1932

Abstract

The intersection of women's health and data science is a field of research that has historically trailed other fields, but more recently it has gained momentum. This growth is being driven not only by new investigators who are moving into this area but also by the significant opportunities that have emerged in new methodologies, resources, and technologies in data science. Here, we describe some of the resources and methods being used by women's health researchers today to meet challenges in biomedical data science. We also describe the opportunities and limitations of applying these approaches to advance women's health outcomes and the future of the field, with emphasis on repurposing existing methodologies for women's health.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-biodatasci-020722-105958
2023-08-10
2024-05-04
Loading full text...

Full text loading...

/deliver/fulltext/biodatasci/6/1/annurev-biodatasci-020722-105958.html?itemId=/content/journals/10.1146/annurev-biodatasci-020722-105958&mimeType=html&fmt=ahah

Literature Cited

  1. 1.
    Cao L. 2017. Data science: a comprehensive overview. ACM Comput. Surv. 50:43
    [Google Scholar]
  2. 2.
    Ober C, Loisel DA, Gilad Y. 2008. Sex-specific genetic architecture of human disease. Nat. Rev. Genet. 9:911–22
    [Google Scholar]
  3. 3.
    Arnold AP. 2017. A general theory of sexual differentiation. J. Neurosci. Res. 95:291–300
    [Google Scholar]
  4. 4.
    Karp NA, Mason J, Beaudet AL, Benjamini Y, Bower L et al. 2017. Prevalence of sexual dimorphism in mammalian phenotypic traits. Nat. Commun. 8:15475
    [Google Scholar]
  5. 5.
    Natri H, Garcia AR, Buetow KH, Trumble BC, Wilson MA. 2019. The pregnancy pickle: Evolved immune compensation due to pregnancy underlies sex differences in human diseases. Trends Genet. 35:478–88
    [Google Scholar]
  6. 6.
    Fathalla MF. 2019. Impact of reproductive evolutionary mismatch on women's health and the need for action and research. Int. J. Gynaecol. Obstet. 144:129–34
    [Google Scholar]
  7. 7.
    Whitacre CC. 2001. Sex differences in autoimmune disease. Nat. Immunol. 2:777–80
    [Google Scholar]
  8. 8.
    Willemars MMA, Nabben M, Verdonschot JAJ, Hoes MF. 2022. Evaluation of the interaction of sex hormones and cardiovascular function and health. Curr. Heart Fail. Rep. 19:200–12
    [Google Scholar]
  9. 9.
    Ying W, Catov JM, Ouyang P. 2018. Hypertensive disorders of pregnancy and future maternal cardiovascular risk. J. Am. Heart Assoc. 7:e009382
    [Google Scholar]
  10. 10.
    Rana S, Lemoine E, Granger JP, Karumanchi SA. 2019. Preeclampsia: pathophysiology, challenges, and perspectives. Circ. Res. 124:1094–112
    [Google Scholar]
  11. 11.
    Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, Brinton RD, Carrero JJ et al. 2020. Sex and gender: modifiers of health, disease, and medicine. Lancet 396:565–82
    [Google Scholar]
  12. 12.
    WHO (World Health Organ.) 2021. Endometriosis Fact Sheet, WHO Geneva: https://www.who.int/news-room/fact-sheets/detail/endometriosis
  13. 13.
    Chapron C, Marcellin L, Borghese B, Santulli P. 2019. Rethinking mechanisms, diagnosis and management of endometriosis. Nat. Rev. Endocrinol. 15:666–82
    [Google Scholar]
  14. 14.
    Landman A, Don EE, Vissers G, Ket HCJ, Oudijk MA et al. 2022. The risk of preterm birth in women with uterine fibroids: a systematic review and meta-analysis. PLOS ONE 17:e0269478
    [Google Scholar]
  15. 15.
    Stewart EA, Cookson CL, Gandolfo RA, Schulze-Rath R. 2017. Epidemiology of uterine fibroids: a systematic review. BJOG 124:1501–12
    [Google Scholar]
  16. 16.
    WHO (World Health Organ.) 2021. Breast cancer Fact Sheet, WHO Geneva: https://www.who.int/news-room/fact-sheets/detail/breast-cancer
  17. 17.
    Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I et al. 2021. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71:209–49
    [Google Scholar]
  18. 18.
    WHO (World Health Organ.) 2022. Cervical cancer Fact Sheet, WHO Geneva: https://www.who.int/news-room/fact-sheets/detail/cervical-cancer
  19. 19.
    Mejia AF, Culp TD, Cladel NM, Balogh KK, Budgeon LR et al. 2006. Preclinical model to test human papillomavirus virus (HPV) capsid vaccines in vivo using infectious HPV/cottontail rabbit papillomavirus chimeric papillomavirus particles. J. Virol. 80:12393–97
    [Google Scholar]
  20. 20.
    Danska JS. 2014. Sex matters for mechanism. Sci. Transl. Med. 6:258fs40
    [Google Scholar]
  21. 21.
    Schmidt A, Morales-Prieto DM, Pastuschek J, Frohlich K, Markert UR. 2015. Only humans have human placentas: molecular differences between mice and humans. J. Reprod. Immunol. 108:65–71
    [Google Scholar]
  22. 22.
    Carter AM. 2022. Evolution of placental hormones: implications for animal models. Front. Endocrinol. 13:891927
    [Google Scholar]
  23. 23.
    Gatford KL, Andraweera PH, Roberts CT, Care AS. 2020. Animal models of preeclampsia: causes, consequences, and interventions. Hypertension 75:1363–81
    [Google Scholar]
  24. 24.
    Elsakr JM, Zhao SK, Ricciardi V, Dean TA, Takahashi DL et al. 2021. Western-style diet consumption impairs maternal insulin sensitivity and glucose metabolism during pregnancy in a Japanese macaque model. Sci. Rep. 11:12977
    [Google Scholar]
  25. 25.
    Elsakr JM, Dunn JC, Tennant K, Zhao SK, Kroeten K et al. 2019. Maternal Western-style diet affects offspring islet composition and function in a non-human primate model of maternal over-nutrition. Mol. Metab. 25:73–82
    [Google Scholar]
  26. 26.
    Boktor JC, Adame MD, Rose DR, Schumann CM, Murray KD et al. 2022. Global metabolic profiles in a non-human primate model of maternal immune activation: implications for neurodevelopmental disorders. Mol. Psychiatry 27:4959–73
    [Google Scholar]
  27. 27.
    Comm. Women's Health Res 2010. Methodologic issues in women's health research. Women's Health Research: Progress, Pitfalls, and Promise221–39. Washington, DC: Natl. Acad. Press
    [Google Scholar]
  28. 28.
    Mazure CM, Jones DP. 2015. Twenty years and still counting: including women as participants and studying sex and gender in biomedical research. BMC Women's Health 15:94
    [Google Scholar]
  29. 29.
    Nadel MV. 1990. National Institutes of Health: problems in implementing policy on women in study populations Statement presented before Subcomm. Hous. Consumer Interest, Sel. Comm. Aging, U.S. House of Representatives Washington, DC:
  30. 30.
    Women's Health Inititat. Study Group 1998. Design of the Women's Health Initiative clinical trial and observational study. Control. Clin. Trials 19:61–109
    [Google Scholar]
  31. 31.
    Pinn VW. 1994. The role of the NIH's Office of Research on Women's Health. Acad. Med. 69:698–702
    [Google Scholar]
  32. 32.
    FDA (US Food Drug Admin.) 2019. Office of Women's Health Web Resour., FDA Silver Spring, MD: https://www.fda.gov/about-fda/office-commissioner/office-womens-health
  33. 33.
    NIH Revitalization Act of 1993, Pub. L. No. 103-43, 107 Stat. 122 1993. Subtitle B, Sections:131–33. https://www.congress.gov/103/statute/STATUTE-107/STATUTE-107-Pg122.pdf
  34. 34.
    Heinrich J 2001. Drug safety: most drugs withdrawn in recent years had greater health risks for women Letter to Hon. T Harkin, Hon. OJ Snowe, Hon. BA Mikulski, Hon. HA Waxman Washington, DC: U.S. Gen. Account. Off https://www.gao.gov/assets/gao-01-286r.pdf
  35. 35.
    Clayton JA, Collins FS. 2014. Policy: NIH to balance sex in cell and animal studies. Nature 509:282–83
    [Google Scholar]
  36. 36.
    21st Century Cures Act, Pub. L. No. 114-255, 130 Stat. 1033 2016. https://www.congress.gov/114/statute/STATUTE-130/STATUTE-130-Pg1033.pdf
  37. 37.
    Courvoisier N, Storari C, Lesage S, Vittoz L, Barbieux C et al. 2022. Facilitators and barriers of women's participation in HIV clinical research in Switzerland: a qualitative study. HIV Med. 23:441–47
    [Google Scholar]
  38. 38.
    Myles S, Tocci C, Falk M, Lynch S, Torres C et al. 2018. A multicenter investigation of factors influencing women's participation in clinical trials. J. Women's Health 27:258–70
    [Google Scholar]
  39. 39.
    van der Zande ISE, van der Graaf R, Hooft L, van Delden JJM. 2018. Facilitators and barriers to pregnant women's participation in research: a systematic review. Women Birth 31:350–61
    [Google Scholar]
  40. 40.
    Goldstein E, Bakhireva LN, Nervik K, Hagen S, Turnquist A et al. 2021. Recruitment and retention of pregnant women in prospective birth cohort studies: a scoping review and content analysis of the literature. Neurotoxicol. Teratol. 85:106974
    [Google Scholar]
  41. 41.
    Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C et al. 2002. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women's Health Initiative randomized controlled trial. JAMA 288:321–33
    [Google Scholar]
  42. 42.
    Boardman HM, Hartley L, Eisinga A, Main C, Roque i Figuls M et al. 2015. Hormone therapy for preventing cardiovascular disease in post-menopausal women. Cochrane Database Syst. Rev. 2015:3CD002229
    [Google Scholar]
  43. 43.
    Cagnacci A, Venier M. 2019. The controversial history of hormone replacement therapy. Medicina 55:9602
    [Google Scholar]
  44. 44.
    Manson JE, Chlebowski RT, Stefanick ML, Aragaki AK, Rossouw JE et al. 2013. Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials. JAMA 310:1353–68
    [Google Scholar]
  45. 45.
    Rossouw JE, Prentice RL, Manson JE, Wu L, Barad D et al. 2007. Postmenopausal hormone therapy and risk of cardiovascular disease by age and years since menopause. JAMA 297:1465–77
    [Google Scholar]
  46. 46.
    Salpeter SR, Walsh JM, Greyber E, Ormiston TM, Salpeter EE. 2004. Mortality associated with hormone replacement therapy in younger and older women: a meta-analysis. J. Gen. Intern. Med. 19:791–804
    [Google Scholar]
  47. 47.
    Salpeter SR, Walsh JM, Greyber E, Salpeter EE. 2006. Brief report: coronary heart disease events associated with hormone therapy in younger and older women. A meta-analysis. J. Gen. Intern. Med. 21:363–66
    [Google Scholar]
  48. 48.
    Schierbeck LL, Rejnmark L, Tofteng CL, Stilgren L, Eiken P et al. 2012. Effect of hormone replacement therapy on cardiovascular events in recently postmenopausal women: randomised trial. BMJ 345:e6409
    [Google Scholar]
  49. 49.
    Salsberry PJ, Reagan PB, Fang MZ. 2013. Disparities in women's health across a generation: a mother–daughter comparison. J. Women's Health 22:617–24
    [Google Scholar]
  50. 50.
    Sutton MY, Anachebe NF, Lee R, Skanes H. 2021. Racial and ethnic disparities in reproductive health services and outcomes, 2020. Obstet. Gynecol. 137:225–33
    [Google Scholar]
  51. 51.
    Hornbuckle LM, Amutah-Onukagha N, Bryan A, Skidmore Edwards E, Madzima T et al. 2017. Health disparities in women. Clin. Med. Insights Women's Health 10:1179562X17709546
    [Google Scholar]
  52. 52.
    Ranji U, Rosenzweig C, Salganicoff A. 2018. Women's coverage, access, and affordability: key findings from the 2017 Kaiser Women's Health Survey Issue Brief, Kaiser Family Found. San Francisco: https://www.kff.org/womens-health-policy/issue-brief/womens-coverage-access-and-affordability-key-findings-from-the-2017-kaiser-womens-health-survey
  53. 53.
    Johnston EM, Strahan AE, Joski P, Dunlop AL, Adams EK 2018. Impacts of the Affordable Care Act's Medicaid expansion on women of reproductive age: differences by parental status and state policies. Women's Health Issues 28:122–29
    [Google Scholar]
  54. 54.
    Hallam L, Vassallo A, Pinho-Gomes A-C, Carcel C, Woodward M. 2022. Does journal content in the field of women's health represent women's burden of disease? A review of publications in 2010 and 2020. J. Women's Health 31:611–19
    [Google Scholar]
  55. 55.
    Sauer CM, Chen LC, Hyland SL, Girbes A, Elbers P, Celi LA. 2022. Leveraging electronic health records for data science: common pitfalls and how to avoid them. Lancet Digit. Health 4:12893–98
    [Google Scholar]
  56. 56.
    Farmer R, Mathur R, Bhaskaran K, Eastwood SV, Chaturvedi N, Smeeth L. 2018. Promises and pitfalls of electronic health record analysis. Diabetologia 61:1241–48
    [Google Scholar]
  57. 57.
    Taksler GB, Dalton JE, Perzynski AT, Rothberg MB, Milinovich A et al. 2021. Opportunities, pitfalls, and alternatives in adapting electronic health records for health services research. Med. Decis. Making 41:133–42
    [Google Scholar]
  58. 58.
    Callahan A, Shah NH, Chen JH. 2020. Research and reporting considerations for observational studies using electronic health record data. Ann. Intern. Med. 172:S79–84
    [Google Scholar]
  59. 59.
    Benchimol EI, Smeeth L, Guttmann A, Harron K, Moher D et al. 2015. The REporting of studies Conducted using Observational Routinely-collected health Data (RECORD) statement. PLOS Med. 12:e1001885
    [Google Scholar]
  60. 60.
    von Elm E, Altman DG, Egger M, Pocock SJ, Gotzsche PC et al. 2007. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. Ann. Intern. Med. 147:573–77
    [Google Scholar]
  61. 61.
    Baird DD, Dunson DB, Hill MC, Cousins D, Schectman JM. 2003. High cumulative incidence of uterine leiomyoma in black and white women: ultrasound evidence. Am. J. Obstet. Gynecol. 188:100–7
    [Google Scholar]
  62. 62.
    Feingold-Link L, Edwards TL, Jones S, Hartmann KE, Velez Edwards DR. 2014. Enhancing uterine fibroid research through utilization of biorepositories linked to electronic medical record data. J. Women's Health 23:1027–32
    [Google Scholar]
  63. 63.
    Sudlow C, Gallacher J, Allen N, Beral V, Burton P et al. 2015. UK Biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLOS Med. 12:e1001779
    [Google Scholar]
  64. 64.
    Leitsalu L, Haller T, Esko T, Tammesoo ML, Alavere H et al. 2015. Cohort profile: Estonian Biobank of the Estonian Genome Center, University of Tartu. Int. J. Epidemiol. 44:1137–47
    [Google Scholar]
  65. 65.
    Nagai A, Hirata M, Kamatani Y, Muto K, Matsuda K et al. 2017. Overview of the BioBank Japan Project: study design and profile. J. Epidemiol. 27:S2–8
    [Google Scholar]
  66. 66.
    Knudsen LB, Borlum Kristensen F. 1986. Monitoring perinatal mortality and perinatal care with a national register: content and usage of the Danish Medical Birth Register. Community Med. 8:29–36
    [Google Scholar]
  67. 67.
    Locke AE, Steinberg KM, Chiang CWK, Service SK, Havulinna AS et al. 2019. Exome sequencing of Finnish isolates enhances rare-variant association power. Nature 572:323–28
    [Google Scholar]
  68. 68.
    Gordis L. 2014. Epidemiology Philadelpia: Elsevier
  69. 69.
    Meinert CL. 2012. Clinical Trials: Design, Conduct, and Analysis New York: Oxford Univ. Press
  70. 70.
    Carnes M, Morrissey C, Geller SE. 2008. Women's health and women's leadership in academic medicine: hitting the same glass ceiling?. J. Women's Health 17:1453–62
    [Google Scholar]
  71. 71.
    Zeng Z, Deng Y, Li X, Naumann T, Luo Y. 2019. Natural language processing for EHR-based computational phenotyping. IEEE/ACM Trans. Comput. Biol. Bioinform. 16:139–53
    [Google Scholar]
  72. 72.
    Wang P, Garza M, Zozus M. 2019. Cancer phenotype development: a literature review. Stud. Health Technol. Inform. 257:468–72
    [Google Scholar]
  73. 73.
    Sharifi-Heris Z, Laitala J, Airola A, Rahmani AM, Bender M. 2022. Machine learning approach for preterm birth prediction using health records: systematic review. JMIR Med. Inform. 10:e33875
    [Google Scholar]
  74. 74.
    Sufriyana H, Husnayain A, Chen YL, Kuo CY, Singh O et al. 2020. Comparison of multivariable logistic regression and other machine learning algorithms for prognostic prediction studies in pregnancy care: systematic review and meta-analysis. JMIR Med. Inform. 8:e16503
    [Google Scholar]
  75. 75.
    Espinosa C, Becker M, Marić I, Wong RJ, Shaw GM et al. 2021. Data-driven modeling of pregnancy-related complications. Trends Mol. Med. 27:762–76
    [Google Scholar]
  76. 76.
    Bertini A, Salas R, Chabert S, Sobrevia L, Pardo F. 2022. Using machine learning to predict complications in pregnancy: a systematic review. Front. Bioeng. Biotechnol. 9:780389
    [Google Scholar]
  77. 77.
    Davidson L, Boland MR. 2021. Towards deep phenotyping pregnancy: a systematic review on artificial intelligence and machine learning methods to improve pregnancy outcomes. Brief. Bioinform. 22:5bbaa369
    [Google Scholar]
  78. 78.
    Uffelmann E, Huang QQ, Munung NS, de Vries J, Okada Y et al. 2021. Genome-wide association studies. Nat. Rev. Methods Primers 1:59
    [Google Scholar]
  79. 79.
    Romualdo Cardoso S, Gillespie A, Haider S, Fletcher O 2021. Functional annotation of breast cancer risk loci: current progress and future directions. Br. J. Cancer 126:981–93
    [Google Scholar]
  80. 80.
    Low S-K, Zembutsu H, Nakamura Y. 2017. Breast cancer: the translation of big genomic data to cancer precision medicine. Cancer Sci. 109:3497–506
    [Google Scholar]
  81. 81.
    Buniello A, MacArthur JAL, Cerezo M, Harris LW, Hayhurst J et al. 2019. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 47:D1005–12
    [Google Scholar]
  82. 82.
    Mlodawska OW, Saini P, Parker JB, Wei JJ, Bulun SE et al. 2022. Epigenomic and enhancer dysregulation in uterine leiomyomas. Hum. Reprod. Update 28:518–47
    [Google Scholar]
  83. 83.
    Mohamad MA, Mohd Manzor NF, Zulkifli NF, Zainal N, Hayati AR, Ahmad Asnawi AW. 2020. A review of candidate genes and pathways in preeclampsia—an integrated bioinformatical analysis. Biology 9:462
    [Google Scholar]
  84. 84.
    Zhang J, Bajari R, Andric D, Gerthoffert F, Lepsa A et al. 2019. The International Cancer Genome Consortium Data Portal. Nat. Biotechnol. 37:367–69
    [Google Scholar]
  85. 85.
    GTEx Consort 2020. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369:1318–30
    [Google Scholar]
  86. 86.
    Kim G, Jang G, Song J, Kim D, Lee S et al. 2022. A transcriptome-wide association study of uterine fibroids to identify potential genetic markers and toxic chemicals. PLOS ONE 17:e0274879
    [Google Scholar]
  87. 87.
    Li B, Ritchie MD. 2021. From GWAS to gene: transcriptome-wide association studies and other methods to functionally understand GWAS discoveries. Front. Genet. 12:713230
    [Google Scholar]
  88. 88.
    Barbeira AN, Dickinson SP, Bonazzola R, Zheng J, Wheeler HE et al. 2018. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat. Commun. 9:1825
    [Google Scholar]
  89. 89.
    Gusev A, Ko A, Shi H, Bhatia G, Chung W et al. 2016. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 48:245–52
    [Google Scholar]
  90. 90.
    Gamazon ER, Wheeler HE, Shah KP, Mozaffari SV, Aquino-Michaels K et al. 2015. A gene-based association method for mapping traits using reference transcriptome data. Nat. Genet. 47:1091–98
    [Google Scholar]
  91. 91.
    Edwards TL, Giri A, Hellwege JN, Hartmann KE, Stewart EA et al. 2019. A trans-ethnic genome-wide association study of uterine fibroids. Front. Genet. 10:511
    [Google Scholar]
  92. 92.
    Hellwege JN, Jeff JM, Wise LA, Gallagher CS, Wellons M et al. 2017. A multi-stage genome-wide association study of uterine fibroids in African Americans. Hum. Genet. 136:1363–73
    [Google Scholar]
  93. 93.
    Chen H, Fan S, Stone J, Thompson DJ, Douglas J et al. 2022. Genome-wide and transcriptome-wide association studies of mammographic density phenotypes reveal novel loci. Breast Cancer Res. 24:27
    [Google Scholar]
  94. 94.
    Feng H, Gusev A, Pasaniuc B, Wu L, Long J et al. 2020. Transcriptome-wide association study of breast cancer risk by estrogen-receptor status. Genet. Epidemiol. 44:442–68
    [Google Scholar]
  95. 95.
    Wu L, Shi W, Long J, Guo X, Michailidou K et al. 2018. A transcriptome-wide association study of 229,000 women identifies new candidate susceptibility genes for breast cancer. Nat. Genet. 50:968–78
    [Google Scholar]
  96. 96.
    Shi J, Wu L, Li B, Lu Y, Guo X et al. 2019. Transcriptome-wide association study identifies susceptibility loci and genes for age at natural menopause. Reprod. Sci. 26:496–502
    [Google Scholar]
  97. 97.
    Guintivano J, Aberg KA, Clark SL, Rubinow DR, Sullivan PF et al. 2022. Transcriptome-wide association study for postpartum depression implicates altered B-cell activation and insulin resistance. Mol. Psychiatry 27:2858–67
    [Google Scholar]
  98. 98.
    Lu M, Feng R, Qin Y, Deng H, Lian B et al. 2022. Identifying environmental endocrine disruptors associated with the age at menarche by integrating a transcriptome-wide association study with chemical-gene-interaction analysis. Front. Endocrinol. 13:836527
    [Google Scholar]
  99. 99.
    Gusev A, Lawrenson K, Lin X, Lyra PC Jr., Kar S et al. 2019. A transcriptome-wide association study of high-grade serous epithelial ovarian cancer identifies new susceptibility genes and splice variants. Nat. Genet. 51:815–23
    [Google Scholar]
  100. 100.
    Lu Y, Beeghly-Fadiel A, Wu L, Guo X, Li B et al. 2018. A transcriptome-wide association study among 97,898 women to identify candidate susceptibility genes for epithelial ovarian cancer risk. Cancer Res. 78:5419–30
    [Google Scholar]
  101. 101.
    Zhou T, Huang L, Wang M, Chen D, Chen Z, Jiang SW 2020. A critical review of proteomic studies in gestational diabetes mellitus. J. Diabetes Res. 2020:6450352
    [Google Scholar]
  102. 102.
    Penick ER, Bateman NW, Rojas C, Magana C, Conrads K et al. 2022. Proteomic alterations associated with residual disease in neoadjuvant chemotherapy treated ovarian cancer tissues. Clin. Proteom. 19:35
    [Google Scholar]
  103. 103.
    Duda JM, Twigg CAI, Thomas SN. 2022. Differential histone deacetylase inhibitor-induced perturbations of the global proteome landscape in the setting of high-grade serous ovarian cancer. Proteomics 4:e2100372
    [Google Scholar]
  104. 104.
    Schon SB, Yang K, Schindler R, Jiang L, Neff LM et al. 2022. Obesity-related alterations in protein expression in human follicular fluid from women undergoing in-vitro fertilization (IVF). F&S Sci. 3:4331–39
    [Google Scholar]
  105. 105.
    Wang X, Zhao M, Guo Z, Song S, Liu S et al. 2022. Urinary proteomic analysis during pregnancy and its potential application in early prediction of gestational diabetes mellitus and spontaneous abortion. Ann. Transl. Med. 10:736
    [Google Scholar]
  106. 106.
    Alesi S, Ghelani D, Mousa A. 2021. Metabolomic biomarkers in polycystic ovary syndrome: a review of the evidence. Semin. Reprod. Med. 39:102–10
    [Google Scholar]
  107. 107.
    Craciunas L, Chu J, Pickering O, Mohiyiddeen L, Coomarasamy A. 2022. The metabolomic profile of endometrial receptivity in recurrent miscarriage. Minerva Obstet. Gynecol. https://doi.org/10.23736/S2724-606X.22.05151-X
    [Crossref] [Google Scholar]
  108. 108.
    Troisi J, Mollo A, Lombardi M, Scala G, Richards SM et al. 2022. The metabolomic approach for the screening of endometrial cancer: validation from a large cohort of women scheduled for gynecological surgery. Biomolecules 12:91229
    [Google Scholar]
  109. 109.
    Yu X, Chen Y, He L, Liu H, Yang Z, Lin Y 2022. Transcriptome and metabolome analyses reveal the interweaving of immune response and metabolic regulation in pelvic organ prolapse. Int. Urogynecol. J. https://doi.org/10.1007/s00192-022-05357-5
    [Crossref] [Google Scholar]
  110. 110.
    Ahmeidat A, Kotts WJ, Wong J, McLernon DJ, Black M. 2021. Predictive models of individual risk of elective caesarean section complications: a systematic review. Eur. J. Obstet. Gynecol. Reprod. Biol. 262:248–55
    [Google Scholar]
  111. 111.
    Black N, Henderson I, Al Wattar BH, Quenby S 2022. Predictive models for estimating the probability of successful vaginal birth after cesarean delivery: a systematic review. Obstet. Gynecol. 140:5821–41
    [Google Scholar]
  112. 112.
    Deng B, Li Y, Chen JY, Guo J, Tan J et al. 2022. Prediction models of vaginal birth after cesarean delivery: a systematic review. Int. J. Nurs. Stud. 135:104359
    [Google Scholar]
  113. 113.
    Mazo C, Aura C, Rahman A, Gallagher WM, Mooney C. 2022. Application of artificial intelligence techniques to predict risk of recurrence of breast cancer: a systematic review. J. Pers. Med. 12:191496
    [Google Scholar]
  114. 114.
    Zhong M, Zhang H, Yu C, Jiang J, Duan X. 2022. Application of machine learning in predicting the risk of postpartum depression: a systematic review. J. Affect. Disord. 318:364–79
    [Google Scholar]
  115. 115.
    Carr BL, Jahangirifar M, Nicholson AE, Li W, Mol BW, Licqurish S. 2022. Predicting postpartum haemorrhage: a systematic review of prognostic models. Aust. N.Z. J. Obstet. Gynaecol. 62:6813–25
    [Google Scholar]
  116. 116.
    Raeisi-Dehkordi H, Kummer S, Francis Raguindin P, Dejanovic G, Eylul Taneri P et al. 2022. Risk prediction models of natural menopause onset: a systematic review. J. Clin. Endocrinol. Metab. 107:2934–44
    [Google Scholar]
  117. 117.
    Joo YY, Actkins K, Pacheco JA, Basile AO, Carroll R et al. A polygenic and phenotypic risk prediction for polycystic ovary syndrome evaluated by phenome-wide association studies. J. Clin. Endocrinol. Metab. 105:61918–36
    [Google Scholar]
  118. 118.
    Svensson A, Garcia-Etxebarria K, Åkesson A, Borgfeldt C, Roth B et al. 2022. Applicability of polygenic risk scores in endometriosis clinical presentation. BMC Women's Health 22:208
    [Google Scholar]
  119. 119.
    Piekos JA, Hellwege JN, Zhang Y, Torstenson ES, Jarvik GP et al. 2022. Uterine fibroid polygenic risk score (PRS) associates and predicts risk for uterine fibroid. Hum. Genet. 141:111739–48
    [Google Scholar]
  120. 120.
    Dareng EO, Tyrer JP, Barnes DR, Jones MR, Yang X et al. 2022. Polygenic risk modeling for prediction of epithelial ovarian cancer risk. Eur. J. Hum. Genet. 30:349–62
    [Google Scholar]
  121. 121.
    Yanes T, Young M-A, Meiser B, James PA. 2020. Clinical applications of polygenic breast cancer risk: a critical review and perspectives of an emerging field. Breast Cancer Res. 22:21
    [Google Scholar]
  122. 122.
    Lee A, Mavaddat N, Cunningham A, Carver T, Ficorella L et al. 2022. Enhancing the BOADICEA cancer risk prediction model to incorporate new data on RAD51C, RAD51D, BARD1 updates to tumour pathology and cancer incidence. J. Med. Genet. 59:121206–18
    [Google Scholar]
  123. 123.
    Lee A, Mavaddat N, Wilcox AN, Cunningham AP, Carver T et al. 2019. BOADICEA: a comprehensive breast cancer risk prediction model incorporating genetic and nongenetic risk factors. Genet. Med. 21:1708–18
    [Google Scholar]
  124. 124.
    Yang X, Eriksson M, Czene K, Lee A, Leslie G et al. 2022. Prospective validation of the BOADICEA multifactorial breast cancer risk prediction model in a large prospective cohort study. J. Med. Genet. 59:1196–205
    [Google Scholar]
  125. 125.
    Rafnar T, Gunnarsson B, Stefansson OA, Sulem P, Ingason A et al. 2018. Variants associating with uterine leiomyoma highlight genetic background shared by various cancers and hormone-related traits. Nat. Commun. 9:3636
    [Google Scholar]
  126. 126.
    Day F, Karaderi T, Jones MR, Meun C, He C et al. 2018. Large-scale genome-wide meta-analysis of polycystic ovary syndrome suggests shared genetic architecture for different diagnosis criteria. PLOS Genet. 14:e1007813
    [Google Scholar]
  127. 127.
    Adewuyi EO, Mehta D, Int. Endogene Consort., 23andMe Res. Team, Nyholt DR. 2022. Genetic overlap analysis of endometriosis and asthma identifies shared loci implicating sex hormones and thyroid signalling pathways. Hum. Reprod. 37:366–83
    [Google Scholar]
  128. 128.
    Olafsdottir T, Thorleifsson G, Sulem P, Stefansson OA, Medek H et al. 2020. Genome-wide association identifies seven loci for pelvic organ prolapse in Iceland and the UK Biobank. Commun. Biol. 3:129
    [Google Scholar]
  129. 129.
    Steinthorsdottir V, McGinnis R, Williams NO, Stefansdottir L, Thorleifsson G et al. 2020. Genetic predisposition to hypertension is associated with preeclampsia in European and Central Asian women. Nat. Commun. 11:5976
    [Google Scholar]
  130. 130.
    Zhang G, Feenstra B, Bacelis J, Liu X, Muglia LM et al. 2017. Genetic associations with gestational duration and spontaneous preterm birth. N. Engl. J. Med. 377:1156–67
    [Google Scholar]
  131. 131.
    Mirin AA. 2021. Gender disparity in the funding of diseases by the US National Institutes of Health. J. Women's Health 30:956–63
    [Google Scholar]
  132. 132.
    King Thomas J, Mir H, Kapur N, Singh S 2019. Racial differences in immunological landscape modifiers contributing to disparity in prostate cancer. Cancers 11:1857
    [Google Scholar]
  133. 133.
    Oliveira DF, Ma Y, Woodruff TK, Uzzi B. 2019. Comparison of National Institutes of Health grant amounts to first-time male and female principal investigators. JAMA 321:898–900
    [Google Scholar]
  134. 134.
    Hechtman LA, Moore NP, Schulkey CE, Miklos AC, Calcagno AM et al. 2018. NIH funding longevity by gender. PNAS 115:7943–48
    [Google Scholar]
  135. 135.
    Knobloch-Westerwick S, Glynn CJ, Huge M. 2013. The Matilda effect in science communication: an experiment on gender bias in publication quality perceptions and collaboration interest. Sci. Commun. 35:603–25
    [Google Scholar]
/content/journals/10.1146/annurev-biodatasci-020722-105958
Loading
/content/journals/10.1146/annurev-biodatasci-020722-105958
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error