1
|
Narula K, Kenkre JS, Loh WJ, Tan T. Obesity, insulin resistance and fertility: unresolved questions and emerging insights. Curr Opin Endocrinol Diabetes Obes 2025; 32:108-114. [PMID: 40125660 DOI: 10.1097/med.0000000000000907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
PURPOSE OF REVIEW Obesity significantly impacts fertility in women, contributing to hormonal imbalances, ovulatory dysfunction, and poor reproductive outcomes. This is especially pronounced in polycystic ovary syndrome (PCOS), where obesity and insulin resistance exacerbate fertility challenges. Moreover, obesity is a risk factor for type 2 diabetes (T2D) and gestational diabetes (GDM), further complicating reproductive health. Effective weight loss interventions before conception are essential to improve fertility and reduce the risks of adverse perinatal outcomes, such as GDM, hypertensive disorders, and neonatal complications. RECENT FINDINGS Lifestyle modifications, including modest calorie restriction and exercise, improve ovulatory function and pregnancy rates but have limited impact on live-birth rates during fertility treatments. Very low-calorie diets (VLCDs) achieve rapid weight loss but raise concerns about maternal nutrition. Pharmacotherapy offers modest benefits for weight loss and fertility, though teratogenic risks persist. Bariatric surgery often results in significant weight loss and enhanced fertility, yet requires careful timing and management of potential nutrient deficiencies. SUMMARY Weight-loss interventions show promise in addressing obesity-related fertility issues, but long-term outcomes and optimal strategies remain unclear. Further research is needed to bridge these gaps and improve reproductive outcomes following weight reduction.
Collapse
Affiliation(s)
| | - Julia S Kenkre
- Imperial College London Division of Experimental Medicine: Imperial College London Faculty of Medicine, London, UK
| | | | - Tricia Tan
- Imperial College London Division of Experimental Medicine: Imperial College London Faculty of Medicine, London, UK
| |
Collapse
|
2
|
Leathersich S, Roche C, Hart R. Minimising OHSS in women with PCOS. Front Endocrinol (Lausanne) 2025; 16:1507857. [PMID: 40182629 PMCID: PMC11966453 DOI: 10.3389/fendo.2025.1507857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Ovarian hyperstimulation syndrome (OHSS) is a serious iatrogenic complication of ovarian stimulation during in vitro fertilisation (IVF) treatment and is associated with significant morbidity and a small risk of mortality. Women with polycystic ovary syndrome (PCOS) are at a substantially increased risk of developing OHSS compared to those without. This paper reviews the current evidence for strategies to mitigate the risk of OHSS in this patient population. In order to minimise the risk of OHSS, clinicians should identify patients at high risk prior to commencing treatment and provide adequate pre-treatment counselling regarding the risks and benefits of IVF treatment, as well as alternative treatment options. Strategies that can reduce the risk of OHSS include co-treatment with metformin in gonadotropin releasing hormone (GnRH) agonist cycles, use of GnRH antagonist or PPOS protocols, appropriate gonadotropin dosing, the use of a GnRH agonist trigger for oocyte maturation in antagonist or PPOS protocols, cryopreservation of all embryos with deferred frozen embryo transfer, and treatment with dopamine-agonists after oocyte collection. In vitro maturation (IVM) offers an alternative with no risk of OHSS, however currently has a lower cumulative live birth rate than conventional IVF. These strategies can prevent significant early and late OHSS in women with PCOS and should be used to optimise the safety of IVF for this high-risk population, striving for OHSS-free treatment for all patients undergoing IVF.
Collapse
Affiliation(s)
- Sebastian Leathersich
- Department of Reproductive Medicine, King Edward Memorial Hospital, Subiaco, WA, Australia
- City Fertility Australia, Claremont, WA, Australia
- Fertility Specialists of Western Australia, Claremont, WA, Australia
- Dexeus Fertility, Dexeus University Hospital, Barcelona, Spain
| | - Caitlin Roche
- Department of Reproductive Medicine, King Edward Memorial Hospital, Subiaco, WA, Australia
| | - Roger Hart
- Department of Reproductive Medicine, King Edward Memorial Hospital, Subiaco, WA, Australia
- City Fertility Australia, Claremont, WA, Australia
- Fertility Specialists of Western Australia, Claremont, WA, Australia
- Division of Obstetrics and Gynaecology, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
3
|
Ma J, Wang M, Zuo Q, Ma H, Wu S. Analysis of use of different rFSHs during IVF/ICSI-assisted conception in elderly population and effect of double trigger on clinical outcomes. J Matern Fetal Neonatal Med 2024; 37:2352790. [PMID: 38777799 DOI: 10.1080/14767058.2024.2352790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE To compare the number of oocytes retrieved and clinical outcomes of ovulation induction in an older population treated with in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) (IVF/ICSI) using different rFSH options and the effectiveness of antagonist treatment to induce ovulation using gonadotropin-releasing hormone agonists (GnRH-a) in combination with an human chorionic gonadotropin (HCG) trigger. METHODS A total of 132 fresh cycles were selected for this study, which were treated with IVF/ICSI in our hospital from March 2022 to December 2022. Observations were made according to different subgroups and the effects of different triggering methods on the number of oocytes obtained, embryo quality, and clinical outcomes. RESULTS The initial gonadotropin (Gn) dose, the number of oocytes, and the number of MII oocytes were higher in group A than in group B (p < .05), and the clinical pregnancy rate was 29.41% in group A. Group B had a clinical pregnancy rate of 27.5%. The double-trigger group was superior to the HCG-trigger group in terms of the number of 2PN, the number of viable embryos, and the number of high-quality embryos (p < .05). The use of a double-trigger regimen (OR = 0.667, 95%CI (0.375, 1.706), p = .024) was a protective factor for the clinical pregnancy rate, whereas AFC (OR = 0.925, 95%CI (0.867, 0.986), p = .017) was an independent factor for the clinical pregnancy rate. CONCLUSIONS The use of a dual-trigger regimen of GnRH-a in combination with HCG using an appropriate antagonist improves pregnancy outcomes in fresh embryo transfer cycles in older patients.
Collapse
Affiliation(s)
- Jianxin Ma
- Department of Reproductive Medicine, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou City, China
| | - Mengna Wang
- Department of Reproductive Medicine, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou City, China
| | - Qianqian Zuo
- Department of Reproductive Medicine, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou City, China
| | - Hong Ma
- Department of Reproductive Medicine, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou City, China
| | - Shangqing Wu
- Department of Reproductive Medicine, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou City, China
| |
Collapse
|
4
|
Neola D, Angelino A, Sirico A, Murolo C, Bartolini G, Vigilante L, Raffone A, Carbone L, Sarno L, Saccone G, Guida M, Maruotti GM. Unveiling therapeutic potentials and exploring maternal-fetal health benefits of metformin in pregnancy: A scoping review. Int J Gynaecol Obstet 2024; 167:538-546. [PMID: 38887906 DOI: 10.1002/ijgo.15728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/20/2024] [Accepted: 06/01/2024] [Indexed: 06/20/2024]
Abstract
This scoping review synthesizes evidence on metformin's use during pregnancy, encompassing diverse conditions like gestational diabetes, type 1 and type 2 diabetes, polycystic ovary syndrome (PCOS), and obesity. Metformin demonstrates comparable efficacy to insulin in gestational diabetes, positive outcomes in type 2 diabetes pregnancies, and potential benefits in reducing complications. The review highlights nuances in its effects across conditions, indicating advantages such as reduced risk of macrosomia and cesarean section in gestational diabetes. However, its prophylactic role in preventing gestational diabetes and associated complications remains inconclusive. In obese pregnant women, mixed results are observed, with potential benefits in reducing pre-eclampsia risk. Metformin shows promise in preventing preterm birth and late miscarriage in PCOS pregnancies. Categorizing patient subgroups is crucial for identifying advantages, especially in gestational diabetes and type 2 diabetes. Challenges arise from study heterogeneity, necessitating standardized indications for dosage, timing, and postpartum follow ups. Efforts to identify patient characteristics influencing metformin efficacy are crucial for tailored therapy. Although metformin emerges as a viable option in complicated pregnancies, comprehensive research, standardized protocols, and subgroup identification efforts will enhance clinical utility, ensuring evidence-based therapies and optimal maternal and fetal outcomes. Bridging existing knowledge gaps remains imperative for advancing metformin's role in pregnancy management.
Collapse
Affiliation(s)
- Daniele Neola
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Antonio Angelino
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Angelo Sirico
- Obstetrics and Gynecology Unit, Sant'Anna e San Sebastiano Hospital, Caserta, Italy
| | - Chiara Murolo
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Giorgia Bartolini
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Luigi Vigilante
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Antonio Raffone
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Luigi Carbone
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Laura Sarno
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Gabriele Saccone
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Maurizio Guida
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
5
|
Jalil AT, Zair MA, Hanthal ZR, Naser SJ, Aslandook T, Abosaooda M, Fadhil A. Role of the AMP-Activated Protein Kinase in the Pathogenesis of Polycystic Ovary Syndrome. Indian J Clin Biochem 2024; 39:450-458. [PMID: 39346714 PMCID: PMC11436500 DOI: 10.1007/s12291-023-01139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/09/2023] [Indexed: 10/01/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a complex disorder characterized by elevated androgen levels, menstrual irregularities, and polycystic morphology of the ovaries. Affecting 6-10% of women in childbearing age, PCOS is a leading cause of infertility worldwide. In recent years, there has been a growing acknowledgment of the involvement of adenosine monophosphate-activated protein kinase (AMPK) in the development of polycystic ovary syndrome (PCOS). The expression of AMPK is diminished in polycystic ovaries, and when AMPK is silenced in human granulosa cells, there is a rise in the expression of steroidogenic enzymes, resulting in increased production of estradiol and progesterone. Additionally, in mouse models, the inhibiting AMPK intensifies the polycystic appearance of ovaries and impairs the process of ovulation. Moreover, it has been shown that AMPK activators like metformin and resveratrol ameliorate PCOS associated signs and symptoms in experimental and clinical studies. These findings, collectively, indicate the key role of AMPK in the pathogenesis of PCOS. Understanding the role of AMPK in PCOS will offer rewarding information on details of PCOS pathogenesis and will provide novel more specific therapeutic approaches. The present review summarizes the latest findings regarding the role of AMPK in PCOS obtained in experimental and clinical studies.
Collapse
Affiliation(s)
- Abduldaheem Turki Jalil
- Department of Medical Laboratories Techniques, Al-Mustaqbal University College, Hilla, Babylon Iraq
| | - Mahdi Abd Zair
- Department of Pharmacy, Kut University College, Kut, Wasit Iraq
| | | | - Sarmad Jaafar Naser
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Tahani Aslandook
- Department of Dentistry, Al-Turath University College, Baghdad, Iraq
| | - Munther Abosaooda
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Ali Fadhil
- Medical Laboratory Technology Department, College of Medical Technology, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
6
|
Gullo G, Cucinella G, Stojanovic V, Stojkovic M, Bruno C, Streva AV, Lopez A, Perino A, Marinelli S. Ovarian Hyperstimulation Syndrome (OHSS): A Narrative Review and Legal Implications. J Pers Med 2024; 14:915. [PMID: 39338169 PMCID: PMC11433561 DOI: 10.3390/jpm14090915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Infertility is a highly meaningful issue with potentially life-changing consequences, and its incidence has been growing worldwide. Assisted reproductive technology (ART) has made giant strides in terms of treating many infertility conditions, despite the risk of developing ovarian hyperstimulation syndrome (OHSS), a potentially life-threatening complication. METHODS This narrative review draws upon scientific articles found in the PubMed database. The search spanned the 1990-2024 period. Search strings used included "OHSS" or "ovarian hyperstimulation" and "IVF" and "GnRH" and "hCG"; 1098 results were retrieved and were ultimately narrowed down to 111 suitable sources, i.e., relevant articles dealing with the condition's underlying dynamics, management pathways, and evidence-based criteria and guidelines, crucial both from a clinical perspective and from the standpoint of medicolegal tenability. RESULTS The following features constitute OHSS risk factors: young age, low body weight, and polycystic ovarian syndrome (PCOS), among others. GnRH antagonist can substantially lower the risk of severe OHSS, compared to the long protocol with a gonadotropin-releasing hormone (GnRH) agonist. However, a mild or moderate form of OHSS is also possible if the antagonist protocol is used, especially when hCG is used for the final maturation of oocytes. For women at risk of OHSS, GnRH agonist trigger and the freeze-all strategy is advisable. OHSS is one of the most frequent complications, with a 30% rate in IVF cycles. CONCLUSION Providing effective care for OHSS patients begins with early diagnosis, while also evaluating for comorbidities and complications. In addition to that, we should pay more attention to the psychological component of this complication and of infertility as a whole. Compliance with guidelines and evidence-based best practices is essential for medicolegal tenability.
Collapse
Affiliation(s)
- Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Gaspare Cucinella
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Vukasin Stojanovic
- Emergency Medicine Center of Montenegro, Faculty of Medicine, University of Montenegro, 81000 Podgorica, Montenegro;
| | - Mirjana Stojkovic
- Clinic of Endocrinology, Diabetes and Metabolic Disorders, University Clinical Center of Serbia, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Carmine Bruno
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Istituto Dermopatico dell’Immacolata (IDI IRCCS), 00167 Rome, Italy
| | - Adriana Vita Streva
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Alessandra Lopez
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Antonio Perino
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF Unit, University of Palermo, 90146 Palermo, Italy; (G.C.); (A.V.S.); (A.L.); (A.P.)
| | - Susanna Marinelli
- School of Law, Polytechnic University of Marche, 60121 Ancona, Italy;
| |
Collapse
|
7
|
Al-Thamarani S, Gad S, Abdel Fattah IO, Hammadi SH, Hammady TM. Comparative analysis of oral and local intraovarian administration of metformin and nanoparticles (NPs11) in alleviating testosterone-induced polycystic ovary syndrome in rats. Tissue Cell 2024; 88:102394. [PMID: 38663112 DOI: 10.1016/j.tice.2024.102394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 06/17/2024]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine and metabolic dysfunction. This study aims to compare the oral and local treatments of metformin or its nanoparticles (NPs11) for ameliorating PCOS in rats. Rats were divided into 4 groups: the control group with no drug treatment; the PCOS group, where subcutaneous testosterone was given (10 mg/kg/day) for 28 days; the MET group, where metformin was administered orally or locally; and the NP group, where metformin NPs11 were also administered orally or locally. Oral administrations were for 21 days, while local injection was performed once surgically. After 7 weeks, all rats were sacrificed; blood glucose and serum hormonal levels and lipid profile were estimated, and the ovaries were assessed by histopathological, Ki-67 immunohistochemical, and histomorphometric evaluations. Blood glucose levels were significantly decreased in groups of orally administered metformin or NPs11 only, while the most efficient option for modulating PCOS-induced hormonal and lipid profile changes was intraovarian injection of NPs11. The ovaries of PCOS rats demonstrated large follicular cysts, massive collagen depositions, and attenuated Ki-67 immunoexpression. Also, the PCOS group revealed a significant decrease in the count of all stages of growing follicles, corpora lutea, granulosa cell layer thickness, and surface area of corpora lutea, in addition to an increase in the number of atretic follicles and follicular cysts, theca cell layer thickness, and surface area of the follicular cysts. All these parameters were recovered with metformin or their NPs11 treatments in different degrees, while local injection of NPs11 was the best option.
Collapse
Affiliation(s)
- Sadeq Al-Thamarani
- Department of Pharmacy, Faculty of Medicine and Health Sciences, Thamar University, Dhamar 87246, Yemen
| | - Shadeed Gad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Islam Omar Abdel Fattah
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Sami H Hammadi
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Taha M Hammady
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
8
|
Xu Q, Zhang J, Lu Y, Wu L. Association of metabolic-dysfunction associated steatotic liver disease with polycystic ovary syndrome. iScience 2024; 27:108783. [PMID: 38292434 PMCID: PMC10825666 DOI: 10.1016/j.isci.2024.108783] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), which has a prevalence of over 25% in adults, encompasses a wide spectrum of liver diseases. Metabolic-dysfunction associated steatotic liver disease (MASLD), the new term for NAFLD, is characterized by steatotic liver disease accompanied by cardiometabolic criteria, showing a strong correlation with metabolic diseases. Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease affecting 4-21% of women of reproductive age. Numerous studies have indicated that NAFLD and PCOS often occur together. However, as MASLD is a new term, there is still a lack of reports describing the effects of MASLD on the development of PCOS. In this review article, we have summarized the complex and multifaceted connections between MASLD and PCOS. Understanding the pathogenesis and treatment methods could not only guide the clinical prevention, diagnosis, and treatment of PCOS in patients with MASLD, but also increase the clinical attention of reproductive doctors to MASLD.
Collapse
Affiliation(s)
- Qiuyu Xu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Feferkorn I, Santos-Ribeiro S, Ubaldi FM, Velasco JG, Ata B, Blockeel C, Conforti A, Esteves SC, Fatemi HM, Gianaroli L, Grynberg M, Humaidan P, Lainas GT, La Marca A, Craig LB, Lathi R, Norman RJ, Orvieto R, Paulson R, Pellicer A, Polyzos NP, Roque M, Sunkara SK, Tan SL, Urman B, Venetis C, Weissman A, Yarali H, Dahan MH. The HERA (Hyper-response Risk Assessment) Delphi consensus for the management of hyper-responders in in vitro fertilization. J Assist Reprod Genet 2023; 40:2681-2695. [PMID: 37713144 PMCID: PMC10643792 DOI: 10.1007/s10815-023-02918-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/15/2023] [Indexed: 09/16/2023] Open
Abstract
PURPOSE To provide agreed-upon guidelines on the management of a hyper-responsive patient undergoing ovarian stimulation (OS) METHODS: A literature search was performed regarding the management of hyper-response to OS for assisted reproductive technology. A scientific committee consisting of 4 experts discussed, amended, and selected the final statements. A priori, it was decided that consensus would be reached when ≥66% of the participants agreed, and ≤3 rounds would be used to obtain this consensus. A total of 28/31 experts responded (selected for global coverage), anonymous to each other. RESULTS A total of 26/28 statements reached consensus. The most relevant are summarized here. The target number of oocytes to be collected in a stimulation cycle for IVF in an anticipated hyper-responder is 15-19 (89.3% consensus). For a potential hyper-responder, it is preferable to achieve a hyper-response and freeze all than aim for a fresh transfer (71.4% consensus). GnRH agonists should be avoided for pituitary suppression in anticipated hyper-responders performing IVF (96.4% consensus). The preferred starting dose in the first IVF stimulation cycle of an anticipated hyper-responder of average weight is 150 IU/day (82.1% consensus). ICoasting in order to decrease the risk of OHSS should not be used (89.7% consensus). Metformin should be added before/during ovarian stimulation to anticipated hyper-responders only if the patient has PCOS and is insulin resistant (82.1% consensus). In the case of a hyper-response, a dopaminergic agent should be used only if hCG will be used as a trigger (including dual/double trigger) with or without a fresh transfer (67.9% consensus). After using a GnRH agonist trigger due to a perceived risk of OHSS, luteal phase rescue with hCG and an attempt of a fresh transfer is discouraged regardless of the number of oocytes collected (72.4% consensus). The choice of the FET protocol is not influenced by the fact that the patient is a hyper-responder (82.8% consensus). In the cases of freeze all due to OHSS risk, a FET cycle can be performed in the immediate first menstrual cycle (92.9% consensus). CONCLUSION These guidelines for the management of hyper-response can be useful for tailoring patient care and for harmonizing future research.
Collapse
Affiliation(s)
- I Feferkorn
- IVF Unit, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | | | - F M Ubaldi
- GeneraLife Centers for Reproductive Medicine, Rome, Italy
| | | | - B Ata
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Koc University School of Medicine, Istanbul, Turkey
- ART Fertility Clinics, Dubai, United Arab Emirates
| | - C Blockeel
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Jette, Belgium
| | - A Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - S C Esteves
- ANDROFERT, Andrology and Human Reproduction Clinic, Av. Dr. Heitor Penteado 1464, Campinas, SP, 13075-460, Brazil
- Department of Surgery (Division of Urology), University of Campinas (UNICAMP), Campinas, SP, Brazil
- Faculty of Health, Aarhus University, C, 8000, Aarhus, Denmark
| | - H M Fatemi
- ART Fertility Clinics, Abu Dhabi, United Arab Emirates
| | - L Gianaroli
- Società Italiana Studi di Medicina della Riproduzione, S.I.S.Me.R. Reproductive Medicine Institute, Bologna, Emilia-Romagna, Italy
| | - M Grynberg
- Department of Reproductive Medicine, Hôpital Antoine-Béclère, University Paris-Sud (Paris XI), Le Kremlin-Bicêtre, Clamart, France
| | - P Humaidan
- The Fertility Clinic, Skive Regional Hospital, Faculty of Health, Aarhus University, Resenvej 25, 7800, Skive, Denmark
| | | | - A La Marca
- Obstetrics, Gynecology and Reproductive Medicine, University of Modena and Reggio Emilia, Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy
| | - L B Craig
- Section of Reproductive Endocrinology & Infertility, Department of Obstetrics & Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - R Lathi
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| | - R J Norman
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
- FertilitySA, Adelaide, South Australia, Australia
- Monash Centre for Health Research and Implementation MCHRI, Monash University, Melbourne, Australia
- NHMRC Centre of Research Excellence in Women's Health in Reproductive Life (CRE-WHiRL), Melbourne, Australia
| | - R Orvieto
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (Tel Hashomer), Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Tarnesby-Tarnowski Chair for Family Planning and Fertility Regulation, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - R Paulson
- University of Southern California, Los Angeles, CA, 90033, USA
| | - A Pellicer
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- IVI Roma Parioli, IVI-RMA Global, Rome, Italy
| | - N P Polyzos
- Department of Reproductive Medicine, Dexeus Mujer, Hospital Universitario Dexeus, Barcelona, Spain
| | - M Roque
- Department of Reproductive Medicine, ORIGEN-Center for Reproductive Medicine, Rio de Janeiro, RJ, Brazil
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - S K Sunkara
- Department of Women and Children's Health, King's College London, London, UK
| | - S L Tan
- OriginElle Fertility Clinic 2110 Boul. Decarie, Montreal, QC, Canada
| | - B Urman
- Department of Obstetrics and Gynecology and Assisted Reproduction, American Hospital, Istanbul, Koc University School of Medicine, Istanbul, Turkey
| | - C Venetis
- Unit for Human Reproduction, 1st Dept of OB/Gyn, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Centre for Big Data Research in Health, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
- Virtus Health, Sydney, Australia
| | - A Weissman
- In Vitro Fertilization Unit, Department of Obstetrics and Gynecology, Edith Wolfson Medical Center, Holon, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - H Yarali
- Division of Reproductive Endocrinology and Infertility, Dept. of Obstetrics and Gynecology, Hacettepe University, School of Medicine, Anatolia IVF and Women's Health Center, Ankara, Turkey
| | - M H Dahan
- Division of Reproductive Endocrinology and Infertility, McGill University Health Care Center, 888 Boul. de Maisonneuve E #200, Montreal, QC, H2L 4S8, Canada
| |
Collapse
|
10
|
Wang C, Yu J, Ding C, Chen C. CangFu Daotan decoction improves polycystic ovarian syndrome by downregulating FOXK1. Gynecol Endocrinol 2023; 39:2244600. [PMID: 37544927 DOI: 10.1080/09513590.2023.2244600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/09/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
Objective: Polycystic ovarian syndrome (PCOS) is a prevalent gynecologic disorder, often associated with abnormal follicular development. Cangfu Daotan decoction (CFD) is a traditional Chinese medicine formula that is effective in alleviating PCOS clinically, but the specific mechanism remains unclear. Forkhead box K1 (FOXK1) is associated with cellular function. This study aimed to explore the effects of CFD and FOXK1 on PCOS.Methods: High-fat diet and letrozole were combined to establish PCOS rat models. Next, primary GCs were extracted from those PCOS rats. Then, GC cells were transfected with si-FOXK1 or oe-FOXK1. CFD-contain serum was prepared, and experiments were conducted to investigate the regulation of FOXK1 by CFD.Results: FOXK1 was highly expressed in GCs of PCOS rats. Further investigation revealed that FOXK1 overexpression resulted in inhibition of proliferation and DNA synthesis, along with promotion of apoptosis and autophagy in GCs. Additionally, it was found that FOXK1 promoted the expressions of the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway-related proteins. Interestingly, treatment with CFD reversed all the effects of FOXK1 overexpression in GCs. Conclusion: This study demonstrated that CFD exerted a protective role in PCOS by inhibiting FOXK1, which provided a research basis for the application of CFD in PCOS, and suggested that FOXK1 is a novel therapeutic target in PCOS treatment.
Collapse
Affiliation(s)
- Chenye Wang
- Department of Reproductive Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou City, China
| | - Jia Yu
- Department of Reproductive Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou City, China
| | - Caifei Ding
- Department of Reproductive Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou City, China
| | - Chunyue Chen
- Department of Reproductive Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou City, China
| |
Collapse
|
11
|
Kanda S, Chatha U, Odoma VA, Pitliya A, AlEdani EM, Bhangu JK, Javed K, Manshahia PK, Nahar S, Hamid P. Effect of Metformin (MTF) Intervention During Pregnancy in Women With Polycystic Ovarian Syndrome (PCOS): A Systematic Review. Cureus 2023; 15:e44166. [PMID: 37753037 PMCID: PMC10519644 DOI: 10.7759/cureus.44166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
Metformin (MTF) is a commonly prescribed medication for women with polycystic ovarian syndrome (PCOS), but its impact on pregnancy outcomes in women with PCOS remains controversial. This systematic review aims to evaluate the effects of MTF intervention on pregnancy outcomes in women with PCOS and the impact of MTF on offspring. A comprehensive search is conducted in PubMed, Google Scholar, and ScienceDirect databases from 2019 up to May 16, 2023. Only review articles and meta-analyses are included, focusing on women with PCOS who received MTF during pregnancy or as part of infertility treatment. The primary outcomes of interest are clinical pregnancy rate (CPR), miscarriage rate, preterm birth rate, and live birth rate. Secondary outcomes are the safety profile of MTF. Data extraction and quality assessment are performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and the assessment using the multiple systematic reviews (AMSTAR) 2 tool, respectively. The initial search produced 1877 studies. Thirteen studies were included in the review. While the use of MTF during pregnancy in women with PCOS may have some benefits in reducing certain pregnancy complications such as pregnancy-induced hypertension (PIH), gestational diabetes mellitus (GDM), preeclampsia, preterm delivery, reducing the risk of ovarian hyperstimulation syndrome (OHSS) in women with PCOS undergoing in vitro fertilization (IVF); however, there is no significant difference in clinical pregnancy and live birth rates overall, but subgroup analysis suggests potential benefits for women with a higher body mass index (BMI). MTF is associated with a larger fetal head circumference and potential long-term effects on offspring's BMI and obesity. Further research is needed to better understand the optimal dosing of MTF, long-term effects, and effects in specific subgroups. The heterogeneity of the included studies limited the ability to analyze the data effectively, leading to challenges in drawing definitive conclusions.
Collapse
Affiliation(s)
- Srishti Kanda
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Uzair Chatha
- Medicine, Lahore Medical and Dental College, Lahore, PAK
| | - Victor A Odoma
- Cardiology/Oncology, Indiana University (IU) Health, Bloomington, USA
| | - Aakanksha Pitliya
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Esraa M AlEdani
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Japneet K Bhangu
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Khalid Javed
- Anesthesiology, Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | - Shamsun Nahar
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Pousette Hamid
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
12
|
Peng Y, Guo R, Shi B, Li D. The role of long non-coding RNA H19 in infertility. Cell Death Discov 2023; 9:268. [PMID: 37507391 PMCID: PMC10382492 DOI: 10.1038/s41420-023-01567-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Infertility is defined as the failure to conceive after at least one year of unprotected intercourse. Long non-coding RNAs (lncRNAs) are transcripts that contain more than 200 nucleotides but do not convert into proteins. LncRNAs, particularly lncRNA H19, have been linked to the emergence and progression of various diseases. This review focuses on the role of H19 in infertility caused by polycystic ovary syndrome, endometriosis, uterine fibroids, diminished ovarian reserve, male factor, and assisted reproductive technology-related pathology, highlighting the potential of H19 as a molecular target for the future treatment of infertility.
Collapse
Affiliation(s)
- Yuanyuan Peng
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China
| | - Renhao Guo
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Bei Shi
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China.
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, 110004, China.
| |
Collapse
|
13
|
Palomba S, Costanzi F, Nelson SM, Caserta D, Humaidan P. Interventions to prevent or reduce the incidence and severity of ovarian hyperstimulation syndrome: a systematic umbrella review of the best clinical evidence. Reprod Biol Endocrinol 2023; 21:67. [PMID: 37480081 PMCID: PMC10360244 DOI: 10.1186/s12958-023-01113-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/21/2023] [Indexed: 07/23/2023] Open
Abstract
Ovarian hyperstimulation syndrome (OHSS) is a potentially life-threating iatrogenic complication of the early luteal phase and/or early pregnancy after in vitro fertilization (IVF) treatment. The aim of the current study was to identify the most effective methods for preventing of and reducing the incidence and severity of OHSS in IVF patients. A systematic review of systematic reviews of randomized controlled trials (RCTs) with meta-analysis was used to assess each potential intervention (PROSPERO website, CRD 268626) and only studies with the highest quality were included in the qualitative analysis. Primary outcomes included prevention and reduction of OHSS incidence and severity. Secondary outcomes were maternal death, incidence of hospital admission, days of hospitalization, and reproductive outcomes, such as incidence of live-births, clinical pregnancies, pregnancy rate, ongoing pregnancy, miscarriages, and oocytes retrieved. A total of specific interventions related to OHSS were analyzed in 28 systematic reviews of RCTs with meta-analyses. The quality assessment of the included studies was high, moderate, and low for 23, 2, and 3 studies, respectively. The certainty of evidence (CoE) for interventions was reported for 37 specific situations/populations and resulted high, moderate, and low-to-very low for one, 5, and 26 cases, respectively, while it was not reported in 5 cases. Considering the effective interventions without deleterious reproductive effects, GnRH-ant co-treatment (36 RCTs; OR 0.61, 95% C 0.51 to 0.72, n = 7,944; I2 = 31%) and GnRH agonist triggering (8 RCTs; OR 0.15, 95% CI 0.05 to 0.47, n = 989; I2 = 42%) emerged as the most effective interventions for preventing OHSS with a moderate CoE, even though elective embryo cryopreservation exhibited a low CoE. Furthermore, the use of mild ovarian stimulation (9 RCTs; RR 0.26, CI 0.14 to 0.49, n = 1,925; I2 = 0%), and dopaminergic agonists (10 RCTs; OR 0.32, 95% CI 0.23 to 0.44, n = 1,202; I2 = 13%) coadministration proved effective and safe with a moderate CoE. In conclusion, the current study demonstrates that only a few interventions currently can be considered effective to reduce the incidence of OHSS and its severity with high/moderate CoE despite the numerous published studies on the topic. Further well-designed RCTs are needed, particularly for GnRH-a down-regulated IVF cycles.
Collapse
Affiliation(s)
- Stefano Palomba
- Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome, Sant'Andrea Hospital, via di Grottarossa, n. 1035/1039, Rome, 00189, Italy.
| | - Flavia Costanzi
- Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome, Sant'Andrea Hospital, via di Grottarossa, n. 1035/1039, Rome, 00189, Italy
| | - Scott M Nelson
- School of Medicine, University of Glasgow, Glasgow, UK
- NIHR Bristol Biomedical Research Centre, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
- TFP, Oxford Fertility, Institute of Reproductive Sciences, Oxford, UK
| | - Donatella Caserta
- Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome, Sant'Andrea Hospital, via di Grottarossa, n. 1035/1039, Rome, 00189, Italy
| | - Peter Humaidan
- The Fertility Clinic, Faculty of Health, Skive Regional Hospital, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
14
|
Tosti G, Barberio A, Tartaglione L, Rizzi A, Di Leo M, Viti L, Sirico A, De Carolis S, Pontecorvi A, Lanzone A, Pitocco D. Lights and shadows on the use of metformin in pregnancy: from the preconception phase to breastfeeding and beyond. Front Endocrinol (Lausanne) 2023; 14:1176623. [PMID: 37409227 PMCID: PMC10319127 DOI: 10.3389/fendo.2023.1176623] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023] Open
Abstract
During pregnancy, the complex hormonal changes lead to a progressive decrease of insulin sensitivity that can drive the onset of gestational diabetes (GDM) or worsen an already-known condition of insulin resistance like type 2 diabetes, polycystic ovarian syndrome (PCOS), and obesity, with complications for the mother and the fetus. Metformin during pregnancy is proving to be safe in a growing number of studies, although it freely crosses the placenta, leading to a fetal level similar to maternal concentration. The aim of this literature review is to analyze the main available evidence on the use of metformin during, throughout, and beyond pregnancy, including fertilization, lactation, and medium-term effects on offspring. Analyzed studies support the safety and efficacy of metformin during pregnancy. In pregnant women with GDM and type 2 diabetes, metformin improves obstetric and perinatal outcomes. There is no evidence that it prevents GDM in women with pregestational insulin resistance or improves lipid profile and risk of GDM in pregnant women with PCOS or obesity. Metformin could have a role in reducing the risk of preeclampsia in pregnant women with severe obesity, the risk of late miscarriages and preterm delivery in women with PCOS, and the risk of ovarian hyperstimulation syndrome, increasing the clinical pregnancy rate in women with PCOS undergoing in vitro fertilization (IVF/FIVET). Offspring of mothers with GDM exposed to metformin have no significant differences in body composition compared with insulin treatment, while it appears to be protective for metabolic and cardiovascular risk.
Collapse
Affiliation(s)
- Giulia Tosti
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Annarita Barberio
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Linda Tartaglione
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Alessandro Rizzi
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Mauro Di Leo
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Luca Viti
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| | - Angelo Sirico
- Catholic University School of Medicine, Rome, Italy
- Department of Woman and Child Health, Woman Health Area Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Sara De Carolis
- Catholic University School of Medicine, Rome, Italy
- Department of Woman and Child Health, Woman Health Area Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Alfredo Pontecorvi
- Catholic University School of Medicine, Rome, Italy
- Department of Endocrinology, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonio Lanzone
- Catholic University School of Medicine, Rome, Italy
- Department of Woman and Child Health, Woman Health Area Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Dario Pitocco
- Diabetes Care Unit, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Catholic University School of Medicine, Rome, Italy
| |
Collapse
|
15
|
Liu J, Zhang M, Deng D, Zhu X. The function, mechanisms, and clinical applications of metformin: potential drug, unlimited potentials. Arch Pharm Res 2023; 46:389-407. [PMID: 36964307 DOI: 10.1007/s12272-023-01445-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 03/08/2023] [Indexed: 03/26/2023]
Abstract
Metformin has been used clinically for more than 60 years. As time goes by, more and more miraculous effects of metformin beyond the clinic have been discovered and discussed. In addition to the clinically approved hypoglycemic effect, it also has a positive metabolic regulation effect on the human body that cannot be ignored. Such as anti-cancer, anti-aging, brain repair, cardiovascular protection, gastrointestinal regulation, hair growth and inhibition of thyroid nodules, and other nonclinical effects. Metformin affects almost the entire body in the situation taking it over a long period, and the preventive effects of metformin in addition to treating diabetes are also beginning to be recommended in some guidelines. This review is mainly composed of four parts: the development history of metformin, the progress of clinical efficacy, the nonclinical efficacy of metformin, and the consideration and prospect of its application.
Collapse
Affiliation(s)
- Jianhong Liu
- Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Ming Zhang
- Department of Physical Medicine and Rehabilitation, Zibo Central Hospital, Zibo, China
| | - Dan Deng
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China.
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China.
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China.
| |
Collapse
|
16
|
Naseri A, Sanaie S, Hamzehzadeh S, Seyedi-Sahebari S, Hosseini MS, Gholipour-Khalili E, Rezazadeh-Gavgani E, Majidazar R, Seraji P, Daneshvar S, Rezazadeh-Gavgani E. Metformin: new applications for an old drug. J Basic Clin Physiol Pharmacol 2023; 34:151-160. [PMID: 36474458 DOI: 10.1515/jbcpp-2022-0252] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022]
Abstract
Metformin is a biguanide, evolved as one of the most widely used medicines. The applications of this component include but are not limited to reducing blood glucose, weight loss, and polycystic ovary syndrome. Studies about other probable indications have emerged, indicating that this agent can also be utilized for other purposes. In this review, applications of metformin are noticed based on the current evidence. Metformin commonly is used as an off-label drug in non-alcoholic fatty liver disease (NAFLD), but it worsens inflammation and should not be used for this purpose, according to the latest research. Metformin decreased the risk of death in patients with liver cirrhosis. It is an effective agent in the prevention and improvement of survival in patients suffering hepatocellular carcinoma. There is evidence of the beneficial effects of metformin in colorectal cancer, early-stage prostate cancer, breast cancer, urothelial cancer, blood cancer, melanoma, and bone cancer, suggesting metformin as a potent anti-tumor agent. Metformin shows neuroprotective effects and provides a potential therapeutic benefit for mild cognitive impairment and Alzheimer's disease (AD). It also has been shown to improve mental function and reduce the incidence of dementia. Another condition that metformin has been shown to slow the progression of is Duchenne muscular dystrophy. Regarding infectious diseases, tuberculosis (TB) and coronavirus disease (COVID-19) are among the conditions suggested to be affected by metformin. The beneficial effects of metformin in cardiovascular diseases were also reported in the literature. Concerning renal function, studies showed that daily oral administration of metformin could ameliorate kidney fibrosis and normalize kidney structure and function. This study reviewed the clinical and preclinical evidence about the possible benefits of metformin based on recent studies. Numerous questions like whether these probable indications of metformin can be observed in non-diabetics, need to be described by future basic experiments and clinical studies.
Collapse
Affiliation(s)
- Amirreza Naseri
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute (JBI) Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sarvin Sanaie
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Hamzehzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | - Ehsan Rezazadeh-Gavgani
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute (JBI) Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Majidazar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parya Seraji
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Daneshvar
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute (JBI) Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
17
|
Polycystic Ovary Syndrome: Challenges and Possible Solutions. J Clin Med 2023; 12:jcm12041500. [PMID: 36836035 PMCID: PMC9967025 DOI: 10.3390/jcm12041500] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in women of reproductive age. This syndrome not only impairs female fertility but also increases the risk of obesity, diabetes, dyslipidemia, cardiovascular diseases, psychological diseases, and other health problems. Additionality, because of the high clinical heterogeneity, the current pathogenesis of PCOS is still unclear. There is still a large gap in precise diagnosis and individualized treatment. We summarize the present findings concerning the genetics, epigenetics, gut microbiota, corticolimbic brain responses, and metabolomics of the PCOS pathogenesis mechanism, highlight the remaining challenges in PCOS phenotyping and potential treatment approaches, and explain the vicious circle of intergenerational transmission of PCOS, which might provide more thoughts for better PCOS management in the future.
Collapse
|
18
|
Ovulation induction using sequential letrozole/gonadotrophin in infertile women with PCOS: a randomized controlled trial. Reprod Biomed Online 2023; 46:352-361. [PMID: 36566146 DOI: 10.1016/j.rbmo.2022.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 02/07/2023]
Abstract
RESEARCH QUESTION Is sequential letrozole/human menopausal gonadotrophin (HMG) superior to letrozole alone in ovulation induction and pregnancy promotion among infertile women with polycystic ovary syndrome (PCOS)? DESIGN This open-label randomized controlled trial comparing sequential letrozole/HMG and letrozole alone included 174 participants enrolled from August 2019 to January 2020 at the Union Hospital of Tongji Medical College, Huazhong University of Science and Technology. Infertile women aged between 18 and 40 years who met Rotterdam criteria for PCOS and without other known causes of infertility were selected for this study. Patients were randomly assigned at a 1:1 ratio to receive 2.5 mg letrozole on cycle days 3-7 (n = 87) or 2.5 mg letrozole on cycle days 3-7 with a sequential injection of 75 IU HMG on cycle days 8-10 for one treatment cycle (n = 87). The pregnancy outcome was recorded after one treatment cycle. RESULTS Women receiving sequential treatment achieved a significantly higher ovulation rate than those in the letrozole group (90.8% versus 70.1%, P = 0.001) and the live birth rate of the sequential group was significantly higher than that of the letrozole protocol (23.0% versus 10.3%, P = 0.025); there was no statistical variation with respect to adverse events. CONCLUSIONS The data suggest that the sequential letrozole/HMG protocol may be superior to the letrozole alone protocol in terms of ovulation induction and pregnancy promotion among infertile women with PCOS.
Collapse
|
19
|
Gao R, Qin L, Li Z, Min W. The homeostasis model assessment of insulin resistance is a judgment criterion for metformin pre-treatment before IVF/ICSI and embryo transfer cycles in patients with polycystic ovarian syndrome. Front Endocrinol (Lausanne) 2023; 14:1106276. [PMID: 36843612 PMCID: PMC9946957 DOI: 10.3389/fendo.2023.1106276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
PURPOSE The aim of this study was to explore the value of the homeostasis model assessment of IR (HOMA-IR) as a judgment criterion for metformin pre-treatment before in vitro fertilization/intracellular sperm injection (IVF/ICSI) and embryo transfer (ET) for polycystic ovarian syndrome (PCOS) patients. MATERIALS AND METHODS The clinical and laboratory information of PCOS patients who received IVF/ICSI-ET from January 2017 to September 2021 was retrospectively analyzed. We compared the clinical pregnancy rate (primary outcome) and controlled ovarian stimulation (COS)-related parameters (secondary outcomes) between patients with and without metformin pre-treatment for all PCOS patients not grouped by HOMA-IR, PCOS patients with HOMA-IR < 2.71, and PCOS patients with HOMA-IR ≥ 2.71. RESULTS A total of 969 PCOS patients who received the GnRH-antagonist protocol were included in this study. For all PCOS patients, the metformin group showed comparable clinical pregnancy rates in fresh ET cycles and frozen ET cycles compared with the control group (55.9% vs. 57.1%, p = 0.821 and 63.8% vs. 60.9%, p = 0.497). For PCOS patients with HOMA-IR < 2.71, the clinical pregnancy rates in both fresh ET cycles and frozen ET cycles were statistically similar between the two groups (61.5% vs. 57.6%, p = 0.658 and 70.6% vs. 66.7%, p = 0.535). For PCOS patients with HOMA-IR ≥ 2.71, the clinical pregnancy rate in fresh ET cycles was comparable between the two groups (51.5% vs. 56.3, p = 0.590), but it was statistically higher in the metformin group than in the control group in frozen ET cycles (57.1% vs. 40.0%, p = 0.023). The metformin group had less oocytes retrieved, a lower cleaved oocyte rate, a lower available D3 embryo rate, a lower blastocyst formation rate, and a lower available blastocyst rate than the control group. CONCLUSION HOMA-IR is a judgment criterion for metformin pre-treatment before IVF/ICSI-ET in patients with PCOS. Metformin pre-treatment could be added for PCOS patients with HOMA-IR ≥ 2.71 during frozen IVF/ICSI-ET cycles to improve the clinical pregnancy rate.
Collapse
Affiliation(s)
- Rui Gao
- The Reproductive Medical Center, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Lang Qin
- The Reproductive Medical Center, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Zhengyu Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Zhengyu Li, ; Wenjiao Min,
| | - Wenjiao Min
- Psychosomatic Department, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- *Correspondence: Zhengyu Li, ; Wenjiao Min,
| |
Collapse
|
20
|
Guo F, Huang Y, Fernando T, Shi Y. Altered Molecular Pathways and Biomarkers of Endometrial Receptivity in Infertile Women with Polycystic Ovary Syndrome. Reprod Sci 2022; 29:3335-3345. [PMID: 35006579 DOI: 10.1007/s43032-022-00845-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/31/2021] [Indexed: 12/14/2022]
Abstract
Anovulation is the most prominent cause of infertility in polycystic ovary syndrome (PCOS) patients. Although ovulation can be corrected pharmacologically, the number of pregnancies remains low. Even if excellent embryos are transferred by IVF, it does not change the high miscarriage rate of PCOS patients. These facts collectively indicate that there is a disorder of endometrial development and receptivity to the embryo in PCOS patients, including the decrease of receptive ability, inhibition of embryo adhesion, undersupply of energy, poor blood perfusion, and pro-inflammatory status in the endometrium. However, it has never received the same attention as ovulatory dysfunction. Here we list some alternations of endometrial receptivity in women with PCOS, discuss the underlying intricate mechanisms, and try to find out the possible therapeutic targets, which may bring new perspectives to those who are able to provide high-quality embryos.
Collapse
Affiliation(s)
- Fei Guo
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Yufan Huang
- Department of Pharmacy, Mindong Hospital, Fujian Medical University, Ningde, 355000, Fujian, China
| | - Taniya Fernando
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Yingli Shi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
21
|
Bahri Khomami M, Teede HJ, Joham AE, Moran LJ, Piltonen TT, Boyle JA. Clinical management of pregnancy in women with polycystic ovary syndrome: An expert opinion. Clin Endocrinol (Oxf) 2022; 97:227-236. [PMID: 35383999 PMCID: PMC9544149 DOI: 10.1111/cen.14723] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/28/2022] [Accepted: 03/06/2022] [Indexed: 12/16/2022]
Abstract
Polycystic ovary syndrome (PCOS) is associated with a higher risk for pregnancy and birth complications according to the specific features associated with PCOS. The features include obesity before and during pregnancy, hyperandrogenism, insulin resistance, infertility, cardiometabolic risk factors, and poor mental health. PCOS is not often recognized as a risk factor for poor pregnancy and birth outcomes in pregnancy care guidelines, while its associated features are. Pregnancy-related risk profile should ideally be assessed for modifiable risk factors (e.g., lifestyle and weight management) at preconception in women with PCOS. Hyperglycaemia should be screened using a 75-g oral glucose tolerance test at preconception or within the first 20 weeks of pregnancy if it has not been performed at preconception and should be repeated at 24-28 weeks of pregnancy. In the absence of evidence of benefit for strategies specific to women with PCOS, the international evidence-based guidelines for the assessment and management of PCOS recommend screening, optimizing, and monitoring risk profile in women with PCOS (at preconception, during and postpregnancy) consistent with the recommendations for the general population. Recommended factors include blood glucose, weight, blood pressure, smoking, alcohol, diet, exercise, sleep and mental health, emotional, and sexual health among women with PCOS. The guidelines recommend Metformin in addition to lifestyle for assisting with weight management and improving cardiometabolic risk factors, particularly in those with overweight or obesity. Letrozole is considered the first-line pharmacological treatment for anovulatory infertility in PCOS. Individualized approach should be considered in the management of pregnancy in PCOS.
Collapse
Affiliation(s)
- Mahnaz Bahri Khomami
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine Nursing and Health SciencesMonash UniversityClaytonLevel 1, 43‐51 Kanooka GroveAustralia
| | - Helena J Teede
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine Nursing and Health SciencesMonash UniversityClaytonLevel 1, 43‐51 Kanooka GroveAustralia
- Monash HealthMelbourneAustralia
| | - Anju E. Joham
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine Nursing and Health SciencesMonash UniversityClaytonLevel 1, 43‐51 Kanooka GroveAustralia
- Monash HealthMelbourneAustralia
| | - Lisa J. Moran
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine Nursing and Health SciencesMonash UniversityClaytonLevel 1, 43‐51 Kanooka GroveAustralia
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University HospitalUniversity of OuluOuluFinland
| | - Jacqueline A. Boyle
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine Nursing and Health SciencesMonash UniversityClaytonLevel 1, 43‐51 Kanooka GroveAustralia
- Monash HealthMelbourneAustralia
| |
Collapse
|
22
|
Study on the Influencing Mechanism of Human Chorionic Gonadotropin (hCG) on Oocyte Maturation in Patients with Polycystic Ovary Syndrome. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7933166. [PMID: 35872929 PMCID: PMC9303116 DOI: 10.1155/2022/7933166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/06/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022]
Abstract
The study was aimed at investigating the influence of human chorionic gonadotropin (hCG) hormone on oocyte maturation in the patients with polycystic ovary syndrome (PCOS). A total of 54 patients with PCOS who received in vitro maturation (IVM) treatment in the Cheeloo College of Medicine, Shandong University, were divided into two groups: one group who underwent hCG injections was the observation group (OG; n = 27) and other was the control group (CG; n = 27) with no hCG injection. The oocyte development and the expression of steroid hormone synthesis-related genes including gonadotropin-releasing hormone receptor (GnRHR), Conexin43, epidermal growth factor-related genes, luteinizing hormone/choriogonadotropin receptor (LHCGR), epiregulin (EREG), and vascular endothelial growth factor (VEGF) were examined. The human ovarian granulosa cell line (SVOG cells) and ovarian epithelial cell line (HOSEpiC cells) were employed to analyze the effect of hCG on the biological behaviour of cells. As a result, OG showed higher normal fertilization, cleavage, and high-qualified embryo rate than CG. Expression levels of GnRHR, Cx43, LHCGR, EREG, and VEGF were significantly elevated in granulosa cells in the OG group. Western blot revealed that phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), and rapamycin (mTOR) proteins were decreased in granulosa cells under hCG intervention. A biological behaviour test indicated that the multiplication capacity of hCG-intervened SVOG and HOSEpiC was increased, while the apoptosis was decreased. In conclusion, hCG could accelerate follicular development and oocyte maturation by activating oocyte maturation genes in PCOS patients, which could significantly improve and popularize the application of IVM technology.
Collapse
|
23
|
Jannatifar R, Piroozmanesh H, Sahraei SS, Asa E. Combination of alpha lipoic acid and metformin supplement improve assisted reproductive technologies outcomes in polycystic ovary syndrome patients. Anat Cell Biol 2022; 55:239-246. [PMID: 35501295 PMCID: PMC9256491 DOI: 10.5115/acb.21.242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 11/27/2022] Open
Abstract
We aimed to investigating the effects of metformin (MET) in combination with alpha lipoic acid (ALA) on hormonal and biochemical parameters, in polycystic ovary syndrome (PCOS) women undergoing intracytoplasmic sperm injection (ICSI). This experimental pilot study with a randomized design was carried out on 40 PCOS women in two groups: (1) MET group, administered 1,500 mg/day MET, and (2) MET (1,500 mg/day)+ALA (1,800 mg/day) group. Drugs were administered from the third day of the previous cycle until the day of oocyte aspiration (six weeks of treatment in total). MET+ALA significantly increased the number of maturated oocytes and the rate of fertilization when compared to the MET group. Combination MET+ALA could increase significantly the number of oocytes retrieval and the number of good-quality embryos. Also, the malondialdehyde (MDA) level decreased significantly in the MET+ALA group and the total antioxidant capacity (TAC) level increased significantly in the MET+ALA group compared to the MET group. Also, fasting blood sugar (FBS), insulin, luteinizing hormone (LH), and LH/follicle stimulating hormone (FSH) levels were significantly lower in the MET+ALA group. The pregnancy outcomes showed no significant difference in the rates of biochemical pregnancy, clinical pregnancy, miscarriage, and live births between the control and study groups. The combination of MET+ALA treatment could moderate the complications of PCOS and subsequently improve oocyte and embryo quality.
Collapse
Affiliation(s)
- Rahil Jannatifar
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran.,Infertility Treatment Center, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Hamid Piroozmanesh
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran.,Infertility Treatment Center, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Seyedeh Saeideh Sahraei
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran.,Infertility Treatment Center, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Elham Asa
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran.,Infertility Treatment Center, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| |
Collapse
|
24
|
Froment P, Plotton I, Giulivi C, Fabre S, Khoueiry R, Mourad NI, Horman S, Ramé C, Rouillon C, Grandhaye J, Bigot Y, Chevaleyre C, Le Guevel R, Mallegol P, Andriantsitohaina R, Guerif F, Tamburini J, Viollet B, Foretz M, Dupont J. At the crossroads of fertility and metabolism: the importance of AMPK-dependent signaling in female infertility associated with hyperandrogenism. Hum Reprod 2022; 37:1207-1228. [PMID: 35459945 DOI: 10.1093/humrep/deac067] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 03/01/2022] [Indexed: 03/25/2024] Open
Abstract
STUDY QUESTION What biological processes are linked to the signaling of the energy sensor 5'-AMP-activated protein kinase (AMPK) in mouse and human granulosa cells (GCs)? SUMMARY ANSWER The lack of α1AMPK in GCs impacted cell cycle, adhesion, lipid metabolism and induced a hyperandrogenic response. WHAT IS KNOWN ALREADY AMPK is expressed in the ovarian follicle, and its activation by pharmacological medications, such as metformin, inhibits the production of steroids. Polycystic ovary syndrome (PCOS) is responsible for infertility in approximately 5-20% of women of childbearing age and possible treatments include reducing body weight, improving lifestyle and the administration of a combination of drugs to improve insulin resistance, such as metformin. STUDY DESIGN, SIZE, DURATION AMPK signaling was evaluated by analyzing differential gene expression in immortalized human granulosa cells (KGNs) with and without silencing α1AMPK using CRISPR/Cas9. In vivo studies included the use of a α1AMPK knock-out mouse model to evaluate the role of α1AMPK in folliculogenesis and fertility. Expression of α1AMPK was evaluated in primary human granulosa-luteal cells retrieved from women undergoing IVF with and without a lean PCOS phenotype (i.e. BMI: 18-25 kg/m2). PARTICIPANTS/MATERIALS, SETTING, METHODS α1AMPK was disrupted in KGN cells and a transgenic mouse model. Cell viability, proliferation and metabolism were evaluated. Androgen production was evaluated by analyzing protein levels of relevant enzymes in the steroid pathway by western blots, and steroid levels obtained from in vitro and in vivo models by mass spectrometry. Differential gene expression in human GC was obtained by RNA sequencing. Analysis of in vivo murine folliculogenesis was performed by histology and immunochemistry, including evaluation of the anti-Müllerian hormone (AMH) marker. The α1AMPK gene expression was evaluated by quantitative RT-PCR in primary GCs obtained from women with the lean PCOS phenotype (n = 8) and without PCOS (n = 9). MAIN RESULTS AND THE ROLE OF CHANCE Silencing of α1AMPK in KGN increased cell proliferation (P < 0.05 versus control, n = 4), promoted the use of fatty acids over glucose, and induced a hyperandrogenic response resulting from upregulation of two of the enzymes involved in steroid production, namely 3β-hydroxysteroid dehydrogenase (3βHSD) and P450 side-chain cleavage enzyme (P450scc) (P < 0.05, n = 3). Female mice deficient in α1AMPK had a 30% decrease in their ovulation rate (P < 0.05, n = 7) and litter size, a hyperandrogenic response (P < 0.05, n = 7) with higher levels of 3βHSD and p450scc levels in the ovaries, and an increase in the population of antral follicles (P < 0.01, n = 10) compared to controls. Primary GCs from lean women with PCOS had lower α1AMPK mRNA expression levels than the control group (P < 0.05, n = 8-9). LARGE SCALE DATA The FastQ files and metadata were submitted to the European Nucleotide Archive (ENA) at EMBL-EBI under accession number PRJEB46048. LIMITATIONS, REASONS FOR CAUTION The human KGN is a not fully differentiated, transformed cell line. As such, to confirm the role of AMPK in GC and the PCOS phenotype, this model was compared to two others: an α1AMPK transgenic mouse model and primary differentiated granulosa-lutein cells from non-obese women undergoing IVF (with and without PCOS). A clear limitation is the small number of patients with PCOS utilized in this study and that the collection of human GCs was performed after hormonal stimulation. WIDER IMPLICATIONS OF THE FINDINGS Our results reveal that AMPK is directly involved in steroid production in human GCs. In addition, AMPK signaling was associated with other processes frequently reported as dysfunctional in PCOS models, such as cell adhesion, lipid metabolism and inflammation. Silencing of α1AMPK in KGN promoted folliculogenesis, with increases in AMH. Evaluating the expression of the α1AMPK subunit could be considered as a marker of interest in infertility cases related to hormonal imbalances and metabolic disorders, including PCOS. STUDY FUNDING/COMPETING INTEREST(S) This study was financially supported by the Institut National de la Recherche Agronomique (INRA) and the national programme « FERTiNERGY » funded by the French National Research Agency (ANR). The authors report no intellectual or financial conflicts of interest related to this work. R.K. is identified as personnel of the International Agency for Research on Cancer/World Health Organization. R.K. alone is responsible for the views expressed in this article and she does not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer/World Health Organization. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Pascal Froment
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Ingrid Plotton
- Molecular Endocrinology and Rare Diseases, University Hospital, Claude Bernard Lyon 1 University, Bron, France
| | - Cecilia Giulivi
- Department of Molecular Biosciences, University of California Davis, School of Veterinary Medicine, Davis, CA, USA
- The MIND Institute, University of California Davis Medical Center, Sacramento, CA, USA
| | - Stephane Fabre
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Rita Khoueiry
- Epigenetics Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Nizar I Mourad
- Pôle de Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Brussels, Belgium
| | - Sandrine Horman
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Christelle Ramé
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | | | - Yves Bigot
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | - Remy Le Guevel
- Plate-forme ImPACcell, Université de Rennes 1, Rennes, France
| | - Patricia Mallegol
- SOPAM, U1063, INSERM, UNIV Angers, Angers, France
- Federative Structure of Research Cellular Interactions and Therapeutic Applications, SFR 4208 ICAT, Univ Angers, Angers, France
| | - Ramaroson Andriantsitohaina
- SOPAM, U1063, INSERM, UNIV Angers, Angers, France
- Federative Structure of Research Cellular Interactions and Therapeutic Applications, SFR 4208 ICAT, Univ Angers, Angers, France
| | | | - Jérôme Tamburini
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Benoit Viollet
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Marc Foretz
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Joelle Dupont
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| |
Collapse
|
25
|
Abruzzese GA, Silva AF, Velazquez ME, Ferrer MJ, Motta AB. Hyperandrogenism and Polycystic ovary syndrome: Effects in pregnancy and offspring development. WIREs Mech Dis 2022; 14:e1558. [PMID: 35475329 DOI: 10.1002/wsbm.1558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/18/2022] [Accepted: 04/01/2022] [Indexed: 11/10/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the major endocrine disorders affecting women of reproductive age. Its etiology remains unclear. It is suggested that environmental factors, and particularly the intrauterine environment, play key roles in PCOS development. Besides the role of androgens in PCOS pathogenesis, exposure to endocrine disruptors, as is Bisphenol A, could also contribute to its development. Although PCOS is considered one of the leading causes of ovarian infertility, many PCOS patients can get pregnant. Some of them by natural conception and others by assisted reproductive technique treatments. As hyperandrogenism (one of PCOS main features) affects ovarian and uterine functions, PCOS women, despite reaching pregnancy, could present high-risk pregnancies, including implantation failure, an increased risk of gestational diabetes, preeclampsia, and preterm birth. Moreover, hyperandrogenism may also be maintained in these women during pregnancy. Therefore, as an altered uterine milieu, including hormonal imbalance, could affect the developing organisms, monitoring these patients throughout pregnancy and their offspring development is highly relevant. The present review focuses on the impact of androgenism and PCOS on fertility issues and pregnancy-related outcomes and offspring development. The evidence suggests that the increased risk of pregnancy complications and adverse offspring outcomes of PCOS women would be due to the factors involved in the syndrome pathogenesis and the related co-morbidities. A better understanding of the involved mechanisms is still needed and could contribute to a better management of these women and their offspring. This article is categorized under: Reproductive System Diseases > Molecular and Cellular Physiology Reproductive System Diseases > Environmental Factors.
Collapse
Affiliation(s)
- Giselle A Abruzzese
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Aimé F Silva
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariela E Velazquez
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria-José Ferrer
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alicia B Motta
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
26
|
Ilias I, Rizzo M, Zabuliene L. Metformin: Sex/Gender Differences in Its Uses and Effects—Narrative Review. Medicina (B Aires) 2022; 58:medicina58030430. [PMID: 35334606 PMCID: PMC8952223 DOI: 10.3390/medicina58030430] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 02/08/2023] Open
Abstract
Metformin (MTF) occupies a major and fundamental position in the therapeutic management of type 2 diabetes mellitus (T2DM). Gender differences in some effects and actions of MTF have been reported. Women are usually prescribed lower MTF doses compared to men and report more gastrointestinal side effects. The incidence of cardiovascular events in women on MTF has been found to be lower to that of men on MTF. Despite some promising results with MTF regarding pregnancy rates in women with PCOS, the management of gestational diabetes, cancer prevention or adjunctive cancer treatment and COVID-19, most robust meta-analyses have yet to confirm such beneficial effects.
Collapse
Affiliation(s)
- Ioannis Ilias
- Department of Endocrinology, Diabetes and Metabolism, Elena Venizelou Hospital, GR-11521 Athens, Greece
- Correspondence: e-mail:
| | - Manfredi Rizzo
- Department of Health Promotion Sciences, Maternal and Infantile Care, Internal Medicine and Medical Specialties (Promise), School of Medicine, University of Palermo, Via del Vespro, 141, 90127 Palermo, Italy;
| | - Lina Zabuliene
- Faculty of Medicine, Vilnius University, M. K. Čiurlionio St. 21/27, LT-03101 Vilnius, Lithuania;
| |
Collapse
|
27
|
Notaro ALG, Neto FTL. The use of metformin in women with polycystic ovary syndrome: an updated review. J Assist Reprod Genet 2022; 39:573-579. [PMID: 35156149 PMCID: PMC8995234 DOI: 10.1007/s10815-022-02429-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 02/04/2022] [Indexed: 10/19/2022] Open
Abstract
PURPOSES Polycystic ovary syndrome (PCOS) is a major cause of female infertility, being present in up to 20% of women of childbearing age. Insulin resistance (IR) plays an important role in the pathophysiology of PCOS; therefore, its treatment may benefit women with the syndrome. The main drug used for IR management is metformin (MT). We aim to review the literature on the use of metformin in women with PCOS. METHODS Using the terms "metformin" and "polycystic ovary syndrome," we conducted a search the PubMed, EMBASE, and Google Scholar databases. The research was restricted to articles published in English. Initially, only published meta-analyses were included, in the absence of meta-analyzes, RCT and well-designed prospective studies were used. RESULTS Metformin increases success rates and decreases complication rates when used as an adjunctive medication for ovulation induction during low complexity assisted reproduction treatments and during ovarian stimulation for in vitro fertilization in women with PCOS. Evidence about the effect of metformin on fetal and obstetric complication rates is conflicting. Metformin is associated with high incidence of gastrointestinal symptoms; however, serious adverse effects are rare and there is no evidence of teratogenicity. CONCLUSION For women with PCOS, metformin is a good adjunctive medication for ovulation induction/stimulation for high and low complexity assisted reproduction therapies. The adverse effects are mostly mild, and there is no risk of teratogenicity, but the risk of long-term complications for the offspring is not yet defined. High heterogeneity of the studies limits extrapolation of findings, and further research is needed to determine which women will benefit most from the medication.
Collapse
Affiliation(s)
- Adriana Leal Griz Notaro
- Amare Clinic – Brazil, Av. República do Líbano, n 251, torre 1, sala 504, Pina, Recife, PE CEP 51110-160 Brazil
| | - Filipe Tenorio Lira Neto
- Andros Recife Clinic - Brazil, Av. Boa Viagem, n 179, apt 901, Pina, Recife, PE, CEP 51011-000, Brazil.
| |
Collapse
|
28
|
Адамян ЛВ, Андреева ЕН, Абсатарова ЮС, Григорян ОР, Дедов ИИ, Мельниченко ГА, Сутурина ЛВ, Филиппов ОС, Шереметьева ЕВ, Чернуха ГЕ, Ярмолинская МИ. [Clinical guidelines «Polycystic Ovary Syndrome»]. PROBLEMY ENDOKRINOLOGII 2022; 68:112-127. [PMID: 35488762 PMCID: PMC9764272 DOI: 10.14341/probl12874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a polygenic endocrine disorder caused by both genetic and epigenetic factors. Depending on the period of a woman's life, the clinical picture, diagnosis, and treatment tactics of the disease are different. PCOS has a complex of reproductive, metabolic and psychological characteristics. The target audience of these clinical recommendations are obstetrician-gynecologists, endocrinologists, general practitioners, general practitioners. In these clinical guidelines, all information is ranked according to the level of persuasiveness of recommendations and the reliability of evidence, depending on the number and quality of studies on this issue.
Collapse
Affiliation(s)
- Л. В. Адамян
- Национальный исследовательский центр акушерства, гинекологии и перинатологии им. В.И. Кулакова;
Московский государственный медико-стоматологический университет им. А.И. Евдокимова
| | - Е. Н. Андреева
- Национальный медицинский исследовательский центр эндокринологии;
Московский государственный медико-стоматологический университет им. А.И. Евдокимова
| | | | - О. Р. Григорян
- Национальный медицинский исследовательский центр эндокринологии
| | - И. И. Дедов
- Национальный медицинский исследовательский центр эндокринологии
| | | | - Л. В. Сутурина
- Научный центр проблем здоровья семьи и репродукции человека
| | - О. С. Филиппов
- Первый Московский государственный медицинский университет им. И.М. Сеченова (Сеченовский Университет);
Министерство здравоохранения Российской Федерации
| | | | - Г. Е. Чернуха
- Национальный исследовательский центр акушерства, гинекологии и перинатологии им. В.И. Кулакова
| | - М. И. Ярмолинская
- Научно-исследовательский институт акушерства, гинекологии и репродукции им. Д.О. Отта;
Северо-Западный государственный медицинский университет им. И.И. Мечникова
| |
Collapse
|
29
|
Wu D, Shi H, Yu Y, Yu T, Zhai J. Comparison of the Effectiveness of Various Medicines in the Prevention of Ovarian Hyperstimulation Syndrome: A Network Meta-Analysis of Randomized Controlled Trials. Front Endocrinol (Lausanne) 2022; 13:808517. [PMID: 35154015 PMCID: PMC8825486 DOI: 10.3389/fendo.2022.808517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Previous studies have described the effects of different drugs in preventing ovarian hyperstimulation syndrome (OHSS). However, the efficacies of those drugs in preventing OHSS remain inconclusive. METHODS We searched the PubMed, Web of Science, Embase, and Cochrane Central Register of Controlled Trials (CENTRAL) databases. A network meta-analysis of randomized controlled trials (RCTs) was performed up to August 2021. We investigated the following drugs in our study: aspirin, albumin, metformin, calcium, cabergoline, quinagolide, letrozole, hydroxyethyl starch (HES), and glucocorticoids. The primary outcome was the incidence rate of moderate-to-severe OHSS, with the results presented as risk ratios (RRs) with 95% confidence intervals (CIs). RESULTS The incidence of moderate-to-severe OHSS was significantly reduced by calcium administration (risk ratios [RR] 0.14, 95% confidence interval [CI]: 0.04, 0.46) (grade: high), HES (RR 0.25, 95% CI 0.07, 0.73) (grade: high), and cabergoline (RR 0.43, 95% CI 0.24, 0.71) (grade: moderate). The surface under the cumulative ranking curve (SUCRA) indicated that calcium (SUCRA, 92.4%) was the most effective intervention for preventing moderate-to-severe OHSS. These drugs were safe and did not affect clinical pregnancy, miscarriage, or live birth rates. CONCLUSION Calcium, HES, and cabergoline could effectively and safely prevent moderate-to-severe OHSS, with calcium as the most effective intervention.
Collapse
Affiliation(s)
- Di Wu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hao Shi
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiping Yu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ting Yu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Zhai
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Jun Zhai,
| |
Collapse
|
30
|
Islam H, Masud J, Islam YN, Haque FKM. An update on polycystic ovary syndrome: A review of the current state of knowledge in diagnosis, genetic etiology, and emerging treatment options. WOMEN'S HEALTH 2022; 18:17455057221117966. [PMID: 35972046 PMCID: PMC9386861 DOI: 10.1177/17455057221117966] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Polycystic ovary syndrome is the most common endocrine disorder in women of reproductive age, which is still incurable. However, the symptoms can be successfully managed with proper medication and lifestyle interventions. Despite its prevalence, little is known about its etiology. In this review article, the up-to-date diagnostic features and parameters recommended on the grounds of evidence-based data and different guidelines are explored. The ambiguity and insufficiency of data when diagnosing adolescent women have been put under special focus. We look at some of the most recent research done to establish relationships between different gene polymorphisms with polycystic ovary syndrome in various populations along with the underestimated impact of environmental factors like endocrine-disrupting chemicals on the reproductive health of these women. Furthermore, the article concludes with existing treatments options and the scopes for advancement in the near future. Various therapies have been considered as potential treatment through multiple randomized controlled studies, and clinical trials conducted over the years are described in this article. Standard therapies ranging from metformin to newly found alternatives based on vitamin D and gut microbiota could shine some light and guidance toward a permanent cure for this female reproductive health issue in the future.
Collapse
Affiliation(s)
- Hiya Islam
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Jaasia Masud
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Yushe Nazrul Islam
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Fahim Kabir Monjurul Haque
- Microbiology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| |
Collapse
|
31
|
Ala M, Ala M. Metformin for Cardiovascular Protection, Inflammatory Bowel Disease, Osteoporosis, Periodontitis, Polycystic Ovarian Syndrome, Neurodegeneration, Cancer, Inflammation and Senescence: What Is Next? ACS Pharmacol Transl Sci 2021; 4:1747-1770. [PMID: 34927008 DOI: 10.1021/acsptsci.1c00167] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Indexed: 12/15/2022]
Abstract
Diabetes is accompanied by several complications. Higher prevalence of cancers, cardiovascular diseases, chronic kidney disease (CKD), obesity, osteoporosis, and neurodegenerative diseases has been reported among patients with diabetes. Metformin is the oldest oral antidiabetic drug and can improve coexisting complications of diabetes. Clinical trials and observational studies uncovered that metformin can remarkably prevent or alleviate cardiovascular diseases, obesity, polycystic ovarian syndrome (PCOS), osteoporosis, cancer, periodontitis, neuronal damage and neurodegenerative diseases, inflammation, inflammatory bowel disease (IBD), tuberculosis, and COVID-19. In addition, metformin has been proposed as an antiaging agent. Numerous mechanisms were shown to be involved in the protective effects of metformin. Metformin activates the LKB1/AMPK pathway to interact with several intracellular signaling pathways and molecular mechanisms. The drug modifies the biologic function of NF-κB, PI3K/AKT/mTOR, SIRT1/PGC-1α, NLRP3, ERK, P38 MAPK, Wnt/β-catenin, Nrf2, JNK, and other major molecules in the intracellular signaling network. It also regulates the expression of noncoding RNAs. Thereby, metformin can regulate metabolism, growth, proliferation, inflammation, tumorigenesis, and senescence. Additionally, metformin modulates immune response, autophagy, mitophagy, endoplasmic reticulum (ER) stress, and apoptosis and exerts epigenetic effects. Furthermore, metformin protects against oxidative stress and genomic instability, preserves telomere length, and prevents stem cell exhaustion. In this review, the protective effects of metformin on each disease will be discussed using the results of recent meta-analyses, clinical trials, and observational studies. Thereafter, it will be meticulously explained how metformin reprograms intracellular signaling pathways and alters molecular and cellular interactions to modify the clinical presentations of several diseases.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), 1416753955 Tehran, Iran
| | - Mahan Ala
- School of Dentistry, Golestan University of Medical Sciences (GUMS), 4814565589 Golestan, Iran
| |
Collapse
|
32
|
Soodi S, Keshavarz SA, Hosseini S, Abbasi B. Dietary diversity score is inversely related to the risk of polycystic ovary syndrome in Tehranian women: a case-control study. Appl Physiol Nutr Metab 2021; 47:1-6. [PMID: 34878921 DOI: 10.1139/apnm-2021-0359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among women of reproductive age and is affected by various dietary factors. Therefore, this study aimed to investigate the relationship between dietary diversity score (DDS) and the risk of PCOS. Our case-control study was conducted in the summer and autumn of 2019 in Taleghani and Arash hospitals in Tehran, Iran. A total of 494 participants (203 cases and 291 controls) were included in the study. Thereafter, their demographic information, dietary intake, and anthropometric and physical activity assessments were gathered. A validated semi-quantitative food frequency questionnaire was then used to calculate the DDS by scoring 5 food groups. To evaluate the risk of PCOS in association with DDS, the subjects were categorized based on the quartile cut-off points of the DDS. The mean ± SD age of the participants in both the case and control groups was 28.98 ± 5.43 and 30.15 ± 6.21 years, while mean ± SD body mass index was 25.74 ± 5.44 and 23.65 ± 3.90 kg/m2, respectively. The comparison between the case and control groups indicated that total DDS was 5.19 ± 1.19 for the cases and 5.51 ± 1.19 for the controls. The comparison of DDS in the highest versus the lowest quartiles showed a decreased risk of PCOS (p < 0.05). We demonstrated an inverse association between DDS and PCOS compared with the control group. Furthermore, a higher DDS was significantly associated with a lower risk of PCOS (odds ratio = 0.40). Novelty: This is the first investigation on the relationship between DDS and PCOS. Results depicted an inverse relationship between DDS and PCOS.
Collapse
Affiliation(s)
- Sahel Soodi
- Department of Nutrition, Electronic Health and Statistics Surveillance Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Ali Keshavarz
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetic, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedighe Hosseini
- Preventive Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnood Abbasi
- Department of Nutrition, Electronic Health and Statistics Surveillance Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
33
|
Li F, Chen Y, Niu A, He Y, Yan Y. Nomogram Model to Predict the Probability of Ovarian Hyperstimulation Syndrome in the Treatment of Patients With Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2021; 12:619059. [PMID: 34421814 PMCID: PMC8377671 DOI: 10.3389/fendo.2021.619059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 07/07/2021] [Indexed: 11/21/2022] Open
Abstract
Objective The objective of this study was to explore the risk factors of ovarian hyperstimulation syndrome (OHSS) in patients with polycystic ovary syndrome (PCOS) undergoing in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) and to establish a nomogram model evaluate the probability of OHSS in PCOS patients. Methods We retrospectively analyzed clinical data from 4,351 patients with PCOS receiving IVF/ICSI in our reproductive medical center. The clinical cases were randomly divided into a modeling group (3,231 cases) and a verification group (1,120 cases) according to a ratio of about 3:1. The independent risk factors correlation with the occurrence of OHSS was identified by logistic regression analysis. Based on the selected independent risk factors and correlated regression coefficients, we established a nomogram model to predict the probability of OHSS in PCOS patients, and the predictive accuracy of the model was measured using the area under the receiver operating curve (AUC). Results Univariate and multivariate logistic regression analyses showed that FSH (OR, 0.901; 95% CI, 0.847-0.958; P<0.001), AMH (OR, 1.259; 95% CI, 1.206-1.315; P<0.001), E2 value on the day of hCG injection (OR, 1.122; 95% CI, 1.021-1.253; P<0.001), total dosage of Gn used (OR, 1.010; 95% CI, 1.002-1.016; P=0.041), and follicle number on the day of hCG injection (OR, 0.134; 95% CI, 1.020-1.261; P=0.020) are the independent risk factors for OHSS in PCOS patients. The AUC of the modeling group is 0.827 (95% CI, 0.795-0.859), and the AUC of the verification group is 0.757 (95% CI, 0.733-0.782). Conclusion The newly established nomogram model has proven to be a novel tool that can effectively, easily, and intuitively predict the probability of OHSS in the patients with PCOS, by which the clinician can set up a better clinical management strategies for conducting a precise personal therapy.
Collapse
Affiliation(s)
- Fei Li
- Center for Reproductive Medicine, The First People’s Hospital of Shangqiu, Henan, China
- Center for Reproductive Medicine, The First Affiliated Hospital of Zheng Zhou University, Henan, China
| | - Ying Chen
- Center for Reproductive Medicine, The First People’s Hospital of Shangqiu, Henan, China
| | - Aiqin Niu
- Center for Reproductive Medicine, The First People’s Hospital of Shangqiu, Henan, China
| | - Yajing He
- Department of Pathology, The First People’s Hospital of Shangqiu, Henan, China
| | - Ying Yan
- Department of Molecular Biology, The First People’s Hospital of Shangqiu, Henan, China
| |
Collapse
|
34
|
Raperport C, Chronopoulou E, Homburg R. Effects of metformin treatment on pregnancy outcomes in patients with polycystic ovary syndrome. Expert Rev Endocrinol Metab 2021; 16:37-47. [PMID: 33634727 DOI: 10.1080/17446651.2021.1889366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/09/2021] [Indexed: 01/28/2023]
Abstract
INTRODUCTION This review covers the current evidence regarding the use of metformin as a therapeutic intervention for optimizing pregnancy outcomes in women with polycystic ovary syndrome (PCOS). AREAS COVERED After searching Medline, Embase and CINAHL, all important large clinical trials and observational studies plus systematic reviews, meta-analyses and Cochrane reviews have been summarized here. The results have been compared to culminate in a thorough review and discussion on the use of metformin in relation to reproductive outcomes for women with PCOS. The role of metformin in PCOS is explored both in terms of achieving conception and during pregnancy. The existing evidence around metformin use is summarized both during the preconceptual period and during pregnancy, in relation to reproductive outcomes. EXPERT OPINION Metformin is a widely used medication, often prescribed to improve reproductive outcomes for women with PCOS. However, the evidence remains equivocal regarding its efficacy both in optimizing fertility and pregnancy outcomes. More research is required with special emphasis on metformin use within different populations, including ethnic groups and women with varying BMI ranges.
Collapse
Affiliation(s)
- Claudia Raperport
- Fertility Unit, Homerton University Hospital, London, UK
- Blizard Institute, Queen Mary University of London, London, UK
| | | | - Roy Homburg
- Fertility Unit, Homerton University Hospital, London, UK
| |
Collapse
|
35
|
Shpakov AO. Improvement Effect of Metformin on Female and Male Reproduction in Endocrine Pathologies and Its Mechanisms. Pharmaceuticals (Basel) 2021; 14:ph14010042. [PMID: 33429918 PMCID: PMC7826885 DOI: 10.3390/ph14010042] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Metformin (MF), a first-line drug to treat type 2 diabetes mellitus (T2DM), alone and in combination with other drugs, restores the ovarian function in women with polycystic ovary syndrome (PCOS) and improves fetal development, pregnancy outcomes and offspring health in gestational diabetes mellitus (GDM) and T2DM. MF treatment is demonstrated to improve the efficiency of in vitro fertilization and is considered a supplementary drug in assisted reproductive technologies. MF administration shows positive effect on steroidogenesis and spermatogenesis in men with metabolic disorders, thus MF treatment indicates prospective use for improvement of male reproductive functions and fertility. MF lacks teratogenic effects and has positive health effect in newborns. The review is focused on use of MF therapy for restoration of female and male reproductive functions and improvement of pregnancy outcomes in metabolic and endocrine disorders. The mechanisms of MF action are discussed, including normalization of metabolic and hormonal status in PCOS, GDM, T2DM and metabolic syndrome and restoration of functional activity and hormonal regulation of the gonadal axis.
Collapse
Affiliation(s)
- Alexander O Shpakov
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 Saint Petersburg, Russia
| |
Collapse
|
36
|
Hussein RS, Elnashar I, Amin AF, Zhao Y, Abdelmagied AM, Abbas AM, Abdelaleem AA, Farghaly TA, Abdalmageed OS, Youssef AA, Badran E, Abou-Taleb HA. Effect of Metformin on Premature Luteinization and Pregnancy Outcomes in Intracytoplasmic Sperm Injection-Fresh Embryo Transfer Cycles: A Randomized Double-Blind Controlled Trial. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2021; 15:108-114. [PMID: 33687163 PMCID: PMC8052800 DOI: 10.22074/ijfs.2020.134643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 09/14/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Premature luteinization (PL) is not unusual in in vitro fertilization (IVF) and could not be wholly avoided by using either gonadotropin-releasing hormone (GnRH) agonists or GnRH antagonist regimens. The study aims to evaluate metformin's efficacy in preventing PL in fresh GnRH antagonist intracytoplasmic sperm injection (ICSI) cycles with cleavage-stage embryo transfer. MATERIALS AND METHODS This randomized, double-blind, placebo-controlled trial was conducted in a tertiary university IVF center. We recruited infertile women who were scheduled to perform their first or second ICSI trial. Eligible women were recruited and randomized in a 1:1 ratio into two groups. Metformin was administered in a dose of 1500 mg per day since the start of contraceptive pills in the cycle antecedent to stimulation cycle until the day of ovulation triggering, while women in the placebo group received a placebo for the same regimen and duration. The primary outcome was the incidence of PL, defined as serum progesterone (P) on the triggering day ≥1.5 ng/mL. Secondary outcomes comprised the live birth, ongoing pregnancy, implantation, and good-quality embryos rates. RESULTS The trial involved 320 eligible participants (n=160 in each group). Both groups had comparable stimulation days, endometrial thickness, peak estradiol levels, number of oocytes retrieved, and number of mature oocytes. Metformin group experienced lower level of serum P (P<0.001) and incidence of PL (10 vs. 23.6%, P=0.001). Moreover, lower progesterone/estradiol (P/E) ratio and progesterone to mature oocyte index (PMOI) (P=0.002 and P=0.002, respectively) were demonstrated in women receiving metformin. Metformin group generated a better rate of goodquality embryos (P=0.005) and ongoing pregnancy (43.8 vs. 31.8%, P=0.026). A similar trend, though of borderline significance, was observed in the live birth rate in favor of metformin administration (38.15 vs. 27.5%, P=0.04). CONCLUSION Metformin could be used in patients with potential PL to improve fresh cycle outcomes by preventing PL (Registration number: NCT03088631).
Collapse
Affiliation(s)
- Reda S. Hussein
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA,Department of Obstetrics and GynecologyFaculty of
MedicineAssiut UniversityAssiutEgypt
| | - Ihab Elnashar
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed F Amin
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Yulian Zhao
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ahmed M. Abdelmagied
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt,Department of Obstetrics and Gynecology, Taibah University, Medina, KSA
| | - Ahmed M. Abbas
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed A. Abdelaleem
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Tarek A. Farghaly
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Osama S Abdalmageed
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed A. Youssef
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Esraa Badran
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Hisham A. Abou-Taleb
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
37
|
Seow KM, Chang YW, Chen KH, Juan CC, Huang CY, Lin LT, Tsui KH, Chen YJ, Lee WL, Wang PH. Molecular Mechanisms of Laparoscopic Ovarian Drilling and Its Therapeutic Effects in Polycystic Ovary Syndrome. Int J Mol Sci 2020; 21:8147. [PMID: 33142702 PMCID: PMC7663012 DOI: 10.3390/ijms21218147] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrinopathy, characterized by chronic anovulation, hyperandrogenism, and multiple small subcapsular cystic follicles in the ovary during ultrasonography, and affects 5-10% of women of reproductive age. PCOS is frequently associated with insulin resistance (IR) accompanied by compensatory hyperinsulinemia and, therefore, presents an increased risk of type 2 diabetes mellitus (DM). The pathophysiology of PCOS is unclear, and many hypotheses have been proposed. Among these hypotheses, IR and hyperandrogenism may be the two key factors. The first line of treatment in PCOS includes lifestyle changes and body weight reduction. Achieving a 5-15% body weight reduction may improve IR and PCOS-associated hormonal abnormalities. For women who desire pregnancy, clomiphene citrate (CC) is the front-line treatment for ovulation induction. Twenty five percent of women may fail to ovulate spontaneously after three cycles of CC treatment, which is called CC-resistant PCOS. For CC-resistant PCOS women, there are many strategies to improve ovulation rate, including medical treatment and surgical approaches. Among the various surgical approaches, one particular surgical method, called laparoscopic ovarian drilling (LOD), has been proposed as an alternative treatment. LOD results in an overall spontaneous ovulation rate of 30-90% and final pregnancy rates of 13-88%. These benefits are more significant for women with CC-resistant PCOS. Although the intra- and post-operative complications and sequelae are always important, we believe that a better understanding of the pathophysiological changes and/or molecular mechanisms after LOD may provide a rationale for this procedure. LOD, mediated mainly by thermal effects, produces a series of morphological and biochemical changes. These changes include the formation of artificial holes in the very thick cortical wall, loosening of the dense and hard cortical wall, destruction of ovarian follicles with a subsequently decreased amount of theca and/or granulosa cells, destruction of ovarian stromal tissue with the subsequent development of transient but purulent and acute inflammatory reactions to initiate the immune response, and the continuing leakage or drainage of "toxic" follicular fluid in these immature and growth-ceased pre-antral follicles. All these factors contribute to decreasing local and systemic androgen levels, the following apoptosis process with these pre-antral follicles to atresia; the re-starting of normal follicular recruitment, development, and maturation, and finally, the normalization of the "hypothalamus-pituitary-ovary" axis and subsequent spontaneous ovulation. The detailed local and systematic changes in PCOS women after LOD are comprehensively reviewed in the current article.
Collapse
Affiliation(s)
- Kok-Min Seow
- Department of Obstetrics and Gynecology, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan;
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan; (C.-Y.H.); (L.-T.L.); (K.-H.T.); (Y.-J.C.)
- Institute of Physiology, National Yang-Ming University, Taipei 112, Taiwan;
| | - Yi-Wen Chang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan;
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Kuo-Hu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei 108, Taiwan;
- School of Medicine, Tzu-Chi University, Hualien 970, Taiwan
| | - Chi-Chang Juan
- Institute of Physiology, National Yang-Ming University, Taipei 112, Taiwan;
| | - Chen-Yu Huang
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan; (C.-Y.H.); (L.-T.L.); (K.-H.T.); (Y.-J.C.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Li-Te Lin
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan; (C.-Y.H.); (L.-T.L.); (K.-H.T.); (Y.-J.C.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan; (C.-Y.H.); (L.-T.L.); (K.-H.T.); (Y.-J.C.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County 907, Taiwan
| | - Yi-Jen Chen
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan; (C.-Y.H.); (L.-T.L.); (K.-H.T.); (Y.-J.C.)
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wen-Ling Lee
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Medicine, Cheng-Hsin General Hospital, Taipei 112, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei 112, Taiwan; (C.-Y.H.); (L.-T.L.); (K.-H.T.); (Y.-J.C.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Female Cancer Foundation, Taipei 104, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|