1
|
Frederick NN, Klosky JL, Meacham L, Quinn GP, Kelvin JF, Cherven B, Freyer DR, Dvorak CC, Brackett J, Ahmed-Winston S, Bryson E, Su HI, Chow EJ, Levine J. Fertility Preservation Practices at Pediatric Oncology Institutions in the United States: A Report From the Children's Oncology Group. JCO Oncol Pract 2023; 19:e550-e558. [PMID: 36763922 PMCID: PMC10113112 DOI: 10.1200/op.22.00349] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/16/2022] [Accepted: 01/06/2023] [Indexed: 02/12/2023] Open
Abstract
PURPOSE Fertility discussions are an integral part of comprehensive care for pediatric, adolescent, and young adult patients newly diagnosed with cancer and are supported by national guidelines. Current institutional practices are poorly understood. METHODS A cross-sectional survey was distributed to 220 Children's Oncology Group member institutions regarding fertility discussion practices. Descriptive statistics were calculated for all variables. The association between specific practices and selected outcomes on the basis of sex was examined via multivariable logistic regression. RESULTS One hundred forty-four programs (65.5%) returned surveys. Of these, 65 (45.1%) reported routine discussions of fertility with all female patients and 55 (38.5%) all male patients (P = .25). Ninety-two (63.8%) reported no specific criteria for offering females fertility preservation (FP), compared with 40 (27.7%) for males (P < .001). Program characteristics associated with fertility discussions included reproductive endocrinology and infertility on site (females odds ratio [OR], 2.1; 95% CI, 1.0 to 4.3), discussion documentation mandate (females OR, 2.3; 95% CI, 1.0 to 5.5; males OR, 3.5; 95% CI, 1.4 to 8.7), and cumulative institution-based FP infrastructure (which included [1] routine practice of documentation, [2] template for documentation, [3] mandate for documentation, and [4] availability of FP navigation; females OR, 1.6; 95% CI, 1.1 to 2.3; males OR, 2.3; 95% CI, 1.6 to 3.4). Utilization of practices unsupported by guidelines included offering sperm banking after treatment initiation (39/135 programs; 28.9%), gonadotropin-releasing hormone analogs for ovarian suppression/FP (75/144 programs; 52.1%), ovarian tissue cryopreservation at diagnosis for patients with leukemia (19/64 programs; 29.7%), and testicular tissue cryopreservation (23/138 programs; 16.7%) not part of a clinical trial. CONCLUSION Despite recommended guidelines, fertility discussions with patients/families before treatment initiation are not routine at Children's Oncology Group institutions. Standard criteria to determine which options should be offered to patients are more common for males than females.
Collapse
Affiliation(s)
- Natasha N. Frederick
- Center for Cancer and Blood Disorders, Connecticut Children's Medical Center, Hartford, CT
- University of Connecticut School of Medicine, Farmington, CT
| | - James L. Klosky
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, Atlanta, GA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Lillian Meacham
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, Atlanta, GA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Gwendolyn P. Quinn
- Division of Medical Ethics, Departments of OB-GYN, Population Health, Grossman School of Medicine, New York University, New York, NY
| | | | - Brooke Cherven
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, Atlanta, GA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - David R. Freyer
- Cancer and Blood Disease Institute, Children's Hospital, Los Angeles, CA
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Christopher C. Dvorak
- Division of Pediatric Allergy, Immunology and Bone Marrow Transplant, University of California San Francisco, San Francisco, CA
| | - Julienne Brackett
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Cancer and Hematology Centers, Houston, TX
| | | | - Elyse Bryson
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta, Atlanta, GA
| | - H. Irene Su
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, CA
| | - Eric J. Chow
- Clinical Research and Public Health Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jennifer Levine
- Division of Pediatric Hematology and Oncology, Weill Cornell Medicine, New York, NY
| |
Collapse
|
2
|
Meisel JL, Harrington A, Whaley N, Vitek W, O'Regan RM. When the personal becomes political: The impact of the Dobbs decision on women with cancer. Cancer 2023; 129:171-174. [PMID: 36345655 DOI: 10.1002/cncr.34522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Jane Lowe Meisel
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Amy Harrington
- Department of Obstetrics and Gynecology, University of Rochester, Rochester, New York, USA
| | - Natalie Whaley
- Department of Obstetrics and Gynecology, University of Rochester, Rochester, New York, USA
| | - Wendy Vitek
- Department of Obstetrics and Gynecology, University of Rochester, Rochester, New York, USA
| | - Ruth M O'Regan
- Department of Medicine, University of Rochester, Rochester, New York, USA.,Wilmot Cancer Institute, University of Rochester, Rochester, New York, USA
| |
Collapse
|
3
|
Mechanistic insights into HuR inhibitor MS-444 arresting embryonic development revealed by low-input RNA-seq and STORM. Cell Biol Toxicol 2022; 38:1175-1197. [PMID: 36085230 DOI: 10.1007/s10565-022-09757-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/26/2022] [Indexed: 01/25/2023]
Abstract
With improvements in the survival rate of patients with cancer, fertility maintenance has become a major concern in terms of cancer treatment for women of reproductive age. Thus, it is important to examine the impact on fertility of anticancer drugs that are used clinically or are undergoing trials. The HuR small-molecule inhibitor MS-444 has been used in many cancer treatment studies, but its reproductive toxicity in females is unknown. Here, we reported that MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA by inhibiting HuR dimerization, resulting in the developmental arrest of 2-cell stage embryos in mouse. Combining analysis of low-input RNA-seq for MS-444-treated 2-cell embryos and mapping binding sites of RNA-binding protein, Agbl2 was predicted to be the target gene of MS-444. For further confirmation, RNAi experiment in wild-type zygotes showed that Agbl2 knockdown reduced the proportion of embryos successfully developed to the blastocyst stage: from 71% in controls to 23%. Furthermore, RNA-FISH and luciferase reporter analyses showed that MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA and reduced its stability by inhibiting HuR dimerization. In addition, optimized stochastic optical reconstruction microscopy (STORM) imaging showed that MS-444 significantly reduced the HuR dimerization, and HuR mainly existed in cluster form in 2-cell stage embryos. In conclusion, this study provides clinical guidance for maintaining fertility during the treatment of cancer with MS-444 in women of reproductive age. And also, our research provides guidance for the application of STORM in nanometer scale studies of embryonic cells. HuR inhibitor MS-444 arrested embryonic development at 2-cell stage. Low-input RNA-seq revealed that Agbl2 was the target gene of MS-444. MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA by inhibiting HuR dimerization and reduced the stability of Agbl2 mRNA. STORM with our optimized protocol showed that HuR tended to form elliptical and dense clusters in 2-cell stage embryos.
Collapse
|
4
|
Xie Y, Duan H, Wang D, Li H, Jia J, Zhang J, Li L. Gonadotropin-releasing hormone agonist protects ovarian function in young patients with ovarian malignancy undergoing platinum-based chemotherapy: A prospective study. Front Oncol 2022; 12:986208. [DOI: 10.3389/fonc.2022.986208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeWe aimed to ascertain the effectiveness of gonadotropin-releasing hormone (GnRH) agonist co-therapy for the preservation of ovarian function in patients with ovarian malignancy who underwent unilateral salpingo-oophorectomy and platinum-based chemotherapy.MethodsWe enrolled 158 patients with ovarian malignancy who underwent fertility preservation surgery and postoperative platinum-based chemotherapy between January 2018 and December 2020. Patients were divided into two groups based on the use of GnRH agonist (GnRHa) during chemotherapy. Two patients withdrew from the study. Laboratory tests (serum follicle-stimulating hormone [FSH], serum luteinizing hormone [LH], and serum anti-Müllerian hormone [AMH]) were performed pre-chemotherapy and one year post-chemotherapy. Data on menstruation resumption, perimenopausal symptoms (modified Kupperman Menopausal Index [KMI]), health-related quality of life (Medical Outcomes Study Short Form-36 [MOS SF-36]), and obstetric outcomes were collected.ResultsOne year post-chemotherapy, the serum AMH level in the GnRHa group was higher than that in the control group (P<0.001), while the serum FSH and FSH/LH levels in the GnRHa group were lower than those in the control group (P<0.001). The mean period from last chemotherapy to menstrual resumption was 3.86 and 5.78 months in the GnRHa and control groups (P<0.001), respectively. The rate of menstrual resumption post-chemotherapy was 93.5% and 82.3% in the GnRHa and control groups (P<0.05), respectively. GnRHa co-administration during chemotherapy reduced the likelihood of low AMH levels post-chemotherapy and was significant in the multivariate analysis (P<0.05). The modified KMI scores and MOS SF-36 scores were better in the GnRHa group than in the control group (both P<0.001).ConclusionGnRHa protects ovarian function during platinum-based adjuvant chemotherapy in young patients with ovarian malignancy. This study provides a therapeutic reference for gynecologists, especially for those in economically and medically underdeveloped areas.Trial registrationChinese Clinical Trial Registry (chiCTR1800019114; October 26, 2018; http://www.chictr.org.cn/index.aspx)
Collapse
|
5
|
Senotherapy Protects against Cisplatin-Induced Ovarian Injury by Removing Senescent Cells and Alleviating DNA Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9144644. [PMID: 35693700 PMCID: PMC9187433 DOI: 10.1155/2022/9144644] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
Abstract
Ovarian damage induced by platinum-based chemotherapy seriously affects young women with cancer, manifesting as infertility, early menopause, and premature ovarian insufficiency. However, effective prevention strategies for such damage are lacking. Senescent cells may be induced by chemotherapeutic agents. We hypothesized that cisplatin can lead to senescence in ovarian cells during the therapeutic process, and senolytic drugs can protect animals against cisplatin-induced ovarian injury. Here, we demonstrated the existence of senescent cells in cisplatin-treated ovaries, identified the senescence-associated secretory phenotype, and observed significant improvement of ovarian function by treatment with metformin or dasatinib and quercetin (DQ) independently or in combination. These senotherapies improved both oocyte quality and fertility, increased the ovarian reserve, and enhanced hormone secretion in cisplatin-exposed mice. Additionally, attenuated fibrosis, reorganized subcellular structure, and mitigated DNA damage were observed in the ovaries of senotherapeutic mice. Moreover, RNA sequencing analysis revealed upregulation of the proliferation-related genes Ki, Prrx2, Sfrp4, and Megfl0; and the antioxidative gene H2-Q10 after metformin plus DQ treatment. Gene ontology analysis further revealed that combining senotherapies enhanced ovarian cell differentiation, development, and communication. In this study, we demonstrated that metformin plus DQ recovered ovarian function to a greater extent compared to metformin or DQ independently, with more follicular reserve, increased pups per litter, and reduced DNA damage. Collectively, our work indicates that senotherapies might prevent cisplatin-induced ovarian injury by removing senescent cells and reducing DNA damage, which represent a promising therapeutic avenue to prevent chemotherapy-induced ovarian damage.
Collapse
|
6
|
Sugishita Y, Meng L, Suzuki-Takahashi Y, Nishimura S, Furuyama S, Uekawa A, Tozawa-Ono A, Migitaka-Igarashi J, Koizumi T, Seino H, Natsuki Y, Kubota M, Koike J, Edashige K, Suzuki N. Quantification of residual cryoprotectants and cytotoxicity in thawed bovine ovarian tissues after slow freezing or vitrification. Hum Reprod 2022; 37:522-533. [PMID: 34928342 DOI: 10.1093/humrep/deab274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 11/17/2021] [Indexed: 09/27/2023] Open
Abstract
STUDY QUESTION How much residual cryoprotectant remains in thawed/warmed ovarian tissues after slow freezing or vitrification? SUMMARY ANSWER After thawing/warming, at least 60 min of diffusion washing in media was necessary to significantly reduce the residual cryoprotectants in ovarian tissues frozen by slow freezing or vitrification. WHAT IS KNOWN ALREADY Ovarian tissue cryopreservation (OTC) by slow freezing has been the conventional method; while the vitrification method has gained popularity for its practicality. The main concern about vitrification is how much potentially toxic residual cryoprotectant remains in the warmed tissues at the time of transplantation. STUDY DESIGN, SIZE, DURATION This was an animal study using the ovarian tissues from 20 bovine ovaries. The duration of this study was from 2018 to 2020. PARTICIPANTS/MATERIALS, SETTING, METHODS Ovarian cortex tissues were prepared from 20 bovine ovaries and assigned randomly to groups of fresh (non-frozen) control, slow freezing with 1.5 M dimethyl sulfoxide (DMSO), 1.5 M 1,2-propanediol (PROH) and vitrification with 35% ethylene glycol (EG). The residual cryoprotectant concentrations in thawed/warmed tissues were measured by gas chromatography at the following time points: frozen (before thawing/warming), 0 min (immediately after thawing/warming), 30, 60 and 120 min after diffusion washing in media. Next, the ultrastructural changes of primordial follicles, granulosa cells, organelles and stromal cells in the ovarian tissues (1 mm × 1 mm × 1 mm) were examined in fresh (non-frozen) control, slow freezing with DMSO or PROH and vitrification with EG groups. Real-time quantitative PCR was carried out to examine the expressions of poly (ADP-ribose) polymerase-1 (PARP1), a DNA damage sensor and caspase-3 (CASP3), an apoptosis precursor, in thawed/warmed ovarian tissues that were washed for either 0 or 120 min and subsequently in tissues that were ex vivo cultured for 24 or 48 h. The same set of tissues were also used to analyze the protein expressions of gamma H2A histone family member X (γH2AX) for DNA double-strand breaks and activated caspase-3 (AC3) for apoptosis by immunohistochemistry. MAIN RESULTS AND THE ROLE OF CHANCE The residual cryoprotectant concentrations decreased with the extension of diffusion washing time. After 60 min washing, the differences of residual cryoprotectant between DMSO, PROH and EG were negligible (P > 0.05). This washing did not affect the tissue integrity or significantly elevate the percentage of AC3 and γH2AX positive cells, indicating that tissues are safe and of good quality for transplantation. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Since the study was performed with ovarian tissues from bovines, generalizability to humans may be limited. Potential changes in ovarian tissue beyond 120 min were not investigated. WIDER IMPLICATIONS OF THE FINDINGS This study addresses concerns about the cytotoxicity of EG in warmed ovarian tissues and could provide insights when devising a standard vitrification protocol for OTC. STUDY FUNDING/COMPETING INTEREST(S) The study was funded by a Grant-in-Aid for Scientific Research (B) from the Japan Society for the Promotion of Science to N.S.
Collapse
Affiliation(s)
- Yodo Sugishita
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Lingbo Meng
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Yuki Suzuki-Takahashi
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Sandy Nishimura
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Sayako Furuyama
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Atsushi Uekawa
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Akiko Tozawa-Ono
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Junko Migitaka-Igarashi
- Department of Anatomy, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Tomoe Koizumi
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
- Center for Reproductive Medicine, Dokkyo Medical University, Saitama Medical Center, Koshigaya, Saitama, Japan
| | - Hibiki Seino
- Department of Pathology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Yasunori Natsuki
- Graduate School Facility of Electron Microscope Research, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Manabu Kubota
- Department of Pathology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Junki Koike
- Department of Anatomy, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| | - Keisuke Edashige
- Faculty of Agriculture and Marine Science, Department of Agricultural Chemistry, Kochi University, Nankoku, Kochi, Japan
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, St. Marianna University, School of Medicine, Kawasaki, Kanagawa, Japan
| |
Collapse
|
7
|
Sugishita Y, Taylan E, Kawahara T, Shahmurzada B, Suzuki N, Oktay K. Comparison of open and a novel closed vitrification system with slow freezing for human ovarian tissue cryopreservation. J Assist Reprod Genet 2021; 38:2723-2733. [PMID: 34398400 DOI: 10.1007/s10815-021-02297-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/03/2021] [Indexed: 12/29/2022] Open
Abstract
PURPOSE To investigate the differences concerning post-thawing/warming follicle survival, DNA damage and apoptosis in human ovarian tissues cryopreserved by slow freezing, open, or closed vitrification methods. METHODS A total of 50 pieces of 5 × 5 × 1 mm ovarian cortical pieces were harvested (5 donor ovaries; mean age 31 ± 6.62 years). From each donor, one cortical piece was used as baseline; the remaining were randomly assigned to slow freezing (SF), vitrification using open device (VF-open), or closed device (VF-closed) groups. After 8-10 weeks of cryostorage, tissues were evaluated 4 h after thawing/warming. Histological analysis was evaluated for follicle survival (primordial and primary follicle densities) by H&E staining. The percentages of primordial and primary follicles with DNA double-strand breaks (γH2AX) and apoptotic cell death pathway activation (AC3) were immunohistochemically assessed. Data were analysed using one-way ANOVA and LSD post hoc comparison. RESULTS Compared to the baseline, primordial follicle (pdf) densities significantly declined in all cryopreserved groups (SF, VF-open, and VF-closed, P < 0.05). However, the total and non-apoptotic pdf densities were similar among SF, VF-open, and VF-closed. SF and VF with either open or closed devices did not increase the percentages of primordial or primary follicles with DNA double-strand breaks (DSBs) or apoptosis compared to the baseline or among the freezing methods in the present study. CONCLUSION Based on the intact primordial follicle survival, DNA damage, and apoptosis rates after thawing/warming, SF vs VF with either open or newly developed closed devices appear to be comparable.
Collapse
Affiliation(s)
- Yodo Sugishita
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, FMB 224, New Haven, CT, 06520, USA
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University, School of Medicine, Kawasaki, Japan
| | - Enes Taylan
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, FMB 224, New Haven, CT, 06520, USA
| | - Tai Kawahara
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, FMB 224, New Haven, CT, 06520, USA
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Bunyad Shahmurzada
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, FMB 224, New Haven, CT, 06520, USA
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kutluk Oktay
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, FMB 224, New Haven, CT, 06520, USA.
| |
Collapse
|
8
|
Song Y, Liu H. A review on the relationship between anti-mullerian hormone and fertility in treating young breast cancer patients. BMC WOMENS HEALTH 2021; 21:295. [PMID: 34376160 PMCID: PMC8353739 DOI: 10.1186/s12905-021-01420-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/06/2021] [Indexed: 11/18/2022]
Abstract
Despite the fact that the long-term survival rate of breast cancer patients had been significantly improved owing to the systemic breast cancer therapies, there are still some side effects such as amenorrhea and fertility retention to be resolved, leaving it an important thing to understand the possible side effects on fertility and fertility preservation strategies while undergoing breast cancer treatment, due to the fact that most young patients hope to become pregnant and have children after breast cancer treatment. With anti-müllerian hormone (AMH) being the most sensitive marker for predicting ovarian function in young premenopausal women with breast cancer, this review is aimed to provide the additional guidance for clinical application of AMH by exploring the impacts of AMH on the fertility of young breast cancer patients, the relationship between AMH and metabolism, and the relationship between BRAC gene mutation and fertility protection strategies.
Collapse
Affiliation(s)
- Yixuan Song
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
9
|
Corey B, Smania MA, Spotts H, Andersen M. Young Women With Breast Cancer: Treatment, Care, and Nursing Implications. Clin J Oncol Nurs 2021; 24:139-147. [PMID: 32196004 DOI: 10.1188/20.cjon.139-147] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Young women with breast cancer (YWBC) are more likely to have aggressive disease, carry mutations for hereditary cancer genes, and experience higher mortality. They also may face reduced fertility because of the toxicity of chemotherapy. OBJECTIVES This article aims to present a review of YWBC treatments, sequelae of treatment, and psychosocial challenges. METHODS The authors performed a review of guideline-supported treatment options, patient resources, and nursing implications. FINDINGS Because of high-risk cancers and a lack of specific treatment guidelines, healthcare providers may consider aggressive treatments for younger patients. However, studies indicate that the foundation for treatment decisions for YWBC are best based on disease stage and National Comprehensive Cancer Network guidelines.
Collapse
|
10
|
Wilkinson AN. Diagnostic de cancer en première ligne. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2021; 67:e99-e103. [PMID: 33853924 PMCID: PMC8324143 DOI: 10.46747/cfp.6704e99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Affiliation(s)
- Anna N Wilkinson
- Professeure adjointe au département de médecine familiale, médecin de famille oncologue, et directrice du programme de troisième année en médecine familiale-oncologie à l'Université d'Ottawa (Ontario); elle préside également le Groupe d'intérêt des membres en soins aux patients atteints du cancer du Collège des médecins de famille du Canada
| |
Collapse
|
11
|
Wilkinson AN. Cancer diagnosis in primary care: Six steps to reducing the diagnostic interval. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2021; 67:265-268. [PMID: 33853914 PMCID: PMC8324147 DOI: 10.46747/cfp.6704265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Anna N Wilkinson
- Assistant Professor in the Department of Family Medicine, an FP oncologist, and Program Director of the third-year FP-Oncology Program at the University of Ottawa in Ontario, and Chair of the College of Family Physicians of Canada's Cancer Care Member Interest Group
| |
Collapse
|
12
|
Floyd JL, Campbell S, Rauh-Hain JA, Woodard T. Fertility preservation in women with early-stage gynecologic cancer: optimizing oncologic and reproductive outcomes. Int J Gynecol Cancer 2021; 31:345-351. [PMID: 32565487 PMCID: PMC12038822 DOI: 10.1136/ijgc-2020-001328] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/22/2022] Open
Abstract
Almost all standard therapies for gynecologic cancer, including surgical intervention, gonadotoxic chemotherapy, and radiation therapy, threaten a woman's childbearing potential. Preservation of fertility should be discussed with premenopausal women with early-stage gynecologic cancer shortly after diagnosis and, for women who desire to preserve fertility, during treatment planning. Many authors have investigated both oncologic and reproductive outcomes following fertility-sparing therapy, and there is ongoing development of assisted reproduction techniques available to cancer patients and survivors. Women with early-stage (IA1-IB1) cervical cancer may be candidates for fertility-sparing cervical conization, simple trachelectomy, or radical trachelectomy. In women with stage I epithelial ovarian cancer, fertility-sparing surgery appears safe overall, although controversy remains in patients with high-risk features (eg, high pathologic grade, clear cell histology, or stage IC disease). In women with low-grade, early-stage endometrial cancer, hormonal therapy has emerged as a viable option. Criteria for patient selection for fertility-sparing therapy are not well defined, thus patients and providers must carefully discuss potential risks and benefits. In general, in carefully selected patients, survival outcomes do not appear to differ significantly between radical and fertility-sparing approaches. Women who undergo fertility-sparing therapies may experience a number of fertility and obstetric complications. Preconception counseling with high-risk obstetric specialists is important to optimize health before a woman attempts to conceive. Identifying appropriate candidates for fertility-sparing treatments, assessing fertility potential, and helping women conceive after cancer treatment is best accomplished through multidisciplinary collaboration between gynecologic oncologists and fertility specialists.
Collapse
Affiliation(s)
- Jessica Lauren Floyd
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Sukhkamal Campbell
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Baylor College of Medicine, Houston, TX, USA
| | - Jose Alejandro Rauh-Hain
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Terri Woodard
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Baylor College of Medicine, Houston, TX, USA
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
13
|
Rosenfield RL, Cooke DW, Radovick S. Puberty in the Female and Its Disorders. SPERLING PEDIATRIC ENDOCRINOLOGY 2021:528-626. [DOI: 10.1016/b978-0-323-62520-3.00016-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
14
|
Vuković P, Peccatori FA, Massarotti C, Miralles MS, Beketić-Orešković L, Lambertini M. Preimplantation genetic testing for carriers of BRCA1/2 pathogenic variants. Crit Rev Oncol Hematol 2020; 157:103201. [PMID: 33333149 DOI: 10.1016/j.critrevonc.2020.103201] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
The detection of germline BRCA1/2 pathogenic variant has relevant implications for the patients and their family members. Family planning, prophylactic surgery and the possibility of preimplantation genetic testing for monogenic disorders (PGT-M) to avoid transmittance of pathogenic variants to the offspring are relevant topics in this setting. PGT-M is valuable option for BRCA carriers, but it remains a controversial and underdiscussed topic. Although the advances in PGT technologies have improved pregnancy rate, there are still several important challenges associated with its use. The purpose of this review is to report the current evidence on PGT-M for BRCA1/2 carriers, ethical concerns and controversy associated with its use, reproductive implications of BRCA pathogenic variants, underlying areas in which an educational effort would be beneficial as well as possibilities for future research efforts in the field.
Collapse
Affiliation(s)
- Petra Vuković
- Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, University Hospital Center Sestre Milosrdnice, Zagreb, 10000, Croatia.
| | - Fedro Alessandro Peccatori
- Fertility and Procreation Unit, Gynecologic Oncology Program, IEO European Institute of Oncology IRCCS, Milan, 20125, Italy.
| | - Claudia Massarotti
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy.
| | | | - Lidija Beketić-Orešković
- Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, University Hospital Center Sestre Milosrdnice, Zagreb, 10000, Croatia; Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, 10000, Croatia.
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, 16126, Italy.
| |
Collapse
|
15
|
Crown A, Muhsen S, Zabor EC, Sevilimedu V, Kelvin J, Goldfarb SB, Gemignani ML. Does Use of Neoadjuvant Chemotherapy Affect the Decision to Pursue Fertility Preservation Options in Young Women with Breast Cancer? Ann Surg Oncol 2020; 27:4740-4749. [PMID: 32767225 PMCID: PMC7554118 DOI: 10.1245/s10434-020-08883-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/23/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND The American Society of Clinical Oncology guidelines recommend early referral to reproductive endocrinology and infertility (REI) specialists for young women diagnosed with breast cancer. Current practice patterns demonstrate an increased utilization of neoadjuvant chemotherapy (NAC). We evaluated premenopausal women with breast cancer after consultation with a Fertility Nurse Specialist (FNS) and determine factors associated with referral to REI specialists. METHODS This retrospective review included all premenopausal women diagnosed at our institution with stage 0-III unilateral breast cancers between 2009 and 2015 who completed an FNS consultation. Clinicopathologic features and factors associated with referral to REI after FNS consultation were analyzed. RESULTS A total of 334 women were identified. Median age was 35 years (interquartile range 32-38). The majority of women were single (n = 198, 59.3%) and nulliparous (n = 239, 71.6%). REI referrals were common (n = 237, 71.0%). The Breast Surgery service was the most frequent referring service (n = 194, 58.1%), with significantly more REI referrals compared to Breast Medicine and Genetics services (p = 0.002). Nulliparity was associated with REI referral (p < 0.0001). Adjuvant chemotherapy (p = 0.003) was associated with pursuing REI referral, whereas NAC (p < 0.001) was associated with declining REI referral. CONCLUSIONS Most women elected to consult with an REI specialist, confirming strong interest in fertility preservation among premenopausal women with breast cancer. However, women receiving NAC more frequently declined referral to REI, suggesting that the need to start NAC may influence decisions regarding fertility preservation. With increasing utilization of NAC, our study supports the need for further counseling and education regarding fertility preservation for women undergoing NAC.
Collapse
Affiliation(s)
- Angelena Crown
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shirin Muhsen
- Clemenceau Medical Center, Johns Hopkins International, Beirut, Lebanon
| | - Emily C Zabor
- Department of Quantitative Health Sciences, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Varadan Sevilimedu
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joanne Kelvin
- Department of Medicine, Survivorship Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shari B Goldfarb
- Breast Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary L Gemignani
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
16
|
Kim SW, Han SJ, Han JY, Kim H, Ku SY, Suh CS. Predictor for supraphysiologic serum estradiol elevation on hCG triggering day of controlled ovarian stimulation using letrozole and gonadotropins in women with estrogen-dependent cancers. PLoS One 2020; 15:e0240870. [PMID: 33085706 PMCID: PMC7577464 DOI: 10.1371/journal.pone.0240870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/04/2020] [Indexed: 11/29/2022] Open
Abstract
The objective of this study was to evaluate predicting factors for supraphysiologic serum estradiol elevation during controlled ovarian stimulation (COS) with administration of letrozole and gonadotropins in patients with estrogen-dependent cancer. Use of aromatase inhibitors is recommended to prevent the potential effects of elevated serum estradiol levels and recurrence of tumor in patients with estrogen-dependent cancers during COS. Although previous studies reported that letrozole have shown an effective lowering of peak estrogen levels, a part of patients shows supraphysiologic levels of estrogen associated with ovarian stimulation despite the administration of letrozole. From January 2009 to December 2019, patients with estrogen-dependent cancer who underwent COS with antagonist protocol using a letrozole (5 mg/ day) to keep estrogen levels low were included in this study. Early monitoring serum estradiol was measured in all patients on the 4-6th day of stimulation. Subjects were classified into two groups according to the serum estradiol level on hCG triggering day, physiologic estradiol group (≤400 pg/mL) and supraphysiologic estradiol group (>400 pg/mL). A total of 96 COS cycles were retrospectively analyzed. Supraphysiologic level of serum estradiol was found in 21.9% of the patients. Mean age, AMH, duration of stimulation, total dose of gonadotropins administered were not different between the two groups. However, early monitoring serum estradiol level was significantly higher in the supraphysiologic estradiol group (67.1±47.9 vs. 115.6±78.1, p = 0.001) and was associated with the occurrence of supraphysiologic elevation of serum estradiol on hCG triggering day. Patients with early monitoring serum estradiol ≥84.5 pg/mL had an odds ratio of 5.376 [95% CI, 1.613-17.913] for supraphysiologic elevation of serum estradiol compared to those with early monitoring serum estradiol below 84.5 pg/mL. In conclusion, early monitoring serum estradiol is an independent predicting factor for supraphysiologic level of serum estradiol on hCG triggering day in the COS cycles using letrozole and gonadotropins.
Collapse
Affiliation(s)
- Sung Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
| | - Soo Jin Han
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
| | - Ji Yeon Han
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Chang Suk Suh
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
| |
Collapse
|
17
|
Bercaire LMN, Cavagna M, Donadio NF, Rocha AR, Portela R, Alves VR, Santos TBB, Cavagna F, Dzik A, Gebrim LH, Nahas EAP. The impact of letrozole administration on oocyte morphology in breast cancer patients undergoing fertility preservation. JBRA Assist Reprod 2020; 24:257-264. [PMID: 32293820 PMCID: PMC7365524 DOI: 10.5935/1518-0557.20200002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Objective: Patients submitted to oncological fertility preservation with letrozole and gonadotropins seem to present a higher rate of immature oocytes and lower fertilization rates in comparison to infertile patients submitted to IVF cycles with gonadotropins. The aim of this study was to evaluate the influence of letrozole on oocyte morphology in patients with breast cancer submitted to fertility preservation. Methods: Retrospective analysis performed at a public tertiary hospital in São Paulo, Brazil. The oocytes were retrieved from patients with breast cancer undergoing fertility preservation (n=69), and from infertile women undergoing in vitro fertilization (n=92). We evaluated 750 oocytes obtained from breast cancer patients submitted to ovarian stimulation with letrozole and gonadotropins, and 699 oocytes from patients without breast cancer submitted to ovarian stimulation for in vitro fertilization with gonadotropins only due to male factor infertility. The mature oocytes retrieved were analyzed for the presence of refractile bodies, ooplasm color and regularity, central granulation degree, cortical granules, zona pellucida staining and regularity, perivitelline space, presence of vacuoles or abnormal smooth-surfaced endoplasmic reticle and oocyte retraction. Results: There was a higher incidence of alterations in oocyte morphology in the letrozole group when compared to the control group: increased perivitelline space (p=0.007), irregular zona pellucida (p<0.001), refractile bodies (p<0.001), dark ooplasm (p<0.001), granular ooplasm (p<0.001), irregular ooplasm (p<0.001) and dense central granulation (p<0.001). Conclusion: Letrozole is a risk factor for worse oocyte morphology. However, the clinical impact of ovarian stimulation protocol with combined use of gonadotropins and letrozole for fertility preservation remains unclear in this setting. These data underline the importance of establishing the predictive potential of morphological dimorphisms of human oocytes in IVF outcomes.
Collapse
Affiliation(s)
- Ludmila M N Bercaire
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil.,Gynecology and Obstetrics Department, Botucatu Medical School, UNESP - Universidade Estadual de São Paulo, Botucatu, SP, Brazil
| | - Mario Cavagna
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Nilka F Donadio
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Andressa R Rocha
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Rafael Portela
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Vanessa R Alves
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Thamara B B Santos
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Felipe Cavagna
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Artur Dzik
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Luiz H Gebrim
- Human Reproduction Department. Women's Health Reference Center - Pérola Byington Hospital, São Paulo, SP, Brazil
| | - Eliana A P Nahas
- Gynecology and Obstetrics Department, Botucatu Medical School, UNESP - Universidade Estadual de São Paulo, Botucatu, SP, Brazil
| |
Collapse
|
18
|
Silvestris E, Dellino M, Cafforio P, Paradiso AV, Cormio G, D'Oronzo S. Breast cancer: an update on treatment-related infertility. J Cancer Res Clin Oncol 2020; 146:647-657. [PMID: 32006118 DOI: 10.1007/s00432-020-03136-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 01/22/2020] [Indexed: 12/19/2022]
Abstract
Breast cancer (BC) is the most common malignancy in women with a significant increasing incidence during the reproductive life. However, based on the newest anti-cancer molecular targeting drugs, successful treatments lead to the disease healing particularly in young patients, thus refreshing their motherhood programs. However, as effect of the BC treatment, a premature depletion of the ovarian follicle reserve occurs in more than one-third of patients resulting in permanent infertility. To prevent the cancer treatment-related infertility (CTRI), several options are today utilized. Besides the ovary suppression by gonadotropin-releasing hormone agonist (GnRHa), other procedures include either oocytes or embryos cryopreservation as well as ovarian cortex cryopreservation that are currently adopted before anti-cancer therapies. These modern techniques appear variably successful in terms of pregnancy rate though their safety concerning the hormonal stimulation to promote the folliculogenesis is still debated in relation to the potential oncogenic risk in patients bearing hormone-sensitive tumors as BC, while the ovarian cortex re-implantation often results in a low number of regenerated follicles including oocytes of unknown quality. Recent studies on ovarian stem cells (OSCs) suggest their use for future application in CTRI. In fact, OSCs from ovarian cortex have been shown to differentiate in vitro into oocyte-like cells (OLCs) and express molecular markers of mature oocytes. Once the OSC technology will be optimized and translated to clinical use, oocytes derived from these cells will be molecularly assessed before fertilization to assure their best embryo quality resulting in a safe procedure to treat CTRI in patients as young women with BC.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy.
| | - Miriam Dellino
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Paola Cafforio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Angelo Virgilio Paradiso
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
- Department of Biomedical Sciences and Human Oncology, Unit of Obstetrics and Gynecology, University of Bari Aldo Moro, Bari, Italy
| | - Stella D'Oronzo
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, Bari, Italy
- IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
19
|
Durrani S, Heena H. Controversies Regarding Ovarian Suppression and Infertility in Early Stage Breast Cancer. Cancer Manag Res 2020; 12:813-817. [PMID: 32104064 PMCID: PMC7008199 DOI: 10.2147/cmar.s231524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/15/2020] [Indexed: 01/24/2023] Open
Abstract
A common side effect of chemotherapy in breast cancer is early menopause in premenopausal patients, which is mainly a result of an indirect form of ovarian ablation, and is associated with substantial impairment of quality of life. Suppressing the production of ovarian estrogen has been shown to reduce the recurrence of hormone receptor-positive early breast cancer in premenopausal women, but whether it has an added advantage over tamoxifen is being discussed. Types of permanent ablation of the ovarian function include surgical oophorectomy and radiation-induced ovarian failure. Both are associated with similar response rates in hormone receptor-positive metastatic breast cancer. Medical castration with luteinizing hormone-releasing hormone analogs (LHRHa) has the benefit of being a reversible approach. Another advantage that premenopausal patients who wish to reduce the risk of developing premature ovarian insufficiency induced by chemotherapy may be offered LHRHa irrespective of whether they desire pregnancy and their age at diagnosis. This also helps reduce the risk of menopausal signs and symptoms as well as the loss of bone density in the long-term, which are primary concerns for women. This is of utmost importance to premenopausal women who do not want to conceive after treatment or are not candidates for fertility preservation strategies because of age. It should be emphasized that for women who are interested in fertility preservation, gamete cryopreservation remains the first option, and LHRHa is not an alternative. During chemotherapy, however, temporary ovarian suppression with LHRHa may be given to women who either have no access to a fertility clinic or who have declined chemotherapy or have contraindications.
Collapse
Affiliation(s)
- Sajid Durrani
- Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Humariya Heena
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Edib Z, Jayasinghe Y, Hickey M, Stafford L, Anderson RA, Su HI, Stern K, Saunders C, Anazodo A, Macheras-Magias M, Chang S, Pang P, Agresta F, Chin-Lenn L, Cui W, Pratt S, Gorelik A, Peate M. Exploring the facilitators and barriers to using an online infertility risk prediction tool (FoRECAsT) for young women with breast cancer: a qualitative study protocol. BMJ Open 2020; 10:e033669. [PMID: 32047016 PMCID: PMC7044829 DOI: 10.1136/bmjopen-2019-033669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/01/2022] Open
Abstract
INTRODUCTION As cancer treatments may impact on fertility, a high priority for young patients with breast cancer is access to evidence-based, personalised information for them and their healthcare providers to guide treatment and fertility-related decisions prior to cancer treatment. Current tools to predict fertility outcomes after breast cancer treatments are imprecise and do not offer individualised prediction. To address the gap, we are developing a novel personalised infertility risk prediction tool (FoRECAsT) for premenopausal patients with breast cancer that considers current reproductive status, planned chemotherapy and adjuvant endocrine therapy to determine likely post-treatment infertility. The aim of this study is to explore the feasibility of implementing this FoRECAsT tool into clinical practice by exploring the barriers and facilitators of its use among patients and healthcare providers. METHODS AND ANALYSIS A cross-sectional exploratory study is being conducted using semistructured in-depth telephone interviews with 15-20 participants each from the following groups: (1) premenopausal patients with breast cancer younger than 40, diagnosed within last 5 years, (2) breast surgeons, (3) breast medical oncologists, (4) breast care nurses (5) fertility specialists and (6) fertility preservation nurses. Patients with breast cancer are being recruited from the joint Breast Service of three affiliated institutions of Victorian Comprehensive Cancer Centre in Melbourne, Australia-Peter MacCallum Cancer Centre, Royal Melbourne Hospital and Royal Women's Hospital, and clinicians are being recruited from across Australia. Interviews are being audio recorded, transcribed verbatim and imported into qualitative data analysis software to facilitate data management and analyses. ETHICS AND DISSEMINATION The study protocol has been approved by Melbourne Health Human Research Ethics Committee, Australia (HREC number: 2017.163). Confidentiality and privacy are maintained at every stage of the study. Findings will be disseminated through peer-reviewed scholarly and scientific journals, national and international conference presentations, social media, broadcast media, print media, internet and various community/stakeholder engagement activities.
Collapse
Affiliation(s)
- Zobaida Edib
- Obstetrics & Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
- Obstetrics & Gynaecology, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Yasmin Jayasinghe
- Obstetrics & Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
- Obstetrics & Gynaecology, The Royal Women's Hospital, Parkville, Victoria, Australia
- Department of Paediatric and Adolescent Gynaecology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Martha Hickey
- Obstetrics & Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
- Obstetrics & Gynaecology, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Lesley Stafford
- Melbourne School of Psychological Sciences, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Women's Mental Health, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, Edinburgh, UK
| | - H Irene Su
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, California, USA
| | - Kate Stern
- Melbourne IVF, East Melbourne, Victoria, Australia
- Reproductive Services, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Christobel Saunders
- School of Surgery, The University of Western Australia, Perth, Western Australia, Australia
| | - Antoinette Anazodo
- Sydney Children's Hospital, School of Women's and Children's Health, UNSW, Sydney, New South Wales, Australia
- Nelune Comprehensive Cancer Centre, Prince of Wales Children's Hospital, Randwick, New South Wales, Australia
| | | | - Shanton Chang
- School of Computing and Information Systems, The University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick Pang
- School of Computing and Information Systems, The University of Melbourne, Melbourne, Victoria, Australia
| | - Franca Agresta
- Melbourne IVF, East Melbourne, Victoria, Australia
- Reproductive Services, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Laura Chin-Lenn
- Department of General Surgery, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Wanyuan Cui
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sarah Pratt
- Breast Service, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alex Gorelik
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria, Australia
- School of Behavioural and Health Sciences, Australian Catholic University, Melbourne, Victoria, Australia
| | - Michelle Peate
- Obstetrics & Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
- Obstetrics & Gynaecology, The Royal Women's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
21
|
Dong J, Esham KS, Boehm L, Karim SA, Lin M, Mao D, Wang F, Fein D, Wang H, Studenmund C, Weidner RA, Noubary F, Freund KM, Erban JK, Parsons SK. Timeliness of Treatment Initiation in Newly Diagnosed Patients With Breast Cancer. Clin Breast Cancer 2020; 20:e27-e35. [PMID: 31439436 PMCID: PMC11372729 DOI: 10.1016/j.clbc.2019.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/06/2019] [Accepted: 06/25/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Evidence-based timeliness benchmarks have been established to assess quality of breast cancer care, as delays in treatment are associated with poor clinical outcomes. However, few studies have evaluated how current breast cancer care meets these benchmarks and what factors may delay the timely initiation of treatment. PATIENTS AND METHODS Demographic and disease characteristics of 377 newly diagnosed patients with breast cancer who initiated treatment at Tufts Medical Center (2009-2015) were extracted from electronic medical records. Time from diagnosis to initial surgery and time from diagnosis to initiation of hormone therapy were estimated with Kaplan-Meier curves. Multivariable regression analysis was used to identify factors associated with treatment delays. Thematic analysis was performed to categorize reasons for delay. RESULTS Of 319 patients who had surgery recommended as the first treatment, 248 (78%) met the 45-day benchmark (median, 28 days; 25th-75th %, 19-43). After adjusting for potential confounders, multivariable regression analysis revealed that negative hormone receptor status (odds ratio, 3.48; 95% confidence interval, 1.44-8.43) and mastectomy (odds ratio, 4.07; 95% confidence interval, 2.10-8.06) were significantly associated with delays in surgery. Delays were mostly owing to clinical complexity or logistical/financial reasons. Of 241 patients eligible for hormone therapy initiation, 232 (96%) met the 1-year benchmark (median, 147 days; 25th-75th %, 79-217). CONCLUSION Most patients met timeliness guidelines for surgery and initiation of hormone therapy, although risk factors for delay were identified. Knowledge of reasons for breast cancer treatment delay, including clinical complexity and logistical/financial issues, may allow targeting interventions for patients at greatest risk of care delays.
Collapse
Affiliation(s)
- Jinghui Dong
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Kimberly S Esham
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA; Department of Medicine, Tufts University School of Medicine, Boston, MA; Division of Hematology/Oncology, Tufts Medical Center, Boston, MA
| | - Lauren Boehm
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Sabrina A Karim
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Mingqian Lin
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Daqin Mao
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Fengqing Wang
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Daniel Fein
- Department of Medicine, Tufts Medical Center, Boston, MA
| | - Hanyin Wang
- Department of Medicine, Tufts Medical Center, Boston, MA
| | - Christine Studenmund
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Ruth Ann Weidner
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Farzad Noubary
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Karen M Freund
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA; Department of Medicine, Tufts University School of Medicine, Boston, MA; Department of Medicine, Tufts Medical Center, Boston, MA
| | - John K Erban
- Department of Medicine, Tufts University School of Medicine, Boston, MA; Division of Hematology/Oncology, Tufts Medical Center, Boston, MA; Department of Medicine, Tufts Medical Center, Boston, MA
| | - Susan K Parsons
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA; Department of Medicine, Tufts University School of Medicine, Boston, MA; Division of Hematology/Oncology, Tufts Medical Center, Boston, MA; Department of Medicine, Tufts Medical Center, Boston, MA; Department of Pediatrics, Tufts University School of Medicine, Boston, MA.
| |
Collapse
|
22
|
Taylan E, Oktay K. Fertility preservation in gynecologic cancers. Gynecol Oncol 2019; 155:522-529. [PMID: 31604663 DOI: 10.1016/j.ygyno.2019.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022]
Abstract
An increasing number of women in modern societies are delaying childbearing beyond the age of 35, and gynecologic cancers affect a significant proportion of reproductive age women who wish to preserve fertility for a future chance of childbearing. As a result, providing treatment options for fertility preservation in women with gynecologic cancer has become a crucial component of cancer survivorship care. In this review article, we discussed the current knowledge on fertility-sparing surgical approaches, as well as assisted reproductive technologies that can be utilized to preserve reproductive potential in women with cervical, endometrial, and ovarian cancer. A brief section on fertility preservation in pediatric gynecologic malignancies is also provided.
Collapse
Affiliation(s)
- Enes Taylan
- Women's Cancer Program at Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.
| | - Kutluk Oktay
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA; Innovation Institute for Fertility Preservation and IVF, New York, NY, USA; Ovarian Transplantation Program, NYU Winthrop Hospital, Mineola, NY, USA
| |
Collapse
|
23
|
Fertilitätsprotektion – jetzt auch Kassenleistung. GYNAKOLOGISCHE ENDOKRINOLOGIE 2019. [DOI: 10.1007/s10304-019-00269-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
24
|
EUropean REcommendations for female FERtility preservation (EU-REFER): A joint collaboration between oncologists and fertility specialists. Crit Rev Oncol Hematol 2019; 138:233-240. [DOI: 10.1016/j.critrevonc.2019.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/25/2018] [Accepted: 03/20/2019] [Indexed: 11/17/2022] Open
|
25
|
Vuković P, Kasum M, Raguž J, Lonjak N, Bilić Knežević S, Orešković I, Beketić Orešković L, Čehić E. FERTILITY PRESERVATION IN YOUNG WOMEN WITH EARLY-STAGE BREAST CANCER. Acta Clin Croat 2019; 58:147-156. [PMID: 31363337 PMCID: PMC6629203 DOI: 10.20471/acc.2019.58.01.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Although breast cancer (BC) occurs more often in older women, it is the most commonly diagnosed malignancy in women of childbearing age. Owing to the overall advancement of modern medicine and the growing global trend of delaying childbirth until later age, we find ever more younger women diagnosed and treated for BC who have not yet completed their family. Therefore, fertility preservation has emerged as a very important quality of life issue for young BC survivors. This paper reviews currently available options for fertility preservation in young women with early-stage BC and highlights the importance of a multidisciplinary approach to fertility preservation as a very important quality of life issue for young BC survivors. Pregnancy after BC treatment is considered not to be associated with an increased risk of BC recurrence; therefore, it should not be discouraged for those women who want to achieve pregnancy after oncologic treatment. Currently, it is recommended to delay pregnancy for at least 2 years after BC diagnosis, when the risk of recurrence is highest. However, BC patients of reproductive age should be informed about the potential negative effects of oncologic therapy on fertility, as well as on the fertility preservation options available, and if interested in fertility preservation, they should be promptly referred to a reproductive specialist. Early referral to a reproductive specialist is an important factor that increases the likelihood of successful fertility preservation. Embryo and mature oocyte cryopreservation are currently the only established fertility preservation methods but they require ovarian stimulation (OS), which delays initiation of chemotherapy for at least 2 weeks. Controlled OS does not seem to increase the risk of BC recurrence. Other fertility preservation methods (ovarian tissue cryopreservation, cryopreservation of immature oocytes and ovarian suppression with gonadotropin-releasing hormone agonists) do not require OS but are still considered to be experimental techniques for fertility preservation.
Collapse
Affiliation(s)
| | - Miro Kasum
- 1Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 2Department of Obstetrics and Gynecology, Zagreb University Hospital Centre, Zagreb, Croatia; 3Zadar General Hospital, Department of Oncology and Nuclear Medicine, Zadar, Croatia; 4School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, Croatia; 6Human Reproduction Unit, Zenica Cantonal Hospital, Zenica, Bosnia and Herzegovina
| | - Jelena Raguž
- 1Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 2Department of Obstetrics and Gynecology, Zagreb University Hospital Centre, Zagreb, Croatia; 3Zadar General Hospital, Department of Oncology and Nuclear Medicine, Zadar, Croatia; 4School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, Croatia; 6Human Reproduction Unit, Zenica Cantonal Hospital, Zenica, Bosnia and Herzegovina
| | - Nikolina Lonjak
- 1Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 2Department of Obstetrics and Gynecology, Zagreb University Hospital Centre, Zagreb, Croatia; 3Zadar General Hospital, Department of Oncology and Nuclear Medicine, Zadar, Croatia; 4School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, Croatia; 6Human Reproduction Unit, Zenica Cantonal Hospital, Zenica, Bosnia and Herzegovina
| | - Sara Bilić Knežević
- 1Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 2Department of Obstetrics and Gynecology, Zagreb University Hospital Centre, Zagreb, Croatia; 3Zadar General Hospital, Department of Oncology and Nuclear Medicine, Zadar, Croatia; 4School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, Croatia; 6Human Reproduction Unit, Zenica Cantonal Hospital, Zenica, Bosnia and Herzegovina
| | - Ivana Orešković
- 1Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 2Department of Obstetrics and Gynecology, Zagreb University Hospital Centre, Zagreb, Croatia; 3Zadar General Hospital, Department of Oncology and Nuclear Medicine, Zadar, Croatia; 4School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, Croatia; 6Human Reproduction Unit, Zenica Cantonal Hospital, Zenica, Bosnia and Herzegovina
| | - Lidija Beketić Orešković
- 1Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 2Department of Obstetrics and Gynecology, Zagreb University Hospital Centre, Zagreb, Croatia; 3Zadar General Hospital, Department of Oncology and Nuclear Medicine, Zadar, Croatia; 4School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, Croatia; 6Human Reproduction Unit, Zenica Cantonal Hospital, Zenica, Bosnia and Herzegovina
| | - Ermin Čehić
- 1Division of Radiotherapy and Medical Oncology, University Hospital for Tumors, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 2Department of Obstetrics and Gynecology, Zagreb University Hospital Centre, Zagreb, Croatia; 3Zadar General Hospital, Department of Oncology and Nuclear Medicine, Zadar, Croatia; 4School of Medicine, University of Zagreb, Zagreb, Croatia; 5Department of Clinical Oncology, School of Medicine, University of Zagreb, Zagreb, Croatia; 6Human Reproduction Unit, Zenica Cantonal Hospital, Zenica, Bosnia and Herzegovina
| |
Collapse
|
26
|
Heudobler D, Rechenmacher M, Lüke F, Vogelhuber M, Klobuch S, Thomas S, Pukrop T, Hackl C, Herr W, Ghibelli L, Gerner C, Reichle A. Clinical Efficacy of a Novel Therapeutic Principle, Anakoinosis. Front Pharmacol 2018; 9:1357. [PMID: 30546308 PMCID: PMC6279883 DOI: 10.3389/fphar.2018.01357] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
Classic tumor therapy, consisting of cytotoxic agents and/or targeted therapy, has not overcome therapeutic limitations like poor risk genetic parameters, genetic heterogeneity at different metastatic sites or the problem of undruggable targets. Here we summarize data and trials principally following a completely different treatment concept tackling systems biologic processes: the principle of communicative reprogramming of tumor tissues, i.e., anakoinosis (ancient greek for communication), aims at establishing novel communicative behavior of tumor tissue, the hosting organ and organism via re-modeling gene expression, thus recovering differentiation, and apoptosis competence leading to cancer control - in contrast to an immediate, "poisoning" with maximal tolerable doses of targeted or cytotoxic therapies. Therefore, we introduce the term "Master modulators" for drugs or drug combinations promoting evolutionary processes or regulating homeostatic pathways. These "master modulators" comprise a broad diversity of drugs, characterized by the capacity for reprogramming tumor tissues, i.e., transcriptional modulators, metronomic low-dose chemotherapy, epigenetically modifying agents, protein binding pro-anakoinotic drugs, such as COX-2 inhibitors, IMiDs etc., or for example differentiation inducing therapies. Data on 97 anakoinosis inducing schedules indicate a favorable toxicity profile: The combined administration of master modulators, frequently (with poor or no monoactivity) may even induce continuous complete remission in refractory metastatic neoplasia, irrespectively of the tumor type. That means recessive components of the tumor, successively developing during tumor ontogenesis, are accessible by regulatory active drug combinations in a therapeutically meaningful way. Drug selection is now dependent on situative systems characteristics, to less extent histology dependent. To sum up, anakoinosis represents a new substantive therapy principle besides novel targeted therapies.
Collapse
Affiliation(s)
- Daniel Heudobler
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Michael Rechenmacher
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Florian Lüke
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Martin Vogelhuber
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Sebastian Klobuch
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Simone Thomas
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Christina Hackl
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Lina Ghibelli
- Department Biology, Universita' di Roma Tor Vergata, Rome, Italy
| | - Christopher Gerner
- Faculty Chemistry, Institut for Analytical Chemistry, University Vienna, Vienna, Austria
| | - Albrecht Reichle
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
27
|
Weterings MAJ, Glanville E, van Eekelen R, Den Hartog JE, Farquhar C. Interventions for fertility preservation in women with cancer undergoing chemotherapy. Hippokratia 2017. [DOI: 10.1002/14651858.cd012891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Maria AJ Weterings
- Maastrict University Medical Center; P. Debyelaan 25 Maastricht Limburg Netherlands 6229 HX
| | | | - Rik van Eekelen
- Academic Medical Center, Amsterdam; Centre for Reproductive Medicine; Meibergdreef 9 Amsterdam Netherlands 1105 AZ
| | - Janneke Eva Den Hartog
- Maastricht University Medical Centre; Department of Gynecology and Obstetrics; P. Debyelaan 25 Maastricht Netherlands 6229 HX
| | - Cindy Farquhar
- University of Auckland; Department of Obstetrics and Gynaecology; FMHS Park Road Grafton Auckland New Zealand 1003
| |
Collapse
|