Next Article in Journal
Characterization, In Vitro Biological Activity and In Vivo Cardioprotective Properties of Trametes versicolor (L.:Fr.) Quél. Heteropolysaccharides in a Rat Model of Metabolic Syndrome
Next Article in Special Issue
Sex-Related Differences in the Pharmacological Response in SARS-CoV-2 Infection, Dyslipidemia, and Diabetes Mellitus: A Narrative Review
Previous Article in Journal
Compounded Effervescent Magnesium for Familial Hypomagnesemia: A Case Report
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evaluation of Sex Differences in Preclinical Pharmacology Research: How Far Is Left to Go?

Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, 10043 Orbassano, Italy
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(6), 786; https://doi.org/10.3390/ph16060786
Submission received: 20 April 2023 / Revised: 10 May 2023 / Accepted: 23 May 2023 / Published: 24 May 2023
(This article belongs to the Special Issue Sex Differences in Pharmaceutical Practice)

Abstract

:
Until the last quarter of the 20th century, sex was not recognized as a variable in health research, nor was it believed to be a factor that could affect health and illness. Researchers preferred studying male models for a variety of reasons, such as simplicity, lower costs, hormone confounding effects, and fear of liability from perinatal exposure in case of pregnancy. Equitable representation is imperative for determining the safety, effectiveness, and tolerance of therapeutic agents for all consumers. Decades of female models’ underrepresentation in preclinical studies has resulted in inequality in the understanding, diagnosis, and treatment of disease between the sexes. Sex bias has been highlighted as one of the contributing factors to the poor translation and replicability of preclinical research. There have been multiple calls for action, and the inclusion of sex as a biological variable is increasingly supported. However, although there has been substantial progress in the efforts to include more female models in preclinical studies, disparities today remain. In the present review, we consider the current standard practice of the preclinical research setting, why the sex bias exists, why there is the need to include female models, and what risks may arise from continuing this exclusion from experimental design.

1. Therapeutic Agent Development Is a Significant Challenge

Therapeutic agent development is driven by medical need, disease prevalence, and the likelihood of success. Therapeutic agent candidate selection is an iterative process between chemistry and biology, whose aim is to refine the molecular properties until a compound suitable for advancing to humans is found. It is an expensive, long, and high-risk process which takes from 10 to 15 years, and it is associated with a high attrition rate; 80 to 90% of research projects fail before they ever get tested in humans, and for every therapeutic agent that gains Food and Drug Administration (FDA) approval, more than 1000 were developed but failed. Prior to administration to humans, the pharmacology and biochemistry of the therapeutic agent is established using an extensive range of in vitro and in vivo test procedures during the preclinical studies [1]. Indeed, starting from the study of the medicine effects on cell cultures, the process goes through animal experimentation, ending with its use in the human species in different clinical phases. It is also an FDA regulatory requirement that the therapeutic agent is administered to animals to assess its safety. Later-stage animal testing is also required to assess the carcinogenicity and the effects on the reproductive system. The goal of preclinical studies is to accurately model, in animals, the desired biological effect of a medicine. This allows doctors to predict the treatment outcome in patients, and to identify toxicities associated with a therapeutic agent, with the aim of predicting adverse events in people [2].
There has been a revolution within clinical trials to include females in the research pipeline. However, there has been limited change in the preclinical arena, yet the preclinical research lays the groundwork for the subsequent clinical trials. In 1993, the Council for International Organizations of Medical Sciences proposed a law, approved by the FDA, requiring women to be included in clinical trials funded by the National Institutes of Health (NIH). Women of “child-bearing potential” were indeed excluded from clinical trials until 1993; until 1993, zero women have ever been enrolled in clinical trials. The NIH issued a mandate in 2014 to guarantee that both male and female subjects are represented in preclinical studies [3]. A comparable program by the Canadian Institute of Health that mandated inquiries about sex and gender during the application process for research funding resulted in a marked rise in the number of applications that took into account both the male and female sexes (from 26 to 48%) [4]. They made a point to mention that scientists working in the biological field were the least likely to admit to including sex in their study. The prospects for policy measures to address sex bias are highlighted by these findings. Unfortunately, not all research is sponsored through these channels, and the inclusion of both the male and female sexes in research pipelines is not yet achieved.
Kim and colleagues looked at the sex annotation of cells in pertinent articles published in the same journal in 2018 to determine how far the reporting of cell sex in studies has come since the examination in 2013. A total of 53 of the 107 papers describing cell experiments reported the sex of the cells; 12 studies only used female cells, 18 used both male and female cells, and 23 only used male cells. Cell lines were employed more frequently than primary cells, which resulted in more instances of sex omission. More than half of the investigations in the articles discussing trials utilizing mouse primary cells solely employed male cells [5]. In other studies, the authors described the 10-year results and lessons discovered from applicant forms, the development of resources for applicants and evaluators, and the grant review requirements in an effort to inform the implementation of scientific policy. The group included all participants in 15 Canadian Institutes of Health Research contests that were started by investigators between 2011 and 2019, as well as grant evaluators between 2018 and 2019. A total of 39,390 applications have been submitted since 2011. The percentage of reports integrating gender and sex increased from 12 to 33% and 22 to 83%, respectively. Applications in population health research gave gender the most consideration (82%). Applications with main investigators who were female were more likely to integrate sex (and gender) than those with male principal investigators throughout every competition. Since 2018, applications with strong sex integration scores and high gender integration scores have a higher chance of receiving funding. Qualitative observations showed that sex and gender were frequently confused [6]. A series of publications look at the preclinical research setting in existing normative practice, the drive to include women, and the reasons behind the discrepancy. They investigate organizational change theory as a tool for developing the institutional and individual strategies required to alter the present situation to establish a scientific environment where sex-sensitive methods are automatically implemented in preclinical research [7,8,9].

2. Methods of Research

The search was performed in PubMed (https://pubmed.ncbi.nlm.nih.gov/, accessed on 1 November 2022) databases starting from November 2022. The following terms were used to find papers that were pertinent: “sex”, “preclinical studies”, “pharmacokinetics”, “pharmacodinamics”, “animal models”, “cellular culture”, “male”, and “female” and “transegender”. The considered timeline is 1973-–2022. Through article search, approximately 1600 papers were initially identified; duplicate and unrelated articles were deleted. Of the 89 remaining articles after assessing the full text for eligible articles, 46 were excluded, leaving 43 in the study.
We looked through the references in the review articles to find any additional qualifying original manuscripts. Only English-language articles were chosen. In this review, we summarize the studies on sex evaluation in preclinical research pharmacological studies, sex differences in animal models used in pharmacological research, sex differences in cellular models used in pharmacological research, related adverse reactions, and patients’ age.
Clinical trial-related publications were eliminated since they failed to relate to the goal of the study that is presented.

3. Gender-Specific Pharmacokinetics and Pharmacodynamics

Women and men respond differently to treatments; this mainly depends on physiological, anatomical, and hormonal characteristics. The existence of the differences in therapeutic agent pharmacokinetics and pharmacodynamics influences the response to treatments [10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45]. Although this was already known since 1932, the year in which the first study on the gender difference in the pharmacology of barbiturates in rats was reported, full awareness of the relevance of the role of gender pharmacology only came at the end of the last century [16,26,27,46,47]. By pharmacokinetics, we mean the study of the following four phases of a medicine transition in the body: absorption, distribution, metabolism, and elimination. These four stages are primarily influenced by age and hormones, thus showing significant differences related to sex [48]. Sex hormones may interact negatively with drugs and their metabolic pathways through different mechanisms; these include absorption, competition for transporters, competition and/or regulation of expression of drug-metabolizing enzymes and sex steroids, and drug interactions with pharmacodynamics [49]. In females, drug effectiveness and adverse drug reactions may be impacted by variations in endogenous sex steroid hormones that happen naturally during the menstrual cycle, during pregnancy, and during the transition to menopause [50]. Exogenous hormones are additionally used by women as a contraceptive, as a treatment for hot flashes, nocturnal sweats, vaginal dryness, and a number of other conditions. Hence, these hormone therapies could be viewed as both medical treatments and a cause of adverse drug reactions, as well as a modulator of the effects of other medications. In other words, while drug metabolic pathways may have an impact on exogenous hormones used for therapy, exogenous hormones may also have an impact on other drugs via modifying metabolism. There may be variations in how each person reacts to exogenous hormones due to inter-individual variations in metabolic pathway components (pharmacogenetics) [49].
Male reproductive aging does not result in a complete cessation of testosterone production or spermatogenesis, unlike female reproductive aging (menopause) or organic androgen lack in males (caused by diseases of the brain, pituitary, or testes). In fact, the decrease in circulating testosterone levels brought on by aging in and of itself is moderate, and testosterone levels are typically in the low–normal range in men. However, a small percentage of aging men may experience testosterone deficiency, which is influenced by the presence of comorbidities. According to recent research, elderly men who maintain their health and fitness typically keep their serum testosterone levels regular. The terms andropause, viropause, partial androgen deficit in the aging male, and late-onset hypogonadism have all been used to describe age-related low testosterone in men [51]. Additionally, the gonadal axis is suppressed by the use of some medications, including glucocorticoids and opioids [52,53,54]. Considering pharmacogenetics, it has been observed that cells that carry the UGT1A4*1a allele may have decreased clearance of testosterone as compared to those with the *3a allele [55].
Pharmacodynamics, on the other hand, indicates the effect of a therapeutic agent on bodies and studies the biochemical and physiological effects and their mechanism of action. There are numerous pharmacodynamic differences depending on sex, mainly mediated by hormones, genes, and the environment. The primary organ for excreting drug metabolites or parent drug molecules is the kidney. All three of the main renal processes—glomerular filtration, tubular secretion, and tubular reabsorption—show documented sex differences. Men often have a higher renal clearance than women. The hepatic enzyme activity is altered by elevated levels of estrogen and progesterone, which can lead to an increased drug buildup or decreased drug clearance in some cases. Autoimmunity is influenced by prolactin and female steroid hormones [37]. The incidence and severity of autoimmune/inflammatory illnesses are two to ten times higher in females than in males due to the hypothalamic–pituitary–adrenal and hypothalamic–pituitary–gonadal axes’ regulation of immunity. Females of reproductive age are the ones who typically have autoimmune illnesses. Hormone levels that fluctuate during menstruation, when using oral contraceptives, during pregnancy, or throughout menopause can also affect metabolic alterations. For instance, some asthmatic women experience symptoms that intensify before or during their periods. Intensive physical activity has been linked to an increase in oxidative stress. Oxidative stress has been linked to gender disparities, particularly as people get older. Studies to investigate this have produced contradictory results, despite the fact that sex hormones are thought to have a major role in modifying sex-based differences in pharmacokinetics. Midazolam clearance, for example, which measures the CYP3A4 metabolic activity, did not change during the course of the menstrual cycle [56]; in addition, studies on eletriptan (used to treat migraines) also showed no changes in response to sex or menstrual cycle [57].
While the pharmacokinetic differences are simpler to analyze, the pharmacodynamic differences are more difficult to detect [26]. However, both deserve a worthy study in the preclinical phase in terms of gender differences, or the resulting clinical phase will be limited and approximate.

4. The Current Standard Practice of Pharmacological Preclinical Research

The main goal of the preclinical studies is to define the therapeutic agent pharmacokinetics and pharmacodynamics, anticipating the active level in the target compartment, to predict the safe starting dose and dose escalation scheme for phase 1 clinical trials. Preclinical research exhibits an endemic persistent sex bias that mostly concentrates on male animals [58,59]; this bias is still visible even when the disease of interest is a disorder that is more common in women. Yoon et al. discovered that only 12% of papers looking at illnesses common in women included studies of females or both the male and female sexes [60].
However, the gender bias is not only observed in the in vivo studies; the in vitro studies have historically ignored the relevance of sex of origin, and it is often still ignored. In the preclinical experimentation phase, the use of female animals is relatively low and the cells are usually considered asexual, since the sex of the donor is not reported, not considering, instead, that even cultured cells have a sex, at least during the first maintenance steps [58,61,62,63]. Despite this, pharmacodynamic differences are increasingly emerging, concerning increasingly relevant pharmacological targets [18,27,46,63,64,65,66]. Hormones have a key role in the modulation of pharmacodynamic reactions [49,67,68]; they regulate many functions within our organism, thanks to the interaction with specific intracellular receptors [69]. Estrogens and androgens behave differently, inducing highly variable responses from a molecular point of view [70,71,72,73]. Furthermore, hormonal changes depend on the age and reproductive life of the woman as they are conditioned by fertile age, pregnancy, postnatal period, and menopause [74]. In addition to hormones, genetics also regulate responses to treatment differently between the sexes.
However, recently, something has been changing. In 2018, Cvitanovíc Tomas and colleagues modified the SteatoNet in silico model to build the computer model LiverSex [75,76], which accounts for sex differences in the liver. Sex-related effects on growth hormone release are included in the data from estrogen and androgen receptor responses. The model has not yet been validated in people, but only in mice. In 2020, Thiele et al. developed sex-specific whole-body metabolic models [77]; 20 organs, 6 sex organs, 6 different blood cell types, systemic blood circulation, the blood–brain barrier, and the gastrointestinal lumen, including the microbiome, were used to illustrate the male and female physiologies.

5. Why the Sex Bias Exists (and It Is Very Robust)

In humans, sex is determined by sex chromosomes. The X and Y chromosomes carry different numbers and sets of genes; about 1000 genes are carried on the X chromosome, and only a few dozen genes are carried on the Y chromosome. A series of recombination events, followed by the loss of genetic material on the Y gene, leads to the morphological differentiation of the sex chromosomes [3]. Most genes on the sex chromosomes are not directly involved in sex determination, and the development of male or female, for a long time, has been mainly attributed to the presence of a single locus, the sex-determining region gene on the Y chromosome (SRY). Other studies have shown that this concept is much more variable than hypothesized, and alternative mechanisms can play a role on sexual development, widely different from that expected by the karyotype. Thus, there is growing proof that the sex-limited chromosome in some systems evolved independently and has no connection to the X or Z chromosomes. For instance, B chromosomes, which are small, unimportant chromosomes that frequently transmit selfishly, serve as the Y chromosome in Rhinocola aceris and Cacopsylla peregrina as well as a W chromosome in several Lake Malawi cichlids [78,79]. Strong evidence suggests that the Lepidoptera W chromosome evolved after the Z chromosome, likely from a B chromosome [80]. In the case of the pill bug (Armadillium vulgare), a Wolbachia feminizer that has been integrated into the nuclear genome gave rise to the W chromosome [81]. This opened the interesting potential that cytoplasmic male sterility factors, which are widespread in both insects and plants, may offer opportunities for the creation of non-homologous W chromosomes when they are transported to the nuclear genome.
Furthermore, the deep genetic difference between the sexes is also indicated by older studies. In 1961, Mary Lyon suggested that one of the two X chromosomes in females becomes genetically silent early in a female embryo’s development. Additionally, this happens randomly from cell to cell, connoting biological females with incredible genetic mosaicism [82].
A striking number of exceptions, and thus, a parallel diversity of underlying mechanisms, are revealed by the diversity of the sex chromosomes. This variability shows that the laws governing sex chromosomal evolution are complex and not universal. The systems that diverge or turn over most frequently may be the most instructive going forward, as comparisons can be made to separate cause from effect [83].
The International Mouse Phenotyping Consortium revealed that sex was a substantial source of variance within the control data and as a modulator of a treatment effect by analyzing data from 10 institutes, 14,000 wild type mice, and 40 thousand knockout mice for 234 features [84]. Sexually dimorphic effects in a wide range of biological systems highlights the importance that we should always examine both the male and female sexes and take sex into account as a source of variation.
Enzymes involved in therapeutic agent metabolism have sexually dimorphic expression patterns in a variety of species, which has an impact on its metabolism. In humans, the mRNA and protein measurements of the cytochrome p450 (CYP) 3A4 in the liver reveal higher levels in females [85]. Additionally, studies found that this enzyme is more active in females [86]. Rat and mouse livers exhibit a high level of sexually dimorphic gene expression [87,88,89,90,91]. For instance, rats express a male-specific CYP2C11 pattern, whereas the CYP2C12 is exclusive to women [92]. When comparing age-matched male and female mouse liver microsomes, the CYP1A2 was consistently more prevalent in males. Male and female mice 3 to 4 weeks of age had higher levels of hepatic expression of the CYP2B9 than mice of other ages, and the CYP2B9 was the only enzyme that was found in pregnant mouse liver microsomes at higher levels than age-matched females. It is interesting to note that only the kidney showed a sexually dimorphic expression of the CYP2B9, 2D26, 2E1, and 4B1 [93]. The complexity of the metabolic pathways emphasizes the significance of comprehending medicine exposure in each sex, at the correct time point, and in the relevant tissue when conducting pharmacodynamic assessments, even though rodent sex differences may not always transfer into similar patterns in humans.
The female estrous cycle is partly responsible for the exclusion of females from some research. Rats go through their estrous cycle for 4–5 days. Progesterone rises quickly, starting early in the metestrum phase of the post-ovulatory cycle on day 1, and falls quickly in the diestrum phase on day 2. Ovulation brings a significant increase in progesterone release and estrogen levels that occur during proestrum. When estrous occurs on day 4, the hormone levels return to normal after a transient temporary rise in estradiol [94]. It is believed that the estrous cycle introduces unpredictability into experimental procedures. The therapeutic agent responsiveness in females can also be impacted by hormonal fluctuations during the menstrual cycle [95,96]. To eliminate the effects of the estrous cycle, it was suggested that pharmacological tests be administered only during diestrum, that only male models be used, or that a counterbalanced design be used to average any changes [97]. Ovariectomy and castration procedures may be used to remove gonadal influences from the female and male rats to examine the effect of hormonal changes on experimental outcomes. To research the physiological effects of circulating hormones, these animals’ behavioral and neurochemical profiles can be compared to those of properly cycling females and intact males. Exogenous hormonal therapies, such as estradiol, progesterone, or both, and, if required, testosterone, may also be given to castrated and ovariectomized animals.

6. Excursus on Preclinical Pharmacology Research: Sex Disaggregated Data or Not?

Drug use and abuse differ in men and women [98,99,100]. Women seem to be more susceptible to drug use acquisition, maintenance, regulation, and relapse phases. This increased susceptibility may be facilitated by the ovarian cycle, as higher estrogen levels were linked to higher levels of illicit drug use among women [101]. The influence of sex and gonadal hormones on the behavioral and neurochemical reactions to addictive substances was better understood using animal models. In Table 1, we summarize the preclinical pharmacological studies which include sex evaluation.
Psychedelics, commonly referred to as hallucinogens, have an impact on sensory processing, perception, and cognition, primarily through the serotonin 5-HT2A receptor (5-HT2AR). As it relates to mental illnesses including depression and substance use disorders, this class of psychoactive drugs—which includes lysergic acid diethylamide, psilocybin, mescaline, and the substituted amphetamine 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI)—is gaining increasing attention. Sex is frequently left out of research that examines the possible clinical effects of hallucinogenic drugs on human subjects. Rodent models have contributed much to our understanding of the pharmacology of psychedelics, yet most preclinical research has just looked at male mice. Jaster and colleagues examined the impact of DOI on male and female mice’s head-twitch behavior, a rodent behavioral model of the potential of psychedelic drugs in humans. In the C57BL/6J mice, DOI causes more ovariectomized behavior in females than in males, a sex-specific increased behavior that was not even seen in 129S6/SvEv animals. In both the male and female C57BL/6J mice, volinanserin, a 5-HT2AR antagonist, completely inhibited the DOI-induced ovariectomized behavior. There was no sex-related difference in the amount of inositol monophosphate that accumulated in the frontal cortex after the DOI treatment in the C57BL/6J mice. However, the pharmacokinetic characteristics of the DOI varied between the sexes; the female C57BL/6J mice had lower brain and plasma levels of DOI 30 and 60 min after treatment than the male C57BL/6J mice [102].
Understanding how sex affects cannabinoid pharmacology is crucial given the growing popularity of cannabis edibles for medicinal purposes and the continuous abuse of these products. Female rats appear to be more sensitive to several consequences of cannabinoid use, such as anti-nociception, discriminative stimulus, and reinforcing effects, according to the studies that have examined the sex differences in the behavioral effects of cannabinoids [103,104,105,106,107]. Interestingly, for the optimum acquisition and preservation of stimulus control in delta-9-tetrahydrocannabinol (THC) discrimination protocols, the female rats required a lower THC training dose than the male rats [106,108]. When the rats of either sex were trained to distinguish between identical THC doses, THC was also more effective at inducing discriminative stimulus effects in the female Sprague Dawley rats than in the males of the same strain. However, the THC potency to produce discriminative stimulus effects was comparable in the C57BL/6J mice of both the male and female sexes treated to distinguish 5.6 mg/kg of THC from the vehicle as well as in the mice trained to distinguish a higher dose of THC (30 mg/kg) [109]. In 2020, Wiley et al. investigated the effects of the intraperitoneal administration of THC and its main psychoactive metabolite, 11-OH-THC, in rodent models of psycho-activity and molecular assays of cannabinoid receptor type-1 pharmacology on the effects of age, sex, and rodent species. They observed that both 11-OH-THC and THC functioned as partial agonists in guanosine triphosphate, labeled on the gamma phosphate group with equal intensity in both the species and the male and female sexes [110].
Cannabinoids also demonstrated promise in the treatment of several challenging pain conditions and may provide a non-opioid alternative for the long-term control of chronic inflammatory pain [111,112]. In comparison to men, women report a higher prevalence of chronic pain as well as higher levels of experimentally produced and postoperative pain [113,114,115]. In a mouse model of inflammatory pain, LaFleur et al. observed that, in comparison to the male mice, the female mice had a lower susceptibility to the effects of Δ-9-THC and a synthetic cannabinoid. The attenuation of nociceptive behaviors for both agonists in both the male and female sexes was exacerbated by the S426A/S430A mutation. Compared to the male mice, the female mice showed a delayed tolerance to Δ-9-THC, whereas the S426A/S430A mutation caused a delayed tolerance in both the male and female sexes. Compared to the wild-type controls, the male S426A/S430A mutant mice exhibited a resistance to tolerance to the synthetic cannabinoid [116].
A preclinical investigation on female and male Sprague Dawley rats and male CD1 mice evaluated NKTR-181 (the novel mu-opioid receptor agonist NKTR-181 with a limited entry into the brain) preclinical pharmacology in comparison to commonly utilized mu-opioid receptor agonists; in the hot-water tail-flick test, NKTR-181 demonstrated dose- and time-related anti-nociception with peak effects comparable to morphine, without sex or species differences [117].
In preclinical diabetes research, animal models are crucial. As one of the most often carried out studies in metabolic research, glucose tolerance tests (GTTs) are used to test new antidiabetic therapies on raised blood glucose concentrations [118]. Female mice are frequently excluded from studies on diabetes because they lose some of their glucose intolerance and insulin resistance [119,120,121,122]. In fact, it is generally accepted that this relative lack of phenotypic behavior reduces their value in treatment efficacy trials by introducing preclinical bias and possibly obstructing translation to a different clinical population. It might also be the reason why researchers are hesitant to investigate female mice because of their perceived higher variability in blood glucose levels throughout the estrous cycle [123,124]. Despite a rising emphasis on the necessity to take into account sex as a biological variable in preclinical studies, the effect of sex and the estrous cycle on blood glucose fluctuation in the GTT has not previously been examined in depth [3,8,124]. The effects of sex, dosage method, length of fasting, and acute habituation stress was examined by Kennard and colleagues in relation to glucose tolerance test (GTT) measures used in the preclinical assessment of putative glucose-modulating therapies. When starting a fast, the researchers noticed that the female mice were less susceptible to human involvement. After a 6 h fast, both the male and female mice’s basal blood glucose levels stabilize more quickly when the bedding is kept intact while the cage base is changed. Sixteen hours of continuous fasting produced an inflated GTT response but a substantial basal hypoglycemia. Exendin-4 and metformin had a similar effect after GTT protocol optimization, with the female mice exhibiting a more moderate but repeatable GTT response [125].
For the treatment of obesity and diabetes, unimolecular peptides that target the glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors (GLP-1/GIP co-agonist) are now being tested in clinical settings [126,127]. From rodent models of obesity to non-human primates and humans, their effectiveness to enhance body weight, glucose management, and lipid metabolism outperforms best-in-class GLP-1 monotherapies [126,127,128]. Sachs et al. sought to identify biomarkers to promote non-invasive metabolic monitoring of compound treatment efficacy and research of additional treatment effects on an individual basis despite sex-specific plasma proteome profiling differences. In comparison to mono-agonist therapies, the GLP-1R/GIPR co-agonist significantly reduced obesity, glucose intolerance, non-alcoholic fatty liver disease, and dyslipidemia in both the male and female mice. In comparison to mono-agonist treatments, proteome profiling differences in both the male and female mice indicated larger alterations in plasma proteins after the GLP-1/GIP co-agonist [129].
Table 1. Summary of the preclinical pharmacological studies which include sex evaluation.
Table 1. Summary of the preclinical pharmacological studies which include sex evaluation.
Ref #First AuthorYearJournalTopic
[38]Song, D.2018Pharmaceutical ResearchInflammation
[102]Jaster, A.M.2022Neuroscience LettersPsychiatry
[103]Craft, R.M.2013Life SciencesCannabinoid
[104]McGregor, I.S.2007British Journal of PharmacologyCannabinoid
[105]Cooper, Z.D.2018NeuropsychopharmacologyCannabinoid
[106]Wiley, J.L.2017Drug and Alcohol DependenceCannabinoid
[107]Craft, R.M.2013PainCannabinoid
[108]Winsauer, P.J.2012Pharmacology, Biochemistry and BehaviorCannabinoid
[109]Wiley, J.L.2011Behavioural PharmacologyCannabinoid
[110]Wiley, J.L.2021Progress in Neuro-Psychopharmacology & Biological PsychiatryCannabinoid
[111]Blake, D.R.2006Rheumatology (Oxford)Cannabinoid
[112]Johnson, J.R.2013Journal of Pain and Symptom ManagementCannabinoid
[113]Nahin, R.L.2012The Journal of PainPain
[114]Riley, J.L.1998PainPain
[115]Aubrun, F.2005AnesthesiologyAnalgesia
[116]LaFleur, R.A.2018NeuroreportCannabinoid
[117]Kopruszinski, C.M.2021Cellular and Molecular NeurobiologyAnalgesia
[118]Pacini, G.2013Journal of Diabetes ResearchEndocrinology
[119]Nyavor, Y.2019Cell and Tissue ResearchEndocrinology
[120]Kaikaew, K.2019EndocrinologyEndocrinology
[121]Pettersson, U.S.2012PLoS OneEndocrinology
[122]Rebolledo-Solleiro, D.2018Physiology and BehaviorEndocrinology
[123]Bartke, A.1973EndocrinologyEndocrinology
[124]Beery, A.K.2018Current Opinion in Behavioral SciencesEndocrinology
[125]Kennard, M.R.2022Diabetes, Obesity and MetabolismEndocrinology
[129]Sachs, S.2021Diabetes, Obesity and MetabolismEndocrinology
[130]Kremer, J.J.2015Journal of Pharmacological and Toxicological MethodsCardiology
[131]Bourdi, M.2020Regulatory Toxicology and PharmacologyCancer
[132]Ewertz, M.2015Acta OncologicaCancer
[133]Kotaka, M.2020Cancer Chemotherapy and PharmacologyCancer
[134]Minami, T.2020European Journal of PharmacologyCancer
[135]Earp, J.C.2009Pharmaceutical ResearchRheumatology
[136]Dubois, D.C.2008Journal of Pharmacology and Experimental TherapeuticsRheumatology
[137]Earp, J.C.2008Journal of Pharmacology and Experimental TherapeuticsRheumatology
[138]Fletcher, C.V.2014Proceedings of the National Academy of SciencesAntiretroviral drugs
[139]Thompson, C.G.2015Antimicrobial Agents and ChemotherapyAntiretroviral drugs
[140]Cottrell, M.L.2016The Journal of Infectious DiseasesAntiretroviral drugs
[141]Dimopoulos, Y.2017Current HIV/AIDS ReportsAntiretroviral drugs
[142]Burgunder, E.2019Journal of Pharmacology and Experimental TherapeuticsAntiretroviral drugs
# reference number.
In a study on dog models by Kremer et al. beagles that were given etilefrine, sotalol, and hydralazine were implanted with a miniature telemetry blood pressure transmitter to monitor their blood pressure. Both the males and female beagles reported changes in blood pressure as a consequence of the etilefrine. Both the hydralazine and sotalol had similar effects on both the male and female beagles and continued for 19 h post-dose. The exposure levels were dose-dependent and quantifiable between 7 and 7.5 h post-dose in the dogs that were given etilefrine, sotalol, and hydralazine. The male and female beagles exposed to 10 mg/kg of etilefrine experienced different concentrations (169 vs. 268 ng/mL or 69%), but generally, there were only small differences [130].
Bourdi and colleagues conducted in vitro and in vivo studies on metarrestin safety evaluation in beagles; they observed a dose-related increase in systemic exposure with no sex difference on days 1 and 27. From day 1 to day 27, metarrestin accumulated in both the male and female dogs and at all dose levels. No adverse effects were reported on the other days’ dosage for 28 days; the level in the dogs was estimated to be 0.25 mg/kg of metarrestin, with a mean male and female maximum concentration of 82.5 ng/mL and an exposure of 2521 h·ng/mL on day 27 [131].
The use of the platinum-based chemotherapeutic oxaliplatin for the treatment of colorectal cancer comes with a major dose-limiting adverse effect, peripheral neuropathy [132]. In a recent clinical phase 2 research, thrombomodulin alfa, a recombinant human soluble thrombomodulin, was demonstrated to inhibit oxaliplatin-induced peripheral neuropathy [133]. A preclinical pharmacology on rats intravenously treated with oxaliplatin (6 mg/kg) was conducted; treatment-induced mechanical hyperalgesia was inhibited by a single intravenous infusion of thrombomodulin alfa in a dose-dependent manner, with no sex-related differences in the effectiveness [134].
Preclinical pharmacologic studies on rheumatoid arthritis frequently employ collagen-induced arthritic rats [135,136,137]. In the first analysis on sex differences in collagen-induced arthritic rats, the male rats reported 43% larger dexamethasone clearances; in the female rats, the temporal patterns of paw edema showed earlier development, earlier times of peak edema, and earlier illness remission. In both the male and female rats with comparable capacity values, dexamethasone effectively reduced paw edema, but the dexamethasone potency was lower in the females [38].
Low-level viral replication within tissues as a result of insufficient antiretroviral (ARV) penetration is one potential reason of viral rebound [138]. Studies on therapeutic agent concentrations in colorectal and female genital tract tissues revealed significantly varied ARV penetration in the field of HIV prophylaxis [139,140]. Even though these tissues are essential to the pathogenesis of HIV, there are a lot fewer data on medication exposure for the presumed lymph node reservoir [141]. Preclinical models including HIV-infected humanized mice and nonhuman primates (NHPs) with reverse transcriptase simian/HIV revealed that sex had no impact on the ARV pharmacokinetics in the collected lymph [142].

7. Why There Is the Need to Include Female Models

In general, a survey of the literature showed that preclinical research has mainly employed males (80%). The 44% of the papers on preclinical research models for medications created for diseases mostly affecting women did not mention the sex of the animals utilized; of those that did, 88% researched solely male animals [58,60]. The poor translation of animal findings to humans was therefore attributed to sex bias in preclinical investigations [7]. Although regulatory toxicity studies must use both the male and female sexes (perhaps as compensation?) prior to the first in-human studies, the scientist is required to “consider” sex when designing safety pharmacology studies that look at the short-term side effects on physiological functions. However, animal models have other drawbacks besides sex bias.
There are other factors than the X and Y chromosomes that distinguish females from males. There has been a prevalent, false notion that male and female rodents have similar characteristics in preclinical investigations [124]. Males and females differ in a variety of physiological phenotypes, including basic physiological traits such as body weight, lean and fat mass, as well as several neuroendocrine, immunological, and behavioral traits outside of reproductive behaviors [84,97]. Additionally, many human diseases have different effects on men and women, and sex differences can have an impact on treatment effectiveness, symptom progression, and disease susceptibility. Cardiovascular illness, autoimmune disorders, chronic pain, and neuropsychiatric disorders have well-established sex disparities, with females typically experiencing higher incidences than males [65]. A study design, phenotypes, pharmacokinetics, pharmacodynamic measurements, or the interpretation of results without considering sex as a covariate are all examples of areas where accuracy in reporting the data is lacking [8,9,10,15,16,20,23,26,27,37,41,48,74,86]. Sex distinctions in disease onset and development have also been noted in relation to animal models used in pharmacological investigations. Furthermore, it should be useful to consider sex distinctions in disaggregated data by age. For humans, especially for women, different periods of life can influence health outcome and treatment response. A preclinical research model that is truly transferable to human reality should also consider the age variable [143,144,145].

8. What Risks May Arise from Continuing Female Exclusion from Experimental Design

There have been ups and downs in the sex/gender debate throughout history, such as when the FDA banned women who may become pregnant from participating in phase 1 and phase 2 trials in response to the thalidomide (Contergan) and diethylstilbestrol scares in 1977. After studies revealed that women were underrepresented in clinical studies, revisions to these recommendations were made in 1993. From 1997 to 2000, eight out of ten prescription drugs were then taken off the market because they posed a greater health risk to women [146,147].
Sex is a crucial biological factor that has significant effects. Preclinical research on females has underrepresented female cells and animals, which led to a worse understanding of the pathophysiological, physiological, and biochemical pathways in females than in males. It is impossible to know whether the results acquired in male cells and animals also apply to female cells and animals without data from females [5,7,148,149]. The pipeline for developing new therapies and finding new molecules is provided by basic science. The application of these preclinical discoveries to the health of men and women is therefore impacted by the discovery of sex differences in pathophysiologic pathways in animal models of disease [150].
The interpretation of sex differences in scientific results presents difficulties, and the field is burdened by inconsistent results. The results could be affected by factors such as rat age and strain; in addition, the data could be susceptible to things such as the time of day that the data were collected and methodological variations between laboratories [149,151,152]. Estrus cycle determination and its appraisal in result interpretation are crucial for furthering the investigation of sex differences with the involvement of a male comparison group. Additionally, the continued use of studies on hormonal and neutering treatments adds to the expanding understanding and appreciation of the significance of sex differences in pharmacology. However, the exclusion of female animals from preclinical studies may lead to the waste of resources on treatments that inevitably do not receive approval. Increased sample sizes will be needed in studies that include females, but the effect of sex on pathological processes and treatment responses shows that differences between the sexes are crucial factors to consider in preclinical research.

9. What about Transgender Models?

If preclinical research still hesitates to enroll the female model, it is almost absent in terms of the study of diseases and drug response in the context of the transgender model. Yet, data from the international scientific literature suggest that the percentage of the transgender population is between 0.5 and 1.2% of the total population. In Italy, for example, the transgender population consists of 400,000 people [153]. There must therefore be a growing interest of the scientific world regarding the health of transgender people. Especially in recent years, several works have been published on this segment of the population, but the small number of subjects studied does not allow us to reach certain conclusions regarding the susceptibility and risk factors for chronic–degenerative diseases not allowing a specific health plan in these population groups. Preclinical research must also adapt to this as soon as possible, starting to propose an experimental transgender model in which to test when a mixed pool of hormones can influence the course of diseases and responses to therapy. The scientific literature related to preclinical research in this field is currently scarce.
While transgender individuals frequently receive gender-affirming/confirming hormone therapy, no hormonal agents or clinical protocols for transgender medicine have yet received approval from international medical regulatory agencies, such as the European Medicines Agency or the Food and Drug Administration. “Off-label” hormone therapies are thus based on recommendations from Endocrine Societies or organizations of a similar nature [154]. The primary goals of human hormone therapies, which are often given throughout life, are to reduce secondary sex traits and return sex hormone levels to the normal range for the matching cisgender. For transgender men, hormone therapy entails injecting testosterone (intramuscularly or subcutaneously); more recently, transdermal administration (patches or gel) of a longer acting form has been suggested because it is appropriate for long-term use. For transgender women, hormone therapy typically consists of β-estradiol that is given transdermally, orally, or intravenously, either on its own or in combination with medications that suppress androgen levels. Depending on how β-estradiol is administered, different dosage levels and administration windows apply. Cyproterone acetate (50 mg daily) is the most widely used anti-androgen medication in Europe [155]. There is a lack of information currently available regarding the potential impacts of HT on the health of transgender individuals as well as its potential long-term consequences. Since one of the main objectives of risk assessment is to characterize the chemical risks in potentially sensitive sub-population groups and to ensure the selection of appropriate safety factors [155], toxicological issues should also be included in this context. Endocrine disruptors, which share targets and modes of action with hormone therapy, are among the environmental and food pollutants that transgender people, like the general population, are exposed to on a regular basis, making transgender people a subgroup of the population that is more susceptible and vulnerable to their effects. To accurately perform a danger identification for transgender people undergoing hormone therapy in this context, specialized animal models should be established and employed [155].
Targeted animal models must be used in toxicological investigations in order to gather reliable information for identifying chemical hazards [156]. People with TG who underwent HT demonstrated distinctive characteristics in terms of particular susceptibility and vulnerability to chemical contaminants; as a result, they require appropriate animal models based on pertinent and novel biomarkers [157].
A significant model for determining whether the sex variations in the phenotypes are brought on by the complement of the sex chromosomes (XX vs. XY), gonadal hormones, or both, is the “four core genotypes” (FCG) mouse model. In the model, a Sry transgene is inserted onto an autosome after the testis-determining gene Sry is deleted from the Y chromosome. In order to compare XX and XY mice with the same type of gonad and determine the phenotypic consequences of sex chromosomal complement in cells and tissues, it breeds XX and XY mice with testes and XX and XY mice with ovaries [158,159,160,161,162,163]. The number of X chromosomes (including X dose, X imprint, or indirect effects of X inactivation), the presence or absence of the Y chromosome, or both, may be the cause of a sex chromosome effect (XX not equal to XY) in FCG mice [159,164]. So, to distinguish between these options, the XY* model is helpful. In XY* mice, which were first discovered by Eicher et al. a defective pseudo-autosomal region on the Y chromosome recombines improperly with the X chromosome [160,162]. When XX females and XY* fathers are mated, mice comparable to XX and XO gonadal females, as well as XY and XXY gonadal males, are produced [159]. When comparing XO vs. XX females or XY vs. XXY males, one can determine the impact of having one X chromosome against two. Comparisons of XY vs. XO and XXY vs. XX are used to assess the impact of having one Y chromosome vs. none. Mice with a Y chromosome are gonadal males according to the XY* paradigm.
A very recent study evaluated the ovarian dynamics after testosterone cessation using a mouse model that mimicked trans-masculine testosterone therapy. During six weeks, they administered testosterone enanthate or a vehicle control injection of 0.9 mg once per week to post-pubertal female C57BL/6N mice 9–10 weeks of age. Within 1 week of beginning the testosterone treatment, all the testosterone-treated mice ceased cycling and displayed chronic diestrum, whereas the control mice cycled regularly. The age-matched vehicle-treated diestrum controls and a group of mice treated with testosterone for 6 weeks each were slaughtered. The age-matched vehicle-treated controls and a different group of mice that had been receiving testosterone therapy were sacrificed together in diestrum four cycles after the cycle had resumed. Comparing the post-testosterone group to both the age-matched controls and the mice at 6 weeks on testosterone demonstrated stromal alterations with clusters of big round cells. Periodic acid-Schiff staining, which was documented in multinucleated macrophages in aging mouse ovaries, was present in these clusters. A significant number of these cells also showed staining for the macrophage markers CD68 and CD11b. Comparing the age-matched controls and ovaries at 6 weeks on testosterone, the ovarian ribonucleic acid-sequencing revealed that the immunological pathways were upregulated post-testosterone [165].

10. Take Home Message

The inclusion of both the male and female sexes is a valid approach to improve heterogeneity [149]. If researchers fail to disclose the sex utilized, or if they let sex be an uncontrolled variable throughout the experiment, or if they omit to account for sex during the study and sex is a substantial source of variance, sex bias may contribute to difficulties in replication.
Female cells function differently from male cells, which affects how sensitive they are to stressors and how susceptible they are to disease. This has an impact on how they respond to medication. Therefore, research on gender disparities and cell injury mechanisms seems essential for creating novel, more effective therapeutic approaches.
The capacity of scientists to properly consider these variations in diseases and patient populations is essential to the success of drug development. This subsequently enables us to create new pharmaceuticals by starting a symphony of preclinical research and safety studies customized precisely to the features of the new drug, and to wisely recognize where sex-specific considerations are required.

11. Conclusions

The knowledge of sex influence and differences in pharmacological studies should be taken into account in terms of treatment personalization and precision medicine.
Despite the fact that many diseases are known to display sexual dimorphism, the majority of preclinical research studies do not consider sex. Sex-based statistical analyses are still rare in preclinical investigations. In addition, despite the well-known sexual dimorphism, women are frequently underrepresented in various therapeutic trials. A delay in the diagnosis and treatment for both the male and female sexes may also result from societal representations of certain pathologies as “male” or “female” diseases, which are sensitive to social representations of certain pathologies. Finally, there may be sex differences in the efficacy and safety of numerous pharmacological classes. For instance, as a result of differing pharmacokinetic characteristics, women present more adverse effects than men. To improve the translation of observed results and advance customized therapy, sex must be taken into account as a variable starting in the preclinical stage [166].
Further works which aim to highlight sex differences in specific areas/diseases are needed to deeply explore different therapeutic strategies.

Author Contributions

Conceptualization, S.A. and S.D.F.; data curation, F.C., D.D.G. and M.G.; writing—original draft preparation, S.A.; writing—review and editing, F.C. and S.D.F.; supervision, S.D.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

Sarah Allegra was supported by Fondazione Umberto Veronesi.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tamimi, N.A.; Ellis, P. Drug development: From concept to marketing! Nephron Clin. Pract. 2009, 113, c125–c131. [Google Scholar] [CrossRef] [PubMed]
  2. Polson, A.G.; Fuji, R.N. The successes and limitations of preclinical studies in predicting the pharmacodynamics and safety of cell-surface-targeted biological agents in patients. Br. J. Pharmacol. 2012, 166, 1600–1602. [Google Scholar] [CrossRef] [PubMed]
  3. Clayton, J.A.; Collins, F.S. Policy: NIH to balance sex in cell and animal studies. Nature 2014, 509, 282–283. [Google Scholar] [CrossRef] [PubMed]
  4. Johnson, J.; Sharman, Z.; Vissandjée, B.; Stewart, D.E. Does a change in health research funding policy related to the integration of sex and gender have an impact? PLoS ONE 2014, 9, e99900. [Google Scholar] [CrossRef]
  5. Kim, J.Y.; Min, K.; Paik, H.Y.; Lee, S.K. Sex omission and male bias are still widespread in cell experiments. Am. J. Physiol. Cell Physiol. 2021, 320, C742–C749. [Google Scholar] [CrossRef]
  6. Haverfield, J.; Tannenbaum, C. A 10-year longitudinal evaluation of science policy interventions to promote sex and gender in health research. Health Res. Policy Syst. 2021, 19, 94. [Google Scholar] [CrossRef]
  7. Karp, N.A.; Reavey, N. Sex bias in preclinical research and an exploration of how to change the status quo. Br. J. Pharmacol. 2019, 176, 4107–4118. [Google Scholar] [CrossRef]
  8. Docherty, J.R.; Stanford, S.C.; Panattieri, R.A.; Alexander, S.P.H.; Cirino, G.; George, C.H.; Hoyer, D.; Izzo, A.A.; Ji, Y.; Lilley, E.; et al. Sex: A change in our guidelines to authors to ensure that this is no longer an ignored experimental variable. Br. J. Pharmacol. 2019, 176, 4081–4086. [Google Scholar] [CrossRef]
  9. Gogos, A.; Langmead, C.; Sullivan, J.C.; Lawrence, A.J. The importance of sex differences in pharmacology research. Br. J. Pharmacol. 2019, 176, 4087–4089. [Google Scholar] [CrossRef]
  10. Beierle, I.; Meibohm, B.; Derendorf, H. Gender differences in pharmacokinetics and pharmacodynamics. Int. J. Clin. Pharmacol. Ther. 1999, 37, 529–547. [Google Scholar]
  11. Bies, R.R.; Bigos, K.L.; Pollock, B.G. Gender differences in the pharmacokinetics and pharmacodynamics of antidepressants. J. Gend. Specif. Med. 2003, 6, 12–20. [Google Scholar] [PubMed]
  12. Bigos, K.L.; Pollock, B.G.; Stankevich, B.A.; Bies, R.R. Sex differences in the pharmacokinetics and pharmacodynamics of antidepressants: An updated review. Gend. Med. 2009, 6, 522–543. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, M.L. Confounding factors for sex differences in pharmacokinetics and pharmacodynamics: Focus on dosing regimen, dosage form, and formulation. Clin. Pharmacol. Ther. 2005, 78, 322–329. [Google Scholar] [CrossRef] [PubMed]
  14. Dawkins, K.; Potter, W.Z. Gender differences in pharmacokinetics and pharmacodynamics of psychotropics: Focus on women. Psychopharmacol. Bull. 1991, 27, 417–426. [Google Scholar] [PubMed]
  15. Dawkins, K.; Rudorfer, M.V.; Potter, W.Z. Comments on gender differences in pharmacokinetics and pharmacodynamics. Am. J. Psychiatry 1993, 150, 678–679. [Google Scholar] [CrossRef]
  16. Gandhi, M.; Aweeka, F.; Greenblatt, R.M.; Blaschke, T.F. Sex differences in pharmacokinetics and pharmacodynamics. Annu. Rev. Pharmacol. Toxicol. 2004, 44, 499–523. [Google Scholar] [CrossRef]
  17. Marazziti, D.; Baroni, S.; Picchetti, M.; Piccinni, A.; Carlini, M.; Vatteroni, E.; Falaschi, V.; Lombardi, A.; Dell’Osso, L. Pharmacokinetics and pharmacodynamics of psychotropic drugs: Effect of sex. CNS Spectr. 2013, 18, 118–127. [Google Scholar] [CrossRef]
  18. Regitz-Zagrosek, V. Sex and gender differences in pharmacotherapy. Bundesgesundh. Gesundh. Gesundh. 2014, 57, 1067–1073. [Google Scholar] [CrossRef]
  19. Zucker, I.; Prendergast, B.J. Sex differences in pharmacokinetics predict adverse drug reactions in women. Biol. Sex Differ. 2020, 11, 32. [Google Scholar] [CrossRef]
  20. Fletcher, C.V.; Acosta, E.P.; Strykowski, J.M. Gender differences in human pharmacokinetics and pharmacodynamics. J. Adolesc. Health 1994, 15, 619–629. [Google Scholar] [CrossRef]
  21. Franconi, F.; Campesi, I. Sex Impact on Biomarkers, Pharmacokinetics and Pharmacodynamics. Curr. Med. Chem. 2017, 24, 2561–2575. [Google Scholar] [CrossRef] [PubMed]
  22. Frost, C.E.; Song, Y.; Shenker, A.; Wang, J.; Barrett, Y.C.; Schuster, A.; Harris, S.I.; LaCreta, F. Effects of age and sex on the single-dose pharmacokinetics and pharmacodynamics of apixaban. Clin. Pharmacokinet. 2015, 54, 651–662. [Google Scholar] [CrossRef] [PubMed]
  23. Anderson, G.D. Gender differences in pharmacological response. Int. Rev. Neurobiol. 2008, 83, 1–10. [Google Scholar] [CrossRef]
  24. Anthony, M.; Berg, M.J. Biologic and molecular mechanisms for sex differences in pharmacokinetics, pharmacodynamics, and pharmacogenetics: Part II. J. Womens Health Gend. Based Med. 2002, 11, 617–629. [Google Scholar] [CrossRef] [PubMed]
  25. Flores Pérez, J.; Juárez Olguín, H.; Flores Pérez, C.; Pérez Guillé, G.; Guillé Pérez, A.; Camacho Vieyra, A.; Toledo López, A.; Carrasco Portugal, M.; Lares Asseff, I. Effects of gender and phase of the menstrual cycle on the kinetics of ranitidine in healthy volunteers. Chronobiol. Int. 2003, 20, 485–494. [Google Scholar]
  26. Franconi, F.; Brunelleschi, S.; Steardo, L.; Cuomo, V. Gender differences in drug responses. Pharmacol. Res. 2007, 55, 81–95. [Google Scholar] [CrossRef] [PubMed]
  27. Franconi, F.; Carru, C.; Spoletini, I.; Malorni, W.; Vella, S.; Mercuro, G.; Deidda, M.; Rosano, G. A GENS-based approach to cardiovascular pharmacology: Impact on metabolism, pharmacokinetics and pharmacodynamics. Ther. Deliv. 2011, 2, 1437–1453. [Google Scholar] [CrossRef]
  28. Greenblatt, D.J.; Harmatz, J.S.; von Moltke, L.L.; Wright, C.E.; Shader, R.I. Age and gender effects on the pharmacokinetics and pharmacodynamics of triazolam, a cytochrome P450 3A substrate. Clin. Pharmacol. Ther. 2004, 76, 467–479. [Google Scholar] [CrossRef]
  29. Greenblatt, D.J.; Harmatz, J.S.; Singh, N.N.; Steinberg, F.; Roth, T.; Moline, M.L.; Harris, S.C.; Kapil, R.P. Gender differences in pharmacokinetics and pharmacodynamics of zolpidem following sublingual administration. J. Clin. Pharmacol. 2014, 54, 282–290. [Google Scholar] [CrossRef]
  30. Gupta, S.K.; Atkinson, L.; Tu, T.; Longstreth, J.A. Age and gender related changes in stereoselective pharmacokinetics and pharmacodynamics of verapamil and norverapamil. Br. J. Clin. Pharmacol. 1995, 40, 325–331. [Google Scholar] [CrossRef]
  31. Harris, R.Z.; Benet, L.Z.; Schwartz, J.B. Gender effects in pharmacokinetics and pharmacodynamics. Drugs 1995, 50, 222–239. [Google Scholar] [CrossRef] [PubMed]
  32. Jiang, X.L.; Samant, S.; Lesko, L.J.; Schmidt, S. Clinical pharmacokinetics and pharmacodynamics of clopidogrel. Clin. Pharmacokinet. 2015, 54, 147–166. [Google Scholar] [CrossRef] [PubMed]
  33. Krecic-Shepard, M.E.; Barnas, C.R.; Slimko, J.; Jones, M.P.; Schwartz, J.B. Gender-specific effects on verapamil pharmacokinetics and pharmacodynamics in humans. J. Clin. Pharmacol. 2000, 40, 219–230. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, Y.S.; Han, K.S.; Lee, M.G. Gender differences in pharmacokinetics and pharmacodynamics of azosemide in rats. Biopharm. Drug Dispos. 1999, 20, 225–229. [Google Scholar] [CrossRef]
  35. Luzier, A.B.; Killian, A.; Wilton, J.H.; Wilson, M.F.; Forrest, A.; Kazierad, D.J. Gender-related effects on metoprolol pharmacokinetics and pharmacodynamics in healthy volunteers. Clin. Pharmacol. Ther. 1999, 66, 594–601. [Google Scholar] [CrossRef]
  36. Patrick, K.S.; Straughn, A.B.; Minhinnett, R.R.; Yeatts, S.D.; Herrin, A.E.; DeVane, C.L.; Malcolm, R.; Janis, G.C.; Markowitz, J.S. Influence of ethanol and gender on methylphenidate pharmacokinetics and pharmacodynamics. Clin. Pharmacol. Ther. 2007, 81, 346–353. [Google Scholar] [CrossRef]
  37. Soldin, O.P.; Mattison, D.R. Sex differences in pharmacokinetics and pharmacodynamics. Clin. Pharmacokinet. 2009, 48, 143–157. [Google Scholar] [CrossRef]
  38. Song, D.; DuBois, D.C.; Almon, R.R.; Jusko, W.J. Modeling Sex Differences in Anti-inflammatory Effects of Dexamethasone in Arthritic Rats. Pharm. Res. 2018, 35, 203. [Google Scholar] [CrossRef]
  39. Swan, S.K.; Hursting, M.J. The pharmacokinetics and pharmacodynamics of argatroban: Effects of age, gender, and hepatic or renal dysfunction. Pharmacotherapy 2000, 20, 318–329. [Google Scholar] [CrossRef]
  40. Tamargo, J.; Rosano, G.; Walther, T.; Duarte, J.; Niessner, A.; Kaski, J.C.; Ceconi, C.; Drexel, H.; Kjeldsen, K.; Savarese, G.; et al. Gender differences in the effects of cardiovascular drugs. Eur. Heart J. Cardiovasc. Pharmacother. 2017, 3, 163–182. [Google Scholar] [CrossRef]
  41. Thürmann, P.A.; Hompesch, B.C. Influence of gender on the pharmacokinetics and pharmacodynamics of drugs. Int. J. Clin. Pharmacol. Ther. 1998, 36, 586–590. [Google Scholar] [PubMed]
  42. Ueno, K.; Sato, H. Sex-related differences in pharmacokinetics and pharmacodynamics of anti-hypertensive drugs. Hypertens. Res. 2012, 35, 245–250. [Google Scholar] [CrossRef] [PubMed]
  43. Vachharajani, N.N.; Raymond, R.H.; Shyu, W.C.; Stouffer, B.C.; Boulton, D.W. The effects of age and gender on the pharmacokinetics and pharmacodynamics in healthy subjects of the plasminogen activator, lanoteplase. Br. J. Clin. Pharmacol. 2011, 72, 775–786. [Google Scholar] [CrossRef] [PubMed]
  44. Yonkers, K.A.; Kando, J.C.; Cole, J.O.; Blumenthal, S. Gender differences in pharmacokinetics and pharmacodynamics of psychotropic medication. Am. J. Psychiatry 1992, 149, 587–595. [Google Scholar] [CrossRef]
  45. Yoon, S.; Jeong, S.; Jung, E.; Kim, K.S.; Jeon, I.; Lee, Y.; Cho, J.Y.; Oh, W.Y.; Chung, J.Y. Effect of CYP3A4 metabolism on sex differences in the pharmacokinetics and pharmacodynamics of zolpidem. Sci. Rep. 2021, 11, 19150. [Google Scholar] [CrossRef] [PubMed]
  46. Franconi, F.; Carru, C.; Malorni, W.; Vella, S.; Mercuro, G. The effect of sex/gender on cardiovascular pharmacology. Curr. Pharm. Des. 2011, 17, 1095–1107. [Google Scholar] [CrossRef]
  47. Anderson, G.D. Pregnancy-induced changes in pharmacokinetics: A mechanistic-based approach. Clin. Pharmacokinet. 2005, 44, 989–1008. [Google Scholar] [CrossRef]
  48. Spoletini, I.; Vitale, C.; Malorni, W.; Rosano, G.M. Sex differences in drug effects: Interaction with sex hormones in adult life. In Sex and Gender Differences in Pharmacology; Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2012; Volume 2014, pp. 91–105. [Google Scholar] [CrossRef]
  49. Moyer, A.M.; Matey, E.T.; Miller, V.M. Individualized medicine: Sex, hormones, genetics, and adverse drug reactions. Pharmacol. Res. Perspect. 2019, 7, e00541. [Google Scholar] [CrossRef]
  50. Mitchell, S.C.; Smith, R.L.; Waring, R.H. The menstrual cycle and drug metabolism. Curr. Drug Metab. 2009, 10, 499–507. [Google Scholar] [CrossRef]
  51. Figueiredo, M.G.; Gagliano-Jucá, T.; Basaria, S. Male Reproduction and Aging. Endocrinol. Metab. Clin. N. Am. 2023, 52, 211–228. [Google Scholar] [CrossRef]
  52. Wu, F.C.; Tajar, A.; Pye, S.R.; Silman, A.J.; Finn, J.D.; O’Neill, T.W.; Bartfai, G.; Casanueva, F.; Forti, G.; Giwercman, A.; et al. Hypothalamic-pituitary-testicular axis disruptions in older men are differentially linked to age and modifiable risk factors: The European Male Aging Study. J. Clin. Endocrinol. Metab. 2008, 93, 2737–2745. [Google Scholar] [CrossRef] [PubMed]
  53. Bawor, M.; Bami, H.; Dennis, B.B.; Plater, C.; Worster, A.; Varenbut, M.; Daiter, J.; Marsh, D.C.; Steiner, M.; Anglin, R.; et al. Testosterone suppression in opioid users: A systematic review and meta-analysis. Drug Alcohol Depend. 2015, 149, 1–9. [Google Scholar] [CrossRef] [PubMed]
  54. de Vries, F.; Bruin, M.; Lobatto, D.J.; Dekkers, O.M.; Schoones, J.W.; van Furth, W.R.; Pereira, A.M.; Karavitaki, N.; Biermasz, N.R.; Zamanipoor Najafabadi, A.H. Opioids and Their Endocrine Effects: A Systematic Review and Meta-analysis. J. Clin. Endocrinol. Metab. 2020, 105, 1020–1029. [Google Scholar] [CrossRef]
  55. Zhou, J.; Argikar, U.A.; Remmel, R.P. Functional analysis of UGT1A4(P24T) and UGT1A4(L48V) variant enzymes. Pharmacogenomics 2011, 12, 1671–1679. [Google Scholar] [CrossRef] [PubMed]
  56. Kharasch, E.D.; Mautz, D.; Senn, T.; Lentz, G.; Cox, K. Menstrual cycle variability in midazolam pharmacokinetics. J. Clin. Pharmacol. 1999, 39, 275–280. [Google Scholar] [CrossRef] [PubMed]
  57. Shah, A.K.; Laboy-Goral, L.; Scott, N.; Morse, T.; Apseloff, G. Pharmacokinetics and safety of oral eletriptan during different phases of the menstrual cycle in healthy volunteers. J. Clin. Pharmacol. 2001, 41, 1339–1344. [Google Scholar] [CrossRef]
  58. Beery, A.K.; Zucker, I. Sex bias in neuroscience and biomedical research. Neurosci. Biobehav. Rev. 2011, 35, 565–572. [Google Scholar] [CrossRef]
  59. Prendergast, B.J.; Onishi, K.G.; Zucker, I. Female mice liberated for inclusion in neuroscience and biomedical research. Neurosci. Biobehav. Rev. 2014, 40, 1–5. [Google Scholar] [CrossRef]
  60. Yoon, D.Y.; Mansukhani, N.A.; Stubbs, V.C.; Helenowski, I.B.; Woodruff, T.K.; Kibbe, M.R. Sex bias exists in basic science and translational surgical research. Surgery 2014, 156, 508–516. [Google Scholar] [CrossRef]
  61. Taylor, K.E.; Vallejo-Giraldo, C.; Schaible, N.S.; Zakeri, R.; Miller, V.M. Reporting of sex as a variable in cardiovascular studies using cultured cells. Biol. Sex Differ. 2011, 2, 11. [Google Scholar] [CrossRef]
  62. Vallabhajosyula, S.; Ponamgi, S.P.; Shrivastava, S.; Sundaragiri, P.R.; Miller, V.M. Reporting of sex as a variable in cardiovascular studies using cultured cells: A systematic review. FASEB J. 2020, 34, 8778–8786. [Google Scholar] [CrossRef] [PubMed]
  63. Maselli, A.; Matarrese, P.; Straface, E.; Canu, S.; Franconi, F.; Malorni, W. Cell sex: A new look at cell fate studies. FASEB J. 2009, 23, 978–984. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, T.Y.; Angiolillo, D.J.; Cushman, M.; Sabatine, M.S.; Bray, P.F.; Smyth, S.S.; Dauerman, H.L.; French, P.A.; Becker, R.C. Platelet biology and response to antiplatelet therapy in women: Implications for the development and use of antiplatelet pharmacotherapies for cardiovascular disease. J. Am. Coll. Cardiol. 2012, 59, 891–900. [Google Scholar] [CrossRef] [PubMed]
  65. Regitz-Zagrosek, V. Sex and gender differences in health. Science & Society Series on Sex and Science. EMBO Rep. 2012, 13, 596–603. [Google Scholar] [CrossRef] [PubMed]
  66. Legato, M.J. Gender-specific medicine in the genomic era. Clin. Sci. 2016, 130, 1–7. [Google Scholar] [CrossRef]
  67. Li, J.; Ma, Z.; Jiang, R.W.; Wu, B. Hormone-related pharmacokinetic variations associated with anti-breast cancer drugs. Expert Opin. Drug Metab. Toxicol. 2013, 9, 1085–1095. [Google Scholar] [CrossRef]
  68. Damoiseaux, V.A.; Proost, J.H.; Jiawan, V.C.; Melgert, B.N. Sex differences in the pharmacokinetics of antidepressants: Influence of female sex hormones and oral contraceptives. Clin. Pharmacokinet. 2014, 53, 509–519. [Google Scholar] [CrossRef]
  69. Romano, S.N.; Gorelick, D.A. Crosstalk between nuclear and G protein-coupled estrogen receptors. Gen. Comp. Endocrinol. 2018, 261, 190–197. [Google Scholar] [CrossRef]
  70. Hernandez, J.P.; Mota, L.C.; Huang, W.; Moore, D.D.; Baldwin, W.S. Sexually dimorphic regulation and induction of P450s by the constitutive androstane receptor (CAR). Toxicology 2009, 256, 53–64. [Google Scholar] [CrossRef]
  71. Nugent, B.M.; McCarthy, M.M. Epigenetic underpinnings of developmental sex differences in the brain. Neuroendocrinology 2011, 93, 150–158. [Google Scholar] [CrossRef]
  72. Trout, K.K.; Rickels, M.R.; Schutta, M.H.; Petrova, M.; Freeman, E.W.; Tkacs, N.C.; Teff, K.L. Menstrual cycle effects on insulin sensitivity in women with type 1 diabetes: A pilot study. Diabetes Technol. Ther. 2007, 9, 176–182. [Google Scholar] [CrossRef] [PubMed]
  73. Bilik, D.; McEwen, L.N.; Brown, M.B.; Pomeroy, N.E.; Kim, C.; Asao, K.; Crosson, J.C.; Duru, O.K.; Ferrara, A.; Hsiao, V.C.; et al. Thiazolidinediones and fractures: Evidence from translating research into action for diabetes. J. Clin. Endocrinol. Metab. 2010, 95, 4560–4565. [Google Scholar] [CrossRef] [PubMed]
  74. Franconi, F.; Raparelli, V.; Regitz-Zagrosek, V. Sex and gender landscape in pharmacology. Pharmacol. Res. 2017, 123, 93–94. [Google Scholar] [CrossRef] [PubMed]
  75. Cvitanović Tomaš, T.; Urlep, Ž.; Moškon, M.; Mraz, M.; Rozman, D. Computational Model: Sexual Aspects in Hepatic Metabolism and Abnormalities. Front. Physiol. 2018, 9, 360. [Google Scholar] [CrossRef] [PubMed]
  76. Naik, A.; Rozman, D.; Belič, A. SteatoNet: The first integrated human metabolic model with multi-layered regulation to investigate liver-associated pathologies. PLoS Comput. Biol. 2014, 10, e1003993. [Google Scholar] [CrossRef]
  77. Thiele, I.; Sahoo, S.; Heinken, A.; Hertel, J.; Heirendt, L.; Aurich, M.K.; Fleming, R.M. Personalized whole-body models integrate metabolism, physiology, and the gut microbiome. Mol. Syst. Biol. 2020, 16, e8982. [Google Scholar] [CrossRef]
  78. Nokkala, S.; Grozeva, S.; Kuznetsova, V.; Maryanska-Nadachowska, A. The origin of the achiasmatic XY sex chromosome system in Cacopsylla peregrina (Frst.) (Psylloidea, Homoptera). Genetica 2003, 119, 327–332. [Google Scholar] [CrossRef]
  79. Nokkala, S.; Kuznetsova, V.; Maryańska-Nadachowska, A. Achiasmate segregation of a B chromosome from the X chromosome in two species of psyllids (Psylloidea, Homoptera). Genetica 2000, 108, 181–189. [Google Scholar] [CrossRef]
  80. Fraïsse, C.; Picard, M.A.L.; Vicoso, B. The deep conservation of the Lepidoptera Z chromosome suggests a non-canonical origin of the W. Nat. Commun. 2017, 8, 1486. [Google Scholar] [CrossRef]
  81. Leclercq, S.; Thézé, J.; Chebbi, M.A.; Giraud, I.; Moumen, B.; Ernenwein, L.; Grève, P.; Gilbert, C.; Cordaux, R. Birth of a W sex chromosome by horizontal transfer of Wolbachia bacterial symbiont genome. Proc. Natl. Acad. Sci. USA 2016, 113, 15036–15041. [Google Scholar] [CrossRef]
  82. Lyon, M.F. Gene action in the X-chromosome of the mouse (Mus musculus L.). Nature 1961, 190, 372–373. [Google Scholar] [CrossRef]
  83. Furman, B.L.S.; Metzger, D.C.H.; Darolti, I.; Wright, A.E.; Sandkam, B.A.; Almeida, P.; Shu, J.J.; Mank, J.E. Sex Chromosome Evolution: So Many Exceptions to the Rules. Genome Biol. Evol. 2020, 12, 750–763. [Google Scholar] [CrossRef] [PubMed]
  84. Karp, N.A.; Mason, J.; Beaudet, A.L.; Benjamini, Y.; Bower, L.; Braun, R.E.; Brown, S.D.M.; Chesler, E.J.; Dickinson, M.E.; Flenniken, A.M.; et al. Prevalence of sexual dimorphism in mammalian phenotypic traits. Nat. Commun. 2017, 8, 15475. [Google Scholar] [CrossRef] [PubMed]
  85. Wolbold, R.; Klein, K.; Burk, O.; Nüssler, A.K.; Neuhaus, P.; Eichelbaum, M.; Schwab, M.; Zanger, U.M. Sex is a major determinant of CYP3A4 expression in human liver. Hepatology 2003, 38, 978–988. [Google Scholar] [CrossRef] [PubMed]
  86. Hunt, C.M.; Westerkam, W.R.; Stave, G.M. Effect of age and gender on the activity of human hepatic CYP3A. Biochem. Pharmacol. 1992, 44, 275–283. [Google Scholar] [CrossRef]
  87. Clodfelter, K.H.; Holloway, M.G.; Hodor, P.; Park, S.H.; Ray, W.J.; Waxman, D.J. Sex-dependent liver gene expression is extensive and largely dependent upon signal transducer and activator of transcription 5b (STAT5b): STAT5b-dependent activation of male genes and repression of female genes revealed by microarray analysis. Mol. Endocrinol. 2006, 20, 1333–1351. [Google Scholar] [CrossRef]
  88. Clodfelter, K.H.; Miles, G.D.; Wauthier, V.; Holloway, M.G.; Zhang, X.; Hodor, P.; Ray, W.J.; Waxman, D.J. Role of STAT5a in regulation of sex-specific gene expression in female but not male mouse liver revealed by microarray analysis. Physiol. Genom. 2007, 31, 63–74. [Google Scholar] [CrossRef]
  89. Yang, X.; Schadt, E.E.; Wang, S.; Wang, H.; Arnold, A.P.; Ingram-Drake, L.; Drake, T.A.; Lusis, A.J. Tissue-specific expression and regulation of sexually dimorphic genes in mice. Genome Res. 2006, 16, 995–1004. [Google Scholar] [CrossRef]
  90. Robertson, J.A.; Haldosén, L.A.; Wood, T.J.; Steed, M.K.; Gustafsson, J.A. Growth hormone pretranslationally regulates the sexually dimorphic expression of the prolactin receptor gene in rat liver. Mol. Endocrinol. 1990, 4, 1235–1239. [Google Scholar] [CrossRef]
  91. Wauthier, V.; Waxman, D.J. Sex-specific early growth hormone response genes in rat liver. Mol. Endocrinol. 2008, 22, 1962–1974. [Google Scholar] [CrossRef]
  92. Shapiro, B.H.; Agrawal, A.K.; Pampori, N.A. Gender differences in drug metabolism regulated by growth hormone. Int. J. Biochem. Cell Biol. 1995, 27, 9–20. [Google Scholar] [CrossRef] [PubMed]
  93. Hersman, E.M.; Bumpus, N.N. A targeted proteomics approach for profiling murine cytochrome P450 expression. J. Pharmacol. Exp. Ther. 2014, 349, 221–228. [Google Scholar] [CrossRef] [PubMed]
  94. Emanuele, M.A.; Wezeman, F.; Emanuele, N.V. Alcohol’s effects on female reproductive function. Alcohol Res. Health 2002, 26, 274–281. [Google Scholar] [PubMed]
  95. Becker, J.B.; Cha, J.H. Estrous cycle-dependent variation in amphetamine-induced behaviors and striatal dopamine release assessed with microdialysis. Behav. Brain Res. 1989, 35, 117–125. [Google Scholar] [CrossRef] [PubMed]
  96. Hu, M.; Becker, J.B. Effects of sex and estrogen on behavioral sensitization to cocaine in rats. J. Neurosci. 2003, 23, 693–699. [Google Scholar] [CrossRef]
  97. Hughes, R.N. Sex does matter: Comments on the prevalence of male-only investigations of drug effects on rodent behaviour. Behav. Pharmacol. 2007, 18, 583–589. [Google Scholar] [CrossRef]
  98. Becker, J.B.; Molenda, H.; Hummer, D.L. Gender differences in the behavioral responses to cocaine and amphetamine. Implications for mechanisms mediating gender differences in drug abuse. Ann. N. Y. Acad. Sci. 2001, 937, 172–187. [Google Scholar] [CrossRef]
  99. Becker, J.B.; Hu, M. Sex differences in drug abuse. Front. Neuroendocrinol. 2008, 29, 36–47. [Google Scholar] [CrossRef]
  100. Berry, A.; Raggi, C.; Borgi, M.; Cirulli, F. Sex-driven vulnerability in stress and drug abuse. Ann. Ist. Super. Sanita 2016, 52, 167–175. [Google Scholar] [CrossRef]
  101. Anker, J.J.; Carroll, M.E. Females are more vulnerable to drug abuse than males: Evidence from preclinical studies and the role of ovarian hormones. In Biological Basis of Sex Differences in Psychopharmacology; Current Topics in Behavioral Neurosciences; Springer: Berlin/Heidelberg, Germany, 2011; Volume 8, pp. 73–96. [Google Scholar] [CrossRef]
  102. Jaster, A.M.; Younkin, J.; Cuddy, T.; de la Fuente Revenga, M.; Poklis, J.L.; Dozmorov, M.G.; González-Maeso, J. Differences across sexes on head-twitch behavior and 5-HT. Neurosci. Lett. 2022, 788, 136836. [Google Scholar] [CrossRef]
  103. Craft, R.M.; Marusich, J.A.; Wiley, J.L. Sex differences in cannabinoid pharmacology: A reflection of differences in the endocannabinoid system? Life Sci. 2013, 92, 476–481. [Google Scholar] [CrossRef] [PubMed]
  104. McGregor, I.S.; Arnold, J.C. Cannabis reward: Biased towards the fairer sex? Br. J. Pharmacol. 2007, 152, 562–564. [Google Scholar] [CrossRef] [PubMed]
  105. Cooper, Z.D.; Craft, R.M. Sex-Dependent Effects of Cannabis and Cannabinoids: A Translational Perspective. Neuropsychopharmacology 2018, 43, 34–51. [Google Scholar] [CrossRef] [PubMed]
  106. Wiley, J.L.; Lefever, T.W.; Marusich, J.A.; Craft, R.M. Comparison of the discriminative stimulus and response rate effects of. Drug Alcohol Depend. 2017, 172, 51–59. [Google Scholar] [CrossRef] [PubMed]
  107. Craft, R.M.; Kandasamy, R.; Davis, S.M. Sex differences in anti-allodynic, anti-hyperalgesic and anti-edema effects of Δ9-tetrahydrocannabinol in the rat. Pain 2013, 154, 1709–1717. [Google Scholar] [CrossRef]
  108. Winsauer, P.J.; Filipeanu, C.M.; Bailey, E.M.; Hulst, J.L.; Sutton, J.L. Ovarian hormones and chronic administration during adolescence modify the discriminative stimulus effects of delta-9-tetrahydrocannabinol (Δ9-THC) in adult female rats. Pharmacol. Biochem. Behav. 2012, 102, 442–449. [Google Scholar] [CrossRef]
  109. Wiley, J.L.; Matthew Walentiny, D.; Vann, R.E.; Baskfield, C.Y. Dissimilar cannabinoid substitution patterns in mice trained to discriminate Δ9-tetrahydrocannabinol or methanandamide from vehicle. Behav. Pharmacol. 2011, 22, 480–488. [Google Scholar] [CrossRef]
  110. Wiley, J.L.; Barrus, D.G.; Farquhar, C.E.; Lefever, T.W.; Gamage, T.F. Sex, species and age: Effects of rodent demographics on the pharmacology of ∆9-tetrahydrocanabinol. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 106, 110064. [Google Scholar] [CrossRef]
  111. Blake, D.R.; Robson, P.; Ho, M.; Jubb, R.W.; McCabe, C.S. Preliminary assessment of the efficacy, tolerability and safety of a cannabis-based medicine (Sativex) in the treatment of pain caused by rheumatoid arthritis. Rheumatology 2006, 45, 50–52. [Google Scholar] [CrossRef]
  112. Johnson, J.R.; Lossignol, D.; Burnell-Nugent, M.; Fallon, M.T. An open-label extension study to investigate the long-term safety and tolerability of THC/CBD oromucosal spray and oromucosal THC spray in patients with terminal cancer-related pain refractory to strong opioid analgesics. J. Pain Symptom Manag. 2013, 46, 207–218. [Google Scholar] [CrossRef]
  113. Nahin, R.L. Estimates of pain prevalence and severity in adults: United States, 2012. J. Pain 2015, 16, 769–780. [Google Scholar] [CrossRef]
  114. Riley, J.L.; Robinson, M.E.; Wise, E.A.; Myers, C.D.; Fillingim, R.B. Sex differences in the perception of noxious experimental stimuli: A meta-analysis. Pain 1998, 74, 181–187. [Google Scholar] [CrossRef] [PubMed]
  115. Aubrun, F.; Salvi, N.; Coriat, P.; Riou, B. Sex- and age-related differences in morphine requirements for postoperative pain relief. Anesthesiology 2005, 103, 156–160. [Google Scholar] [CrossRef]
  116. LaFleur, R.A.; Wilson, R.P.; Morgan, D.J.; Henderson-Redmond, A.N. Sex differences in antinociceptive response to Δ9-tetrahydrocannabinol and CP 55,940 in the mouse formalin test. Neuroreport 2018, 29, 447–452. [Google Scholar] [CrossRef] [PubMed]
  117. Kopruszinski, C.M.; Swiokla, J.; Lee, Y.S.; Navratilova, E.; VanderVeen, L.; Yang, M.; Liu, Y.; Miyazaki, T.; Schmidt, W.K.; Zalevsky, J.; et al. Preclinical Assessment of the Analgesic Pharmacology of NKTR-181 in Rodents. Cell Mol. Neurobiol. 2021, 41, 949–960. [Google Scholar] [CrossRef]
  118. Pacini, G.; Omar, B.; Ahrén, B. Methods and models for metabolic assessment in mice. J. Diabetes Res. 2013, 2013, 986906. [Google Scholar] [CrossRef] [PubMed]
  119. Nyavor, Y.; Estill, R.; Edwards, H.; Ogden, H.; Heideman, K.; Starks, K.; Miller, C.; May, G.; Flesch, L.; McMillan, J.; et al. Intestinal nerve cell injury occurs prior to insulin resistance in female mice ingesting a high-fat diet. Cell Tissue Res. 2019, 376, 325–340. [Google Scholar] [CrossRef]
  120. Kaikaew, K.; Steenbergen, J.; van Dijk, T.H.; Grefhorst, A.; Visser, J.A. Sex Difference in Corticosterone-Induced Insulin Resistance in Mice. Endocrinology 2019, 160, 2367–2387. [Google Scholar] [CrossRef]
  121. Pettersson, U.S.; Waldén, T.B.; Carlsson, P.O.; Jansson, L.; Phillipson, M. Female mice are protected against high-fat diet induced metabolic syndrome and increase the regulatory T cell population in adipose tissue. PLoS ONE 2012, 7, e46057. [Google Scholar] [CrossRef] [PubMed]
  122. Rebolledo-Solleiro, D.; Fernández-Guasti, A. Influence of sex and estrous cycle on blood glucose levels, body weight gain, and depressive-like behavior in streptozotocin-induced diabetic rats. Physiol. Behav. 2018, 194, 560–567. [Google Scholar] [CrossRef]
  123. Bartke, A.; Steele, R.E.; Musto, N.; Caldwell, B.V. Fluctuations in plasma testosterone levels in adult male rats and mice. Endocrinology 1973, 92, 1223–1228. [Google Scholar] [CrossRef]
  124. Beery, A.K. Inclusion of females does not increase variability in rodent research studies. Curr. Opin. Behav. Sci. 2018, 23, 143–149. [Google Scholar] [CrossRef] [PubMed]
  125. Kennard, M.R.; Nandi, M.; Chapple, S.; King, A.J. The glucose tolerance test in mice: Sex, drugs and protocol. Diabetes Obes. Metab. 2022, 24, 2241–2252. [Google Scholar] [CrossRef]
  126. Frias, J.P.; Bastyr, E.J.; Vignati, L.; Tschöp, M.H.; Schmitt, C.; Owen, K.; Christensen, R.H.; DiMarchi, R.D. The Sustained Effects of a Dual GIP/GLP-1 Receptor Agonist, NNC0090-2746, in Patients with Type 2 Diabetes. Cell Metab. 2017, 26, 343–352.e2. [Google Scholar] [CrossRef]
  127. Frias, J.P.; Nauck, M.A.; Van, J.; Kutner, M.E.; Cui, X.; Benson, C.; Urva, S.; Gimeno, R.E.; Milicevic, Z.; Robins, D.; et al. Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: A randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet 2018, 392, 2180–2193. [Google Scholar] [CrossRef] [PubMed]
  128. Finan, B.; Ma, T.; Ottaway, N.; Müller, T.D.; Habegger, K.M.; Heppner, K.M.; Kirchner, H.; Holland, J.; Hembree, J.; Raver, C.; et al. Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans. Sci. Transl. Med. 2013, 5, 209ra151. [Google Scholar] [CrossRef]
  129. Sachs, S.; Niu, L.; Geyer, P.; Jall, S.; Kleinert, M.; Feuchtinger, A.; Stemmer, K.; Brielmeier, M.; Finan, B.; DiMarchi, R.D.; et al. Plasma proteome profiles treatment efficacy of incretin dual agonism in diet-induced obese female and male mice. Diabetes Obes. Metab. 2021, 23, 195–207. [Google Scholar] [CrossRef] [PubMed]
  130. Kremer, J.J.; Bills, A.J.; Hanke, N.J.; Chen, H.; Meier, W.A.; Osinski, M.A.; Foley, C.M. Evaluation of cardiovascular changes in dogs administered three positive controls using jacketed external telemetry-blood pressure (JET-BP). J. Pharmacol. Toxicol. Methods 2015, 75, 27–37. [Google Scholar] [CrossRef]
  131. Bourdi, M.; Rudloff, U.; Patnaik, S.; Marugan, J.; Terse, P.S. Safety assessment of metarrestin in dogs: A clinical candidate targeting a subnuclear structure unique to metastatic cancer cells. Regul. Toxicol. Pharmacol. 2020, 116, 104716. [Google Scholar] [CrossRef]
  132. Ewertz, M.; Qvortrup, C.; Eckhoff, L. Chemotherapy-induced peripheral neuropathy in patients treated with taxanes and platinum derivatives. Acta Oncol. 2015, 54, 587–591. [Google Scholar] [CrossRef]
  133. Kotaka, M.; Saito, Y.; Kato, T.; Satake, H.; Makiyama, A.; Tsuji, Y.; Shinozaki, K.; Fujiwara, T.; Mizushima, T.; Harihara, Y.; et al. A placebo-controlled, double-blind, randomized study of recombinant thrombomodulin (ART-123) to prevent oxaliplatin-induced peripheral neuropathy. Cancer Chemother. Pharmacol. 2020, 86, 607–618. [Google Scholar] [CrossRef] [PubMed]
  134. Minami, T.; Takeda, M.; Sata, M.; Kato, H.; Yano, K.; Sakai, T.; Tsujita, R.; Kawasaki, K.; Ito, A. Thrombomodulin alfa prevents oxaliplatin-induced neuropathic symptoms through activation of thrombin-activatable fibrinolysis inhibitor and protein C without affecting anti-tumor activity. Eur. J. Pharmacol. 2020, 880, 173196. [Google Scholar] [CrossRef] [PubMed]
  135. Earp, J.C.; Dubois, D.C.; Almon, R.R.; Jusko, W.J. Quantitative dynamic models of arthritis progression in the rat. Pharm. Res. 2009, 26, 196–203. [Google Scholar] [CrossRef]
  136. Earp, J.C.; Dubois, D.C.; Molano, D.S.; Pyszczynski, N.A.; Keller, C.E.; Almon, R.R.; Jusko, W.J. Modeling corticosteroid effects in a rat model of rheumatoid arthritis I: Mechanistic disease progression model for the time course of collagen-induced arthritis in Lewis rats. J. Pharmacol. Exp. Ther. 2008, 326, 532–545. [Google Scholar] [CrossRef]
  137. Earp, J.C.; Dubois, D.C.; Molano, D.S.; Pyszczynski, N.A.; Almon, R.R.; Jusko, W.J. Modeling corticosteroid effects in a rat model of rheumatoid arthritis II: Mechanistic pharmacodynamic model for dexamethasone effects in Lewis rats with collagen-induced arthritis. J. Pharmacol. Exp. Ther. 2008, 326, 546–554. [Google Scholar] [CrossRef]
  138. Fletcher, C.V.; Staskus, K.; Wietgrefe, S.W.; Rothenberger, M.; Reilly, C.; Chipman, J.G.; Beilman, G.J.; Khoruts, A.; Thorkelson, A.; Schmidt, T.E.; et al. Persistent HIV-1 replication is associated with lower antiretroviral drug concentrations in lymphatic tissues. Proc. Natl. Acad. Sci. USA 2014, 111, 2307–2312. [Google Scholar] [CrossRef]
  139. Thompson, C.G.; Bokhart, M.T.; Sykes, C.; Adamson, L.; Fedoriw, Y.; Luciw, P.A.; Muddiman, D.C.; Kashuba, A.D.; Rosen, E.P. Mass spectrometry imaging reveals heterogeneous efavirenz distribution within putative HIV reservoirs. Antimicrob. Agents Chemother. 2015, 59, 2944–2948. [Google Scholar] [CrossRef] [PubMed]
  140. Cottrell, M.L.; Yang, K.H.; Prince, H.M.; Sykes, C.; White, N.; Malone, S.; Dellon, E.S.; Madanick, R.D.; Shaheen, N.J.; Hudgens, M.G.; et al. A Translational Pharmacology Approach to Predicting Outcomes of Preexposure Prophylaxis Against HIV in Men and Women Using Tenofovir Disoproxil Fumarate With or Without Emtricitabine. J. Infect. Dis. 2016, 214, 55–64. [Google Scholar] [CrossRef]
  141. Dimopoulos, Y.; Moysi, E.; Petrovas, C. The Lymph Node in HIV Pathogenesis. Curr. HIV/AIDS Rep. 2017, 14, 133–140. [Google Scholar] [CrossRef]
  142. Burgunder, E.; Fallon, J.K.; White, N.; Schauer, A.P.; Sykes, C.; Remling-Mulder, L.; Kovarova, M.; Adamson, L.; Luciw, P.; Garcia, J.V.; et al. Antiretroviral Drug Concentrations in Lymph Nodes: A Cross-Species Comparison of the Effect of Drug Transporter Expression, Viral Infection, and Sex in Humanized Mice, Nonhuman Primates, and Humans. J. Pharmacol. Exp. Ther. 2019, 370, 360–368. [Google Scholar] [CrossRef]
  143. Tannenbaum, C.; Day, D.; Alliance, M. Age and sex in drug development and testing for adults. Pharmacol. Res. 2017, 121, 83–93. [Google Scholar] [CrossRef] [PubMed]
  144. Jackson, S.J.; Andrews, N.; Ball, D.; Bellantuono, I.; Gray, J.; Hachoumi, L.; Holmes, A.; Latcham, J.; Petrie, A.; Potter, P.; et al. Does age matter? The impact of rodent age on study outcomes. Lab. Anim. 2017, 51, 160–169. [Google Scholar] [CrossRef] [PubMed]
  145. Sukoff Rizzo, S.J.; McTighe, S.; McKinzie, D.L. Genetic Background and Sex: Impact on Generalizability of Research Findings in Pharmacology Studies. In Good Research Practice in Non-Clinical Pharmacology and Biomedicine; Handbook of Experimental Pharmacology; Springer: Cham, Switzerland, 2020; Volume 257, pp. 147–162. [Google Scholar] [CrossRef]
  146. Liu, K.A.; Mager, N.A. Women’s involvement in clinical trials: Historical perspective and future implications. Pharm. Pract. 2016, 14, 708. [Google Scholar] [CrossRef] [PubMed]
  147. Carey, J.L.; Nader, N.; Chai, P.R.; Carreiro, S.; Griswold, M.K.; Boyle, K.L. Drugs and Medical Devices: Adverse Events and the Impact on Women’s Health. Clin. Ther. 2017, 39, 10–22. [Google Scholar] [CrossRef] [PubMed]
  148. Buoncervello, M.; Marconi, M.; Carè, A.; Piscopo, P.; Malorni, W.; Matarrese, P. Preclinical models in the study of sex differences. Clin. Sci. 2017, 131, 449–469. [Google Scholar] [CrossRef]
  149. Voelkl, B.; Vogt, L.; Sena, E.S.; Würbel, H. Reproducibility of preclinical animal research improves with heterogeneity of study samples. PLoS Biol. 2018, 16, e2003693. [Google Scholar] [CrossRef]
  150. Sandberg, K.; Umans, J.G.; Group, G.C.C.W. Recommendations concerning the new U.S. National Institutes of Health initiative to balance the sex of cells and animals in preclinical research. FASEB J. 2015, 29, 1646–1652. [Google Scholar] [CrossRef]
  151. Voelkl, B.; Altman, N.S.; Forsman, A.; Forstmeier, W.; Gurevitch, J.; Jaric, I.; Karp, N.A.; Kas, M.J.; Schielzeth, H.; Van de Casteele, T.; et al. Reproducibility of animal research in light of biological variation. Nat. Rev. Neurosci. 2020, 21, 384–393. [Google Scholar] [CrossRef]
  152. Freedman, L.P.; Cockburn, I.M.; Simcoe, T.S. The Economics of Reproducibility in Preclinical Research. PLoS Biol. 2015, 13, e1002165. [Google Scholar] [CrossRef]
  153. Istituto Superiore di Sanità. Standard di Cura per la Salute di Persone Transessuali, Transgender e di Genere Non-Conforme. Available online: https://www.iss.it/identit%C3%A0-di-genere/-/asset_publisher/xqprREqYmtu7/content/standard-di-cura-per-la-salute-di-persone-transessuali-transgender-e-di-genere-non-conforme (accessed on 10 November 2022).
  154. T’Sjoen, G.; Arcelus, J.; Gooren, L.; Klink, D.T.; Tangpricha, V. Endocrinology of Transgender Medicine. Endocr. Rev. 2019, 40, 97–117. [Google Scholar] [CrossRef]
  155. Pettit, S.D. ToxPoint: Health Disparities, COVID-19, and Owning Our Share. Toxicol. Sci. 2021, 179, 147–148. [Google Scholar] [CrossRef]
  156. Rusyn, I.; Chiu, W.A.; Wright, F.A. Model systems and organisms for addressing inter- and intra-species variability in risk assessment. Regul. Toxicol. Pharmacol. 2022, 132, 105197. [Google Scholar] [CrossRef] [PubMed]
  157. Tassinari, R.; Cordelli, E.; Eleuteri, P.; Villani, P.; Pacchierotti, F.; Narciso, L.; Tait, S.; Valeri, M.; Martinelli, A.; Di Felice, G.; et al. Effects of sub-chronic oral exposure to pyrogenic synthetic amorphous silica (NM-203) in male and female Sprague-Dawley rats: Focus on reproductive systems. Reprod. Toxicol. 2021, 105, 17–24. [Google Scholar] [CrossRef]
  158. Arnold, A.P. Four Core Genotypes and XY* mouse models: Update on impact on SABV research. Neurosci. Biobehav. Rev. 2020, 119, 1–8. [Google Scholar] [CrossRef] [PubMed]
  159. Burgoyne, P.S.; Arnold, A.P. A primer on the use of mouse models for identifying direct sex chromosome effects that cause sex differences in non-gonadal tissues. Biol. Sex Differ. 2016, 7, 68. [Google Scholar] [CrossRef]
  160. Burgoyne, P.S.; Mahadevaiah, S.K.; Perry, J.; Palmer, S.J.; Ashworth, A. The Y* rearrangement in mice: New insights into a perplexing PAR. Cytogenet. Genome Res. 1998, 80, 37–40. [Google Scholar] [CrossRef]
  161. De Vries, G.J.; Rissman, E.F.; Simerly, R.B.; Yang, L.Y.; Scordalakes, E.M.; Auger, C.J.; Swain, A.; Lovell-Badge, R.; Burgoyne, P.S.; Arnold, A.P. A model system for study of sex chromosome effects on sexually dimorphic neural and behavioral traits. J. Neurosci. 2002, 22, 9005–9014. [Google Scholar] [CrossRef] [PubMed]
  162. Eicher, E.M.; Hale, D.W.; Hunt, P.A.; Lee, B.K.; Tucker, P.K.; King, T.R.; Eppig, J.T.; Washburn, L.L. The mouse Y* chromosome involves a complex rearrangement, including interstitial positioning of the pseudoautosomal region. Cytogenet. Genome Res. 1991, 57, 221–230. [Google Scholar] [CrossRef] [PubMed]
  163. Mahadevaiah, S.K.; Odorisio, T.; Elliott, D.J.; Rattigan, A.; Szot, M.; Laval, S.H.; Washburn, L.L.; McCarrey, J.R.; Cattanach, B.M.; Lovell-Badge, R.; et al. Mouse homologues of the human AZF candidate gene RBM are expressed in spermatogonia and spermatids, and map to a Y chromosome deletion interval associated with a high incidence of sperm abnormalities. Humam Mol. Genet. 1998, 7, 715–727. [Google Scholar] [CrossRef] [PubMed]
  164. Arnold, A.P. A general theory of sexual differentiation. J. Neurosci. Res. 2017, 95, 291–300. [Google Scholar] [CrossRef]
  165. Kinnear, H.M.; Hashim, P.H.; Dela Cruz, C.; Chang, F.L.; Rubenstein, G.; Nimmagadda, L.; Elangovan, V.R.; Jones, A.; Brunette, M.A.; Hannum, D.F.; et al. Presence of ovarian stromal aberrations after cessation of testosterone therapy in a transgender mouse model. Biol. Reprod. 2023, 108, 802–813. [Google Scholar] [CrossRef] [PubMed]
  166. Barus, R.; Bergeron, S.; Chen, Y.; Gautier, S. Sex differences: From preclinical pharmacology to clinical pharmacology. Therapie 2023, 78, 189–194. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Allegra, S.; Chiara, F.; Di Grazia, D.; Gaspari, M.; De Francia, S. Evaluation of Sex Differences in Preclinical Pharmacology Research: How Far Is Left to Go? Pharmaceuticals 2023, 16, 786. https://doi.org/10.3390/ph16060786

AMA Style

Allegra S, Chiara F, Di Grazia D, Gaspari M, De Francia S. Evaluation of Sex Differences in Preclinical Pharmacology Research: How Far Is Left to Go? Pharmaceuticals. 2023; 16(6):786. https://doi.org/10.3390/ph16060786

Chicago/Turabian Style

Allegra, Sarah, Francesco Chiara, Daniela Di Grazia, Marco Gaspari, and Silvia De Francia. 2023. "Evaluation of Sex Differences in Preclinical Pharmacology Research: How Far Is Left to Go?" Pharmaceuticals 16, no. 6: 786. https://doi.org/10.3390/ph16060786

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop