran Lipid and Glucose Study. Aging Male 2020; 23:243-250.
  65. Zhao D, Guallar E, Ballantyne CE, et al. Sex hormones and incident heart failure in men and postmenopausal women: the Atherosclerosis Risk in Communities Study. J Clin Endocrinol Metab. 2020; 105: e3789-e3807.
  66. Collett T-H, Ewing SK, Ensrud KE, et al. Endogenous testosterone levels and the risk of cardiovascular events in elderly men: the MrOS Prospective Study. J Endocr Soc 2020; 4: bvaa038.
  67. Boden WE, Miller MG, McBride R, et al. Testosterone concentrations and risk of cardiovascular events in androgen-deficient men with atherosclerotic cardiovascular disease. Am Heart J 2020; 224: 65-76.
  68. Schafer S, Aydin MA, Appelbaum S, et al. Low testosterone concentrations and prediction of future heart failure in men and in women: evidence from the large FINRISK97 study. ESC Heart Failure 2021; 8:2485-2491.
  69. Yeap BB, Marriott RJ, Antonio L, et al. Associations of serum testosterone and sex hormone-binding globulin with incident cardiovascular events in middle-aged to older men. Ann Intern Med 2022; 175: 159-170.
  70. Dittadi R, Matteucci M, Meneghetti E, Ndreu R. Reassessment of the Access Testosterone chemiluminescence assay and comparison with LC-MS method. J Clin Lab Anal. 2018; 32: e22286.
  71. Fry A, Littlejohns TJ, Sudlow C, et al. Comparison of sociodemographic and health-related characteristics of UK Biobank participants with those of the general population. Am J Epidemiol 2017; 186: 1026-1034.
  72. Khaw K-T, Dowsett M, Folkerd E, et al. Endogenous testosterone and mortality due to all causes, cardiovascular disease, and cancer in men. European Prospective Investigation into Cancer in Norfolk (EPIC-Norfolk) prospective population study. Circulation 2007; 116: 2694-2701.
  73. Araujo AB, Kupelian V, Page ST, et al. Sex steroids and cause-specific mortality in men. Arch Intern Med 2007; 167: 1252-1260.
  74. Laughlin GA, Barrett-Connor E, Bergstrom J. Low serum testosterone and mortality in older men. J Clin Endocrinol Metab 2008; 93: 68-75.
  75. Tivesten A, Vandenput L, Labrie F, et al. Low serum testosterone and estradiol predict mortality in elderly men. J Clin Endocrinol Metab 2009; 94: 2482-2488.
  76. Menke A, Guallar E, Rohrmann S, et al. Sex steroid concentrations and risk of death in US men. Am J Epidemiol 2010; 171: 583-592.
  77. Haring R, Volzke H, Steveling A, et al. Low serum testosterone levels are associated with increased risk of mortality in a population-based cohort of men aged 20-79. Eur Heart J 2010; 31: 1494-1501.
  78. Hyde Z, Norman PE, Flicker L, et al. Low free testosterone predicts mortality from cardiovascular disease but not other causes: the Health In Men Study. J Clin Endocrinol Metab 2012; 97: 179-189.
  79. Yeap BB, Alfonso H, Chubb SAP, et al. In older men an optimal plasma testosterone is associated with reduced all-cause mortality, and higher dihydrotestosterone with reduced ischaemic heart disease mortality, while estradiol levels do not predict mortality. J Clin Endocrinol Metab 2014; 99: E9-E18.
  80. Pye SR, Huhtaniemi IT, Finn JD, et al. Late-onset hypogonadism and mortality in ageing men. J Clin Endocrinol Metab 2014; 99: 1357-1366.
  81. Holmboe SA, Vradi E, Jensen TK, et al. The association of reproductive hormone levels and all-cause, cancer, and cardiovascular disease mortality in men. J Clin Endocrinol Metab 2015; 100: 4472-4480.
  82. Hsu B, Cumming RG, Naganathan V, et al. Temporal changes in androgens and estrogens are associated with al-cause and cause-specific mortality in older men. J Clin Endocrinol Metab 2016; 101: 2201-2210.
  83. Chasland LC, Knuiman MW, Divitini ML, et al. Greater physical activity and higher androgen concentrations are independently associated with lower cardiometabolic risk in men. Clin Endocrinol 2017; 87: 466-474.
  84. Yeap BB, Marriott RJ, Antonio L, et al. Serum testosterone is inversely and sex hormone-binding globulin is directly associated with all-cause mortality in men. J Clin Endocrinol Metab 2021; 106: e625-e637.
  85. Takov K, Wu J, Denvir MA, Smith LB, Hadoke PWF. The role of androgen receptors in atherosclerosis. Mol Cell Endocrinol 2018; 465: 82-91.
  86. Jones TH, Kelly DM. Randomized controlled trials – mechanistic studies of testosterone and the cardiovascular system. Asian J Androl 2018; 20: 120-130.
  87. Larsen BA, Nordestgaard BG, Stender A, Kjeldsen K. Effect of testosterone on atherogenesis in cholesterol-fed rabbits with similar plasma cholesterol levels. Atherosclerosis 1993; 99: 79-86.
  88. Bruck B, Brehme U, Gugel N, et al. Gender-specific differences in the effects of testosterone and estrogen on the development of atherosclerosis in rabbits. Arterioscler Thromb Vasc Biol 1997; 17: 2192-2199.
  89. Alexandersen P, Haarbo J, Byrjalsen I, Lawaetz H, Christiansen C. Natural androgens inhibit male atherosclerosis: a study in castrated, cholesterol-fed rabbits. Circ Res 1999; 84: 813-819.
  90. Li SJ, Li XY, Li Y. Regulation of atherosclerotic plaque growth and stability by testosterone via influence of inflammatory reaction. Vascul Pharmacol 2008; 49: 14-18.
  91. Deng L, Fu D, Zhu L, Huang J, Ling Y, Cai Z. Testosterone deficiency accelerates early stage atherosclerosis in miniature pigs fed a high-fat and high-cholesterol diet: urine 1H NMR metabolomics targeted analysis. Mol Cell Biochem 2021; 476: 1245-1255.
  92. Nathan L, Shi W, Dinh H, et al. Testosterone inhibits early atherogenesis by conversion to estradiol: Critical role of aromatase. PNAS 2001; 98: 3589-3593.
  93. McRobb L, Handelsman DJ, Heather AK. Androgen-induced progression of arterial calcification in Apolipoprotein E-null mice is uncoupled from plaque growth and lipid levels. Endocrinol 2009; 150: 841-848.
  94. Nettleship JE, Jones TH, Channer KS, Jones RD. Physiological testosterone replacement therapy attenuates fatty streak formation and improves high-density lipoprotein cholesterol in the Tfm mouse. Circulation 2007; 116: 2427-2434.
  95. Bourghardt J, Wilhelmson ASK, Alexanderson C, et al. Androgen receptor-dependent and independent atheroprotection by testosterone in male mice. Endocrinol 2010; 151: 5428-5437.
  96. Hanke H, Lenz C, Hess B, Spindler KD, Weidemann W. effect of testosterone on plaque development and androgen receptor expression in the arterial wall. Circulation 2001; 103: 1382-1385.
  97. Tharp DL, Masseau I, Ivey J, Ganjam VK, Bowles DK. Endogenous testosterone attenuates neointima formation after moderate coronary balloon injury in male swine. Cardiovasc Res 2009; 82: 152-160.
  98. Wu J, Hadoke PWF, Mair I, et al. Modulation of neointimal lesion formation by endogenous androgens in independent of vascular androgen receptor. Cardiovasc Res 2014; 103: 281-190.
  99. Bentzon JF, Otsuka F, Virmani R, Falk E. Mechanisms of plaque formation and rupture. Circ Res 2014; 114: 1852-1866.
  100. Rana K, Lee NKL, Zajac JD, MacLean HE. Expression of androgen receptor target genes in skeletal muscle. Asian J Androl 2014; 16: 675-683.
  101. Rana K, Chiu MWS, Russel PK, et al. Muscle-specific androgen receptor deletion shows limited actions in myoblasts but not in myofibers in different muscles in vivo. J Mol Endocrinol 2016; 57: 125-138.
  102. Williams MRI, Ling S, Dawood T, et al. Dehydroepiandrosterone inhibits human vascular smooth muscle cell proliferation independent of ARs and ERs. J Clin Endocrinol Metab 2002; 87: 176-181.
  103. Lopes RAM, Neves KB, Pestana CR, et al. Testosterone induces apoptosis in vascular smooth muscle cells via extrinsic apoptotic pathway with mitochondria-generated reactive oxygen species involvement. Am J Physiol Heart Circ Physiol 2014; 306: H1485-H1494.
  104. Huang C-K, Pang H, Wang L, et al. New therapy in targeting androgen receptor in monocytes/macrophages to battle atherosclerosis. Hypertension 2014; 63: 1345-1353.
  105. Zhu D, Hadoke PWF, Wu J, et al. Ablation of the androgen receptor from vascular smooth muscle cells demonstrate a role for testosterone in vascular calcification. Sci Rep 2016; 6: 24807.
  106. Libby P, Loscalzo J, Ridker P, et al. Inflammation, immunity, and infection in atherothrombosis. J Am Coll Cardiol 2018; 72: 2071-2081.
  107. Moriya J. Critical roles of inflammation in atherosclerosis. J Cardiol 2019; 73: 22-27.
  108. Ridker PM, Danielson E, Fonseca FAH, et al. Rosuvastatin to prevent events in men and women with elevated C-reactive protein. N Engl J Med 2008; 359: 2195-2207.
  109. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. New Eng J Med 2017; 377: 1119-1131.
  110. Ridker PM, Everett BM, Pradhan A, et al. Low-dose methotrexate for the prevention of atherosclerotic events. New Eng J Med 2018; doi:10.1056/NEJMoa1809798.
  111. Tardiff J-C, Kouz S, Waters DD, et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N Engl J Med 2019; 381: 2497-2505.
  112. Nidorf SM, Fiolet ATL, Mosterd A, et al. Colchicine in patients with chronic coronary coronary disease. N Engl J Med 2020; 383: 1838-1847.
  113. Li ZG, Danis VA, Brooks PM. Effect of gonadal steroids on the production of IL-1 and IL-6 by blood mononuclear cells in vitro. Clin Exp Rheumatol 1993; 11: 157-162.
  114. D’Agostino P, Milano S, Barbera C, et al. Sex hormones modulate inflammatory mediators produced by macrophages. Ann NY Acad Sci 1999; 876: 426-429.
  115. Hatakeyama H, Nishizawa M, Nakagawa A, Nakano S, Kigoshi T, Uchida K. Testosterone inhibits tumor necrosis factor-α-induced vascular cells adhesion moleculre-1 expression in human aortic endothelial cells. FEBS Lett 2002; 530: 129-132.
  116. Wilhelmson AS, Rodriguez ML, Eriksson ES, et al. Testosterone protects against atherosclerosis in male mice by targeting thymic epithelial cells-brief report. Arterioscler Thromb Vasc Biol 2018; 38: 1519-1527.
  117. Khosla S, Atkinson EJ, Dunstan CR, O’Fallon WM. Effect of estrogen versus testosterone on circulating osteoprotegerin and other cytokine levels in normal elderly men. J Clin Endocrinol Metab 2001; 87: 1550-1554.
  118. Malkin CJ, Pugh PJ, Jones RD, Kapoor D, Channer KS, Jones TH. The effect of testosterone replacement on endogenous inflammatory cytokines and lipid profiles in hypogonadal men. J Clin Endocrinol Metab 2004; 89: 3213-3318.
  119. Kapoor D, Clarke S, Stanworth R, Channer KS, Jones TH. The effect of testosterone replacement therapy on adipocytokines and C-reactive protein in hypogonadal men with type 2 diabetes. Eur J Endocrinol 2007; 156: 595-602.
  120. Mohler ER, Ellenbery SS, Lewis CE, et al. The effect of testosterone on cardiovascular biomarkers in the Testosterone Trials. J Clin Endocrinol Metab 2018; 103: 681-688.
  121. Ng MK, Liu PY, Williams AJ, et al. Prospective study of effect of androgens on serum inflammatory markers in men. Arterioscler Thromb Vasc Biol 2002; 22: 1136-1141.
  122. Aversa A, Bruzziches R, Francomano D, et al. Effects of testosterone undecanoate on cardiovascular risk factors and atherosclerosis in middle-aged men with late-onset hypogonadism and metabolic syndrome: results from a 24-month, randomized, double-blind, placebo-controlled trial. J Sex Med 2010; 7: 3495-3503.
  123. Khripun I, Vorobyev S, Belousov I, Kogan M, Zitzmann M. Influence of testosterone substitution on glycemic control and endothelial markers in men with newly diagnosed functional hypogonadism and type 2 diabetes mellitus: a randomized controlled trial. Aging Male 2019; 22: 241-249.
  124. Lesser MA. The treatment of angina pectoris with testosterone propionate. New Engl J Med 1942; 226: 51-54.
  125. Levine SA, Likoff WB. The therapeutic value of testosterone propionate in angina pectoris. New Engl J Med 1943; 229; 770-772.
  126. Lesser MA. Testosterone propionate therapy in one hundred cases of angina pectoris. J Clin Endocrinol (Metab) 1946; 6: 549-557.
  127. The Coronary Drug Project Research Group. Initial findings leading to modifications of its research protocol. JAMA 1970; 214: 1303-1313.
  128. Yeap BB, Grossmann M, McLachlan RI, et al. Endocrine Society of Australia position statement on male hypogonadism (part 2): treatment and therapeutic considerations. Med J Aust 2016; 205: 228-231.
  129. Jaffe MD. Effect of testosterone cypionate on postexercise ST segment depression, Brit Heart J 1977; 39: 1217-1222.
  130. English KM, Steeds RP, Hugh Jones T, et al. Low-dose transdermal testosterone therapy improves angina threshold in men with chronic stable angina. A randomized, double-blind, placebo-controlled study. Circulation 2000; 102: 1906-1911.
  131. Webb CM, Elkington AG, Kraidly MM, et al. Effects of oral testosterone treatment on myocardial perfusion and vascular function in men with low plasma testosterone and coronary heart disease. Am J Cardiol 2008; 101: 618-624.
  132. Mathur A, Malkin C, Saeed B, et al. Long-term benefits of testosterone replacement therapy on angina threshold and atheroma in men. Eur J Endocrinol 2009; 161: 443-449.
  133. Cornoldi A, Caminiti G, Marazzi G, et al. Effects of chronic testosterone administration on myocardial ischemia, lipid metabolism and insulin resistance in elderly male diabetic patients with coronary artery disease. Int J Cardiol 2010; 142: 50-55.
  134. Takase B, Uebata A, Akima T, et al. Endothelium-dependent flow-mediated vasodilatation in coronary and brachial arteries in suspected coronary artery disease. Am J Cardiol 1998; 82: 1535-1539.
  135. Ben-Shlomo Y, Spears M, Boustred C, et al. Aortic pulse wave velocity improves cardiovascular event prediction: an individual participant meta-analysis of prospective observational data from 17,635 subjects. J Am Coll Cardiol 2014; 63: 636-646.
  136. Vlachopoulos C, Aznaouridis K, Stefanidis C. Aortic stiffness for cardiovascular risk prediction: just measure it, just do it! J Am Coll Cardiol 2014; 63: 647-649.
  137. Sader MA, Griffiths KA, Skilton MR, Wishart SM, Handelsman DJ, Celermajer DS. Physiological testosterone replacement and arterial endothelial function in men. Clin Endocrinol 2003; 59: 62-67.
  138. Shoskes DA, Tucky B, Polackwich AS. Improvement of endothelial function following initiation of testosterone replacement therapy. Trans Androl Urol 2016; 5: 819-823.
  139. Yaron M, Greenman Y, Rosenfeld JB, et al. Effects of testosterone replacement therapy in arterial stiffness in older hypogonadal men. Eur J Endocrinol 2009; 160: 839-846.
  140. Groti K, Zuran I, Antonic B, Forsnaric L, Pfeifer M. The impact of testosterone replacement therapy on glycemic control, vascular function, and components of the metabolic syndrome in obese hypogonadal men with type 2 diabetes. Aging Male 2018; 21: 158-169.
  141. Hildreth KL, Schwartz RS, van de Griend J, Kohrt WM, Blatchford PJ, Moreau KL. Effects of testosterone and progressive resistance exercise on vascular function in older men. J Appl Physiol 2018; 125: 1693-1701.
  142. Chasland LC, Naylor LH, Yeap BB, Maiorana AJ, Green DJ. Testosterone and exercise in middle-to-older aged men: combined and independent effects on vascular function. Hypertension 2021; 77: 1095-1105.
  143. Yu J, Akishita M, Eto M, et al. Androgen receptor-dependent activation of endothelial nitric oxide synthase in vascular endothelial cells: role of phosphatidylinositol 3-kinase/Akt pathway. Endocrinology 2010; 151: 1822-1828.
  144. Isidori A, Giannetta E, Greco EA, et al. Effects of testosterone on body composition, bone metabolism and serum lipid profile in middle-aged men: a meta-analysis. Clin Endocrinol 2005; 63: 280-293.
  145. Guo C, Gu W, Liu M, et al. Efficacy and safety of testosterone replacement therapy in men with hypogonadism: a meta-analysis study of placebo-controlled trials. Exp Ther Med 2016; 11: 853-863.
  146. Corona G, Giagulli VA, Maseroli E, et al. Testosterone supplementation an body composition: results from a meta-analysis study. Eur J Endocrinol 2016; 174: R99-R116.
  147. Fernandez-Balsells MM, Murad MH, Lane M, et al. Adverse effects of testosterone therapy in adult men: a systematic review and meta-analysis. J Clin Endocrinol metab. 2010; 95: 2560-2575.
  148. Khera AV, Cuchel M, de la Llera-Moya M, et al. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N Engl J Med 2011; 364: 127-135.
  149. Rohatgi A, Khera A, Berry JD, et al. HDL cholesterol efflux capacity and incident cardiovascular events. N Engl J Med 2014; 371: 2383-2393.
  150. Rubinow KB, Vaisar T, Chao JH, Heinecke JW, Page ST. Sex steroids mediate discrete effects on HDL efflux capacity and particle concentration in healthy men. J Clin Lipidol 2018; 12: 1072-1082.
  151. Makinen JI, Perheentupa A, Irjala K, et al. Endogenous testosterone and serum lipids in middle-aged men. Atherosclerosis 2008; 197: 688-693.
  152. Haring R, Baumeister SE, Volzke H, et al. Prospective association of low total testosterone concentrations with adverse lipid profiles and increased incident dyslipidemia. Eur J Cardiovasc Prev Rehab 2011; 18: 86-96.
  153. Lorenz MW, Markus HS, Bots ML, Rosvall M, Sitzer M. Prediction of clinical cardiovascular events with carotid intima-media thickness. Circulation 2007; 115: 459-467.
  154. Van den Beld AW, Bots ML, Janssen JAMLL, Pols HAP, Lamberts SWJ, Grobbee DE. Endogenous hormones and carotid atherosclerosis in elderly men. Am J Epidemiol. 2003; 157: 25-31.
  155. Svartberg J, von Muhlen D, Mathiesen E, Joakimsen O, Bonaa KH, Stensland-Bugge E. Low testosterone levels are associated with carotid atherosclerosis in men. J Intern Med 2006; 259: 576-582.
  156. Muller M, van den Beld AW, Bots ML, Grobbee DE, Lamberts SWJ, van der Schouw YT. Endogenous sex hormones and progression of carotid atherosclerosis in elderly men. Circulation 2004; 109: 2074-2079.
  157. Tivesten A, Hulthe J, Wallenfeldt K, Wikstrand J, Ohlsson C, Fagerberg B. Circulating estradiol is an independent predictor of progression of carotid artery intima-media thickness in middle-aged men. J Clin Endocrinol Metab 2006; 91: 4423-4437.
  158. Vikan T, Johnsen SH, Schirmer H, Njolstad I, Svartberg J. Endogenous testosterone and the prospective association with carotid atherosclerosis in men: the Tromso study. Eur J Epidemiol 2009; 24: 289-295.
  159. Soisson V, Brailly-Tabard S, Empana J-P, et al. Low plasma testosterone and elevated carotid intima-media thickness: importance of low-grade inflammation in elderly men. Atherosclerosis 2012; 223: 244-249.
  160. Dorr M, Wallaschofski H, Friedrich N. Association of low total testosterone levels and prevalent carotid plaques: result of the study of health in Pomerania. Eur J Epidemiol 2009; 24: 389-391.
  161. Chan YX, Knuiman MW, Hung J, et al. Testosterone, dihydrotestosterone and estradiol are differentially associated with carotid intima-media thickness and the presence of carotid plaque in men with and without coronary artery disease. Endocr J 2015; 62: 777-786.
  162. Glisic M, Mujaj B, Rueda-Ochoa OL, et al. Associations of endogenous estradiol and testosterone levels with plaque composition and risk of stroke in subjects with carotid atherosclerosis. Circ Res 2018; 122: 97-105.
  163. van Koeverden ID, de Bakker M, Haitjema S, et al. Testosterone to oestradiol ratio reflects systemic and plaque inflammation and predicts future cardiovascular events in men with severe atherosclerosis. Cardiovasc Res 2019; 115: 453-462.
  164. Basaria S, Harman SM, Travison TG, et al. Effects of testosterone administration for 3 years on subclinical atherosclerosis progression in older men with low or low-normal testosterone levels: a randomized clinical trial. JAMA 2015; 314: 570-581.
  165. Hulten EA, Carbonaro S, Petrillo SP, Mitchell JD, Villines TC. Prognostic value of cardiac computed tomography angiography. J Am Coll Cardiol 2011; 57: 1237-1247.
  166. Bamberg F, Summer WH, Hoffmann V, et al. meta-analysis and systematic review of the long-term predictive value of assessment of coronary atherosclerosis by contrast-enhanced coronary computed tomography angiography. J Am Coll Cardiol 2011; 57: 2426-2436.
  167. Nakazato R, Arsanjani R, Achenbach S, et al. Age-related risk of major adverse cardiac event risk and coronary artery disease extent and severity by coronary CT angiography: results from 15,187 patients from the International Multisite CONFIRM Study. Eur Heart J Cardiovasc Imaging 2014; 15: 586-594.
  168. Budoff MJ, Ellenberg SS, Lewis CE, et al. Testosterone treatment and coronary artery plaque volume in older men with low testosterone. JAMA 2017; 317: 708-716.
  169. Yeap BB, Page ST, Grossmann M. Testosterone treatment in older men: clinical implications and unresolved questions from the Testosterone Trials. Lancet Diabetes Endocrinol 2018; 6: 659-672.
  170. Dhindsa S, Wilson MF, Dandona P. Letter to the Editor on “Changes in coronary artery plaque with testosterone therapy”. JAMA 2017; 317: 2450.
  171. Oikonomou EK, Marwan M, Desai M, et al. Non-invasive detection of coronary inflammation using computed tomography and prediction of residual cardiovascular risk (the CRISP CT study): a post-hoc analysis of prospective outcome data. Lancet 2018; 392: 929-939.
  172. Basaria S, Coviello AD, Travison TG, et al. Adverse events associated with testosterone administration. New Engl J Med 2010; 363: 109-122.
  173. Srinivas-Shankar U, Roberts SA, Connolly MJ, et al. Effects of testosterone on muscle strength, physical function, body composition, and quality of life in intermediate-frail and frail elderly men: a randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab 2010: 95: 639-650.
  174. Snyder PJ, Bhasin S, Cunningham GR, et al. Effects of testosterone treatment in older men. N Engl J Med 2016; 374: 611-624.
  175. Wittert G, Bracken K, Robledo KP, et al. Testosterone treatment to prevent or revert type 2 diabetes in men enrolled in a lifestyle programme (T4DM): a randomised, double blind, placebo-controlled, 2-year, phase 3b trial. Lancet Diabetes Endocrinol 2021; 9: 32-45.
  176. Snyder PJ, Bhasin S, Cunningham GR, et al. Lessons from the Testosterone Trials. Endocr Rev 2018; 39: 369-386.
  177. Blair SN, Kampert JB, Kohl HW, et al. Influences of cardiorespiratory fitness and other precursors on cardiovascular disease and all-cause mortality in men and women. JAMA 1996; 276: 205-10.
  178. Sui X, LaMonte MJ, Laditka JN, et al. Cardiorespiratory fitness and adiposity as mortality predictors in older adults. JAMA 2007; 298: 2507-2516.
  179. Kodama S, Saito K, Tanaka S, et al. Cardiorespiratory fitness as a quantitative predictor of all-cause mortality and cardiovascular events in health men and women: a meta-analysis. JAMA 2009; 301: 2024-2035.
  180. Ross R, Blair SN, Arena R, et al. Importance of assessing cardiorespiratory fitness in clinical practice: a case for fitness as a clinical vital sign: a scientific statement from the American Heart Association. Circulation 2016; 134: e653-e699.
  181. Chasland LC, Yeap BB, Maiorana AJ, et al. Testosterone and exercise: effects on fitness, body composition, and strength in middle-to-older aged men with low normal serum testosterone levels. Am J Physiol Heart Circ Physiol 2021; 320: H1985-H1998.
  182. Traustadottir T, Harman SM, Tsitouras P, et al. Long-term testosterone supplementation in older men attenuates age-related decline in aerobic capacity. J Clin Endocrinol Metab 2018; 103: 2861-2869.
  183. Bhasin S, Woodhouse L, Casaburi R, et al. Older men are as responsive as young men to the anabolic effects of graded doses of testosterone on the skeletal muscle. J Clin Endocrinol Metab 2005; 90: 678-688.
  184. Page ST, Amory JK, Bowman FD, et al. Exogenous testosterone (T) alone or with finasteride increases physical performance, grip strength, and lean body mass in older men with low serum T. J Clin Endocrinol Metab 2005; 90: 1502-1510.
  185. Haddad RM, Kennedy CC, Caples SM, et al. Testosterone and cardiovascular risk in men: a systematic review and meta-analysis of randomized placebo-controlled trials. Mayo Clin Proc 2007; 82: 29-39.
  186. Xu L, Freeman G, Cowling BJ, Schooling CM. Testosterone therapy and cardiovascular events among men: a systematic review and meta-analysis of placebo-controlled randomized trials. BMC Med 2013; 11: 108.
  187. Ruige JB, Ouwens DM, Kaufman J-M. Beneficial and adverse effects of testosterone on the cardiovascular system in men. J Clin Endocrinol Metab 2013; 98: 4300-4310.
  188. Corona G, Maseroli E, Rastrelli G, et al. Cardiovascular risk associated with testosterone-boosting medications: a systematic review and meta-analysis. Expert Opin Drug Saf 2014; 13: 1327-1351.
  189. Borst SE, Shuster JJ, Zou B, et al. Cardiovascular risks and elevation of serum DHT vary by route of testosterone administration: a systematic review and meta-analysis. BMC Medicine 2014; 12: 211.
  190. Alexander GC, Iyer G, Lucas E, Lin D, Singh S. Cardiovascular risks of exogenous testosterone use among men: a systematic review and meta-analysis. Am J Med 2017; 130: 293-305.
  191. Elliott J, Kelly SE, Millar AC, et al. Testosterone therapy in hypogonadal men: a systematic review and network meta-analysis. BMJ Open 2017; 7: e015284.
  192. Corona G, Rastrelli G, Di Pasquale G, et al. Testosterone and cardiovascular risk: meta-analysis of interventional studies. J Sex Med 2018; 15: 820-838.
  193. Diem SJ, Greer NL, MacDonald R, et al. Efficacy and safety of testosterone treatment in men: an evidence report for a clinical practice guideline by the American College of Physicians. Ann Intern Med 2020; 172: 105-118.
  194. Hudson J, Cruickshank M, Quinton R, et al. Adverse cardiovascular events and mortality in men during testosterone treatment: an individual patient and aggregate data data meta-analysis. Lancet Healthy Longev 2022; 3: e381-393.
  195. Bhasin S, Lincoff AM, Basaria S, et al. Effects of long-term testosterone treatment on cardiovascular outcomes in men with hypogonadism: Rationale and design of the TRAVERSE study. Am J Heart 2022; 245: 41-50.
  196. Shores MM. Testosterone treatment and cardiovascular events in prescription database studies. Asian J Androl 2018; 20: 138-144.
  197. Shores MM, Smith NL, Forsberg CW, et al. Testosterone treatment and mortality in men with low testosterone levels. J Clin Endocrinol Metab 2012; 97: 2050-2058.
  198. Muraleedharan V, Marsh H, Kapoor D, et al. Testosterone deficiency is associated with increased risk of mortality and testosterone replacement improves survival in men with type 2 diabetes. Eur J Endocrinol 2013; 169: 725-733.
  199. Vigen R, O’Donnell CI, Baron AE, et al. Association of testosterone therapy with mortality, myocardial infarction, and stroke in men with low testosterone levels. JAMA 2013; 310: 1829-1836.
  200. Finkle WD, Greenland S, Ridgeway GK, et al. Increased risk of non-fatal myocardial infarction following testosterone therapy prescription in men. PLoS One 2014; 9: e85805.
  201. Baillargeon J, Urban RJ, Kuo Y-F, et al. Risk of myocardial infarction in older men receiving testosterone therapy. Ann Pharmacother 2014; 48: 1138-1144.
  202. Sharma R, Oni OA, Gupta K, et al. Normalization of testosterone level is associated with reduced incidence of myocardial infarction and mortality in men. Eur Heart J 2015; 36: 2706-2715.
  203. Anderson JL, May HT, Lappe DL, et al. Impact of testosterone replacement therapy on myocardial infarction, stroke and death in men with low testosterone concentrations in an integrated health care system. Am J Cardiol 2016; 117: 794-799.
  204. Wallis CJD, Lo K, Lee Y, et al. Survival and cardiovascular events in men treated with testosterone replacement therapy: an intention-to-treat observational cohort study. Lancet Diabetes Endocrinol 2016; 4: 498-506.
  205. Cheetham TC, An JJ, Jacobsen SJ, et al. Association of testosterone replacement with cardiovascular outcomes among men with androgen deficiency. JAMA Intern Med 2017; 177: 491-499.
  206. Loo SY, Azoulay L, Nie R, et al. Cardiovascular and cerebrovascular safety of testosterone replacement therapy among aging men with low testosterone levels: a cohort study. Am J Med 2019; 132: 1069-1077.
  207. Oni OA, Dehkordi SHH, Jazayeri M-A, et al. Relation of testosterone normalization to mortality and myocardial infarction in men with previous myocardial infarction. Am J Cardiol 2019; 124: 1171-1178.
  208. Shores MM, Walsh TJ, Korpak A, et al. Association between testosterone treatment and risk of incident cardiovascular events among US male veterans with low testosterone levels and multiple medical comorbidities. J Am Heart Assoc 2021; 10: e020562.
  209. Morgentaler A, Lunenfeld B. Testosterone and cardiovascular risk: world’s experts take unprecedented action to correct misinformation. Aging Male 2014; 17: 63-65.
  210. Vigen R, O’Donnell CI, Baron AE, et al. Association of testosterone therapy with mortality, myocardial infarction, and stroke in men with low testosterone levels. JAMA 2013; 310: 1829-1836. Erratum published in JAMA 2014; 311: 967.
  211. Swerdloff RS, Pak Y, Wang C, et al. Serum testosterone (T) level variability in T gel-treated older hypogonadal men: treatment monitoring implications. J Clin Endocrinol Metab 2015; 100: 3280-3287.
  212. Layton JB, Meier CR, Sharpless JL, et al. Comparative safety of testosterone dosage forms. JAMA Intern Med 2015; 175: 1187-1196.
  213. Martinez C, Suissa S, Rietbrock S, et al. Testosterone treatment and risk of venous thromboembolism. BMJ 2016; 355: i5968.
  214. Houghton DE, Alsawas M, Barrioneuvo P, et al. Testosterone therapy and venous thromboembolism: a systematic review and meta-analysis. Thromb Res 2018; 172: 94-102.
  215. Nieschlag E, Vorona E. Medical consequences of doping with anabolic androgenic steroids: effects in reproductive functions. Eur J Endocrinol 2015; 173: R47-R58.
  216. Goldman AL, Pope HG, Bhasin S. The health threat posed by the hidden epidemic of anabolic steroid use and body image disorders among young men. J Clin Endocrinol Metab 2018; doi:10.1210/jc.2018-01706.
  217. Baggish AL, Weiner RB, Kanayama G, et al. Cardiovascular toxicity of illicit anabolic-androgenic steroid use. Circulation 2017; 135: 1991-2002.
  218. Ng MTF, Hoermann R, Bracken K, et al. Effect of testosterone treatment on bone microarchitecture and bone mineral density in men: a 2-year RCT. J Clin Endocrinol Metab 2021; 106: e3143-3158.