Skip to main content
International Journal of Epidemiology logoLink to International Journal of Epidemiology
. 2019 Jul 10;48(3):830–833. doi: 10.1093/ije/dyz141

Commentary: Mendelian randomization and women’s health

Jenny C Censin 1,2,, Jonas Bovijn 1,2, Michael V Holmes 1,3,4,5, Cecilia M Lindgren 1,2,5,6
PMCID: PMC6659371  PMID: 31292646

Women and men are not biologically identical; differences in body shapes and compositions, hormone levels, enzymes, lifestyles and other factors lead to alterations in the presentation, diagnosis and natural history of disease, as well as drug efficacy and safety.1–3 Yet, such differences have historically been disregarded and women’s health conditions continue to be under-researched, under-diagnosed and under-treated.

Estimates suggest that up to 10% of women between 18 and 45 years are affected by polycystic ovarian syndrome (PCOS), making it the most common endocrinopathy among women of reproductive age.4,5 Despite this, most PCOS studies have had small sample sizes,6–10 and survey data suggest that over a third of PCOS patients have to wait more than 2 years for diagnosis.11 At the same time, comorbidities are under-diagnosed and under-treated,7–9 despite substantial effects on patient health and quality of life.10,12

Women are disproportionately affected by common diseases such as Alzheimer's and osteoarthritis, as compared with men, and experience more disease-related disability.13 In addition to such disease that affects both women and men, it is estimated that 5% of disability-adjusted life years (DALYs) in women arises from diseases specific to women, with the corresponding value being almost 10-fold lower in men at 0.7% of DALYS.13 Despite this, therapeutic options for women’s health conditions remain limited. In the 5-year period between 2014 and 2018, the US Food and Drug Administration (FDA) approved 213 novel drugs.14 Of these, only seven (3.3%) drugs were for female-specific indications, with a further five for breast cancer-related indications.14 The corresponding value for male-specific indications was two (both related to prostate cancer), a value that is iniquitous to the proportion of DALYs arising from sex-specific disease.14 Historically, the FDA also excluded women of childbearing potential from participating in clinical trials, largely driven by concerns of teratogenicity and a perceived difficulty in accounting for female-specific hormonal fluctuations.1,15 Although guidelines and policies established in the 1990s and 2000s15,16 led to greater inclusion of women in clinical trials,1 a recent study found that 72% of clinical trials did not provide sex-specific outcome data.17 With pharmacokinetic and pharmacodynamic differences evident between men and women for many drugs,18 and with most drugs withdrawn from the market having greater health risks in women,18,19 a detailed analysis of sex-specific effects of drug efficacy and safety is essential.

Genetic epidemiology may potentially offer solutions here. Mendelian randomization (MR) presents an opportunity to explore causal relationships between risk factors (e.g. obesity), therapeutic targets (e.g. proteins, metabolites and hormones) and disease risk.20 In MR, genetic variants associated with altered levels of an exposure are used as a proxy for the risk factor or therapeutic target.20 Causal effects of the exposure on the outcome can be estimated by assessing how these variants affect the outcome.20 Since genetic variants are randomly allocated at conception, estimates from MR should be less affected by confounding, and the non-modifiable nature of genotype abrogates reverse causation.20 Most large genome-wide association studies (GWAS) include a large proportion of women (48–100%),21 and sex-specific effect estimates are increasingly reported,22,23 albeit still only for a minority of GWAS. By leveraging these sex-specific genetic data, MR could ameliorate the historical under-representation of women in research studies in a safe and cost-effective manner. With increasingly large GWAS and biobank datasets publicly available for risk factors, potential drug targets (e.g. protein concentrations) and disease outcomes, this represents an opportune moment to identify causal risk factors and novel drug targets in a sex-stratified manner.

In this MR-themed special issue of the journal, the three articles by Dimou et al.24, Shu et al.25 and Harris et al.26 together advance our knowledge of how metabolic risk factors and sex hormones affect female health.

First, Dimou et al. show that sex hormone-binding globulin (SHBG) may have opposing effects on estrogen receptor-positive and- negative breast cancer risk. Their results provide evidence for higher SHBG levels decreasing risk of estrogen receptor-positive breast cancer and increasing risk of estrogen receptor-negative breast cancer, although they note that the latter needs to be confirmed in future studies. They included body mass index (BMI) in their multivariable analyses as it might be a confounder—and metabolic traits might have an impact on SHBG.27,28 Other hormones such as insulin have also been linked to SHBG levels and breast cancer risk,29,30 and further work is needed to tease apart the causal relationships between specific obesity traits (e.g. fat distribution), glycaemic traits (e.g. insulin levels), sex hormones and SHBG, and their respective roles in the development of different breast cancer subtypes.

BMI has been suggested as having opposing effects on breast cancer risk depending on menopausal status (higher BMI being associated with lower risk of breast cancer risk in premenopausal women and with higher risk of breast cancer in postmenopausal women).31–33 Contradicting this, the study by Shu et al., suggests that BMI is protective for both pre- and postmenopausal breast cancer, in keeping with a previous large MR study.34 The explanation for this inconsistency is unclear, but it might be due to differences in whether the BMI measurement, or the genetic instrument for BMI, largely captures ‘lifetime’ adult BMI (including early life BMI), BMI after menopause or postmenopausal weight gain.34 This discrepancy highlights the need for more precise measurements of risk factors to tease apart these relationships; e.g. the effects of weight gain and body fat composition and distribution, and the interplay of these at different ages and for various durations.

Next, Harris et al. corroborate previous evidence linking genetic liability to PCOS to lower risk of ovarian cancer, and show that the causal link between these two conditions may only be limited to certain histological subtypes (particularly endometrioid tumours). However, the associations are still relatively weak and it remains unclear whether this protective effect, if real, is mediated by oligo-anovulation, hyperandrogenism, or other PCOS-related mechanisms.

Collectively, these three contributions highlight the need for more precise and specific definitions of phenotypes, and equally important, larger sample sizes when assessing robust causal relationships. It is evident from these studies and the broader literature that metabolic traits play a major role in female health, though causality and directions of effects remain largely unexplored in the complex interplay between metabolic traits, hormones and female-specific diseases. With the rising obesity epidemic,35 it is ever more important to elucidate the precise nature of these associations to reliably inform public health policies.

As we improve our understanding of the biology underlying women’s health, we should advocate for improvements in women’s health not only from a biological perspective but more holistically. For instance, recent legislation in several US states has restricted access to abortion, despite evidence that shows improving such access leads to better maternal health outcomes.36 Thus, a broader approach to prioritize and advance women’s health which encompasses biological, political and social aspects should be embraced by all.

Funding

J.C.C. is funded by the Oxford Medical Research Council Doctoral Training Partnership (Oxford MRC DTP) and the Nuffield Department of Clinical Medicine, University of Oxford. J.B. is supported by funding from the Rhodes Trust, Clarendon Fund and the Medical Sciences Doctoral Training Centre, University of Oxford. M.V.H. works in a unit that receives funding from the MRC and is supported by a British Heart Foundation Intermediate Clinical Research Fellowship (FS/18/23/33512) and the National Institute for Health Research Oxford Biomedical Research Centre. C.M.L. is supported by the Li Ka Shing Foundation, WT-SSI/John Fell funds, the NIHR Biomedical Research Centre, Oxford Widenlife and NIH (5P50HD028138-27).

Conflict of interest: M.V.H has collaborated with Boehringer Ingelheim in research, and in accordance with the policy of the Clinical Trial Service Unit and Epidemiological studies Unit (University of Oxford), did not accept any personal payment. C.M.L has collaborated with Novo Nordisk and Bayer in research, and in accordance with the policy of the University of Oxford, did not accept any personal payment. None of other authors declared.

References

  • 1. Liu KA, Mager N.. Women’s involvement in clinical trials: historical perspective and future implications. Pharm Pract (Granada) 2016;14:708.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Ober C, Loisel DA, Gilad Y.. Sex-specific genetic architecture of human disease. Nat Rev Genet 2008;9:911–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Doyal L. Sex, gender, and health: the need for a new approach. BMJ 2001;323:1061–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. McCartney CR, Marshall JC, Clinical P.. Polycystic Ovary Syndrome. N Engl J Med 2016;375:54–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Bozdag G, Mumusoglu S, Zengin D, Karabulut E, Yildiz BO.. The prevalence and phenotypic features of polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod 2016;31:2841–55. [DOI] [PubMed] [Google Scholar]
  • 6. Moran LJ, Hutchison SK, Norman RJ, Teede HJ.. Lifestyle changes in women with polycystic ovary syndrome. Cochrane Database Syst Rev 2011;CD007506. [DOI] [PubMed] [Google Scholar]
  • 7. Wolf WM, Wattick RA, Kinkade ON, Olfert MD.. The current description and future need for multidisciplinary PCOS clinics. J Clin Med Res 2018;7:395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Broder-Fingert S, Shah B, Kessler M, Pawelczak M, David R.. Evaluation of adolescents for polycystic ovary syndrome in an urban population. J Clin Res Pediatr Endocrinol 2009;1:188–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Sivayoganathan D, Maruthini D, Glanville JM, Balen AH.. Full investigation of patients with polycystic ovary syndrome (PCOS) presenting to four different clinical specialties reveals significant differences and undiagnosed morbidity. Hum Fertil 2011;14:261–65. [DOI] [PubMed] [Google Scholar]
  • 10. Coffey S, Bano G, Mason HD.. Health-related quality of life in women with polycystic ovary syndrome: a comparison with the general population using the polycystic ovary syndrome questionnaire (PCOSQ) and the short form-36 (SF-36). Gynecol Endocrinol 2006;22:80–86. [DOI] [PubMed] [Google Scholar]
  • 11. Gibson-Helm M, Teede H, Dunaif A, Dokras A.. Delayed diagnosis and a lack of information associated with dissatisfaction in women with polycystic ovary syndrome. J Clin Endocrinol Metab 2017;102:604–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Hart R, Doherty DA.. The potential implications of a PCOS diagnosis on a woman’s long-term health using data linkage. J Clin Endocrinol Metab 2015;100:911–19. [DOI] [PubMed] [Google Scholar]
  • 13. Buvinić M, Medici A, Fernández E, Torres AC.. Gender differentials in health In: Jamison DT, Breman JG, Measham AR. et al. (eds). Disease Control Priorities in Developing Countries. Washington (DC: ): World Bank, 2011. [Google Scholar]
  • 14.Center for Drug Evaluation, Research; U.S. Food and Drug Administration. CDER’s New Molecular Entities and New Therapeutic Biological Products http://www.fda.gov/drugs/development-approval-process-drugs/new-drugs-fda-cders-new-molecular-entities-and-new-therapeutic-biological-products (29 May 2019, date last accessed).
  • 15.Food and Drug Administration. Guideline for the study and evaluation of gender differences in the clinical evaluation of drugs; notice. Fed Regist 1993;58:39406–16. [PubMed] [Google Scholar]
  • 16.Food and Drug Administration. Investigational new drug applications and new drug applications. Fed Regist 1998;63:6854–62. [PubMed] [Google Scholar]
  • 17. Geller SE, Koch AR, Roesch P, Filut A, Hallgren E, Carnes M.. The more things change, the more they stay the same: a study to evaluate compliance with inclusion and assessment of women and minorities in randomized controlled trials. Acad Med 2018;93:630–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Tamargo J, Rosano G, Walther T. et al. Gender differences in the effects of cardiovascular drugs. Eur Heart J Cardiovasc Pharmacother 2017;3:163–82. [DOI] [PubMed] [Google Scholar]
  • 19.US General Accounting Office (GAO). Drug Safety: Most Drugs Withdrawn in Recent Years had Greater Health Risks for Women https://www.gao.gov/assets/100/90642.pdf (19 January 2001, date last accessed).
  • 20. Davies NM, Holmes MV, Davey Smith G.. Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. BMJ 2018;362:k601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Mills MC, Rahal C.. A scientometric review of genome-wide association studies. Commun Biol 2019;2:9.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Morris AP, Voight BF, Teslovich TM. et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet 2012;44:981–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Pulit SL, Stoneman C, Morris AP. et al. Meta-analysis of genome-wide association studies for body fat distribution in 694 649 individuals of European ancestry. Hum Mol Genet 2019;28:166–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Dimou NL, Papadimitriou N, Gill D. et al. Sex hormone binding globulin and risk of breast cancer: a Mendelian randomization study. Int J Epidemiol 2019;48:807–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Shu X, Wu L, Khankari NK. et al. Associations of obesity and circulating insulin and glucose with breast cancer risk: a Mendelian randomization analysis. Int J Epidemiol 2019;48:1013–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Harris HR, Cushing-Haugen KL, Webb PM. et al. Association between genetically predicted polycystic ovary syndrome and ovarian cancer: a Mendelian randomization study. Int J Epidemiol 2019;48:822–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Seyfart T, Friedrich N, Kische H. et al. Association of sex hormones with physical, laboratory, and imaging markers of anthropometry in men and women from the general population. PLoS One 2018;13:e0189042.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Azrad M, Gower BA, Hunter GR, Nagy TR.. Intra-abdominal adipose tissue is independently associated with sex-hormone binding globulin in premenopausal women. Obesity 2012;20:1012–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Maggio M, Lauretani F, Basaria S. et al. Sex hormone binding globulin levels across the adult lifespan in women - the role of body mass index and fasting insulin. J Endocrinol Invest 2008;31:597–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Goodwin PJ. Insulin resistance in breast cancer: relevance and clinical implications. Breast Cancer Res 2011;13:7. [Google Scholar]
  • 31. Al-Ajmi K, Lophatananon A, Ollier W, Muir KR.. Risk of breast cancer in the UK biobank female cohort and its relationship to anthropometric and reproductive factors. PLoS One 2018;13:e0201097.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Bhaskaran K, Douglas I, Forbes H, dos-Santos SI, Leon DA, Smeeth L.. Body-mass index and risk of 22 specific cancers: a population-based cohort study of 5·24 million UK adults. Lancet 2014;384:755–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Pa van den B, Spiegelman D, Yaun SS. et al. Pooled analysis of prospective cohort studies on height, weight, and breast cancer risk. Am J Epidemiol 2000;152:514–27. [DOI] [PubMed] [Google Scholar]
  • 34. Guo Y, Warren Andersen S, Shu X-O. et al. Genetically predicted body mass index and breast cancer risk: Mendelian randomization analyses of data from 145, 000 women of European descent. PLoS Med 2016;13:e1002105.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.GBD 2015 Obesity Collaborators, Afshin A, Forouzanfar MH. et al. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med 2017;377:13–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lancet ed. We must all support women in the fight for abortion. Lancet 2019;393:2099. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Epidemiology are provided here courtesy of Oxford University Press

RESOURCES