Next Article in Journal
Ventricular Tachycardia Catheter Ablation: Retrospective Analysis and Prospective Outlooks—A Comprehensive Review
Previous Article in Journal
An Investigation of Metabolic Risk Factors and Gut Microbiota in Unexplained Syncope
Previous Article in Special Issue
Sex Differences in Oxycodone/Naloxone vs. Tapentadol in Chronic Non-Cancer Pain: An Observational Real-World Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Gender Medicine and Pharmacology

Laboratory of Clinical Pharmacology Service “Franco Ghezzo”, Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga Hospital, 10043 Orbassano, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(2), 265; https://doi.org/10.3390/biomedicines12020265
Submission received: 12 January 2024 / Revised: 18 January 2024 / Accepted: 22 January 2024 / Published: 24 January 2024
(This article belongs to the Special Issue Gender Medicine and Pharmacology)
Gender-specific medicine consists of a transversal methodological approach that aims to study the influence of sex and gender on diseases. Gender-specific medicine aims to understand how diseases of all organs and systems manifest themselves in the two sexes and, above all, to evaluate the differences in symptoms, the need for different diagnostic paths and interpretations of results, the differences in response to drugs, or even the need to use different drugs. Again, there are differences related to the prevention of all diseases. In fact, treatment response varies between men and women; these differences are mostly due to anatomical, physiological, and hormonal factors. The response to treatments is influenced by variations in the pharmacokinetics and pharmacodynamics of therapeutic agents [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36]. Age and hormones play an important role in drug adsorption, distribution, metabolism, and elimination [37]. Considering females, changes in endogenous sex steroid hormones that occur naturally during the menstrual cycle, during pregnancy, and during the menopausal transition may have an impact on treatment effectiveness and adverse drug reactions [38]. Unlike female reproductive aging (menopause) or organic androgen deficiency in males (due to disorders of the brain, pituitary, or testes), male reproductive aging does not lead to a total cessation of testosterone production or of spermatogenesis. In addition, testosterone insufficiency affects a tiny percentage of aged men and is determined by the existence of comorbidities. Serum testosterone levels are generally normal in older men who maintain good health, and age-related low testosterone has been referred to andropause, viropause, partial androgen deficit in the aging man, and late-onset hypogonadism [39]. Testosterone injection is used to virilize gender-incongruent patients who were given the female gender at birth (AFAB). Even when the same hormone formulations and regimens are utilized, inter-individual variations in the timing and acquisition of phenotypic traits are commonly noted. Pallotti et al. investigated the potential associations between the CpG methylation profile of the H19 and estrogen receptor 2 gene (ESR2) and their impact on phenotypic changes in a group of 13 individuals with AFAB at baseline (T0) and following six months (T6) and twelve months (T12) of testosterone treatment [40]. They speculated that exogenous testosterone treatment may alter the ESR2 methylation pattern. Additionally, they noted that epigenetic modifications seemed to be controlled, implying that the regulation of estrogen signaling is important from the start of the treatment until T6, at which point it ceases to be significantly altered until T12. Moreover, it appears that exogenous testosterone administration and subject age are related to estrogen receptor methylation, as a marker of androgenic therapy. Furthermore, the gonadal axis is inhibited when certain drugs, such as opioids and glucocorticoids, are taken [41,42,43]. Eventually, older men with higher body mass index are more susceptible to oxidative stress, linked to insulin resistance, low sperm concentration, and erectile dysfunction [44]. Considering pharmacodynamics, many variations exist based on sex, mostly caused by hormones, genetics, and environmental factors [28]. Prolactin and female steroid hormones influence immunity. The hypothalamic–pituitary–adrenal and hypothalamic–pituitary–gonadal axes regulate immunity, resulting in two to ten times higher prevalence and severity of inflammatory and autoimmune diseases in females than in males. It is mostly females of reproductive age who suffer from autoimmune diseases. The role of sex is also debated in a number of allergy disorders and their treatment with omalizumab, the first biologic medication approved for use in the field of allergology [45]. In particular, at a 12-month follow-up visit, females’ general evaluation of their asthma was much worse than that of boys, even when their symptoms improved after starting omalizumab medication. Omalizumab treatment resulted in a better response in females than in males, but its safety and tolerability did not seem not gender-specific. Omalizumab probably has an effect on sex hormones directly or indirectly through the downstream processes they mediate.
It is known that pain varies across the sexes, but how it translates to practical pain management is not. Barrachina et al. compared the analgesic response of tapentadol (TAP) and oxycodone/naloxone (OXN) between sexes, in contrast to other opioids (OPO) that are frequently recommended for chronic non-cancer pain (CNCP) [46]. Enrolling 571 patients, they observed a significant gender disparity, with a higher hospital frequency and lower female tolerability, particularly in the OXN group. In this instance, compared to OXN men, females demonstrated higher hospital utilization, necessitating a higher morphine equivalent daily dose. Across all opioid groups, CNCP women were three years older than men, used benzodiazepines at a significantly higher rate, experienced more constipation and headaches as adverse events, and were more likely to report sexual dysfunction and losing interest. Women were also significantly older than men. Sex disparities were shown to be associated with higher hospital resources and worse drug acceptability in female OXN users. Also, Escorial and colleagues investigated the possible sex-related disparities among outpatients with CNCP, with an observational cross-sectional study on 806 patients [47]. Of these patients, 137 had an opioid use disorder diagnosis; the other 669 were considered as the control group. In comparison to the male controls, the female controls were older, underwent less rigorous pain therapy, and had more psychiatric prescriptions and visits. Comparing the patients to controls, the results showed lower age, greater job impairment, double the daily dose of morphine equivalent, and usage of benzodiazepines. Moreover, males reported the highest number of adverse drug responses and exhibited a higher usage of fentanyl, whereas females demonstrated a higher rate of substance use disorder and lower rate of tramadol use.
In the field of cancer treatment and related toxicity, in our previous study, we highlighted the variations in lipid profiles according to sex in patients who underwent radical surgery for adrenocortical cancer while receiving adjuvant mitotane treatment. The results showed that the lipid profiles of men and women, as well as pre- and postmenopausal women, differed. In light of the effects of mitotane on lipid levels, higher drug concentrations were associated with higher levels of HDL in all groups, total cholesterol in both males and females, triglycerides in postmenopausal females, and LDL in male patients. An increase in mitotane active metabolite (o,p’-DDE) correlated positively with HDL levels and negatively with LDL in all the evaluated groups. Additionally, increases in o,p’-DDE were positively correlated with total cholesterol in pre- and postmenopausal women and with triglycerides in premenopausal females. Considering another type of cancer, cholangiocellular adenocarcinoma, palliative chemotherapy is still the only available treatment. To evaluate the effectiveness of chemotherapy, Jordens and colleagues assessed bone mineral density (BMD) as a predictive tool [48]. Their study included 75 patients who received treatment, and they observed that BMD declined with age but that there was no difference between men and women. The predictive value of BMD was only seen in females, indicating sex-specific differences. In conclusion, BMD appears to be an independent, useful, and easily accessible predictive measure for overall survival in patients with advanced cholangiocellular adenocarcinoma. A study by Damanskiene on pediatric glioma cells evaluated the histone deacetylase inhibitor valproic acid (VPA) as an anticancer and immunomodulatory drug. They examined the impact of 0.5 and 0.75 mM VPA on the gene expressions of the co-transporters NKCC1 (SLC12A2), KCC2 (SLC12A5), and SLC5A8 (SLC5A8), specifically in SF8628 (girls) and PBT24 (boys) cells, using an RT-PCR technique [49]. Between the PBT24 and SF8628 controls, there was no difference in the expressions of SLC12A2 and SLC5A8. Compared to the SF8628 control, the PBT24 control had substantially higher levels of SLC12A5 expression. In PBT24, VPA treatment dramatically enhanced SLC12A2 expression; however, SF8628 cells were unaffected. Compared to matching SF8628 groups, the PBT24-treated cells had a considerably higher level of SLC12A5 expression. The expression of SLC5A8 in PBT24-treated cells was noticeably higher than in the corresponding SF8628 groups. The observed differences point to possible gender-related variations in tumor cell biology. Another work conducted by Salas and colleagues observed that myometrial and MED12 mutant leiomyoma cells repopulated cell-depleted tissue slices after 20 days of culture. They detected clusters of CD49b+ cells in tumor slices using immunofluorescence and quantitative PCR of stem cell and undifferentiated cell markers [50]. However, there were only a few CD49b+ cells found in the myometrial slices. While only a small percentage of myometrial cells were stained for this proliferation marker, almost all leiomyoma cells showed significant expression of Ki67. While the mesenchymal stem cell receptor KIT was only found in normal cells, the CD73 marker was exclusively expressed in tumor cells. Compared to myometrial cells, leiomyoma cells exhibited greater signal intensity and broader expression patterns for HMGA2 and CD24. According to the proposed research, asymmetrical division caused by the activation of CD49b+ stem cells in the myometrium results in the development of transit-amplifying KIT+ cells, which subsequently differentiate into smooth muscle cells. Activated leiomyoma CD49b+ cells, on the other hand, divide symmetrically to create clusters of stem cells that can proliferate and develop into smooth muscle cells. In conclusion, they speculated that both normal and mutant stem cells may multiply and differentiate in an organ culture over an extended period of time, providing a useful platform for the development of new therapeutics.
Studying the gender differences found during the COVID-19 epidemic is essential, and analyzing the positive and negative effects of medications on sex as well as their effectiveness in clinical trials is crucial for treating COVID-19 [51]. One possible medication that may be developed to stop the spread and complications of SARS-CoV-2 infection is VPA, a histone deacetylase inhibitor. Preclinical studies by Stakistaitis indicate that the pharmacological effects of VPA may be relevant to the pathogenetic pathways of COVID-19. Through mechanisms that may be connected to sex, VPA lowers inflammatory tissue and organ damage, suppresses the pro-inflammatory immune cell and cytokine response to infection, and prevents the entry of the SARS-CoV-2 virus. The potential benefits of VPA for treating COVID-19 include its antithrombotic, antiplatelet, anti-inflammatory, immunomodulatory, and blood serum effects on glucose and testosterone levels.
There are significant sex variations in the etiology, epidemiology, and prognosis of cardiovascular diseases (CVDs) between men and women, which is also the case when they are linked to ambient particulate matter (PM) exposure. A study conducted by Liu and colleagues on male and female C57BL/6 mice (8–10 weeks) reported that intranasal PM exposure dramatically reduced the numbers of endothelial progenitor cells (EPCs) in both blood and bone marrow, with an increase in reactive oxygen species (ROS) production in men but not in females, and males had higher serum levels of IL-6 and IL-1 than females. After being exposed to PM, males’ pulmonary expression of the antioxidant enzyme SOD1 dramatically decreased, but females’ did not. Therefore, the authors suggested that male mice’s lower expression of pulmonary SOD1 may be the cause of the PM exposure-induced drop in the EPC population [52].
Due to increased knowledge and accessibility to healthcare facilities, the gender-diverse and transgender community is a small patient group that is being seen in clinical settings more frequently [53]. Gender-affirming therapy, which involves both hormone medication and surgical intervention, typically results in partial impairment to full removal of reproductive potential and outcomes depending on the treatment modality. Prior to undergoing a medical or surgical gender transformation, transgender individuals should receive fertility preservation counseling, even though this is rarely implemented in the professional setting. Data and clinical information on transgender people’s ability to preserve their fertility are rather scarce. Despite the fact that there are several choices for fertility preservation, there are significant obstacles for the transgender population because of a lack of information and a clinical narrative that is unfamiliar to doctors and other healthcare professionals. More knowledge of this clinical agenda and the required procedures will eventually lead to far more specialized and thorough care for transgender patients who are desperately in need of fertility preservation treatments or counseling.
In conclusion, though specialized studies aimed at addressing gender inequalities are rare, gender disparities are becoming more apparent in every aspect of medicine, including drug therapy [45]. One essential variable that cannot be overlooked is sex. Gender pharmacology must always be considered while adjusting therapy in order to increase drug safety and effectiveness. Understanding gender differences encourages more health protection for both genders as well as the right use of medicines. In order to uncover potential hazards and benefits that can differ between genders, further gender study would enable reports on differences in the assimilation and response of the female organism compared to the male. Moreover, a deeper comprehension of the effects of sex and gender on pharmacological activity might facilitate the creation of customized, “tailor-made” medications.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Beierle, I.; Meibohm, B.; Derendorf, H. Gender differences in pharmacokinetics and pharmacodynamics. Int. J. Clin. Pharmacol. Ther. 1999, 37, 529–547. [Google Scholar] [PubMed]
  2. 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]
  3. 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]
  4. 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]
  5. 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]
  6. 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] [PubMed]
  7. 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]
  8. 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]
  9. Regitz-Zagrosek, V. Sex and gender differences in pharmacotherapy. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2014, 57, 1067–1073. [Google Scholar] [CrossRef]
  10. Zucker, I.; Prendergast, B.J. Sex differences in pharmacokinetics predict adverse drug reactions in women. Biol. Sex. Differ. 2020, 11, 32. [Google Scholar] [CrossRef]
  11. 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] [PubMed]
  12. Franconi, F.; Campesi, I. Sex Impact on Biomarkers, Pharmacokinetics and Pharmacodynamics. Curr. Med. Chem. 2017, 24, 2561–2575. [Google Scholar] [CrossRef] [PubMed]
  13. 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]
  14. Anderson, G.D. Gender differences in pharmacological response. Int. Rev. Neurobiol. 2008, 83, 1–10. [Google Scholar] [CrossRef]
  15. 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]
  16. Pérez, J.F.; Olguín, H.J.; Pérez, C.F.; Guillé, G.P.; Pérez, A.G.; Vieyra, A.C.; López, A.T.; Portugal, M.C.; Asseff, I.L. 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] [CrossRef]
  17. Franconi, F.; Brunelleschi, S.; Steardo, L.; Cuomo, V. Gender differences in drug responses. Pharmacol. Res. 2007, 55, 81–95. [Google Scholar] [CrossRef]
  18. 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]
  19. 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]
  20. 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]
  21. 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] [PubMed]
  22. Harris, R.Z.; Benet, L.Z.; Schwartz, J.B. Gender effects in pharmacokinetics and pharmacodynamics. Drugs 1995, 50, 222–239. [Google Scholar] [CrossRef]
  23. 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]
  24. 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]
  25. 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]
  26. 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] [PubMed]
  27. 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] [PubMed]
  28. Soldin, O.P.; Mattison, D.R. Sex differences in pharmacokinetics and pharmacodynamics. Clin. Pharmacokinet. 2009, 48, 143–157. [Google Scholar] [CrossRef]
  29. 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]
  30. 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]
  31. 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] [PubMed]
  32. 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]
  33. 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]
  34. 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]
  35. 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] [PubMed]
  36. 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]
  37. Spoletini, I.; Vitale, C.; Malorni, W.; Rosano, G.M. Sex differences in drug effects: Interaction with sex hormones in adult life. Handb. Exp. Pharmacol. 2012, 214, 91–105. [Google Scholar] [CrossRef]
  38. 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]
  39. Figueiredo, M.G.; Gagliano-Jucá, T.; Basaria, S. Male Reproduction and Aging. Endocrinol. Metab. Clin. N. Am. 2023, 52, 211–228. [Google Scholar] [CrossRef]
  40. Pallotti, F.; Senofonte, G.; Konstantinidou, F.; Di Chiano, S.; Faja, F.; Rizzo, F.; Cargnelutti, F.; Krausz, C.; Paoli, D.; Lenzi, A.; et al. Epigenetic Effects of Gender-Affirming Hormone Treatment: A Pilot Study of the ESR2 Promoter’s Methylation in AFAB People. Biomedicines 2022, 10, 459. [Google Scholar] [CrossRef]
  41. 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]
  42. 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]
  43. 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] [PubMed]
  44. Wonisch, W.; Falk, A.; Sundl, I.; Winklhofer-Roob, B.M.; Lindschinger, M. Oxidative stress increases continuously with BMI and age with unfavourable profiles in males. Aging Male 2012, 15, 159–165. [Google Scholar] [CrossRef] [PubMed]
  45. Sirufo, M.M.; De Pietro, F.; Ginaldi, L.; De Martinis, M. Sex, Allergic Diseases and Omalizumab. Biomedicines 2022, 10, 328. [Google Scholar] [CrossRef] [PubMed]
  46. Barrachina, J.; Margarit, C.; Muriel, J.; López-Gil, V.; López-Gil, S.; Ballester, P.; Mira-Lorente, L.; Agulló, L.; Peiró, A.M. Sex Differences in Oxycodone/Naloxone vs. Tapentadol in Chronic Non-Cancer Pain: An Observational Real-World Study. Biomedicines 2022, 10, 2468. [Google Scholar] [CrossRef]
  47. Escorial, M.; Muriel, J.; Margarit, C.; Agulló, L.; Morales, D.; Peiró Peiró, A.M. Sex-Differences in Pain and Opioid Use Disorder Management: A Cross-Sectional Real-World Study. Biomedicines 2022, 10, 2302. [Google Scholar] [CrossRef]
  48. Jördens, M.S.; Wittig, L.; Loberg, C.; Heinrichs, L.; Keitel, V.; Schulze-Hagen, M.; Antoch, G.; Knoefel, W.T.; Fluegen, G.; Loosen, S.H.; et al. Bone Mineral Density Is a Predictor of Mortality in Female Patients with Cholangiocellular Carcinoma Undergoing Palliative Treatment. Biomedicines 2022, 10, 1660. [Google Scholar] [CrossRef]
  49. Damanskienė, E.; Balnytė, I.; Valančiūtė, A.; Alonso, M.M.; Stakišaitis, D. Different Effects of Valproic Acid on SLC12A2 SLC12A5 and SLC5A8 Gene Expression in Pediatric Gliablastoma cells as an Approach to Personalized Therapy. Biomedicines 2022, 10, 968. [Google Scholar] [CrossRef]
  50. Salas, A.; Beltrán-Flores, S.; Évora, C.; Reyes, R.; Montes de Oca, F.; Delgado, A.; Almeida, T.A. Stem Cell Growth and Differentiation in Organ Culture: New Insights for Uterine Fibroid Treatment. Biomedicines 2022, 10, 1542. [Google Scholar] [CrossRef]
  51. Stakišaitis, D.; Kapočius, L.; Valančiūtė, A.; Balnytė, I.; Tamošuitis, T.; Vaitkevičius, A.; Sužiedėlis, K.; Urbonienė, D.; Tatarūnas, V.; Kilimaitė, E.; et al. SARS-CoV-2 Infection, Sex-Related Differences, and a Possible Personalized Treatment Approach with Valproic Acid: A Review. Biomedicines 2022, 10, 962. [Google Scholar] [CrossRef] [PubMed]
  52. Liu, X.; Wang, A.; Chen, Z.; Cui, Y.; Hao, H.; Domeier, T.L.; Sun, Q.; Liu, Z. Tempol Preserves Endothelial Progenitor Cells in Male Mice with Ambient Fine Particulate Matter Exposure. Biomedicines 2022, 10, 327. [Google Scholar] [CrossRef] [PubMed]
  53. Choi, J.Y.; Kim, T.J. Fertility Preservation and Reproductive Potential in Transgender and Gender Fluid Population. Biomedicines 2022, 10, 2279. [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.; De Francia, S. Gender Medicine and Pharmacology. Biomedicines 2024, 12, 265. https://doi.org/10.3390/biomedicines12020265

AMA Style

Allegra S, Chiara F, De Francia S. Gender Medicine and Pharmacology. Biomedicines. 2024; 12(2):265. https://doi.org/10.3390/biomedicines12020265

Chicago/Turabian Style

Allegra, Sarah, Francesco Chiara, and Silvia De Francia. 2024. "Gender Medicine and Pharmacology" Biomedicines 12, no. 2: 265. https://doi.org/10.3390/biomedicines12020265

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