1
|
Wallace WH, Kelsey TW, Morrison D, Anderson RA. Live birth and maternity outcome in childhood and adolescent cancer survivors under 18 years at diagnosis: a 40-year population-based cohort study. Br J Cancer 2024; 131:1309-1319. [PMID: 39266623 PMCID: PMC11473688 DOI: 10.1038/s41416-024-02818-0] [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: 10/24/2023] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Survival from childhood and adolescent cancer has increased, but the chance of a livebirth in female survivors under 18 years at diagnosis may be reduced. METHODS We performed a national population-based analysis, including all female cancer survivors diagnosed in Scotland before the age of 18 years between 1981 and 2012. Scottish Cancer Registry records were linked to Scottish maternity records. Females from the exposed group with no pregnancies before cancer diagnosis (n = 2118) were compared with three general population controls matched for age and year of diagnosis. FINDINGS The cumulative incidence of a livebirth for all diagnoses was reduced to 37% (95% CI 33-40%) for cancer survivors at 30 years of age vs 58% (57-60%) for controls. The deficit varying by diagnosis: for lymphoid leukaemia, the cumulative incidence at 30 years was 29% (23-36%) vs 57% (52-61%) for controls with similar deficits in CNS tumours and retinoblastoma. There was a steady improvement in the chance of livebirth in those diagnosed more recently. INTERPRETATION We have shown a reduced chance of livebirth in female survivors of cancer diagnosed before age 18. The deficit is present for all diagnoses.
Collapse
Affiliation(s)
- W H Wallace
- Department of Paediatric Haematology and Oncology, Royal Hospital for Children and Young People and University of Edinburgh, Edinburgh, UK.
| | - T W Kelsey
- School of Computer Science, University of St. Andrews, North Haugh, St. Andrews, UK
| | - D Morrison
- Scottish Cancer Registry, Public Health Scotland, 1 South Gyle Crescent, Edinburgh, UK
| | - R A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Hosoi H, Miyachi M, Teramukai S, Sakabayashi S, Tsuchiya K, Kuwahara Y, Onodera R, Matsuyama K, Yokota I, Hojo H, Okita H, Hata JI, Hamasaki M, Tsuneyoshi M, Oda Y, Nakazawa A, Kato M, Takimoto T, Horibe K, Hara JI, Suita S, Hanada R, Masaki H, Nozaki M, Ikeda H, Kishimoto S, Kaneko M, Kawai A, Morikawa Y. Results of the JRS-I LRA0401 and LRB0402 Japan Rhabdomyosarcoma Study Group trials for low-risk embryonal rhabdomyosarcoma. Int J Clin Oncol 2024; 29:1746-1755. [PMID: 39177879 DOI: 10.1007/s10147-024-02608-x] [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: 01/04/2024] [Accepted: 08/04/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Failure-free survival (FFS) rates of low-risk patients with rhabdomyosarcoma improved in Intergroup Rhabdomyosarcoma Study IV after the escalation of cyclophosphamide total dose to 26.4 g/m2. However, this dose may increase the risk of adverse events, including infertility, in some patients. The JRS-I LRA0401 and LRB0402 protocols aimed to reduce the cyclophosphamide dose to 9.6 g/m2 and 17.6 g/m2, respectively, without decreasing the FFS rates. METHODS Subgroup-A patients received eight cycles (24 weeks) of vincristine, actinomycin D, and 1.2 g/m2/cycle cyclophosphamide. Subgroup-B patients received eight cycles (24 weeks) of vincristine, actinomycin D, and 2.2 g/m2/cycle cyclophosphamide, followed by six cycles (24 weeks) of vincristine and actinomycin D. Group II/III patients in both subgroups received radiotherapy. RESULTS In subgroup A (n = 12), the 3-year FFS rate was 83% (95% confidence interval [CI], 48-96), and the 3-year overall survival (OS) rate was 100%. Only one isolated local recurrence was observed (8.3%). There were no unexpected grade-4 toxicities and no deaths. In subgroup B (n = 16), the 3-year FFS and OS rates were 88% (95% CI, 59-97) and 94% (95% CI, 63-99), respectively. There were no unexpected grade 4 toxicities and no deaths. CONCLUSIONS Shorter duration therapy using vincristine, actinomycin D, and lower dose cyclophosphamide with or without radiotherapy for patients with low-risk subgroup A rhabdomyosarcoma (JRS-I LRA0401 protocol) and moderate reduction of cyclophosphamide dose for patients with low-risk subgroup B rhabdomyosarcoma (JRS-I LRB0402 protocol) did not compromise FFS.
Collapse
Affiliation(s)
- Hajime Hosoi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine (KPUM), Kyoto, Japan.
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan.
- Department of Nursing, Doshisha Women's College of Liberal Arts, Kyoto, Japan.
| | - Mitsuru Miyachi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine (KPUM), Kyoto, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Satoshi Teramukai
- Department of Biostatistics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
- Department of Clinical Trial Design and Management, Translational Research Center, Kyoto University Hospital, Kyoto, Japan
| | - Satomi Sakabayashi
- Department of Biostatistics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kunihiko Tsuchiya
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine (KPUM), Kyoto, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Yasumichi Kuwahara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine (KPUM), Kyoto, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Rie Onodera
- Translational Research Informatics Center, Kobe, Japan
- Department of Clinical Trial Design and Management, Translational Research Center, Kyoto University Hospital, Kyoto, Japan
- Health and Medical Innovation, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kotone Matsuyama
- Translational Research Informatics Center, Kobe, Japan
- Department of Health Policy and Management, Nippon Medical School, Tokyo, Japan
| | - Isao Yokota
- Department of Biostatistics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Biostatistics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Hiroshi Hojo
- Department of Diagnostic Pathology, School of Medicine, Fukushima Medical University, Fukushima, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Hajime Okita
- Division of Diagnostic Pathology, Keio University School of Medicine, Tokyo, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Jun-Ichi Hata
- Experimental Medicine and Life Science, Kanagawa, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Minori Hamasaki
- Department of Pathology, Shizuoka Children's Hospital, Shizuoka, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Masazumi Tsuneyoshi
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
- Department of Diagnostic Pathology, Fukuoka Sanno Hospital, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Atsuko Nakazawa
- Department of Pathology, Saitama Children's Medical Center, Saitama, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Miho Kato
- Childhood Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Tetsuya Takimoto
- Childhood Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Keizo Horibe
- Clinical Research Center, NHO Nagoya Medical Center, Aichi, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Jun-Ichi Hara
- Department of Pediatric Hematology/Oncology, Children's Medical Center, Osaka City General Hospital, Osaka, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Sachiyo Suita
- Department of Pediatric Surgery, Faculty of Medicine, Kyushu University, Fukuoka, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Ryoji Hanada
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Hidekazu Masaki
- Department of Radiology, National Center for Child Health and Development (NCCHD), Tokyo, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Miwako Nozaki
- Department of Radiology, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Hitoshi Ikeda
- Department of Pediatric Surgery, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Seiji Kishimoto
- Department of Otolaryngology, Head and Neck Surgery, Kameda Medical Center, Chiba, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Michio Kaneko
- Department of Pediatric Surgery, Institute of Clinical Medicine, University of Tsukuba, Ibaraki, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| | - Yasuhide Morikawa
- Department of Pediatric Surgery, International University of Health and Welfare Hospital, Tokyo, Japan
- Japan Rhabdomyosarcoma Study Group (JRSG), Tokyo, Japan
| |
Collapse
|
3
|
Robinson M, Meller L, Patterson M. Premature ovarian insufficiency in pediatric cancer patients: a 10 year Rady Children's hospital experience. J Pediatr Endocrinol Metab 2024:jpem-2024-0207. [PMID: 39295284 DOI: 10.1515/jpem-2024-0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024]
Abstract
OBJECTIVES To highlight the occurrence of premature ovarian insufficiency in pediatric cancer patients and determine which patient characteristics or treatment modalities are associated with ovarian failure and recovery. METHODS Between August 2011-August 2021, 36 of 2,661 patients with cancer were identified to have subsequent ovarian failure. Data collected included cancer type, diagnosis age, types of chemotherapy, bone marrow transplant or radiation treatment, peak FSH, peak AMH, GnRHa treatment, type of hormone replacement therapy, and if ovarian function recovery occurred. RESULTS The most common cancer type identified was ALL. The mean age of diagnosis was 8.5±4.3 years and mean age of peak FSH value was 12.6±2.8 years. Most patients (97.2 %) were treated with alkylating agents and 72.2 % received radiation. Most patients (72.2 %) received hormone therapy, and 15.8 % of patients received GnRHa Lupron. Ten patients (27.8 %) had ovarian function recovery. Diagnosis age and treatment type were recovery predictors in multivariate regression modeling. Each year older in age was associated with a 30 % decrease in odds of recovery (OR: 0.7, CI: 0.5-0.95, p=0.035), and alkylating agent treatment without transplant was associated with a 3-fold increase in odds of recovery (OR: 3, CI: 2.7-564, p=0.007). CONCLUSIONS This retrospective review demonstrates that POI can occur in pediatric cancer survivors, emphasizing the importance of educating patients on potential long-term effects of cancer treatment and importance of routine surveillance. This study confirmed that recovery of ovarian function is possible, especially when diagnosed at a younger age, making continued monitoring essential.
Collapse
Affiliation(s)
| | - Leo Meller
- UC San Diego School of Medicine, La Jolla, CA, USA
| | - Mary Patterson
- UC San Diego School of Medicine, La Jolla, CA, USA
- Pediatric Endocrinology, Rady Children's Hospital, San Diego, CA, USA
| |
Collapse
|
4
|
Ali A, Lee YK, Alias H, Zainuddin AA. Fertility preservation in Malaysian pediatric cohort: a survey of healthcare providers' knowledge, practice, attitude, perceptions and barriers. Front Pediatr 2024; 12:1419515. [PMID: 39363970 PMCID: PMC11446869 DOI: 10.3389/fped.2024.1419515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/30/2024] [Indexed: 10/05/2024] Open
Abstract
Introduction Impaired future fertility potential secondary to gonadotoxic therapies for childhood cancer is a shattering aftermath faced by childhood cancer survivors. Fertility preservation (FP) has emerged as a key to mitigate this unwelcomed sequelae. FP services catering to the needs of children and adolescents (C&A) population in developing countries are limited. Malaysia recently launched its pioneering pediatrics FP services. Aims of study To evaluate healthcare providers' (HCPs) FP knowledge, practice behaviors, attitudes, perceptions, and barriers towards FP counseling/services (C/S) for the C&A cohort. Methods A questionnaire-based study was conducted utilizing a questionnaire consisting of 51 items which was adapted from G.Quinn et al. The questionnaire was distributed both online and physically amongst HCPs in a tertiary center. Ethical committee approval was granted by the Research Ethical Committee, Universiti Kebangsaan Malaysia. Results A total of 102 HCPs completed the questionnaires. The majority of respondents were Malays (74.5%), females (80.4%), gynecology/pediatrics specialty (76.5%), and had children (88.2%). Nearly 72% of HCPs demonstrated good knowledge of FP. Almost 73% of HCPs consulted reproductive specialists (RES) on potential fertility issues and over 80% of HCPs referred patients who enquired on fertility issues to RES. Only 17% of HCPs practiced FP discussion, 12% reported no available person to discuss FP, and 10% of HCPs were unaware of who to discuss FP with. Patients' inability to afford FP (30.4%) tops the list of barriers to FP C/S, followed by limited available information on FP for patients (17.6%) and patients too ill to delay treatment (12.7%). Most HCPs (88.2%) demonstrated unfavorable attitudes towards FP C/S. Discussions In general, the majority of our HCP respondents demonstrated good current FP knowledge and practice behaviors. Mitigating several controversial issues in FP would improve HCPs' attitude towards FP. Main barriers to the uptake of FP C/S for C&A were patient and resource barriers. Addressing these issues by funding aid for FP procedures, increasing FP knowledge dispersion, as well as developing age-appropriate FP-related educational materials would improve FP service provision for C&A in the future. Conclusions In conclusion, successful corrective action combined with strategic planning points to a promising future for Malaysia's FP services provision for C&A.
Collapse
Affiliation(s)
- Anizah Ali
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
- Paediatrics and Adolescent Gynaecology (PAG) Unit, Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Yew Kong Lee
- Department of Primary Care Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hamidah Alias
- Department of Paediatric, Faculty of Medicine, Hospital Tunku Ampuan Besar Tuanku Aishah Rohani, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Ani Amelia Zainuddin
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
- Paediatrics and Adolescent Gynaecology (PAG) Unit, Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
Jimenez-Kurlander L, DeRosa A, Kostrzewa CE, Moskowitz CS, Bogardus K, Antal Z, Wolden S, La Quaglia MP, Basu EM, Cardenas FI, Kramer K, Kushner BH, Cheung NKV, Modak S, Friedman DN. Ovarian function in female survivors of high-risk neuroblastoma. Pediatr Blood Cancer 2024; 71:e31181. [PMID: 38967225 PMCID: PMC11269001 DOI: 10.1002/pbc.31181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION Data on ovarian function in neuroblastoma survivors are limited. We sought to determine the prevalence of ovarian dysfunction in a cohort of high-risk neuroblastoma survivors and compare outcomes among survivors treated with and without autologous stem cell rescue (ASCR) preceded by myeloablative chemotherapy. METHODS Retrospective review of female survivors of high-risk neuroblastoma ≥5 years from diagnosis, diagnosed between 1982 and 2014, and followed in a tertiary cancer center. Participants were divided into two groups: individuals treated with conventional chemotherapy ± radiation ("non-ASCR") (n = 32) or with chemotherapy ± radiation followed by myeloablative chemotherapy with ASCR ("ASCR") (n = 51). Ovarian dysfunction was defined as follicle-stimulating hormone ≥15 mU/mL, while premature ovarian insufficiency (POI) was defined as persistent ovarian dysfunction requiring hormone replacement therapy. Poisson models were used to determine prevalence ratios of ovarian dysfunction and POI. RESULTS Among 83 females (median attained age: 19 years [range, 10-36]; median follow-up: 15 years [range, 7-36]), 49 (59%) had ovarian dysfunction, and 34 (41%) developed POI. Survivors treated with ASCR were 3.2-fold more likely to develop ovarian dysfunction (95% CI: 1.8-6.0; p < 0.001) and 4.5-fold more likely to develop POI (95% CI: 1.7-11.7; p = 0.002) when compared with those treated with conventional chemotherapy, after adjusting for attained age. Two participants in the non-ASCR group and six in the ASCR group achieved at least one spontaneous pregnancy. DISCUSSION Ovarian dysfunction is prevalent in female high-risk neuroblastoma survivors, especially after ASCR. Longitudinal follow-up of larger cohorts is needed to inform counseling about the risk of impaired ovarian function after neuroblastoma therapy.
Collapse
Affiliation(s)
| | - Amelia DeRosa
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caroline E. Kostrzewa
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chaya S. Moskowitz
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kimberly Bogardus
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Zoltan Antal
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Suzanne Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering, New York, NY, USA
| | | | - Ellen M. Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian H. Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Nai-Kong V. Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Danielle Novetsky Friedman
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
6
|
Zhou B, Kwan B, Desai MJ, Nalawade V, Henk J, Viravalli N, Murphy JD, Nathan PC, Ruddy KJ, Shliakhtsitsava K, Su HI, Whitcomb BW. Association of platinum-based chemotherapy with live birth and infertility in female survivors of adolescent and young adult cancer. Fertil Steril 2024; 121:1020-1030. [PMID: 38316209 PMCID: PMC11128346 DOI: 10.1016/j.fertnstert.2024.01.039] [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: 09/21/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
OBJECTIVE To estimate the effect of platinum-based chemotherapy on live birth (LB) and infertility after cancer, in order to address a lack of treatment-specific fertility risks for female survivors of adolescent and young adult cancer, which limits counseling on fertility preservation decisions. DESIGN Retrospective cohort study. SETTING US administrative database. PATIENTS We identified incident breast, colorectal, and ovarian cancer cases in females aged 15-39 years who received platinum-based chemotherapy or no chemotherapy and matched them to females without cancer. INTERVENTION Platinum-based chemotherapy. MAIN OUTCOME MEASURES We estimated the effect of chemotherapy on the incidence of LB and infertility after cancer, overall, and after accounting for competing events (recurrence, death, and sterilizing surgeries). RESULTS There were 1,287 survivors in the chemotherapy group, 3,192 in the no chemotherapy group, and 34,147 women in the no cancer group, with a mean age of 33 years. Accounting for competing events, the overall 5-year LB incidence was lower in the chemotherapy group (3.9%) vs. the no chemotherapy group (6.4%). Adjusted relative risks vs. no chemotherapy and no cancer groups were 0.61 (95% confidence interval [CI] 0.42-0.82) and 0.70 (95% CI 0.51-0.93), respectively. The overall 5-year infertility incidence was similar in the chemotherapy group (21.8%) compared with the no chemotherapy group (20.7%). The adjusted relative risks vs. no chemotherapy and no cancer groups were 1.05 (95% CI 0.97-1.15) and 1.42 (95% CI 1.31-1.53), respectively. CONCLUSIONS Cancer survivors treated with platinum-based chemotherapy experienced modestly increased adverse fertility outcomes. The estimated effects of platinum-based chemotherapy were affected by competing events, suggesting the importance of this analytic approach for interpretations that ultimately inform clinical fertility preservation decisions.
Collapse
Affiliation(s)
- Beth Zhou
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, California
| | - Brian Kwan
- Division of Biostatistics and Bioinformatics, Herbert Wertheim School of Public Health, University of California San Diego, San Diego, California; Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, California
| | - Milli J Desai
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, California
| | - Vinit Nalawade
- Moores Cancer Center, University of California San Diego, San Diego, California; Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California
| | - Joe Henk
- OptumLabs, Eden Prarie, Minnesota
| | | | - James D Murphy
- Moores Cancer Center, University of California San Diego, San Diego, California; Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California
| | - Paul C Nathan
- Department of Paediatrics, Division of Haematology/Oncology, the Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Ksenya Shliakhtsitsava
- Division of Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - H Irene Su
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, California; Moores Cancer Center, University of California San Diego, San Diego, California
| | - Brian W Whitcomb
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts.
| |
Collapse
|
7
|
Romero SAD, Au L, Flores-Ortega RE, Helsten T, Palomino H, Kaiser BN, Echevarria M, Lukas K, Freeman K, Zou J, Aristizabal P, Armenian S, Su HI. Let's TOC Fertility: A stepped wedge cluster randomized controlled trial of the Telehealth Oncofertility Care (TOC) intervention in children, adolescent and young adult cancer survivors. Contemp Clin Trials 2024; 141:107537. [PMID: 38614445 PMCID: PMC11520196 DOI: 10.1016/j.cct.2024.107537] [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: 12/15/2023] [Revised: 03/20/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
INTRODUCTION Children, adolescent, and young adult cancer survivors experience overall increased risks of infertility that are preventable through effective fertility preservation services prior to starting cancer treatment. Oncofertility care is the evidence-based practice of informing newly diagnosed cancer patients about their reproductive risks and supporting shared decision-making on fertility preservation services. Despite longstanding clinical guidelines, oncofertility care delivery continues to be limited and highly variable across adult and pediatric oncology settings. MATERIALS AND METHODS We describe the design of a stepped wedge cluster randomized clinical trial to evaluate the effectiveness of the multi-component Telehealth Oncofertility Care (TOC) intervention conducted in 20 adult and pediatric oncology clinics across three health systems in Southern California. Intervention components are: 1) electronic health record-based oncofertility needs screen and referral pathway to a virtual oncofertility hub; 2) telehealth oncofertility counseling through the hub; and 3) telehealth oncofertility financial navigation through the hub. We hypothesize the intervention condition will be associated with increased proportions of patients who engage in goal-concordant oncofertility care (i.e., engagement in reproductive risk counseling and fertility preservation services that meet the patient's fertility goals) and improved patient-reported outcomes, compared to the usual care control condition. We will also evaluate intervention implementation in a mixed-methods study guided by implementation science frameworks. DISCUSSION Our overall goal is to speed implementation of a scalable oncofertility care intervention at cancer diagnosis for children, adolescent and young adult cancer patients to improve their future fertility and quality of life. TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT05443737.
Collapse
Affiliation(s)
- Sally A D Romero
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, School of Medicine, United States of America; Herbert Wertheim School of Public Health and Human Longevity Science, University of California at San Diego, United States of America.
| | - Lauren Au
- Department of Medicine, University of Hawai'i at Mānoa John A Burns School of Medicine, United States of America
| | - Ricardo E Flores-Ortega
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, School of Medicine, United States of America
| | - Teresa Helsten
- Department of Medicine, University of California San Diego, School of Medicine, United States of America; Moores Cancer Center, University of California San Diego, United States of America
| | - Helen Palomino
- Cancer Resource Center of the Desert, United States of America
| | - Bonnie N Kaiser
- Department of Anthropology and Global Health Program, University of California San Diego, United States of America
| | | | - Kara Lukas
- City of Hope Comprehensive Cancer Center, United States of America
| | - Kendall Freeman
- City of Hope Comprehensive Cancer Center, United States of America
| | - Jingjing Zou
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California at San Diego, United States of America
| | - Paula Aristizabal
- Moores Cancer Center, University of California San Diego, United States of America; Department of Pediatrics, University of California San Diego, School of Medicine, United States of America
| | - Saro Armenian
- City of Hope Comprehensive Cancer Center, United States of America
| | - H Irene Su
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, School of Medicine, United States of America; Moores Cancer Center, University of California San Diego, United States of America
| |
Collapse
|
8
|
Dorfman CS, Stalls JM, Shelby RA, Arthur SS, Acharya K, Davidson B, Corbett C, Greenup RA. Impact of Financial Costs on Patients' Fertility Preservation Decisions: An Examination of Qualitative Data from Female Young Adults with Cancer and Oncology Providers. J Adolesc Young Adult Oncol 2024; 13:502-513. [PMID: 38294823 DOI: 10.1089/jayao.2023.0108] [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] [Indexed: 02/01/2024] Open
Abstract
Purpose: To examine the impact of financial costs on fertility preservation decisions among female young adults (YAs) with cancer. Methods: Female YAs (N = 18; aged 21-36) with a history of cancer and oncology providers (N = 12) were recruited from an National Cancer Institute-designated comprehensive cancer center in a state without insurance coverage for fertility preservation. YAs and providers completed individual interviews and a brief online assessment. Qualitative description using thematic analysis was used to identify, analyze, and report common themes. Descriptive statistics was used to characterize the sample. Results: Female YAs and oncology providers highlighted the critical role that high out-of-pocket costs play in YAs' fertility preservation decisions along with the value that enhanced insurance coverage for fertility preservation would have for increasing female YAs' access to and utilization of fertility preservation. Although providers were concerned about preservation costs for their patients, they reported that their concerns did not impact whether they referred interested female YAs to reproductive specialists. Oncology providers expressed concern about inequities in utilization of fertility preservation for female and racially/ethnically minoritized YAs that were exacerbated by the high out-of-pocket fertility preservation costs. Conclusion: Cost is a significant barrier to fertility preservation for female YA cancer patients. Female YAs of reproductive age may benefit from decision support tools to assist with balancing the cost of fertility preservation with their values and family building goals. Policy-relevant interventions may mitigate cost barriers and improve access to care.
Collapse
Affiliation(s)
- Caroline S Dorfman
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
- Supportive Care and Survivorship Center, Duke Cancer Institute, Durham, North Carolina, USA
| | - Juliann M Stalls
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
- Supportive Care and Survivorship Center, Duke Cancer Institute, Durham, North Carolina, USA
| | - Rebecca A Shelby
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
- Supportive Care and Survivorship Center, Duke Cancer Institute, Durham, North Carolina, USA
| | - Sarah S Arthur
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Kelly Acharya
- Division of Reproductive Endocrinology and Infertility; Department of Obstetrics and Gynecology, Duke University, Durham, North Carolina, USA
| | - Brittany Davidson
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology, Duke University, Durham, North Carolina, USA
| | - Cheyenne Corbett
- Supportive Care and Survivorship Center, Duke Cancer Institute, Durham, North Carolina, USA
| | - Rachel A Greenup
- Division of Surgical Oncology, Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
9
|
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
|
10
|
Seok J, Park HS, Cetin E, Ghasroldasht MM, Liakath FB, Al-Hendy A. The potent paracrine effect of umbilical cord mesenchymal stem cells mediates mitochondrial quality control to restore chemotherapy-induced damage in ovarian granulosa cells. Biomed Pharmacother 2024; 172:116263. [PMID: 38350369 DOI: 10.1016/j.biopha.2024.116263] [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: 12/08/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
The basic principle of chemotherapy is to attack cells with fast growth, and cancer cells are targeted by anticancer drugs because they have a faster growth rate than normal cells. High doses of anticancer drugs may cause an irreversible decline in reproductive capacity, and novel approaches for fertility preservation and/or restoration after anticancer treatment are urgently needed. Here, we provide important insights into the recovery of human reproductive cells damaged by chemotherapy. We performed a detailed screening of the cytokines of various human mesenchymal stem cells (hMSCs) to select superior MSCs. Also, we analyzed the Ovarian granulosa cell (OGC)-)-specific functions for restoring function, apoptosis, and mitochondrial functions to confirm the recovery mechanism in damaged OGCs. As a result, we demonstrated that conditioned media (CM) of Umbilical cord mesenchymal stem cells (UC-MSCs) could restore the functions of damaged OGCs primarily through antiapoptotic and antioxidant effects. Furthermore, CM changed the phenotype of damaged OGCs to an energetic status by restoring mitochondrial function and enhanced the mitochondrial metabolic activity decreased by chemotherapy. Finally, we demonstrated that the restoration of mitochondrial function in damaged OGCs was mediated through mitochondrial autophagy (mitophagy). Our findings offer new insights into the potential of stem cell-based therapy for fertility preservation and/or restoration in female cancer patients.
Collapse
Affiliation(s)
- Jin Seok
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Hang-Soo Park
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Esra Cetin
- Department of Obstetrics and Gynecology, Hurley Medical Center, Michigan State University, 1 Hurley Plaza, Flint, MI 48503, USA
| | | | - Farzana Begum Liakath
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA.
| |
Collapse
|
11
|
Tan CY, Francis-Levin N, Stelmak D, Iannarino NT, Zhang A, Herrel L, Ellman E, Walling E, Moravek MB, Chugh R, Zebrack B. Differentiating gender-based reproductive concerns among adolescent and young adult cancer patients: A mixed methods study. J Psychosoc Oncol 2024; 42:526-542. [PMID: 38164962 DOI: 10.1080/07347332.2023.2291798] [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] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Few studies have examined the distinct reproductive concerns (RC) of men and women in the adolescent and young adult (AYA) cancer patient population. The purpose of this mixed-methods study was to explore and differentiate the RC of AYAs. METHODS Participants completed the Reproductive Concerns After Cancer (RCAC) scale and participated in a semistructured interview. Interviews were deductively coded based on an analytic schema derived from the RCAC. RESULTS After identifying participants through the electronic health record, 27 younger AYAs, ages 12-25, enrolled in the study. Four inductive themes emerged and differed by gender. These include differential temporality, acceptance, and openness to alternatives, partner influence, and parental/guardian influence. AYA men reported fewer RC (M = 49.4, SD = 9.6) compared to AYA women (M = 56.8, SD = 8.4). CONCLUSIONS Oncofertility care providers are advised to account for short- and long-ranging concerns based on AYAs' gender. Future evaluations of patient-reported outcome measures specific to AYA RC are recommended.
Collapse
Affiliation(s)
- Chiu Yi Tan
- School of Social Policy and Practice, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nina Francis-Levin
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan, Ann Arbor, Michigan, USA
| | - Daria Stelmak
- School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas T Iannarino
- Department of Language, Culture, and the Arts, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Anao Zhang
- School of Social Work, University of Michigan, Ann Arbor, Michigan, USA
| | - Lindsey Herrel
- Department of Urology, University of Michigan, Ann Arbor, Michigan, USA
| | - Erin Ellman
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Emily Walling
- Department of Pediatric Hematology/Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Molly B Moravek
- Department of Urology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Division of Reproductive Endocrinology and Infertility, University of Michigan, Ann Arbor, Michigan, USA
| | - Rashmi Chugh
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Brad Zebrack
- School of Social Work, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Pei C, Todorov P, Cao M, Kong Q, Isachenko E, Rahimi G, Mallmann-Gottschalk N, Uribe P, Sanchez R, Isachenko V. Comparative Transcriptomic Analyses for the Optimization of Thawing Regimes during Conventional Cryopreservation of Mature and Immature Human Testicular Tissue. Int J Mol Sci 2023; 25:214. [PMID: 38203385 PMCID: PMC10778995 DOI: 10.3390/ijms25010214] [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: 11/09/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Cryopreservation of human testicular tissue, as a key element of anticancer therapy, includes the following stages: saturation with cryoprotectants, freezing, thawing, and removal of cryoprotectants. According to the point of view existing in "classical" cryobiology, the thawing mode is the most important consideration in the entire process of cryopreservation of any type of cells, including cells of testicular tissue. The existing postulate in cryobiology states that any frozen types of cells must be thawed as quickly as possible. The technologically maximum possible thawing temperature is 100 °C, which is used in our technology for the cryopreservation of testicular tissue. However, there are other points of view on the rate of cell thawing, according to how thawing should be carried out at physiological temperatures. In fact, there are morphological and functional differences between immature (from prepubertal patients) and mature testicular tissue. Accordingly, the question of the influence of thawing temperature on both types of tissues is relevant. The purpose of this study is to explore the transcriptomic differences of cryopreserved mature and immature testicular tissue subjected to different thawing methods by RNA sequencing. Collected and frozen testicular tissue samples were divided into four groups: quickly (in boiling water at 100 °C) thawed cryopreserved mature testicular tissue (group 1), slowly (by a physiological temperature of 37 °C) thawed mature testicular tissue (group 2), quickly thawed immature testicular tissue (group 3), and slowly thawed immature testicular tissue (group 4). Transcriptomic differences were assessed using differentially expressed genes (DEG), the Kyoto Encyclopedia of Genes and Genomes (KEGG), gene ontology (GO), and protein-protein interaction (PPI) analyses. No fundamental differences in the quality of cells of mature and immature testicular tissue after cryopreservation were found. Generally, thawing of mature and immature testicular tissue was more effective at 100 °C. The greatest difference in the intensity of gene expression was observed in ribosomes of cells thawed at 100 °C in comparison with cells thawed at 37 °C. In conclusion, an elevated speed of thawing is beneficial for frozen testicular tissue.
Collapse
Affiliation(s)
- Cheng Pei
- Department of Obstetrics and Gynecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (C.P.); (Q.K.); (E.I.); (N.M.-G.)
| | - Plamen Todorov
- Institute of Biology and Immunology of Reproduction of Bulgarian Academy of Sciences (BAS), 1113 Sofia, Bulgaria;
| | - Mengyang Cao
- Department of Obstetrics and Gynecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (C.P.); (Q.K.); (E.I.); (N.M.-G.)
| | - Qingduo Kong
- Department of Obstetrics and Gynecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (C.P.); (Q.K.); (E.I.); (N.M.-G.)
| | - Evgenia Isachenko
- Department of Obstetrics and Gynecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (C.P.); (Q.K.); (E.I.); (N.M.-G.)
| | - Gohar Rahimi
- Department of Obstetrics and Gynecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (C.P.); (Q.K.); (E.I.); (N.M.-G.)
- Medizinisches Versorgungszentrum AMEDES für IVF- und Pränatalmedizin in Köln GmbH, 50968 Cologne, Germany
| | - Nina Mallmann-Gottschalk
- Department of Obstetrics and Gynecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (C.P.); (Q.K.); (E.I.); (N.M.-G.)
| | - Pamela Uribe
- Center of Excellence in Translational Medicine, Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Temuco 4810296, Chile; (P.U.); (R.S.)
- Department of Internal Medicine, Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile
| | - Raul Sanchez
- Center of Excellence in Translational Medicine, Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Temuco 4810296, Chile; (P.U.); (R.S.)
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile
| | - Volodimir Isachenko
- Department of Obstetrics and Gynecology, Medical Faculty, Cologne University, 50931 Cologne, Germany; (C.P.); (Q.K.); (E.I.); (N.M.-G.)
| |
Collapse
|
13
|
Kipling LM, Shandley LM, Mertens AC, Spencer JB, Howards PP. The use of fertility treatments among reproductive-aged women after cancer. Fertil Steril 2023:S0015-0282(23)02079-4. [PMID: 38103881 DOI: 10.1016/j.fertnstert.2023.12.012] [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: 07/14/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVE To examine whether female cancer survivors are more likely to pursue care for infertility after cancer than women without cancer. DESIGN Population-based cohort study involving detailed interviews regarding reproductive history. SETTING Not applicable. PATIENTS Female cancer survivors aged 22-45 years, who were at least 2 years after a cancer diagnosis between the ages of 20 and 35 years (n = 1,036), and age-matched comparison women with no cancer history (n = 1,026). EXPOSURE History of cancer vs. no history of cancer. MAIN OUTCOME MEASURE(S) Each cancer survivor was randomly matched to a comparison woman, who was assigned an artificial age at cancer diagnosis equal to that of her match. Matching was repeated 1,000 times. Outcomes of visiting a doctor for help becoming pregnant or undergoing fertility treatment were modeled using Cox proportional hazards regression, comparing survivors after a cancer diagnosis to age-matched comparison women, adjusted for race, income, residence, education, and parity. RESULTS Only 25.5% of cancer survivors reported meeting their desired family size before a cancer diagnosis. The median time from diagnosis to interview among survivors was 7 (interquartile range 5-11) years. Cancer survivors were more likely to report having no children (32.6%) at the interview compared with women with no cancer history (19.5%). Survivors were not more likely to visit a doctor for help becoming pregnant compared with women without a cancer history, matched on birth year and followed by the age at which cancer survivors received their diagnosis (hazard ratio [HR] 1.16, 95% simulation interval [SI] 0.78-1.74). Compared with cancer-free women, cancer survivors had similar probabilities of pursuing any treatment (adjusted HR [aHR] 0.88, 95% SI 0.46-1.56), using hormones or medications (aHR 0.86, 95% SI 0.46-1.63), or undergoing intrauterine insemination (aHR 1.26, 95% SI 0.40-5.88) to conceive. Cancer survivors were slightly more likely to pursue surgical interventions to become pregnant (HR 1.55, 95% SI 0.67-3.71). Of those who visited a doctor but declined to pursue fertility treatment, one-quarter of women reported declining treatment due to cost. CONCLUSION Cancer survivors did not use fertility treatments at higher rates than the general population. Further counseling and education surrounding fertility options are recommended for young adult female cancer patients after treatment is completed.
Collapse
Affiliation(s)
- Lauren M Kipling
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Lisa M Shandley
- Division of Reproductive Endocrinology and Infertility, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia.
| | - Ann C Mertens
- Department of Pediatrics, Aflac Cancer Center, Emory University School of Medicine, Atlanta, Georgia
| | - Jessica B Spencer
- Division of Reproductive Endocrinology and Infertility, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| | - Penelope P Howards
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| |
Collapse
|
14
|
Nerb L, Yang E, Exume D, Dornisch A, Zhou B, Helsten T, Kaiser BN, Romero SA, Su HI. Development, Usability Testing, and Implementation Assessment of Cancer Related Infertility Score Predictor, an Online Cancer Related Infertility Risk Counseling Tool. J Adolesc Young Adult Oncol 2023; 12:843-850. [PMID: 37184539 PMCID: PMC10739786 DOI: 10.1089/jayao.2022.0187] [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] [Indexed: 05/16/2023] Open
Abstract
Purpose: Oncofertility counseling of female cancer patients lacks efficient access to tailored and valid infertility risk estimates to support shared decision-making on fertility preservation treatments. The objective was to develop, conduct user-centered design, and plan clinic-based implementation of the Cancer Related Infertility Score Predictor (CRISP), a web-based tool to support infertility risk counseling. Methods: Using a mixed methods design, literature review was undertaken to abstract data on infertility, primary ovarian insufficiency, and amenorrhea risks of common cancer treatments. The CRISP website was programmed to take user input about patient ages and cancer treatments and generate a risk summary. Using user experience methodology and semistructured interviews, usability testing and implementation assessment were conducted with 12 providers recruited from 5 medical centers in Southern California. Results: The web-based CRISP tool encompasses infertility risk data for 60 treatment regimens among 10 cancer types. Usability testing demonstrated that the tool is intuitive and informed minor modifications, including adding crowd-sourced submission of additional cancer treatments. Participants rated the tool as credible, advantageous over current provider methods to ascertain infertility risks, and useful for tailoring treatment planning and counseling patients. A key barrier was lack of information on some cancer treatments. Fit within clinical workflow was feasible, particularly with electronic health record integration. Conclusions: The novel, web-based CRISP tool is a feasible, acceptable, and appropriate tool to address provider knowledge gap about cancer related infertility risks and use for patient counseling. CRISP has significant potential to support tailored oncofertility counseling in the heterogeneous young cancer patient population.
Collapse
Affiliation(s)
- Laura Nerb
- School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Emily Yang
- School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Dominique Exume
- Department of Obstetrics and Gynecology, Penn State Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Anna Dornisch
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | - Beth Zhou
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Teresa Helsten
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Bonnie N. Kaiser
- Department of Anthropology and Global Health Program, University of California San Diego, La Jolla, California, USA
| | - Sally A.D. Romero
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - H. Irene Su
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
15
|
Drechsel KCE, Broer SL, Stoutjesdijk FS, Twisk JWR, van den Berg MH, Lambalk CB, van Leeuwen FE, Overbeek A, van den Heuvel-Eibrink MM, van Dorp W, de Vries ACH, Loonen JJ, van der Pal HJ, Kremer LC, Tissing WJ, Versluys B, Kaspers GJL, van Dulmen-den Broeder E, Veening MA. Clinical and self-reported markers of reproductive function in female survivors of childhood Hodgkin lymphoma. J Cancer Res Clin Oncol 2023; 149:13677-13695. [PMID: 37522923 PMCID: PMC10590326 DOI: 10.1007/s00432-023-05035-z] [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: 05/22/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023]
Abstract
PURPOSE To evaluate the impact of treatment for Hodgkin lymphoma (HL) on clinical reproductive markers and pregnancy outcomes. METHODS This study was embedded within the DCOG LATER-VEVO study; a Dutch, multicenter, retrospective cohort study between 2004 and 2014. Serum anti-Müllerian hormone (AMH), follicle stimulating hormone (FSH), inhibin B, antral follicle count (AFC), and self-reported (first) pregnancy outcomes were evaluated in female childhood HL survivors and controls. RESULTS 84 HL survivors and 798 controls were included, aged 29.6 and 32.7 years old at time of assessment. Median age at HL diagnosis was 13.4 years. Cyclophosphamide equivalent dose (CED-score) exceeded 6000 mg/m2 in 56 women and 14 survivors received pelvic irradiation. All clinical markers were significantly deteriorated in survivors (odds-ratio for low AMH (< p10) 10.1 [95% CI 4.9; 20.6]; low AFC (< p10) 4.6 [95% CI 2.1; 9.9]; elevated FSH (> 10 IU/l) 15.3 [95% CI 5.7; 41.1], low Inhibin B (< 20 ng/l) 3.6 [ 95% CI 1.7; 7.7], p < 0.001). Pregnancy outcomes were comparable between survivors and controls (± 80% live birth, ± 20% miscarriage). However, survivors were significantly younger at first pregnancy (27.0 years vs 29.0 years, P = 0.04). Adjusted odds-ratio for time to pregnancy > 12 months was 2.5 [95% CI 1.1; 5.6] in survivors, p = 0.031. Adverse outcomes were specifically present after treatment with procarbazine and higher CED-score. CONCLUSION HL survivors appear to have an impaired ovarian reserve. However, chance to achieve pregnancy seems reassuring at a young age. Additional follow-up studies are needed to assess fertile life span and reproductive potential of HL survivors, in particular for current HL treatments that are hypothesized to be less gonadotoxic.
Collapse
Affiliation(s)
- K C E Drechsel
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands.
| | - S L Broer
- Department of Reproductive Medicine & Gynecology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F S Stoutjesdijk
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - J W R Twisk
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M H van den Berg
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
| | - C B Lambalk
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - F E van Leeuwen
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Overbeek
- Department of Obstetrics and Gynaecology, Northwest Clinics, Alkmaar, The Netherlands
| | - M M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Paediatric Hemato-Oncology, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - W van Dorp
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A C H de Vries
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Paediatric Hemato-Oncology, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J J Loonen
- Department of Hematology, Radboudumc Center of Expertise for Cancer Survivorship, Radboud University Medical Center, Nijmegen, The Netherlands
| | - H J van der Pal
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - L C Kremer
- Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - W J Tissing
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B Versluys
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Heamatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - G J L Kaspers
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - E van Dulmen-den Broeder
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
| | - M A Veening
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
16
|
Amargant F, Zhou LT, Yuan Y, Nahar A, Krisher RL, Spate LD, Roberts RM, Prather RS, Rowell EE, Laronda MM, Duncan FE. FGF2, LIF, and IGF1 (FLI) supplementation during human in vitro maturation enhances markers of gamete competence. Hum Reprod 2023; 38:1938-1951. [PMID: 37608600 DOI: 10.1093/humrep/dead162] [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: 02/07/2023] [Revised: 07/05/2023] [Indexed: 08/24/2023] Open
Abstract
STUDY QUESTION Does a chemically defined maturation medium supplemented with FGF2, LIF, and IGF1 (FLI) improve in vitro maturation (IVM) of cumulus-oocyte complexes (COCs) obtained from children, adolescents, and young adults undergoing ovarian tissue cryopreservation (OTC)? SUMMARY ANSWER Although FLI supplementation did not increase the incidence of oocyte meiotic maturation during human IVM, it significantly improved quality outcomes, including increased cumulus cell expansion and mitogen-activated protein kinase (MAPK) expression as well as enhanced transzonal projection retraction. WHAT IS KNOWN ALREADY During OTC, COCs, and denuded oocytes from small antral follicles are released into the processing media. Recovery and IVM of these COCs is emerging as a complementary technique to maximize the fertility preservation potential of the tissue. However, the success of IVM is low, especially in the pediatric population. Supplementation of IVM medium with FLI quadruples the efficiency of pig production through improved oocyte maturation, but whether a similar benefit occurs in humans has not been investigated. STUDY DESIGN, SIZE, DURATION This study enrolled 75 participants between January 2018 and December 2021 undergoing clinical fertility preservation through the Fertility & Hormone Preservation & Restoration Program at the Ann & Robert H. Lurie Children's Hospital of Chicago. Participants donated OTC media, accumulated during tissue processing, for research. PARTICIPANTS/MATERIALS, SETTING, METHODS Participants who underwent OTC and include a pediatric population that encompassed children, adolescents, and young adults ≤22 years old. All participant COCs and denuded oocytes were recovered from media following ovarian tissue processing. IVM was then performed in either a standard medium (oocyte maturation medium) or one supplemented with FLI (FGF2; 40 ng/ml, LIF; 20 ng/ml, and IGF1; 20 ng/ml). IVM outcomes included meiotic progression, cumulus cell expansion, transzonal projection retraction, and detection of MAPK protein expression. MAIN RESULTS AND THE ROLE OF CHANCE The median age of participants was 6.3 years, with 65% of them classified as prepubertal by Tanner staging. Approximately 60% of participants had been exposed to chemotherapy and/or radiation prior to OTC. On average 4.7 ± 1 COCs and/or denuded oocytes per participant were recovered from the OTC media. COCs (N = 41) and denuded oocytes (N = 29) were used for IVM (42 h) in a standard or FLI-supplemented maturation medium. The incidence of meiotic maturation was similar between cohorts (COCs: 25.0% vs 28.6% metaphase II arrested eggs in Control vs FLI; denuded oocytes: 0% vs 5.3% in Control vs FLI). However, cumulus cell expansion was 1.9-fold greater in COCs matured in FLI-containing medium relative to Controls and transzonal projection retraction was more pronounced (2.45 ± 0.50 vs 1.16 ± 0.78 projections in Control vs FLIat 16 h). Additionally, MAPK expression was significantly higher in cumulus cells obtained from COCs matured in FLI medium for 16-18 h (chemiluminescence corrected area 621,678 vs 2,019,575 a.u., P = 0.03). LIMITATIONS, REASONS FOR CAUTION Our samples are from human participants who exhibited heterogeneity with respect to age, diagnosis, and previous treatment history. Future studies with larger sample sizes, including adult participants, are warranted to determine the mechanism by which FLI induces MAPK expression and activation. Moreover, studies that evaluate the developmental competence of eggs derived from FLI treatment, including assessment of embryos as outcome measures, will be required prior to clinical translation. WIDER IMPLICATIONS OF THE FINDINGS FLI supplementation may have a conserved beneficial effect on IVM for children, adolescents, and young adults spanning the agricultural setting to clinical fertility preservation. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by Department of Obstetrics and Gynecology startup funds (F.E.D.), Department of Surgery Faculty Practice Plan Grant and the Fertility & Hormone Preservation & Restoration Program at the Ann & Robert H. Lurie Children's Hospital of Chicago (M.M.L. and E.E.R.). M.M.L. is a Gesualdo Foundation Research Scholar. Y.Y.'s research is supported by the internal research funds provided by Colorado Center of Reproductive Medicine. Y.Y., L.D.S., R.M.R., and R.S.P. have a patent pending for FLI. The remaining authors have no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Farners Amargant
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Luhan T Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ye Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| | - Asrafun Nahar
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| | | | - Lee D Spate
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - R Michael Roberts
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Erin E Rowell
- Division of Pediatric Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Monica M Laronda
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
17
|
Din HN, Singh-Carlson S, Corliss HL, Hartman SJ, Strong D, Madanat H, Su HI. Perceived and Objective Fertility Risk Among Female Survivors of Adolescent and Young Adult Cancer. JAMA Netw Open 2023; 6:e2337245. [PMID: 37819662 PMCID: PMC10568355 DOI: 10.1001/jamanetworkopen.2023.37245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023] Open
Abstract
Importance Fertility is important to many survivors of adolescent and young adult (AYA) cancer, yet data on this population's fertility perceptions and their alignment with objective infertility risk are scant. Objective To assess whether estimated treatment gonadotoxicity and posttreatment menstrual pattern are associated with higher infertility risk perception. Design, Setting, and Participants This retrospective cohort study included female young adult survivors of cancer diagnosed between ages 15 and 39 years were recruited between March 25, 2015, and September 24, 2018, from 2 state cancer registries, social media, and clinician referrals to participate in a study of posttreatment ovarian function. Data analysis occurred between March 1 and September 1, 2022. Exposures Participants reported their menstrual pattern. Estimated treatment gonadotoxicity was ascertained through medical record review. Main Outcomes and Measures Participants reported infertility risk perception and were categorized as increased risk (feeling less fertile or unable to become pregnant) or no increased risk (feeling more or as fertile) compared with female individuals their age. Objective infertility risk was determined by estimated gonadotoxicity, menstrual pattern, and ovarian reserve testing of self-collected dried blood spots. Multivariable logistic regression identified factors associated with perceived infertility and underestimation or overestimation of infertility risk. Results This study included 785 female participants with a mean (SD) age of 33.2 (4.8) years at enrollment and 25.9 (5.7) years at diagnosis. Most participants self-identified their race and ethnicity as White (585 [74.5%]) and non-Hispanic (628 [78.7%]). Most participants (483 [61.5%]) perceived a higher risk of infertility compared with female participants their age. Prior exposure to moderate- or high-gonadotoxicity treatments was associated with higher odds of perceiving increased infertility risk compared with exposure to low-gonadotoxicity treatments (adjusted odds ratio [AOR], 2.73 [95% CI, 1.87-3.97] and 15.39 [95% CI, 5.52-42.96], respectively). Amenorrhea and irregular cycles were associated with higher odds of perceiving increased infertility risk (AOR, 3.98 [95% CI, 2.13-7.41] and 1.69 [95% CI, 1.19-2.40], respectively). Perceived infertility risk had minimal agreement with objective risk (κ = 0.19). Multiparity (AOR, 4.17 [95% CI, 2.61-6.64]) was associated with increased odds of underestimation, while older age (AOR, 0.94 [95% CI, 0.89-0.98]), endocrine comorbidity (AOR, 0.35 [95% CI, 0.18-0.69]), and prior infertility (AOR, 0.16 [95% CI, 0.07-0.38]) were associated with lower odds of underestimation. Multiparity (AOR, 0.48 [95% CI, 0.27-0.86]), breast cancer (AOR, 0.38 [95% CI, 0.20-0.73]), and skin cancer (AOR, 0.24 [95% CI, 0.11-0.51]) were associated with lower odds of overestimation. Conclusions and Relevance In this cohort study, survivors of AYA cancer had high rates of perceiving increased infertility risk but frequently overestimated or underestimated their risk. These findings suggest that counseling on infertility risk throughout survivorship may reduce misalignment between perceptions and actual risk, decrease fertility-related psychological distress, and inform family planning decisions.
Collapse
Affiliation(s)
- Hena Naz Din
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla
- School of Public Health, San Diego State University, San Diego, California
| | | | - Heather L. Corliss
- School of Public Health, San Diego State University, San Diego, California
- Center for Research on Sexuality and Sexual Health, San Diego State University, San Diego, California
- Institute for Behavioral and Community Health, San Diego State University, San Diego, California
| | - Sheri J. Hartman
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla
- Moores Cancer Center, University of California San Diego, La Jolla
| | - David Strong
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla
- Moores Cancer Center, University of California San Diego, La Jolla
| | - Hala Madanat
- School of Public Health, San Diego State University, San Diego, California
- Division of Research and Innovation, San Diego State University, San Diego, California
- Institute for Behavioral and Community Health, San Diego State University, San Diego, California
| | - H. Irene Su
- Moores Cancer Center, University of California San Diego, La Jolla
- Division of Reproductive Endocrinology and Infertility, University of California San Diego, La Jolla
| |
Collapse
|
18
|
Peipert BJ, Potapragada NR, Lantos PM, Harris BS, Reinecke J, Goldman KN. A Geospatial Analysis of Disparities in Access to Oncofertility Services. JAMA Oncol 2023; 9:1364-1370. [PMID: 37561485 PMCID: PMC10416086 DOI: 10.1001/jamaoncol.2023.2780] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/24/2023] [Indexed: 08/11/2023]
Abstract
Importance Fertility preservation (FP), including oocyte and embryo cryopreservation prior to gonadotoxic therapy, is an urgent and essential component of comprehensive cancer care. Geographic proximity to a center offering FP is a critical component of ensuring equitable access for people with cancer desiring future fertility. Objective To characterize the distribution of centers offering FP services in the US, quantify the number of self-identified reproductive-age female individuals living outside of geographically accessible areas, and investigate the association between geographic access and state FP mandates. Design, Setting, and Participants This cross-sectional analysis calculated 2-hour travel time isochrone maps for each center based on latitude and longitude coordinates. Population-based geospatial analysis in the US was used in this study. Fertility clinics identified through the 2018 Centers for Disease Control and Prevention Fertility Clinic Success Rates Report were defined as oncofertility centers by meeting 4 criteria: (1) offered oocyte and embryo cryopreservation, (2) performed at least 1 FP cycle in 2018, (3) served people without partners, and (4) had an accredited laboratory. County-level data were obtained from the 2020 US Census, with the primary at-risk population identified as reproductive-age female individuals aged 15 years to 44 years. The analysis was performed from 2021 to 2022. Exposures Location outside of 2-hour travel time isochrone of an oncofertility center. Main Outcomes and Measures Oncofertility centers were compared with centers not meeting criteria and were classified by US region, state FP mandate status, number of assisted reproductive technology cycles performed, and number of FP cycles performed. The number and percentage of at-risk patients, defined as those living outside of accessible service areas by state, were identified. Results Among 456 Centers for Disease Control and Prevention-reporting fertility clinics, 86 (18.9%) did not meet the criteria as an oncofertility center. A total of 3.63 million (5.70%) reproductive-age female individuals lack geographic access to an oncofertility center. States with FP mandates have the highest rates of eligible female patients with geographic access (98.54%), while states without active or pending legislation have the lowest rates (79.57%). The greatest disparities in geographic access to care are most concentrated in the Mountain West and West North Central regions. Conclusions and Relevance Patients face numerous barriers to comprehensive cancer care, including a lack of geographic access to centers capable of offering FP services. This cross-sectional study identified disparities in geographic access and potential opportunities for strategic expansion.
Collapse
Affiliation(s)
- Benjamin J Peipert
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, North Carolina
| | - Nivedita R Potapragada
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Paul M Lantos
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Benjamin S Harris
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, North Carolina
| | - Joyce Reinecke
- Alliance for Fertility Preservation, Lafayette, California
| | - Kara N Goldman
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
19
|
Gupta D, Singh S, Shukla S, Shrivastava S. Oncofertility: Treatment options from bench to bedside. CANCER PATHOGENESIS AND THERAPY 2023; 1:284-289. [PMID: 38327602 PMCID: PMC10846294 DOI: 10.1016/j.cpt.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 02/09/2024]
Abstract
In recent years, there has been continuous improvement in the treatment and diagnosis of cancer, which has led to a significant improvement in the survival rate of cancer patients. Treatments that include chemotherapy, radiotherapy, surgery, or combined therapy have several side effects that may lead to premature ovarian insufficiency in females or substantial male germ cell loss. Reproductive biologists recommend that all patients who are diagnosed with a malignant tumor must undergo a consultation for fertility protection and preservation. In this review, we discuss the background knowledge, methods, and options for fertility preservation and how these new strategies help oncologists, surgeons, pediatricians, and hematologists, conserve fertility and be aware of the concepts, methods, and importance of fertility guards. This review may aid in the advancement of novel personalized methods for fertility preservation according to patients' conditions.
Collapse
Affiliation(s)
- Divya Gupta
- Reproductive Biology and Toxicology Lab, SOS in Zoology, Jiwaji University, Gwalior, Madhya Pradesh 474011, India
| | - Shubham Singh
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur (C.G.) 495009, India
| | - Sangeeta Shukla
- Reproductive Biology and Toxicology Lab, SOS in Zoology, Jiwaji University, Gwalior, Madhya Pradesh 474011, India
| | - Sadhana Shrivastava
- Reproductive Biology and Toxicology Lab, SOS in Zoology, Jiwaji University, Gwalior, Madhya Pradesh 474011, India
| |
Collapse
|
20
|
Abstract
Importance An estimated 15 000 children and adolescents aged 0 to 19 years are diagnosed with cancer each year in the US, and more than 85% survive for at least 5 years. By 45 years of age, approximately 95% of people who survive childhood cancer will develop a significant health problem related to the childhood cancer diagnosis or its treatment. Observations Approximately 500 000 people currently alive in the US have survived childhood cancer. The most common severe or life-threatening chronic health problems related to childhood cancer or its treatment are endocrine disorders such as hypothyroidism or growth hormone deficiency (44%), subsequent neoplasms such as breast cancer or thyroid cancer (7%), and cardiovascular disease such as cardiomyopathy or congestive heart failure, coronary artery disease, and cerebrovascular disease (5.3%). Medical conditions related to a cancer diagnosis during childhood or adolescence are most commonly caused by the radiation therapy and the chemotherapies used to treat cancer and may develop at varying lengths of time after exposure to these treatments. Individuals at highest risk for developing treatment-related health problems include patients with brain cancer treated with cranial irradiation (approximately 70% develop severe or life-threatening health problems) and allogeneic hematopoietic stem cell transplant recipients (approximately 60% develop severe or life-threatening health problems). Individuals at the lowest risk for developing treatment-related health problems include those who survived solid tumors (such as Wilms tumor) treated with surgical resection alone or with minimal chemotherapy, for whom the prevalence of subsequent health problems is similar to people who did not have cancer during childhood or adolescence. People diagnosed with childhood cancer in the 1990s who survived for at least 5 years after the cancer diagnosis have a shorter lifespan (by about 9 years) vs children who were not diagnosed with cancer in the 1990s. Conclusions and Relevance Approximately 500 000 individuals currently alive in the US have survived childhood cancer. The most common adverse effects in individuals who survived childhood cancer are endocrine disorders, subsequent neoplasms, and cardiovascular disease. There is a need for clinicians and patients to have heightened awareness of these complications.
Collapse
Affiliation(s)
- Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, Heersink School of Medicine, University of Alabama at Birmingham
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Heersink School of Medicine, University of Alabama at Birmingham
| | - Emily S Tonorezos
- Office of Cancer Survivorship, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, Heersink School of Medicine, University of Alabama at Birmingham
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Heersink School of Medicine, University of Alabama at Birmingham
| |
Collapse
|
21
|
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
|
22
|
Moravek MB, Pavone ME, Burns K, Kashanian JA, Anderson RA, Klosky JL, Rotz SJ, Stern CJ, Rodriguez-Wallberg KA, Levine JM, Meacham LR. Fertility assessment and treatment in adolescent and young adult cancer survivors. Pediatr Blood Cancer 2023; 70 Suppl 5:e28854. [PMID: 37381152 DOI: 10.1002/pbc.28854] [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: 07/01/2020] [Revised: 11/23/2020] [Accepted: 12/04/2020] [Indexed: 06/30/2023]
Abstract
In the survivorship setting, adolescent and young adult (AYA) cancer survivors frequently demonstrate little knowledge of infertility risk, are unclear regarding their fertility status, and may under- or overestimate their treatment-related risk for infertility. In female AYA survivors, ovarian function usually parallels fertility, and can be assessed with serum hormone levels and ultrasonography. Posttreatment fertility preservation may be appropriate for survivors at risk for primary ovarian insufficiency. In male AYA survivors, fertility and gonadal function are not always equally affected, and can be assessed with a semen analysis and serum hormones, respectively. As reproductive health issues are commonly cited as an important concern by survivors of AYA cancer, multidisciplinary care teams including oncology, endocrinology, psychology, and reproductive medicine are advocated, with the aim of optimal provision of fertility advice and care for AYA cancer survivors.
Collapse
Affiliation(s)
- Molly B Moravek
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Mary Ellen Pavone
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Karen Burns
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | | | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - James L Klosky
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Seth J Rotz
- Division of Pediatric Hematology, Oncology, and Bone Marrow Transplantation, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Catharyn J Stern
- Melbourne IVF, East Melbourne, Victoria, Australia
- The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Kenny A Rodriguez-Wallberg
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Solna, Sweden
| | - Jennifer M Levine
- Division of Pediatric Hematology/Oncology, Cornell University Joan and Sanford I Weill Medical College, New York, New York
| | - Lillian R Meacham
- Aflac Cancer Center, Children's Healthcare of Atlanta, Atlanta, Georgia
- Department of Pediatrics, Division of Hematology/Oncology and Endocrinology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
23
|
Close A, Burns K, Bjornard K, Webb M, Chavez J, Chow EJ, Meacham L. Fertility preservation in pediatric leukemia and lymphoma: A report from the Children's Oncology Group. Pediatr Blood Cancer 2023; 70:e30407. [PMID: 37194406 PMCID: PMC10525075 DOI: 10.1002/pbc.30407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/18/2023]
Abstract
Certain chemotherapy agents, radiation, and surgery can all negatively impact future fertility. Consults regarding treatment-related risk for infertility and gonadal late effects of these agents should occur at the time of diagnosis as well as during survivorship. Counseling on fertility risk has traditionally varied significantly across providers and institutions. We aim to provide a guide to standardize the assignment of gonadotoxic risk, which can be used in counseling patients both at the time of diagnosis and in survivorship. Gonadotoxic therapies were abstracted from 26 frontline Children's Oncology Group (COG) phase III protocols for leukemia/lymphoma, in use from 2000-2022. A stratification system based on gonadotoxic therapies, sex, and pubertal status was used to assign treatments into minimal, significant, and high level of increased risk for gonadal dysfunction/infertility. Risk levels were assigned to protocols and different treatment arms to aid oncologists and survivor care providers in counseling patients regarding treatment-related gonadotoxicity. Males were most commonly at high risk, with at least one high-risk arm in 14/26 protocols (54%), followed by pubertal females (23% of protocols) and prepubertal females (15% of protocols). All patients who received direct gonadal radiation or hematopoietic stem cell transplant (HSCT) were considered at high risk. Partnering with patients and their oncology/survivorship team is imperative for effective fertility counseling both prior to and post treatment, and this comprehensive guide can be used as a tool to standardize and improve reproductive health counseling in patients undergoing COG-based leukemia/lymphoma care.
Collapse
Affiliation(s)
- Allison Close
- Helen DeVos Children’s Hospital, Division of Hematology/Oncology, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Karen Burns
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Kari Bjornard
- Department of Pediatrics, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA; Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA
| | - Martine Webb
- University of California, Los Angeles, Division of Internal Medicine, Los Angeles, CA, USA
| | - Josuah Chavez
- Helen DeVos Children’s Hospital, Division of Hematology/Oncology, Grand Rapids, MI, USA
| | - Eric J. Chow
- Fred Hutchinson Cancer Center, Seattle Children’s Hospital, Seattle, WA, USA
| | - Lillian Meacham
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Pediatric Hematology/Oncology/BMT, Emory University Atlanta, GA USA
| |
Collapse
|
24
|
Nelson M, Levine J. Current Issues in Fertility Preservation Among Pediatric and Adolescent Cancer Patients. Curr Oncol Rep 2023; 25:793-802. [PMID: 37036623 DOI: 10.1007/s11912-023-01401-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2023] [Indexed: 04/11/2023]
Abstract
PURPOSE OF REVIEW Although fertility preservation is not yet fully integrated into the comprehensive cancer care of pediatric and adolescent patients, advances continue to take place. This review summarizes recent updates and trends for health care professionals caring for these patients. RECENT FINDINGS The creation of standardized infertility risk assessment guidelines offers the opportunity to provide greater consistency in clinical care and to provide a current baseline for future research studies seeking to refine risk stratification for individual patients. New agents are being introduced into cancer care; as their use increases, information about their impact on fertility is being studied. Ovarian tissue cryopreservation offers a new standard of care option for fertility preservation, but additional studies are needed to further assess efficacy and impact on ovarian reserve among pediatric and adolescent patients. Standardization of fertility preservation recommendations among certain sub-populations may also make it easier to provide greater consistency in clinical care. Advances continue to be made in the field of fertility preservation, but dissemination of this information is critical to moving toward fertility preservation truly being a part of comprehensive cancer care.
Collapse
Affiliation(s)
- Marie Nelson
- Children's National Medical Center, Center for Cancer and Blood Disorders, 111 Michigan Avenue NW, Room W4-604, Washington, D.C., 20010, USA
| | - Jennifer Levine
- Children's National Medical Center, Center for Cancer and Blood Disorders, 111 Michigan Avenue NW, Room W4-604, Washington, D.C., 20010, USA.
| |
Collapse
|
25
|
Cherven B, Ivankova NV, Spencer JB, Fitzpatrick AM, Burns KC, Demedis J, Hoefgen HR, Mertens AC, Klosky JL. Examining decisional needs and contextual factors influencing fertility status assessment among young female survivors of childhood cancer: A sequential mixed methods study protocol. PLoS One 2023; 18:e0286511. [PMID: 37315007 PMCID: PMC10266625 DOI: 10.1371/journal.pone.0286511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023] Open
Abstract
INTRODUCTION Female cancer survivors who received gonadotoxic cancer treatment are at risk for profound diminished ovarian reserve and/or primary ovarian insufficiency with resulting infertility, which can be associated with distress and decreased quality of life.. Despite prioritizing future parenthood, many survivors are unsure of the impact of their treatment on their future fertility, and little is known about the perceived reproductive health needs and factors associated with receipt of a fertility status assessment (FSA). There is a lack of developmentally appropriate reproductive health decisional support interventions available for emerging adult cancer survivors. This study will explore the perceived reproductive health needs of emerging adult female survivors of childhood cancer and to identify decisional and contextual factors that influence pursuit of FSA using an explanatory sequential quantitative to qualitative mixed methods design. METHODS AND ANALYSIS This study will enroll 325 female survivors (aged 18 to 29 years and >1-year post treatment; diagnosed with cancer < age 21 years) from four cancer centers in the United States. Sociodemographic and developmental factors, reproductive knowledge and values, decisional needs, and receipt of an FSA will be assessed through a web-based survey. Informed by survey findings, a subset of participants will be recruited for qualitative interviews to explore decisional factors associated with uptake of an FSA. Clinical data will be abstracted from the medical records. Multivariable logistic regression models will be developed to identify factors associated with FSA and qualitative descriptive analysis will be used to develop themes from the interviews. Quantitative and qualitative findings will be merged using a joint display to develop integrated study conclusions and direct future interventional research.
Collapse
Affiliation(s)
- Brooke Cherven
- Aflac Cancer and Blood Disorders Center at Children’s Healthcare of Atlanta, Atlanta, GA, United States of America
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Nataliya V. Ivankova
- Department of Health Services Administration, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Jessica B. Spencer
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Anne M. Fitzpatrick
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Karen C. Burns
- University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Jenna Demedis
- Center for Cancer and Blood Disorders at Children’s Hospital Colorado, Aurora, CO, United States of America
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Holly R. Hoefgen
- Aflac Cancer and Blood Disorders Center at Children’s Healthcare of Atlanta, Atlanta, GA, United States of America
- Washington University School of Medicine, St. Louis, MO, United States of America
| | - Ann C. Mertens
- Aflac Cancer and Blood Disorders Center at Children’s Healthcare of Atlanta, Atlanta, GA, United States of America
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - James L. Klosky
- Aflac Cancer and Blood Disorders Center at Children’s Healthcare of Atlanta, Atlanta, GA, United States of America
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
26
|
Yang E, Dornisch A, Nerb L, Helsten T, Kaiser BN, Aristizabal P, Armenian S, Torno LL, Baca NM, Genensen MC, Su HI, Romero SA. A Multicomponent Telehealth Intervention to Improve Oncofertility Care Delivery Among Young Cancer Patients: A Pilot Study. J Adolesc Young Adult Oncol 2023; 12:241-249. [PMID: 35639102 PMCID: PMC10124175 DOI: 10.1089/jayao.2021.0224] [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] [Indexed: 11/13/2022] Open
Abstract
Purpose: Oncofertility care for pediatric, adolescent, and young adult cancer patients remains under-implemented across adult and pediatric oncology settings. We pilot tested an electronic health record (EHR)-enabled multicomponent oncofertility intervention (including screening, referral, and fertility consult) in an adult academic oncology program and systematically assessed intervention fit to pediatric and community oncology programs. Methods: Using surveys (n = 33), audits (n = 143), and interviews (n = 21) guided by implementation science frameworks, we pilot tested the EHR-enabled intervention for oncofertility care in young cancer patients at an adult oncology program and evaluated implementation outcomes. We interviewed health care providers from seven regional oncology and fertility programs about intervention fit to their clinical contexts. Results: We recruited 33 health care providers from an adult oncology setting and 15 health care providers from seven additional oncology and fertility settings. At the adult oncology setting, the intervention was found to be appropriate, acceptable, and feasible and improved the screening of fertility needs (from 30% pre- to 51% post-intervention); yet, some patients did not receive appropriate referrals to fertility consults. Providers across all settings suggested content and context modifications, such as adding options to the intervention or allowing the screening component to pop up at a second visit, to improve and adapt the intervention to better fit their clinical care contexts. Conclusions: We found that the EHR-enabled intervention increased the rate of goal-concordant oncofertility care delivery at an adult oncology program. We also identified facilitators, barriers, and needed adaptations to the intervention required for implementation and scaling-up across diverse oncology settings.
Collapse
Affiliation(s)
- Emily Yang
- School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Anna Dornisch
- School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Laura Nerb
- School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Teresa Helsten
- Moores Cancer Center, and University of California San Diego, La Jolla, California, USA
| | - Bonnie N. Kaiser
- Department of Anthropology and Global Health Program, University of California San Diego, La Jolla, California, USA
| | | | - Saro Armenian
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Lilibeth L. Torno
- Hyundai Cancer Institute, Children's Health of Orange County, Orange, California, USA
| | - Nicole M. Baca
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California, USA
| | - Mark C. Genensen
- Eisenhower Lucy Curci Cancer Center, Rancho Mirage, California, USA
| | - H. Irene Su
- Moores Cancer Center, and University of California San Diego, La Jolla, California, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Sally A.D. Romero
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
27
|
Clasen NHZ, van der Perk MEM, Neggers SJCMM, Bos AME, van den Heuvel-Eibrink MM. Experiences of Female Childhood Cancer Patients and Survivors Regarding Information and Counselling on Gonadotoxicity Risk and Fertility Preservation at Diagnosis: A Systematic Review. Cancers (Basel) 2023; 15:cancers15071946. [PMID: 37046607 PMCID: PMC10093478 DOI: 10.3390/cancers15071946] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Childhood cancer patients and their families are increasingly offered oncofertility care including information regarding their risk of gonadal damage by paediatric oncologists, fertility counselling by fertility specialists and fertility preservation options. However, experiences regarding oncofertility care are underreported. We aimed to summarize the available evidence of experiences of female childhood cancer patients and survivors regarding oncofertility care. Methods: Manuscripts were systematically identified using the PubMed and Embase database. From, respectively, 1256 and 3857 manuscripts, 7 articles were included and assessed, including risk of bias assessment. Outcome measures included data describing experiences of female childhood cancer patients and survivors, regarding fertility information, counselling and/or preservation. Results: Female patients and survivors are variably satisfied with fertility information, report challenges in communication with healthcare professionals and prefer to receive general information at diagnosis and detailed fertility information later. Regrets after fertility counselling are underreported, but are associated with refusing fertility preservation. Lastly, regardless of counselling, female patients and survivors report fertility concerns about their future children’s health and effect on relationships. Conclusion: Currently, the satisfaction with oncofertility care varies and female patients or survivors report regrets and concerns regardless of receiving fertility information or counselling. These results may help to improve the content of fertility information, communication skills of healthcare professionals and timing of counselling.
Collapse
Affiliation(s)
- Nikita H Z Clasen
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, The Netherlands
- Faculty of Medicine, Vrije Universiteit Amsterdam, 1081CX Amsterdam, The Netherlands
| | | | - Sebastian J C M M Neggers
- Department of Internal Medicine, Section Endocrinology, Erasmus Medical Center, 3015GD Rotterdam, The Netherlands
| | - Annelies M E Bos
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, The Netherlands
- Division of Child Health, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584EA Utrecht, The Netherlands
| |
Collapse
|
28
|
Griffiths MJ, Marshall SA, Cousins FL, Alesi LR, Higgins J, Giridharan S, Sarma UC, Menkhorst E, Zhou W, Care AS, Donoghue JF, Holdsworth-Carson SJ, Rogers PA, Dimitriadis E, Gargett CE, Robertson SA, Winship AL, Hutt KJ. Radiotherapy exposure directly damages the uterus and causes pregnancy loss. JCI Insight 2023; 8:163704. [PMID: 36946464 PMCID: PMC10070119 DOI: 10.1172/jci.insight.163704] [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: 07/18/2022] [Accepted: 02/01/2023] [Indexed: 03/23/2023] Open
Abstract
Female cancer survivors are significantly more likely to experience infertility than the general population. It is well established that chemotherapy and radiotherapy can damage the ovary and compromise fertility, yet the ability of cancer treatments to induce uterine damage, and the underlying mechanisms, have been understudied. Here, we show that in mice total-body γ-irradiation (TBI) induced extensive DNA damage and apoptosis in uterine cells. We then transferred healthy donor embryos into ovariectomized adolescent female mice that were previously exposed to TBI to study the impacts of radiotherapy on the uterus independent from effects to ovarian endocrine function. Following TBI, embryo attachment and implantation were unaffected, but fetal resorption was evident at midgestation in 100% of dams, suggesting failed placental development. Consistent with this hypothesis, TBI impaired the decidual response in mice and primary human endometrial stromal cells. TBI also caused uterine artery endothelial dysfunction, likely preventing adequate blood vessel remodeling in early pregnancy. Notably, when pro-apoptotic protein Puma-deficient (Puma-/-) mice were exposed to TBI, apoptosis within the uterus was prevented, and decidualization, vascular function, and pregnancy were restored, identifying PUMA-mediated apoptosis as a key mechanism. Collectively, these data show that TBI damages the uterus and compromises pregnancy success, suggesting that optimal fertility preservation during radiotherapy may require protection of both the ovaries and uterus. In this regard, inhibition of PUMA may represent a potential fertility preservation strategy.
Collapse
Affiliation(s)
- Meaghan J Griffiths
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Sarah A Marshall
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Fiona L Cousins
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Lauren R Alesi
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jordan Higgins
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Saranya Giridharan
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Urooza C Sarma
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Ellen Menkhorst
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Wei Zhou
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Alison S Care
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Jacqueline F Donoghue
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Sarah J Holdsworth-Carson
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
- Epworth HealthCare, Richmond, Victoria, Australia
| | - Peter Aw Rogers
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Caroline E Gargett
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Amy L Winship
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Karla J Hutt
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
29
|
Ida H, Alicia GK, Anna F, Irene L, Anna N, Helena M, Maria E, Emir H, Hannah N. Quality of life among female childhood cancer survivors with and without premature ovarian insufficiency. J Cancer Surviv 2023; 17:101-109. [PMID: 33469826 PMCID: PMC9971100 DOI: 10.1007/s11764-021-00987-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/02/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE Due to an increase in survival, a growing population of childhood cancer survivors (CCS) is present. However, female CCS are at risk of developing premature ovarian insufficiency (POI) after cancer treatment. POI involves a decreased chance of conceiving and the increased infertility state has a large impact on affected individuals' health and mental life. The objective of this study was to investigate health state and well-being among female CCS with and without POI and healthy controls (HC). METHODS Female CCS treated in southern Sweden between 1964 and 2008 were included. Each patient was matched with a HC. The final study population included 167 female CCS and 164 HC that were examined between October 2010 and January 2015 at the Reproductive Medicine Centre at Skåne University Hospital in Malmö, Sweden. All participants, except for two HCs, answered an EQ-5D-3L questionnaire for measuring health state including a visual analogue scale (VAS) for estimating well-being. RESULTS There were 22 CCS with POI, none of the HC had POI. The mean health state differed among groups (unadjusted: P = 0.002; adjusted: P = 0.007). A difference in mean experienced well-being among groups was noted (unadjusted: P = 0.003; adjusted: P = 0.012). Lowest well-being was found in the CCS group with POI (P = 0.024). CONCLUSIONS Female CCS have a significantly decreased health state and well-being. Female CCS with POI additionally have the lowest self-estimated well-being. IMPLICATIONS FOR CANCER SURVIVORS Female CCS with POI should be identified early in order to give them adequate information and support.
Collapse
Affiliation(s)
- Hjelmér Ida
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Malmo, Sweden
| | - Gustafsson Kylberg Alicia
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Malmo, Sweden
| | - Fridenborg Anna
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Malmo, Sweden
| | - Leijonhufvud Irene
- Department of Translational Medicine, Reproductive Medicine Centre, Skåne University Hospital, Lund University, Malmo, Sweden
| | - Nyström Anna
- Department of Pediatrics, Pediatric Endocrinology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Mörse Helena
- Department of Pediatrics, Pediatric Oncology and Hematology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Elfving Maria
- Department of Pediatrics, Pediatric Endocrinology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Henic Emir
- Department of Translational Medicine, Reproductive Medicine Centre, Skåne University Hospital, Lund University, Malmo, Sweden
| | - Nenonen Hannah
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Malmo, Sweden.
| |
Collapse
|
30
|
Investigation of the female infertility risk associated with anti-cancer therapy. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023:10.1007/s12094-023-03087-8. [PMID: 36689055 DOI: 10.1007/s12094-023-03087-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023]
Abstract
Female infertility is a significant health issue worldwide with a rising incidence. Anti-cancer therapy is one of the most important reasons for increasing infertility. Although anti-cancer treatment increases the rate of survival, it decreases the quality of life through its side effects. The most substantial side effects are sexual dysfunction and infertility. Breast cancer is the most common cancer. The first-line treatment of breast cancer is chemotherapy by alkylating agents like cyclophosphamide, which leads to infertility. For instance, persistent chemotherapy-induced amenorrhea among breast cancer patients could affect almost half of the patients that undergo such therapy. However, some agents or therapeutic methods can ameliorate these intoxicating effects. Chemotherapy plus gonadotropin-releasing hormone agonist, in breast cancer patients, can not only improve overall survival but also reduce ovarian toxicity. Age plays an essential role in chemotherapy-induced amenorrhea. Chemotherapy at a younger age can reduce the risk of infertility. Gynecological cancers including uterine and ovarian cancer, which have high mortality rates, are the most related cancers to infertility. Surgery is the primary treatment of gynecological cancers. Studies demonstrated that fertility-sparing surgery is a better option than radical surgery. In addition, neoadjuvant chemotherapy is mostly a better option than primary cytoreductive surgery in terms of survival and fertility. Immune checkpoint inhibitors (ICIs) have recently played a major role in treating various cancer types. However, ICIs are associated with hypophysitis, which affects ovaries and can lead to infertility. There are some options for ovarian preservation such as embryo cryopreservation, oocyte cryopreservation, ovarian transposition, ovarian tissue cryopreservation, and ovarian suppression by GnRH agonists. Anti-müllerian hormone level can be utilized to monitor the ovarian reserve. Moreover, to avoid fertility loss, approaches such as using transplantation of human placenta mesenchymal stem cells, administrating anti-inflammatory agents and hormone therapy are under investigation.
Collapse
|
31
|
Harris ML, Feyissa TR, Bowden NA, Gemzell-Danielsson K, Loxton D. Contraceptive use and contraceptive counselling interventions for women of reproductive age with cancer: a systematic review and meta-analysis. BMC Med 2022; 20:489. [PMID: 36528586 PMCID: PMC9759910 DOI: 10.1186/s12916-022-02690-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND A lack of clarity exists regarding contraceptive uptake and counselling among women with cancer, despite these women having unique family planning needs. This study aimed to systematically review the available literature and produce an overall summary estimate of contraceptive use and counselling among women with cancer across the cancer care continuum. METHODS A systematic search of articles reporting on contraceptive counselling and/or contraceptive use among women of reproductive age (15-49 years) with cancer across the cancer care continuum (e.g. diagnosis, treatment, survivorship) was conducted in MEDLINE, Embase, CINAHL, Maternity and Infant Care and Cochrane Library. Two independent reviewers conducted the data screening, data extraction and risk of bias assessment. Qualitative synthesis and meta-analyses were conducted to summarise the key findings. RESULTS We included 21 articles involving 3835 participants in this review. Studies varied according to the cancer population and time along the cancer care continuum it was assessed. Of the studies that reported the overall contraceptive prevalence among women diagnosed with cancer (n = 8), contraceptive use ranged from 25 to 92%. Of the four studies that focused on cancer survivors, the contraceptive prevalence ranged from 47 to 84%. When the prevalence of these studies was pooled, a crude summary prevalence of 64% (62% among women with cancer versus 68% among cancer survivors) was found. The rate of contraceptive counselling was assessed in ten studies. A pooled prevalence of 50% (44% among women with cancer versus 58% among cancer survivors) was found, with the prevalence ranging from 12 to 78% among individual studies depending on the point in the cancer care continuum that it was provided. When contraceptive counselling was provided, it was found to significantly increase contraceptive use although biases were identified in its application. CONCLUSIONS Contraceptive counselling interventions as part of standard cancer care have the potential to not only empower women with cancer and cancer survivors to make informed choices regarding their reproductive health but also provide the ability to plan future pregnancies for times of better health.
Collapse
Affiliation(s)
- Melissa L Harris
- Centre for Women's Health Research, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia.
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia.
| | - Tesfaye R Feyissa
- Centre for Women's Health Research, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Medicine, Faculty of Health, Deakin University, Geelong, Australia
| | - Nikola A Bowden
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- Centre for Drug Repurposing and Medicines Research, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
| | - Kristina Gemzell-Danielsson
- Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Deborah Loxton
- Centre for Women's Health Research, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
32
|
Raimondo D, Raspollini A, Vicenti R, Renzulli F, Magnani V, Franceschini C, Raffone A, Mollo A, Casadio P, Seracchioli R. The use of near-infrared imaging with indocyanine green in the ovarian tissue transplantation: a case report. Facts Views Vis Obgyn 2022; 14:353-356. [PMID: 36724430 PMCID: PMC10364327 DOI: 10.52054/fvvo.14.4.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The request for fertility preservation has consistently increased in recent years. To our knowledge this case report is the first to describe the application of near-infrared intraoperative imaging using indocyanine green (NIR-ICG) during ovarian tissue transplantation (OTT), to assist surgeon choosing the site of implantation of ovarian fragments. OTT was performed in a 42-year-old woman using NIR-ICG to evaluate the vascularisation of peritoneal area as the site of implantation for the ovarian graft. we believe this new approach could be useful in identifying the best reimplantation site.
Collapse
|
33
|
Cherven B, Kelling E, Lewis RW, Pruett M, Meacham L, Klosky JL. Fertility-related worry among emerging adult cancer survivors. J Assist Reprod Genet 2022; 39:2857-2864. [PMID: 36447078 PMCID: PMC9790831 DOI: 10.1007/s10815-022-02663-1] [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: 07/12/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
PURPOSE Cancer survivors with a history of gonadotoxic treatment are at risk for future infertility and reproductive concerns, including worry about infertility. The purpose of this study was to describe factors associated with fertility-related worry among emerging adult survivors of childhood cancer. METHODS This chart review included patients aged 18.00-25.99 years and > 1 year from cancer treatment completion with a history of gonadotoxic treatment. Survivors were offered structured fertility-focused discussions at age ≥ 18 years, which assessed worry about future infertility. Data from this discussion (i.e., reported fertility-related worry (yes/no), sociodemographic, and clinical characteristics were abstracted from the medical record. Multivariable logistic regression with backwards elimination was used to calculate odds ratios (OR) and 95% confidence intervals (95%CI) for factors associated with fertility-related worry. RESULTS Survivors (N = 249) were a mean age of 19.1 ± 1.2 years at initial fertility discussion; 55.8% were male, 58.2% non-Hispanic White, and 27.3% were at high risk for future treatment-related infertility. Fertility-related worry was reported by 66.3% of survivors. Factors related to worry on multivariable analysis included female sex (OR: 2.64, 95%CI: 1.44-4.96, p = .002), solid tumor diagnosis (OR: 2.31, 95%CI: 1.15-4.71, p = .019), moderate and high risk of infertility (OR: 2.94, 95%CI: 1.23-7.64, p = .02; OR: 3.25, 95%CI: 1.55-7.17, p = .002), and ≥ 2 fertility discussions during survivorship care OR: 2.71, 95%CI: 1.46-5.20, p = .002). CONCLUSIONS Two-thirds of emerging adult cancer survivors expressed worry about future infertility, which has been linked to a variety of adverse quality of life outcomes. Survivors who are worried about infertility may benefit from psychological interventions.
Collapse
Affiliation(s)
- Brooke Cherven
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, 2015 Uppergate Dr, 4th Fl, Atlanta, GA, 30322, USA.
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| | - Erin Kelling
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, 2015 Uppergate Dr, 4th Fl, Atlanta, GA, 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca Williamson Lewis
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, 2015 Uppergate Dr, 4th Fl, Atlanta, GA, 30322, USA
| | - Megan Pruett
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, 2015 Uppergate Dr, 4th Fl, Atlanta, GA, 30322, USA
| | - Lillian Meacham
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, 2015 Uppergate Dr, 4th Fl, Atlanta, GA, 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - James L Klosky
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, 2015 Uppergate Dr, 4th Fl, Atlanta, GA, 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
34
|
Li Y, Cai X, Dong B, Wang Q, Yang X, Yu A, Wei H, Ke Z, Sun P, Zheng B, Sun Y. The Impact of Malignancy on Assisted Reproductive Outcomes for Cancer Survivors: A Retrospective Case–Control Study. Front Oncol 2022; 12:941797. [PMID: 36185197 PMCID: PMC9523265 DOI: 10.3389/fonc.2022.941797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundRelated studies have shown that it is safe for cancer patients to undergo assisted reproduction. However, studies on whether a history of cancer affects long-term reproductive outcomes in women who undergo assisted reproductive technology (ART) are scarce. In this study, we evaluated the long-term reproductive outcomes of patients with malignant tumors undergoing ART treatment and explored the impact of malignancy history on ART outcomes.MethodsThis retrospective study analyzed the clinical outcomes of patients with malignant tumors undergoing their first in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) cycles compared with those of age-matched healthy infertile women at Fujian Maternity and Child Health Hospital between January 2003 and October 2020. We evaluated ovarian stimulation outcome, the pregnancy rate, the live birth rate, the risk of adverse obstetric outcomes and birth outcomes.ResultsThis study included 59 patients in the cancer group for data analysis who had a history of malignancy. By matching, a total of 118 healthy infertile women were included in the control group. No statistically significant association was found in terms of age, duration of infertility, BMI, or insemination type between the two groups of patients. Thyroid cancer(45.8%) and gynecologic malignancies (44.07%) were the major cancer types in this study. There were statistically significant differences in the antral follicle count (AFC) (12.00 ± 7.86 vs. 14.90 ± 8.71, P=0.033), length of ovarian stimulation (9.98 ± 2.68 vs. 11.42 ± 2.43, P=0.033) and endometrial thickness on the trigger day (10.16 ± 3.11 vs. 10.84 ± 2.17, P<0.001) between the two groups. The total gonadotropin dose, number of oocytes retrieved, fertilization rate, cleavage rate, high-quality embryo rate, blastocyst rate and first-time embryo-transfer (ET) implantation rate were nonsignificantly lower in the cancer group than in the control group (P>0.05). There were no significant differences in the clinical pregnancy rate per ET cycle (32% vs. 40.39%, P=0.156), live birth rate per ET cycle (27% vs. 35.96%, P=0.119), miscarriage rate per ET cycle (5% vs. 4.43%, P=0.779), or preterm delivery rate per ET cycle (11.11% vs. 17.80%, P=0.547) between the two groups. Additionally, regression analysis showed that a history of malignancy was not a risk factor for reproductive outcomes.ConclusionsOverall, it is feasible for women with a history of cancer to conceive using ART is feasible and their long-term reproductive outcomes are similar to these of healthy infertile women. A history of cancer does not decrease the number of retrieved oocytes, increase the risk of adverse obstetric outcomes or affect birth outcomes.
Collapse
Affiliation(s)
- Yuehong Li
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Xuefen Cai
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Binhua Dong
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Qi Wang
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Xiaohui Yang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Aili Yu
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Huijuan Wei
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Zhanghong Ke
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Pengming Sun, ; Beihong Zheng, ; Yan Sun,
| | - Beihong Zheng
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Pengming Sun, ; Beihong Zheng, ; Yan Sun,
| | - Yan Sun
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Pengming Sun, ; Beihong Zheng, ; Yan Sun,
| |
Collapse
|
35
|
Rallis KS, George AM, Wozniak AM, Bigogno CM, Chow B, Hanrahan JG, Sideris M. Molecular Genetics and Targeted Therapies for Paediatric High-grade Glioma. Cancer Genomics Proteomics 2022; 19:390-414. [PMID: 35732328 PMCID: PMC9247880 DOI: 10.21873/cgp.20328] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 04/30/2022] [Accepted: 05/09/2022] [Indexed: 11/10/2022] Open
Abstract
Brain tumours are the leading cause of paediatric cancer-associated death worldwide. High-grade glioma (HGG) represents a main cause of paediatric brain tumours and is associated with poor prognosis despite surgical and chemoradiotherapeutic advances. The molecular genetics of paediatric HGG (pHGG) are distinct from those in adults, and therefore, adult clinical trial data cannot be extrapolated to children. Compared to adult HGG, pHGG is characterised by more frequent mutations in PDGFRA, TP53 and recurrent K27M and G34R/V mutations on histone H3. Ongoing trials are investigating novel targeted therapies in pHGG. Promising results have been achieved with BRAF/MEK and PI3K/mTOR inhibitors. Combination of PI3K/mTOR, EGFR, CDK4/6, and HDAC inhibitors are potentially viable options. Inhibitors targeting the UPS proteosome, ADAM10/17, IDO, and XPO1 are more novel and are being investigated in early-phase trials. Despite preclinical and clinical trials holding promise for the discovery of effective pHGG treatments, several issues persist. Inadequate blood-brain barrier penetration, unfavourable pharmacokinetics, dose-limiting toxicities, long-term adverse effects in the developing child, and short-lived duration of response due to relapse and resistance highlight the need for further improvement. Future pHGG management will largely depend on selecting combination therapies which work synergistically based on a sound knowledge of the underlying molecular target pathways. A systematic investigation of multimodal therapy with chemoradiotherapy, surgery, target agents and immunotherapy is paramount. This review provides a comprehensive overview of pHGG focusing on molecular genetics and novel targeted therapies. The diagnostics, genetic discrepancies with adults and their clinical implications, as well as conventional treatment approaches are discussed.
Collapse
Affiliation(s)
- Kathrine S Rallis
- Barts Cancer Institute, Queen Mary University of London, London, U.K.;
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Alan Mathew George
- Liverpool School of Medicine, University of Liverpool, Liverpool, U.K
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - Anna Maria Wozniak
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Carola Maria Bigogno
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Barbara Chow
- UCL Cancer Institute, University College London, London, U.K
- GKT School of Medicine, King's College London, London, U.K
| | | | - Michail Sideris
- Women's Health Research Unit, Queen Mary University of London, London, U.K
| |
Collapse
|
36
|
Effects of Chemotherapy on Fertility Preservation in Patients with Tumors of the Hematopoietic and Lymphoid Tissues. REPRODUCTIVE MEDICINE 2022. [DOI: 10.3390/reprodmed3020012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Fertility preservation is an important concern for young cancer patients. Oocyte or embryo cryopreservation prior to chemotherapy administration is desirable but often difficult for patients with hematopoietic and lymphoid tissue tumors. In this study, we examined the results of fertility preservation therapy in patients with hematopoietic and lymphoid tissue tumors. We retrospectively examined hematopoietic and lymphoid tissue tumors of five patients who underwent oocyte cryopreservation as a fertility preservation therapy after chemotherapy, at Showa University Hospital from February 2017 to September 2020. Eleven treatment cycles were administered (one of which was cancelled). The mean age of the patients was 28.6 years. The mean controlled ovarian stimulation duration for 10 cycles was 15.9 days, the mean total gonadotropin dose was 3705 IU, and the mean peak E2 was 502.8 (pg/mL). The mean number of eggs retrieved was 3.2, the mean number of mature oocytes was 2.1, and the mean maturation rate (mature oocytes/returned oocytes) was 70.7%. Fertility preservation procedures in the early period after chemotherapy may be viable because they allow for the acquisition of mature oocytes, even though the procedures may take longer and yield fewer oocytes.
Collapse
|
37
|
Dargan C, Mc Dermott S, Chesbro S, Trout A, Terwilliger N, Hilyard T, Flynn A, Fulbright J. Standardization of Fertility Preservation Discussion Among Pediatric Oncology and Bone Marrow Transplant Patients: A Single Institution Experience. J Adolesc Young Adult Oncol 2022; 12:259-265. [PMID: 35675684 DOI: 10.1089/jayao.2022.0003] [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: 11/13/2022] Open
Abstract
Purpose: Infertility is an impactful late effect of cancer therapy. Options for fertility preservation exist, however, barriers remain. Within our division, we lacked a standard approach to discussing fertility preservation. Methods: During the time period of 2014-2020, a fertility preservation program was developed with program improvements implemented over time and provider comfort with fertility identified and addressed through educational intervention. To evaluate how our improvements affected frequency of documented reproductive health discussions, 474 pubertal pediatric patients with new oncological diagnoses were reviewed. Descriptive analysis of sociodemographic determinants was performed. Results: One hundred seventy-five patients met inclusion criteria. Racial/ethnic composition was similar in those receiving and not receiving a fertility consult. Although 19.3% of Caucasians pursued fertility preservation, none of the eight African Americans or five Hispanic females did. Division feedback identified a lack of knowledge regarding available fertility preservation options and diagnoses that should prompt this conversation as barriers to the consulting fertility preservation team. Pre- and posteducation assessments demonstrated increased comfort in discussing fertility preservation and knowledge regarding diagnoses at higher risk of infertility. Integration of a standardized fertility preservation process and addressing barriers identified led to a 33.6% increase in fertility discussions. Conclusion: The establishment of a fertility preservation process and team and division-wide education has led to improvement in rates of fertility discussion in pediatric and adolescent and young adult cancer patients. Similar to young adult data, our data suggest that some racial health disparities may exist in the utilization of fertility preservation in the pediatric oncology population.
Collapse
Affiliation(s)
- Chandni Dargan
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplant, Children's Mercy Kansas City, Missouri, USA
| | - Sarah Mc Dermott
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplant, Children's Mercy Kansas City, Missouri, USA
| | - Shelby Chesbro
- Department of Pediatrics, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Amanda Trout
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplant, Children's Mercy Kansas City, Missouri, USA
| | - Nancy Terwilliger
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplant, Children's Mercy Kansas City, Missouri, USA
| | - Tennille Hilyard
- Division of Pediatric and Adolescent Gynecology, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Ashley Flynn
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplant, Children's Mercy Kansas City, Missouri, USA
| | - Joy Fulbright
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplant, Children's Mercy Kansas City, Missouri, USA
| |
Collapse
|
38
|
Di Tucci C, Galati G, Mattei G, Chinè A, Fracassi A, Muzii L. Fertility after Cancer: Risks and Successes. Cancers (Basel) 2022; 14:2500. [PMID: 35626104 PMCID: PMC9139810 DOI: 10.3390/cancers14102500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 12/22/2022] Open
Abstract
The incidence of cancer in reproductive-aged women is 7%, but, despite the increased number of cancer cases, advances in early diagnosis and treatment have raised the survival rate. Furthermore, in the last four decades, there has been a rising trend of delaying childbearing. There has been an increasing number of couples referred to Reproductive Medicine Centers for infertility problems after one partner has been treated for cancer. In these cases, the main cause of reduced fertility derives from treatments. In this review, we describe the effects and the risks of chemotherapy, radiotherapy, and surgery in women with cancer, and we will focus on available fertility preservation techniques and their efficacy in terms of success in pregnancy and live birth rates.
Collapse
Affiliation(s)
- Chiara Di Tucci
- Department of Obstetrics and Gynecology, “Sapienza” University, 00185 Rome, Italy; (G.G.); (G.M.); (A.C.); (A.F.); (L.M.)
| | | | | | | | | | | |
Collapse
|
39
|
Cioffi R, Fais ML, Bergamini A, Vanni VS, Pagliardini L, Papaleo E, Mangili G, Candiani M. Ovarian failure risk in post-pubertal patients with cancer: a prognostic model. Future Oncol 2022; 18:2391-2400. [PMID: 35469452 DOI: 10.2217/fon-2022-0078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To develop a predictive model for ovarian failure (OF) after chemotherapy in young post-pubertal women with cancer. Methods: Retrospective, monocentric cohort study including 348 patients referring to the Oncofertility Unit of San Raffaele Hospital (Milan, Italy) from August 2011 to January 2020. A predictive model was constructed by multivariate logistic regression and receiver operating characteristic analysis. Results: Data about menstrual function resumption were available for 184 patients. The best predictive model for OF was identified by the combination of age; number of chemotherapy lines; vincristine, adriamycin, ifosphamide/adriamycin, ifosphamide; capecitabine; adriamycin, bleomycine, vinblastine, doxorubicin (area under the curve= 0.906, CI 95% 0.858-0.954, p = 0.0001). Conclusions: The model predicts the probability of loss of ovarian function at cancer diagnosis and with every change of treatment.
Collapse
Affiliation(s)
- Raffaella Cioffi
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Maria Luisa Fais
- Obstetrics & Gynecology Unit, University of Cagliari, Cagliari, 09124, Italy
| | - Alice Bergamini
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Valeria Stella Vanni
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Luca Pagliardini
- Division of Genetics & Cell Biology, Reproductive Sciences Laboratory, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Enrico Papaleo
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Giorgia Mangili
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Massimo Candiani
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| |
Collapse
|
40
|
Unlaid Eggs: Ovarian Damage after Low-Dose Radiation. Cells 2022; 11:cells11071219. [PMID: 35406783 PMCID: PMC8997758 DOI: 10.3390/cells11071219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/24/2022] [Accepted: 04/02/2022] [Indexed: 11/17/2022] Open
Abstract
The total body irradiation of lymphomas and co-irradiation in the treatment of adjacent solid tumors can lead to a reduced ovarian function, premature ovarian insufficiency, and menopause. A small number of studies has assessed the radiation-induced damage of primordial follicles in animal models and humans. Studies are emerging that evaluate radiation-induced damage to the surrounding ovarian tissue including stromal and immune cells. We reviewed basic laboratory work to assess the current state of knowledge and to establish an experimental setting for further studies in animals and humans. The experimental approaches were mostly performed using mouse models. Most studies relied on single doses as high as 1 Gy, which is considered to cause severe damage to the ovary. Changes in the ovarian reserve were related to the primordial follicle count, providing reproducible evidence that radiation with 1 Gy leads to a significant depletion. Radiation with 0.1 Gy mostly did not show an effect on the primordial follicles. Fewer data exist on the effects of radiation on the ovarian microenvironment including theca-interstitial, immune, endothelial, and smooth muscle cells. We concluded that a mouse model would provide the most reliable model to study the effects of low-dose radiation. Furthermore, both immunohistochemistry and fluorescence-activated cell sorting (FACS) analyses were valuable to analyze not only the germ cells but also the ovarian microenvironment.
Collapse
|
41
|
Voigt P, Persily J, Blakemore JK, Licciardi F, Thakker S, Najari B. Sociodemographic differences in utilization of fertility services among reproductive age women diagnosed with cancer in the USA. J Assist Reprod Genet 2022; 39:963-972. [PMID: 35316438 PMCID: PMC9051007 DOI: 10.1007/s10815-022-02455-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/04/2022] [Indexed: 10/18/2022] Open
Abstract
PURPOSE To determine whether sociodemographic differences exist among female patients accessing fertility services post-cancer diagnosis in a representative sample of the United States population. METHODS All women ages 15-45 with a history of cancer who responded to the National Survey for Family Growth (NSFG) from 2011 to 2017 were included. The population was then stratified into 2 groups, defined as those who did and did not seek infertility services. The demographic characteristics of age, legal marital status, education, race, religion, insurance status, access to healthcare, and self-perceived health were compared between the two groups. The primary outcome measure was the utilization of fertility services. The complex sample analysis using the provided sample weights required by the NSFG survey design was used. RESULTS Five hundred forty-five women reported a history of cancer and were included in this study. Forty-three (7.89%) pursued fertility services after their cancer diagnosis. Using the NSFG sample weights, this equates to a population of 161,500.7 female cancer survivors in the USA who did utilize fertility services and 1,811,955.3 women who did not. Using multivariable analysis, household income, marital status, and race were significantly associated with women utilizing fertility services following a cancer diagnosis. CONCLUSIONS In this nationally representative cohort of reproductive age women diagnosed with cancer, there are marital, socioeconomic, and racial differences between those who utilized fertility services and those who did not. This difference did not appear to be due to insurance coverage, access to healthcare, or perceived health status.
Collapse
Affiliation(s)
- Paxton Voigt
- NYU Grossman School of Medicine, NY, New York, USA.
| | - Jesse Persily
- NYU Langone Department of Urology, New York, NY, USA
| | | | | | | | - Bobby Najari
- NYU Langone Department of Urology, New York, NY, USA
| |
Collapse
|
42
|
Hessels AC, Langendijk JA, Gawryszuk A, A.A.M. Heersters M, van der Salm NL, Tissing WJ, van der Weide HL, Maduro JH. Review – late toxicity of abdominal and pelvic radiotherapy for childhood cancer. Radiother Oncol 2022; 170:27-36. [DOI: 10.1016/j.radonc.2022.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 11/15/2022]
|
43
|
Bottini AL, Reis VMSFD, Capp E, Silva ISBD, Kliemann LM, Corleta HVE. Ovarian Tissue Culture to Preserve Fertility in Transgender Male Patients after Hormonal Treatment. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRICIA : REVISTA DA FEDERACAO BRASILEIRA DAS SOCIEDADES DE GINECOLOGIA E OBSTETRICIA 2022; 44:251-257. [PMID: 35139567 PMCID: PMC9948281 DOI: 10.1055/s-0042-1742410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To evaluate the reproductive and histological characteristics of fresh cultured ovarian tissue from transgender male patients. METHODS An in vitro pilot study in which samples were collected during sex reassignment surgery for transgender male patients. The ovarian cortex was cut into fragments of 2 mm, 3 mm, and 4 mm, and placed in a 96-well plate suitable for cultivation at days 0, 2, 4, 6, and 8, when the histology was analyzed. RESULTS Stromal hyperplasia was observed in all samples, and it was not associated with the obtainment of primordial or primary follicles. Peripheral reduction in cell count was also a recurrent finding. Primordial and primary follicles were identified with a heterogeneous pattern in fragments from the same patient and from different patients, and follicles in more advanced stages of development (secondary and antral) were not found. There was an association between the diameter of the ovarian fragments and the identification of primary follicles (p = 0.036). The number of days in culture was associated with histological signs of tissue damaging in the fragments (p = 0.002). The total number of follicles identified in the samples with 2 mm in diameter was significantly lower than in those that measured 4 mm in diameter (p = 0.031). CONCLUSION A diameter of 4 mm is suitable for ovarian tissue culture with the benefit of ease of handling. Even after prolonged exposure to testosterone, the ovarian fragments presented primordial and primary follicles, maintaining viability throughout the days they were exposed to the culture. Freezing the ovarian cortex of transgender patients who will undergo surgery for gender reassignment would be an interesting option, in the future, for the preservation of fertility.
Collapse
Affiliation(s)
- Alessandra Leal Bottini
- Programa de Pós-Graduação em Ginecologia e Obstetrícia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Edison Capp
- Programa de Pós-Graduação em Ginecologia e Obstetrícia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Gynecology and Obstetrics, School of Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre/RS, Brazil
| | - Ilma Simoni Brum da Silva
- Programa de Pós-Graduação em Ginecologia e Obstetrícia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lúcia Maria Kliemann
- Programa de Pós-Graduação em Ginecologia e Obstetrícia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Gynecology and Obstetrics, School of Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre/RS, Brazil
| | - Helena von Eye Corleta
- Programa de Pós-Graduação em Ginecologia e Obstetrícia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Gynecology and Obstetrics, School of Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre/RS, Brazil.,Generar, Porto Alegre, Brazil
| |
Collapse
|
44
|
Farland LV, Stern JE, Hwang SS, Liu CL, Cabral H, Knowlton R, Gershman ST, Coddington CC, Missmer SA. History of cancer and fertility treatment outcomes: a registry linkage study in Massachusetts. J Assist Reprod Genet 2022; 39:517-526. [PMID: 35037166 PMCID: PMC8956755 DOI: 10.1007/s10815-021-02376-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 12/06/2021] [Indexed: 02/03/2023] Open
Abstract
PURPOSE To investigate assisted reproductive technology (ART) outcomes among adolescent and young-adult female cancer survivors. METHODS The Society for Assisted Reproductive Technology Clinic Outcome Reporting System (SART CORS) data were linked to the Massachusetts Cancer Registry for 90,928 ART cycles in Massachusetts to women ≥ 18 years old from 2004 to 2013. To estimate relative risks (RR) and 95% confidence intervals (CI), we used generalized estimating equations with a log link that accounted for multiple cycles per woman and a priori adjusted for maternal age and cycle year. The main outcomes of interest were ART treatment patterns; number of autologous oocytes retrieved, fertilized, and transferred; and rates of implantation, clinical intrauterine gestation (CIG), live birth, and pregnancy loss. RESULTS We saw no difference in number of oocytes retrieved (aRR: 0.95 (0.89-1.02)) or proportion of autologous oocytes fertilized (aRR: 0.99 (0.95-1.03)) between autologous cycles with and without a history of cancer; however, cancer survivors required a higher total FSH administered (aRR: 1.12 (1.06-1.19)). Among autologous cycle starts, cycles in women with a history of cancer were less likely to result in CIG compared to no history of cancer (aRR: 0.73 (0.65-0.83)); this relationship was absent from donor cycles (aRR: 1.01 (0.85-1.20)). Once achieving CIG, donor cycles for women with a history of cancer were two times more likely to result in pregnancy loss (aRR: 1.99 (1.26-3.16)). CONCLUSIONS Our analysis suggests that cancer may influence ovarian stimulation response, requiring more FSH and resulting in lower CIG among cycle starts.
Collapse
Affiliation(s)
- Leslie V Farland
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA.
| | - Judy E Stern
- Department of Obstetrics and Gynecology, Dartmouth-Hitchcock, Lebanon, NH, USA
| | - Sunah S Hwang
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Chia-Ling Liu
- Massachusetts Department of Public Health, Bureau of Family Health and Nutrition, Boston, MA, USA
| | - Howard Cabral
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Richard Knowlton
- Massachusetts Cancer Registry, Office of Data Management and Outcomes Assessment, Office of Population Health, Massachusetts Department of Public Health, Boston, MA, USA
| | - Susan T Gershman
- Massachusetts Cancer Registry, Office of Data Management and Outcomes Assessment, Office of Population Health, Massachusetts Department of Public Health, Boston, MA, USA
| | | | - Stacey A Missmer
- Department of Obstetrics and Gynecology, Carolinas Medical Center/Atrium Health, Charlotte, NC, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
45
|
Anderson RA, Kelsey TW, Morrison DS, Wallace WHB. Family size and duration of fertility in female cancer survivors: a population-based analysis. Fertil Steril 2022; 117:387-395. [PMID: 34933761 PMCID: PMC8865032 DOI: 10.1016/j.fertnstert.2021.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To assess family size and timescale for achieving pregnancy in women who remain fertile after cancer. DESIGN Population-based analysis. SETTING National databases. PATIENT(S) All women diagnosed with cancer before the age of 40 years in Scotland, 1981-2012 (n = 10,267) with no previous pregnancy; each was matched with 3 population controls. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) The number and timing of pregnancy and live birth after cancer diagnosis, to 2018. RESULT(S) In 10,267 cancer survivors, the hazard ratio for a subsequent live birth was 0.56 (95% confidence interval, 0.53-0.58) overall. In women who achieved a subsequent pregnancy, age at live birth increased (mean ± SD, 31.2 ± 5.5 vs. 29.7 ± 6.1 in controls), and the family size was lower (2.0 ± 0.8 vs. 2.3 ± 1.1 live births). These findings were consistent across several diagnoses. The interval from diagnosis to last pregnancy was similar to that of controls (10.7 ± 6.4 vs. 10.9 ± 7.3 years) or significantly increased, for example, after breast cancer (6.2 ± 2.8 vs. 5.3 ± 3.3 years) and Hodgkin lymphoma (11.1 ± 5.1 vs. 10.1 ± 5.8 years). CONCLUSION(S) These data quantify the reduced chance of live birth after cancer. Women who subsequently conceived achieved a smaller family size than matched controls, but the period of time after cancer diagnosis across which pregnancies occurred was similar or, indeed, increased. Thus, we did not find evidence that women who were able to achieve a pregnancy after cancer had a shorter timescale over which they have pregnancies.
Collapse
Affiliation(s)
- Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | - Tom W Kelsey
- School of Computer Science, University of St Andrews, St Andrews, United Kingdom
| | | | - W Hamish B Wallace
- Department of Hematology and Oncology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| |
Collapse
|
46
|
Kaneva K, Erickson L, Rowell E, Badawy SM. Fertility preservation education for pediatric hematology-oncology fellows, faculty and advanced practice providers: a pilot study. Pediatr Hematol Oncol 2022; 39:68-73. [PMID: 34028331 DOI: 10.1080/08880018.2021.1928348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Infertility secondary to chemotherapy, myeloablative conditioning regimens prior to stem cell transplantation, radiation therapy, and/or surgery is an important cause of morbidity and psychosocial distress among pediatric cancer patients. Known options exist for fertility preservation; however, knowledge among providers varies. We conducted a pilot study with an educational intervention over one-hour for hematology-oncology faculty, fellows, and advanced practice providers. Participants completed pre-/post-test assessment on fertility preservation knowledge. Participants' pretest mean (SD) score was 53% (17%), which significantly increased to 72% (11%) in the post-test (p = 0.0004). We demonstrated that a fertility education intervention could improve knowledge regarding infertility risk assessment and fertility preservation options.
Collapse
Affiliation(s)
- Kristiyana Kaneva
- Division of Hematology, Oncology, Neuro-Oncology & Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Laura Erickson
- Division of Pediatric Surgery and Fertility & Hormone Preservation & Restoration Program, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Erin Rowell
- Division of Pediatric Surgery and Fertility & Hormone Preservation & Restoration Program, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA.,Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sherif M Badawy
- Division of Hematology, Oncology, Neuro-Oncology & Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
47
|
Barnett S, Holden V, Campbell-Hewson Q, Veal GJ. Perspectives and Expertise in Establishing a Therapeutic Drug Monitoring Programme for Challenging Childhood Cancer Patient Populations. Front Oncol 2022; 11:815040. [PMID: 35071019 PMCID: PMC8770741 DOI: 10.3389/fonc.2021.815040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
The utility of Therapeutic Drug Monitoring (TDM) in the setting of childhood cancer is a largely underused tool, despite the common use of cytotoxic chemotherapeutics. While it is encouraging that modern advances in chemotherapy have transformed outcomes for children diagnosed with cancer, this has come at the cost of an elevated risk of life-changing long-term morbidity and late effects. This concern can limit the intensity at which these drugs are used. Widely used chemotherapeutics exhibit marked inter-patient variability in drug exposures following standard dosing, with fine margins between exposures resulting in toxicity and those resulting in potentially suboptimal efficacy, thereby fulfilling criteria widely accepted as fundamental for TDM approaches. Over the past decade in the UK, the paediatric oncology community has increasingly embraced the potential benefits of utilising TDM for particularly challenging patient groups, including infants, anephric patients and those receiving high dose chemotherapy. This has been driven by a desire from paediatric oncologists to have access to clinical pharmacology information to support dosing decisions being made. This provides the potential to modify doses between treatment cycles based on a comprehensive set of clinical information, with individual patient drug exposures being used alongside clinical response and tolerability data to inform dosing for subsequent cycles. The current article provides an overview of recent experiences of conducting TDM in a childhood cancer setting, from the perspectives of the clinicians, scientists and pharmacists implementing TDM-based dosing recommendations. The ongoing programme of work has facilitated investigations into the validity of current approaches to dosing for some of the most challenging childhood cancer patient groups, with TDM approaches now being expanded from well-established cytotoxic drugs through to newer targeted treatments.
Collapse
Affiliation(s)
- Shelby Barnett
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | - Gareth J Veal
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
48
|
Ozcan MCH, Snegovskikh V, Adamson GD. Oocyte and embryo cryopreservation before gonadotoxic treatments: Principles of safe ovarian stimulation, a systematic review. WOMEN'S HEALTH (LONDON, ENGLAND) 2022; 18:17455065221074886. [PMID: 35130799 PMCID: PMC8829712 DOI: 10.1177/17455065221074886] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/10/2021] [Accepted: 01/04/2022] [Indexed: 05/05/2023]
Abstract
OBJECTIVE Review the safety of fertility preservation through ovarian stimulation with oocyte or embryo cryopreservation, including cycle and medication options. EVIDENCE REVIEW A systematic review of peer-reviewed sources revealed 2 applicable randomized control trials and 60 cohort studies as well as 20 additional expert opinions or reviews. RESULTS The capacity for future family building is important for the majority of reproductive age people, despite life-altering medical or oncologic diagnosis. Modern fertility preservation generates a high rate of oocyte yield while utilizing protocols that can be started at multiple points in the menstrual cycle and suppressing supra-physiologic levels of estrogen. Finally, more than one quarter of fertility preservation patients will return to later utilize fertility services. CONCLUSION For most patients, fertility preservation can safely be pursued and completed within 2 weeks without affecting disease severity or long-term survival.
Collapse
Affiliation(s)
- Meghan CH Ozcan
- Department of Obstetrics and Gynecology, Warren Alpert Medical School of Brown University, Women & Infants Hospital, Providence, RI, USA
| | - Victoria Snegovskikh
- Department of Obstetrics and Gynecology, Warren Alpert Medical School of Brown University, Women & Infants Hospital, Providence, RI, USA
| | | |
Collapse
|
49
|
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: 1.5] [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]
|
50
|
Ruan X, Cheng J, Du J, Jin F, Gu M, Li Y, Ju R, Wu Y, Wang H, Yang W, Cheng H, Li L, Bai W, Kong W, Yang X, Lv S, Wang Y, Yang Y, Xu X, Jiang L, Li Y, Mueck AO. Analysis of Fertility Preservation by Ovarian Tissue Cryopreservation in Pediatric Children in China. Front Endocrinol (Lausanne) 2022; 13:930786. [PMID: 35846295 PMCID: PMC9277002 DOI: 10.3389/fendo.2022.930786] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Ovarian tissue cryopreservation (OTC) is the only method of fertility preservation (FP) in prepubertal girls, but the experience remains limited. This study investigates the effectiveness and feasibility of FP of OTC in children facing gonadotoxicity treatment in Chinese first ovarian tissue cryobank. PROCEDURE OTC and evaluation of 49 children ≤14 years old in the cryobank of Beijing Obstetrics and Gynecology Hospital, Capital Medical University, from July 2017 to May 19, 2022, were analyzed retrospectively. We compared children's general characteristics, follicle numbers, and hormone levels with and without chemotherapy before OTC. RESULTS The age of 49 children at the time of OTC was 7.55 (1-14) years old. There were 23 cases of hematological non-malignant diseases, eight cases of hematological malignant diseases, four cases of gynecological malignant tumors, one case of neurological malignant tumors, one case of bladder cancer, five cases of sarcoma, three cases of mucopolysaccharidosis, one case of metachromatic leukodystrophy, two cases of dermatomyositis, one case of Turner's syndrome. The median follicular count per 2-mm biopsy was 705. Age and AMH were not correlated (r = 0.084, P = 0.585). Age and follicle count per 2-mm biopsy was not correlated (r = -0.128, P = 0.403). Log10 (follicle count per 2-mm biopsy) and Log10 (AMH) were not correlated (r = -0.118, P = 0.456). Chemotherapy before OTC decreased AMH levels but had no significant effect on the number of follicles per 2-mm biopsy. CONCLUSIONS OTC is the only method to preserve the fertility of prepubertal girls, and it is safe and effective. Chemotherapy before OTC is not a contraindication to OTC.
Collapse
Affiliation(s)
- Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- *Correspondence: Xiangyan Ruan,
| | - Jiaojiao Cheng
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Juan Du
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fengyu Jin
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Muqing Gu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanglu Li
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Rui Ju
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yurui Wu
- Department of Thoracic Surgery and Surgical Oncology, Children’s Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Huanmin Wang
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Wei Yang
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Haiyan Cheng
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Long Li
- Department of Pediatric Surgery, Children’s Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Wenpei Bai
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Weimin Kong
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xin Yang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Shulan Lv
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yuejiao Wang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yu Yang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xin Xu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Lingling Jiang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanqiu Li
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Alfred O. Mueck
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department of Women’s Health, University of Tuebingen, University Women’s Hospital and Research Centre for Women’s Health, Tuebingen, Germany
| |
Collapse
|