1
|
Balachandren N, Seshadri S, Yasmin E, Saab W, Gates C, Sayar Z, Cohen H, Webber L. Venous thromboembolism associated with medically assisted reproduction (MAR): British fertility society policy and practice guidance for assessment and prevention. HUM FERTIL 2024; 27:2352387. [PMID: 38804228 DOI: 10.1080/14647273.2024.2352387] [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: 12/27/2023] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
The association between Medically Assisted Reproduction (MAR) and thromboembolic complications has been reported widely in multiple published studies. Although venous thromboembolism (VTE) is not thought to be a common complication of MAR, it is associated with high morbidity and is often preventable. Since VTE usually occurs after completion of MAR treatment and is often managed outside of the treating fertility unit, these complications are likely to be underreported and there may be limited awareness of the risks among clinicians. As we continue to see a rise in the total number of MAR treatment cycles, particularly in women over 40 years of age, along with a steady increase in the number of fertility preservation cycles for both medical and social indications, it is likely that we will see an increase in absolute numbers of VTE complications. Currently, there is a lack of management guidance and reporting of VTE events associated with assisted conception treatment. The aim of this guidance is to provide clinicians with information on VTE risk factors, guidance on assessing VTE risk and the best practice recommendations on risk reducing strategies for individuals at risk of VTE undergoing ovarian stimulation and embryo transfer cycles.
Collapse
Affiliation(s)
| | | | - Ephia Yasmin
- Reproductive Medicine Unit, University College London Hospital, London, UK
| | - Wael Saab
- The Centre for Reproductive & Genetic Health, London, UK
| | - Carolyn Gates
- Department of Haematology, University College London Hospital, London, UK
| | - Zara Sayar
- Department of Haematology, University College London Hospital, London, UK
- Department of Haematology, Whittington Health NHS Trust, London, UK
| | - Hannah Cohen
- Department of Haematology, University College London Hospital, London, UK
| | - Lisa Webber
- Gynaecology & Reproductive Medicine, Imperial College Healthcare NHS Trust, St Mary's Hospital, London, W2 1NY and Department of Obstetrics & Gynaecology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
2
|
Tanaka Y, Amano T, Nakamura A, Takahashi A, Takebayashi A, Hanada T, Tsuji S, Murakami T. Balancing Fertility Preservation and Treatment Efficacy in (Neo)adjuvant Therapy for Adolescent and Young Adult Breast Cancer Patients: a Narrative Review. Curr Oncol Rep 2024; 26:1563-1574. [PMID: 39499484 DOI: 10.1007/s11912-024-01615-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2024] [Indexed: 11/07/2024]
Abstract
PURPOSE OF REVIEW Adolescent and young adult (AYA) breast cancer survivors face a significant risk of infertility due to the gonadotoxic effects of (neo)adjuvant therapy, which complicates their ability to conceive post-treatment. While (neo)adjuvant therapy primarily aims to improve recurrence-free and overall survival, fertility preservation strategies should also be considered for young patients. This narrative review explores recent advancements in fertility preservation techniques, such as oocyte, embryo, and ovarian tissue cryopreservation, and evaluates the feasibility of modifying breast cancer (neo)adjuvant therapy to preserve fertility without compromising survival outcomes. RECENT FINDINGS Our review highlights that clinical trials with co-primary endpoints of oncological safety and fertility preservation are limited, and substituting standard treatment regimens solely for fertility preservation is currently not recommended. Nevertheless, new clinical studies have emerged that either exclude highly ovarian-toxic agents, such as cyclophosphamide, or omit adjuvant therapy altogether, even if fertility preservation is not their primary endpoint. Unfortunately, many of these trials have not evaluated ovarian toxicity. Notably, since 2020, major oncology organizations, including the American Society of Clinical Oncology (ASCO), the European Society of Medical Oncology (ESMO) have advocated for the routine assessment of ovarian toxicity in all clinical trials. The review underscores the importance of incorporating ovarian toxicity as a standard endpoint in future trials involving premenopausal breast cancer patients to identify treatment regimens that can effectively balance fertility preservation with treatment efficacy.
Collapse
Affiliation(s)
- Yuji Tanaka
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan.
| | - Tsukuru Amano
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akiko Nakamura
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akimasa Takahashi
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akie Takebayashi
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| |
Collapse
|
3
|
Grynberg M, Zeghari F, Peigné M, Benoit A, Rakrouki S, Sifer C, Mayeur A, Saïs E, Sonigo C. Effect of breast cancer prognostic factors on ovarian reserve and response in fertility preservation. Reprod Biomed Online 2024; 49:104109. [PMID: 39182453 DOI: 10.1016/j.rbmo.2024.104109] [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: 01/25/2024] [Revised: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 08/27/2024]
Abstract
RESEARCH QUESTION Do breast cancer prognostic factors influence ovarian reserve and response to ovarian stimulation in the context of fertility preservation? DESIGN Observational, bicentric retrospective study of 352 women with breast cancer who underwent ovarian stimulation using a random start gonadotrophin releasing hormone antagonist protocol and vitrified oocytes between November 2015 and August 2022. Serum anti-Müllerian hormone (AMH) levels and antral follicle count (AFC) were measured. The number of oocytes recovered, maturation rate and follicular output rate (FORT) were analysed according to patients' characteristics and breast cancer prognostic factors. RESULTS Median age was 34 years (31.1-37.1). Median AFC and serum AMH level were 17 (12-26) follicles and 2 (1.2-3.4) ng/ml, respectively. After ovarian stimulation, 10.5 (6.0-16.0) oocytes were recovered, with eight (4-13) being mature. Mean oocyte maturation rate was 79% (62-92). Antral follicle count (>12) significantly affected the risk of recovering fewer than eight mature oocytes (P < 0.0001, multivariate analysis). Follicular responsiveness to FSH, assessed by the follicular output rate (FORT index) and number of oocytes recovered, were 31% (21-50) and 10.5% (6.0-16.0), respectively. FORT index and ovarian stimulation outcomes were not influenced by breast cancer prognostic factors. CONCLUSION Breast cancer prognostic factors do not influence ovarian reserve markers or response to ovarian stimulation in fertility preservation. Therefore, tumour grade, triple-negative status, HER2 overexpression and high Ki67 should not alter the fertility-preservation strategy when considering ovarian stimulation for oocyte vitrification.
Collapse
Affiliation(s)
- Michaël Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; Department of Reproductive Medicine and Fertility Preservation, Université Sorbonne Paris Nord, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France; INSERM, U1133, Université Paris Diderot, France.
| | - Fayçal Zeghari
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France
| | - Maeliss Peigné
- Department of Reproductive Medicine and Fertility Preservation, Université Sorbonne Paris Nord, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
| | - Alexandra Benoit
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France
| | - Sophia Rakrouki
- Department of Reproductive Medicine and Fertility Preservation, Université Sorbonne Paris Nord, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
| | | | - Anne Mayeur
- Department of Biology of Reproduction and CECOS, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclere Hospital, 92140, Clamart, France
| | - Emine Saïs
- Department of Biology of Reproduction and CECOS, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclere Hospital, 92140, Clamart, France
| | - Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; Unité Inserm U1185, Université Paris-Sud, Le Kremlin Bicêtre, France
| |
Collapse
|
4
|
Shapira M, Sella T, Safrai M, Villain E, Lifshitz D, Orvieto R, Gal-Yam E, Meirow D. Long-term safety of controlled ovarian stimulation for fertility preservation before chemotherapy treatment in patients with breast cancer. Fertil Steril 2024:S0015-0282(24)02308-2. [PMID: 39427822 DOI: 10.1016/j.fertnstert.2024.10.014] [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: 05/13/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024]
Abstract
OBJECTIVE To evaluate the long-term safety of controlled ovarian stimulation for fertility preservation before breast cancer chemotherapy treatment. DESIGN Retrospective observational cohort. SETTING Tertiary medical center. PATIENT(S) Two hundred thirteen women aged 18 to 43 years with newly diagnosed stage I-III breast cancer treated with systemic chemotherapy during 2015-2019. Of those, 74 underwent controlled ovarian stimulation for fertility preservation recipients, and 141 did not (controls). INTERVENTION(S) controlled ovarian stimulation for fertility preservation. MAIN OUTCOME MEASURE(S) Invasive disease-free survival, calculated from the time of surgery to the time of detection of breast cancer recurrence or death, whichever came first. RESULT(S) At diagnosis, fertility preservation recipients were significantly younger than controls (32.7 vs. 38.5 years), were less likely to be partnered (44.4% vs. 90.1%) or parous (38.9% vs. 95%), and were more likely to harbor a BRCA germline mutation (36.5% vs. 14.2%). Disease characteristics and treatment modalities were comparable between groups, apart from tumor staging, with maximal tumor diameter being >5 cm in 22.2% of fertility preservation recipients as opposed to 5.7% of controls. Mean follow-up was 60.9 and 65.4 months for fertility preservation recipients and controls, respectively. Five-year invasive disease-free survival was 80% for fertility preservation recipients and 86% for controls. In a multivariate analysis adjusted for statistically significant covariates, invasive disease-free survival remained similar between the groups (hazards ratio [HR], 0.86; 95% confidence interval [CI], 0.4-1.87). Invasive disease-free survival rates were not statistically different in clinically relevant subgroups, including patients receiving neoadjuvant chemotherapy (HR, 1.57; 95% CI, 0.62-3.99) and those cotreated with tamoxifen during stimulation because of an estrogen receptor positive disease (HR, 1.66; 95% CI, 0.67-3.49). CONCLUSION(S) Fertility preservation with controlled ovarian stimulation for patients with breast cancer was not found to impair long-term oncologic outcomes, including in emergent clinically relevant subgroups.
Collapse
Affiliation(s)
- Moran Shapira
- Fertility Preservation Center, IVF Unit, Obstetrics and Gynecology Department, Sheba Medical Center, Ramat Gan, Israel; Obstetrics and Gynecology Department, IVF Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Science, Tel Aviv University, Tel Aviv, Israel.
| | - Tal Sella
- Department of Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Myriam Safrai
- Fertility Preservation Center, IVF Unit, Obstetrics and Gynecology Department, Sheba Medical Center, Ramat Gan, Israel; Obstetrics and Gynecology Department, IVF Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Evyatar Villain
- Faculty of Medical and Health Science, Tel Aviv University, Tel Aviv, Israel
| | - Dror Lifshitz
- Obstetrics and Gynecology Department, IVF Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Raoul Orvieto
- Fertility Preservation Center, IVF Unit, Obstetrics and Gynecology Department, Sheba Medical Center, Ramat Gan, Israel; Obstetrics and Gynecology Department, IVF Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Science, Tel Aviv University, Tel Aviv, Israel
| | - Einav Gal-Yam
- Faculty of Medical and Health Science, Tel Aviv University, Tel Aviv, Israel; Department of Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Dror Meirow
- Fertility Preservation Center, IVF Unit, Obstetrics and Gynecology Department, Sheba Medical Center, Ramat Gan, Israel; Obstetrics and Gynecology Department, IVF Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Science, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
5
|
Gotschel F, Sonigo C, Becquart C, Sellami I, Mayeur A, Grynberg M. New Insights on In Vitro Maturation of Oocytes for Fertility Preservation. Int J Mol Sci 2024; 25:10605. [PMID: 39408934 PMCID: PMC11477201 DOI: 10.3390/ijms251910605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
In the last decade, the evolution of oncofertility has sparked a resurgence of interest in in vitro maturation (IVM) due to its suitability in certain oncological scenarios where controlled ovarian hyperstimulation may not be feasible. The retrieval of immature cumulus-oocyte complexes from small antral follicles, regardless of the menstrual cycle phase, presents a swift opportunity to vitrify mature oocytes or embryos post-IVM in urgent situations or when stimulation is not advisable. Harvesting immature cumulus-oocyte complexes and immature oocytes can be achieved transvaginally or directly in the laboratory from extracorporeal ovarian tissue. Although IVM has transitioned from an experimental status due to safety validations, it relies on the intricate process of oocyte maturation. Despite successful live births resulting from IVM in fertility preservation contexts, the comparatively lower developmental competence of in vitro matured oocytes highlights the necessity to enhance IVM culture systems. Recent advancements in IVM systems hold promise in bolstering oocyte competence post-IVM, thereby narrowing the gap between IVM and outcomes from ovarian stimulation. Additionally, for optimizing the chances of conception in cancer survivors, the combination of IVM and ovarian tissue cryopreservation stands as the favored choice when ovarian stimulation is unfeasible.
Collapse
Affiliation(s)
- Flavie Gotschel
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
- Inserm, Physiologie et Physiopathologie Endocrinienne, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Celeste Becquart
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Ines Sellami
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Anne Mayeur
- Histology-Embryology-Cytogenetic Laboratory, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France;
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
- Department of Reproductive Medicine and Fertility Preservation, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
- Unité Inserm U1133, Université Paris-Diderot, 75013 Paris, France
| |
Collapse
|
6
|
Liu SM, Huang SY, Wu HM, Chang CL, Huang HY. Ovarian stimulation response and fertility outcomes in patients with breast cancer across different stages, grades, and hormone receptor status for fertility preservation. J Formos Med Assoc 2024:S0929-6646(24)00399-1. [PMID: 39214749 DOI: 10.1016/j.jfma.2024.08.031] [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: 03/01/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND This study aimed to explore the potential impact of stage, grade, and hormone receptor profile on ovarian stimulation response and fertility preservation outcomes. METHODS This retrospective cohort study evaluated data from breast cancer patients who underwent fertility preservation at a tertiary medical center between 2014 and 2022. The outcomes of women with low-stage cancer (stages I and II) were compared with those of women with high-stage disease (stages III and IV or lymph node metastasis). Similarly, we compared those with low-grade (grades 1 and 2) and high-grade (grade 3) malignancies. In addition, we compared different hormone statuses of breast cancer (1) estrogen receptor (ER) positive vs. ER-negative and (2) triple-negative breast cancer (TNBC) vs. non-TNBC. The primary outcome measured was the number of mature oocytes, while the secondary outcomes included the numbers of total oocytes retrieved, peak estradiol levels, and subsequent fertility preservation outcomes. RESULTS A total of 47 patients were included. Patients with high-grade tumors had a comparable number of mature oocytes (8 vs. 10, p = 0.08) compared to patients with low grade cancers. The stage-based analysis revealed a similar number of mature oocytes (8 vs. 10, p = 0.33) between high/low stage patients. In the hormone receptor-based analysis, no differences were seen in mature oocytes collected between the ER-positive/ER-negative group (9 vs. 9, p = 0.87) and the TNBC/non-TNBC group (11 vs. 9, p = 0.13). The utilization rate was 27.6% (13/47). CONCLUSION Our study showed similar ovarian stimulation response and fertility preservation outcomes among breast cancer patients with different prognostic factors.
Collapse
Affiliation(s)
- Shang-Min Liu
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, 5 Fu-Shin Street, Kwei-Shan, Tao-Yuan, Taiwan
| | - Shang-Yu Huang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, 5 Fu-Shin Street, Kwei-Shan, Tao-Yuan, Taiwan; Chang Gung University College of Medicine, 259 Wen-Hua 1st Road, Kwei-shan, Tao-Yuan, Taiwan
| | - Hsien-Ming Wu
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, 5 Fu-Shin Street, Kwei-Shan, Tao-Yuan, Taiwan; Chang Gung University College of Medicine, 259 Wen-Hua 1st Road, Kwei-shan, Tao-Yuan, Taiwan
| | - Chia-Lin Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, 5 Fu-Shin Street, Kwei-Shan, Tao-Yuan, Taiwan; Chang Gung University College of Medicine, 259 Wen-Hua 1st Road, Kwei-shan, Tao-Yuan, Taiwan
| | - Hong-Yuan Huang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, 5 Fu-Shin Street, Kwei-Shan, Tao-Yuan, Taiwan; Chang Gung University College of Medicine, 259 Wen-Hua 1st Road, Kwei-shan, Tao-Yuan, Taiwan.
| |
Collapse
|
7
|
Abel MK, Wang A, Letourneau JM, Melisko ME, Cedars MI, Rosen MP. Changing the Perspective on Fertility Preservation for Women with Metastatic or Advanced Stage Cancer. Curr Oncol Rep 2024; 26:583-592. [PMID: 38639793 DOI: 10.1007/s11912-024-01530-9] [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] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
PURPOSE OF REVIEW In this Perspective we share the personal story of a 33-year-old patient diagnosed with metastatic breast cancer and her journey through fertility preservation, surrogacy, and eventually motherhood, highlighting misconceptions about fertility preservation in this population. RECENT FINDINGS There are nearly 1 million women under the age of 50 diagnosed and living with cancer in the USA. These patients are met with life-altering decisions, including those that may limit their reproductive ability. While there have been tremendous advances and advocacy in the field of oncofertility, there has been limited focus on patients with advanced stage or metastatic cancer. We describe five key misconceptions surrounding fertility preservation in patients with advanced stage cancer, offering a review of the literature and our approach to challenging topics like desiring fertility preservation in the face of Stage 4 disease, the safety and timing of ovarian stimulation during cancer treatment, and passing away following fertility preservation. We review the importance of assessing perceptions of fertility preservation in patients with metastatic cancer and highlight the lack of research in this area as a call to action.
Collapse
Affiliation(s)
- Mary Kathryn Abel
- San Francisco School of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
- Deparment of Obstetrics and Gynecology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, 75 Francis Street, 3rd Floor, Boston, MA, 02115, USA.
| | - Ange Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Joseph M Letourneau
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Michelle E Melisko
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Marcelle I Cedars
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Mitchell P Rosen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
Roberts JE, Benoit J, Foong S, Saumet J, Korkidakis A, Marr K, McQuillan S, Todd N. Fertility preservation in patients undergoing gonadotoxic treatments: a Canadian Fertility and Andrology Society clinical practice guideline. Reprod Biomed Online 2024; 48:103767. [PMID: 38458057 DOI: 10.1016/j.rbmo.2023.103767] [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: 08/12/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 03/10/2024]
Abstract
The management of young patients with cancer presents several unique challenges. In general, these patients are ill prepared for the diagnosis and the impact on their fertility. With the improved survival for all tumour types and stages, the need for adequate fertility counselling and a multidisciplinary approach in the reproductive care of these patients is paramount. Recent advances in cryopreservation techniques allow for the banking of spermatozoa, oocytes, embryos and ovarian tissue without compromising survival. This Canadian Fertility and Andrology Society (CFAS) guideline outlines the current understanding of social and medical issues associated with oncofertility, and the medical and surgical technologies available to optimize future fertility.
Collapse
Affiliation(s)
- Jeffrey E Roberts
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, B.C., Canada.
| | - Janie Benoit
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| | - Shu Foong
- Department of Obstetrics and Gynecology, University of Calgary, Calgary, AB, Canada
| | - Julio Saumet
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| | - Ann Korkidakis
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard University, Boston, MA, USA
| | - Kristin Marr
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, B.C., Canada
| | - Sarah McQuillan
- Department of Obstetrics and Gynecology, University of Calgary, Calgary, AB, Canada
| | - Nicole Todd
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, B.C., Canada
| |
Collapse
|
9
|
Takae S, Harada M, Nakamura K, Furuyama S, Ono M, Osuga Y, Suzuki N. Survey on the implementation status and reproductive outcomes of oocyte and ovarian tissue cryopreservation in Japan: Historical comparison with nationwide surveys. J Obstet Gynaecol Res 2024; 50:709-718. [PMID: 38263596 DOI: 10.1111/jog.15893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024]
Abstract
PURPOSE To clarify the reproductive outcomes of fertility preservation (FP) treatment. METHODS We conducted a mailed-in questionnaire survey at institutions certified by the Japan Society of Obstetrics and Gynecology to investigate the number of oocyte cryopreservations (OC) and ovarian tissue cryopreservations (OTC) performed from December 2016 to the end of 2020. And, we conducted a detailed investigation of cases in which frozen specimens were used during the investigation period, and made historical comparisons with previous nationwide studies. RESULTS Responses were received from 114 out of 150 facilities (response rate: 76.0%) for OC and 43 out of 51 for OTC (response rate: 84.3%). Breast cancer was the most common disease among patients whose FP specimens were used. During the study period, 1237 OCs and 198 OTCs were performed. In addition, 57 cycles of embryo transfer (ET) using cryopreserved oocytes and 12 cases of ovarian tissue transplantation (OTT) were performed. The mean age of patients who underwent ET using cryopreserved oocytes was 34.8 (±5.8) years, with a median age of 36 years. The pregnancy rate per ET using cryopreserved oocytes was 26.3% and the live birth rate (LBR) was 17.5%. Further, the LBR per patient was 43.3%, and the pregnancy rate following OTTs was 33.3%. Also, controlled ovarian stimulation using the random start method or the combination of aromatase inhibitors had no effect on pregnancy outcome. CONCLUSION Implementation of both OCs and OTCs have markedly increased over time in Japan, with comparable reproductive outcomes as other reports.
Collapse
Affiliation(s)
- Seido Takae
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki City, Kanagawa, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kentaro Nakamura
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki City, Kanagawa, Japan
| | - Sayako Furuyama
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki City, Kanagawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki City, Kanagawa, Japan
| |
Collapse
|
10
|
Nair N, More A, Singh BR, Wadkar A, Tilak P. A Successful Pregnancy Following Intracytoplasmic Sperm Injection in a Breast Cancer Survivor: A Case Report. Cureus 2024; 16:e55756. [PMID: 38586701 PMCID: PMC10998928 DOI: 10.7759/cureus.55756] [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: 02/06/2024] [Accepted: 03/07/2024] [Indexed: 04/09/2024] Open
Abstract
This report documents the case of a 36-year-old female diagnosed with stage I invasive ductal carcinoma of the left breast who, alongside her 39-year-old husband, sought fertility assistance at our center due to primary infertility. Having survived cancer twice in the span of their seven-year marriage, the couple faced the challenge of overcoming both the repercussions of cancer treatment and difficulties in conceiving. Initial attempts through three intrauterine insemination (IUI) cycles proved unsuccessful, leading the couple to opt for in vitro fertilization (IVF). The fertility assessment of the husband revealed the presence of several pus cells and a high sperm DNA fragmentation index (DFI). To address this, a medication regimen was administered to improve sperm quality. Concurrently, the female underwent controlled ovarian stimulation (COS) with the anti-estrogen agent letrozole to mitigate the risk of estrogen surges that could compromise her health. Subsequently, oocytes were retrieved from the female, and intracytoplasmic sperm injection (ICSI) was used to facilitate fertilization with her husband's sperm. Following successful embryo development, the patient underwent embryo transfer (ET), resulting in a positive beta-human chorionic gonadotropin (beta-hCG) result, signifying a successful conception. This case report highlights the intricate challenges faced by individuals with a history of breast cancer, emphasizing the delicate balance required in managing infertility in such circumstances. The described approach, involving personalized treatments and meticulous care, underscores the possibility of achieving successful conception for females struggling with fertility issues post-cancer survival. The documented journey serves as a testament to the resilience of individuals facing the dual challenges of cancer survival and infertility, offering insights into the complexities of their reproductive healthcare.
Collapse
Affiliation(s)
- Nancy Nair
- Clinical Embryology, School of Allied Health Sciences, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Akash More
- Clinical Embryology, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Brij Raj Singh
- Anatomy, Datta Meghe Institute of Medical Science, Wardha, IND
| | - Achyut Wadkar
- Anatomy, Datta Meghe Institute of Medical Science, Wardha, IND
| | - Priyal Tilak
- Clinical Embryology, School of Allied Health Sciences, Datta Meghe Institute of higher Education and Research, Wardha, IND
| |
Collapse
|
11
|
Hatirnaz S, Hatirnaz E, Urkmez SS, Calıskan CS, Celik S, Hatirnaz K, Cao M, Tan SL, Dahan MH. Oocyte in-vitro maturation primed with letrozole-HCG versus FSH-HCG in women with oocyte maturation abnormalities: a retrospective study. Reprod Biomed Online 2024; 48:103620. [PMID: 38194886 DOI: 10.1016/j.rbmo.2023.103620] [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: 07/24/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 01/11/2024]
Abstract
RESEARCH QUESTION Are there differences between in-vitro maturation (IVM) primed with letrozole-human chorionic gonadotrophin (HCG) and IVM primed with FSH-HCG in women with oocyte maturation abnormalities (OMAs), defined as at least two failed IVF cycles where immature oocytes were retrieved? DESIGN This retrospective study was conducted at a private fertility clinic from January 2009 to April 2023. The final analysis included 75 women in Group 1 (IVM primed with FSH-HCG) and 52 women in Group 2 (IVM primed with letrozole-HCG). RESULTS A significantly higher median number of oocytes was obtained in Group 1 compared with Group 2 {9 [interquartile range (IQR) 1-5] versus 5 (IQR 1-18); P < 0.001}. However, no differences in oocyte maturation stage at collection were found between the groups (P > 0.05). At the end of IVM, Group 1 had 73/666 mature oocytes and Group 2 had 106/322 mature oocytes, and the median metaphase II oocyte rate per patient was higher in Group 2 [33.3% (IQR 66.7-100.0%) versus 0.0% (IQR 0.0-22.2%); P < 0.001]. Moreover, Group 2 demonstrated a higher median fertilization rate [66.7% (IQR 50.0-100.0%) versus 50.0% (IQR 0.0-66.7%); P = 0.027]. Group 2 had a higher proportion of Grade 2 embryos (58.5% versus 6.3%), and Group 1 had a higher proportion of Grade 3 embryos (93.8% vs 24.4%; P < 0.001). Notably, all pregnancies obtained in the study were in Group 2 (5 versus 0; P = 0.042). CONCLUSIONS IVM primed with letrozole-HCG in women with prior failed IVF cycles due to OMAs may result in mature oocytes, clinical pregnancies and live births. The effectiveness of letrozole priming for the subtypes of OMAs needs further investigation, with studies including greater numbers of cases.
Collapse
Affiliation(s)
| | | | - Sebati Sinan Urkmez
- Department of Biochemistry, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey.
| | - Canan Soyer Calıskan
- Department of Obstetrics and Gynaecology, Samsun Training and Research Hospital, Samsun, Turkey
| | - Samettin Celik
- Department of Obstetrics and Gynaecology, Samsun Training and Research Hospital, Samsun, Turkey
| | - Kaan Hatirnaz
- Department of Molecular Biology and Genetics, Faculty of Science, Ondokuz Mayıs University, Samsun, Turkey
| | - Mingju Cao
- OriginElle Fertility Clinic and Women's Health Center, Montreal, Quebec, Canada
| | - Seang Lin Tan
- OriginElle Fertility Clinic and Women's Health Center, Montreal, Quebec, Canada
| | - Michael H Dahan
- Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Taniskidou AM, Voultsos P, Tarlatzis V, Timotheadou E. Perceptions and experiences of fertility preservation in female patients with cancer in Greece. BMC Womens Health 2024; 24:108. [PMID: 38336757 PMCID: PMC10858603 DOI: 10.1186/s12905-024-02955-x] [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: 07/06/2023] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND As advances in oncology have led to remarkable and steady improvements in the survival rates of patients with cancer and anticancer treatment can cause premature ovarian failure in women, fertility preservation (FP) has become a global public health concern and an integral part of the care for women diagnosed with cancer during reproductive age. However, for various reasons, FP remains underutilized for patients with cancer. There are substantial gaps in our knowledge about women's experiences and perceptions of the issue. This study aims to contribute to bridging that gap. METHODS This prospective qualitative study was conducted from March 2018 to February 2023. A combination of purposive and snowball sampling was used. Data were collected by semistructured interviews with nineteen reproductive-age women who had been recently diagnosed with cancer. Data were classified and analysed with a thematic analysis approach. RESULTS A variety of distinct themes and subthemes emerged from the analysis of the interview data. The cancer diagnosis emerged as a factor that considerably affects the women's attitudes towards biological parenthood: It can further increase their (strong) previous desire or decrease their previous (weak) desire. Women with a recent cancer diagnosis had not received adequate and multidisciplinary counselling, including clear and sufficient information. However, participants felt satisfied with the information they received because they either received the information they requested or remained in denial about the need to be informed (i.e., because they felt overwhelmed after the cancer diagnosis). Embryo cryopreservation emerged as a less desirable FP option for women with cancer. Participants showed respect for human embryos, not always for religious reasons. Surrogacy emerged as the last resort for most participants. Religious, social or financial factors did play a secondary (if any) role in women's decision-making about FP. Finally, male partners' opinions played a secondary role in most participants' decision-making about FP. If embryo cryopreservation was the selected option, partners would have a say because they were contributing their genetic material. CONCLUSIONS The findings that emerged from the data analysis were partly consistent with prior studies. However, we identified some interesting nuances that are of clinical importance. The results of this study may serve as a starting point for future research.
Collapse
Affiliation(s)
- Anna-Maria Taniskidou
- Laboratory of Forensic Medicine & Toxicology (Division: Medical Law and Ethics), School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, Thessaloniki, GR, 54124, Greece
| | - Polychronis Voultsos
- Laboratory of Forensic Medicine & Toxicology (Division: Medical Law and Ethics), School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, Thessaloniki, GR, 54124, Greece.
| | - Vasileios Tarlatzis
- 1st Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, Thessaloniki, GR, 54124, Greece
| | - Eleni Timotheadou
- Department of Medical Oncology, School of Medicine, Faculty of Health Sciences, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
13
|
Gao F, Han J, Jia L, He J, Wang Y, Chen M. Circ_0001982 aggravates breast cancer development through the circ_0001982-miR-144-3p-GSE1 axis. J Biochem Mol Toxicol 2024; 38:e23565. [PMID: 37867456 DOI: 10.1002/jbt.23565] [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: 07/07/2022] [Revised: 11/29/2022] [Accepted: 10/09/2023] [Indexed: 10/24/2023]
Abstract
This study was designed to explore the role of circ_0001982 in breast cancer (BC) development. Quantitative real-time polymerase chain reaction and western blot analysis assays were used to determine circ_0001982, miR-144-3p, and gse1 coiled-coil protein (GSE1) expression. Functional assays were performed to evaluate cell proliferation, apoptosis, migration, and invasion. The glycolysis was analyzed with commercial kits. Dual-luciferase reporter assay and RNA immunoprecipitation assays were conducted to analyze the relationships among circ_0001982, miR-144-3p, and GSE1. A murine xenograft model assay was performed to determine circ_0001982-induced effects on BC cell tumor properties in vivo. Circ_0001982 expression was upregulated, but miR-144-3p was reduced in BC tissues and cells in comparison with normal breast tissues and normal human mammary epithelial cells. Circ_0001982 knockdown or miR-144-3p overexpression inhibited BC cell proliferation, glycolysis, migration and invasion, and promoted apoptosis. Circ_0001982 sponged miR-144-3p and negatively regulated miR-144-3p expression in BC cells. In addition, GSE1 was identified as a target mRNA of miR-144-3p. Ectopic GSE1 expression relieved circ_0001982 depletion-induced effects on BC cell tumor properties. Furthermore, circ_0001982 absence suppressed BC cell tumor properties in vivo. Circ_0001982 contributed to the BC cell tumor properties by regulating the miR-144-3p-GSE1 axis.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
| | - Jianjun Han
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
| | - Li Jia
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
| | - Jun He
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
| | - Yun Wang
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
| | - Mi Chen
- Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
| |
Collapse
|
14
|
Sii S, Polyakov A, Rozen G, Agresta F, Stern K. Controlled ovarian hyperstimulation in breast cancer patients: Does oestrogen receptor status make a difference? Aust N Z J Obstet Gynaecol 2023; 63:774-779. [PMID: 37317583 DOI: 10.1111/ajo.13721] [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: 12/20/2022] [Accepted: 06/01/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND The presence of different breast cancer receptor status may impact ovarian stimulation outcomes. AIM To study the association between oestrogen receptor (ER) status in breast cancer patients and fertility preservation outcomes in a major tertiary referral centre. MATERIALS AND METHODS Women who underwent fertility preservation following the diagnosis of breast cancer from 2008 to 2018 were included in the study. Patient age, ovarian stimulation parameters and laboratory outcomes were recorded and compared between the ER positive and negative groups. The primary outcome was total number of oocytes frozen. Secondary outcomes included total number of oocytes collected, mature oocytes, and embryos frozen. RESULTS The women included in the study (n = 214) were analysed in the following groups based on their fertility preservation method: oocyte freezing (n = 131), embryo freezing (n = 70), and both embryo and oocyte freezing (n = 13). There was an increase in the mean (but not mature) number of oocytes frozen (12.4 and 9.2, P-value = 0.03) favouring the ER positive group, even though the women in this group were older (35.0 and 33.4, P-value of 0.03). There is no difference in the starting follicle-stimulating hormone dose, duration of stimulation, mature oocytes collected, and embryos frozen in both groups. CONCLUSION Patients with ER positive breast cancer may have more positive ovarian stimulation outcomes.
Collapse
Affiliation(s)
| | - Alex Polyakov
- Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Genia Rozen
- Royal Women's Hospital, Melbourne, Victoria, Australia
| | | | - Kate Stern
- Royal Women's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Harris CJ, Rowell EE, Jayasinghe Y, Cost C, Childress KJ, Frederick NN, McNally O, Appiah L, Anazodo A. Pediatric, adolescent, and young adult breast and reproductive tumors. Pediatr Blood Cancer 2023; 70 Suppl 5:e29422. [PMID: 36458682 DOI: 10.1002/pbc.29422] [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: 06/17/2020] [Revised: 08/03/2021] [Accepted: 08/17/2021] [Indexed: 12/04/2022]
Abstract
Tumors of the breast and reproductive organs that occur in children, adolescents, and young adults (AYA) have different biological features and can present special challenges. Although prognosis for these tumors is generally favorable, the long-term effects of treatment can be debilitating. Treatments are often multimodal and may include surgery as well as chemotherapy and/or radiation, which can cause considerable distress and anxiety related to loss of femininity or masculinity, concern over future fertility, or sexual dysfunction. Thus, tumors of the reproductive organs in pediatric/AYA patients require special consideration of the treatment effects beyond the intended oncologic outcome. Multidisciplinary teams should be involved in their care and address issues of fertility, sexual dysfunction, and psychosexual concerns before treatment begins. This review addresses histology, risk factors, prognosis, staging and treatment of gynecologic, breast and testicular cancers in pediatric and AYA patients.
Collapse
Affiliation(s)
- Courtney J Harris
- Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Erin E Rowell
- Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Yasmin Jayasinghe
- Royal Women's Hospital, Parkville, Victoria, Australia
- Royal Children's Hospital, Parkville, Victoria, Australia
- University of Melbourne, Parkville, Victoria, Australia
| | - Carrye Cost
- Children's Hospital Colorado, Aurora, Colorado
| | - Krista J Childress
- Children's Healthcare of Atlanta, Atlanta, Georgia
- Emory University, Atlanta, Georgia
| | - Natasha N Frederick
- Department of Pediatrics and the Center for Cancer and Blood Disorders, Connecticut Children's Medical Center, Hartford, Connecticut
- University of Connecticut School of Medicine, Farmington, Connecticut
| | - Orla McNally
- Royal Women's Hospital, Parkville, Victoria, Australia
- University of Melbourne, Parkville, Victoria, Australia
| | | | - Antoinette Anazodo
- Sydney Children's Hospital, Sydney, New South Wales, Australia
- Prince of Wales Hospital, Sydney, New South Wales, Australia
- School of Women's and Children's, University of New South Wales, High St Kensington, New South Wales, Australia
| |
Collapse
|
16
|
Romero SAD, Palomino H, Ahmed SH, Peacher D, Urias A, Ramirez L, Yocupicio J, Gutierrez P, Flores Ortega RE, Reyes B, Kaiser BN, Hoyt H, Su HI. Intervening on women's health for rural young breast cancer survivors: A study protocol. Contemp Clin Trials 2023; 130:107215. [PMID: 37164298 PMCID: PMC10723631 DOI: 10.1016/j.cct.2023.107215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
INTRODUCTION From diagnosis to post-treatment, many young breast cancer survivors (YBCS) experience infertility, limited contraception choices, concern about pregnancy safety, and menopausal symptoms. Clinical guidelines recommend oncofertility care (counseling and/or clinical services that meet fertility, contraception, pregnancy health and/or menopausal symptom management needs) throughout the cancer care continuum. However, significant oncofertility care gaps exist in rural, community oncology settings. MATERIALS AND METHODS We describe the design of an interrupted time series, effectiveness-implementation hybrid clinical trial that evaluates a multi-component intervention to improve YBCS engagement in oncofertility care. The intervention is comprised of 1) oncology clinic-based oncofertility needs screen; 2) a women's health survivorship care plan in Spanish and English; 3) remote patient navigation; and 4) telehealth oncofertility consultation. During the pre-intervention period (12 months), usual care will be delivered. During the intervention period (15 months), the multi-component intervention will be implemented at two rural oncology clinics with largely Latina, Spanish-speaking populations. The primary outcome of YBCS (n = 135) engagement in oncofertility care will be collected from medical record review. We will also collect validated patient-reported outcomes. Informed by the Exploration Preparation Implementation Sustainment (EPIS) implementation science framework, we will integrate qualitative and quantitative data to explore whether and how the intervention was effective, acceptable, appropriate, and delivered with fidelity. DISCUSSION Our overall goal is to speed implementation of a scalable oncofertility care intervention for YBCS in underserved areas to reduce disparities and improve reproductive health and quality of life. TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT05414812.
Collapse
Affiliation(s)
- Sally A D Romero
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, 9300 Campus Point, MC 7433, La Jolla, CA 92037-7433, United States of America.
| | - Helen Palomino
- Cancer Resource Center of the Desert, 444 S 8th St Ste B3, El Centro, CA 92243, United States of America
| | - Syed H Ahmed
- El Centro Regional Medical Center, 1415 Ross Avenue, El Centro, CA 92243, United States of America
| | - Diana Peacher
- Cancer Resource Center of the Desert, 444 S 8th St Ste B3, El Centro, CA 92243, United States of America
| | - Aday Urias
- Cancer Resource Center of the Desert, 444 S 8th St Ste B3, El Centro, CA 92243, United States of America
| | - Lourdes Ramirez
- Cancer Resource Center of the Desert, 444 S 8th St Ste B3, El Centro, CA 92243, United States of America
| | - Jessica Yocupicio
- Cancer Resource Center of the Desert, 444 S 8th St Ste B3, El Centro, CA 92243, United States of America
| | - Priscilla Gutierrez
- Cancer Resource Center of the Desert, 444 S 8th St Ste B3, El Centro, CA 92243, United States of America; Pioneers Memorial Healthcare District, 197 W Legion Rd, Brawley, CA 92227, United States of America
| | - Ricardo E Flores Ortega
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, 9300 Campus Point, MC 7433, La Jolla, CA 92037-7433, United States of America
| | - Breanna Reyes
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, 9300 Campus Point, MC 7433, La Jolla, CA 92037-7433, United States of America
| | - Bonnie N Kaiser
- Department of Anthropology and Global Health Program, University of California San Diego, 9500 Gilman Drive, MC 0532, La Jolla, CA 92093-0532, United States of America
| | - Helina Hoyt
- San Diego State University, Imperial Valley, School of Nursing, 560 CA-78, Brawley, CA 92227, United States of America
| | - H Irene Su
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, 9300 Campus Point, MC 7433, La Jolla, CA 92037-7433, United States of America; Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, MC 0901, La Jolla, CA 92093-0901, United States of America
| |
Collapse
|
17
|
Hong YH, Park C, Paik H, Lee KH, Lee JR, Han W, Park S, Chung S, Kim HJ. Fertility Preservation in Young Women With Breast Cancer: A Review. J Breast Cancer 2023; 26:221-242. [PMID: 37387349 DOI: 10.4048/jbc.2023.26.e28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 07/01/2023] Open
Abstract
Fertility preservation is a major concern in young patients diagnosed with breast cancer and planning to receive multimodality treatment, including gonadotoxic chemotherapy with or without age-related decline through long-term endocrine therapy. Most breast cancer patients undergo multimodality treatments; many short-term and long-term side effects arise during these therapies. One of the most detrimental side effects is reduced fertility due to gonadotoxic treatments with resultant psychosocial stress. Cryopreservation of oocytes, embryos, and ovarian tissue are currently available fertility preservation methods for these patients. As an adjunct to these methods, in vitro maturation or gonadotropin-releasing hormone agonist could also be considered. It is also essential to communicate well with patients in the decision-making process on fertility preservation. It is essential to refer patients diagnosed with breast cancer on time to fertility specialists for individualized treatment, which may lead to desirable outcomes. To do so, a multimodal team-based approach and in-depth discussion on the treatment of breast cancer and fertility preservation is crucial. This review aims to summarize infertility risk related to currently available breast cancer treatment, options for fertility preservation and its details, barriers to oncofertility counseling, and psychosocial issues.
Collapse
Affiliation(s)
- Yeon Hee Hong
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Changhee Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Haerin Paik
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Jung Ryeol Lee
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Wonshik Han
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seho Park
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seockhoon Chung
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Hee Jeong Kim
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
18
|
Castiglione Morelli MA, Iuliano A, Matera I, Viggiani L, Schettini SCA, Colucci P, Ostuni A. A Pilot Study on Biochemical Profile of Follicular Fluid in Breast Cancer Patients. Metabolites 2023; 13:metabo13030441. [PMID: 36984881 PMCID: PMC10054828 DOI: 10.3390/metabo13030441] [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: 02/15/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Breast cancer (BC) is the most common type of cancer among women in almost all countries worldwide and is one of the oncological pathologies for which is indicated fertility preservation, a type of procedure used to help keep a person's ability to have children. Follicular fluid (FF) is a major component of oocyte microenvironment, which is involved in oocyte growth, follicular maturation, and in communication between germ and somatic cells; furthermore, it accumulates all metabolites during oocytes growth. To obtain information about changes on fertility due to cancer, we aimed at investigating potential biomarkers to discriminate between FF samples obtained from 16 BC patients and 10 healthy women undergoing in vitro fertilization treatments. An NMR-based metabolomics approach was performed to investigate the FF metabolic profiles; ELISA and western blotting assays were used to investigate protein markers of oxidative and inflammatory stress, which are processes closely related to cancer. Our results seem to suggest that FFs of BC women display some significant metabolic alterations in comparison to healthy controls, and these variations are also related with tumor staging.
Collapse
Affiliation(s)
| | - Assunta Iuliano
- Center for Reproductive Medicine of "San Carlo" Hospital, 85100 Potenza, Italy
| | - Ilenia Matera
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| | - Licia Viggiani
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| | | | - Paola Colucci
- Center for Reproductive Medicine of "San Carlo" Hospital, 85100 Potenza, Italy
| | - Angela Ostuni
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| |
Collapse
|
19
|
Suzuki R, Horage-Okutsu Y, Kawahara T, Nakamura K, Shiraishi E, Iwahata H, Suzuki-Takahashi Y, Sugishita Y, Takae S, Suzuki N. The effect of aromatase inhibitor on controlled ovarian stimulation for oocyte cryopreservation in adolescent and young cancer patients. J Obstet Gynaecol Res 2023; 49:973-979. [PMID: 36606606 DOI: 10.1111/jog.15535] [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: 09/28/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023]
Abstract
AIM To determine the impact of aromatase inhibitor (AI) use in oocyte cryopreservation among Japanese adolescent and young adult (AYA) cancer patients for fertility preservation, we evaluated the oocyte cryopreservation outcomes following AI therapy in combination with the follicular phase start (FPS) and random start (RS) protocols. METHODS This retrospective study included 81 cycles of controlled ovarian stimulation (COS) among 73 AYA patients with cancer who underwent oocyte cryopreservation to maintain fertility. The outcome measures were the total number of matured oocytes that were retrieved and cryopreserved, as well as their maturation rates. The AI (+) and AI (-) groups were compared using the RS and FPS protocols. RESULTS Our results showed that the combined use of AI and COS decreases serum E2 levels and maintains the number of retrieved and cryopreserved mature oocytes. We also confirmed the efficacy of the RS protocol, which was found to have comparable outcomes to that of the FPS protocol in both AI (+) and AI (-) groups. CONCLUSION The combined use of AI and COS is beneficial for oocyte cryopreservation in patients with estrogen-sensitive cancer, regardless of the menstrual cycle phase of COS initiation.
Collapse
Affiliation(s)
- Reiko Suzuki
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| | - Yuki Horage-Okutsu
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| | - Tai Kawahara
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| | - Kentaro Nakamura
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| | - Eriko Shiraishi
- Department of Obstetrics and Gynecology, The Jikei University school of medicine, Tokyo, Japan
| | - Hideyuki Iwahata
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| | - Yuki Suzuki-Takahashi
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| | - Yodo Sugishita
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| | - Seido Takae
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, Saint Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
20
|
Sonigo C, Amsellem N, Mayeur A, Laup L, Pistilli B, Delaloge S, Eustache F, Sifer C, Rakrouki S, Benoit A, Peigné M, Grynberg M. Disease-free survival does not differ according to fertility preservation technique for young women with breast cancer. Fertil Steril 2023; 119:465-473. [PMID: 36473609 DOI: 10.1016/j.fertnstert.2022.11.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022]
Abstract
OBJECTIVE To study whether fertility preservation strategies using ovarian stimulation or without using it impact long-term disease-free survival of patients with breast cancer. DESIGN Retrospective bicentric cohort study. SETTING Two university hospitals. PATIENT(S) In this study, 740 women with breast cancer, aged 18-43 years, who received primary fertility preservation between 2013 and 2019 after a diagnosis of localized breast cancer were included. INTERVENTION(S) Overall, 328 patients underwent at least 1 ovarian stimulation cycle (STIM group) and 412 had a technique without hormonal administration (no STIM group). MAIN OUTCOME MEASURE(S) Disease-free survival and overall survival up to May 2021 were compared between the 2 groups by log-rank test. Cox proportional-hazard regression model was used for multivariable analyses. RESULT(S) Out of the 740 women who underwent fertility preservation, follow-up data were available for 269 women in the STIM group (82%) and 330 (80%) in the no STIM group. Kaplan-Meier estimates of disease-free survival at 4 years were 87.9% (82.8%-92.2%) and 83.1% (78.4%-87.3%) in the STIM and no STIM groups, respectively. After adjustment on prognostic parameters, no significant difference in breast cancer recurrence rate was observed between the STIM and no STIM groups (hazard ratios, 0.83 [0.64-1.08]). Kaplan-Meier estimate of overall survival at 4 years was 97.6% (95.3%-99.2%) and 93.6% (90.9%-95.9%) in the STIM and no STIM groups, respectively. Overall survival was higher in the STIM group than no STIM group (log-rank test). After adjustment on prognostic parameters, the risk of death remained significantly lower in the STIM group (Hazard Ratio, 0.55 [0.35-0.85]). CONCLUSION(S) In our cohort, STIM for fertility preservation in breast cancer did not significantly impact disease-free survival but was associated with higher overall survival. The disease-free survival and overall survival of young patients with breast cancer were not impacted by fertility preservation techniques irrespective of the timing of chemotherapy (neoadjuvant or adjuvant) and the use of ovarian stimulation. Nevertheless, because death and recurrence were rare events, these results should be taken with caution.
Collapse
Affiliation(s)
- Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France; Universite Paris-Saclay, Inserm, Physiologie et physiopathologie endocrinienne, Le Kremlin-Bicetre, France.
| | - Noémi Amsellem
- Department of Reproductive Medicine and Fertility Preservation, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France
| | - Anne Mayeur
- Histology-Embryology-Cytogenetic Laboratory, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France
| | - Laetitia Laup
- Department of Reproductive Medicine and Fertility Preservation, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Barbara Pistilli
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| | - Suzette Delaloge
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| | - Florence Eustache
- Department of Biology of Reproduction and CECOS, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Christophe Sifer
- Department of Biology of Reproduction and CECOS, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Sophia Rakrouki
- Department of Reproductive Medicine and Fertility Preservation, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Alexandra Benoit
- Department of Reproductive Medicine and Fertility Preservation, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France
| | - Maeliss Peigné
- Department of Reproductive Medicine and Fertility Preservation, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Universite Paris-Saclay, Assistance Publique Hopitaux de Paris, Antoine Beclere Hospital, Clamart, France; Department of Reproductive Medicine and Fertility Preservation, Universite Sorbonne Paris Nord, Assistance Publique - Hopitaux de Paris, Jean Verdier Hospital, Bondy, France
| |
Collapse
|
21
|
Osuka S, Kasahara Y, Iyoshi S, Sonehara R, Myake N, Muraoka A, Nakamura T, Iwase A, Kajiyama H. Follicle development and its prediction in patients with primary ovarian insufficiency: Possible treatments and markers to maximize the ability to conceive with residual follicles. Reprod Med Biol 2023; 22:e12556. [PMID: 38144239 PMCID: PMC10746865 DOI: 10.1002/rmb2.12556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/16/2023] [Accepted: 12/11/2023] [Indexed: 12/26/2023] Open
Abstract
Background Primary ovarian insufficiency (POI) is characterized by the development of hypergonadotropic hypogonadism before 40 years of age and leads to intractable infertility. Although in vitro fertilization and embryo transfer with donated eggs enables pregnancy, not a few patients desire pregnancy using their oocytes. However, follicular development is rare and unpredictable in patients with POI. Thus, there is a need for treatments that promote the development of residual follicles and methods to accurately predict infrequent ovulation. Methods This review discusses the effects of various treatments for obtaining eggs from POI patients. Furthermore, this study focused a potential marker for predicting follicular growth in patients with POI. Main Findings Different treatments such as hormone-replacement therapy, dehydroepiandrosterone supplementation, platelet-rich plasma injection, and in vitro activation have shown varying degrees of effectiveness in retrieving oocytes from patients with POI. To predict follicle development in the cycle, elevated serum estradiol and reduced follicle-stimulating hormone (FSH) levels are important. However, these markers are not always reliable under continuous estradiol-replacement therapy. As a novel marker for predicting follicle growth, serum anti-Müllerian hormone (AMH) levels, measured using the picoAMH enzyme-linked immunosorbent assay, were found to predict follicle growth in patients and the cycle. Conclusion This review highlights the challenges and available interventions for achieving pregnancy using a patient's oocytes in cases of POI. We believe that a combination of currently available treatments and prediction methods is the best strategy to enable patients with POI to conceive using their own eggs. Although AMH levels may predict follicle growth, further research is necessary to improve the chances of successful follicular development and conception in patients with POI.
Collapse
Affiliation(s)
- Satoko Osuka
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | | | - Shohei Iyoshi
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Advanced ResearchNagoya UniversityNagoyaJapan
| | - Reina Sonehara
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Natsuki Myake
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Ayako Muraoka
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Perinatology, Center for Maternal‐Neonatal CareNagoya University HospitalNagoyaJapan
| | - Tomoko Nakamura
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Perinatology, Center for Maternal‐Neonatal CareNagoya University HospitalNagoyaJapan
| | - Akira Iwase
- Department of Obstetrics and GynecologyGunma University Graduate School of MedicineMaebashiJapan
| | - Hiroaki Kajiyama
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
22
|
Meng Y, Cao J, Li Y, Duan S, Zhou Z, Li J, Ousmane D, Ou C, Wang J. Emerging role of ferroptosis-related circular RNA in tumor metastasis. Front Pharmacol 2023; 14:1168458. [PMID: 37168995 PMCID: PMC10164976 DOI: 10.3389/fphar.2023.1168458] [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: 02/17/2023] [Accepted: 04/14/2023] [Indexed: 05/13/2023] Open
Abstract
Tumor metastasis is an important factor that contributes to the poor prognosis of patients with tumors. Therefore, to solve this problem, research on the mechanism of metastasis is essential. Ferroptosis, a new mode of cell death, is characterized by membrane damage due to lipid peroxidation caused by iron overload. Many studies have shown that excessive ferroptosis can affect tumor metastasis and thus inhibit tumor progression. Recently, circular RNA (circRNA), a type of non-coding RNA, has been shown to be associated with the progression of ferroptosis, thus influencing tumor development. However, the specific mechanisms by which circRNAs affect the progression of ferroptosis and their roles in tumor metastasis are not known. In this review, we systematically discuss the role of circRNAs in regulating tumor ferroptosis and their mechanism of action through sponging miRNAS in various tumors, thereby impacting metastasis. This review helps elucidate the relationship and role of ferroptosis-related circRNAs in tumor metastasis and may provide future researchers with new ideas and directions for targeted therapies.
Collapse
Affiliation(s)
- Yifei Meng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- Department of Pathology, Ultrapathology (Biomedical Electron Microscopy) Center, Xiangya Hospital, Central South University, Changsha City, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jingdong Cao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- Department of Pathology, Ultrapathology (Biomedical Electron Microscopy) Center, Xiangya Hospital, Central South University, Changsha City, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yidan Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- Department of Pathology, Ultrapathology (Biomedical Electron Microscopy) Center, Xiangya Hospital, Central South University, Changsha City, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Saili Duan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- Department of Pathology, Ultrapathology (Biomedical Electron Microscopy) Center, Xiangya Hospital, Central South University, Changsha City, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zongjiang Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
| | - Jinghe Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
| | - Diabate Ousmane
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Junpu Wang, ; Chunlin Ou,
| | - Junpu Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- Department of Pathology, Ultrapathology (Biomedical Electron Microscopy) Center, Xiangya Hospital, Central South University, Changsha City, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Junpu Wang, ; Chunlin Ou,
| |
Collapse
|
23
|
Rives N, Courbière B, Almont T, Kassab D, Berger C, Grynberg M, Papaxanthos A, Decanter C, Elefant E, Dhedin N, Barraud-Lange V, Béranger MC, Demoor-Goldschmidt C, Frédérique N, Bergère M, Gabrel L, Duperray M, Vermel C, Hoog-Labouret N, Pibarot M, Provansal M, Quéro L, Lejeune H, Methorst C, Saias J, Véronique-Baudin J, Giscard d'Estaing S, Farsi F, Poirot C, Huyghe É. What should be done in terms of fertility preservation for patients with cancer? The French 2021 guidelines. Eur J Cancer 2022; 173:146-166. [PMID: 35932626 DOI: 10.1016/j.ejca.2022.05.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 11/03/2022]
Abstract
AIM To provide practice guidelines about fertility preservation (FP) in oncology. METHODS We selected 400 articles after a PubMed review of the literature (1987-2019). RECOMMENDATIONS Any child, adolescent and adult of reproductive age should be informed about the risk of treatment gonadotoxicity. In women, systematically proposed FP counselling between 15 and 38 years of age in case of treatment including bifunctional alkylating agents, above 6 g/m2 cyclophosphamide equivalent dose (CED), and for radiation doses on the ovaries ≥3 Gy. For postmenarchal patients, oocyte cryopreservation after ovarian stimulation is the first-line FP technique. Ovarian tissue cryopreservation should be discussed as a first-line approach in case of treatment with a high gonadotoxic risk, when chemotherapy has already started and in urgent cases. Ovarian transposition is to be discussed prior to pelvic radiotherapy involving a high risk of premature ovarian failure. For prepubertal girls, ovarian tissue cryopreservation should be proposed in the case of treatment with a high gonadotoxic risk. In pubertal males, sperm cryopreservation must be systematically offered to any male who is to undergo cancer treatment, regardless of toxicity. Testicular tissue cryopreservation must be proposed in males unable to cryopreserve sperm who are to undergo a treatment with intermediate or severe risk of gonadotoxicity. In prepubertal boys, testicular tissue preservation is: - recommended for chemotherapy with a CED ≥7500 mg/m2 or radiotherapy ≥3 Gy on both testicles. - proposed for chemotherapy with a CED ≥5.000 mg/m2 or radiotherapy ≥2 Gy. If several possible strategies, the ultimate choice is made by the patient.
Collapse
Affiliation(s)
- Nathalie Rives
- Normandie Univ, UNIROUEN, Team "Adrenal and Gonadal Physiopathology" Inserm U1239 Nordic, Rouen University Hospital, Biology of Reproduction-CECOS Laboratory, Rouen, France
| | - Blandine Courbière
- Reproductive Medicine and Biology Department, Assistance Publique Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Thierry Almont
- Cancerology, Urology, Hematology Department, Centre Hospitalier Universitaire de Martinique, Fort-de-France, Martinique, France; General Cancer Registry of Martinique UF1441, Centre Hospitalier Universitaire de Martinique, Fort-de-France, Martinique, France
| | - Diana Kassab
- Methodology Unit, Association Française d'Urologie, Paris, Ile-de-France, France
| | - Claire Berger
- Department of Pediatric Hematology and Oncology, University-Hospital of Saint-Etienne, Hospital, Nord Saint-Etienne cedex 02, France 42055; Childhood Cancer Registry of the Rhône-Alpes Region, University of Saint-Etienne, 15 rue Ambroise Paré, Saint-Etienne cedex 02, France 42023
| | - Michaël Grynberg
- Reproductive Medicine and Fertility Department, Hôpital Antoine-Beclère, Clamart, Île-de-France, France
| | - Aline Papaxanthos
- Reproductive Medicine and Biology Department, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, Aquitaine, France
| | - Christine Decanter
- Medically Assisted Procreation and Fertility Preservation Department, Centre Hospitalier Régional Universitaire de Lille, Lille, Hauts-de-France, France
| | - Elisabeth Elefant
- Reference Center for Teratogenic Agents, Hôpital Armand-Trousseau Centre de Référence sur les Agents Tératogènes, Paris, Île-de-France, France
| | - Nathalie Dhedin
- Adolescents and Young Adults Unit, Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris, France
| | - Virginie Barraud-Lange
- Reproductive Medicine and Biology Department, Hôpital Cochin, Paris, Île-de-France, France
| | | | | | - Nicollet Frédérique
- Information and Promotion Department, Association Laurette Fugain, Paris, France
| | - Marianne Bergère
- Human Reproduction, Embryology and Genetics Directorate, Agence de la biomédecine, La Plaine Saint-Denis, France
| | - Lydie Gabrel
- Good Practices Unit - Guidelines and Medicines Directorate, Institut National du Cancer, Billancourt, Île-de-France, France
| | - Marianne Duperray
- Guidelines and Drug Directorate, Institut National du Cancer, Billancourt, Île-de-France, France
| | - Christine Vermel
- Expertise Quality and Compliance Mission - Communication and Information Directorate, Institut National du Cancer, Billancourt, Île-de-France, France
| | - Natalie Hoog-Labouret
- Research and Innovation, Institut National du Cancer, Billancourt, Île-de-France, France
| | - Michèle Pibarot
- OncoPaca-Corse Regional Cancer Network, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Magali Provansal
- Medical Oncology Department, Institut Paoli-Calmettes, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Laurent Quéro
- Cancerology and Radiotherapy Department, Hôpital Saint Louis, AP-HP, Paris, France
| | - Hervé Lejeune
- Reproductive Medicine and Biology Department, Hospices Civils de Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Charlotte Methorst
- Reproductive Medicine and Biology Department, Centre Hospitalier des Quatre Villes - Site de Saint-Cloud, Saint-Cloud, France
| | - Jacqueline Saias
- Reproductive Medicine and Biology Department, Assistance Publique Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Jacqueline Véronique-Baudin
- Cancerology, Urology, Hematology Department, Centre Hospitalier Universitaire de Martinique, Fort-de-France, Martinique, France; General Cancer Registry of Martinique UF1441, Centre Hospitalier Universitaire de Martinique, Fort-de-France, Martinique, France
| | - Sandrine Giscard d'Estaing
- Reproductive Medicine and Biology Department, Hospices Civils de Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Fadila Farsi
- Regional Cancer Network, Réseau Espace Santé Cancer, Lyon, Rhône-Alpes, France
| | - Catherine Poirot
- Adolescents and Young Adults Unit, Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris, France
| | - Éric Huyghe
- Urology Department, Centre Hospitalier Universitaire de Toulouse, Toulouse, France; Laboratoire Développement Embryonnaire, Fertilité et Environnement (DEFE) UMR 1203, Université Toulouse 3 Paul Sabatier, Toulouse, France.
| |
Collapse
|
24
|
Marklund A, Lekberg T, Hedayati E, Liljegren A, Bergh J, Lundberg FE, Rodriguez-Wallberg KA. Relapse Rates and Disease-Specific Mortality Following Procedures for Fertility Preservation at Time of Breast Cancer Diagnosis. JAMA Oncol 2022; 8:1438-1446. [PMID: 36006625 PMCID: PMC9412846 DOI: 10.1001/jamaoncol.2022.3677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Question Is fertility preservation at time of breast cancer diagnosis associated with any increased risk of disease-specific relapse or mortality? Findings In this population-based Swedish nationwide cohort study that included 1275 women with breast cancer, fertility preservation at time of breast cancer diagnosis was not statistically significantly associated with any increased risk of disease-specific mortality or relapse. Meaning Findings of this study support the safety of fertility preservation in women with breast cancer, which is highly relevant for reproductive counseling of women with breast cancer diagnosed at a young age. Importance Breast cancer (BC) is the most common indication for fertility preservation (FP) in women of reproductive age. Procedures for FP often include hormonal stimulation, but current data are scarce regarding whether using hormonal stimulation for FP is associated with any deterioration in BC prognosis. Objective To investigate the risk of disease-specific mortality and relapse in women who underwent FP with or without hormonal stimulation compared with women who did not at time of BC diagnosis. Design, Setting, and Participants This Swedish nationwide prospective cohort study was conducted to assess the safety of hormonal and nonhormonal FP procedures indicated by BC in Sweden from January 1, 1994, through June 30, 2017. Women were identified from any of the regional FP programs located at Swedish university hospitals. A total of 425 women were found to have undergone FP, and 850 population comparators who had not undergone FP were sampled from regional BC registers and matched on age, calendar period of diagnosis, and region. Relapse-free survival was assessed in a subcohort of 241 women who underwent FP and 482 women who had not, with complete data. Nationwide demographic and health care registers provided data on outcome, disease- and treatment-related variables, and socioeconomic characteristics. Data analyses were performed between November 2021 and March 2022 and completed in June 2022. Main Outcomes and Measures Relapse and disease-specific mortality after a diagnosis of BC. Results The final study population included 1275 women (mean [SD] age, 32.9 [3.8] years) at the time of BC diagnosis. After stratification by the matching variables age, calendar period, and region, and adjustment for country of birth, education, parity at diagnosis, tumor size, number of lymph node metastases, and estrogen receptor status, disease-specific mortality was similar in women who underwent hormonal FP (adjusted hazard ratio [aHR], 0.59; 95% CI, 0.32-1.09), women who underwent nonhormonal FP (aHR, 0.51; 95% CI, 0.20-1.29), and women who were not exposed to FP (reference). In a subcohort with detailed data on relapse, adjusted rate of disease-specific mortality and relapse were also similar among the groups who underwent hormonal FP (aHR, 0.81; 95% CI, 0.49-1.37), underwent nonhormonal FP (aHR, 0.75; 95% CI, 0.35-1.62), and were not exposed to FP (reference). Conclusions and Relevance In this cohort study, FP with or without hormonal stimulation was not associated with any increased risk of relapse or disease-specific mortality in women with BC. Results of this study provide much needed additional evidence on the safety of FP procedures in women with BC and may influence current health care practice to the benefit of young women with BC who wish to preserve their fertility.
Collapse
Affiliation(s)
- Anna Marklund
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Solna, Sweden.,Division of Gynecology and Reproduction, Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Tobias Lekberg
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Solna, Sweden.,Breast Cancer Theme Center, Karolinska University Hospital and Karolinska Comprehensive Cancer Centre, Stockholm, Sweden
| | - Elham Hedayati
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Solna, Sweden.,Breast Cancer Theme Center, Karolinska University Hospital and Karolinska Comprehensive Cancer Centre, Stockholm, Sweden
| | - Annelie Liljegren
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Solna, Sweden.,Department of Internal Medicine, Southern Hospital, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Solna, Sweden.,Breast Cancer Theme Center, Karolinska University Hospital and Karolinska Comprehensive Cancer Centre, Stockholm, Sweden
| | - Frida E Lundberg
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Solna, Sweden.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Solna, Sweden.,Division of Gynecology and Reproduction, Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| |
Collapse
|
25
|
Clinical outcome of embryo cryopreservation in Japanese breast cancer patients: pregnancy rates after transfer of thawed embryos. J Assist Reprod Genet 2022; 39:1769-1777. [PMID: 35980490 PMCID: PMC9428083 DOI: 10.1007/s10815-022-02575-0] [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: 08/10/2021] [Accepted: 07/11/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE To examine pregnancy outcomes after cryopreserved embryo transfer (ET) in breast cancer patients and to investigate the effect of controlled ovarian hyperstimulation (COH) as well as that of aromatase inhibitor (AI) administration and of the random start (RS) ovarian stimulation method. METHODS This retrospective study covered 126 patients who underwent embryo cryopreservation between 2010 and 2019. Thirty-one patients underwent frozen embryo transfer (FET), and we examined resulting pregnancy rates (PRs) and live birth rates (LBRs) in those who did and did not undergo COH and in relation to the AI and RS methods. RESULTS PR and LBR per patient were higher among patients who underwent COH than among those who did not. PR per ET did not differ from that documented for non-cancer infertility patients, after adjustment for age. The PR and LBR did not differ between use and non-use of AI (27.8% vs 35.2%). In addition, there was no significant difference in the PR or LBR between RS and conventional start ovarian stimulation (33.3% vs 30.8%). No prenatal fetal abnormalities were observed in 8 cases (including 5 AI cases and 2 RS cases). CONCLUSIONS This study showed that the outcome of FET after FP was equivalent to that seen in non-cancer patients. Further, neither use of AI nor the RS method influenced LBR. COH including use of AI and the RS method are useful in FP for collecting and freezing many embryos within a short period and for increasing the per patient LBR after cancer treatment.
Collapse
|
26
|
Wang Y, Tesch ME, Lim C, Xu YH, Lee S, Perdizet K, Yokom D, Warner E, Roberts J, Lohrisch CA. Risk of recurrence and pregnancy outcomes in young women with breast cancer who do and do not undergo fertility preservation. Breast Cancer Res Treat 2022; 195:201-208. [PMID: 35908122 DOI: 10.1007/s10549-022-06650-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE To assess the impact of fertility preservation (FP) requiring ovarian stimulation on breast cancer outcomes and pregnancy after breast cancer. METHODS Women aged ≤ 40 years diagnosed with stage I-III breast cancer between 2007 and 2018 and referred for FP consultation prior to systemic therapy were identified from a British Columbia fertility center database. The primary endpoint was invasive breast cancer-free survival (iBCFS) and secondary endpoints were overall survival (OS) and achievement of pregnancy. Survival and pregnancy endpoints were compared using Cox and logistic regression analyses, respectively, for patients who did and did not undergo FP. RESULTS The study included 153 patients, with 71 (46%) in the FP group and 82 (54%) in the non-FP group. Patients who underwent FP were more likely to be ECOG 0 (99% vs. 88%, p = 0.011) and receive chemotherapy (93% vs. 67%, p < 0.001), but had similar ER positivity status to non-FP patients (70% vs. 79%, p = 0.21). Over a median follow-up of 4.1 years, there were no differences in iBCFS (HR 1.006, 95% CI 0.416-2.438, p = 0.988) or OS (HR 0.789, 95% CI 0.210-2.956, p = 0.725) between FP and non-FP groups. Patients who underwent FP had higher odds of conceiving at least once (OR 3.024, 95% CI 1.312-6.970, p = 0.008). CONCLUSION At a median follow-up of 4.1 years, FP did not impact iBCFS or OS, supporting its safety in young women with breast cancer. In addition, patients who underwent FP were more likely to become pregnant after breast cancer, highlighting the value of pre-oncologic treatment FP in survivorship family planning.
Collapse
Affiliation(s)
- Ying Wang
- Department of Medical Oncology, British Columbia Cancer, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada
| | - Megan E Tesch
- Department of Medical Oncology, British Columbia Cancer, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada
| | - Chloe Lim
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ying Hui Xu
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Shaina Lee
- Division of Oncology and Department of Gynecology, Allan Blair Cancer Centre, Regina, SK, Canada
| | | | - Dan Yokom
- Trillium Health Partners, Credit Valley Hospital, Mississauga, ON, Canada
| | - Ellen Warner
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, Toronto, ON, Canada
| | - Jeffery Roberts
- Pacific Centre for Reproductive Medicine, Vancouver, BC, Canada
| | - Caroline A Lohrisch
- Department of Medical Oncology, British Columbia Cancer, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada.
| |
Collapse
|
27
|
Balkenende EME, Dahhan T, Beerendonk CCM, Fleischer K, Stoop D, Bos AME, Lambalk CB, Schats R, Smeenk JMJ, Louwé LA, Cantineau AEP, Bruin JPD, Linn SC, van der Veen F, van Wely M, Goddijn M. Fertility preservation for women with breast cancer: a multicentre randomized controlled trial on various ovarian stimulation protocols. Hum Reprod 2022; 37:1786-1794. [PMID: 35776109 PMCID: PMC9340107 DOI: 10.1093/humrep/deac145] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/01/2022] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Does ovarian stimulation with the addition of tamoxifen or letrozole affect the number of cumulus-oocyte complexes (COCs) retrieved compared to standard ovarian stimulation in women with breast cancer who undergo fertility preservation? SUMMARY ANSWER Alternative ovarian stimulation protocols with tamoxifen or letrozole did not affect the number of COCs retrieved at follicle aspiration in women with breast cancer. WHAT IS KNOWN ALREADY Alternative ovarian stimulation protocols have been introduced for women with breast cancer who opt for fertility preservation by means of banking of oocytes or embryos. How these ovarian stimulation protocols compare to standard ovarian stimulation in terms of COC yield is unknown. STUDY DESIGN, SIZE, DURATION This multicentre, open-label randomized controlled superiority trial was carried out in 10 hospitals in the Netherlands and 1 hospital in Belgium between January 2014 and December 2018. We randomly assigned women with breast cancer, aged 18–43 years, who opted for banking of oocytes or embryos to one of three study arms; ovarian stimulation plus tamoxifen, ovarian stimulation plus letrozole or standard ovarian stimulation. Standard ovarian stimulation included GnRH antagonist, recombinant FSH and GnRH agonist trigger. Randomization was performed with a web-based system in a 1:1:1 ratio, stratified for oral contraception usage at start of ovarian stimulation, positive estrogen receptor (ER) status and positive lymph nodes. Patients and caregivers were not blinded to the assigned treatment. The primary outcome was number of COCs retrieved at follicle aspiration. PARTICIPANTS/MATERIALS, SETTING, METHODS During the study period, 162 women were randomly assigned to one of three interventions. Fifty-four underwent ovarian stimulation plus tamoxifen, 53 ovarian stimulation plus letrozole and 55 standard ovarian stimulation. Analysis was according to intention-to-treat principle. MAIN RESULTS AND THE ROLE OF CHANCE No differences among groups were observed in the mean (±SD) number of COCs retrieved: 12.5 (10.4) after ovarian stimulation plus tamoxifen, 14.2 (9.4) after ovarian stimulation plus letrozole and 13.6 (11.6) after standard ovarian stimulation (mean difference −1.13, 95% CI −5.70 to 3.43 for tamoxifen versus standard ovarian stimulation and 0.58, 95% CI −4.03 to 5.20 for letrozole versus standard ovarian stimulation). After adjusting for oral contraception usage at the start of ovarian stimulation, positive ER status and positive lymph nodes, the mean difference was −1.11 (95% CI −5.58 to 3.35) after ovarian stimulation plus tamoxifen versus standard ovarian stimulation and 0.30 (95% CI −4.19 to 4.78) after ovarian stimulation plus letrozole versus standard ovarian stimulation. There were also no differences in the number of oocytes or embryos banked. There was one serious adverse event after standard ovarian stimulation: one woman was admitted to the hospital because of ovarian hyperstimulation syndrome. LIMITATIONS, REASONS FOR CAUTION The available literature on which we based our hypothesis, power analysis and sample size calculation was scarce and studies were of low quality. Our study did not have sufficient power to perform subgroup analysis on follicular, luteal or random start of ovarian stimulation. WIDER IMPLICATIONS OF THE FINDINGS Our study showed that adding tamoxifen or letrozole to a standard ovarian stimulation protocol in women with breast cancer does not impact the effectiveness of fertility preservation and paves the way for high-quality long-term follow-up on breast cancer treatment outcomes and women’s future pregnancy outcomes. Our study also highlights the need for high-quality studies for all women opting for fertility preservation, as alternative ovarian stimulation protocols have been introduced to clinical practice without proper evidence. STUDY FUNDING/COMPETING INTEREST(S) The study was supported by a grant (2011.WO23.C129) of ‘Stichting Pink Ribbon’, a breast cancer fundraising charity organization in the Netherlands. M.G., C.B.L. and R.S. declared that the Center for Reproductive Medicine, Amsterdam UMC (location VUMC) has received unconditional research and educational grants from Guerbet, Merck and Ferring, not related to the presented work. C.B.L. declared a speakers fee for Inmed and Yingming. S.C.L. reports grants and non-financial support from Agendia, grants, non-financial support and other from AstraZeneca, grants from Eurocept-pharmaceuticals, grants and non-financial support from Genentech/Roche and Novartis, grants from Pfizer, grants and non-financial support from Tesaro and Immunomedics, other from Cergentis, IBM, Bayer, and Daiichi-Sankyo, outside the submitted work; In addition, S.C.L. has a patent UN23A01/P-EP pending that is unrelated to the present work. J.M.J.S. reported payments and travel grants from Merck and Ferring. C.C.M.B. reports her role as unpaid president of the National guideline committee on Fertility Preservation in women with cancer. K.F. received unrestricted grants from Merck Serono, Good Life and Ferring not related to present work. K.F. declared paid lectures for Ferring. D.S. declared former employment from Merck Sharp & Dohme (MSD). K.F. declared paid lectures for Ferring. D.S. reports grants from MSD, Gedeon Richter and Ferring paid to his institution; consulting fee payments from MSD and Merck Serono paid to his institution; speaker honoraria from MSD, Gedeon Richter, Ferring Pharmaceuticals and Merck Serono paid to his institution. D.S. has also received travel and meeting support from MSD, Gedeon Richter, Ferring Pharmaceuticals and Merck Serono. No payments are related to present work. TRIAL REGISTRATION NUMBER NTR4108. TRIAL REGISTRATION DATE 6 August 2013. DATE OF FIRST PATIENT’S ENROLMENT 30 January 2014.
Collapse
Affiliation(s)
- Eva M E Balkenende
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Taghride Dahhan
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Catharina C M Beerendonk
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kathrin Fleischer
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dominic Stoop
- Center for Reproductive Medicine, UZ Brussel, Free University of Brussels, Brussels, Belgium.,Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Annelies M E Bos
- Department of Reproductive Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cornelis B Lambalk
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Roel Schats
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jesper M J Smeenk
- Department of Obstetrics and Gynaecology, St Elisabeth Hospital, Tilburg, The Netherlands
| | - Leonie A Louwé
- Department of Gynaecology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Astrid E P Cantineau
- Center for Reproductive Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Peter de Bruin
- Department of Obstetrics and Gynecology, Jeroen Bosch Hospital, Den Bosch, The Netherlands
| | - Sabine C Linn
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Fulco van der Veen
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Madelon van Wely
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Mariëtte Goddijn
- Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Pleasant V, Ulrich N, Pearlman MD, Moravek MB. Reproductive Considerations for Patients with Early-Onset Breast Cancer. CURRENT BREAST CANCER REPORTS 2022; 14:37-45. [PMID: 39525552 PMCID: PMC11548833 DOI: 10.1007/s12609-022-00445-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
Abstract
Purpose of Review Early-onset breast cancer presents various challenges in regard to reproductive health. This review examines the current data pertaining to issues such as fertility preservation, preimplantation genetics, the impact of chemotherapy and endocrine therapy, contraception, and pregnancy in the young breast cancer population. Recent Findings Many breast cancer treatments such as chemotherapy and endocrine therapy can be gonadotoxic or teratogenic. Multiple fertility preservation options are now available to young breast cancer patients, including embryo and oocyte preservation, ovarian tissue preservation, and ovarian suppression with GnRH agonists. In patients with known cancer germline mutations, preimplantation genetic testing is an option. Summary Fertility preservation and other reproductive services are expanding and offer great promise to the early-onset breast cancer community. Providers must be knowledgeable about the various options in order to better empower patients.
Collapse
Affiliation(s)
- Versha Pleasant
- Cancer Genetics & Breast Health, Department of Obstetrics and Gynecology, University of Michigan, L4204 UH South, 1500 E. Medical Center Dr., SPC 5276, Ann Arbor, MI 48109-5276, USA
| | - Nicole Ulrich
- Audobon Fertility, New Orleans, LA, USA
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Mark D. Pearlman
- Cancer Genetics & Breast Health, Department of Obstetrics and Gynecology, University of Michigan, L4204 UH South, 1500 E. Medical Center Dr., SPC 5276, Ann Arbor, MI 48109-5276, USA
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Molly B. Moravek
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Department of Urology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
29
|
Salama M, Lambertini M, Christianson MS, Jayasinghe Y, Anazodo A, De Vos M, Amant F, Stern C, Appiah L, Woodard TL, Anderson RA, Westphal LM, Leach RE, Rodriguez-Wallberg KA, Patrizio P, Woodruff TK. Installing oncofertility programs for breast cancer in limited versus optimum resource settings: Empirical data from 39 surveyed centers in Repro-Can-OPEN Study Part I & II. J Assist Reprod Genet 2022; 39:505-516. [PMID: 35032286 PMCID: PMC8760079 DOI: 10.1007/s10815-022-02394-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/05/2022] [Indexed: 01/09/2023] Open
Abstract
PURPOSE As a further step to elucidate the actual diverse spectrum of oncofertility practices for breast cancer around the globe, we present and discuss the comparisons of oncofertility practices for breast cancer in limited versus optimum resource settings based on data collected in the Repro-Can-OPEN Study Part I & II. METHODS We surveyed 39 oncofertility centers including 14 in limited resource settings from Africa, Asia & Latin America (Repro-Can-OPEN Study Part I), and 25 in optimum resource settings from the United States, Europe, Australia and Japan (Repro-Can-OPEN Study Part II). Survey questions covered the availability of fertility preservation and restoration options offered to young female patients with breast cancer as well as the degree of utilization. RESULTS In the Repro-Can-OPEN Study Part I & II, responses for breast cancer and calculated oncofertility scores showed the following characteristics: (1) higher oncofertility scores in optimum resource settings than in limited resource settings especially for established options, (2) frequent utilization of egg freezing, embryo freezing, ovarian tissue freezing, GnRH analogs, and fractionation of chemo- and radiotherapy, (3) promising utilization of oocyte in vitro maturation (IVM), (4) rare utilization of neoadjuvant cytoprotective pharmacotherapy, artificial ovary, and stem cells reproductive technology as they are still in preclinical or early clinical research settings, (5) recognition that technical and ethical concerns should be considered when offering advanced and innovative oncofertility options. CONCLUSIONS We presented a plausible oncofertility best practice model to guide oncofertility teams in optimizing care for breast cancer patients in various resource settings.
Collapse
Affiliation(s)
- Mahmoud Salama
- Oncofertility Consortium, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316 USA
| | - M. Lambertini
- Department of Medical Oncology, UOC Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - MS Christianson
- Division of Reproductive Endocrinology and Infertility, Johns Hopkins Fertility Center, Johns Hopkins University School of Medicine, 10751 Falls Road, Suite 280, Lutherville, MD 21093 USA
| | - Y. Jayasinghe
- Royal Children’s Hospital, Flemington Rd, Parkville, Melbourne, Vic 3054 Australia
- Department of Obstetrics & Gynecology, Royal Womens Hospital Melbourne, Parkville, Australia
| | - A. Anazodo
- Fertility Research Centre, Royal Hospital for Women, Barker Street, Sydney, Australia
| | - M. De Vos
- Centre for Reproductive Medicine of UZ Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - F. Amant
- Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - C. Stern
- Fertility Preservation Service, Reproductive Services Unit, Royal Women’s Hospital, Parkville, 3051 Australia
- Fertility Preservation Service, Melbourne IVF, East Melbourne, 3002 Australia
| | - L. Appiah
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO USA
| | - T. L. Woodard
- Department of Gynecologic Oncology and Reproductive Medicine, MD Anderson Oncofertility Program, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - R. A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - L. M. Westphal
- Stanford University Hospital, 300 Pasteur Drive, Stanford, CA USA
| | - R. E. Leach
- Oncofertility Consortium, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316 USA
| | - K. A. Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska University Hospital, 14186 Stockholm, Sweden
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - P. Patrizio
- Yale Fertility Center and Yale Fertility Preservation Program, 200 West Campus Dr, Orange, CT 06477 USA
| | - Teresa K. Woodruff
- Oncofertility Consortium, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316 USA
| |
Collapse
|
30
|
Ulrich ND, Raja NS, Moravek MB. A Review of Fertility Preservation in Patients with Breast Cancer. Best Pract Res Clin Obstet Gynaecol 2022; 82:60-68. [DOI: 10.1016/j.bpobgyn.2022.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 12/20/2022]
|
31
|
Female Oncofertility: Current Understandings, Therapeutic Approaches, Controversies, and Future Perspectives. J Clin Med 2021; 10:jcm10235690. [PMID: 34884393 PMCID: PMC8658080 DOI: 10.3390/jcm10235690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Recent advances in early detection and oncological therapies have ameliorated the survival rate of young cancer patients. Yet, ovarian impairment induced by chemotherapy and radiotherapy is still a challenging issue. This review, based on clinical and lab-based studies, summarizes the evidence of gonadotoxicity of chemoradiotherapy, the recent approaches, ongoing controversies, and future perspectives of fertility preservation (FP) in female patients who have experienced chemo- or radio-therapy. Existing data indicate that chemotherapeutic agents induce DNA alterations and massive follicle activation via the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Meanwhile, the radiation causes ionizing damage, leading to germ cell loss. In addition to the well-established methods, numerous therapeutic approaches have been suggested, including minimizing the follicle loss in cryopreserved ovarian grafts after transplantation, in vitro activation or in vitro growing of follicles, artificial ovarian development, or fertoprotective adjuvant to prevent ovarian damage from chemotherapy. Some reports have revealed positive outcomes from these therapies, whereas others have demonstrated conflictions. Future perspectives are improving the live birth rate of FP, especially in patients with adverse ovarian reserve, eliminating the risk of malignancy reintroducing, and increasing society’s awareness of FP importance.
Collapse
|
32
|
Clinical Pregnancy and Incidence of Ovarian Hyperstimulation Syndrome in High Ovarian Responders Receiving Different Doses of hCG Supplementation in a GnRH-Agonist Trigger Protocol. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2180933. [PMID: 34733337 PMCID: PMC8560257 DOI: 10.1155/2021/2180933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/27/2021] [Indexed: 11/18/2022]
Abstract
Objective Ovarian hyperstimulation syndrome (OHSS) is a side effect of the exogenous human chorionic gonadotropin (hCG) hormones used to trigger oocyte maturation. High ovarian responders represent a population with a higher risk of OHSS and are characterized by an exaggerated response to gonadotropin administration. In this study, we compared clinical pregnancy and incidence of OHSS in high ovarian responders receiving different doses of hCG supplementation in a GnRH-agonist trigger protocol. Methods A total of 205 high ovarian responders who were to undergo in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) cycles were recruited and randomly assigned to receive different doses of hCG supplementation in a GnRH-agonist trigger protocol: GnRH-a (n = 42), GnRH-a + 1000 IU hCG (n = 49), GnRH-a + 2000 IU hCG (n = 54), and GnRH-a + 3000 IU hCG (n = 60) groups. Results The GnRH-a + 1000 IU hCG, GnRH-a + 2000 IU hCG, and GnRH-a + 3000 IU hCG groups had more oocytes retrieved, embryos, high-quality embryos, and a higher rate of high-quality embryos than the GnRH-a group (p < 0.05). The GnRH-a + 1000 IU hCG group demonstrated more oocytes retrieved, embryos, high-quality embryos, and a higher rate of high-quality embryos than the GnRH-a + 2000 IU hCG and GnRH-a + 3000 IU hCG groups (p < 0.05). No moderate and severe OHSS cases occurred in the GnRH-a and GnRH-a + 1000 IU hCG groups. The incidence rate of moderate and severe OHSS was remarkably lower in the GnRH-a group and GnRH-a + 1000 IU hCG groups than in the GnRH-a + 2000 IU hCG and GnRH-a + 3000 IU hCG groups (p < 0.05). The GnRH-a + 1000 IU hCG, GnRH-a + 2000 IU hCG, and GnRH-a + 3000 IU hCG groups had a higher clinical pregnancy rate than the GnRH-a group, showing no significant difference (p > 0.05). The GnRH-a + 1000 IU hCG, GnRH-a + 2000 IU hCG, and GnRH-a + 3000 IU hCG groups had a lower abortion rate than the GnRH-a group (p < 0.05). Conclusion Based on the data obtained from this prospective study, we recommend 1000 IU hCG supplementation in a GnRH-agonist trigger protocol for high ovarian responders during IVF/ICSI cycles considering a higher rate of high-quality embryos, a lower incidence rate of moderate and severe OHSS, and a lower abortion rate.
Collapse
|
33
|
Zhang XQ, Zhang LJ, Zhu XL, Xu H, Luo YQ, Yao L, Huang QW, Nong YQ, Liu WJ, Liu FH. The Clinical Efficacy of Three Different Follicle-Stimulating Hormones for Follicle Growth and Development in Long-Protocol Controlled Ovarian Hyperstimulation Treatment. Drug Des Devel Ther 2021; 15:3573-3580. [PMID: 34429586 PMCID: PMC8378929 DOI: 10.2147/dddt.s316189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/08/2021] [Indexed: 12/05/2022] Open
Abstract
Objective To compare the use and clinical efficacy of three different follicle-stimulating hormones (FSHs) for follicle growth and development in long-protocol controlled ovarian hyperstimulation (COH). Methods A total of 540 gonadotropin-releasing hormone (GnRH) agonists’ long protocol treatment cycles at our hospital between January 2015 and May 2020 and met the inclusion criteria were retrospectively analyzed. The cycles were divided into three groups based on their indexes (groups A, B, and C). Each of the groups received a different type of FSH during treatment. A cross-group comparison was then undertaken to evaluate the growth and development of the three largest follicles and the patients’ pregnancy-related indexes between the normal-response and high-response populations. Results In the normal-response populations, the number of high-quality embryos obtained in groups A and B was significantly higher than in group C, and the FSH dosage was significantly lower than in group C (P < 0.05). There were more follicles with a diameter of 16–18 mm found in group A than in group C on the day of hCG injection (hCG day) (P < 0.05), but there were no significant differences in the groups in other indicators. In the high-response populations, the number of oocytes retrieved and high-quality embryos obtained in group A were significantly higher than in group C (P < 0.05), and the total dosage and duration of FSH stimulation in group C were significantly higher than groups A and B (P < 0.05). Conclusion Three different types of FSH led to comparable growth rates of the three largest follicles and clinical pregnancy rates per fresh cycle in long-protocol COH treatment.
Collapse
Affiliation(s)
- Xi-Qian Zhang
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Li-Jia Zhang
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Xiu-Lan Zhu
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Hong Xu
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Yan-Qun Luo
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Li Yao
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Qian-Wen Huang
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Ying-Qi Nong
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Wen-Juan Liu
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| | - Feng-Hua Liu
- Reproductive Medical Center, Guangdong Women and Children Hospital, Guangzhou, 511442, People's Republic of China
| |
Collapse
|
34
|
Sahin G, Goker ENT, Gokmen E, Yeniay L, Acet F, Zekioglu O, Tavmergen E. Controlled ovarian stimulation outcomes of fertility preservation procedures in newly diagnosed breast cancer patients: a retrospective study from a single-tertiary-IVF centre. J OBSTET GYNAECOL 2021; 42:518-523. [PMID: 34382483 DOI: 10.1080/01443615.2021.1931067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The aim of this study was to evaluate the effectiveness of controlled ovarian stimulation (COS) using the letrozole-supplemented stimulation protocol in breast cancer (BC) patients prior to their cancer treatment. Sixty-one BC patients (Stages 0-3) who were referred to a university IVF unit for fertility preservation (FP) and underwent embryo and/or oocyte cryopreservation between 2008 - 2020 were included in this retrospective study. Time intervals between breast surgery and initial fertility consultation (IFC)/completion of FP procedures were evaluated. COS outcomes were assessed and compared between the early follicular phase (EFP) and the random-start (RS) protocols. The patients' mean age was 33.3 ± 4.9 years. The mean time interval between breast surgery and IFC was 20.6 ± 11 (day, mean ± SD) and from IFC to completion of FP procedure was 14.7 ± 5.3. Overall, 9.1 ± 5.9 mature oocytes were obtained, with a peak serum oestradiol level of 388 ± 358 pg/mL. The number of oocytes obtained (11.5 ± 9.3 vs. 10.9 ± 6.9, p = .9) and maturation rates (84.3 ± 17.5% vs. 89.2 ± 11.7, p = .5) were not statistically different between the EPF and RS protocols. The study results support that oocyte or embryo freezing can be performed effectively in a limited time period with letrozole-supplemented COS protocols before the initiation of oncological treatments in breast cancer patients.Impact statementWhat is already known on this subject? Currently, embryo and oocyte freezing are considered the most established fertility preservation (FP) methods for newly diagnosed cancer patients.What do the results of this study add? This study reports the COS outcomes of newly diagnosed breast cancer patients for FP over a period of twelve years from a single IVF unit. The results support that a considerable number of oocytes can be harvested with letrozole-supplemented COS protocol, which appears to be an effective protocol for BC patients.What are the implications of these findings for clinical practice and/or further research? There is a need for additional studies evaluating long-term follow-up of patients with their pregnancy outcomes.
Collapse
Affiliation(s)
- Gulnaz Sahin
- Ege University, Family Planning and Infertility Research and Practice Center, Izmir, Turkey
| | - Ege Nazan Tavmergen Goker
- Ege University, Family Planning and Infertility Research and Practice Center, Izmir, Turkey.,Department of Obstetrics and Gynecology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Erhan Gokmen
- Department of Medical Oncology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Levent Yeniay
- Department of General Surgery, Ege University, Faculty of Medicine, Izmir, Turkey
| | - Ferruh Acet
- Ege University, Family Planning and Infertility Research and Practice Center, Izmir, Turkey.,Department of Obstetrics and Gynecology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Osman Zekioglu
- Department of Pathology, Ege University, Faculty of Medicine, Izmir, Turkey
| | - Erol Tavmergen
- Ege University, Family Planning and Infertility Research and Practice Center, Izmir, Turkey.,Department of Obstetrics and Gynecology, Ege University Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
35
|
Huang H, Takai Y, Samejima K, Gomi Y, Narita T, Ichinose S, Itaya Y, Ono Y, Matsunaga S, Saitoh M, Seki H. Severe hemoperitoneum resulting from restart of letrozole after oocyte retrieval procedure: a case report. J Med Case Rep 2021; 15:327. [PMID: 34174941 PMCID: PMC8236175 DOI: 10.1186/s13256-021-02938-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the field of oncofertility, patients with breast cancer are often administered letrozole as an adjuvant drug before and after oocyte retrieval to prevent an increase in circulating estradiol. CASE PRESENTATION We report a case of abdominal hemorrhage due to an ovarian rupture in a 29-year-old Japanese patient who restarted letrozole 2 days after an oocyte retrieval procedure in which 14 mature oocytes were retrieved. The patient had sought embryo cryopreservation as a fertility preservation option before undergoing treatment for recurrent breast cancer. A day after restarting letrozole treatment, the patient unexpectedly developed severe abdominal pain. Laparoscopic hemostasis was performed to manage the ovarian swelling and hemorrhage. CONCLUSIONS The ovaries can be restimulated by restart letrozole after an oocyte retrieval procedure. Therefore, reproductive-medicine practitioners should understand the potential complications of letrozole administration in such cases and take steps to ensure that they are minimized.
Collapse
Affiliation(s)
- Haipeng Huang
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan.
| | - Yasushi Takai
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Kouki Samejima
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Yosuke Gomi
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Tatsuya Narita
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Shunichiro Ichinose
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Yukiko Itaya
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Yosihisa Ono
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Sigetaka Matsunaga
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Masahiro Saitoh
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| | - Hiroyuki Seki
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe City, Saitama, 350-3550, Japan
| |
Collapse
|
36
|
Luo X, Wang H, Ji D. Carbon nanotubes (CNT)-loaded ginsenosides Rb3 suppresses the PD-1/PD-L1 pathway in triple-negative breast cancer. Aging (Albany NY) 2021; 13:17177-17189. [PMID: 34111025 PMCID: PMC8312428 DOI: 10.18632/aging.203131] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/29/2021] [Indexed: 12/16/2022]
Abstract
Carbon nanotubes (CNTs), as advanced nanotechnology with specific properties and structures, have presented practical drug delivery properties. Ginsenoside Rg3 is a component of puffed ginseng and demonstrates anti-cancer activities. To explore the effect of CNTs-loaded Rg3 (Rg3-CNT) on the PD-1/PD-L1 signaling and the development of triple-negative breast cancer (TNBC). Our data revealed that Rg3 inhibited the cell viability of TNBC cells, in which Rg3-CNT further enhanced this effect in the system. Similarly, the colony formation of TNBC cells was decreased by Rg3, while Rg3-CNT could reinforce its effect in the cells. Besides, the treatment of Rg3 induced apoptosis of TNBC cells, in which Rg3-CNT treatment further increased the phenotype in the cells. Remarkably, Rg3-CNT, but not Rg3, attenuated PD-L1 expression in TNBC cells. Rg3-CNT decreased the PD-L1 upregulation induced by interferon-γ (IFN-γ) in breast cancer cells. Importantly, Rg3-CNT was able to reduce PD-1 expression in activated T cells. Specifically, Rg3-CNT reduced the PD-1/PD-L1 axis in a T cell/triple-negative TNBC cell co-culture system. Moreover, the levels of IFN-γ, interleukins-2 (IL-2), interleukins-9 (IL-9), interleukins-10 (IL-10), interleukins-22 (IL-22), and interleukins-23 (IL-23) were significantly stimulated in the activated T cells, while the treatment of Rg3-CNT could reverse these phenotypes in the cells. Rg3-CNT attenuated the TNBC cell growth in vivo. The Rg3-CNT improved the anti-cancer effect of Rg3 toward TNBC by inhibiting the PD-1/PD-L1 axis. Our finding provides new insights into the mechanism by which Rg3-CNT attenuates the development of TNBC. Rg3-CNT may be applied as the potential therapeutic strategy for immunotherapy of TNBC.
Collapse
Affiliation(s)
- Xiao Luo
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Hui Wang
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Degang Ji
- Department of Hepatobiliary Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| |
Collapse
|
37
|
Gonadotropin-Releasing Hormone Agonist Versus Recombinant Human Chorionic Gonadotropin Triggering in Fertility Preservation Cycles. Reprod Sci 2021; 28:3390-3396. [PMID: 34076872 DOI: 10.1007/s43032-021-00622-2] [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: 04/01/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
The purpose of this research is to study the efficacy of GnRH-a versus r-hCG triggering in patients who go through fertility preservation cycles. This retrospective cohort study was performed in a tertiary university-affiliated medical center. It includes 191 patients undergoing fertility preservation cycles between May 2013 and September 2018, in which ovulation was induced by either GnRH-a or r-hCG. Main outcome measures were number and rate of mature oocyte. Among treatment cycles with medical indication, GnRH agonist significantly increases the odds for high mature rate by 3.55 (1.30-9.66), while in treatment cycles with social indication, there is no significant effect of the triggering agent. An advantage for GnRH-a triggering was observed in medically indicated preservation cycles.
Collapse
|
38
|
De Vos M, Grynberg M, Ho TM, Yuan Y, Albertini DF, Gilchrist RB. Perspectives on the development and future of oocyte IVM in clinical practice. J Assist Reprod Genet 2021; 38:1265-1280. [PMID: 34218388 PMCID: PMC8266966 DOI: 10.1007/s10815-021-02263-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
Oocyte in vitro maturation (IVM) is an assisted reproductive technology designed to obtain mature oocytes following culture of immature cumulus-oocyte complexes collected from antral follicles. Although IVM has been practiced for decades and is no longer considered experimental, the uptake of IVM in clinical practice is currently limited. The purpose of this review is to ensure reproductive medicine professionals understand the appropriate use of IVM drawn from the best available evidence supporting its clinical potential and safety in selected patient groups. This group of scientists and fertility specialists, with expertise in IVM in the ART laboratory and/or clinic, explore here the development of IVM towards acquisition of a non-experimental status and, in addition, critically appraise the current and future role of IVM in human ART.
Collapse
Affiliation(s)
- Michel De Vos
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
- Department of Obstetrics, Gynecology, Perinatology and Reproductology, Institute of Professional Education, Sechenov University, Moscow, Russia
| | - Michaël Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Antoine Béclère University Hospital, Clamart, Clamart, France
- Paris-Sud University, Le Kremlin Bicêtre, France
| | - Tuong M Ho
- IVFMD, My Duc Hospital, Ho Chi Minh City, Vietnam
| | - Ye Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA
| | - David F Albertini
- Bedford Research Foundation, 124 South Road, Bedford, MA, 01730, USA
| | - Robert B Gilchrist
- Fertility & Research Centre, School of Women's and Children's Health, University of New South Wales Sydney, Sydney, NSW, Australia.
| |
Collapse
|
39
|
Delattre S, Segers I, Van Moer E, Drakopoulos P, Mateizel I, Enghels L, Tournaye H, De Vos M. Combining fertility preservation procedures to spread the eggs across different baskets: a feasibility study. Hum Reprod 2021; 35:2524-2536. [PMID: 32951035 DOI: 10.1093/humrep/deaa193] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/02/2020] [Indexed: 12/18/2022] Open
Abstract
STUDY QUESTION What is the reproductive potential following combinations of ovarian stimulation, IVM and ovarian tissue cryopreservation (OTC) in female patients seeking fertility preservation (FP)? SUMMARY ANSWER In selected patients, combining different FP procedures is a feasible approach and reproductive outcomes after FP in patients who return to attempt pregnancy are promising. WHAT IS KNOWN ALREADY FP is increasingly performed in fertility clinics but an algorithm to select the most suitable FP procedure according to patient characteristics and available timeframe is currently lacking. Vitrification of mature oocytes (OV) and OTC are most commonly performed, although in some clinical scenarios a combination of procedures including IVM, to spread the sources of gametes, may be considered in order to enhance reproductive options for the future. STUDY DESIGN, SIZE, DURATION Retrospective, observational study in a university-based, tertiary fertility centre involving all female patients who underwent urgent medical FP between January 2012 and December 2018. Descriptive analysis of various FP procedures, either stand-alone or combined, was performed, and reproductive outcomes of patients who attempted pregnancy in the follow-up period were recorded. PARTICIPANTS/MATERIALS, SETTING, METHODS In total, 207 patients underwent medical FP. Patient-tailored strategies and procedures were selected after multidisciplinary discussion. When deemed feasible, FP procedures were combined to cryopreserve different types of reproductive tissue for future use. The main primary outcome measure was the number of mature oocytes. Live birth rates were evaluated in patients who returned for reproductive treatment. MAIN RESULTS AND THE ROLE OF CHANCE Among patients seeking FP, 95/207 (46%) had breast cancer, 43/207 (21%) had haematological malignancies and 31/207 (15%) had a gynaecological tumour. Mean ± SD age was 27.0 ± 8.3 years. Eighty-five (41.1%) patients underwent controlled ovarian stimulation (COS), resulting in 10.8 ± 7.1 metaphase II (MII) oocytes for vitrification. Eleven (5.3%) patients had multiple COS cycles. Transvaginal oocyte retrieval for IVM was performed in 17 (8.2%) patients, yielding 9.2 ± 10.1 MII oocytes. Thirty-four (16.4%) patients underwent OTC combined with IVM of oocytes retrieved from ovarian tissue 'ex vivo' (OTO-IVM), yielding 4.0 ± 4.3 MII oocytes in addition to ovarian fragments. Seventeen (8.2%) patients had OTC combined with OTO-IVM and transvaginal retrieval of oocytes for IVM from the contralateral ovary, resulting in 13.5 ± 9.7 MII oocytes. In 13 (6.3%) patients, OTC with OTO-IVM was followed by controlled stimulation of the contralateral ovary, yielding 11.3 ± 6.6 MII oocytes in total. During the timeframe of the study, 31/207 (15%) patients have returned to the fertility clinic with a desire for pregnancy. Of those, 12 (38.7%) patients had preserved ovarian function and underwent ART treatment with fresh oocytes, resulting in nine (75%) livebirth. The remaining 19 (61.3%) patients requested warming of their cryopreserved material because of ovarian insufficiency. Of those, eight (42.1%) patients had a livebirth, of whom three after OTO-IVM. To date, 5/207 patients (2.4%) achieved an ongoing pregnancy or livebirth after spontaneous conception. LIMITATIONS, REASONS FOR CAUTION Our FP programme is based on a patient-tailored approach rather than based on an efficiency-driven algorithm. The data presented are descriptive, which precludes firm conclusions. WIDER IMPLICATIONS OF THE FINDINGS Combining different FP procedures is likely to enhance the reproductive fitness of patients undergoing gonadotoxic treatment but further follow-up studies are needed to confirm this. STUDY FUNDING/COMPETING INTEREST(S) No external funding was used for this study and the authors have no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- S Delattre
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
| | - I Segers
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
| | - E Van Moer
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
| | - P Drakopoulos
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
| | - I Mateizel
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
| | - L Enghels
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
| | - H Tournaye
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
| | - M De Vos
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium.,Follicle Biology Laboratory (FOBI), UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Obstetrics, Gynecology, Perinatology and Reproductology, Institute of Professional Education, Sechenov University, Moscow, Russia
| |
Collapse
|
40
|
Marklund A, Eloranta S, Wikander I, Kitlinski ML, Lood M, Nedstrand E, Thurin-Kjellberg A, Zhang P, Bergh J, Rodriguez-Wallberg KA. Efficacy and safety of controlled ovarian stimulation using GnRH antagonist protocols for emergency fertility preservation in young women with breast cancer-a prospective nationwide Swedish multicenter study. Hum Reprod 2021; 35:929-938. [PMID: 32313940 PMCID: PMC7192532 DOI: 10.1093/humrep/deaa029] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/21/2020] [Indexed: 11/30/2022] Open
Abstract
STUDY QUESTION How efficacious and safe are the current approaches to controlled ovarian stimulation (COS) aimed at fertility preservation (FP) in women with breast cancer (BC)? SUMMARY ANSWER In women with BC undergoing COS aiming at egg/embryo cryopreservation, letrozole-based protocols and those randomly started were equally effective compared with conventional COS, and the overall survival was similar between the women that proceeded to FP and those who did not. WHAT IS KNOWN ALREADY Cryopreservation of oocytes and embryos is an established method for FP in women with BC. Recent improvements to COS protocols include concomitant use of letrozole, random-cycle start day of stimulation and the use of GnRHa for the egg maturation trigger. To date, limited sample size of the available studies has not allowed investigation of differences in the efficacy of the different approaches to COS for FP in this patient population. STUDY DESIGN, SIZE, DURATION A prospective multicenter study with national coverage including 610 women with BC counseled between 1 January 1995 and 30 June 2017 at six Swedish FP regional programs. PARTICIPANTS/MATERIALS, SETTING, METHODS After counseling, 401 women elected to undergo COS. Treatments differed in the use or not of concomitant letrozole, a conventional or random-cycle day COS initiation and the use of hCG versus GnRHa trigger for oocyte maturation. Numbers of cryopreserved oocytes and embryos were defined as primary outcome. Pregnancy attempts, reproductive outcomes and long-term survival, investigated by the linking of individuals of the cohort to the total population register of the Swedish Tax Agency (up to 25 November 2018), were evaluated. MAIN RESULTS AND THE ROLE OF CHANCE Using letrozole or not resulted in similar numbers of oocytes and embryos cryopreserved (meanoocytes = 9.7 versus 10 and meanembryos 4.0 versus 5.3, respectively), similar to COS with random versus conventional start (meanoocytes 9.0 versus 10.6 and meanembryos 4.8 versus 4.8). In COS with letrozole, a GnRHa trigger was associated with a higher number of oocytes retrieved (P < 0.05) and embryos cryopreserved (P < 0.005), compared with conventional hCG trigger. Of 99 women who returned to fertility clinics after cancer treatment, 32 proceeded to thawing of oocytes or embryos and 10 of them had live births. The all-cause survival between the women that underwent COS and those who did not was similar and did not differ between the two groups. LIMITATIONS, REASONS FOR CAUTION Data on tumor characteristics and estrogen receptor (ER) status were not known for all women at the time of FP counseling and planning of COS, thus protocols with letrozole have been used for both estrogen-sensitive and non-estrogen-sensitive BC. For the same reason, subsequent adjustment for ERs in the BC or tumor characteristics as potential confounders were not performed as these parameters were not available and did not influence the provision of FP through COS. WIDER IMPLICATIONS OF THE FINDINGS The results of our study support the premise that recently introduced potential improvements to COS protocols for FP in women with BC are efficacious and safe. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by research grants from the Swedish Cancer Society, the Stockholm County Council, the Percy Falk Stiftelsen, Radiumhemmets Forskningsfonder, The Swedish Breast Cancer Association and Karolinska Institutet to K.A.R.W. J.B. reports grants from Amgen, AstraZeneca, Pfizer, Roche, Sanofi-Aventis and Merck, outside the submitted work, and payment from UpToDate to Asklepios Medicine HB for a chapter on BC prediction and prognostication. All the other authors have no competing interests to report.
Collapse
Affiliation(s)
- Anna Marklund
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Obstetrics and Gynecology, Södersjukhuset, Stockholm, Sweden
| | - Sandra Eloranta
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ida Wikander
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
| | | | - Mikael Lood
- Department of Obstetrics and Gynecology, Fertility Unit, Örebro University Hospital, Örebro, Sweden
| | - Elizabeth Nedstrand
- Department of Obstetrics and Gynecology, Linköping University Hospital, Linköping, Sweden
| | - Ann Thurin-Kjellberg
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Sweden and Reproductive Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pu Zhang
- Reproduction Center, Department of Obstetrics and Gynecology, Uppsala University Hospital, Uppsala, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Breast Cancer Flow, Patient Area of Breast Cancer Sarcoma and Endocrine Tumours, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum J5:30, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Success and risks of pregnancy after breast cancer. Breast Cancer Res Treat 2021; 188:593-600. [PMID: 33884537 DOI: 10.1007/s10549-021-06232-5] [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: 01/13/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Breast cancer is the most common cancer in women worldwide. The number of childbearing-age women diagnosed with early breast cancer (eBC) is increasing, raising questions over their subsequent fertility. PURPOSE The main objective of this study was therefore to assess, in a cohort of eBC patients with pregnancy desire, the rate of live births achieved spontaneously or by assisted reproductive technology. METHODS We conducted an observational, descriptive, retrospective study including patients aged 18-40, treated for eBC at the Institut de Cancérologie de l'Ouest (ICO) Pays de Loire between July 2010 and July 2016, with pregnancy desire. The primary outcome was the rate of live births. Secondary outcomes were overall survival, disease-free survival, time to conception, and spontaneous or assisted pregnancy rate. RESULTS 61 patients were included, with a live birth rate of 19.7% (12/61). We observed no recurrence or death in women with a pregnancy. Pregnancy started with a median time of 36.4 months after the end of treatment (4.1-51.3 months). All pregnancies in this cohort were achieved spontaneously. CONCLUSION The results of our cohort are consistent with previous results showing that spontaneous pregnancy remains possible after treatment for eBC without increasing the risk of recurrence or death.
Collapse
|
42
|
Rodriguez-Wallberg KA, Hao X, Marklund A, Johansen G, Borgström B, Lundberg FE. Hot Topics on Fertility Preservation for Women and Girls-Current Research, Knowledge Gaps, and Future Possibilities. J Clin Med 2021; 10:jcm10081650. [PMID: 33924415 PMCID: PMC8069871 DOI: 10.3390/jcm10081650] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/18/2022] Open
Abstract
Fertility preservation is a novel clinical discipline aiming to protect the fertility potential of young adults and children at risk of infertility. The field is evolving quickly, enriched by advances in assisted reproductive technologies and cryopreservation methods, in addition to surgical developments. The best-characterized target group for fertility preservation is the patient population diagnosed with cancer at a young age since the bulk of the data indicates that the gonadotoxicity inherent to most cancer treatments induces iatrogenic infertility. Since improvements in cancer therapy have resulted in increasing numbers of long-term survivors, survivorship issues and the negative impact of infertility on the quality of life have come to the front line. These facts are reflected in an increasing number of scientific publications referring to clinical medicine and research in the field of fertility preservation. Cryopreservation of gametes, embryos, and gonadal tissue has achieved quality standards for clinical use, with the retrieval of gonadal tissue for cryopreservation being currently the only method feasible in prepubertal children. Additionally, the indications for fertility preservation beyond cancer are also increasing since a number of benign diseases and chronic conditions either require gonadotoxic treatments or are associated with premature follicle depletion. There are many remaining challenges, and current research encompasses clinical health care and caring sciences, ethics, societal, epidemiological, experimental studies, etc.
Collapse
Affiliation(s)
- Kenny A. Rodriguez-Wallberg
- Department of Oncology and Pathology, Karolinska Institutet, SE-171 64 Stockholm, Sweden; (X.H.); (A.M.); (G.J.); (B.B.); (F.E.L.)
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, SE-141 86 Stockholm, Sweden
- Correspondence:
| | - Xia Hao
- Department of Oncology and Pathology, Karolinska Institutet, SE-171 64 Stockholm, Sweden; (X.H.); (A.M.); (G.J.); (B.B.); (F.E.L.)
| | - Anna Marklund
- Department of Oncology and Pathology, Karolinska Institutet, SE-171 64 Stockholm, Sweden; (X.H.); (A.M.); (G.J.); (B.B.); (F.E.L.)
| | - Gry Johansen
- Department of Oncology and Pathology, Karolinska Institutet, SE-171 64 Stockholm, Sweden; (X.H.); (A.M.); (G.J.); (B.B.); (F.E.L.)
| | - Birgit Borgström
- Department of Oncology and Pathology, Karolinska Institutet, SE-171 64 Stockholm, Sweden; (X.H.); (A.M.); (G.J.); (B.B.); (F.E.L.)
| | - Frida E. Lundberg
- Department of Oncology and Pathology, Karolinska Institutet, SE-171 64 Stockholm, Sweden; (X.H.); (A.M.); (G.J.); (B.B.); (F.E.L.)
| |
Collapse
|
43
|
Kauffman RP, Young C, Castracane VD. Perils of prolonged ovarian suppression and hypoestrogenism in the treatment of breast cancer: Is the risk of treatment worse than the risk of recurrence? Mol Cell Endocrinol 2021; 525:111181. [PMID: 33529690 DOI: 10.1016/j.mce.2021.111181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/04/2021] [Accepted: 01/21/2021] [Indexed: 01/18/2023]
Abstract
Premenopausal breast cancer is usually estrogen receptor positive, and hence, prolonged ovarian suppression by medical or surgical means to prevent recurrence has become standard of management to improve disease-free survival. Ten-year adjuvant tamoxifen therapy is associated with 3.5% fewer recurrences compared to five years. The SOFT trial demonstrated small but statistically significant incremental improvements in long-term disease-free survival by the addition of gonadotropin-releasing hormone analog treatment (triptorelin) to an aromatase inhibitor (exemestane). Profound hypoestrogenism in the premenopausal age group may not be well tolerated due to a host of bothersome side effects (primarily vasomotor symptoms, musculoskeletal complaints, genitourinary syndrome of menopause, and mood disorders). Prolonged hypoestrogenism in younger women is associated with premature development of cardiovascular disease, bone loss, cognitive decline, and all-cause mortality. This paper explores multi-system consequences of prolonged hypoestrogenism in premenopausal women derived from studies of women with and without breast cancer. Pretreatment counseling in estrogen receptor positive breast cancer should emphasize the benefit of prolonged estrogen suppression on breast cancer recurrence and established risks of lifelong hypoestrogenism on quality of life and all-cause mortality. Future genomic research may help identify the best candidates for extended ovarian suppression to avoid treating many women when only a minority benefit.
Collapse
Affiliation(s)
- Robert P Kauffman
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA.
| | - Christina Young
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA
| | - V Daniel Castracane
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA
| |
Collapse
|
44
|
Zhang H, Ge Z, Wang Z, Gao Y, Wang Y, Qu X. Circular RNA RHOT1 promotes progression and inhibits ferroptosis via mir-106a-5p/STAT3 axis in breast cancer. Aging (Albany NY) 2021; 13:8115-8126. [PMID: 33686957 PMCID: PMC8034942 DOI: 10.18632/aging.202608] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
To explore the effect of circRHOT1 on breast cancer progression and the underlying mechanism. Significantly, our data revealed that the depletion of circRHOT1 was able to repress the proliferation and induce the apoptosis of breast cancer cells. CircRHOT1 knockdown could remarkably inhibit the invasion and migration in the breast cancer cells. Meanwhile, the depletion of circRHOT1 enhanced the erastin-induced inhibition effect on cell growth of breast cancer cells. The circRHOT1 knockdown notably increased the levels of reactive oxygen species (ROS), iron, and Fe2+ in breast cancer cells. Mechanically, circRHOT1 was able to sponge microRNA-106a-5p (miR-106a-5p) and inhibited ferroptosis by down-regulating miR-106a-5p in breast cancer cells. Besides, miR-106a-5p induced ferroptosis by targeting signal transducer and activator of transcription 3 (STAT3) in the system. Moreover, the overexpression of STAT3 and miR-106a-5p inhibitor could reverse circRHOT1 knockdown-mediated breast cancer progression. Functionally, circRHOT1 promoted the tumor growth of breast cancer in vivo. In conclusion, we discovered that circRHOT1 contributed to malignant progression and attenuated ferroptosis in breast cancer by the miR-106a-5p/STAT3 axis. Our finding provides new insights into the mechanism by which circRHOT1 promotes the development of breast cancer. CircRHOT1 and miR-106a-5p may serve as potential targets for breast cancer therapy.
Collapse
Affiliation(s)
- Huiming Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhicheng Ge
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zihan Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yinguang Gao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yang Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiang Qu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
45
|
Buonomoa B, Peccatorib FA. Fertility preservation in endocrine responsive breast cancer: data and prejudices. Ecancermedicalscience 2021; 14:1157. [PMID: 33574902 PMCID: PMC7864682 DOI: 10.3332/ecancer.2020.1157] [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: 05/18/2020] [Indexed: 12/21/2022] Open
Abstract
Even if current guidelines suggest an early referral of young breast cancer (BC) patients to fertility preservation counselling, physicians still lack knowledge about the different available strategies. Hormonal stimulation to harvest mature oocytes is considered unsafe by many oncologists and experts in reproductive medicine, particularly in the setting of oestrogen receptor-positive BC. The aim of this mini-review is to provide an overview on the available data about this topic in order to clarify potential misunderstandings and to highlight the new trends in the oncofertility field with their pros and limitations.
Collapse
|
46
|
Fredriksson A, Rosenberg E, Einbeigi Z, Bergh C, Strandell A. Gonadotrophin stimulation and risk of relapse in breast cancer. Hum Reprod Open 2021; 2021:hoaa061. [PMID: 33501382 PMCID: PMC7810817 DOI: 10.1093/hropen/hoaa061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/18/2020] [Indexed: 01/09/2023] Open
Abstract
STUDY QUESTION Is gonadotrophin stimulation as part of IVF associated with an increased risk of relapse in breast cancer? SUMMARY ANSWER Controlled ovarian stimulation (COS) in connection with IVF in women with previous breast cancer was not associated with an increased risk of breast cancer relapse. WHAT IS KNOWN ALREADY Breast cancer is the most common malignancy among women worldwide and the leading cause of cancer death among females. The use of COS with gonadotrophins with subsequent cryopreservation of oocytes or embryos in order to enhance the chances of pregnancy after cancer treatment is the current most established fertility preservation method for women with breast cancer. To date, there are only a few small retrospective hospital-based controlled studies evaluating the risk of breast cancer relapse in patients undergoing fertility preservation with or without COS, showing no evident risk of relapse in breast cancer after the use of gonadotoxic agents. STUDY DESIGN, SIZE, DURATION This was a retrospective, population-based cohort study comprising 5857 women with previous breast cancer of whom 337 were exposed to COS. Exposure (COS) and outcomes (relapse and death) were identified for all patients from 2005 to 2014 by assessing the National Quality Register for Assisted Reproduction, the Swedish Medical Birth Register, the National Patient Register, the Swedish Prescribed Drug Register, the Swedish Cause of Death Register, the National Breast Cancer Register and the Swedish Cancer Register. Matching according to set criteria was possible for 334 women, who constituted the control group. A total of 274 women had undergone IVF after completing breast cancer treatment and 63 women had undergone COS for fertility preservation at the time of breast cancer diagnosis. PARTICIPANTS/MATERIALS, SETTING, METHODS Women aged 20–44 years previously diagnosed with breast cancer and exposed to COS were matched for age at breast cancer diagnosis ±5 years, tumour size and lymph node involvement with a non-exposed control group, including women with known T- and N-stages. In a subsequent analysis, the matched cohort was assessed by also including women with unknown T- and N-stages. A secondary analysis comprised the entire non-matched cohort, including all women with known T- and N-stages. Also here, a subsequent analysis included women with missing data for T- and N-stages. The risk of relapse in breast cancer was estimated as crude hazard ratios (HRs) and 95% CI using Cox proportional hazards models in the primary and secondary analyses where T- and N-stages were known: otherwise the risks of relapse were only given descriptively. MAIN RESULTS AND THE ROLE OF CHANCE In the primary matched analysis, relapse occurred in 20 of 126 women exposed to COS (15.9%) compared with 39 of 126 (31.0%) in the control cohort (HR = 0.70; 95% CI 0.39–1.45; P = 0.22). In the subsequent analysis, also including women with unknown T- and N-stages, relapse occurred in 27 of 337 (8.0%) women having undergone COS compared with 71/334 (21.3%) among the non-exposed. In the secondary adjusted analysis, relapse occurred in 20 of 126 (15.9%) exposed women and in 918 of 3729 (24.6%) non-exposed women (HR = 0.81; 95% CI 0.49–1.33; P = 0.70). In the subsequent analysis, including unknown T- and N-stages, relapse occurred in 27 of 337 (8.0%) women in the exposed group and 1176 of 5520 (21.3%) in the non-exposed cohort. LIMITATIONS, REASONS FOR CAUTION A substantial degree of missing data on important prognostic variables was a limitation, particularly when analysing the total cohort. Furthermore, data on confounding factors, such as BMI, were not completely covered. Another limitation was that a pre-specified variable for relapse was not in use for the majority of the National Breast Cancer Register. Furthermore, the follow-up time from available register data (2005–2014) is rather short. Finally, we cannot be sure whether the prognostic information from receptor status, showing a lower incidence in the exposed group, is representative. Information on T- and N-stages was missing in more than half of the patients. WIDER IMPLICATIONS OF THE FINDINGS In this large, retrospective, matched cohort study, we found no increased risk of relapse in breast cancer among women who had been exposed to gonadotrophins as part of IVF. This is reassuring but might be confounded by the selection of a group of women with a more favourable prognosis than those not undergoing IVF. The present study strengthens previous findings by being large, national and register based. Its results are applicable to women undergoing fertility preservation as well as to those undergoing regular IVF treatment. STUDY FUNDING/COMPETING INTEREST(S) Supported in part by grants from the Swedish state under the agreement between the Swedish government and the county councils the ALF-agreement (ALFGBG-720291), The Assar Gabrielsson Fund (FB 15-20), The Breast Cancer Fund and the Swedish Association of Local authorities and Regions, SKR. There are no conflicts of interest to declare. TRIAL REGISTRATION N/A
Collapse
Affiliation(s)
- A Fredriksson
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, SE 413 45, Sweden
| | - E Rosenberg
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, SE 413 45, Sweden
| | - Z Einbeigi
- Department of Medicine, Southern Älvsborg Hospital, Borås, SE 501 82, Sweden
| | - C Bergh
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, SE 413 45, Sweden
| | - A Strandell
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, SE 413 45, Sweden
| |
Collapse
|
47
|
Shulman Y, Almog B, Kalma Y, Fouks Y, Azem F, Cohen Y. Effects of letrozole or tamoxifen coadministered with a standard stimulation protocol on fertility preservation among breast cancer patients. J Assist Reprod Genet 2021; 38:743-750. [PMID: 33409757 DOI: 10.1007/s10815-020-02030-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To assess the effects of letrozole or tamoxifen coadministration on fertility preservation treatment outcomes. METHODS Retrospective cohort study of 118 breast cancer patients undergoing fertility preservation treatment between 2008 and 2018. Patients who received letrozole (n = 36) or tamoxifen (n = 30) were compared to controls (n = 52) who underwent standard ovarian stimulation protocols. The primary outcome measures included the number of retrieved oocytes, mature oocytes (MII), fertilization, and top-quality embryo rates. The secondary outcome measures included duration of stimulation, gonadotropin dose and peak estradiol level. RESULTS The number of oocytes retrieved, MII oocytes, fertilization rate, duration of stimulation, or gonadotropin dose were similar in the letrozole and tamoxifen groups, compared to controls. Top-quality embryo rate was lower in the tamoxifen group compared to controls (25% vs 39.4%, respectively, P = 0.034). The abnormal fertilization rate was higher in the letrozole group compared to controls (7.8% vs 3.60%, respectively, P = 0.015). A stepwise logistic regression analysis revealed that letrozole and peak estradiol were significantly associated with abnormal fertilization (OR 11.94; 95% CI 2.35-60.4, P = 0.003 for letrozole and OR 1.075; 95% CI 1.024-1.12, P = 0.004 per 100 unit change in estradiol). CONCLUSIONS There may be a negative effect of letrozole or tamoxifen on fertilization and embryo quality, in fertility preservation cycles. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Yael Shulman
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel.
| | - Benny Almog
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Yael Kalma
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Yuval Fouks
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Foad Azem
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Yoni Cohen
- IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| |
Collapse
|
48
|
Predictors and outcomes in breast cancer patients who did or did not pursue fertility preservation. Breast Cancer Res Treat 2021; 186:429-437. [PMID: 33392838 DOI: 10.1007/s10549-020-06031-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Breast cancer is the most common cancer in reproductive age women, and treatment can affect fertility; however, there is often concern regarding the safety of increased estradiol (E2) levels and potential delays in treatment with ovarian stimulation for fertility preservation (FP). The aim of this study was to compare recurrence and survival in breast cancer patients who pursued FP without concurrent letrozole to those who did not (non-FP). METHODS We reviewed charts of women with breast cancer who contacted the FP patient navigator (PN) at Northwestern University from 01/2005-01/2018. Oncology and fertility outcome data were collected. Data were analyzed by Chi-square test or regression, as appropriate. Kaplan-Meier curves were used to examine breast cancer recurrence and survival. Statistical analyses were performed with SPSS IBM Statistics 26.0 for Windows. RESULTS 332 patients were included, of which 157 (47.3%) underwent FP. Median days to treatment after consulting the PN was 35 in the FP group and 21 in non-FP (p < 0.05). Cancer recurrence was noted in 7 (4.7%) FP patients and 13 (7.9%) non-FP patients (NS), and mortality in 5 (3.2%) FP patients and 7 (4.2%) non-FP patients (NS). Within the FP group, no significant differences were found in recurrence or mortality based on ER status, age, BMI, peak E2 level or total gonadotropin dose. Likelihood of pursuing FP was primarily a function of age and parity, and was not affected by breast cancer stage. To date, 21 have used cryopreserved specimens, and 13 (62%) had a live birth. CONCLUSIONS FP is safe and effective in breast cancer patients, regardless of receptor status; E2 elevations and the 2-week delay in treatment start are unlikely to be clinically significant. These findings are unique in that our institution does not use concomitant letrozole during stimulation to minimize E2 elevations in breast cancer patients.
Collapse
|
49
|
Marklund A, Lundberg FE, Eloranta S, Hedayati E, Pettersson K, Rodriguez-Wallberg KA. Reproductive Outcomes After Breast Cancer in Women With vs Without Fertility Preservation. JAMA Oncol 2021; 7:86-91. [PMID: 33211089 PMCID: PMC7677871 DOI: 10.1001/jamaoncol.2020.5957] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Question What are the long-term reproductive outcomes after breast cancer in women with vs without a history of fertility preservation? Findings In this population-based nationwide cohort study of 425 Swedish women with breast cancer who underwent fertility preservation, fertility preservation at the time of breast cancer diagnosis was associated with a significantly higher rate of postdiagnosis live births and assisted reproduction treatments, without any negative association with all-cause survival following fertility preservation. Meaning The findings of this study may be relevant for reproductive counseling of women with breast cancer diagnosed at reproductive age. Importance The practice of fertility preservation (FP) in women with breast cancer (BC) is spreading, but long-term reproductive outcomes after FP are largely unknown. Objective To investigate the long-term reproductive outcomes in women who did or did not undergo FP at the time of BC diagnosis. Design, Setting, and Participants A Swedish nationwide cohort study was conducted to investigate the long-term reproductive outcomes of women with BC receiving FP at 1 of the regional FP programs from 1994 to 2017 (n = 425). Population comparators with BC but without history of FP (n = 850) were sampled from regional BC registers, matched on age, calendar period of diagnosis, and county. Data on live births, assisted reproductive technology (ART) use, and mortality were retrieved from population-based registers. Data analysis was performed from January to September 2020. Exposures History of having received FP compared with no history of FP (unexposed). Main Outcomes and Measures The primary outcome was hazard ratios (HRs) of live births and ART treatments following BC in women with vs without FP and the cumulative incidence of these events in the presence of the competing risk of death. Results Women who had undergone FP (n = 425) had lower parity (302 [71.1%] were nulliparous compared with 171 [20.1%] in the unexposed group), were younger (mean [SD] age, 32.1 [4.0] vs 33.3 [3.6] years), more often had estrogen receptor–positive tumors (289 [68.0%] vs 515 [60.6%]), and were more often scheduled for chemotherapy (399 [93.9%] vs 745 [87.7%]). Of 425 women exposed to FP, 97 (22.8%) had at least 1 post-BC live birth (mean follow-up, 4.6 years), compared with 74 of 850 women (8.7%) unexposed to FP (mean follow-up, 4.8 years). Overall, live birth rates after BC were significantly higher among women with FP (adjusted hazard ratio [aHR], 2.3; 95% CI, 1.6-3.3). The 5-year and 10-year cumulative incidence of post-BC live births was 19.4% and 40.7% among FP-exposed women vs 8.6% and 15.8% among comparators, respectively. Rates of ART use were also higher in the FP group (aHR, 4.8; 95% CI, 2.2-10.7). The all-cause mortality rate was lower in women exposed to FP (aHR, 0.4; 95% CI, 0.3-0.7), with 5-year cumulative incidence of death of 5.3% (95% CI, 3.1%-9.0%) vs 11.1% (95% CI, 8.7%-14.1%) for women with vs without FP. Conclusions and Relevance In this cohort study of Swedish women after a BC diagnosis, successful pregnancy after BC was possible both in women with and without FP at the time of diagnosis, but a significantly higher likelihood of post-BC live births and ART treatments was observed in women who underwent FP, without any negative association with all-cause survival. This information is valuable for health care clinicians responsible for oncologic treatment and reproductive counseling of women diagnosed with breast cancer at reproductive age.
Collapse
Affiliation(s)
- Anna Marklund
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Obstetrics and Gynecology, Södersjukhuset, Stockholm, Sweden
| | - Frida E Lundberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Eloranta
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Elham Hedayati
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Medical Unit of Breast Cancer Sarcoma and Endocrine Tumors, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Karin Pettersson
- Department of Women's Health, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum J 5:30, Stockholm, Sweden
| |
Collapse
|
50
|
Xing Z, Wang X, Liu J, Zhang M, Feng K, Wang X. Hsa_circ_0069094 accelerates cell malignancy and glycolysis through regulating the miR-591/HK2 axis in breast cancer. Cell Signal 2020; 79:109878. [PMID: 33309838 DOI: 10.1016/j.cellsig.2020.109878] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 01/08/2023]
Abstract
Circular RNAs (circRNAs) are implicated in the initiation and advancement of diverse tumors. CircRNA hsa_circ_0069094 (circ_0069094) has been reported to be upregulated in BC, but the biological role of circ_0069094 in BC is indistinct. Hence, we aimed to survey the biological role of circ_0069094 in BC. In the present study, we verified that circ_0069094 was upregulated in BC tissues and cells. BC patients with high circ_0069094 expression had a poor prognosis. Functional analysis revealed that circ_0069094 silencing induced apoptosis, curbed proliferation, and reduced glycolysis in BC cells in vitro, but circ_0069094 overexpression had an opposing influence. Also, circ_0069094 knockdown reduced BC growth in vivo. Mechanically, circ_0069094 was validated as a decoy for miR-591, which targeted HK2. Importantly, circ_0069094 sponged miR-591 to regulate HK2 expression. Both miR-591 silencing and HK2 overexpression counteracted circ_0069094 inhibition-mediated influence on cell proliferation, apoptosis, and glycolysis in BC cells. In conclusion, these results indicated that circ_0069094 facilitated cell malignancy and glycolysis by upregulating HK2 through adsorbing miR-591, suggesting that circ_0069094 might be a prognostic biomarker and therapeutic target for BC.
Collapse
Affiliation(s)
- Zeyu Xing
- Department of Breast Cancer, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xin Wang
- Department of Breast Cancer, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiaqi Liu
- Department of Breast Cancer, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Menglu Zhang
- Department of Breast Cancer, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Kexin Feng
- Department of Breast Cancer, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiang Wang
- Department of Breast Cancer, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|