Int J Sports Med 2023; 44(02): 81-94
DOI: 10.1055/a-1909-1196
Review

Clinical Concerns on Sex Steroids Variability in Cisgender and Transgender Women Athletes

Luigi Di Luigi
1   Department of Movement, Human and Health Sciences, Università degli Studi di Roma 'Foro Italico', Rome, Italy
,
Emanuela A Greco
1   Department of Movement, Human and Health Sciences, Università degli Studi di Roma 'Foro Italico', Rome, Italy
2   Department of Science of Movement, Università degli Studi Niccolò Cusano, Rome, Italy
,
Chiara Fossati
1   Department of Movement, Human and Health Sciences, Università degli Studi di Roma 'Foro Italico', Rome, Italy
,
Antonio Aversa
3   Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
,
Paolo Sgrò
1   Department of Movement, Human and Health Sciences, Università degli Studi di Roma 'Foro Italico', Rome, Italy
,
Cristina Antinozzi
1   Department of Movement, Human and Health Sciences, Università degli Studi di Roma 'Foro Italico', Rome, Italy
› Author Affiliations
 

Abstract

In the female athletic community, there are several endogenous and exogenous variables that influence the status of the hypothalamus-pituitary-ovarian axis and serum sex steroid hormones concentrations (e. g., 17β-estradiol, progesterone, androgens) and their effects. Moreover, female athletes with different sex chromosome abnormalities exist (e. g., 46XX, 46XY, and mosaicism). Due to the high variability of sex steroid hormones serum concentrations and responsiveness, female athletes may have different intra- and inter-individual biological and functional characteristics, health conditions, and sports-related health risks that can influence sports performance and eligibility. Consequently, biological, functional, and/or sex steroid differences may exist in the same and in between 46XX female athletes (e. g., ovarian rhythms, treated or untreated hypogonadism and hyperandrogenism), between 46XX and 46XY female athletes (e. g., treated or untreated hyperandrogenism/disorders of sexual differentiation), and between transgender women and eugonadal cisgender athletes. From a healthcare perspective, dedicated physicians need awareness, knowledge, and an understanding of sex steroid hormones’ variability and related health concerns in female athletes to support physiologically healthy, safe, fair, and inclusive sports participation. In this narrative overview, we focus on the main clinical relationships between hypothalamus-pituitary-ovarian axis function, endogenous sex steroids and health status, health risks, and sports performance in the heterogeneous female athletic community.


#

Introduction

From a clinical perspective, female athletes represent a highly heterogeneous community ([Table 1]). Due to both physiological and non-physiological factors, there exists different hypothalamus-pituitary-ovarian (HPO) axis and sex steroid hormone statuses. In addition, there are female athletes with different sex chromosomes (e. g., 46XX, 46XY, and mosaicism) and endocrine profiles due to endocrine diseases with altered genetic expression such as hyperandrogenism, with or without disorders of sexual differentiation (DSD), and due to gender dysphoria ([Table 2]). Consequently, the genetic sex and general features of female athletes should not be taken for granted. Furthermore, due to genetic traits, HPO-axis status, and sex steroid hormone variability, these athletes will have different intra- and inter-individual biological and functional characteristics, and thus different gonadal statuses, health risks, and exercise capacities.

Table 1 The female athletic community: a very heterogeneous genetic and biological environment.

Eugonadal Female Athletes (46 XX)

Non-eugonadal Female Athletes (46 XX)

Non-eugonadal Female Athletes (46 XY)

Transgender Women Athletes (46 XY)

Childhood (<8 years)
Adolescence (9–18 years)

  • Pre-puberty

  • Puberty

  • Post-puberty

Early Middle adulthood

  • Reproductive phase±OC

  • Pregnancy

  • Post-menopause±HRT

Later adulthood (>65 years)

Primary hypogonadismsSecondary hypogonadisms Sportrelated hypogonadismsHyperandrogenisms/DSD:

  • PCOS

  • CAH

  • Ovarian/Adrenal tumors

  • Idiopathic

  • AAS abuse

Ovotestis DSD Genetic mosaicisms

Hyperandrogenisms/DSD:

  • 5α-reductase deficiency

  • 17β-HSD3 deficiency

  • CAIS, PAIS

Gonadal dysgenesis Ovotestis DSD Genetic mosaicisms

Treated before puberty

  • GnRH analogues

  • GAHT

  • ±Anti-androgens

  • ±Surgery

Treated after puberty

  • GAHT

  • ±Anti-androgens

  • ±Surgery

Untreated (Not GAHT±anti-androgens)

AAS, androgenic anabolic steroids; CAH, classic and late onset congenital adrenal hyperplasia; CAIS, complete androgen insensitivity; DSD, disorder of sexual differentiation; GnRH, gonadotropin-releasing hormone; GAHT, gender-affirming hormone therapy (i. e., exogenous 17β-estradiol or other estrogens); HRT, hormone replacement therapy; OC, oral contraceptive; PAIS, partial androgen insensitivity; PCOS, polycystic ovary syndrome, 17β-HSD3, 17β-hydroxysteroid dehydrogenase type 3. Please note all reported female athletes could abuse with AAS and other substances influencing the hypothalamus-pituitary-gonadal axis.

Table 2 Examples of treatment-related variability in serum steroid hormone concentrations in female athletes affected by sport-related hypogonadism, in some diseases causing hyperandrogenism, and in transgender women athletes treated with gender-affirming hormonal therapy (e. g., exogenous 17β-estradiol or other estrogens,±anti-androgens) or untreated.

HORMONES

Sport-Related Hypogonadism

CAH/PCOS

Transgender Women Athletes (Treated/Untreated)

Estrogens

decreased/normal*

decreased/normal*

increased/normal#

Progestogens

decreased/normal*

decreased/normal*

normal

17OH Progesterone

normal

increased/normal*

normal

Testosterone

decreased/normal

increased/normal*

decreased/normal#

DHT

decreased/normal

increased/normal*

decreased/normal#

Androstenedione

decreased/normal

increased/normal*

decreased/normal#

DHEA, DHEAS

decreased/normal

increased/normal*

decreased/normal#

Cortisol

normal

decreased/normal*

normal

Aldosterone

normal

decreased/normal*

normal

CAH, classic and late-onset congenital adrenal hyperplasia; DHEA, dehydroepiandrosterone; DHEAS, dehydroepiandrosterone sulphate; DHT, dihydrotestosterone; Estrogens, endogenous or exogenous 17β-estradiol or other estrogens; PCOS, polycystic ovary syndrome. Please note all reported female athletes may assume, for therapy and/or doping purposes, prohibited and non-prohibited substances further influencing their steroid hormone status. *Depending on diseases and performed therapy. #With respect to the physiological levels in untreated 46XY males.

Given this context, there are many unresolved clinical and ethical concerns that have been reported in the female athletic community. For instance, in sports-related hypogonadism, there are concerns regarding responsibility, health prevention, and early differential diagnosis. Moreover, concerns regarding health protection, sports eligibility, and fairness have been remarked upon for female athletes affected by untreated hyperandrogenism and those treated for gender dysphoria, particularly when they compete against eugonadal cisgender athletes.

Due to the relative novelty of these issues and difficulties in experimental standardization, few reproducible and comparable scientific investigations that are useful for a systematic analysis are available. Furthermore, for ethical and technical reasons, not all clinical conditions can be investigated in all sports. In existing observational studies, the data are often controversial, and some conditions, such as the effects of hyperandrogenism, are likely to be underestimated due to collider bias [1]. Unfortunately, in this area of investigation, demagogic, economic, legal, opportunistic, political, social, and/or speculative issues often dominate over health-related issues and sports medicine competences and responsibilities.

To sensitize sport physicians in supporting safe and physiologically healthy sports participation, this narrative overview focuses on several clinical issues concerning sex steroid hormones, gonadal status, and exercise performance in three groups of adult female athletes: a) 46XX normo-androgenic, b) 46XX and 46XY individuals affected by hyperandrogenism/DSD, and c) 46XY male-to-female transgender women (TW). The links between sex steroid hormones and exercise performance are considered of clinical relevance even because, in some clinical situations, the normalization of hormonal imbalance could worsen individual sport performance, thus representing a possible negative factor for some female athletes, even if necessary for health and wellness.

As this is a controversial topic, we attempted to restrict our focus to the clinical context of sports endocrinology, without discussing ethical, legal, political, or sports regulations that have been adequately discussed elsewhere [2] [3] [4] [5] [6] [7] [8] [9] [10].


#

46XX normo-androgenic adult female athletes

The pleiotropic effects of sex steroid hormones

Following puberty, female sex steroid hormones are secreted by the ovaries in a cyclical, monthly rhythm (17β-estradiol mainly during the follicular phase and progesterone during the luteal phase) under the pulsatile control of the hypothalamic gonadotropin-releasing hormone (GnRH) and the pituitary gonadotropins, such as luteinizing hormone (LH) and follicle-stimulating hormone (FSH) [11]. Depending on exercise intensity and HPO-axis status, serum 17β-estradiol and progesterone may rapidly increase after exercise [12]. Moreover, exercise directly activates estrogen receptor alpha (ERα) in muscles, up-regulating myogenic-related gene expression independently of the serum 17β-estradiol levels [13].

In normo-androgenic, eugonadal female athletes, androgens are physiologically produced, with serum concentrations like adult males for androstenedione, dehydroepiandrosterone (DHEA), and dehydroepiandrosterone sulphate (DHEAS), and at substantially lower concentrations for dihydrotestosterone (DHT) and testosterone. Half of circulating testosterone is secreted in equal amounts by the ovaries and adrenal glands, and the other 50% is produced by the peripheral conversion of ovarian and adrenal androstenedione [14]. Testosterone is transformed into DHT by the enzyme 5α-reductase in different organs and tissues. Under physiological conditions, DHEA and DHEAS, which are produced by the adrenal glands (90%) and ovaries (10%), are converted to 17β-estradiol, testosterone, and DHT [14].

The 17β-estradiol and progesterone, by acting on respective receptors (e. g., ERα, ERβ, PR) – and the variations of 17β-estradiol/progesterone ratio – exert different pleiotropic effects influencing exercise physiology and sporting performance ([Table 3]) [15] [16] [17] [18] [19] [20]. In terms of exercise physiology, 17β-estradiol: a) increases 5' AMP-activated protein kinase, glucose transporter type 4, and insulin receptor substrate activation, increasing insulin sensitivity; b) increases glucose uptake and glycogen storage in muscles by reducing glycogen utilization/kinetics and liver gluconeogenesis; c) controls mitochondrial biogenesis, oxygen consumption, and mitochondrial DNA transcription; d) increases the contraction-stimulated glucose uptake in type I muscle fibers, which is beneficial during high-intensity aerobic exercise; e) favorably affects motor behavior and muscle-force generation by acting directly on tendon and muscle proteins, independent of physical activity; f) blunts muscle response to intense exercise and preserves muscle function after exercise-induced muscle damage; g) induces the normal growth hormone response to resistance exercise; and h) increases epinephrine response to exercise, whereas cortico-adrenal response to exercise seems to be uninfluenced by circulating estrogen serum concentrations [15] [17] [21] [22] [23] [24] [25]. Taken together, the metabolic and bio-energetic effects of 17β-estradiol appear to primarily support sports endurance. In addition, progesterone decreases insulin sensitivity, the glycogen-sparing effect of 17β-estradiol and liver gluconeogenesis by decreasing contraction-stimulated glucose uptake in type I muscle fibers [16] [17]. Female sex steroid hormones decrease the osmotic threshold for arginine vasopressin and thirst stimulation and led to greater fluid retention during hypertonic saline infusion. Moreover, female sexual hormones concentrations positively correlate with ventilatory parameters (e. g., tidal volume, inspiratory and expiratory times) and peak expiratory flow, playing a positive role in respiratory control and thoracic pump muscles in the luteal phase [26] [27].

Table 3 Examples of non-reproductive biological effects of physiological female sex steroid hormones concentrations (17β-estradiol and progesterone) that could influence health status, exercise capacity and sporting performance in female athletes (in alphabetical order).

Sex Hormone

Glucose Metabolism

Lipid and Protein Metabolisms

Other Effects*

17β-estradiol

Increases
AMPK/GLUT4/IRS1 activation
Glycogen storage
Insulin sensitivity and signaling
Muscles glucose uptake
Type I fibers glucose uptake
(Contraction-stimulated)

Increases
FFA availability
FFA oxidation
FFA uptake
Lipolysis
Muscles lipid stores
Liver lipogenesis
WAT lipogenesis

Increases
Bone mineral density
GH-IGF-I secretion
GH response to exercise
Mood state
Motor/walking behavior
Muscles mass
Muscle force generation
Recovery from muscle damage
Sense of well-being

Decreases
Glycogen utilization
Glucose kinetic
Liver gluconeogenesis

Decreases
Protein catabolism
Protein oxidation

Regulates
CNS neurotransmission (i. e., dopamine)
CNS reward responses
Mood state
Osmotic threshold for AVP
Renal fluid/sodium absorption
Sodium appetite
Stress hormones responses
Thirst control
Vascular tone

Progesterone

Increases
Insulin resistance

Increases
Fat utilizationProtein catabolism

Increases
CNS sensitivity to PaCO2

Decreases
Glycogen-sparing effects of 17β-E2
Glucose kinetic
Hepatic gluconeogenesis
Type I fibers glucose uptake
(Contraction-stimulated)

Regulates
Osmotic threshold for AVP
Renal fluid/sodium absorption (i. e., anti-aldosterone effect)
Sodium appetite
Thirst control Ventilatory parameters (e. g., tidal volume, inspiratory and expiratory times) and peak expiratory flow

17β-estradiol/ Progesterone ratio

Influences/Regulates
Motor behavior, body composition, body temperature, breathing, cardiovascular system, cognitive processes, fluid intake, muscle metabolism and bioenergetics, mood state, substrate metabolism, thermoregulation, water, and salt balance

AMPK, 5' AMP-activated protein kinase; AVP, arginine-vasopressin; CNS, central nervous system; FFA, free fatty acids; GH, growth hormone; GLUT4, glucose transporter type 4; IGF-I, insulin growth factors-I; IRS1, insulin receptor substrate 1; PaCO2, carbon dioxide partial pressure; WAT, white adipose tissue; 17β-E2, 17β-estradiol. *Related to direct effects of 17β-estradiol and/or progesterone and/or 17β-estradiol/progesterone ratio.

In female athletes, endogenous androgens also exert pleiotropic effects that may influence exercise physiology and performance depending on their serum concentrations and androgen-receptor (AR) distribution and sensitivity [28] ([Table 4]).

Table 4 Non-reproductive biological effects of testosterone influencing health status, exercise capacity, and sporting performance in male and female athletes (in alphabetical order). (Modified from Sgrò P, Di Luigi L. Sport and male sexuality. J Endocrinol Invest 2017; 40: 911–923 [28]).

Somatic Growth Body Composition

Metabolisms

Behaviour

Increases
Bone mineral density
Epiphyseal cartilage closure
Erythropoiesis and hemoglobin
Muscle mass and male distribution
Secondary sexual characteristics
Somatic masculinization

Increases
Anaerobic glycolytic capacity
Enzyme’s activity in mitochondria
Phosphocreatine content in muscles
Protein anabolism
Sarco-tubular enzymes activity

Increases
Aggressiveness
Dominance
Inclination to command

Decreases
Fat mass

Decreases
Protein catabolism

Decreases
Empathy
Negative reaction to external stimuli Perception of negative emotions Sense of fatigue

Endocrine system
Psycho-Physical Stress

Functional Skills

Psycho-Functional Skills

Growth control
Inhibits CRH-ACTH- Cortisol responses to stress
Insulin-like effects
Peripheral anti-glucocorticoids effects (for competition at receptor level)
Pro-insulin effects
Synergistic effects with growth hormone

Increases
Aerobic and anaerobic capacity
Cardiovascular efficiency
Explosive strength
Muscle strength
Muscle adaptation to training
Neuromuscular excitability
Neuromuscular conduction

Increases
Aggressiveness in competition
Motivation to compete
Resistance to fatigue
Visual-spatial orientation

Decreases
Negative reaction to alarms

ACTH, adrenocorticotropin hormone; CRH, corticotropin-releasing hormone

Due to physiological variability and the balance of 17β-estradiol and progesterone, fine-tuning exercise performance may change according to HPO-cycle phases, with monthly variations in different sport performance measures such as maximal aerobic capacity (VO2max), endurance capacity, time to exhaustion, time trial performance, training strain and monotony [17] [29] [30] [31]. Furthermore, menstrual bleeding, body weight increases in the mid-late luteal phase, mood fluctuations, and/or premenstrual-syndrome (e. g., anxiety, breast tenderness, headache, inflammatory state, irritability, mood disorders, and weight increase) can negatively affect sports performance in athletes [30] [31] [32] [33]. These data suggest that monitoring HPO-cycle phases and symptoms might provide useful feedback for supporting the health status in female athletes in devising training and predicting results in competitions [30] [32].


#

Sports-related hypothalamus-pituitary-ovarian axis alterations

The HPO axis in adult 46XX female athletes can be altered by common diseases such as primary or secondary hypogonadism and/or by sports-related factors, such as energy deficiency due to the combination of high energy expenditure (during exercise training) and insufficient caloric intake. The hormonal profile in elite athletes, both males and females, is largely influenced by the type of sport [9]. In particular, it has been reported that a high percentage of female elite athletes (up to 60% of gymnasts and dancers) have serious reproductive disorders such as oligomenorrhea, functional hypothalamic amenorrhea, anovulation, and infertility due to sports-related hypogonadism caused by energy deficiency and marked alterations in body composition, which in turn determine an increased activity of the hypothalamic-pituitary-adrenal (HPA) axis [34]. Indeed, the main sports-related factors that, either alone or in association with other complex mechanisms, inhibit the HPO axis are relative energy deficiency in sports (RED-S) and psycho-physical stress [35] [36] [37] [38] [39]. In addition to the reproductive consequences of HPA inhibition ([Table 5]), RED-S could be associated with decreased bone mineral density, impaired bone microarchitecture, and decreased bone strength, increasing the risk of bone stress fractures in athletes. Moreover, RED-S could be responsible per se for other non-reproductive health consequences: iron deficiency anemia, growth retardation in adolescence because of a GH secretion disorder, endothelial dysfunction, gastrointestinal disorder (e. g., constipation), or increased susceptibility to gastrointestinal and respiratory tract illness due to alteration of the immunological system. The main strategy to prevent/counteract the complications of RED-S is nutritional education, and changes in food choice and intake in relation to individual energy expenditure; particularly, supplementation with vitamin D and calcium should be considered, independently of normal nutrition practices in order to prevent bone tissue alterations [37] [38]. Apart from RED-S and psycho-physical stress, the etiology of sports-related hypogonadism is multi-factorial and complex, and includes age, drugs, epigenetics and genetic factors, social issues, type of sport and training, and the abuse of prohibited substances (i. e., mainly androgenic anabolic steroids (AAS)), which all exhibit substantial inter-individual variability [40]. In most cases, the first alteration is progressively altered GnRH secretions, which reduce LH pulse frequency and cause a progressive and often biphasic reduction of female sex steroid hormones (i. e., firstly progesterone and successively 17β-estradiol) and a reduction of LH-dependent androgens concentration.

Table 5 Symptoms and health complications in female athletes affected by sport-related hypogonadism and by diseases causing hyperandrogenism/disorders of sexual differentiation, and in transgender women athletes treated with gender-affirming hormone therapy (e. g., exogenous 17β-estradiol or other estrogens,±anti-androgens) or untreated (in alphabetical order).

SYMPTOMS AND HEALTH COMPLICATIONS

Sport-Related Female Hypogonadism (+/− RED-S)

Female Athletes with Hyperandrogenism/DSD

Transgender Women Athletes* (Treated with GAHT/Untreated)

Reproduction
Amenorrhea
Anovulation
Infertility
Oligomenorrhea

Reproduction/Sexuality
Erectile Dysfunction#
Genital Pain
Infertility

General Health
Anxiety
Cardiovascular Diseases
Depression
Eating Disorders
Fat Mass Increase
Hypertension
Hypertriglyceridemia
Iatrogenic Risk
Impaired Fasting Glycemia
Inguinal Hernia (?)
Insulin Resistance
Muscle Hypotrophy
Prolactinomas
Substance Abuse
Suicidality
Thromboembolism

Reproduction/Sexuality
Breast Atrophy
Delayed Puberty

General Health
Arrhythmias
Cardiovascular Diseases
Cognitive Impairment
Depression
Early Atherosclerosis
Eating Disorders
Endothelial Dysfunction
Gastrointestinal Illness
Iatrogenic Risks
Increased Inflammation
Insulin Resistance
Iron Deficiency, Anemia
Low IgA
Low BMI
Reduced Feminization
Reduced Linear Growth
Reduced Peak BMD, Osteopenia, Osteoporosis,
and Fractures
Reduced RMR
Respiratory Illness

Reproduction/Sexuality
Acne
Androgenic Alopecia
Breast Atrophy
Clitoromegaly
Common Urogenital Sinus
Delayed Puberty
Fused Labia Majora
Hirsutism
Male Muscle Distribution
Pseudo-Puberty
Reduced Feminization
Voice Deepening
Delayed Puberty

General Health
Adrenal Insufficiency
Cardiovascular Diseases
Diabetes
Gonadal Cancer
Hyperinsulinemia
Hypertension
Iatrogenic Risks
Insulin Resistance
Obesity
Salt Wasting
Short Stature

BMI, body mass index; BMD, bone mineral density; DSD, disorders of sexual differentiation; GAHT, gender-affirming hormone therapy; iatrogenic risk: all reported female athletes may assume, as therapy or for doping purposes, prohibited and non-prohibited substances that could further influence health status for their side effects; IgA, immunoglobulin A; RED-S, relative energy deficit in sport; RMR, resting metabolic rate. *The symptoms in transgender women athletes are multifactorial because they could be related to the assumed drugs, to hypoandrogenism/hyperestrogenism and to a multitude of social reasons, independently of GAHT, see text. #in the absence of surgical sex reassignment.

Where health risks are concerned, sports-related female hypogonadism, especially in cases of delayed treatment, is associated with serious reproductive symptoms and non-reproductive complications, particularly in the presence of RED-S ([Table 5]) [36] [37] [38].

Furthermore, it is not well known if and how the reduction of sex steroid hormones influences the fine adjustment of sports performance. For instance, in amenorrheic athletes, neuromuscular alterations have been reported that include reduced knee muscular strength/endurance and prolonged reaction times, [41]. In cases of oligomenorrhea and in presence of an increased 17β-estradiol/progesterone ratio, due to progesterone reduction, a negative effect on sports performance may be related to water retention, increased body weight, and other unknown mechanisms. Moreover, the absence of 17β-estradiol and progesterone could lead to advantages in sports performance due to the absence of menstrual bleeding, body weight and mood fluctuations, and premenstrual syndrome [30] [31] [32] [33].


#
#

46XX and 46XY female athletes affected by hyperandrogenism

The clinical profile of hyperandrogenism

Female athletes can be affected by a wide spectrum of genetic diseases that influence ovarian and/or adrenal steroid hormone production, and they are characterized by variable supra-physiological serum concentrations of testosterone, DHT, androstenedione, DHEA and/or DHEAS, as well as variable decreases in 17β-estradiol, progesterone, cortisol, and aldosterone serum concentrations, also depending on performed therapy ([Tables 1] [2]).

Health complications may be observed, which depend on the features of the primary disease, the time of onset of hyperandrogenism (e. g., in the fetus, at birth, at puberty, in adulthood), AR distribution and sensitivity, time of diagnosis, therapy (treated or untreated), specific symptoms and different endocrinological situations. Generally, female athletes may have symptoms of abnormal gonadal and DSD with ambiguous genitalia, which may range from a nearly male appearance to minimal clitoromegaly, and/or symptoms of virilization, differently associated symptoms of female hypogonadism, altered adrenal steroid secretion (e. g., aldosterone, cortisol), and increased risk of general short- and long-term health complications, particularly if they are not adequately diagnosed and treated ([Table 5]).

When hyperandrogenism is associated with DSD, a different phenotype at birth may be observed and when ambiguous genitalia lead to difficulties in diagnosis and final sex assignment, a female gender is assigned at birth in most cases. The conditions of hyperandrogenism associated with DSD are genetic disorders such as classic congenital adrenal hyperplasia (CAH), 5α-reductase deficiency, 17β-hydroxysteroid dehydrogenase type 3 (17β-HSD3) deficiency, partial or complete androgen insensitivity syndrome (AIS), and ovotestis DSD. The symptoms of virilization may be observed at birth or puberty and are often associated with primary amenorrhea. In some cases of DSD associated with a completely normal female phenotype, a diagnosis is not made until they are investigated for primary amenorrhea or found to have a high serum testosterone concentration when blood is analyzed for clinical or anti-doping purposes.


#

Congenital adrenal hyperplasia

Classic CAH comprises a group of autosomal recessive disorders that cause the deficiency of specific enzymes involved in the adrenal steroidogenesis. The common form is 21-hydroxylase deficiency (21-OHD) due to mutations in the 21-hydroxylase (CYP21A2) gene; other virilizing forms include 3β-hydroxysteroid dehydrogenase and 11β-hydroxylase deficiencies, associated with mutations in the 3β-hydroxysteroid dehydrogenase (HSD3B2) and 11β-hydroxylase (CYP11B1) genes, respectively [42]. The features associated with classic CAH encompass a wide clinical spectrum reflecting the specific mutation, and the clinical manifestations of 21-OHD deficiency range from salt-losing syndrome and severe virilizing forms to the mild forms. Classic CAH generally appears in the neonatal period and the presentations of clinical features differ depending on the chromosomal sex of the affected infant. Salt-losing CAH is a medical emergency because of the risk of hyponatremia, hyperkalemia, hypotension, and fatal outcome within the first 2–3 weeks of life if not recognized. In addition, the extent of prenatal virilization can lead to mis-assignment of sex at birth. Infant females with classic CAH generally present ambiguous genitalia in the neonatal period, from a nearly male appearance with penile urethra and bilateral undescended testes to minimal clitoromegaly, with normal female internal genitalia. The most common physical findings in affected girls include clitoromegaly, fused labia majora, and a single perineal orifice; occasionally, the minimally virilized girl may not be identified until progressive clitoromegaly prompts a medical evaluation. Both male and female children with CAH can present with premature pubarche, tall stature, accelerated linear growth velocity, and advanced skeletal maturation. Symptoms of CAH in adolescent females include hirsutism, irregular menses, chronic anovulation, acne, and infertility, with hirsutism being the most common presenting feature [42] [43] [44]


#

5α-Reductase deficiency

The 5α-reductase deficiency is a very rare autosomal recessive condition causing an altered conversion of testosterone into DHT. Since during fetal life the development of male external genitalia is DHT-dependent, this condition leads to a male’s under-virilization with identification as a female gender at birth, despite the presence of testes and normal testosterone production. During and after puberty, when circulating levels of testosterone rise in the normal adult male serum concentrations, the female individuals will undergo increasing virilization and more than half will change their gender identity, becoming males [6].


#

17β-Hydroxysteroid Dehydrogenase Type 3 Gene Mutation

A mutation of the 17β-HSD3 gene leads a reduced conversion of androstenedione to testosterone. These 46XY individuals develop under-virilized external genitalia, with some being identified as female at birth. As in the case of 5α-reductase deficiency, when the testes start to produce androgens at puberty, such individuals undergo marked virilization and approximately half of them will change their gender identity to male (6).


#

Androgen insensitivity syndrome

AIS, caused by a mutation in the androgen receptor gene, causes various features of under-virilization in 46XY individuals. In the case of complete AIS (CAIS), even though they have testes (undescended) and normal levels of serum testosterone at a different age, they respond very little or not at all to androgens and will therefore appear as fully female. In the case of partial AIS (PAIS), the phenotype will vary from that of a virilized woman to an under-virilized man (6). Moreover, distinguishing the degree of androgen insensitivity is difficult and somewhat controversial. Many cases of AIS are completely normal anatomical females and not diagnosed until adulthood. Those who are diagnosed before adulthood have severe AIS, but proving someone has complete AIS is not possible. By definition, a person undiagnosed before adulthood, e. g., those identified via a blood test for an athletic biological passport, have a CAIS.


#

Ovotestis DSD

The ovotestis DSD (true hermaphroditism) is a very rare condition with varying karyotype, although 46XX is common. These individuals develop both ovarian and testicular tissue; they can have an ovary on one side and a testis on the other or combined tissue, so-called ovotestis. Depending on their gonadal tissue, their clinical features varied from that of a normal man to normal woman, although the external genitalia are generally ambiguous, and little is known about their final gender identity [6].

When the hyperandrogenism is not associated with DSD, the symptoms of virilization and female hypogonadism are often observed during or immediately after puberty, or even later. Conditions of female hyperandrogenism without DSD are polycystic ovary syndrome (PCOS), late-onset CAH, adrenal/ovarian androgen-secreting tumors, and idiopathic hyperandrogenism.


#

Polycystic ovary syndrome

PCOS is the most frequent endocrine disorder among general female population, and particularly elite female athletes [6] [45]. PCOS is characterized by high ovarian production of androgens, disorders of ovulation (anovulatory cycles), and polycystic ovarian morphology. Although the etiology of PCOS remains largely unclear, probably it is based on a genetic predisposition. Hyperandrogenism and insulin resistance are the typical endocrine features of PCOS that alter the pulsatile GnRH secretion, resulting in high LH secretion and relative FSH deficiency. The altered LH/FSH ratio causes the characteristic polycystic aspect of the ovaries, anovulation, menstrual disorders, and reduced fertility as well as hirsutism and acne [45]. In addition, women with PCOS often show insulin resistance with secondary insulin hypersecretion, which directly stimulates ovarian androgen production. Moreover, insulin inhibits hepatic synthesis of sex hormone-binding globulin (SHBG), resulting in increased levels of free (bioavailable) testosterone [44]. Insulin resistance in PCOS is associated with abdominal obesity, type 2 diabetes, dyslipidemia, and increased cardiovascular risk. However, in athletes affected by PCOS, training per se or reported sports-related factors inducing hypogonadism may reduce symptoms and androgen secretion [46] [47] [48].


#

Late-onset congenital adrenal hyperplasia

Late-onset CAH is also known as the non-classic form, and it is considered the mild form of CAH. However, this classification system is somewhat artificial because disease severity is better represented as a continuum based on residual enzyme activity [42]. Late-onset CAH is more common than the classic forms, with an incidence of 1:1000 vs. an incidence range of 1:5000 to 1:15,000, respectively [49]. Female children with late-onset CAH can present with premature pubarche and clitoromegaly [50]. After puberty, the clinical manifestations of late-onset CAH include hirsutism, irregular menses, chronic anovulation, acne, and infertility; whereas hirsutism is the most common clinical symptom [44] [50]. Due to similar clinical features, it may be difficult to distinguish late-onset CAH from PCOS. Generally, women with late-onset CAH present higher 17-OHP and progesterone concentrations than women with PCOS, while insulin resistance, obesity, polycystic ovary morphology, and elevated LH/FSH ratio is more common among women with PCOS. However, none of these features clearly differentiate women with late-onset CAH from those with PCOS [51] nor do Anti-Mullerian hormone serum concentrations [52].


#

Androgen-secreting tumors

Tumors secreting androgens are a rare cause of hyperandrogenism and can originate from ovarian or adrenal tissue; unfortunately, they are malignant in more than 50% of cases [53]. The typical clinical presentation is a very rapid onset of female virilization, whereas small tumors can have more indolent presentations. Physical examination generally reveals abdominal or pelvic masses, and if they are adrenal in origin can be associated with increased DHEA and DHEA-S levels, and hypercortisolemia, also leading to Cushing’s syndrome.


#

Idiopathic hyperandrogenism

Idiopathic hyperandrogenism is characterized by no secondary causes, no genetic alterations, normal menses, normal ovaries on ultrasonography, and elevated androgen levels that are generally the cause of acne and hirsutism [54].

The therapy for diseases causing hyperandrogenism, including cyproterone acetate, fludrocortisone, GnRH analogues, glucocorticoids, oral contraceptives, spironolactone, or surgery should begin immediately following a differential diagnosis, and depending on symptom severity, should be well-tailored to improve the risk/benefit ratio. Female athletes affected by severe hyperandrogenism/DSD and/or adrenal insufficiency should have been diagnosed and treated early in life. When hyperandrogenism appears at puberty or later, therapy should start only after a differential diagnosis is made and in accordance with good clinical practice criteria. In female athletes treated with prohibited drugs (i. e., fludrocortisone, glucocorticoids, and spironolactone), a therapeutic use exemption (TUE) must be requested according to World Anti-Doping Agency criteria [55] [56].

Interestingly, from our unpublished clinical experience, a dichotomic approach to medical therapy for the same type of hyperandrogenism exists. Female non-athletes more often search for the best therapy to reduce their serum androgens, while female athletes more often prefer to avoid any therapy, thereby maintaining their hyperandrogenism and consequently increasing their health complications. We do not know if the stance of female athletes is based on fear of iatrogenic risks or against a normo-androgenic status.


#

Female hyperandrogenism and sports performance

When female diseases inducing hyperandrogenism are undiagnosed or not adequately treated, high concentrations of serum testosterone (serum total testosterone>1.8 nmol/L) and/or of other androgens could be observed given high individual variability [57]. Aside from clinical aspects, an ethical concern regarding fairness with respect to normo-androgenic female athletes also remains. This is particularly true as untreated diseases that induce hyperandrogenism may lend an unfair advantage in sports performance, and female athletes might refuse medical therapy to maintain a potential athletic advantage.

Testosterone has many dose-dependent pleiotropic effects that influence sports performance ([Table 4]), and the sexual dimorphism in physical performance between eugonadal adult 46XY males and 46XX females is related to both genetics and greater testosterone levels in males [58] [59]. Weak androgens such as androstenedione, DHEA, and DHEAS are also correlated with lean mass and physical performance in normo-androgenic females. Furthermore, a positive correlation between increased DHEA, lean mass and explosive performance has been reported in female Olympic athletes [60] [61] [62]. However, no studies have evaluated the influence of weak androgens on increasing sports performance under different hyperandrogenisms, particularly when the potential role of DHEAS is considered [63].

While few would doubt the evidence indicating that administering AAS to female athletes can improve their performance, there are only a few, and controversial, studies that have evaluated exercise performance in females after testosterone administration or in female athletes affected by hyperandrogenism. A 10-week testosterone treatment at 10 mg/day in young, physically active women found that increasing total serum testosterone from 0.9±0.4 to 4.3±2.8 nmol/L increased aerobic running time to exhaustion (+8%) and lean mass (+2%) [63]. The administration of testosterone (150/300 µg/day for 12 months) in androgen-deficient women with hypopituitarism to restore physiological female total and free testosterone serum levels significantly increased fat-free mass (+3.4%), cross-sectional muscle area (+6.6%), and behavioral parameters (arousal, behavior/experience, and cognition) [64]. In addition, testosterone administration at different doses (3–25 mg/week) for six months in postmenopausal women (reaching a maximal serum total testosterone level of 7.3 nmol/L) resulted in dose-dependent increases in muscle mass (+4.4%) and strength (+12–26%) [65].

The majority of studies concerning hyperandrogenism in female athletes have evaluated PCOS, which has a high incidence in the general population (4–12%) and even higher in elite athletes (15–31%) [66] [67] [68] [69]; nonetheless, few studies have evaluated CAH [66]. Interestingly, in the athletic community, the prevalence of hyper-androgenic 46XY DSD is seven per 1000, which is 140 times higher than in the general population [70]. Even if hyperandrogenism plays a role in the decision to participate in sports by influencing dominance and competitiveness, the high prevalence of DSD among elite athletes has also been attributed to factors that are associated with the Y chromosome (other than testosterone) [71].

In untreated athletes with PCOS and hyperandrogenism, increased muscle mass and strength, explosive strength, vertical jumping ability, lower limb power, muscle strength in response to resistance training, visuospatial ability, and VO2max have been reported [46] [47] [54] [72] [73] [74] [75]. For many scientists, female athletes with high serum testosterone concentrations have an estimated competitive advantage of at least 2–5% over normo-androgenic athletes, except for CAIS athletes. Moreover, decreased sports performance (by about 6% over two years) in female athletes treated for hyperandrogenism appears to support a previously testosterone-related advantage in performance [58] [59] [61] [76] [77]. A potential advantage in sports could also be related to the integrated effects of untreated hyperandrogenism plus female hypogonadism, and/or to possible TUE abuse and/or to the possible effects on the endocrine system of supplements and/or drug abuse; for example, we highlight that female athletes affected by CAH and authorized (i. e., TUE) for glucocorticoid treatment might increase the dose of glucocorticoids for doping purposes [55] [56] [78] [79] [80] [81] [82].

As sport endocrinologists, we cannot affirm at which exact supra-physiological testosterone concentration each female athlete might start to have specific advantages for all different psycho-physical capacities influencing all different sports performances. The pleiotropic effects of androgens, intra-individual AR distributions and sensitivity, the large intra- and inter-disease variability of weak androgens, and the fact that testosterone is not always assayed by liquid or gas chromatography-mass spectrometry all largely influence this type of evaluation. Moreover, we cannot affirm that differences in testosterone-related health risks and sports performance exist between female individuals with identical supra-physiological testosterone concentrations due to a disease or to exogenous testosterone abuse. Therefore, a generalized cut-off value for serum total testosterone concentrations that is higher than the upper normal female testosterone level (1.8 nmol/L) to grant sports eligibility in all types of female hyperandrogenism could be misleading, unfair and may facilitate short- and/or long-term health complications, particularly in female athletes that refuse therapy to maintain hyperandrogenism-related sports advantages. In theory, a case-by-case clinical assessment of health status, endocrine profile, and iatrogenic risks, and a balance between untreated disease-related risks and therapy-related risks would be necessary to provide advice regarding possible treatments, as in the general population, and evaluate sports eligibility.


#
#

Transgender women athletes

Actually, to avoid gender discrimination, male athletes affected by gender dysphoria can participate in sports as females once their medical (gender-affirming hormone therapy (GAHT) with exogenous 17β-estradiol or other estrogens±anti-androgens,±gender-affirming surgery) and bureaucratic transition process from male-to-female gender is completed, according to respective national laws on gender identity changes (please note in some countries GAHT and/or gender-affirming surgery are not mandatory). According to the International Olympic Committee (IOC) and other sports organization regulations, TW can participate in sports, independently of gender-affirming surgery for anatomical sexual reassignment, once their eligibility has been established by respective sports federations and, when necessary, after a TUE for prohibited drugs is obtained (i. e., for spironolactone). Given these stipulations, there should be no concerns regarding the participation of TW athletes in sports, respecting the previous IOC criteria for inclusion of TW athletes in female sports categories that required testosterone suppression below 10 nmol/L for 12 months prior to and during competition [83].

Unfortunately, worldwide debate continues regarding possible sports advantages of being a TW athlete as opposed to a cisgender female athlete, particularly in sports such as track and field, cycling, and weight-bearing sports. Due to the social and political stance against gender discrimination, the scientific evaluation of this question is very difficult. Particularly, the new IOC framework on fairness, inclusion, and non-discrimination based on gender identity and sex variations in sports has generated a very serious debate and discussion in sport and exercise medicine and we are waiting for possible new sport federations’ regulations for sport inclusion in these athletes [84] [85] [86]. In this sense, serious concerns will arise because in some countries it is possible to be a TW athlete without assuming GAHT, thus remaining, from a functional and endocrinological point of view, a male individual (in Switzerland, for example). Moreover, it will be very difficult to regulate sport participation because of the added complexity of gender non-binary people who were presumed male at birth, who may be prescribed GAHT, and who do not neatly fit into the binary categorization of most sports at the elite level.

It is not disputed that, apart from the psychological and social formation of gender and sexual orientation, an objective evaluation of the anatomical, endocrinological, and functional status of TW athletes is complex. From a mechanistic point of view, they are 46XY individuals who developed as males, often for many years, before becoming severely hypogonadal (i. e., low serum testosterone concentration and effects) ([Table 2]) due to continuous estrogen administration at female replacement doses that inhibit endogenous GnRH secretion. Furthermore, GAHT with estrogen therapy induces male hyperestrogenism, and anti-androgen drugs reduce all peripheral effects of androgens [87].

Aside from possible drug-related side effects due to the association between GAHT-related severe hypoandrogenism and relative hyperestrogenism in 46XY individuals, with respect to eugonadal cisgender athletes, TW athletes are at increased risk for cardiovascular diseases, endocrine-metabolic alterations, and potential lower abdominal muscle fibrosis and hypotrophy, which could lead to inguinal hernias, as observed in an animal model ([Table 5]). In addition, independently of possible side effects of GAHT on mood state, TW shows an increased rate of depression, anxiety, and suicides due to a multitude of reasons, including social and institutional discrimination, bullying, poor access to gender-affirming care, and so forth ([Table 5]) [88] [89] [90] [91] [92] [93] [94] [95].

In gender medicine, it is widely accepted that, due to genetic/cellular mechanisms (e. g., cell memory, enzymes, hormone receptor pathways) and endocrine-metabolic factors (e. g., sex steroid hormones concentrations, receptor distribution and sensitivity), a sex-chromosome-related dimorphism in exercise performance between 46XX and 46XY individuals exists [58] [96] [97]. In athletes, the 46XX/46XY dimorphism influences the psyche and behavior, stress and immune responses, disease prevalence and clinical expression, pain perception, and drug metabolism/effects. It also leads to gender-specific differences in health risks, exercise potential, and sports performance [39] [98] [99] [100]. In comparison with eugonadal adult 46XX individuals, eugonadal adult 46XY individuals have a taller stature, larger bone fulcrum, greater leverage for muscular-limb power, greater muscle area (+4–50%) and strength (+20–40%), greater aerobic capacity and anaerobic power, and better performance in running (+10–12%) and jumping [58] [96]. However, eugonadal 46XX individuals have greater flexibility, knee and hip flexion, increased shoulder extension and external rotation, reduced knee and hip extension, better balance, and more body fat [58] [96]. Many gender divergences in physical performance begin at puberty and reach a plateau in late adolescence, with the timing and tempo closely connected to the rise in serum testosterone concentrations in boys [101].

In terms of the anatomical and functional status in TW athletes, the main issue requiring resolution is whether the genetic expression and all the factors characterizing sports performance in eugonadal 46XY individuals (e. g., behavior, bio-energetics, body composition, body temperature, bone tissue, breathing, cardiovascular system, cognitive processes, CNS, mood, muscle physiology, skeleton, substrate metabolism, stress adaptation) could be significantly modified only by a suppressed testosterone concentration plus estrogen treatment to make them as similar as possible to cisgender female athletes [102] [103].

The male-to-female transition process is associated with a reduction in lean body mass (−2.4 kg), muscle mass (−9%), hand-grip strength (−9.5%), total thigh muscle volume (−5%), and quadriceps sectional area (−4%) [104] [105] [106] [107] [108]. Moreover, it has been found that after 12 months of estrogen treatment for transition, the total thigh volume and lower limb strength (both absolute and height-adjusted) in TWs are higher than in cisgender individuals and in female-to-male transgender (TM) individuals [108]. Gooren et al. has reported that testosterone deprivation in TWs decreased muscle mass, thereby increasing the overlap with untreated TM individuals, but mean muscle mass remained significantly higher in TWs than in TMs. They concluded that androgen deprivation in TWs increased the overlap in muscle mass with women but does not reverse it [109]. Moreover, other longitudinal studies examining the effects of testosterone suppression on muscle mass and strength in TW athletes show very modest changes after 12 months of treatment (approximately 5%), in terms of lean body mass and muscle size, suggesting that the muscle advantage enjoyed by TW athletes is only minimally reduced when testosterone is suppressed. In fact, given the large baseline differences in muscle mass between males and females (approximately 40%), the reduction achieved by 12 months of testosterone suppression can reasonably be assessed as small relative to the initial superior mass [83] [110] [111] [112] [113] [114] [115] [116] [117] [118].

It is likely that in TW athletes, and also depending on individual AR responsiveness [119], testosterone reduction cannot reset muscles to a female phenotype [102] [103] [108] [109] as testosterone suppression must not necessarily reach female serum testosterone concentrations (i. e., <1.8 nmol/L) in all TW athletes, both prior to and during competitions, and because muscle memory for testosterone still stimulates fibers hypertrophy in response to mechanical loads [120] [121]. Furthermore, estrogen administration at relatively high doses for the 46XY gender also has a strong influence on muscle structure and physiology, attenuating the devirilization of the body’s muscles [15] [25] [122] [123]. Many biological and functional differences between treated TW and eugonadal cisgender athletes exist ([Table 6]), as not all gene-related gender characteristics can be partially or completely reversed by GAHT. In addition, due to gender-related drug metabolism, possible abuse with prohibited substances may induce different effects in treated TW athletes with respect to cisgender athletes and male athletes.

Table 6 Genetic-, endocrine- and treatment-related differences influencing health status, exercise physiology, and sporting performance between adult transgender women athletes treated with gender-affirming hormone therapy (e. g., exogenous 17β-estradiol or other estrogens;±anti-androgens) and respective eugonadal cisgender adult athletes (in alphabetical order).

Transgender Women Athletes Treated with GAHT

Eugonadal Cisgender Athletes

Legal, Psychological and Social Gender

Female

Female

Sex chromosomes

46 XY

46 XX

Biomechanics of movements*

Male Phenotype*

Female Phenotype

Body fat %

Bone mineral density

E2 and P adaptive responses to exercise

Female sex hormone serum levels

Hypothalamus-pituitary-gonadal axis

Inhibited

Active

Menstrual cycle

Absent

Present

Muscle cell memory for male’s T levels*

Present*

Absent

Muscles distribution*

Male Phenotype*

Female Phenotype

Muscle mass and strength*

Higher vs. Female Phenotype*

Female Phenotype

Muscles response to exercise load*

Increased*

Physiological

Muscles-tendon anatomical structure*

Male Phenotype*

Female Phenotype

Peripheral responsiveness to androgens

Inhibited**/Male Phenotype

Female Phenotype

Pre-menstrual syndrome

Absent

Absent/Present

Skeletal anatomy*

Male Phenotype*

Female Phenotype

T and other androgen serum levels

±Female range

Female range

Cellular enzymes and metabolisms (e. g., heart, kidney, liver, muscles, neurons, and so forth)

Male Phenotype with Hypo-Testosteronemia*/**/***

Female Phenotype

Central nervous system physiology

Drug metabolism/effects (i. e., gender-related pharmacology/toxicology)

Hormone receptors (e. g., AR, ER, GR, PR)

Hormone responses to psycho-physical stress

AR, androgen receptors; E2, 17β-estradiol; EE, exogenous estrogens; ER, estrogen receptors; GAHT, gender-affirming hormonal treatment; GR, glucocorticoid receptors; P, progesterone; PR, progesterone receptors; T, testosterone. *These differences could be partially attenuated in the case of pre-puberty GnRH analogue therapy. ** Depending on anti-androgen assumption. *** Many cellular functions and metabolisms can be differently influenced by low serum T concentrations, as observed in male hypogonadism, and by a relatively high serum exogenous 17β-estradiol (or other estrogens) concentration.

We hypothesize that minimal differences between TW athletes and cisgender athletes related to behavior, mood, biomechanics of movement, functional and metabolic mechanisms, stress adaptation and fatigue perception ([Table 6]), are also associated with differences in sport performance. These could be small differences, yet significant enough to separate a champion from other finishing positions.

When possible differences and advantages in sports for adult TW athletes are evaluated with respect to cisgender athletes, we highlight that transgender girls may start a treatment with GnRH analogues before puberty to suppress male pubertal development, and then later commence GAHT (±anti-androgens); if they became athletes, these TWs would not be expected to have the same differences vs. cisgender athletes and possible advantages in sports that a TW athlete may have when GAHT was commenced post-completion of male puberty ([Table 6]).


#

Conclusions

From a healthcare perspective, sport physicians need awareness, knowledge, and understanding of sex steroid hormone variability and health concerns in the female athletic community ([Tables 1] [2] [5]). In this context, the main issue should be to protect general and reproductive health by a) reducing all factors causing sports-related hypogonadism; b) monitoring reproductive/sexual symptoms; c) favoring specialized counseling for diagnosing and treating sport-related hypogonadism, diseases causing hyperandrogenism/DSD, and iatrogenic complications in TW athletes according to gender-based medical criteria; and d) evaluating sports eligibility when health complications appear. In addition, support staff should undertake HPO-axis-cycle profiling in athletes to support possible menstrual cycle-related sports performance issues [32].

In our opinion, it could be quite difficult to implement these roles for sports physicians as several factors (e. g., athletes’ negligence, concomitant AAS abuse, ethical concerns, inefficient health care services, opportunistic behavior, personal choice, and socioeconomic aspects) can lead to the symptoms of hypogonadism and/or hyperandrogenism/DSD in female athletes not being declared. Moreover, due to possible health complications in TW athletes ([Table 5]), difficulties in the standardization of the criteria for granting sports eligibility may exist in countries where a pre-participation medical evaluation is mandatory.

Even with individual variability and depending on performed therapy ([Tables 2] [6]), hyper-androgenic females and TW athletes might have small but useful advantages during sports competition. However, it is necessary to gather proportionate, evidence- based, and reproducible data to draw definitive conclusions [86] [124]. Specific case-by-case and sport-by-sport investigations are recommended and all genetic-, endocrine-, and treatment-related factors potentially influencing exercise physiology and the final result in competitions should be addressed ([Tables 3] [4] [6]). Two serious obstacles should be taken into consideration: the final result in competitions is related to many difficult-to-measure factors, and it is challenging to detect the insignificant differences in performance (often less than 0.5–1.0%) that separate the winner from others.

Only when we have gathered sufficient observational data on the number of female athletes with untreated hyperandrogenism and TW athletes who win first place in sports competitions will we have an idea of the true scale of this phenomenon. Moreover, observational clinical investigations on the prevalence of health complications in such populations are warranted. Even if difficult, we hope to rapidly reach a clinical and scientific consensus for health protection and sports eligibility in the female athletic community to guarantee inclusion, fairness, and safe case-by-case sports participation between cisgender and transgender female athletes and also between well-treated and untreated hyperandrogenic and transgender female athletes while simultaneously avoiding gender-related discrimination and abuse.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

  • References

  • 1 Borgen NT. Collider bias (aka sample selection bias) in observational studies: why the effects of hyperandrogenism in elite women's sport are likely underestimated. Br J Sports Med 2020; 54: 750-752
  • 2 Camporesi S.. A question of ’fairness’: why ethics should factor in the court of arbitration for sport’s decision on the IAAF Hyperandrogenism Regulations. Br J Sports Med 2019; 53: 797-798
  • 3 Franklin S, Ospina Betancurt J, Camporesi S.. What statistical data of observational performance can tell us and what they cannot: the case of Dutee Chand v. AFI & IAAF. Br J Sports Med 2018; 52: 420-421
  • 4 Harper J, Lima G, Kolliari-Turner A. et al. The fluidity of gender and implications for the biology of inclusion for transgender and intersex athletes. Curr Sports Med Rep 2018; 17: 467-472
  • 5 Harper J, Martinez-Patino M-J, Pigozzi F. et al. Implications of a third gender for elite sports. Curr Sports Med Rep 2018; 7: 42-44
  • 6 Hirschberg AL.. Hyperandrogenism in female athletes. J Clin Endocrinol Metab 2019; 104: 503-505
  • 7 Sönksen PH, Bavington LD, Boehning T. et al. Hyperandrogenism controversy in elite women’s sport: an examination and critique of recent evidence. Br J Sports Med 2018; 52: 1481-1482
  • 8 Sonksen P, Ferguson-Smith MA, Bavington LD. et al. Medical and ethical concerns regarding women with hyperandrogenism and elite sport. J Clin Endocrinol Metab 2015; 100: 825-827
  • 9 Sönksen PH, Holt RIG, Böhning W. et al. Why do endocrine profiles in elite athletes differ between sports?. Clin Diabetes Endocrinol 2018; 4: 3
  • 10 Tannenbaum C, Bekker S.. Sex, gender, and sports. BMJ 2019; 364: I1120
  • 11 Holesh JE, Bass AN, Lord M.. Physiology, Ovulation. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022. Jan. 2022 May 8
  • 12 Cano Sokoloff N, Misra M, Ackerman KE.. Exercise, training, and the hypothalamic-pituitary-gonadal axis in men and women. Front Horm Res 2016; 47: 27-43
  • 13 Haines M, McKinley-Barnard SK, Andre TL. et al. Skeletal muscle estrogen receptor activation in response to eccentric exercise up-regulates myogenic-related gene expression independent of differing serum estradiol levels occurring during the human menstrual cycle. J Sports Sci Med 2018; 17: 31-39
  • 14 Hirschberg AL.. Female hyperandrogenism and elite sport. Endocr Connect 2020; 9: R81-R92
  • 15 Chidi-Ogbolu N, Baar K.. Effect of estrogen on musculoskeletal performance and injury risk. Front Physiol 2019; 9: 1834
  • 16 Guillaume M, Montagner A, Fontaine C. et al. Nuclear and membrane actions of estrogen receptor alpha: contribution to the regulation of energy and glucose homeostasis. Adv Exp Med Biol 2017; 1043: 401-426
  • 17 Oosthuyse T, Bosch AN.. The effect of the menstrual cycle on exercise metabolism: implications for exercise performance in eumenorrhoeic women. Sports Med 2010; 40: 207-227
  • 18 Rettberg JR, Yao J, Brinton RD.. Estrogen: a master regulator of bioenergetic systems in the brain and body. Front Neuroendocrinol 2014; 35: 8-30
  • 19 Smith JR, Koepp KE, Berg JD. et al. Influence of sex, menstrual cycle, and menopause status on the exercise pressor reflex. Med Sci Sports Exerc 2019; 51: 874-881
  • 20 Melanson EL, Lyden K, Gibbons E. et al. Influence of estradiol status on physical activity in premenopausal women. Med Sci Sports Exerc 2018; 50: 1704-1709
  • 21 Minahan C, Joyce S, Bulmer AC. et al. The influence of estradiol on muscle damage and leg strength after intense eccentric exercise. J Appl Physiol (1985) 2015; 115: 1493-1500
  • 22 Hansen M.. Female hormones: do they influence muscle and tendon protein metabolism?. Proc Nutr Soc 2018; 77: 32-41
  • 23 Klinge CM.. Estrogenic control of mitochondrial function and biogenesis. J Cell Biochem 2008; 105: 1342-1351
  • 24 Kraemer RR, Francois M, Castracane VD.. Estrogen mediation of hormone responses to exercise. Metabolism 2012; 61: 1337-1346
  • 25 Spangenburg EE, Geiger PC, Leinwand LA. et al. Regulation of physiological and metabolic function of muscle by female sex steroids. Med Sci Sports Exerc 2012; 44: 1653-1662
  • 26 da Silva SB, de Sousa Ramalho Viana E. et al. Changes in peak expiratory flow and respiratory strength during the menstrual cycle. Respir Physiol Neurobiol 2006; 150: 211-219
  • 27 Stachenfeld NS.. Sex hormone effects on body fluid regulation. Exerc Sport Sci Rev 2008; 36: 152-159
  • 28 Sgrò P, Di Luigi L.. Sport and male sexuality. J Endocrinol Invest 2017; 40: 911-923
  • 29 Julian R, Hecksteden A, Fullagar HH. et al. The effects of menstrual cycle phase on physical performance in female soccer players. PLoS One 2017; 12: e0173951
  • 30 Cristina-Souza G, Santos-Mariano AC, Souza-Rodrigues CC. et al. Menstrual cycle alters training strain, monotony, and technical training length in young. J Sports Sci 2019; 37: 1824-1830
  • 31 Janse DE, Jonge X, Thompson B, Han A.. Methodological recommendations for menstrual cycle research in sports and exercise. Med Sci Sports Exerc 2019; 51: 2610-2617
  • 32 Findlay RJ, Macrae EHR, Whyte IY. et al. How the menstrual cycle and menstruation affect sporting performance: experiences and perceptions of elite female rugby players. Br J Sports Med 2020; 54: 1108-1113
  • 33 Foster R, Vaisberg M, Bachi ALL. et al. Premenstrual syndrome, inflammatory status, and mood states in soccer players. Neuroimmunomodulation 2019; 26: 1-6
  • 34 Hakimi O, Cameron LC.. Effect of exercise on ovulation: a systematic review. Sports Med 2017; 47: 1555-1567
  • 35 Ackerman KE, Holtzman B, Cooper KM. et al. Low energy availability surrogates correlate with health and performance consequences of relative energy deficiency in sport. Br J Sports Med 2019; 53: 628-633
  • 36 De Souza MJ, Koltun KJ, Williams NI.. The role of energy availability in reproductive function in the female athlete triad and extension of its effects to men: an initial working model of a similar syndrome in male athletes. Sports Med 2019; 49: 125-137
  • 37 Dipla K, Kraemer RR, Constantini NW. et al. Relative energy deficiency in sports (RED-S): elucidation of endocrine changes affecting the health of males and females. Hormones (Athens) 2021; 20: 35-47
  • 38 Mountjoy M, Sundgot-Borgen JK, Burke LM. et al. IOC consensus statement on relative energy deficiency in sport (RED-S): 2018 update. Br J Sports Med 2018; 52: 687-697
  • 39 Toufexis D, Rivarola MA, Lara H. et al. Stress and the reproductive axis. J Neuroendocrinol 2014; 26: 573-586
  • 40 Vorona E, Nieschlag E.. Adverse effects of doping with anabolic androgenic steroids in competitive athletics, recreational sports and bodybuilding. Minerva Endocrinol 2018; 43: 476-488
  • 41 Tornberg ÅB, Melin A, Koivula FM. et al. Reduced neuromuscular performance in amenorrheic elite endurance athletes. Med Sci Sports Exerc 2017; 49: 2478-2485
  • 42 Witchel SF.. Congenital adrenal hyperplasia. Pediatr Adolesc Gynecol 2017; 30: 520-534
  • 43 Moran C, Azziz R, Weintrob N. et al. Reproductive outcome of women with 21-hydroxylase-deficient nonclassic adrenal hyperplasia. J Clin Endocrinol Metab 2006; 91: 3451-3456
  • 44 Bidet M, Bellanne-Chantelot C, Galand-Portier MB. et al. Fertility in women with nonclassical congenital adrenal hyperplasia due to 21-hydroxylase deficiency. J Clin Endocrinol Metab 2010; 95: 1182-1190
  • 45 Aversa A, La Vignera S, Rago R. et al. Fundamental concepts and novel aspects of polycystic ovarian syndrome: expert consensus resolutions. Fornt Endocrinol (Lausanne) 2020; 11: 516
  • 46 Kogure GS, Silva RC, Miranda-Furtado CL. et al. Hyperandrogenism enhances muscle strength after progressive resistance training, independent of body composition, in women with polycystic ovary syndrome. J Strength Cond Res 2018; 32: 2642-2651
  • 47 Dos Santos IK, de Lima Nunes R, Soares GM. et al. Exercise and reproductive function in polycystic ovary syndrome: protocol of a systematic review. Syst Rev 2017; 6: 264
  • 48 Kogure GS, Silva RC, Picchi Ramos FK. et al. Women with polycystic ovary syndrome have greater muscle strength irrespective of body composition. Gynecol Endocrinol 2015; 31: 237-242
  • 49 Speiser PW, Dupont B, Rubinstein P. et al. High frequency of nonclassical steroid 21-hydroxylase deficiency. Am J Hum Genet 1985; 37: 650-667
  • 50 Moran C, Azziz R, Carmina E. et al. 21-hydroxylase-deficient nonclassic adrenal hyperplasia is a progressive disorder: a multicenter study. Am J Obstet Gynecol 2000; 183: 1468-1474
  • 51 Pignatelli D.. Non-classic adrenal hyperplasia due to the deficiency of 21-hydroxylase and its relation to polycystic ovarian syndrome. Front Horm Res 2013; 40: 158-170
  • 52 Oncul M, Sahmay S, Tuten A. et al. May AMH levels distinguish LOCAH from PCOS among hirsute women?. Eur J Obstet Gynecol Reprod Biol 2014; 178: 183-187
  • 53 Martin KA, Anderson RR, Chang RJ. et al. Evaluation and treatment of hirsutism in premenopausal women: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2018; 103: 1233-1257
  • 54 Escobar-Morreale HF, Carmina E, Dewailly D. et al. Epidemiology, diagnosis and management of hirsutism: a consensus statement by the Androgen Excess and Polycystic Ovary Syndrome Society. Hum Reprod Update 2012; 18: 146-170
  • 55 Di Luigi L, Pigozzi F, Sgrò P. et al. The use of prohibited substances for therapeutic reasons in athletes affected by endocrine diseases and disorders: the therapeutic use exemption (TUE) in clinical endocrinology. J Endocrinol Invest 2020; 43: 563-573
  • 56 World Anti-doping Agency (WADA). Therapeutic Use Exemption (TUE Available from https://www.wada-ama.org/en/what-we-do/science-medical/therapeutic-use-exemptions Accessed: May 2022
  • 57 Clark RV, Wald JA, Swerdloff RS. et al. Large divergence in testosterone concentrations between men and women: frame of reference for elite athletes in sex-specific competition in sports, a narrative review. Clin Endocrinol (Oxf) 2019; 90: 15-22
  • 58 Handelsman DJ, Hirschberg AL, Bermon S.. Circulating testosterone as the hormonal basis of sex differences in athletic performance. Endocr Rev 2018; 39: 803-829
  • 59 Ristori J, Cocchetti C, Romani A. et al. Brain sex differences related to gender identity development: genes or hormones?. Int J Mol Sci 2020; 21: 2123
  • 60 Eklund E, Berglund B, Labrie F. et al. Serum androgen profile and physical performance in women Olympic athletes. Br J Sports Med 2017; 51: 1301-1308
  • 61 Hirschberg AL, Elings Knutsson J, Helge T. et al. Effects of moderately increased testosterone concentration on physical performance in young women: a double blind, randomised, placebo-controlled study. Br J Sports Med 2020; 54: 599-604
  • 62 Labrie F, Martel C, Bélanger A. et al. Androgens in women are essentially made from DHEA in each peripheral tissue according to intracrinology. J Steroid Biochem Mol Biol 2017; 168: 9-18
  • 63 Huang G, Basaria S.. Do anabolic-androgenic steroids have performance-enhancing effects in female athletes?. Mol Cell Endocrinol 2018; 15: 56-64
  • 64 Miller KK, Biller BM, Beauregard C. et al. Effects of testosterone replacement in androgen-deficient women with hypopituitarism: a randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab 2006; 91: 1683-1690
  • 65 Huang G, Basaria S, Travison TG. et al. Testosterone dose-response relationships in hysterectomized women with or without oophorectomy: effects on sexual function, body composition, muscle performance and physical function in a randomized trial. Menopause 2014; 21: 612-623
  • 66 Eliakim A, Nemet D.. Endogenous hyperandrogenism and exercise capacity lessons from the exercise-congenital adrenal hyperplasia model. J Pediatr Endocrinol Metab 2010; 23: 1213-1219
  • 67 Hagmar M, Berglund B, Brismar K. et al. Hyperandrogenism may explain reproductive dysfunction in olympic athletes. Med Sci Sports Exerc 2009; 41: 1241-1248
  • 68 Kogure GS, Miranda-Furtado CL, Silva RC. et al. Resistance exercise impacts lean muscle mass in women with polycystic ovary syndrome. Med Sci Sports Exerc 2016; 48: 589-598
  • 69 Costa EC, Sá JCF DE, Stepto NK. et al. Aerobic training improves quality of life in women with polycystic ovary syndrome. Med Sci Sports Exerc 2018; 50: 1357-1366
  • 70 Knochenhauer ES, Key TJ, Kahsar-Miller M. et al. Prevalence of the polycystic ovary syndrome in unselected black and white women of the southeastern United States: a prospective study. J Clin Endocrinol Metab 1998; 83: 3078-3082
  • 71 Ferguson-Smith MA, Bavington LD.. Natural selection for genetic variants in sport: the role of Y chromosome genes in elite female athletes with 46,XY DSD1. Sports Med 2014; 44: 1629-1634
  • 72 Bermon S, Garnier PY, Hirschberg AL. et al. Serum androgen levels in elite female athletes. J Clin Endocrinol Metab 2014; 99: 4328-4335
  • 73 Caliskan Guzelce E, Eyupoglu D, Torgutalp S. et al. Is muscle mechanical function altered in polycystic ovary syndrome?. Arch Gynecol Obstet 2019; 300: 771-776
  • 74 Cardinale M, Stone MH.. Is testosterone influencing explosive performance?. J Strength Cond Res 2006; 20: 103-107
  • 75 Douchi T, Yamamoto S, Oki T. et al. Serum androgen levels and muscle mass in women with polycystic ovary syndrome. Obstet Gynecol 1999; 94: 337-340
  • 76 Rickenlund A, Carlström K, Ekblom B. et al. Hyperandrogenicity is an alternative mechanism underlying oligomenorrhea or amenorrhea in female athletes and may improve physical performance. Fertil Steril 2003; 79: 947-955
  • 77 Bermon S.. Androgens and athletic performance of elite female athletes. Curr Opin Endocrinol Diabetes Obes 2017; 24: 246-251
  • 78 Conte D, Romanelli F, Fillo S. et al. Aspirin inhibits androgen response to chorionic gonadotropin in humans. Am J Physiol 1999; 277: e1032
  • 79 Di Luigi L, Rossi C, Sgrò P. et al. Do non-steroidal anti-inflammatory drugs influence the steroid hormone milieu in male athletes?. Int J Sports Med 2007; 28: 809-814
  • 80 Di Luigi L, Guidetti L, Romanelli F. et al. Acetylsalicylic acid inhibits the pituitary response to exercise-related stress in humans. Med Sci Sports Exerc 2001; 33: 2029-2035
  • 81 Di Luigi L, Guidetti L, Pigozzi F. et al. Acute amino acids supplementation enhances pituitary responsiveness in athletes. Med Sci Sports Exerc 1999; 31: 1748-1754
  • 82 Di Luigi L.. Supplements and the endocrine system in athletes. Clin Sports Med 2008; 27: 131-51
  • 83 Hilton EN, Lundberg TR.. Transgender women in the female category of sport: perspectives on testosterone suppression and performance advantage. Sports Med 2021; 51: 199-214
  • 84 Jones BA, Arcelus J, Bouman WP. et al. Sport and transgender people: a systematic review of the literature relating to sport participation and competitive sport policies. Sports Med 2017; 47: 701-716
  • 85 International Olympic Committee. Framework on Fairness, Inclusion and Non-discrimination on the Basis of Gender Identity and Sex Variations, 2021 Available from https://stillmed.olympics.com/media/Documents/News/2021/11/IOC-Framework Fairness-Inclusion-Nondiscrimination-2021.pdf Accessed: May 2022
  • 86 Pigozzi F, Bigard X, Steinacker J. , on behalf of the International Federation of Sports Medicine (FIMS) and the European Federation of Sports Medicine Associations (EFSMA) et al. Joint position statement of the International Federation of Sports Medicine (FIMS) and European Federation of Sports Medicine Associations (EFSMA) on the IOC framework on fairness, inclusion and non-discrimination based on gender identity and sex variations. BMJ Open Sport Exerc Med 2022; 8: e001273
  • 87 Iuliano S, Izzo G, Zagari MC. et al. Endocrine management of transgender adults: a clinical approach. Sexes 2021; 2: 104-118
  • 88 Bermon S, Garnier PY.. Serum androgen levels and their relation to performance in track and field: mass spectrometry results from 2127 observations in male and female elite athletes. Br J Sports Med 2017; 51: 1309-1314
  • 89 Dubon ME, Abbott K, Carl RL.. Care of the transgender athlete. Curr Sports Med Rep 2018; 17: 410-418
  • 90 Gooren LJ, Giltay EJ, Bunck MC.. Long-term treatment of transsexuals with cross-sex hormones: extensive personal experience. J Clin Endocrinol Metab 2008; 93: 19-25
  • 91 van Dijk D, Dekker MJHJ, Conemans EB. et al. Explorative prospective evaluation of short-term subjective effects of hormonal treatment in trans people–results from the European network for the investigation of gender incongruence. J Sex Med 2019; 16: 1297-1309
  • 92 Zhao H, Zhou L, Li L. et al. Shift from androgen to estrogen action causes abdominal muscle fibrosis, atrophy, and inguinal hernia in a transgenic male mouse model. Proc Natl Acad Sci USA 2018; 115: e10427-e10436
  • 93 Catelan RF, Saadeh A, Lobato MIR. et al. Depression, self-esteem, and resilience and its relationship with psychological features of sexuality among transgender men and women from Brazil. Arch Sex Behav 2022; 51: 1993-2002
  • 94 Sarno EL, Dyar C, Newcomb ME. et al. Relationship quality and mental health among sexual and gender minorities. J Fam Psychol 2022; 36: 770-779
  • 95 Romani A, Mazzoli F, Ristori J. et al. Psychological wellbeing and perceived social acceptance in gender diverse individuals. J Sex Med 2021; 18: 1933-1944
  • 96 Allison KF, Keenan KA, Sell TC. et al. Musculoskeletal, biomechanical, and physiological gender differences in the US military. US Army Med Dep J 2015; 22-32
  • 97 Ngun TC, Ghahramani N, Sánchez FJ. et al. The genetics of sex differences in brain and behavior. Front Neuroendocrinol 2011; 32: 227-246
  • 98 Zheng D, Wang X, Antonson P. et al. Genomics of sex hormone receptor signaling in hepatic sexual dimorphism. Mol Cell Endocrinol 2018; 15: 33-41
  • 99 Boldt P, Knechtle B, Nikolaidis P. et al. Sex differences in the health status of endurance runners: results from the NURMI study (step 2). J Strength Cond Res 2019; 33: 1929-1940
  • 100 Lepers R, Knechtle B, Stapley PJ.. Trends in triathlon performance: effects of sex and age. Sports Med 2013; 43: 851-863
  • 101 Handelsman DJ.. Sex differences in athletic performance emerge coinciding with the onset of male puberty. Clin Endocrinol (Oxf) 2017; 87: 68-72
  • 102 Oyola MG, Handa RJ.. Hypothalamic-pituitary-adrenal and hypothalamic-pituitary-gonadal axes: sex differences in regulation of stress responsivity. Stress 2017; 20: 476-494
  • 103 Roberts TA, Smalley J, Ahrendt D.. Effect of gender affirming hormones on athletic performance in transwomen and transmen: implications for sporting organisations and legislators. Br J Sports Med . 2020
  • 104 Elbers JM, de Jong S, Teerlink T. et al. Changes in fat cell size and in vitro lipolytic activity of abdominal and gluteal adipocytes after a one-year cross-sex hormone administration in transsexuals. Metabolism 1999; 48: 1371-1377
  • 105 Klaver M, de Blok CJM, Wiepjes CM. et al. Changes in regional body fat, lean body mass and body shape in trans persons using cross-sex hormonal therapy: results from a multicenter perspective study. Eur J Endocrinol 2018; 178: 163-171
  • 106 Klaver M, Dekker MJHJ, de Mutsert R. et al. Cross-sex hormone therapy in transgender persons affects total body weight, body fat and lean body mass: a meta-analysis. Andrologia 2017; 49: e12660
  • 107 Van Caenegem E, Wierckx K, Taes Y. et al. Preservation of volumetric bone density and geometry in trans women during cross-sex hormonal therapy: a prospective observational study. Osteoporos Int 2015; 26: 35-47
  • 108 Wiik A, Lundberg TR, Rullman E. et al. Muscle strength, size, and composition following 12 months of gender-affirming treatment in transgender individuals. J Clin Endocrinol Metab 2020; 105: dgz247
  • 109 Gooren LJ, Bunck MC.. Transsexuals and competitive sports. Eur J Endocrinol 2004; 151: 425-429
  • 110 Haraldsen IR, Haigh E, Falch J. et al. Cross-sex pattern of bone mineral density in early onset gender identity dsorder. Horm Behav 2007; 52: 334-343
  • 111 Mueller A, Zollver H, Kronawitter D. et al. Body composition and bone mineral density in male-to-female transexuals during cross-sex hormone therapy using gonadotrophin-releasing hormone agonist. Exp Clin Endocrinol Diabetes 2011; 119: 95-100
  • 112 Wierckx K, Van Caenegem E, Schreiner T. et al. Cross-sex hormone therapy in trans persons is safe and effective at short-time follow-up: results from the European network for the investigation of gender incongruence. J Sex Med 2014; 11: 1999-2011
  • 113 Gava G, Cerpolini S, Martelli V. et al. Cyproterone acetate vs leuprolide acetate in combination with transdermal oestradiol in transwomen: a comparison of safety and effectiveness. Clin Endocrinol (Oxf) 2016; 85: 239-246
  • 114 Auer MK, Ebert T, Pietzner M. et al. Effects of sex hormone treatment on the metabolic syndrome in transgender individuals: focus on metabolic cytokines. J Clin Endocrinol Metab 2018; 103: 790-802
  • 115 Fighera TM, da Silva E, Lindenau JDR. et al. Impact of cross-sex hormone therapy on bone mineral density and body composition in transwomen. Clin Endocrinol (Oxf) 2018; 88: 856-862
  • 116 Scharff M, Wiepjes CM, Klaver M. et al. Change in grip strength in trans people and its association with lean body mass and bone density. Endocr Connect 2019; 8: 1020-1028
  • 117 Tack LJW, Craen M, Lapauw B. et al. Proandrogenic and antiandrogenic progestins in transgender youth: differential effects on body composition and bone metabolism. J Clin Endocrinol Metab 2018; 103: 2147-2156
  • 118 Polderman KH, Gooren LJG, Asscheman H. et al. Induction of insulin resistance by androgens and estrogens. J Clin Endocrinol Metab 1994; 79: 265-271
  • 119 D'Andrea S, Pallotti F, Senofonte G. et al. Polymorphic cytosine-adenine-guanine repeat length of androgen receptor gene and gender incongruence in trans women: a systematic review and meta-analysis of case-control studies. J Sex Med 2020; 17: 543-550
  • 120 Egner IM, Bruusgaard JC, Eftestøl E. et al. A cellular memory mechanism aids overload hypertrophy in muscle long after an episodic exposure to anabolic steroids. J Physiol 2013; 591: 6221-6230
  • 121 Gundersen K.. Muscle memory and a new cellular model for muscle atrophy and hypertrophy. J Exp Biol 2016; 219: 235-242
  • 122 Nicoll JX, Fry AC, Mosier EM.. Sex-based differences in resting MAPK, androgen, and glucocorticoid receptor phosphorylation in human skeletal muscle. Steroids 2019; 141: 23-29
  • 123 Scott NL, Abreu MR, Cates BE. et al. Prolonged effects of elevated 17β-estradiol on physical activity after orchidectomy. Med Sci Sports Exerc 2018; 50: 1588-1595
  • 124 Hamilton BR, Martinez-Patiño MJ, Barrett J.. et al. Response to the United Nations Human Rights Council’s report on race and gender discrimination in sport: an expression of concern and a call to prioritise research. Sports Med 2021; 51: 839-842

Correspondence

Prof. Luigi Di Luigi
Università degli Studi di Roma "Foro Italico", Scienze Motorie, Umane e della Salute, piazza Lauro de Bosis 6
00135 Rome
Italy   
Phone: +390636733563   
Fax: +390636733231   

Publication History

Received: 01 February 2022

Accepted: 08 July 2022

Article published online:
29 September 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Borgen NT. Collider bias (aka sample selection bias) in observational studies: why the effects of hyperandrogenism in elite women's sport are likely underestimated. Br J Sports Med 2020; 54: 750-752
  • 2 Camporesi S.. A question of ’fairness’: why ethics should factor in the court of arbitration for sport’s decision on the IAAF Hyperandrogenism Regulations. Br J Sports Med 2019; 53: 797-798
  • 3 Franklin S, Ospina Betancurt J, Camporesi S.. What statistical data of observational performance can tell us and what they cannot: the case of Dutee Chand v. AFI & IAAF. Br J Sports Med 2018; 52: 420-421
  • 4 Harper J, Lima G, Kolliari-Turner A. et al. The fluidity of gender and implications for the biology of inclusion for transgender and intersex athletes. Curr Sports Med Rep 2018; 17: 467-472
  • 5 Harper J, Martinez-Patino M-J, Pigozzi F. et al. Implications of a third gender for elite sports. Curr Sports Med Rep 2018; 7: 42-44
  • 6 Hirschberg AL.. Hyperandrogenism in female athletes. J Clin Endocrinol Metab 2019; 104: 503-505
  • 7 Sönksen PH, Bavington LD, Boehning T. et al. Hyperandrogenism controversy in elite women’s sport: an examination and critique of recent evidence. Br J Sports Med 2018; 52: 1481-1482
  • 8 Sonksen P, Ferguson-Smith MA, Bavington LD. et al. Medical and ethical concerns regarding women with hyperandrogenism and elite sport. J Clin Endocrinol Metab 2015; 100: 825-827
  • 9 Sönksen PH, Holt RIG, Böhning W. et al. Why do endocrine profiles in elite athletes differ between sports?. Clin Diabetes Endocrinol 2018; 4: 3
  • 10 Tannenbaum C, Bekker S.. Sex, gender, and sports. BMJ 2019; 364: I1120
  • 11 Holesh JE, Bass AN, Lord M.. Physiology, Ovulation. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022. Jan. 2022 May 8
  • 12 Cano Sokoloff N, Misra M, Ackerman KE.. Exercise, training, and the hypothalamic-pituitary-gonadal axis in men and women. Front Horm Res 2016; 47: 27-43
  • 13 Haines M, McKinley-Barnard SK, Andre TL. et al. Skeletal muscle estrogen receptor activation in response to eccentric exercise up-regulates myogenic-related gene expression independent of differing serum estradiol levels occurring during the human menstrual cycle. J Sports Sci Med 2018; 17: 31-39
  • 14 Hirschberg AL.. Female hyperandrogenism and elite sport. Endocr Connect 2020; 9: R81-R92
  • 15 Chidi-Ogbolu N, Baar K.. Effect of estrogen on musculoskeletal performance and injury risk. Front Physiol 2019; 9: 1834
  • 16 Guillaume M, Montagner A, Fontaine C. et al. Nuclear and membrane actions of estrogen receptor alpha: contribution to the regulation of energy and glucose homeostasis. Adv Exp Med Biol 2017; 1043: 401-426
  • 17 Oosthuyse T, Bosch AN.. The effect of the menstrual cycle on exercise metabolism: implications for exercise performance in eumenorrhoeic women. Sports Med 2010; 40: 207-227
  • 18 Rettberg JR, Yao J, Brinton RD.. Estrogen: a master regulator of bioenergetic systems in the brain and body. Front Neuroendocrinol 2014; 35: 8-30
  • 19 Smith JR, Koepp KE, Berg JD. et al. Influence of sex, menstrual cycle, and menopause status on the exercise pressor reflex. Med Sci Sports Exerc 2019; 51: 874-881
  • 20 Melanson EL, Lyden K, Gibbons E. et al. Influence of estradiol status on physical activity in premenopausal women. Med Sci Sports Exerc 2018; 50: 1704-1709
  • 21 Minahan C, Joyce S, Bulmer AC. et al. The influence of estradiol on muscle damage and leg strength after intense eccentric exercise. J Appl Physiol (1985) 2015; 115: 1493-1500
  • 22 Hansen M.. Female hormones: do they influence muscle and tendon protein metabolism?. Proc Nutr Soc 2018; 77: 32-41
  • 23 Klinge CM.. Estrogenic control of mitochondrial function and biogenesis. J Cell Biochem 2008; 105: 1342-1351
  • 24 Kraemer RR, Francois M, Castracane VD.. Estrogen mediation of hormone responses to exercise. Metabolism 2012; 61: 1337-1346
  • 25 Spangenburg EE, Geiger PC, Leinwand LA. et al. Regulation of physiological and metabolic function of muscle by female sex steroids. Med Sci Sports Exerc 2012; 44: 1653-1662
  • 26 da Silva SB, de Sousa Ramalho Viana E. et al. Changes in peak expiratory flow and respiratory strength during the menstrual cycle. Respir Physiol Neurobiol 2006; 150: 211-219
  • 27 Stachenfeld NS.. Sex hormone effects on body fluid regulation. Exerc Sport Sci Rev 2008; 36: 152-159
  • 28 Sgrò P, Di Luigi L.. Sport and male sexuality. J Endocrinol Invest 2017; 40: 911-923
  • 29 Julian R, Hecksteden A, Fullagar HH. et al. The effects of menstrual cycle phase on physical performance in female soccer players. PLoS One 2017; 12: e0173951
  • 30 Cristina-Souza G, Santos-Mariano AC, Souza-Rodrigues CC. et al. Menstrual cycle alters training strain, monotony, and technical training length in young. J Sports Sci 2019; 37: 1824-1830
  • 31 Janse DE, Jonge X, Thompson B, Han A.. Methodological recommendations for menstrual cycle research in sports and exercise. Med Sci Sports Exerc 2019; 51: 2610-2617
  • 32 Findlay RJ, Macrae EHR, Whyte IY. et al. How the menstrual cycle and menstruation affect sporting performance: experiences and perceptions of elite female rugby players. Br J Sports Med 2020; 54: 1108-1113
  • 33 Foster R, Vaisberg M, Bachi ALL. et al. Premenstrual syndrome, inflammatory status, and mood states in soccer players. Neuroimmunomodulation 2019; 26: 1-6
  • 34 Hakimi O, Cameron LC.. Effect of exercise on ovulation: a systematic review. Sports Med 2017; 47: 1555-1567
  • 35 Ackerman KE, Holtzman B, Cooper KM. et al. Low energy availability surrogates correlate with health and performance consequences of relative energy deficiency in sport. Br J Sports Med 2019; 53: 628-633
  • 36 De Souza MJ, Koltun KJ, Williams NI.. The role of energy availability in reproductive function in the female athlete triad and extension of its effects to men: an initial working model of a similar syndrome in male athletes. Sports Med 2019; 49: 125-137
  • 37 Dipla K, Kraemer RR, Constantini NW. et al. Relative energy deficiency in sports (RED-S): elucidation of endocrine changes affecting the health of males and females. Hormones (Athens) 2021; 20: 35-47
  • 38 Mountjoy M, Sundgot-Borgen JK, Burke LM. et al. IOC consensus statement on relative energy deficiency in sport (RED-S): 2018 update. Br J Sports Med 2018; 52: 687-697
  • 39 Toufexis D, Rivarola MA, Lara H. et al. Stress and the reproductive axis. J Neuroendocrinol 2014; 26: 573-586
  • 40 Vorona E, Nieschlag E.. Adverse effects of doping with anabolic androgenic steroids in competitive athletics, recreational sports and bodybuilding. Minerva Endocrinol 2018; 43: 476-488
  • 41 Tornberg ÅB, Melin A, Koivula FM. et al. Reduced neuromuscular performance in amenorrheic elite endurance athletes. Med Sci Sports Exerc 2017; 49: 2478-2485
  • 42 Witchel SF.. Congenital adrenal hyperplasia. Pediatr Adolesc Gynecol 2017; 30: 520-534
  • 43 Moran C, Azziz R, Weintrob N. et al. Reproductive outcome of women with 21-hydroxylase-deficient nonclassic adrenal hyperplasia. J Clin Endocrinol Metab 2006; 91: 3451-3456
  • 44 Bidet M, Bellanne-Chantelot C, Galand-Portier MB. et al. Fertility in women with nonclassical congenital adrenal hyperplasia due to 21-hydroxylase deficiency. J Clin Endocrinol Metab 2010; 95: 1182-1190
  • 45 Aversa A, La Vignera S, Rago R. et al. Fundamental concepts and novel aspects of polycystic ovarian syndrome: expert consensus resolutions. Fornt Endocrinol (Lausanne) 2020; 11: 516
  • 46 Kogure GS, Silva RC, Miranda-Furtado CL. et al. Hyperandrogenism enhances muscle strength after progressive resistance training, independent of body composition, in women with polycystic ovary syndrome. J Strength Cond Res 2018; 32: 2642-2651
  • 47 Dos Santos IK, de Lima Nunes R, Soares GM. et al. Exercise and reproductive function in polycystic ovary syndrome: protocol of a systematic review. Syst Rev 2017; 6: 264
  • 48 Kogure GS, Silva RC, Picchi Ramos FK. et al. Women with polycystic ovary syndrome have greater muscle strength irrespective of body composition. Gynecol Endocrinol 2015; 31: 237-242
  • 49 Speiser PW, Dupont B, Rubinstein P. et al. High frequency of nonclassical steroid 21-hydroxylase deficiency. Am J Hum Genet 1985; 37: 650-667
  • 50 Moran C, Azziz R, Carmina E. et al. 21-hydroxylase-deficient nonclassic adrenal hyperplasia is a progressive disorder: a multicenter study. Am J Obstet Gynecol 2000; 183: 1468-1474
  • 51 Pignatelli D.. Non-classic adrenal hyperplasia due to the deficiency of 21-hydroxylase and its relation to polycystic ovarian syndrome. Front Horm Res 2013; 40: 158-170
  • 52 Oncul M, Sahmay S, Tuten A. et al. May AMH levels distinguish LOCAH from PCOS among hirsute women?. Eur J Obstet Gynecol Reprod Biol 2014; 178: 183-187
  • 53 Martin KA, Anderson RR, Chang RJ. et al. Evaluation and treatment of hirsutism in premenopausal women: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2018; 103: 1233-1257
  • 54 Escobar-Morreale HF, Carmina E, Dewailly D. et al. Epidemiology, diagnosis and management of hirsutism: a consensus statement by the Androgen Excess and Polycystic Ovary Syndrome Society. Hum Reprod Update 2012; 18: 146-170
  • 55 Di Luigi L, Pigozzi F, Sgrò P. et al. The use of prohibited substances for therapeutic reasons in athletes affected by endocrine diseases and disorders: the therapeutic use exemption (TUE) in clinical endocrinology. J Endocrinol Invest 2020; 43: 563-573
  • 56 World Anti-doping Agency (WADA). Therapeutic Use Exemption (TUE Available from https://www.wada-ama.org/en/what-we-do/science-medical/therapeutic-use-exemptions Accessed: May 2022
  • 57 Clark RV, Wald JA, Swerdloff RS. et al. Large divergence in testosterone concentrations between men and women: frame of reference for elite athletes in sex-specific competition in sports, a narrative review. Clin Endocrinol (Oxf) 2019; 90: 15-22
  • 58 Handelsman DJ, Hirschberg AL, Bermon S.. Circulating testosterone as the hormonal basis of sex differences in athletic performance. Endocr Rev 2018; 39: 803-829
  • 59 Ristori J, Cocchetti C, Romani A. et al. Brain sex differences related to gender identity development: genes or hormones?. Int J Mol Sci 2020; 21: 2123
  • 60 Eklund E, Berglund B, Labrie F. et al. Serum androgen profile and physical performance in women Olympic athletes. Br J Sports Med 2017; 51: 1301-1308
  • 61 Hirschberg AL, Elings Knutsson J, Helge T. et al. Effects of moderately increased testosterone concentration on physical performance in young women: a double blind, randomised, placebo-controlled study. Br J Sports Med 2020; 54: 599-604
  • 62 Labrie F, Martel C, Bélanger A. et al. Androgens in women are essentially made from DHEA in each peripheral tissue according to intracrinology. J Steroid Biochem Mol Biol 2017; 168: 9-18
  • 63 Huang G, Basaria S.. Do anabolic-androgenic steroids have performance-enhancing effects in female athletes?. Mol Cell Endocrinol 2018; 15: 56-64
  • 64 Miller KK, Biller BM, Beauregard C. et al. Effects of testosterone replacement in androgen-deficient women with hypopituitarism: a randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab 2006; 91: 1683-1690
  • 65 Huang G, Basaria S, Travison TG. et al. Testosterone dose-response relationships in hysterectomized women with or without oophorectomy: effects on sexual function, body composition, muscle performance and physical function in a randomized trial. Menopause 2014; 21: 612-623
  • 66 Eliakim A, Nemet D.. Endogenous hyperandrogenism and exercise capacity lessons from the exercise-congenital adrenal hyperplasia model. J Pediatr Endocrinol Metab 2010; 23: 1213-1219
  • 67 Hagmar M, Berglund B, Brismar K. et al. Hyperandrogenism may explain reproductive dysfunction in olympic athletes. Med Sci Sports Exerc 2009; 41: 1241-1248
  • 68 Kogure GS, Miranda-Furtado CL, Silva RC. et al. Resistance exercise impacts lean muscle mass in women with polycystic ovary syndrome. Med Sci Sports Exerc 2016; 48: 589-598
  • 69 Costa EC, Sá JCF DE, Stepto NK. et al. Aerobic training improves quality of life in women with polycystic ovary syndrome. Med Sci Sports Exerc 2018; 50: 1357-1366
  • 70 Knochenhauer ES, Key TJ, Kahsar-Miller M. et al. Prevalence of the polycystic ovary syndrome in unselected black and white women of the southeastern United States: a prospective study. J Clin Endocrinol Metab 1998; 83: 3078-3082
  • 71 Ferguson-Smith MA, Bavington LD.. Natural selection for genetic variants in sport: the role of Y chromosome genes in elite female athletes with 46,XY DSD1. Sports Med 2014; 44: 1629-1634
  • 72 Bermon S, Garnier PY, Hirschberg AL. et al. Serum androgen levels in elite female athletes. J Clin Endocrinol Metab 2014; 99: 4328-4335
  • 73 Caliskan Guzelce E, Eyupoglu D, Torgutalp S. et al. Is muscle mechanical function altered in polycystic ovary syndrome?. Arch Gynecol Obstet 2019; 300: 771-776
  • 74 Cardinale M, Stone MH.. Is testosterone influencing explosive performance?. J Strength Cond Res 2006; 20: 103-107
  • 75 Douchi T, Yamamoto S, Oki T. et al. Serum androgen levels and muscle mass in women with polycystic ovary syndrome. Obstet Gynecol 1999; 94: 337-340
  • 76 Rickenlund A, Carlström K, Ekblom B. et al. Hyperandrogenicity is an alternative mechanism underlying oligomenorrhea or amenorrhea in female athletes and may improve physical performance. Fertil Steril 2003; 79: 947-955
  • 77 Bermon S.. Androgens and athletic performance of elite female athletes. Curr Opin Endocrinol Diabetes Obes 2017; 24: 246-251
  • 78 Conte D, Romanelli F, Fillo S. et al. Aspirin inhibits androgen response to chorionic gonadotropin in humans. Am J Physiol 1999; 277: e1032
  • 79 Di Luigi L, Rossi C, Sgrò P. et al. Do non-steroidal anti-inflammatory drugs influence the steroid hormone milieu in male athletes?. Int J Sports Med 2007; 28: 809-814
  • 80 Di Luigi L, Guidetti L, Romanelli F. et al. Acetylsalicylic acid inhibits the pituitary response to exercise-related stress in humans. Med Sci Sports Exerc 2001; 33: 2029-2035
  • 81 Di Luigi L, Guidetti L, Pigozzi F. et al. Acute amino acids supplementation enhances pituitary responsiveness in athletes. Med Sci Sports Exerc 1999; 31: 1748-1754
  • 82 Di Luigi L.. Supplements and the endocrine system in athletes. Clin Sports Med 2008; 27: 131-51
  • 83 Hilton EN, Lundberg TR.. Transgender women in the female category of sport: perspectives on testosterone suppression and performance advantage. Sports Med 2021; 51: 199-214
  • 84 Jones BA, Arcelus J, Bouman WP. et al. Sport and transgender people: a systematic review of the literature relating to sport participation and competitive sport policies. Sports Med 2017; 47: 701-716
  • 85 International Olympic Committee. Framework on Fairness, Inclusion and Non-discrimination on the Basis of Gender Identity and Sex Variations, 2021 Available from https://stillmed.olympics.com/media/Documents/News/2021/11/IOC-Framework Fairness-Inclusion-Nondiscrimination-2021.pdf Accessed: May 2022
  • 86 Pigozzi F, Bigard X, Steinacker J. , on behalf of the International Federation of Sports Medicine (FIMS) and the European Federation of Sports Medicine Associations (EFSMA) et al. Joint position statement of the International Federation of Sports Medicine (FIMS) and European Federation of Sports Medicine Associations (EFSMA) on the IOC framework on fairness, inclusion and non-discrimination based on gender identity and sex variations. BMJ Open Sport Exerc Med 2022; 8: e001273
  • 87 Iuliano S, Izzo G, Zagari MC. et al. Endocrine management of transgender adults: a clinical approach. Sexes 2021; 2: 104-118
  • 88 Bermon S, Garnier PY.. Serum androgen levels and their relation to performance in track and field: mass spectrometry results from 2127 observations in male and female elite athletes. Br J Sports Med 2017; 51: 1309-1314
  • 89 Dubon ME, Abbott K, Carl RL.. Care of the transgender athlete. Curr Sports Med Rep 2018; 17: 410-418
  • 90 Gooren LJ, Giltay EJ, Bunck MC.. Long-term treatment of transsexuals with cross-sex hormones: extensive personal experience. J Clin Endocrinol Metab 2008; 93: 19-25
  • 91 van Dijk D, Dekker MJHJ, Conemans EB. et al. Explorative prospective evaluation of short-term subjective effects of hormonal treatment in trans people–results from the European network for the investigation of gender incongruence. J Sex Med 2019; 16: 1297-1309
  • 92 Zhao H, Zhou L, Li L. et al. Shift from androgen to estrogen action causes abdominal muscle fibrosis, atrophy, and inguinal hernia in a transgenic male mouse model. Proc Natl Acad Sci USA 2018; 115: e10427-e10436
  • 93 Catelan RF, Saadeh A, Lobato MIR. et al. Depression, self-esteem, and resilience and its relationship with psychological features of sexuality among transgender men and women from Brazil. Arch Sex Behav 2022; 51: 1993-2002
  • 94 Sarno EL, Dyar C, Newcomb ME. et al. Relationship quality and mental health among sexual and gender minorities. J Fam Psychol 2022; 36: 770-779
  • 95 Romani A, Mazzoli F, Ristori J. et al. Psychological wellbeing and perceived social acceptance in gender diverse individuals. J Sex Med 2021; 18: 1933-1944
  • 96 Allison KF, Keenan KA, Sell TC. et al. Musculoskeletal, biomechanical, and physiological gender differences in the US military. US Army Med Dep J 2015; 22-32
  • 97 Ngun TC, Ghahramani N, Sánchez FJ. et al. The genetics of sex differences in brain and behavior. Front Neuroendocrinol 2011; 32: 227-246
  • 98 Zheng D, Wang X, Antonson P. et al. Genomics of sex hormone receptor signaling in hepatic sexual dimorphism. Mol Cell Endocrinol 2018; 15: 33-41
  • 99 Boldt P, Knechtle B, Nikolaidis P. et al. Sex differences in the health status of endurance runners: results from the NURMI study (step 2). J Strength Cond Res 2019; 33: 1929-1940
  • 100 Lepers R, Knechtle B, Stapley PJ.. Trends in triathlon performance: effects of sex and age. Sports Med 2013; 43: 851-863
  • 101 Handelsman DJ.. Sex differences in athletic performance emerge coinciding with the onset of male puberty. Clin Endocrinol (Oxf) 2017; 87: 68-72
  • 102 Oyola MG, Handa RJ.. Hypothalamic-pituitary-adrenal and hypothalamic-pituitary-gonadal axes: sex differences in regulation of stress responsivity. Stress 2017; 20: 476-494
  • 103 Roberts TA, Smalley J, Ahrendt D.. Effect of gender affirming hormones on athletic performance in transwomen and transmen: implications for sporting organisations and legislators. Br J Sports Med . 2020
  • 104 Elbers JM, de Jong S, Teerlink T. et al. Changes in fat cell size and in vitro lipolytic activity of abdominal and gluteal adipocytes after a one-year cross-sex hormone administration in transsexuals. Metabolism 1999; 48: 1371-1377
  • 105 Klaver M, de Blok CJM, Wiepjes CM. et al. Changes in regional body fat, lean body mass and body shape in trans persons using cross-sex hormonal therapy: results from a multicenter perspective study. Eur J Endocrinol 2018; 178: 163-171
  • 106 Klaver M, Dekker MJHJ, de Mutsert R. et al. Cross-sex hormone therapy in transgender persons affects total body weight, body fat and lean body mass: a meta-analysis. Andrologia 2017; 49: e12660
  • 107 Van Caenegem E, Wierckx K, Taes Y. et al. Preservation of volumetric bone density and geometry in trans women during cross-sex hormonal therapy: a prospective observational study. Osteoporos Int 2015; 26: 35-47
  • 108 Wiik A, Lundberg TR, Rullman E. et al. Muscle strength, size, and composition following 12 months of gender-affirming treatment in transgender individuals. J Clin Endocrinol Metab 2020; 105: dgz247
  • 109 Gooren LJ, Bunck MC.. Transsexuals and competitive sports. Eur J Endocrinol 2004; 151: 425-429
  • 110 Haraldsen IR, Haigh E, Falch J. et al. Cross-sex pattern of bone mineral density in early onset gender identity dsorder. Horm Behav 2007; 52: 334-343
  • 111 Mueller A, Zollver H, Kronawitter D. et al. Body composition and bone mineral density in male-to-female transexuals during cross-sex hormone therapy using gonadotrophin-releasing hormone agonist. Exp Clin Endocrinol Diabetes 2011; 119: 95-100
  • 112 Wierckx K, Van Caenegem E, Schreiner T. et al. Cross-sex hormone therapy in trans persons is safe and effective at short-time follow-up: results from the European network for the investigation of gender incongruence. J Sex Med 2014; 11: 1999-2011
  • 113 Gava G, Cerpolini S, Martelli V. et al. Cyproterone acetate vs leuprolide acetate in combination with transdermal oestradiol in transwomen: a comparison of safety and effectiveness. Clin Endocrinol (Oxf) 2016; 85: 239-246
  • 114 Auer MK, Ebert T, Pietzner M. et al. Effects of sex hormone treatment on the metabolic syndrome in transgender individuals: focus on metabolic cytokines. J Clin Endocrinol Metab 2018; 103: 790-802
  • 115 Fighera TM, da Silva E, Lindenau JDR. et al. Impact of cross-sex hormone therapy on bone mineral density and body composition in transwomen. Clin Endocrinol (Oxf) 2018; 88: 856-862
  • 116 Scharff M, Wiepjes CM, Klaver M. et al. Change in grip strength in trans people and its association with lean body mass and bone density. Endocr Connect 2019; 8: 1020-1028
  • 117 Tack LJW, Craen M, Lapauw B. et al. Proandrogenic and antiandrogenic progestins in transgender youth: differential effects on body composition and bone metabolism. J Clin Endocrinol Metab 2018; 103: 2147-2156
  • 118 Polderman KH, Gooren LJG, Asscheman H. et al. Induction of insulin resistance by androgens and estrogens. J Clin Endocrinol Metab 1994; 79: 265-271
  • 119 D'Andrea S, Pallotti F, Senofonte G. et al. Polymorphic cytosine-adenine-guanine repeat length of androgen receptor gene and gender incongruence in trans women: a systematic review and meta-analysis of case-control studies. J Sex Med 2020; 17: 543-550
  • 120 Egner IM, Bruusgaard JC, Eftestøl E. et al. A cellular memory mechanism aids overload hypertrophy in muscle long after an episodic exposure to anabolic steroids. J Physiol 2013; 591: 6221-6230
  • 121 Gundersen K.. Muscle memory and a new cellular model for muscle atrophy and hypertrophy. J Exp Biol 2016; 219: 235-242
  • 122 Nicoll JX, Fry AC, Mosier EM.. Sex-based differences in resting MAPK, androgen, and glucocorticoid receptor phosphorylation in human skeletal muscle. Steroids 2019; 141: 23-29
  • 123 Scott NL, Abreu MR, Cates BE. et al. Prolonged effects of elevated 17β-estradiol on physical activity after orchidectomy. Med Sci Sports Exerc 2018; 50: 1588-1595
  • 124 Hamilton BR, Martinez-Patiño MJ, Barrett J.. et al. Response to the United Nations Human Rights Council’s report on race and gender discrimination in sport: an expression of concern and a call to prioritise research. Sports Med 2021; 51: 839-842