1
|
Ma S, Zhou L, Liu Y, Jie H, Yi M, Guo C, Mei J, Li C, Zhu L, Deng S. Identification of a novel chemotherapy benefit index for patients with advanced ovarian cancer based on Bayesian network analysis. PLoS One 2025; 20:e0322130. [PMID: 40424327 PMCID: PMC12112407 DOI: 10.1371/journal.pone.0322130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/16/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND This study aims to evaluate the efficacy of chemotherapy and optimize treatment strategies for patients with advanced ovarian cancer. METHODS Based on The Cancer Genome Atlas (TCGA) transcriptome data, we conducted correlation and Bayesian network analyses to identify key genes strongly associated with chemotherapy prognosis. Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR) was used to verify the expression of these key genes. The Chemotherapy Benefit Index (CBI) was developed using these genes via multivariable Cox regression analysis, and validated using both internal and external validation sets (GSE32062 and GSE30161) with a random forest model. Subsequently, we analyzed distinct molecular characteristics and explored additional immunotherapy in CBI-high and CBI-low subgroups. RESULTS Based on the network and machine learning analyses, CBI was developed from the following ten genes: COL6A3, SPI1, HSF1, CD3E, PIK3R4, MZB1, FERMT3, GZMA, PSMB9 and RSF1. Significant differences in overall survival were observed among the CBI-high, medium, and low subgroups (P < 0.001), which were consistent with the two external validation sets (P < 0.001 and P = 0.003). The AUC of internal validation and two external validation cohorts were 0.87, 0.71 and 0.70, respectively. Molecular function analysis indicated that the CBI-low subgroup is characterized by the activation of cancer-related signaling pathways, immune-related biological processes, higher TP53 mutation rate, particularly with a better response to immune checkpoint blockade (ICB) treatment, while the CBI-high subgroup is characterized by inhibition of cell cycle, less response to ICB treatment, and potential therapeutic targets. CONCLUSIONS This study provided a novel CBI for patients with advanced ovarian cancer through network analyses and machine learning. CBI could serve as a prognostic prediction tool for patients with advanced ovarian cancer, and also as a potential indicator for immunotherapy.
Collapse
Affiliation(s)
- Shuxiao Ma
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Zhou
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Jie
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Min Yi
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Chenglin Guo
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiandong Mei
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Chuan Li
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Zhu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Senyi Deng
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Ewy J, Senapati S, Ali ND, Gracia CR. Special considerations in assisted reproductive technology for patients with hematologic disease. Fertil Steril 2025:S0015-0282(25)00240-7. [PMID: 40288481 DOI: 10.1016/j.fertnstert.2025.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Populations with hematologic disorders seeking fertility care often present with a complex clinical picture, including disease-specific sequelae and comorbid conditions. Limited literature exists to help guide fertility clinics on the management of these patients, many of whom require multidisciplinary care coordination centered on patient-specific fertility goals. Thanks to advancements in life-prolonging therapies for hematologic disorders, growing numbers of affected individuals are seeking assisted reproductive technologies for family building. Thus, it is important to be aware of the unique considerations and risks of assisted reproductive technologies for these populations and develop evidence-based care guidelines to optimize outcomes.
Collapse
Affiliation(s)
- Joshua Ewy
- Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Suneeta Senapati
- Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nadia D Ali
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Clarisa R Gracia
- Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Su HI, Lacchetti C, Letourneau J, Partridge AH, Qamar R, Quinn GP, Reinecke J, Smith JF, Tesch M, Wallace WH, Wang ET, Loren AW. Fertility Preservation in People With Cancer: ASCO Guideline Update. J Clin Oncol 2025; 43:1488-1515. [PMID: 40106739 DOI: 10.1200/jco-24-02782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/09/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE To provide updated fertility preservation (FP) recommendations for people with cancer. METHODS A multidisciplinary Expert Panel convened and updated the systematic review. RESULTS One hundred sixty-six studies comprise the evidence base. RECOMMENDATIONS People with cancer should be evaluated for and counseled about reproductive risks at diagnosis and during survivorship. Patients interested in or uncertain about FP should be referred to reproductive specialists. FP approaches should be discussed before cancer-directed therapy. Sperm cryopreservation should be offered to males before cancer-directed treatment, with testicular sperm extraction if unable to provide semen samples. Testicular tissue cryopreservation in prepubertal males is experimental and should be offered only in a clinical trial. Males should be advised of potentially higher genetic damage risks in sperm collected soon after cancer-directed therapy initiation and completion. For females, established FP methods should be offered, including embryo, oocyte, and ovarian tissue cryopreservation (OTC), ovarian transposition, and conservative gynecologic surgery. In vitro maturation of oocytes may be offered as an emerging method. Post-treatment FP may be offered to people who did not undergo pretreatment FP or cryopreserve enough oocytes or embryos. Gonadotropin-releasing hormone agonist (GnRHa) should not be used in place of established FP methods but may be offered as an adjunct to females with breast cancer. For patients with oncologic emergencies requiring urgent oncologic therapy, GnRHa may be offered for menstrual suppression. Established FP methods in children who have begun puberty should be offered with patient assent and parent/guardian consent. The only established method for prepubertal females is OTC. Oncology teams should ensure prompt access to a multidisciplinary FP team. Clinicians should advocate for comprehensive FP services coverage and help patients access benefits.Additional information is available at www.asco.org/survivorship-guidelines.
Collapse
Affiliation(s)
- H Irene Su
- University of California, San Diego, San Diego, CA
| | | | | | | | | | | | | | - James F Smith
- University of California, San Francisco, San Francisco, CA
| | | | - W Hamish Wallace
- Royal Hospital for Children & Young People & University of Edinburgh, Edinburgh, United Kingdom
| | | | - Alison W Loren
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
4
|
Feng C, Jiang Y, Wang Y, Zhang Y, Liu Y, Li J. Protocatechualdehyde improves cyclophosphamide-induced premature ovarian insufficiency by inhibiting granulosa cell apoptosis and senescence through the SIRT1/p53 axis. Reprod Toxicol 2025; 135:108903. [PMID: 40228705 DOI: 10.1016/j.reprotox.2025.108903] [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: 12/30/2024] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025]
Abstract
Premature ovarian insufficiency (POI) is a prevalent gynecological disorder. Cyclophosphamide (CP), as a chemotherapeutic drug, particularly plays an important role in inducing POI. Protocatechualdehyde (PCA) is a major phenolic acid in Chinese herb Danshen, and has been reported to have beneficial effects on anti-inflammatory, anti-apoptotic, and anti-oxidant functions. We aimed to investigate the effect of different doses of PCA on ovarian function and the underlying molecular mechanisms. PCA administration reduced estrous cycle disorders, increased ovarian weight, promoted the secretion of serum hormone levels, and improved the CP-damaged ovarian microenvironment. Importantly, the administration of PCA contributed to the recovery of ovarian function with POI by inhibiting the senescence and apoptosis of granulosa cells. In vitro assay further confirmed the protective effect of PCA on CP-induced senescence and apoptosis of granulosa cells. Mechanistically, both in vivo and in vitro experiments proved that PCA administration promoted activation of the Sirt1/p53 signaling cascade, ultimately improving ovarian function. In conclusion, PCA might protect against ovarian damage in CP-induced POI that might be related to its activity on senescence and apoptosis of granulosa cells by the Sirt1/p53 pathway.
Collapse
Affiliation(s)
- Cong Feng
- Department of Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yue Jiang
- Department of Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yi Wang
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yuehui Zhang
- Department of Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yu Liu
- Department of Plastic and Maxillofacial Surgery, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Jia Li
- Department of Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
5
|
Hawazie A, Druce M. Breast Cancer Risk and Management in the Endocrine Clinic: A Comprehensive Review. Clin Endocrinol (Oxf) 2025. [PMID: 39905814 DOI: 10.1111/cen.15209] [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: 04/09/2024] [Revised: 12/30/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVE This review seeks to provide endocrine clinicians with a comprehensive analysis of breast cancer risk, diagnostic modalities and management strategies in women with endocrine disorders, with particular emphasis on the influence of metabolic factors such as diabetes and obesity, and the role of Menopausal Hormone Therapy (MHT). DESIGN The review examines a spectrum of endocrine disorders commonly encountered in clinical practice, including Multiple Endocrine Neoplasia Types 1 (MEN1), 2 (MEN2) and 4 (MEN4), Von Hippel-Lindau syndrome (VHL), Pheochromocytoma and Paraganglioma (PPGL), Acromegaly, Hyperprolactinaemia, Polycystic Ovary Syndrome (PCOS), Congenital Adrenal Hyperplasia (CAH), Turner Syndrome, alongside metabolic conditions such as diabetes and obesity and the effects of MHT. The review critically appraises each disorder's association with breast cancer risk, screening implications and therapeutic management. PATIENTS This analysis focuses on women with the aforementioned endocrine and metabolic disorders, assessing their specific breast cancer risk profiles, informed by the latest clinical evidence and molecular insights. MEASUREMENTS The review comprehensively evaluates current evidence-based approaches to screening, diagnostic accuracy and treatment in this patient cohort. Emphasis is placed on the metabolic derangements, hormonal influences and genetic predispositions that modulate breast cancer risk, providing disorder-specific recommendations for individualised care. RESULTS The findings indicate a significantly elevated breast cancer risk in patients with MEN1, necessitating early initiation of MRI screening by age 40. In MEN2, emerging evidence suggests that combining RET inhibitors with endocrine therapy may yield clinical benefits, although further research is needed to validate this approach. The breast cancer risk associated with MEN4 and VHL syndromes, while documented, remains less well-characterised, requiring further investigation. Diabetes and obesity are confirmed as major modifiable risk factors, particularly in postmenopausal women, where hyperinsulinemia and metabolic dysfunction contribute to increased incidence and poorer outcomes, notably in triple-negative breast cancer (TNBC). The role of MHT, particularly combined oestrogen-progestogen therapy, is strongly associated with increased breast cancer risk, particularly for hormone receptor-positive malignancies, necessitating cautious use and personalised treatment planning. In contrast, oestrogen-only MHT appears to confer a reduced risk in women post-hysterectomy. For patients with PCOS, CAH and Turner Syndrome, while definitive evidence of elevated breast cancer risk is lacking, individualised screening strategies and careful hormone therapy management remain essential due to the complex interplay of hormonal and metabolic factors. CONCLUSIONS The review highlights the need for personalised breast cancer screening and management protocols in women with endocrine and metabolic disorders. For high-risk groups such as MEN1 patients, early initiation of MRI screening is warranted. In women with diabetes and obesity, targeted interventions addressing hyperinsulinemia and metabolic dysfunction are critical to mitigating their increased cancer risk. The association between MHT and breast cancer underscores the importance of individualised risk stratification in hormone therapy administration, particularly in women with predisposing genetic or endocrine conditions. Enhanced surveillance tailored to the unique risk profiles of endocrine disorder patients will facilitate early detection and improve clinical outcomes. However, further large-scale studies are necessary to refine these associations and develop robust, evidence-based guidelines.
Collapse
Affiliation(s)
- Arie Hawazie
- Centre for Endocrinology, Queen Mary University, London, UK
| | - Maralyn Druce
- Centre for Endocrinology, Queen Mary University, London, UK
| |
Collapse
|
6
|
Chevillon F, Rebotier M, Dhédin N, Bruno B, Cacciatore C, Charbonnier A, Joseph L, Le Bourgeois A, Talouarn M, Magro L, Barraud Lange V. [Fertility preservation and hematopoietic stem cell transplantation (SFGM-TC)]. Bull Cancer 2025; 112:S24-S35. [PMID: 38918137 DOI: 10.1016/j.bulcan.2024.04.009] [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: 02/12/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 06/27/2024]
Abstract
Conditioning regimen prior to hematopoietic stem cell transplantation have an impact on patient fertility through the use of gonadal irradiation and/or bifunctional alkylating agents. Their impact on fertility depends mainly on the dose used and, in women, on age at the time of treatment. All patients should benefit before treatment from a consultation informing them of the potential impact on fertility and of fertility preservation techniques. In the absence of contraindications, the major toxicity of myeloablative conditioning regimen justifies fertility preservation. There are few data concerning fertility after reduced-intensity conditioning. Despite lower theoretical gonadotoxicity, we also recommend fertility preservation, if possible before transplantation. The fertility preservation techniques used depend on the patient's age, pathology and conditioning. In the event of subsequent use of harvested gonadal tissue in the context of acute leukemia or aggressive lymphoma, it is advisable to assess the risk of reintroduction of tumor cells. Finally, it is recommended to assess gonadal function after transplant, especially after reduced conditioning. If there is persistent residual gonadal function, post-treatment fertility preservation should be discuss.
Collapse
Affiliation(s)
- Florian Chevillon
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France.
| | - Marine Rebotier
- Service oncogynécologie, centre Leon-Berard et IHOPe, 28, promenade Léa et Napoléon Bullukian, 69008 Lyon, France
| | - Nathalie Dhédin
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - Bénédicte Bruno
- Service hématologie pédiatrique, hôpital Jeanne-de-Flandre, avenue Eugène-Avinée, 59037 Lille, France
| | - Carlotta Cacciatore
- Service de médecine interne, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | | | - Laure Joseph
- Service hématologie, département de biothérapie, hôpital Necker-enfants malades, AP-HP, 149, rue de Sèvres, 75015 Paris, France
| | - Amandine Le Bourgeois
- Service d'hématologie, CHU de Nantes, 1, place Alexis-Ricordeau, 44000 Nantes, France
| | - Marie Talouarn
- Service d'hématologie, hôpital Saint-Antoine, AP-HP, 184, rue du faubourg Saint-Antoine, 75012 Paris, France
| | - Leonardo Magro
- Service d'hématologie, CHU de Lille, 2, avenue Oscar-Lambret, 59000 Lille, France
| | - Virginie Barraud Lange
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service biologie de la reproduction, hôpital Cochin Port Royal, AP-HP, 123, boulevard de Port Royal, 75014 Paris, France
| |
Collapse
|
7
|
Pouladvand N, Azarnia M, Zeinali H, Fathi R, Tavana S. An overview of different methods to establish a murine premature ovarian failure model. Animal Model Exp Med 2024; 7:835-852. [PMID: 39219374 PMCID: PMC11680483 DOI: 10.1002/ame2.12477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/14/2024] [Indexed: 09/04/2024] Open
Abstract
Premature ovarian failure (POF)is defined as the loss of normal ovarian function before the age of 40 and is characterized by increased gonadotropin levels and decreased estradiol levels and ovarian reserve, often leading to infertility. The incomplete understanding of the pathogenesis of POF is a major impediment to the development of effective treatments for this disease, so the use of animal models is a promising option for investigating and identifying the molecular mechanisms involved in POF patients and developing therapeutic agents. As mice and rats are the most commonly used models in animal research, this review article considers studies that used murine POF models. In this review based on the most recent studies, first, we introduce 10 different methods for inducing murine POF models, then we demonstrate the advantages and disadvantages of each one, and finally, we suggest the most practical method for inducing a POF model in these animals. This may help researchers find the method of creating a POF model that is most appropriate for their type of study and suits the purpose of their research.
Collapse
Affiliation(s)
- Negar Pouladvand
- Department of Embryology, Reproductive Biomedicine Research CenterRoyan Institute for Reproductive Biomedicine, ACECRTehranIran
- Department of Animal Biology, Faculty of Biological SciencesKharazmi UniversityTehranIran
| | - Mahnaz Azarnia
- Department of Animal Biology, Faculty of Biological SciencesKharazmi UniversityTehranIran
| | - Hadis Zeinali
- Department of Animal Biology, Faculty of Biological SciencesKharazmi UniversityTehranIran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research CenterRoyan Institute for Reproductive Biomedicine, ACECRTehranIran
| | - Somayeh Tavana
- Department of Embryology, Reproductive Biomedicine Research CenterRoyan Institute for Reproductive Biomedicine, ACECRTehranIran
| |
Collapse
|
8
|
Sockel K, Neu A, Goeckenjan M, Ditschkowski M, Hilgendorf I, Kröger N, Ayuk FA, Stoelzel F, Middeke JM, Eder M, Bethge W, Finke J, Bertz H, Kobbe G, Kaufmann M, Platzbecker U, Beverungen D, Schmid C, von Bonin M, Egger-Heidrich K, Heberling L, Trautmann-Grill K, Teipel R, Bug G, Tischer J, Fraccaroli A, Fante M, Wolff D, Luft T, Winkler J, Schäfer-Eckart K, Scheid C, Holtick U, Klein S, Blau IW, Burchert A, Wulf G, Hasenkamp J, Schwerdtfeger R, Kaun S, Junghanss C, Wortmann F, Winter S, Neidlinger H, Theuser C, Beyersmann J, Bornhaeuser M, Schmeller S, Schetelig J. Hope for motherhood: pregnancy after allogeneic hematopoietic cell transplantation (a national multicenter study). Blood 2024; 144:1532-1542. [PMID: 39007722 DOI: 10.1182/blood.2024024342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 07/16/2024] Open
Abstract
ABSTRACT Improved long-term survival rates after allogeneic hematopoietic cell transplantation (alloHCT) make family planning for young adult cancer survivors an important topic. However, treatment-related infertility risk poses challenges. To assess pregnancy and birth rates in a contemporary cohort, we conducted a national multicenter study using data from the German Transplant Registry, focusing on adult women aged 18 to 40 years who underwent alloHCT between 2003 and 2018. Of 2654 women who underwent transplantation, 50 women experienced 74 pregnancies, occurring at a median of 4.7 years after transplant. Fifty-seven of these resulted in live births (77%). The annual first birth rate among HCT recipients was 0.45%, which is >6 times lower than in the general population. The probability of a live birth 10 years after HCT was 3.4%. Factors associated with an increased likelihood of pregnancy were younger age at alloHCT, nonmalignant transplant indications, no total body irradiation or a cumulative dose of <8 Gy, and nonmyeloablative/reduced-intensity conditioning. Notably, 72% of pregnancies occurred spontaneously, with assisted reproductive technologies used in the remaining cases. Preterm delivery and low birth weight were more common than in the general population. This study represents the largest data set reporting pregnancies in a cohort of adult female alloHCT recipients. Our findings underscore a meaningful chance of pregnancy in alloHCT recipients. Assisted reproductive technologies techniques are important and funding should be made available. However, the potential for spontaneous pregnancies should not be underestimated, and patients should be informed of the possibility of unexpected pregnancy despite reduced fertility. Further research is warranted to understand the impact of conditioning decisions on fertility preservation.
Collapse
Affiliation(s)
- Katja Sockel
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Annika Neu
- Altona Children's Hospital, Hamburg, Germany
| | - Maren Goeckenjan
- Department for Gynaecology and Obstetrics, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Markus Ditschkowski
- Department of Hematology and Stem Cell Transplantation, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Inken Hilgendorf
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Jena, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis A Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Stoelzel
- Division of Stem Cell Transplantation and Cellular Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jan Moritz Middeke
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Matthias Eder
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Wolfgang Bethge
- Department of Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Jürgen Finke
- Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Bertz
- Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Martin Kaufmann
- 2nd Department of Internal Medicine, Oncology, and Hematology, Robert Bosch Hospital, Stuttgart, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - David Beverungen
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Christoph Schmid
- Augsburg University Hospital and Medical Faculty and Bavarian Cancer Research Center, Augsburg, Germany
| | - Malte von Bonin
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Katharina Egger-Heidrich
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Lisa Heberling
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Karolin Trautmann-Grill
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Raphael Teipel
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Gesine Bug
- Department of Medicine, Hematology, and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Johanna Tischer
- Department of Medicine III, Ludwig Maximilian University, University Hospital of Munich, Munich, Germany
| | - Alessia Fraccaroli
- Department of Medicine III, Ludwig Maximilian University, University Hospital of Munich, Munich, Germany
| | - Matthias Fante
- Department of Internal Medicine III, Hematology, and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Daniel Wolff
- Department of Internal Medicine III, Hematology, and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Luft
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Heidelberg, Germany
| | - Julia Winkler
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Kerstin Schäfer-Eckart
- Department of Internal Medicine V, Nuremberg Hospital North, Paracelsus Medical University, Nuremberg, Germany
| | - Christof Scheid
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Stefan Klein
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Igor Wolfgang Blau
- Department of BMT, Clinic of Hematology, Oncology, and Tumor Immunology, Charite-University Medicine Berlin, Berlin, Germany
| | - Andreas Burchert
- Department of Hematology, Oncology, and Immunology, Carreras Leukemia Center, Philips University Marburg and University Hospital Gießen and Marburg, Marburg, Germany
| | - Gerald Wulf
- Hematology and Medical Oncology, University Medicine Göttingen, Göttingen, Germany
| | - Justin Hasenkamp
- Hematology and Medical Oncology, University Medicine Göttingen, Göttingen, Germany
| | - Rainer Schwerdtfeger
- Center for Hematopoietic Cell Transplantation, Deutsche Klinik für Diagnostik Helios Klinik, Wiesbaden, Germany
| | - Stephan Kaun
- Hematology, Oncology und Infectiology, Clinic Bremen-Mitte, Bremen, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Friederike Wortmann
- Department of Hematology and Oncology, Medical Center, University of Schleswig-Holstein, Lübeck, Germany
| | - Susann Winter
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Helga Neidlinger
- German Registry of Stem Cell Transplantation (DRST), Ulm, Germany
| | - Catrin Theuser
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | | | - Martin Bornhaeuser
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- National Center for Tumor Diseases, Partner Site Dresden, Dresden, Germany
| | | | - Johannes Schetelig
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| |
Collapse
|
9
|
Berkel C. Inducers and Inhibitors of Pyroptotic Death of Granulosa Cells in Models of Premature Ovarian Insufficiency and Polycystic Ovary Syndrome. Reprod Sci 2024; 31:2972-2992. [PMID: 39026050 PMCID: PMC11438836 DOI: 10.1007/s43032-024-01643-3] [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: 04/04/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024]
Abstract
Granulosa cells (GCs), the largest cell population and primary source of steroid hormones in the ovary, are the important somatic ovarian components. They have critical roles in folliculogenesis by supporting oocyte, facilitating its growth, and providing a microenvironment suitable for follicular development and oocyte maturation, thus having essential functions in maintaining female fertility and in reproductive health in general. Pyroptotic death of GCs and associated inflammation have been implicated in the pathogenesis of several reproductive disorders in females including Premature Ovarian Insufficiency (POI) and Polycystic Ovary Syndrome (PCOS). Here, I reviewed factors, either intrinsic or extrinsic, that induce or inhibit pyroptosis in GCs in various models of these disorders, both in vitro and in vivo, and also covered associated molecular mechanisms. Most of these studied factors influence NLRP3 inflammasome- and GSDMD (Gasdermin D)-mediated pyroptosis in GCs, compared to other inflammasomes and gasdermins (GSDMs). I conclude that a more complete mechanistic understanding of these factors in terms of GC pyroptosis is required to be able to develop novel strategies targeting inflammatory cell death in the ovary.
Collapse
Affiliation(s)
- Caglar Berkel
- Department of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Türkiye.
| |
Collapse
|
10
|
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
|
11
|
Weidlinger S, Graber S, Bratschi I, Pape J, Kollár A, Karrer T, von Wolff M. A Systematic Review of the Gonadotoxicity of Osteosarcoma and Ewing's Sarcoma Chemotherapies in Postpubertal Females and Males. J Adolesc Young Adult Oncol 2024; 13:597-606. [PMID: 38629685 PMCID: PMC11322626 DOI: 10.1089/jayao.2023.0185] [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] [Indexed: 07/18/2024] Open
Abstract
Data on gonadotoxicity of chemotherapies are essential to better counsel young females and males about the risk of infertility and to better indicate fertility preservation measures before cancer therapies. However, such data have not recently been reviewed for bone cancer. Therefore, a systematic literature search was conducted considering papers published since 2000. This study is part of the FertiTOX® project, which aims to improve the lack of data regarding gonadotoxicity of cancer therapies to enable more accurate counseling regarding fertility preservation. Only relapse-free women and men were included. Gonadotoxic therapy-induced suspected infertility was defined as very low anti-mullerian hormone, high gonadotropin concentration, amenorrhea, oligomenorrhea, azoospermia, or oligozoospermia. The quality of the individual studies was assessed using the Newcastle-Ottawa Scale (NOS). In total, 11 out of 831 studies were included in the review. Suspected infertility was found in 10/190 (5.1%, range 0%-66%) of female patients with osteosarcoma (six studies), in 24/46 (52.2%, range 46%-100%) of male patients with osteosarcoma (three studies), in 18/138 (13.0%, range 3%-18%) of female patients with Ewing's sarcoma (three studies), and in 34/38 (89.5%) of male patients with Ewing's sarcoma (one study). A risk calculation in relation to specific chemotherapies was not possible. Risk of suspected infertility tends to be higher in Ewing's sarcoma in which all patients received chemotherapies with alkylating agents. Two of the 11 included studies received a high NOS quality score, whereas the remaining nine studies received a low quality score, mainly because of the lack of a comparator group. Published data are too limited for precise estimation of the gonadotoxicity. However, data indicate clinically relevant risk for infertility, supporting counseling patients before chemotherapy about fertility preservation measures.
Collapse
Affiliation(s)
- Susanna Weidlinger
- Division of Gynecological Endocrinology and Reproductive Medicine, University Women’s Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Satu Graber
- Division of Gynecological Endocrinology and Reproductive Medicine, University Women’s Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Irina Bratschi
- Division of Gynecological Endocrinology and Reproductive Medicine, University Women’s Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Janna Pape
- Division of Gynecological Endocrinology and Reproductive Medicine, University Women’s Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Attila Kollár
- Department of Medical Oncology, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Tanya Karrer
- Medical Library, University Library Bern, University of Bern, Bern, Switzerland
| | - Michael von Wolff
- Division of Gynecological Endocrinology and Reproductive Medicine, University Women’s Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
12
|
Heras M, Alonso-Espias M, Arencibia O, Minig L, Marti L, Diestro MD, Cespedes J, Niguez I, Gil-Ibañez B, Diaz-Feijoo B, Llueca A, Rosado C, Iacoponi S, Lopez de la Manzanara C, Morales S, Fernandez-Galguera MJ, Cano A, Gorostidi M, Zapardiel I. Real Implication of Fertility-Sparing Surgery for Ovarian Cancer: Reproductive Outcomes. Diagnostics (Basel) 2024; 14:1424. [PMID: 39001314 PMCID: PMC11241223 DOI: 10.3390/diagnostics14131424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND to prove the effectivity of fertility-sparing procedures in early-stage ovarian cancer by assessing pregnancy rates and obstetrical outcomes. METHODS we performed a retrospective multicenter study among 55 Spanish hospitals, collecting patients from 18 to 40 years old with diagnosis of early-stage ovarian cancer, epithelial (EOC) or non-epithelial (non-EOC), from January 2010 to December 2019. Data on the use of assisted reproductive techniques, pregnancy attempts and obstetrical outcomes were collected. RESULTS a total of 150 patients met inclusion criteria, 70 (46.6%) EOC and 80 (53.4%) non-EOC. Pregnancy attempts were reported in 51 (34%) patients, with 42 (28%) patients carrying the pregnancy to term. Among them, 30 (71.4%) underwent surgery alone and 12 (28.6%) had additional postoperative chemotherapy. A total of 32 (76.1% patients) had spontaneous pregnancies and 10 (23.9%) required in vitro fertilization. There was only one (2.4%) complication reported. Vaginal delivery was reported in twenty-nine (69%) patients and cesarean section in five (11.9%) patients. CONCLUSIONS fertility-sparing management for ovarian cancer seems to be an option with proven good pregnancy rates and low complications. The selection of patients must consider strict criteria in order to maintain a good prognosis.
Collapse
Affiliation(s)
- Marta Heras
- Gynecology Department, Hospital Universitario Santa Cristina, 28009 Madrid, Spain
| | | | - Octavio Arencibia
- Gynecology Department, Hospital Universitario Materno Infantil, 06010 Las Palmas de Gran Canaria, Spain
| | - Lucas Minig
- Gynecology Department, IMED Hospitales, 46100 Valencia, Spain
| | - Lola Marti
- Gynecology Department, Hospital Universitario Bellvitge, 08907 Barcelona, Spain
| | | | - Juan Cespedes
- Gynecology Department, Hospital Universitario de Donostia, 20014 San Sebastian, Spain
| | - Isabel Niguez
- Gynecology Department, Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Blanca Gil-Ibañez
- Gynecology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Berta Diaz-Feijoo
- Institute Clinic of Gynecology, Obstetrics and Neotatology, Hospital Clinic de Barcelona, 08007 Barcelona, Spain
| | - Antoni Llueca
- Gynecology Department, Hospital de Castellón, 12004 Castellón, Spain
| | - Claudia Rosado
- Gynecology Department, Hospital de Mataró, 08304 Barcelona, Spain
| | - Sara Iacoponi
- Gynecology Department, Hospital Quirón Madrid, 28223 Madrid, Spain
| | | | - Sara Morales
- Gynecology Department, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
| | | | - Ana Cano
- Gynecology Department, Hospital Universitario Fundación Alcorcón, 28922 Madrid, Spain
| | - Mikel Gorostidi
- Gynecology Department, Hospital Universitario de Donostia, 20014 San Sebastian, Spain
| | - Ignacio Zapardiel
- Gynecologic Oncology Unit, La Paz University Hospital, 28046 Madrid, Spain
| |
Collapse
|
13
|
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
|
14
|
Mo X, Zhang Z, Song J, Wang Y, Yu Z. Self-assembly of peptides in living cells for disease theranostics. J Mater Chem B 2024; 12:4289-4306. [PMID: 38595070 DOI: 10.1039/d4tb00365a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
The past few decades have witnessed substantial progress in biomedical materials for addressing health concerns and improving disease therapeutic and diagnostic efficacy. Conventional biomedical materials are typically created through an ex vivo approach and are usually utilized under physiological environments via transfer from preparative media. This transfer potentially gives rise to challenges for the efficient preservation of the bioactivity and implementation of theranostic goals on site. To overcome these issues, the in situ synthesis of biomedical materials on site has attracted great attention in the past few years. Peptides, which exhibit remarkable biocompability and reliable noncovalent interactions, can be tailored via tunable assembly to precisely create biomedical materials. In this review, we summarize the progress in the self-assembly of peptides in living cells for disease diagnosis and therapy. After a brief introduction to the basic design principles of peptide assembly systems in living cells, the applications of peptide assemblies for bioimaging and disease treatment are highlighted. The challenges in the field of peptide self-assembly in living cells and the prospects for novel peptide assembly systems towards next-generation biomaterials are also discussed, which will hopefully help elucidate the great potential of peptide assembly in living cells for future healthcare applications.
Collapse
Affiliation(s)
- Xiaowei Mo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| | - Zeyu Zhang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| | - Jinyan Song
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| | - Yushi Wang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China.
- Haihe Laboratory of Synthetic Biology, 21 West 15th Avenue, Tianjin 300308, China
| |
Collapse
|
15
|
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
|
16
|
Link-Rachner CS, Göbel A, Jaschke NP, Rachner TD. Endocrine health in survivors of adult-onset cancer. Lancet Diabetes Endocrinol 2024; 12:350-364. [PMID: 38604215 DOI: 10.1016/s2213-8587(24)00088-3] [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: 06/19/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 04/13/2024]
Abstract
Long-term survivors of cancer (ie, the patient who is considered cured or for whom the disease is under long-term control and unlikely to recur) are at an increased risk of developing endocrine complications such as hypothalamic-pituitary dysfunctions, hypogonadisms, osteoporosis, or metabolic disorders, particularly when intensive tumour-directed therapies are applied. Symptom severity associated with these conditions ranges from mild and subclinical to highly detrimental, affecting individual health and quality of life. Although they are usually manageable, many of these endocrine pathologies remain underdiagnosed and untreated for years. To address this challenge, a higher degree of awareness, standardised screening tools, comprehensible treatment algorithms, and a close collaborative effort between endocrinologists and oncologists are essential to early identify patients who are at risk, and to implement appropriate treatment protocols. This Review highlights common symptoms and conditions related to endocrine disorders among survivors of adult-onset cancer, provides a summary of the currently available practice guidelines, and proposes a practical approach to diagnose affected patients among this group.
Collapse
Affiliation(s)
- Cornelia S Link-Rachner
- Division of Haematology and Oncology, Department of Medicine I, Technische Universität Dresden, Dresden, Germany
| | - Andy Göbel
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nikolai P Jaschke
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Tilman D Rachner
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
17
|
Hickey M, Basu P, Sassarini J, Stegmann ME, Weiderpass E, Nakawala Chilowa K, Yip CH, Partridge AH, Brennan DJ. Managing menopause after cancer. Lancet 2024; 403:984-996. [PMID: 38458217 DOI: 10.1016/s0140-6736(23)02802-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/18/2023] [Accepted: 12/11/2023] [Indexed: 03/10/2024]
Abstract
Globally, 9 million women are diagnosed with cancer each year. Breast cancer is the most commonly diagnosed cancer worldwide, followed by colorectal cancer in high-income countries and cervical cancer in low-income countries. Survival from cancer is improving and more women are experiencing long-term effects of cancer treatment, such as premature ovarian insufficiency or early menopause. Managing menopausal symptoms after cancer can be challenging, and more severe than at natural menopause. Menopausal symptoms can extend beyond hot flushes and night sweats (vasomotor symptoms). Treatment-induced symptoms might include sexual dysfunction and impairment of sleep, mood, and quality of life. In the long term, premature ovarian insufficiency might increase the risk of chronic conditions such as osteoporosis and cardiovascular disease. Diagnosing menopause after cancer can be challenging as menopausal symptoms can overlap with other common symptoms in patients with cancer, such as fatigue and sexual dysfunction. Menopausal hormone therapy is an effective treatment for vasomotor symptoms and seems to be safe for many patients with cancer. When hormone therapy is contraindicated or avoided, emerging evidence supports the efficacy of non-pharmacological and non-hormonal treatments, although most evidence is based on women older than 50 years with breast cancer. Vaginal oestrogen seems safe for most patients with genitourinary symptoms, but there are few non-hormonal options. Many patients have inadequate centralised care for managing menopausal symptoms after cancer treatment, and more information is needed about cost-effective and patient-focused models of care for this growing population.
Collapse
Affiliation(s)
- Martha Hickey
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and the Royal Women's Hospital, Melbourne, VIC, Australia.
| | - Partha Basu
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, WHO, Lyon, France
| | - Jenifer Sassarini
- Department of Obstetrics and Gynaecology, School of Gynaecology, University of Glasgow, Glasgow, UK
| | - Mariken E Stegmann
- Department of Primary and Long-term Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | | - Cheng-Har Yip
- Department of Surgery, University of Malaya, Kuala Lumpur, Malaysia
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Donal J Brennan
- Gynaecological Oncology Group, UCD School of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland; Systems Biology Ireland, UCD School of Medicine, Dublin, Ireland
| |
Collapse
|
18
|
Keefe KW, Lanes A, Stratton K, Green DM, Chow EJ, Oeffinger KC, Barton S, Diller L, Yasui Y, Leisenring WM, Armstrong GT, Ginsburg ES. Assisted reproductive technology use and outcomes in childhood cancer survivors. Cancer 2024; 130:128-139. [PMID: 37732943 PMCID: PMC10841316 DOI: 10.1002/cncr.34995] [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: 01/31/2023] [Revised: 05/29/2023] [Accepted: 06/15/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Treatment exposures for childhood cancer reduce ovarian reserve. However, the success of assisted reproductive technology (ART) among female survivors is not well established. METHODS Five-year survivors of childhood cancer in the Childhood Cancer Survivor Study were linked to the Society for Assisted Reproductive Technology Clinic Outcome Reporting System, which captures national ART outcomes. The authors assessed the live birth rate, the relative risk (RR) with 95% confidence intervals (95% CIs), and associations with treatment exposure using generalized estimating equations to account for multiple ovarian stimulations per individual. Siblings from a random sample of survivors were recruited to serve as a comparison group. RESULTS Among 9885 female survivors, 137 (1.4%; median age at diagnosis, 10 years [range, 0-20 years]; median years of follow-up after age 18 years, 11 years [range, 2-11 years]) underwent 224 ovarian stimulations using autologous or donor eggs and/or gestational carriers (157 autologous ovarian stimulation cycles, 67 donor ovarian stimulation cycles). In siblings, 33 (1.4%) underwent 51 autologous or donor ovarian stimulations. Of those who used embryos from autologous eggs without using gestational carriers, 97 survivors underwent 155 stimulations, resulting in 49 live births, for a 31.6% chance of live birth per ovarian stimulation (vs. 38.3% for siblings; p = .39) and a 43.9% chance of live birth per transfer (vs. 50.0%; p = .33). Prior treatment with cranial radiation therapy (RR, 0.44; 95% CI, 0.20-0.97) and pelvic radiation therapy (RR, 0.33; 95% CI, 0.15-0.73) resulted in a reduced chance of live birth compared with siblings. The likelihood of live birth after ART treatment in survivors was not affected by alkylator exposure (cyclophosphamide-equivalent dose, ≥8000 mg/m2 vs. none; RR, 1.04; 95% CI, 0.52-2.05). CONCLUSIONS Childhood cancer survivors are as likely to undergo treatment using ART as sibling controls. The success of ART treatment was not reduced after alkylator exposure. The results from the current study provide needed guidance on the use of ART in this population.
Collapse
Affiliation(s)
- Kimberly W Keefe
- Center for Infertility and Reproductive Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Andrea Lanes
- Center for Infertility and Reproductive Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Daniel M Green
- St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Eric J Chow
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Sara Barton
- Colorado Center for Reproductive Medicine, Denver, Colorado, USA
| | - Lisa Diller
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Yutaka Yasui
- St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | - Elizabeth S Ginsburg
- Center for Infertility and Reproductive Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Plotogea M, Zgura A, Mehedințu C, Scurtu F, Petca A, Varlas VN, Bors RG, Edu A, Ionescu OM, Andreescu M, Mateescu RN, Isam AJ. Women's Sexual Dysfunctions Following Stem Cell Transplant and the Impact on Couple Relationship. Life (Basel) 2023; 14:35. [PMID: 38255651 PMCID: PMC10817538 DOI: 10.3390/life14010035] [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: 11/06/2023] [Revised: 12/17/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Stem cell transplant proved its efficacy in increasing the survival rate among young patients diagnosed with hematological malignancies. A transplant conditioning regimen is particularly destructive on the genital system, often determining premature ovarian failure, accompanied by vulvovaginal atrophy and sexual dysfunctions. The aims of the present study were, first, to evaluate sexual dysfunctions among transplanted women, using clinical examination and the female sexual function index (FSFI), and second, to determine their impact on a couple's relationship. A prospective observational comparative study was performed and included 38 patients who underwent allogenic stem cell transplant (SCT) procedures for different hematological malignancies and 38 healthy patients (control group). This study included baseline evaluation, one-year, and three-year follow-up visits. In addition to anamnesis and medically obtained information, FSFI was evaluated to determine the impact of gynecological damage in a subjective manner. In the study group, vulvovaginal atrophy was diagnosed in 76.32%, with subsequent sexual dysfunctions in 92.10% of patients, based on FSFI scoring. Even though the results improved throughout the study, at the last visit, mild vulvovaginal atrophy was diagnosed in 81.58% of patients, and the FSFI score was abnormal for 21.05%. When compared to the control group, both sexual dysfunctions and FSFI results were considerably impaired, with statistical significance. There is a confirmed negative impact of sexual dysfunctions and self-declared FSFI on couple/marital status and couple relationships, with statistical significance, at the last visit. In conclusion, anatomical, functional, and psychological difficulties are a reality of long-term survivors after a stem cell transplant. They should be addressed and assessed equally to other medical conditions, as they may determine serious consequences and impact the sexual quality of life and the couple's relationship.
Collapse
Affiliation(s)
- Mihaela Plotogea
- Department of Obstetrics and Gynecology, “Nicolae Malaxa” Clinical Hospital, 022441 Bucharest, Romania
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Anca Zgura
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Claudia Mehedințu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Obstetrics and Gynecology, “Filantropia” Clinical Hospital, 011179 Bucharest, Romania
| | - Francesca Scurtu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Obstetrics and Gynecology, “Filantropia” Clinical Hospital, 011179 Bucharest, Romania
| | - Aida Petca
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Valentin Nicolae Varlas
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Obstetrics and Gynecology, “Filantropia” Clinical Hospital, 011179 Bucharest, Romania
| | - Roxana Georgiana Bors
- Department of Obstetrics and Gynecology, “Filantropia” Clinical Hospital, 011179 Bucharest, Romania
| | - Antoine Edu
- Department of Obstetrics and Gynecology, “Nicolae Malaxa” Clinical Hospital, 022441 Bucharest, Romania
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Oana-Maria Ionescu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihaela Andreescu
- Faculty of Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Radu Nicolae Mateescu
- Department of Obstetrics and Gynecology, “Nicolae Malaxa” Clinical Hospital, 022441 Bucharest, Romania
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Al Jashi Isam
- Faculty of Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| |
Collapse
|
20
|
Garrido Colino C, González Urdiales P, Molinés Honrubia A, Ortega Acosta MJ, García Abos M. Primary ovarian insufficiency in cancer survivors: Keys to optimal management. An Pediatr (Barc) 2023; 99:385-392. [PMID: 37993293 DOI: 10.1016/j.anpede.2023.11.007] [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: 05/22/2023] [Accepted: 09/24/2023] [Indexed: 11/24/2023] Open
Abstract
INTRODUCTION Primary ovarian insufficiency (POI) carries significant morbidity, causing infertility, sexual disfunction, decreased bone density, cardiovascular risk, emotional distress and early mortality. OBJECTIVE To know the incidence and current management of POI in childhood/adolescent solid tumour survivors. MATERIAL AND METHODS We conducted a multicentre observational study. It included female patients aged 12-18 years with a diagnosis of solid tumour and meeting clinical or biochemical criteria for POI. The risk was estimated based on the criteria of the Pediatric Initiative Network of the Oncofertility Consortium. RESULTS We found an incidence of 1.5 (30 cases of POI): The median age at the time of the event was 14 years (standard deviation, 2.09). The solid tumours associated most frequently with POI were Ewing sarcoma and brain and germ cell tumours. Eighty-three percent of patients did not undergo fertility preservation. Sixty-three percent reported not having undergone menarche at the time of ovarian failure. Ninety-seven percent were at high risk of gonadal toxicity, yet 47% were not monitored before the diagnosis. The median time elapsed to the occurrence of the event was 43.5 months after diagnosis and 29.5 months after completing treatment. The Kaplan-Meier curves showed that approximately 30% of POI cases developed within 2 years of diagnosis and that women at Tanner stage 1 developed insufficiency later than women at Tanner stage 5. CONCLUSIONS There is room for improvement in the follow-up of women at risk of POI in Spain. The tools currently available facilitate risk assessment at the time of treatment planning and allow the implementation of monitoring, education, early diagnosis, fertility preservation, and replacement therapy as needed. All of this would achieve significant improvement in health outcomes.
Collapse
Affiliation(s)
- Carmen Garrido Colino
- Servicio de Pediatría, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
| | | | - Antonio Molinés Honrubia
- Servicio de Hematología, Hospital Insular Las Palmas de Gran Canarias, Las Palmas de Gran Canaria, Las Palmas, Spain
| | | | - Mirian García Abos
- Servicio de Pediatría, Hospital Universitario Donostia, Donostia, Gipuzkoa, Spain
| |
Collapse
|
21
|
Roman E, Kane E, Smith A, Howell D, Sheridan R, Simpson J, Bonaventure A, Kinsey S. Cohort profile: the United Kingdom Childhood Cancer Study (UKCCS) - a UK-wide population-based study examining the health of cancer survivors. BMJ Open 2023; 13:e073712. [PMID: 37977872 PMCID: PMC10660444 DOI: 10.1136/bmjopen-2023-073712] [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: 03/14/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023] Open
Abstract
PURPOSE The United Kingdom Childhood Cancer Study's (UKCCS's) matched cohort was established to examine the longer term morbidity and mortality of individuals previously diagnosed with cancer before 15 years of age, comparing future healthcare patterns in 5-year cancer survivors to baseline activity seen in age- and sex-matched individuals from the general population. PARTICIPANTS Predicated on a national childhood cancer case-control study conducted in the early 1990s (4430 cases, 9753 controls) in England, Scotland and Wales, the case population comprises 3125 cancer survivors (>5 years), and the control population 7156 age- and sex-matched individuals from the general population who did not have cancer as a child. Participants are now being followed up via linkage to national administrative healthcare databases (deaths, cancers and secondary care hospital activity). FINDINGS TO DATE Enabling the creation of cohorts with minimal selection bias and loss to follow-up, the original case-control study registered all newly diagnosed cases of childhood cancer and their corresponding controls, regardless of their family's participation. Early findings based on the registered case population found marked survival variations with age and sex across subtypes and differences with deprivation among acute lymphoblastic leukaemia (ALL) survivors. More recently, comparing the health-activity patterns of the case and control populations revealed that survivors of childhood ALL experienced excess outpatient and inpatient activity across their teenage/young adult years. Adding to increased risks of cancer and death and involving most clinical specialties, excesses were not related to routine follow-up monitoring and showed no signs of diminishing over time. FUTURE PLANS With annual linkage updates, the UKCCS's maturing population-based matched cohorts provide the foundation for tracking the health of individuals through their lifetime. Comparing the experience of childhood cancer survivors to that of unaffected general-population counterparts, this will include examining subsequent morbidity and mortality, secondary care hospital activity and the impact of deprivation on longer term outcomes.
Collapse
Affiliation(s)
- Eve Roman
- Department of Health Sciences, University of York, York, North Yorkshire, UK
| | - Eleanor Kane
- Department of Health Sciences, University of York, York, North Yorkshire, UK
| | - Alexandra Smith
- Department of Health Sciences, University of York, York, North Yorkshire, UK
| | - Debra Howell
- Department of Health Sciences, University of York, York, North Yorkshire, UK
| | - Rebecca Sheridan
- Department of Health Sciences, University of York, York, North Yorkshire, UK
| | - Jill Simpson
- Department of Health Sciences, University of York, York, North Yorkshire, UK
| | - Audrey Bonaventure
- INSERM, Université Paris Cité and Université Sorbonne Paris Nord, Paris, Île-de-France, France
| | - Sally Kinsey
- Paediatric Haematology, Leeds Children's Hospital, Leeds, West Yorkshire, UK
| |
Collapse
|
22
|
Zhang S, Yahaya BH, Pan Y, Liu Y, Lin J. Menstrual blood-derived endometrial stem cell, a unique and promising alternative in the stem cell-based therapy for chemotherapy-induced premature ovarian insufficiency. Stem Cell Res Ther 2023; 14:327. [PMID: 37957675 PMCID: PMC10644549 DOI: 10.1186/s13287-023-03551-w] [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: 12/16/2022] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Chemotherapy can cause ovarian dysfunction and infertility since the ovary is extremely sensitive to chemotherapeutic drugs. Apart from the indispensable role of the ovary in the overall hormonal milieu, ovarian dysfunction also affects many other organ systems and functions including sexuality, bones, the cardiovascular system, and neurocognitive function. Although conventional hormone replacement therapy can partly relieve the adverse symptoms of premature ovarian insufficiency (POI), the treatment cannot fundamentally prevent deterioration of POI. Therefore, effective treatments to improve chemotherapy-induced POI are urgently needed, especially for patients desiring fertility preservation. Recently, mesenchymal stem cell (MSC)-based therapies have resulted in promising improvements in chemotherapy-induced ovary dysfunction by enhancing the anti-apoptotic capacity of ovarian cells, preventing ovarian follicular atresia, promoting angiogenesis and improving injured ovarian structure and the pregnancy rate. These improvements are mainly attributed to MSC-derived biological factors, functional RNAs, and even mitochondria, which are directly secreted or indirectly translocated with extracellular vesicles (microvesicles and exosomes) to repair ovarian dysfunction. Additionally, as a novel source of MSCs, menstrual blood-derived endometrial stem cells (MenSCs) have exhibited promising therapeutic effects in various diseases due to their comprehensive advantages, such as periodic and non-invasive sample collection, abundant sources, regular donation and autologous transplantation. Therefore, this review summarizes the efficacy of MSCs transplantation in improving chemotherapy-induced POI and analyzes the underlying mechanism, and further discusses the benefit and existing challenges in promoting the clinical application of MenSCs in chemotherapy-induced POI.
Collapse
Affiliation(s)
- Shenghui Zhang
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Ying Pan
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, , China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| |
Collapse
|
23
|
Antunes MB, Cardeal SP, Magalhães M, Vale-Fernandes E, Barreiro M, Sá R, Sousa M. Preservation of fertility in female patients with hematologic diseases. Blood Rev 2023; 62:101115. [PMID: 37562987 DOI: 10.1016/j.blre.2023.101115] [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: 04/11/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023]
Abstract
Recent developments of assisted reproduction techniques turned possible to avoid the infertility consequences of oncologic treatments, but fertility preservation (FP) has been somewhat neglected in women with hematologic diseases undergoing gonadotoxic treatments. For these specific cases, the current options for FP include the cryopreservation of embryos, mature oocytes and ovarian tissue, and oocyte in-vitro maturation. We intend to make patients and clinicians aware of this important and relevant issue, and provide hematologists, assisted reproduction physicians and patients, with updated tools to guide decisions for FP. The physicians of the units responsible for female FP should always be available to decide on the best-individualized FP option in strict collaboration with hematologists. With a wide range of options for FP tailored to each case, a greater level of training and information is needed among clinicians, so that patients proposed to gonadotoxic treatments can be previously advised for FP techniques in hematological conditions. ABBREVIATED ABSTRACT: Recent developments of assisted reproduction techniques turned possible to preserve the fertility of women with hematologic diseases undergoing gonadotoxic treatments. Current options for fertility preservation in women with hematologic diseases are presented. It is imperative to offer fertility preservation to all women before starting any gonadotoxic treatment and in some cases after treatment. Fertility preservation methods enable to later achieve the desired pregnancy.
Collapse
Affiliation(s)
- Marika Bini Antunes
- Department of Clinical Hematology, Centro Hospitalar Universitário do Porto, Largo do Professor Abel Salazar, 4099-001 Porto, Portugal; UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Sara Pinto Cardeal
- Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Manuel Magalhães
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Department of Oncology, Centro Hospitalar Universitário do Porto, Largo do Professor Abel Salazar, 4099-001 Porto, Portugal
| | - Emídio Vale-Fernandes
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Centro de Procriação Medicamente Assistida, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Largo da Maternidade, 4050-371, Porto, Portugal
| | - Márcia Barreiro
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Centro de Procriação Medicamente Assistida, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Largo da Maternidade, 4050-371, Porto, Portugal.
| | - Rosália Sá
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Mário Sousa
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
24
|
Emori C, Boucher Z, Bolcun-Filas E. CHEK2 signaling is the key regulator of oocyte survival after chemotherapy. SCIENCE ADVANCES 2023; 9:eadg0898. [PMID: 37862420 PMCID: PMC10588956 DOI: 10.1126/sciadv.adg0898] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 09/06/2023] [Indexed: 10/22/2023]
Abstract
Cancer treatments can damage the ovarian follicle reserve, leading to primary ovarian insufficiency and infertility among survivors. Checkpoint kinase 2 (CHEK2) deficiency prevents elimination of oocytes in primordial follicles in female mice exposed to radiation and preserves their ovarian function and fertility. Here, we demonstrate that CHEK2 also coordinates the elimination of oocytes after exposure to standard-of-care chemotherapy drugs. CHEK2 activates two downstream targets-TAp63 and p53-which direct oocyte elimination. CHEK2 knockout or pharmacological inhibition preserved ovarian follicle reserve after radiation and chemotherapy. However, the lack of specificity for CHEK2 among available inhibitors limits their potential for clinical development. These findings demonstrate that CHEK2 is a master regulator of the ovarian cellular response to damage caused by radiation and chemotherapy and warrant the development of selective inhibitors specific to CHEK2 as a potential avenue for ovario-protective treatments.
Collapse
Affiliation(s)
- Chihiro Emori
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan
| | - Zachary Boucher
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
25
|
Cui W, Rocconi RP, Thota R, Anderson RA, Bruinooge SS, Comstock IA, Denduluri N, Gassman A, Gralow J, Hutt KJ, Amiri-Kordestani L, Lambertini M, Leighton J, Lu KH, Mostoufi-Moab S, Pollastro T, Pradhan S, Saber H, Schenkel C, Spratt D, Wedam S, Phillips KA. Measuring ovarian toxicity in clinical trials: an American Society of Clinical Oncology research statement. Lancet Oncol 2023; 24:e415-e423. [PMID: 37797647 DOI: 10.1016/s1470-2045(23)00390-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 10/07/2023]
Abstract
Anticancer agents can impair ovarian function, resulting in premature menopause and associated long-term health effects. Ovarian toxicity is not usually adequately assessed in trials of anticancer agents, leaving an important information gap for patients facing therapy choices. This American Society of Clinical Oncology (ASCO) statement provides information about the incorporation of ovarian toxicity measures in trial design. ASCO recommends: (1) measurement of ovarian toxicity in relevant clinical trials of anticancer agents that enrol post-pubertal, pre-menopausal patients; (2) collection of ovarian function measures at baseline and at 12-24 months after anticancer agent cessation, as a minimum, and later in line with the trial schedule; and (3) assessment of both clinical measures and biomarkers of ovarian function. ASCO recognises that routine measurement of ovarian toxicity and function in cancer clinical trials will add additional complexity and burden to trial resources but asserts that this issue is of such importance to patients that it cannot continue to be overlooked.
Collapse
Affiliation(s)
- Wanda Cui
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Rodney P Rocconi
- The University of Mississippi Medical Center, Cancer Center & Research Institute, Jackson, MS, USA
| | | | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | | - Ioanna A Comstock
- Office of Rare Diseases, Pediatrics, Urologic and Reproductive Medicine, Division of Urology, Obstetrics, and Gynecology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Audrey Gassman
- Office of Rare Diseases, Pediatrics, Urologic and Reproductive Medicine, Division of Urology, Obstetrics, and Gynecology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Julie Gralow
- American Society of Clinical Oncology, Alexandria, VA, USA
| | - Karla J Hutt
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Laleh Amiri-Kordestani
- Division of Oncology 1, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties, School of Medicine, University of Genova, Genoa, Italy; Department of Medical Oncology, UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - John Leighton
- Division of Hematology Oncology Toxicity, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sogol Mostoufi-Moab
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Shan Pradhan
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Haleh Saber
- Division of Hematology Oncology Toxicity, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Daniel Spratt
- Moffitt Cancer Center and Research Institute, Cleveland, OH, USA
| | - Suparna Wedam
- Division of Oncology 1, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Kelly-Anne Phillips
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
26
|
Song A, Zhang S, Zhao X, Wu S, Qi X, Gao S, Qi J, Li P, Tan J. Exosomes derived from menstrual blood stromal cells ameliorated premature ovarian insufficiency and granulosa cell apoptosis by regulating SMAD3/AKT/MDM2/P53 pathway via delivery of thrombospondin-1. Biomed Pharmacother 2023; 166:115319. [PMID: 37573658 DOI: 10.1016/j.biopha.2023.115319] [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: 06/19/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023] Open
Abstract
Premature ovarian insufficiency (POI) is clinically irreversible and seriously damages female fertility. We previously demonstrated that menstrual blood stromal cells (MenSCs)-derived exosomes (EXOs) effectively improved ovarian functions in the POI rat model. In this study, we investigated whether TSP1 is the key component in EXOs to ameliorate ovarian functions and further explored the molecular mechanism of EXOs in improving granulosa cell (GCs) activities. Our results demonstrated that knockdown TSP1 significantly debilitated the therapeutic effect of EXOs on estrous cyclicity, ovarian morphology, follicle numbers and pregnancy outcomes in 4-vinylcyclohexene diepoxide (VCD) induced POI rat model. In addition, EXOs treatment significantly promoted the activities and inhibited the apoptosis of VCD induced granulosa cells in vitro. Moreover, EXOs stimulation markedly activated the phosphorylation of SMAD3(Ser425) and AKT(Ser473), up-regulated the expressions of BCL2 and MDM2 as well as down-regulated the expressions of CASPASE3, CASPASE8, P53 and BAX. All these effects were supressed by SIS3, a inhibitor of TGF1/SMAD3. Our study revealed the key role of TSP1 in EXOs in improving POI pathology, restoring ovarian functions and GCs activities, andprovided a promising basis for EXOs in the treatment of ovarian dysfunction.
Collapse
Affiliation(s)
- Aixin Song
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China
| | - Siwen Zhang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China
| | - Xinyang Zhao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China
| | - Shanshan Wu
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China
| | - Xiaohan Qi
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China
| | - Shan Gao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China
| | - Jiarui Qi
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China
| | - Pingping Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China
| | - Jichun Tan
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang 110022, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China.
| |
Collapse
|
27
|
Abstract
Breast cancer survivors may experience significant after effects from diagnoses of breast cancer and cancer directed therapies. This review synthesizes the evidence about optimal management of the sequelae of a diagnosis of breast cancer. It describes the side effects of chemotherapy and endocrine therapy and evidence based strategies for management of such effects, with particular attention to effects of therapies with curative intent. It includes strategies to promote health and wellness among breast cancer survivors, along with data to support the use of integrative oncology strategies. In addition, this review examines models of survivorship care and ways in which digital tools may facilitate communication between clinicians and patients. The strategies outlined in this review are paramount to supporting breast cancer survivors' quality of life.
Collapse
|
28
|
Pu X, Zhang L, Zhang P, Xu Y, Wang J, Zhao X, Dai Z, Zhou H, Zhao S, Fan A. Human UC-MSC-derived exosomes facilitate ovarian renovation in rats with chemotherapy-induced premature ovarian insufficiency. Front Endocrinol (Lausanne) 2023; 14:1205901. [PMID: 37564988 PMCID: PMC10411896 DOI: 10.3389/fendo.2023.1205901] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/05/2023] [Indexed: 08/12/2023] Open
Abstract
Premature ovarian insufficiency (POI) induced by chemotherapy is an intractable disorder with a considerable incidence that commonly results in insufficient fertility and concomitant complications in female patients. Due to limitations in the current progress in POI diagnosis and treatment, there is an urgent need to develop novel remedies to improve ovarian function and protect fertility. The ameliorative effect of human umbilical cord mesenchymal stem cells (hUCMSCs) and exosomes derived from them in POI treatment could be a new hope for patients. Herein, we identified exosomes from hUCMSCs (hUCMSC-Exos). Then, systematic infusion of hUCMSC-Exos was accomplished via tail intravenous injection to investigate the feasibility of the treatment of rats with chemotherapy-induced POI by intraperitoneal injection of cyclophosphamide (CTX) and busulfan (BUS). Ovarian functions in the indicated group were evaluated, including oestrous cycle, serum sex hormone levels, follicle counts, ovarian pathological changes, proliferation and apoptosis of granulosa cells (GCs), and reproductive ability testing. Furthermore, the potential influence of hUCMSC-Exos on ovarian tissues was illuminated by conducting RNA-seq and multifaceted bioinformatics analyses. POI rats with hUCMSC-Exos transplantation exhibited a decrease in follicle-stimulating hormone (FSH) and apoptosis of GCs but an increase in oestradiol (E2), anti-Müllerian hormone (AMH), and the number of ovarian follicles and foetuses in the uterus. And the immunomodulation- and cellular vitality-associated gene sets in rats had also undergone moderate changes. Our data indicated the feasibility of hUCMSC-Exos in improving ovarian function and protecting fertility in chemotherapy-induced POI rats. HUCMSC-Exos can improve the local microenvironment of ovarian tissue in POI rats by participating in immune regulation, cellular viability, inflammation regulation, fibrosis and metabolism, and other related signal pathways.
Collapse
Affiliation(s)
- Xiaodi Pu
- Department of Obstetrics and Gynecology, Guizhou Medical University, Guiyang, China
| | - Leisheng Zhang
- Stem Cell Bank of Guizhou Province, Guizhou Health-Biotech Biotechnology Co., Ltd., Guiyang, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, China
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Radiation Technology and Biophysics, Hefei Institute of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Peiyu Zhang
- Department of Obstetrics and Gynecology, Guizhou Medical University, Guiyang, China
| | - Yaqiong Xu
- Department of Obstetrics and Gynecology, Guizhou Medical University, Guiyang, China
| | - Jun Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology of the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiaomei Zhao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology of the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhihua Dai
- Stem Cell Bank of Guizhou Province, Guizhou Health-Biotech Biotechnology Co., Ltd., Guiyang, China
| | - Hua Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology of the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shuyun Zhao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology of the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Anran Fan
- Key Laboratory of Reproductive Medicine, Stem Cell and Tissue Engineering Research Center in Guizhou Province, Guizhou Medical University, Guiyang, China
| |
Collapse
|
29
|
Bentsen L, Pappot H, Hjerming M, Hanghøj S. Thoughts about fertility among female adolescents and young adults with cancer: a qualitative study. Support Care Cancer 2023; 31:421. [PMID: 37357225 PMCID: PMC10290964 DOI: 10.1007/s00520-023-07887-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 06/13/2023] [Indexed: 06/27/2023]
Abstract
PURPOSE Nine hundred female adolescents and young adults (AYAs) aged 15-39 are diagnosed with cancer in Denmark annually. Advances in cancer therapy have led to increased long-term survival; however, a serious side effect of cancer therapy is reduced fertility. The aim of our study was to explore the thoughts about fertility among female AYAs with cancer. METHODS Our study was conducted from September 2020 to March 2021 at the Copenhagen University Hospital, Rigshospitalet. Inclusion criteria were female AYAs with cancer aged 18-39. Twelve individual, semi-structured, qualitative interviews were performed with female AYAs with cancer (20-35 years). Data were analysed using thematic analysis. RESULTS Four main themes were found: (1) the female AYAs held on to a hope of having children in the future; (2) the female AYAs experienced time pressure and waiting time as a sprint as well as a marathon; (3) the female AYAs faced existential and ethical choices about survival and family formation; and (4) the female AYAs felt a loss of control of their bodies. CONCLUSION Our study contributes with knowledge on how important holding on to the hope of children in the future is among female AYAs with cancer. Meanwhile, they are frustrated by the rushed decision on fertility preservation at diagnosis. The female AYAs also have existential and ethical concerns related to the choice of cancer therapy and fertility preservation. Finally, they suffer from altered body image, loss of femininity, and body control due to hormone therapy.
Collapse
Affiliation(s)
- Line Bentsen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Helle Pappot
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Maiken Hjerming
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Signe Hanghøj
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
30
|
Gao Y, Wu T, Tang X, Wen J, Zhang Y, Zhang J, Wang S. Increased cellular senescence in doxorubicin-induced murine ovarian injury: effect of senolytics. GeroScience 2023; 45:1775-1790. [PMID: 36648735 PMCID: PMC10400526 DOI: 10.1007/s11357-023-00728-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
Ovarian injury caused by chemotherapy can lead to early menopause, infertility, and even premature senility in female cancer patients, impairing the quality of life and overall health of the cancer survivors seriously. However, there is still a lack of effective protection strategies against such injury. Cellular senescence can be induced by chemotherapeutic agents in multiple organs and may corrode the structure and function of normal tissues. We hypothesized that the widely used first-line chemotherapy drug, doxorubicin, could increase senescent cell burden in normal ovarian tissue during the therapeutic process and that elimination of senescent cells with senolytics would ameliorate doxorubicin-induced ovarian injury. Here, we demonstrated an accumulation of cellular senescence in doxorubicin-treated ovaries through detecting p16 and p21 expression levels and senescence-associated β-galactosidase (SA-β-gal) activity as well as senescence-associated secretory phenotype (SASP) factors. Short-term intervention with the classic senolytic combination dasatinib and quercetin (DQ) or fisetin significantly reduced the load of senescent cells in ovaries after doxorubicin treatment. However, neither DQ nor fisetin alleviated doxorubicin-related ovarian dysfunction. Further experiments showed that ovarian apoptosis and fibrosis following doxorubicin exposure could not be improved by senolytics. Collectively, our study shows that senolytic treatment can eliminate accumulated senescent cells, but cannot reverse the massive follicle loss and ovarian stromal fibrosis caused by doxorubicin, suggesting that cellular senescence may not be one of the key mechanisms in doxorubicin-induced ovarian injury.
Collapse
Affiliation(s)
- Yueyue Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, Hubei, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, Hubei, China
| | - Xianan Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, Hubei, China
| | - Jingyi Wen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, Hubei, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, Hubei, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, Hubei, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, Hubei, China.
| |
Collapse
|
31
|
Su C, Zhang R, Zhang X, Lv M, Liu X, Ao K, Hao J, Mu YL. Dingkun Pill modulate ovarian function in chemotherapy-induced premature ovarian insufficiency mice by regulating PTEN/PI3K/AKT/FOXO3a signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023:116703. [PMID: 37257704 DOI: 10.1016/j.jep.2023.116703] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dingkun Pill (DKP) is a traditional Chinese medicine that has been shown to have beneficial effects on reproductive function. However, the specific mechanism underlying its effect on POI is not well understood. AIM OF THE STUDY To investigate the effect of different doses of Dingkun Pill on ovarian function in cyclophosphamide (CTX)-induced premature ovarian insufficiency (POI) mice and to explore its molecular mechanism through PTEN/PI3K/AKT/FOXO3a signaling pathway. This study will provide valuable insights into the potential clinical application of Dingkun Pill for the treatment of POI. MATERIALS AND METHODS Fifty female ICR mice were randomly divided into normal control (NC) group, model control (MC) group, and Dingkun Pill low, medium, high dose (DKP-L, M, H) groups. Mice were injected with CTX to construct the POI model. Mice in the DKP-L, M, and H groups were given 0.9 g/kg, 1.8 g/kg, and 3.6 g/kg of Dingkun Pill suspension for 21 days, respectively. Mice in the NC and MC groups were given the same amount of normal saline by gavage. Changes in body weight, estrous cycle and gonadal index were observed in each group of mice. Serum levels of FSH, LH, E2 and AMH were detected by ELISA. Hematoxylin-eosin (HE) staining observed the changes of ovarian pathological morphology and follicle counts at all levels. qRT-PCR was used to measure the levels of the PTEN and FOXO3a genes in ovarian tissue. The expression of PTEN/PI3K/AKT/FOXO3a signaling pathway related proteins were detected by Western-blot and immunohistochemistry(IHC). RESULTS In POI mice, Dingkun Pill increased body weight, promoted the recovery of estrous cycle, increased ovarian index, and improved pathological morphology of the ovaries. The FSH level decreased in the medium dose group (P < 0.05), the LH level reduced significantly in the medium and high dose groups (P < 0.01), and the E2 level in the high dose group increased (P < 0.05). There was no significant difference in AMH levels across all dose groups. The number of growing follicles improved at all levels in the low and medium dose groups, but declined significantly in the high dose group. However, the number of corpus luteum increased significantly in the high dose group (P < 0.001), and the atretic follicles in the three dose groups decreased. Results from qRT-PCR, Western-blot and IHC showed that the moderate dose of Dingkun Pill suppressed the levels of the p-PI3K and p-AKT proteins by upregulating the expression of PTEN in the ovarian tissues of POI mice, thereby inhibiting the expression of the key protein p-FOXO3a. However, the inhibitory effect of the higher dose may be less than that of the lower and intermediate dose groups. CONCLUSIONS The Dingkun Pill modulated hormonal levels, promoted follicle growth and induced ovulation in mice with CTX-induced POI, with better results in the low and moderate dose groups. Its mechanism may be related to the regulation of the PTEN/PI3K/AKT/FOXO3a signaling pathway.
Collapse
Affiliation(s)
- Chan Su
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China; The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Ruihong Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan, 250012, China
| | - Xiujuan Zhang
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China
| | - Mengxiao Lv
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China
| | - Xiang Liu
- The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Kai Ao
- The Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250013, China
| | - Jing Hao
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Medicine, Shandong University, Jinan, 250012, China.
| | - Yu-Lan Mu
- Department of Gynecology, Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, 250098, China.
| |
Collapse
|
32
|
Himpe J, Lammerant S, Van den Bergh L, Lapeire L, De Roo C. The Impact of Systemic Oncological Treatments on the Fertility of Adolescents and Young Adults-A Systematic Review. Life (Basel) 2023; 13:life13051209. [PMID: 37240854 DOI: 10.3390/life13051209] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Over the past decades, advancements in oncological treatments have led to major improvements in survival. Particularly for adolescents and young adults (AYAs), fertility is an important concern in cancer survivorship. The purpose of the review is to provide physicians with a practical overview of the current knowledge about the impact of systemic oncological treatments on the fertility of female and male AYAs. METHODS A systematic review was performed based on relevant articles obtained from 4 databases up until 31 December 2022. RESULTS The mechanisms of gonadotoxicity and the concurrent risk is described for the following categories: chemotherapy, targeted therapy and immunotherapy. For the category "chemotherapy", the specific effects and risks are listed for the different classes and individual chemotherapeutics. In the category "targeted therapy", a distinction was made between tyrosine kinase inhibitors (TKIs) and monoclonal antibodies. Information concerning immunotherapy is scarce. CONCLUSIONS The effects of chemotherapy on fertility are well investigated, but even in this category, results can be conflicting. Insufficient data are available on the fertility effects of targeted therapy and immunotherapy to draw definitive conclusions. More research is needed for these therapies and their evolving role in treating cancers in AYAs. It would be useful to include fertility endpoints in clinical trials that evaluate new and existing oncological treatments.
Collapse
Affiliation(s)
- Justine Himpe
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Sander Lammerant
- Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lore Van den Bergh
- Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lore Lapeire
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- AYA Research Centre and Hub (ARCH), Ghent University, 9000 Ghent, Belgium
| | - Chloë De Roo
- AYA Research Centre and Hub (ARCH), Ghent University, 9000 Ghent, Belgium
- Department of Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
33
|
Ma H, Wang Y, Liu G, Hu Q, Zhu J, Dai Y. Ovarian scaffolds promoted mouse ovary recovery from cyclophosphamide damage. J Reprod Immunol 2023; 157:103950. [PMID: 37079974 DOI: 10.1016/j.jri.2023.103950] [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: 11/13/2022] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/22/2023]
Abstract
There is growing evidence to suggest that scaffold of tissue can promote the tissue reparation. In this study, we investigate the effects of ovarian scaffolds on the reparation of cyclophosphamide (CPA) damaged mice ovaries. The mice were first administered with CPA, was then either transplanted an ovarian scaffold into each ovarian bursa for the experimental group (EG) or underwent sham surgery as the control (CG). To evaluate the extent of ovarian damage caused by CPA, a third group which did not undergo any treatment was included for the normal control (NG). Their ovaries were harvested for examination at day 30, 60, and 90 post CPA injection. We found that in EG, the number of all types of follicles in the ovaries remained almost the same throughout. The numbers of follicles were not significantly different from CG, except at day 60, where in CG the numbers of each type of follicle decreased to basal levels. The decrease in the number of ovarian follicles at day 60 in CG was mirrored by the significant increase in the number of apoptotic granulosa cells in the follicles, and was corroborated further by the basal levels of serum estradiol. Furthermore, we observed a significant decrease in collagen composition preceded by macrophage polarization, and elevation of inflammatory cytokine expression in the ovaries of the EG compared to the CG at day 60. We concluded that ovarian scaffolds can effectively protect primordial follicles from CPA-damage and promote the reparation of CPA-damaged ovaries. This research establishes a proof of concept for the future treatment of chemo-damaged ovaries.
Collapse
Affiliation(s)
- Hongmeng Ma
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Yuxing Wang
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Gang Liu
- Key Laboratory of Medical Cell Biology, Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, Inner Mongolia, China
| | - Qike Hu
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Jie Zhu
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Yanfeng Dai
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China.
| |
Collapse
|
34
|
Abd Karim NA, Adam AHB, Jaafaru MS, Rukayadi Y, Abdull Razis AF. Apoptotic Potential of Glucomoringin Isothiocyanate (GMG-ITC) Isolated from Moringa oleifera Lam Seeds on Human Prostate Cancer Cells (PC-3). Molecules 2023; 28:molecules28073214. [PMID: 37049977 PMCID: PMC10096378 DOI: 10.3390/molecules28073214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 04/14/2023] Open
Abstract
Inhibition of several protein pathways involved in cancer cell regulation is a necessary key in the discovery of cancer chemotherapy. Moringa oleifera Lam is often used in traditional medicine for the treatment of various illnesses. The plant contains glucomoringin isothiocyanate (GMG-ITC) with therapeutic potential against various cancer cells. Therefore, GMG-ITC was evaluated for its cytotoxicity against the PC-3 prostate cancer cell line and its potential to induce apoptosis. GMG-ITC inhibited cell proliferation in the PC-3 cell line with IC50 value 3.5 µg/mL. Morphological changes as a result of GMG-ITC-induced apoptosis showed chromatin condensation, nuclear fragmentation, and membrane blebbing. Additionally, Annexin V assay showed proportion of cells in early and late apoptosis upon exposure to GMG-ITC in a time-dependent manner. Moreover, GMG-ITC induced a time-dependent G2/M phase arrest, with reduction of 39.1% in the PC-3 cell line. GMG-ITC also activates apoptotic genes including caspase, tumor suppressor gene (p53), Akt/MAPK, and Bax of the proapoptotic Bcl family. Early apoptosis proteins (JNK, Bad, Bcl2, and p53) were significantly upregulated upon GMG-ITC treatment. It is concluded that apoptosis induction was observed in PC-3 cells treated with GMG-ITC. These phenomena suggest that GMG-ITC from M. oleifera seeds could be useful as a future cytotoxic agent against prostate cancer.
Collapse
Affiliation(s)
- Nurul Ashikin Abd Karim
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Aziza Hussein Bakheit Adam
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Department of Food Hygiene and Safety, Faculty of Public and Environmental Health, University of Khartoum, Khartoum 11111, Sudan
| | - Mohammed Sani Jaafaru
- Medical Analysis Department, Faculty of Science, Tishk International University, Erbil 44001, Iraq
| | - Yaya Rukayadi
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Ahmad Faizal Abdull Razis
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Laboratory of Food Safety and Food Integrity, Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Serdang 43400, Malaysia
| |
Collapse
|
35
|
Cavestro GM, Mannucci A, Balaguer F, Hampel H, Kupfer SS, Repici A, Sartore-Bianchi A, Seppälä TT, Valentini V, Boland CR, Brand RE, Buffart TE, Burke CA, Caccialanza R, Cannizzaro R, Cascinu S, Cercek A, Crosbie EJ, Danese S, Dekker E, Daca-Alvarez M, Deni F, Dominguez-Valentin M, Eng C, Goel A, Guillem JG, Houwen BBSL, Kahi C, Kalady MF, Kastrinos F, Kühn F, Laghi L, Latchford A, Liska D, Lynch P, Malesci A, Mauri G, Meldolesi E, Møller P, Monahan KJ, Möslein G, Murphy CC, Nass K, Ng K, Oliani C, Papaleo E, Patel SG, Puzzono M, Remo A, Ricciardiello L, Ripamonti CI, Siena S, Singh SK, Stadler ZK, Stanich PP, Syngal S, Turi S, Urso ED, Valle L, Vanni VS, Vilar E, Vitellaro M, You YQN, Yurgelun MB, Zuppardo RA, Stoffel EM. Delphi Initiative for Early-Onset Colorectal Cancer (DIRECt) International Management Guidelines. Clin Gastroenterol Hepatol 2023; 21:581-603.e33. [PMID: 36549470 PMCID: PMC11207185 DOI: 10.1016/j.cgh.2022.12.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND & AIMS Patients with early-onset colorectal cancer (eoCRC) are managed according to guidelines that are not age-specific. A multidisciplinary international group (DIRECt), composed of 69 experts, was convened to develop the first evidence-based consensus recommendations for eoCRC. METHODS After reviewing the published literature, a Delphi methodology was used to draft and respond to clinically relevant questions. Each statement underwent 3 rounds of voting and reached a consensus level of agreement of ≥80%. RESULTS The DIRECt group produced 31 statements in 7 areas of interest: diagnosis, risk factors, genetics, pathology-oncology, endoscopy, therapy, and supportive care. There was strong consensus that all individuals younger than 50 should undergo CRC risk stratification and prompt symptom assessment. All newly diagnosed eoCRC patients should receive germline genetic testing, ideally before surgery. On the basis of current evidence, endoscopic, surgical, and oncologic treatment of eoCRC should not differ from later-onset CRC, except for individuals with pathogenic or likely pathogenic germline variants. The evidence on chemotherapy is not sufficient to recommend changes to established therapeutic protocols. Fertility preservation and sexual health are important to address in eoCRC survivors. The DIRECt group highlighted areas with knowledge gaps that should be prioritized in future research efforts, including age at first screening for the general population, use of fecal immunochemical tests, chemotherapy, endoscopic therapy, and post-treatment surveillance for eoCRC patients. CONCLUSIONS The DIRECt group produced the first consensus recommendations on eoCRC. All statements should be considered together with the accompanying comments and literature reviews. We highlighted areas where research should be prioritized. These guidelines represent a useful tool for clinicians caring for patients with eoCRC.
Collapse
Affiliation(s)
- Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesc Balaguer
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), University of Barcelona, Barcelona, Spain
| | - Heather Hampel
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | - Sonia S Kupfer
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago Medicine, Chicago, Illinois
| | - Alessandro Repici
- Gastrointestinal Endoscopy Unit, Humanitas University, Humanitas Research Hospital, Rozzano, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, and Department of Hematology Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Toni T Seppälä
- Faculty of Medicine and Medical Technology, University of Tampere and TAYS Cancer Centre, Arvo Ylpön katu, Tampere, Finland; Unit of Gastroenterological Surgery, Tampere University Hospital, Elämänaukio, Tampere, Finland; Applied Tumor Genomics Research Program and Department of Surgery, Helsinki University and Helsinki University Hospital, Helsinki, Finland
| | - Vincenzo Valentini
- Department of Radiology, Radiation Oncology and Hematology, Università Cattolica del Sacro Cuore di Roma, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Rome, Italy
| | - Clement Richard Boland
- Department of Medicine, Division of Gastroenterology, University of California San Diego, San Diego, California
| | - Randall E Brand
- Division of Gastroenterology, Hepatology & Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tineke E Buffart
- Department of Medical Oncology. Amsterdam UMC, Location de Boelelaan, Amsterdam, The Netherlands
| | - Carol A Burke
- Department of Gastroenterology, Hepatology and Nutrition, Cleveland Clinic, Cleveland, Ohio
| | - Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Renato Cannizzaro
- SOC Gastroenterologia Oncologica e Sperimentale Centro di Riferimento Oncologico di Aviano (CRO) IRCCS 33081, Aviano, Italy
| | - Stefano Cascinu
- Oncology Department, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emma J Crosbie
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, United Kingdom; Division of Gynaecology, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Silvio Danese
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Evelien Dekker
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Maria Daca-Alvarez
- Department of Gastroenterology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Francesco Deni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mev Dominguez-Valentin
- Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Cathy Eng
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Ajay Goel
- Department of Molecular Diagnostics & Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, California
| | - Josè G Guillem
- Department of Surgery and Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Britt B S L Houwen
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Charles Kahi
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Matthew F Kalady
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center and the Vagelos College of Physicians and Surgeons, New York, New York
| | - Florian Kühn
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Luigi Laghi
- Department of Medicine and Surgery, University of Parma, Parma, and Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Andrew Latchford
- Lynch Syndrome Clinic, Centre for Familial Intestinal Cancer, St Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, United Kingdom
| | - David Liska
- Department of Colorectal Surgery and Edward J. DeBartolo Jr Family Center for Young-Onset Colorectal Cancer, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Patrick Lynch
- Department of Gastroenterology, M. D. Anderson Cancer Center, Houston, Texas
| | - Alberto Malesci
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianluca Mauri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, and Department of Hematology Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Elisa Meldolesi
- Department of Radiology, Radiation Oncology and Hematology, Università Cattolica del Sacro Cuore di Roma, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Rome, Italy
| | - Pål Møller
- Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
| | - Kevin J Monahan
- Lynch Syndrome Clinic, Centre for Familial Intestinal Cancer, St Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, United Kingdom; Faculty of Medicine, Department of Surgery & Cancer, Imperial College, London, United Kingdom
| | - Gabriela Möslein
- Surgical Center for Hereditary Tumors, Ev. BETHESDA Khs. Duisburg, Academic Hospital University of Düsseldorf, Düsseldorf, Germany
| | - Caitlin C Murphy
- School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| | - Karlijn Nass
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Kimmie Ng
- Young-Onset Colorectal Cancer Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Cristina Oliani
- Medical Oncology, AULSS 5 Polesana, Santa Maria Della Misericordia Hospital, Rovigo, Italy
| | - Enrico Papaleo
- Centro Scienze della Natalità, Department of Obstetrics and Gynecology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Swati G Patel
- University of Colorado Anschutz Medical Center and Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Marta Puzzono
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Remo
- Pathology Unit, Mater Salutis Hospital, ULSS9, Legnago, Verona, Italy
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, Universita degli Studi di Bologna, Bologna, Italy
| | - Carla Ida Ripamonti
- Department of Onco-Haematology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, and Department of Hematology Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Satish K Singh
- Department of Medicine, Section of Gastroenterology, VA Boston Healthcare System and Boston University, Boston, Massachusetts
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter P Stanich
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Sapna Syngal
- Brigham and Women's Hospital, Harvard Medical School, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Stefano Turi
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuele Damiano Urso
- Chirurgia Generale 3, Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), University Hospital of Padova, Padova, Italy
| | - Laura Valle
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, Bellvitge Biomedical Research Center (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Madrid, Spain
| | - Valeria Stella Vanni
- Centro Scienze della Natalità, Department of Obstetrics and Gynecology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marco Vitellaro
- Unit of Hereditary Digestive Tract Tumours, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Yi-Qian Nancy You
- Department of Colon & Rectal Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matthew B Yurgelun
- Brigham and Women's Hospital, Harvard Medical School, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Raffaella Alessia Zuppardo
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena M Stoffel
- Division of Gastroenterology and Hepatology, Department of Internal Medicine and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
36
|
Liu X, Song Y, Zhou F, Zhang C, Li F, Hu R, Ma W, Song K, Tang Z, Zhang M. Network and experimental pharmacology on mechanism of Si-Wu-tang improving ovarian function in a mouse model of premature ovarian failure induced by cyclophosphamide. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115842. [PMID: 36265674 DOI: 10.1016/j.jep.2022.115842] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Si-Wu-Tang (SWT) has become a common basic prescription for supplementing blood and regulating menstruation, and enjoys the reputation of "the first prescription in gynecology". It is often reported in the treatment of premature ovarian failure (POF). However, knowledge of its specific mechanism is still limited. AIM OF THE STUDY This study aimed to identify the potential effects and underlying mechanisms of SWT on POF. MATERIALS AND METHODS After confirming the therapeutic effect of SWT on POF mice induced by cyclophosphamide, we further clarified the promoting effect of SWT on ovarian follicle development by detecting the expression of key factors related to follicle development in the ovary in different ways.Then, network pharmacology and gene expression profiling of POF from the GEO database were used to clarify the underlying mechanisms. Molecular biology and molecular docking analysis were applied for final mechanism verification. RESULTS Our results showed that SWT increased body weight, ovarian index, reversed disordered serum hormone levels, and menstrual cycle in POF mice. After SWT treatment, the number of follicles at all levels in mice with POF also recovered. Using molecular biology techniques, it was proven that SWT can improve follicle development and angiogenesis in the microenvironment. The network pharmacology and gene expression profiling from the GEO database indicated that the PI3K/Akt signaling pathway may be the reason why SWT improves ovarian function in mice with POF. Subsequently, further Western blot and immunoprecipitation indicated that SWT indeed inhibited the PI3K/Akt signaling pathway in mice with POF. In addition, this conclusion was further confirmed by molecular docking experiments. CONCLUSIONS SWT can improve ovarian function in POF mice induced by cyclophosphamide, and its mechanism is related to the inhibition of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xia Liu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yufan Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fanru Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fan Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Runan Hu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wenwen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Kunkun Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Mingmin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
37
|
Chen Y, Zhao Y, Miao C, Yang L, Wang R, Chen B, Zhang Q. Quercetin alleviates cyclophosphamide-induced premature ovarian insufficiency in mice by reducing mitochondrial oxidative stress and pyroptosis in granulosa cells. J Ovarian Res 2022; 15:138. [PMID: 36572950 PMCID: PMC9793602 DOI: 10.1186/s13048-022-01080-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Exposure to cyclophosphamide (CTX) induces premature ovarian insufficiency (POI). Quercetin is a natural flavonoid that exhibits anti-inflammatory and antioxidant properties, and its antioxidant activity is correlated with POI. However, the mechanism underlying its protective role in CTX-induced ovarian dysfunction is unclear. This study aimed to explore whether quercetin can protect ovarian reserves by activating mitochondrial biogenesis and inhibiting pyroptosis. METHODS Thirty-six female C57BL/6 mice were randomly subdivided into six groups. Except for the control group, all groups were injected with 90 mg/kg CTX to establish a POI model and further treated with coenzyme 10 or various doses of quercetin. The mice were sacrificed 48 h after 10 IU pregnant mare serum gonadotropin was injected four weeks after treatments. We used enzyme-linked immunosorbent assays to detect serum hormone expression and light and transmission electron microscopy to assess ovarian tissue morphology and mitochondria. Additionally, we tested oxidant and antioxidant levels in ovarian tissues and mitochondrial function in granulosa cells (GCs). The expression of mitochondrial biogenesis and pyroptosis-related proteins and mRNA was analyzed using western blotting and RT-qPCR. RESULTS Quercetin elevated serum anti-Müllerian hormone, estradiol, and progesterone levels, decreased serum follicle-stimulating hormone and luteinizing hormone levels, and alleviated ovarian pathology. It reduced the mitochondrial DNA content and mitochondrial membrane potential. Furthermore, it upregulated ATP levels and the mRNA and protein expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), mitochondrial transcription factor A, and superoxide dismutase 2. In addition, it suppressed NOD-like receptor pyrin domain containing 3, caspase-1, interleukin-1β, and gasdermin D levels in the GCs of POI mice. CONCLUSIONS Quercetin protected the ovarian reserve from CTX-induced ovarian damage by reversing mitochondrial dysfunction and activating mitochondrial biogenesis via the PGC1-α pathway. Moreover, quercetin may improve ovarian functions by downregulating pyroptosis in the CTX-induced POI model. Thus, quercetin can be considered a potential agent for treating POI.
Collapse
Affiliation(s)
- Yun Chen
- grid.268505.c0000 0000 8744 8924Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Xihu District, Hangzhou, 310007 Zhejiang Province China
| | - Ying Zhao
- grid.268505.c0000 0000 8744 8924Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Xihu District, Hangzhou, 310007 Zhejiang Province China
| | - Chenyun Miao
- grid.268505.c0000 0000 8744 8924Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Xihu District, Hangzhou, 310007 Zhejiang Province China
| | - Liuqing Yang
- grid.268505.c0000 0000 8744 8924Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Xihu District, Hangzhou, 310007 Zhejiang Province China
| | - Ruye Wang
- grid.268505.c0000 0000 8744 8924Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Xihu District, Hangzhou, 310007 Zhejiang Province China
| | - Bixia Chen
- grid.268505.c0000 0000 8744 8924Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Xihu District, Hangzhou, 310007 Zhejiang Province China
| | - Qin Zhang
- grid.268505.c0000 0000 8744 8924Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Xihu District, Hangzhou, 310007 Zhejiang Province China
| |
Collapse
|
38
|
Wang Z, Guo X, Hao L, Zhang X, Lin Q, Sheng R. Charge-Convertible and Reduction-Sensitive Cholesterol-Containing Amphiphilic Copolymers for Improved Doxorubicin Delivery. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6476. [PMID: 36143789 PMCID: PMC9504105 DOI: 10.3390/ma15186476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/06/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
For achieving successful chemotherapy against cancer, designing biocompatible drug delivery systems (DDSs) with long circulation times, high cellular endocytosis efficiency, and targeted drug release is of upmost importance. Herein, a well-defined PEG-b-P(MASSChol-co-MANBoc) block copolymer bearing redox-sensitive cholesteryl-side group was prepared via reversible addition-fragmentation chain transfer (RAFT) polymerization (with non-redox PEG-b-P(MACCChol-co-MAN-DCA) as the reference), and 1,2-dicarboxylic-cyclohexene acid (DCA) was then grafted onto the hydrophobic block to endow it with charge-convertible characteristics under a tumor microenvironment. The amphiphilic copolymer could be assembled into polymeric spherical micelles (SSMCs) with polyethylene glycol (PEG) as the corona/shell, and anti-cancer drug doxorubicin (DOX) was successfully encapsulated into the micellar core via strong hydrophobic and electrostatic interactions. This nanocarrier showed high stability in the physiological environment and demonstrated "smart" surface charge conversion from negative to positive in the slightly acidic environment of tumor tissues (pH 6.5~6.8), as determined by dynamic light scattering (DLS). Moreover, the cleavage of a disulfide bond linking the cholesterol grafts under an intracellular redox environment (10 mM GSH) resulted in micellar dissociation and accelerated drug release, with the non-redox-responsive micelles (CCMCs) as the control. Additionally, a cellular endocytosis and tumor proliferation inhibition study against MCF-7 tumor cells demonstrated the enhanced endocytosis and tumor cell inhibitory efficiency of dual-responsive SSMCs/DOX nanomedicines, revealing potentials as multifunctional nanoplatforms for effective oncology treatment.
Collapse
Affiliation(s)
- Zhao Wang
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing 211169, China
| | - Xinyu Guo
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing 211169, China
| | - Lingyun Hao
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing 211169, China
| | - Xiaojuan Zhang
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing 211169, China
| | - Qing Lin
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing 211169, China
| | - Ruilong Sheng
- CQM-Centro de Quimica da Madeira, Campus da Penteada, Universidade da Madeira, 9000390 Funchal, Madeira, Portugal
| |
Collapse
|
39
|
Rives N, Courbière B, Almont T, Kassab D, Berger C, Grynberg M, Papaxanthos A, Decanter C, Elefant E, Dhedin N, Barraud-Lange V, Béranger MC, Demoor-Goldschmidt C, Frédérique N, Bergère M, Gabrel L, Duperray M, Vermel C, Hoog-Labouret N, Pibarot M, Provansal M, Quéro L, Lejeune H, Methorst C, Saias J, Véronique-Baudin J, Giscard d'Estaing S, Farsi F, Poirot C, Huyghe É. What should be done in terms of fertility preservation for patients with cancer? The French 2021 guidelines. Eur J Cancer 2022; 173:146-166. [PMID: 35932626 DOI: 10.1016/j.ejca.2022.05.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 11/03/2022]
Abstract
AIM To provide practice guidelines about fertility preservation (FP) in oncology. METHODS We selected 400 articles after a PubMed review of the literature (1987-2019). RECOMMENDATIONS Any child, adolescent and adult of reproductive age should be informed about the risk of treatment gonadotoxicity. In women, systematically proposed FP counselling between 15 and 38 years of age in case of treatment including bifunctional alkylating agents, above 6 g/m2 cyclophosphamide equivalent dose (CED), and for radiation doses on the ovaries ≥3 Gy. For postmenarchal patients, oocyte cryopreservation after ovarian stimulation is the first-line FP technique. Ovarian tissue cryopreservation should be discussed as a first-line approach in case of treatment with a high gonadotoxic risk, when chemotherapy has already started and in urgent cases. Ovarian transposition is to be discussed prior to pelvic radiotherapy involving a high risk of premature ovarian failure. For prepubertal girls, ovarian tissue cryopreservation should be proposed in the case of treatment with a high gonadotoxic risk. In pubertal males, sperm cryopreservation must be systematically offered to any male who is to undergo cancer treatment, regardless of toxicity. Testicular tissue cryopreservation must be proposed in males unable to cryopreserve sperm who are to undergo a treatment with intermediate or severe risk of gonadotoxicity. In prepubertal boys, testicular tissue preservation is: - recommended for chemotherapy with a CED ≥7500 mg/m2 or radiotherapy ≥3 Gy on both testicles. - proposed for chemotherapy with a CED ≥5.000 mg/m2 or radiotherapy ≥2 Gy. If several possible strategies, the ultimate choice is made by the patient.
Collapse
Affiliation(s)
- Nathalie Rives
- Normandie Univ, UNIROUEN, Team "Adrenal and Gonadal Physiopathology" Inserm U1239 Nordic, Rouen University Hospital, Biology of Reproduction-CECOS Laboratory, Rouen, France
| | - Blandine Courbière
- Reproductive Medicine and Biology Department, Assistance Publique Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Thierry Almont
- Cancerology, Urology, Hematology Department, Centre Hospitalier Universitaire de Martinique, Fort-de-France, Martinique, France; General Cancer Registry of Martinique UF1441, Centre Hospitalier Universitaire de Martinique, Fort-de-France, Martinique, France
| | - Diana Kassab
- Methodology Unit, Association Française d'Urologie, Paris, Ile-de-France, France
| | - Claire Berger
- Department of Pediatric Hematology and Oncology, University-Hospital of Saint-Etienne, Hospital, Nord Saint-Etienne cedex 02, France 42055; Childhood Cancer Registry of the Rhône-Alpes Region, University of Saint-Etienne, 15 rue Ambroise Paré, Saint-Etienne cedex 02, France 42023
| | - Michaël Grynberg
- Reproductive Medicine and Fertility Department, Hôpital Antoine-Beclère, Clamart, Île-de-France, France
| | - Aline Papaxanthos
- Reproductive Medicine and Biology Department, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, Aquitaine, France
| | - Christine Decanter
- Medically Assisted Procreation and Fertility Preservation Department, Centre Hospitalier Régional Universitaire de Lille, Lille, Hauts-de-France, France
| | - Elisabeth Elefant
- Reference Center for Teratogenic Agents, Hôpital Armand-Trousseau Centre de Référence sur les Agents Tératogènes, Paris, Île-de-France, France
| | - Nathalie Dhedin
- Adolescents and Young Adults Unit, Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris, France
| | - Virginie Barraud-Lange
- Reproductive Medicine and Biology Department, Hôpital Cochin, Paris, Île-de-France, France
| | | | | | - Nicollet Frédérique
- Information and Promotion Department, Association Laurette Fugain, Paris, France
| | - Marianne Bergère
- Human Reproduction, Embryology and Genetics Directorate, Agence de la biomédecine, La Plaine Saint-Denis, France
| | - Lydie Gabrel
- Good Practices Unit - Guidelines and Medicines Directorate, Institut National du Cancer, Billancourt, Île-de-France, France
| | - Marianne Duperray
- Guidelines and Drug Directorate, Institut National du Cancer, Billancourt, Île-de-France, France
| | - Christine Vermel
- Expertise Quality and Compliance Mission - Communication and Information Directorate, Institut National du Cancer, Billancourt, Île-de-France, France
| | - Natalie Hoog-Labouret
- Research and Innovation, Institut National du Cancer, Billancourt, Île-de-France, France
| | - Michèle Pibarot
- OncoPaca-Corse Regional Cancer Network, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Magali Provansal
- Medical Oncology Department, Institut Paoli-Calmettes, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Laurent Quéro
- Cancerology and Radiotherapy Department, Hôpital Saint Louis, AP-HP, Paris, France
| | - Hervé Lejeune
- Reproductive Medicine and Biology Department, Hospices Civils de Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Charlotte Methorst
- Reproductive Medicine and Biology Department, Centre Hospitalier des Quatre Villes - Site de Saint-Cloud, Saint-Cloud, France
| | - Jacqueline Saias
- Reproductive Medicine and Biology Department, Assistance Publique Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Jacqueline Véronique-Baudin
- Cancerology, Urology, Hematology Department, Centre Hospitalier Universitaire de Martinique, Fort-de-France, Martinique, France; General Cancer Registry of Martinique UF1441, Centre Hospitalier Universitaire de Martinique, Fort-de-France, Martinique, France
| | - Sandrine Giscard d'Estaing
- Reproductive Medicine and Biology Department, Hospices Civils de Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Fadila Farsi
- Regional Cancer Network, Réseau Espace Santé Cancer, Lyon, Rhône-Alpes, France
| | - Catherine Poirot
- Adolescents and Young Adults Unit, Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris, France
| | - Éric Huyghe
- Urology Department, Centre Hospitalier Universitaire de Toulouse, Toulouse, France; Laboratoire Développement Embryonnaire, Fertilité et Environnement (DEFE) UMR 1203, Université Toulouse 3 Paul Sabatier, Toulouse, France.
| |
Collapse
|
40
|
van der Perk MEM, Cost NG, Bos AME, Brannigan R, Chowdhury T, Davidoff AM, Daw NC, Dome JS, Ehrlich P, Graf N, Geller J, Kalapurakal J, Kieran K, Malek M, McAleer MF, Mullen E, Pater L, Polanco A, Romao R, Saltzman AF, Walz AL, Woods AD, van den Heuvel-Eibrink MM, Fernandez CV. White paper: Onco-fertility in pediatric patients with Wilms tumor. Int J Cancer 2022; 151:843-858. [PMID: 35342935 PMCID: PMC9541948 DOI: 10.1002/ijc.34006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 11/18/2022]
Abstract
The survival of childhood Wilms tumor is currently around 90%, with many survivors reaching reproductive age. Chemotherapy and radiotherapy are established risk factors for gonadal damage and are used in both COG and SIOP Wilms tumor treatment protocols. The risk of infertility in Wilms tumor patients is low but increases with intensification of treatment including the use of alkylating agents, whole abdominal radiation or radiotherapy to the pelvis. Both COG and SIOP protocols aim to limit the use of gonadotoxic treatment, but unfortunately this cannot be avoided in all patients. Infertility is considered one of the most important late effects of childhood cancer treatment by patients and their families. Thus, timely discussion of gonadal damage risk and fertility preservation options is important. Additionally, irrespective of the choice for preservation, consultation with a fertility preservation (FP) team is associated with decreased patient and family regret and better quality of life. Current guidelines recommend early discussion of the impact of therapy on potential fertility. Since most patients with Wilms tumors are prepubertal, potential FP methods for this group are still considered experimental. There are no proven methods for FP for prepubertal males (testicular biopsy for cryopreservation is experimental), and there is just a single option for prepubertal females (ovarian tissue cryopreservation), posing both technical and ethical challenges. Identification of genetic markers of susceptibility to gonadotoxic therapy may help to stratify patient risk of gonadal damage and identify patients most likely to benefit from FP methods.
Collapse
Affiliation(s)
| | - Nicholas G Cost
- Department of Surgery, Division of Urology, University of Colorado School of Medicine and the Surgical Oncology Program of the Children's Hospital Colorado, Aurora, CO, USA
| | - Annelies M E Bos
- University Medical Center Utrecht, Reproductive Medicine and Gynaecology, Utrecht, Netherlands
| | - Robert Brannigan
- Department of Urology, Northwestern University, Chicago, Illinois, USA
| | - Tanzina Chowdhury
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, USA
| | - Najat C Daw
- Department of Pediatrics - Patient Care, MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey S Dome
- Division of Oncology at Children's National Hospital, Washington, DC, USA
| | - Peter Ehrlich
- University of Michigan, C.S. Mott Children's Hospital Section of Pediatric Surgery, Ann Arbor, MI, USA
| | - Norbert Graf
- Department for Pediatric Oncology and Hematology, Saarland University Medical Center, Homburg, Germany
| | - James Geller
- Division of Pediatric Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - John Kalapurakal
- Department of Radiation Oncology, Northwestern University, Chicago, Illinois, USA
| | - Kathleen Kieran
- Department of Urology, University of Washington, and Division of Urology, Seattle Children's Hospital, Seattle, USA
| | - Marcus Malek
- Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Mary F McAleer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth Mullen
- Department of Pediatric Oncology, Children's Hospital Boston/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Luke Pater
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Angela Polanco
- National Cancer Research Institute Children's Group Consumer Representative, London, UK
| | - Rodrigo Romao
- Departments of Surgery and Urology, IWK Health Centre, Dalhousie University, Halifax, Canada
| | | | - Amy L Walz
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
| | - Andrew D Woods
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | | | - Conrad V Fernandez
- Department of Pediatric Hematology/Oncology, IWK Health Centre and Dalhousie University, Halifax, Canada
| |
Collapse
|
41
|
Zidi S, Stayoussef M, Sontini FK, Mezlini A, Yacoubi-Loueslati B, Almawi WY. Decreased risk of ovarian cancer associated with rs9898876 sex hormone-binding globulin gene variant. Mol Biol Rep 2022; 49:4537-4544. [PMID: 35277784 DOI: 10.1007/s11033-022-07297-1] [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: 11/21/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Ovarian cancer (OC) is one of the most common gynecologic cancers,with significant morbidity and mortality. The risk of OC is influenced by hormone status, of which sex hormone-binding globulin (SHBG), which influences the serum availability of steroid sex hormones, is implicated in the pathogenesis and evolution of OC. The aim of this study is to evaluate the involvement of common SHBG gene variants in OC susceptibility and evolution. MATERIALS A case control study including 71 OC patients and 74 cancer-free controls, who were genotyped for rs9898876, rs13894, rs1799941 and rs6257 SHBG SNP. Genotyping was done by the allelic discrimination method, using VIC- and FAM-labeled primers. RESULTS The minor allele frequencies of rs9898876, rs13894, rs1799941 and rs6257 SHBG SNP was comparable between OC cases and control women, implying no significant associations of the tested variants and overall OC risk. Taking homozygous wild-type genotype as reference (OR = 1.00), heterozygous rs9898876 (G/T), and minor allele-carrying genotypes [G/T + T/T] were associated with reduced risk of OC. While rs9898876 heterozygosity (G/T) was predictive of OC occurrence, no significant association of the remaining three tested SNPs was noted with altered risk of OC. Irrespective of FIGO staging, the four tested SHBG SNPs were not associated with the clinical progression of OC. CONCLUSIONS In conclusion, SHBG rs9898876 is associated with a decreased risk of OC, and thus constitutes a potential diagnostic biomarker of OC.
Collapse
Affiliation(s)
- Sabrina Zidi
- Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, 1060, Tunis, Tunisia
| | - Mouna Stayoussef
- Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, 1060, Tunis, Tunisia
| | | | - Amel Mezlini
- Salah Azaiez Institute of Oncology, Tunis, Tunisia
| | - Besma Yacoubi-Loueslati
- Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, 1060, Tunis, Tunisia
| | - Wassim Y Almawi
- Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, 1060, Tunis, Tunisia. .,Department of Biomedical Sciences, Nazarbayev University School of Medicine, Z05K4F4, Nur-Sultan, Kazakhstan.
| |
Collapse
|
42
|
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
|
43
|
Female Oncofertility: Current Understandings, Therapeutic Approaches, Controversies, and Future Perspectives. J Clin Med 2021; 10:jcm10235690. [PMID: 34884393 PMCID: PMC8658080 DOI: 10.3390/jcm10235690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Recent advances in early detection and oncological therapies have ameliorated the survival rate of young cancer patients. Yet, ovarian impairment induced by chemotherapy and radiotherapy is still a challenging issue. This review, based on clinical and lab-based studies, summarizes the evidence of gonadotoxicity of chemoradiotherapy, the recent approaches, ongoing controversies, and future perspectives of fertility preservation (FP) in female patients who have experienced chemo- or radio-therapy. Existing data indicate that chemotherapeutic agents induce DNA alterations and massive follicle activation via the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Meanwhile, the radiation causes ionizing damage, leading to germ cell loss. In addition to the well-established methods, numerous therapeutic approaches have been suggested, including minimizing the follicle loss in cryopreserved ovarian grafts after transplantation, in vitro activation or in vitro growing of follicles, artificial ovarian development, or fertoprotective adjuvant to prevent ovarian damage from chemotherapy. Some reports have revealed positive outcomes from these therapies, whereas others have demonstrated conflictions. Future perspectives are improving the live birth rate of FP, especially in patients with adverse ovarian reserve, eliminating the risk of malignancy reintroducing, and increasing society’s awareness of FP importance.
Collapse
|
44
|
Liu JJ, De Vivo I, Wu CY, Giovannucci E. Subsequent primary urogenital cancers among childhood and adolescent cancer survivors in the United States. Urol Oncol 2021; 40:65.e11-65.e18. [PMID: 34810078 DOI: 10.1016/j.urolonc.2021.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/10/2021] [Accepted: 10/21/2021] [Indexed: 11/15/2022]
Abstract
PURPOSE To conduct an updated and comprehensive study on the risks of subsequent primary urogenital cancers for childhood and adolescent cancer survivors. METHODS This longitudinal study was conducted using 9 cancer registries from the Surveillance, Epidemiology and End Results (SEER) Program with follow-up from 1975 to 2017. There were 43,991 patients diagnosed with first primary cancer from 1975 to 2016 before the age of 20 years who subsequently survived for at least 1 year. Standardized incidence ratios (SIRs) and absolute excess risks (AERs) for urogenital cancers were calculated. RESULTS Compared with the general population, the risk of urinary system cancer was significantly higher in both female (SIR = 5.18, 95% CI: 3.65-7.14) and male (SIR = 2.80, 95% CI: 1.94-3.92) survivors of any first cancer, with shorter median interval length between first cancer and subsequent urinary system cancer for male survivors (19.9 years) than female survivors (29.3 years). Females also had significantly higher SIR than males for subsequent urinary system cancer (SIRfemale:male=1.86, 95% CI: 1.13-3.03) and kidney cancer (SIRfemale:male = 1.97, 95% CI: 1.11-3.53). Compared with the general population, females with any first cancer had significantly higher risks for cancers of the corpus uteri (SIR = 2.32, 95% CI: 1.49-3.45) and vulva (SIR = 4.27, 95% CI: 1.38-9.95). CONCLUSIONS Childhood and adolescent cancer survivors may have greater female susceptibility for developing subsequent urinary system and kidney cancers, and these survivors may have higher risks for specific types of reproductive system cancers. Our findings may lead to better awareness and surveillance for urogenital cancer by these cancer survivors and their physicians.
Collapse
Affiliation(s)
- Jason J Liu
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Chun-Ying Wu
- Division of Translational Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Edward Giovannucci
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
45
|
Johns B, Ficken M, Engberg M, Wecker L, Philpot R. Increasing dietary choline attenuates spatial memory deficits resulting from exposure to the chemotherapeutic agents cyclophosphamide and doxorubicin. J Psychopharmacol 2021; 35:1300-1309. [PMID: 34225528 PMCID: PMC9152698 DOI: 10.1177/02698811211029752] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Choline supplementation (+Ch) improves cognitive function in impaired animals and humans. Chemotherapy-related cognitive deficits (CRCDs) occur in cancer patients, and these deficits persist following treatment, adversely impacting quality of life. To date, there are no approved treatments for this condition. AIM Because +Ch improves impaired memory, it was of interest to determine whether +Ch can attenuate spatial memory deficits induced by the chemotherapeutic agents doxorubicin (DOX) and cyclophosphamide (CYP). METHODS Female BALB/C mice, 64 days of age, were trained in the Morris water maze and baseline performance determined on day 15. Following baseline assessment, mice were placed on +Ch diet (2.0% Ch) or remained on standard diet (0.12% Ch). Mice received intravenous injections of DOX (2.5 mg/kg) and CYP (25 mg/kg), or equivalent volumes of saline (0.9% NaCl), on days 16, 23, 30, and 37, and spatial memory was assessed weekly from day 22 to 71. RESULTS DOX and CYP produced a prolonged impairment in spatial memory as indicated by an increased latency to the correct zone (p < 0.05), and a decrease in time in the correct zone (p < 0.05), % of total swim distance in the correct zone (p < 0.05) and % entries to the correct zone (p < 0.05). These effects were attenuated by +Ch. CONCLUSION Although it remains to be determined whether this effect extends to other cognitive domains and whether +Ch is prophylactic or therapeutic, these findings suggest that +Ch may be an effective intervention for CRCDs.
Collapse
Affiliation(s)
| | | | | | | | - R.M. Philpot
- Corresponding Author: Rex M. Philpot, Ph.D., Assistant Professor, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, 3515 E Fletcher Ave., Tampa, FL 33612,
| |
Collapse
|
46
|
Ryder-Burbidge C, Diaz RL, Barr RD, Gupta S, Nathan PC, McKillop SJ, Fidler-Benaoudia MM. The Burden of Late Effects and Related Risk Factors in Adolescent and Young Adult Cancer Survivors: A Scoping Review. Cancers (Basel) 2021; 13:4870. [PMID: 34638350 PMCID: PMC8508204 DOI: 10.3390/cancers13194870] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022] Open
Abstract
Risk factors associated with late effects in survivors of adolescent and young adult (AYA) cancer are poorly understood. We conducted a systematic scoping review to identify cohort studies published in English from 2010-2020 that included: (1) cancer survivors who were AYAs (age 15-39 years) at diagnosis and (2) outcomes of subsequent malignant neoplasms (SMNs), chronic conditions, and/or late mortality (>5 years postdiagnosis). There were 652 abstracts identified and, ultimately, 106 unique studies were included, of which 23, 34, and 54 studies related to the risk of SMNs, chronic conditions, and mortality, respectively. Studies investigating late effects among survivors of any primary cancer reported that AYA cancer survivors were at higher risk of SMN, chronic conditions, and all-cause mortality compared to controls. There was an indication that the following factors increased risk: radiation exposure (n = 3) for SMNs; younger attained age (n = 4) and earlier calendar period of diagnosis (n = 3) for chronic conditions; and non-Hispanic Black or Hispanic (n = 5), low socioeconomic status (n = 3), and earlier calendar period of diagnosis (n = 4) for late mortality. More studies including the full AYA age spectrum, treatment data, and results stratified by age, sex, and cancer type are needed to advance knowledge about late effects in AYA cancer survivors.
Collapse
Affiliation(s)
- Charlotte Ryder-Burbidge
- Cancer Care Alberta, Alberta Health Services, Holy Cross Centre, Department of Cancer Epidemiology and Prevention Research, 5th Floor, BOX ACB, 2210-2 St. SW, Calgary, AB T2S 3C3, Canada; (C.R.-B.); (R.L.D.)
| | - Ruth L. Diaz
- Cancer Care Alberta, Alberta Health Services, Holy Cross Centre, Department of Cancer Epidemiology and Prevention Research, 5th Floor, BOX ACB, 2210-2 St. SW, Calgary, AB T2S 3C3, Canada; (C.R.-B.); (R.L.D.)
| | - Ronald D. Barr
- Health Sciences Centre 3A, Department of Pediatrics, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada;
| | - Sumit Gupta
- Department of Pediatrics, Division of Hematology/Oncology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; (S.G.); (P.C.N.)
| | - Paul C. Nathan
- Department of Pediatrics, Division of Hematology/Oncology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; (S.G.); (P.C.N.)
| | - Sarah J. McKillop
- Department of Pediatrics, Edmonton Clinic Health Academy, University of Alberta, 11405-87 Avenue, Edmonton, AL T6G 1C9, Canada;
| | - Miranda M. Fidler-Benaoudia
- Cancer Care Alberta, Alberta Health Services, Holy Cross Centre, Department of Cancer Epidemiology and Prevention Research, 5th Floor, BOX ACB, 2210-2 St. SW, Calgary, AB T2S 3C3, Canada; (C.R.-B.); (R.L.D.)
- Departments of Oncology and Community Health Sciences, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
47
|
Sen Halicioglu B, Saadat KASM, Tuglu MI. Adipose-Derived Mesenchymal Stem Cell Transplantation in Chemotherapy-Induced Premature Ovarian Insufficiency: the Role of Connexin and Pannexin. Reprod Sci 2021; 29:1316-1331. [PMID: 34449073 DOI: 10.1007/s43032-021-00718-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 08/14/2021] [Indexed: 10/20/2022]
Abstract
In women undergoing chemotherapy, it is inevitable that infertility risk will increase because of impaired reproductive functions. Premature ovarian insufficiency (POI), which occurs as a devastating result of chemotherapy, is the complete depletion or dysfunction of ovarian follicles. Adipose-derived mesenchymal stem cells (ADMSCs) transplantation is among the alternative treatment methods for POI, which currently do not have an effective treatment method. Apoptosis of granulosa cells in POI is seen as the main mechanism of the disease. It is also reported that in addition to molecules directly associated with apoptosis, connexins, and pannexins are also potential effector molecules in apoptosis. The roles of these molecules in POI, which are known to play a role in many important mechanisms in the ovary, are unknown. In this study, it was aimed to analyze the expressions of Connexin43 and Pannexin1, which are thought to be effective in the formation of POI, and to show the relationship between the antiapoptotic effects of ADMSCs transplantation and these molecules in POI. For this purpose, Caspase3, Connexin43, Pannexin1 proteins, and mRNA expressions were analyzed by immunohistochemistry and RT-qPCR, and AMH levels were measured by ELISA. It was determined that Pannexin1, Caspase3 proteins, and mRNA levels increased in the POI, while Pannexin1 and Caspase3 expressions decreased in the ADMSCs treated group. While Connexin43 level decreased in POI, Connexin43 protein and mRNA levels increased in ADMSCs group. Consequently, this study demonstrated for the first time that Connexin43 and Pannexin1 were associated with apoptosis in POI. In addition, it was revealed that ADMSCs transplantation could produce antiapoptotic effects by modulating these molecules.
Collapse
Affiliation(s)
- Busra Sen Halicioglu
- Faculty of Medicine, Department of Histology and Embryology, Gaziantep University, Gaziantep, Turkey. .,Faculty of Medicine, Department of Histology and Embryology, Manisa Celal Bayar University, Manisa, Turkey.
| | - Khandakar A S M Saadat
- Faculty of Medicine, Department of Medical Biology, Gaziantep University, Gaziantep, Turkey
| | - Mehmet Ibrahim Tuglu
- Faculty of Medicine, Department of Histology and Embryology, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
48
|
Hassel JC, Livingstone E, Allam JP, Behre HM, Bojunga J, Klein HH, Landsberg J, Nawroth F, Schüring A, Susok L, Thoms KM, Kiesel L, Berking C. Fertility preservation and management of pregnancy in melanoma patients requiring systemic therapy. ESMO Open 2021; 6:100248. [PMID: 34438241 PMCID: PMC8390524 DOI: 10.1016/j.esmoop.2021.100248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 01/09/2023] Open
Abstract
Melanoma is one of the most common cancers in adolescents and adults at fertile age, especially in women. With novel and more effective systemic therapies that began to profoundly change the dismal outcome of melanoma by prolonging overall survival, the wish for fertility preservation or even parenthood has to be considered for a growing portion of melanoma patients-from the patients' as well as from the physicians' perspective. The dual blockade of the mitogen-activated protein kinase pathway by B-Raf proto-oncogene serine/threonine kinase and mitogen-activated protein kinase inhibitors and the immune checkpoint inhibition by anti-programmed cell death protein 1 and anti-cytotoxic T-lymphocyte-associated protein-4 monoclonal antibodies constitute the current standard systemic approaches to combat locally advanced or metastatic melanoma. Here, the preclinical data and clinical evidence of these systemic therapies are reviewed in terms of their potential gonadotoxicity, teratogenicity, embryotoxicity and fetotoxicity. Recommendations for routine fertility and contraception counseling of melanoma patients at fertile age are provided in line with interdisciplinary recommendations for the diagnostic work-up of these patients and for fertility-protective measures. Differentiated recommendations for the systemic therapy in both the adjuvant and the advanced, metastatic treatment situation are given. In addition, the challenges of pregnancy during systemic melanoma therapy are discussed.
Collapse
Affiliation(s)
- J C Hassel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - E Livingstone
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - J P Allam
- Department of Andrology, University Hospital Bonn, Bonn, Germany
| | - H M Behre
- Center for Reproductive Medicine and Andrology, University Hospital Halle (Saale), Halle (Saale), Germany
| | - J Bojunga
- Department of Endocrinology, Diabetology and Nutrition Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - H H Klein
- Department of Internal Medicine, Endocrinology & Diabetology & Gastroenterology and Hepatology, BG University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - J Landsberg
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - F Nawroth
- Center for Infertility, Prenatal Medicine, Endocrinology and Osteology, Amedes Medical Center MVZ Hamburg, Hamburg, Germany
| | - A Schüring
- Fertility Center MVZ KITZ Regensburg, Regensburg, Germany
| | - L Susok
- Department of Dermatology, University Hospital Bochum, Bochum, Germany
| | - K M Thoms
- Department of Dermatology, Venereology and Allergy, University Medical Center Goettingen, Goettingen, Germany
| | - L Kiesel
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - C Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen EMN, Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.
| |
Collapse
|
49
|
van den Berg MH, van Dijk M, Byrne J, Berger C, Dirksen U, Winther JF, Fossa SD, Grabow D, Grandage VL, Haupt R, van den Heuvel-Eibrink MM, Kaiser M, Kepak T, van der Kooi ALF, Kremer LCM, Kruseova J, Lambalk CB, van Leeuwen FE, Leiper A, Modan-Moses D, Spix C, Twisk JWR, Ronckers CM, Kaatsch P, van Dulmen-den Broeder E. Treatment-related fertility impairment in long-term female childhood, adolescent and young adult cancer survivors: investigating dose-effect relationships in a European case-control study (PanCareLIFE). Hum Reprod 2021; 36:1561-1573. [PMID: 33744927 DOI: 10.1093/humrep/deab035] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/21/2021] [Indexed: 02/02/2023] Open
Abstract
STUDY QUESTION Which chemotherapeutic agents and body site-specific radiation fields are dose-dependently associated with an increased risk of fertility impairment in long-term female childhood, adolescent and young adulthood (CAYA) cancer survivors? SUMMARY ANSWER Busulfan, lower abdominal radiotherapy (RT) and total body irradiation (TBI) seem to be associated with fertility impairment at any dose, whereas gonadotoxicity of melphalan and procarbazine is suggested at medium/high (>140 mg/m2) or high dose (>5600 mg/m2) therapy, respectively. WHAT IS KNOWN ALREADY Several treatment-related fertility deficits, as assessed by both self-reported outcomes and hormonal markers are known to occur following treatment of CAYA cancer. However, knowledge regarding precise dose-related estimates of these treatment-related risks are scarce. STUDY DESIGN, SIZE, DURATION The current case-control study was nested within the PanCareLIFE cohort study. In total, 1332 CAYA survivors from 8 countries, 9 institutions and 11 cohorts, participated in and contributed data to the study. PARTICIPANTS/MATERIALS, SETTING, METHODS All participants were female 5-year CAYA cancer survivors. In total, 450 cases (fertility impaired survivors) and 882 matched controls (not fertility impaired survivors) were included. Fertility impairment was defined using both questionnaire data (primary or secondary amenorrhea; use of artificial reproductive techniques; unfulfilled wish to conceive) and hormonal data (FSH and anti-Müllerian hormone (AMH)). Multivariable logistic regression models were used to investigate the effect of (i) alkylating agent exposure, and (ii) dose categories for individual chemotherapeutic agents and for RT-exposed body sites. MAIN RESULTS AND THE ROLE OF CHANCE A positive dose-effect relationship between cyclophosphamide equivalent dose (CED) score and fertility impairment was found, with survivors with a CED score > 7121 mg/m2 being at a significantly increased risk of fertility impairment (odds ratio (95% CI) = 2.6 (1.9-3.6) P < 0.001). Moreover, cumulative dose variables of the following treatments were significantly associated with fertility impairment: busulfan, carmustine, cyclophosphamide, melphalan, procarbazine, lower abdominal RT and TBI. Busulfan, lower abdominal RT and TBI seem to be associated with fertility impairment at any dose, whereas gonadotoxicity of melphalan and procarbazine is suggested at medium/high (>140 mg/m2) or high dose (>5600 mg/m2) therapy, respectively. LIMITATIONS, REASONS FOR CAUTION Our study may have been subject to selection bias since data from about half of the original base cohorts were available for the current study. This could impact the generalizability of our study results. WIDER IMPLICATIONS OF THE FINDINGS We identified survivors at high risk for fertility impairment and, consequently, for a reduced or even absent reproductive life span. Both girls and young women who are about to start anti-cancer treatment, as well as adult female survivors, should be counselled about future parenthood and referred to a reproductive specialist for fertility preservation, if desired. STUDY FUNDING/COMPETING INTEREST(S) This study has received funding from the European Union's Seventh Framework Programme for research, technological development and demonstration under grant agreement no. 602030. There are no competing interests. TRIAL REGISTRATION NUMBER n/a.
Collapse
Affiliation(s)
- M H van den Berg
- Department of Paediatrics, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M van Dijk
- Department of Paediatrics, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J Byrne
- Boyne Research Institute, Department of Epidemiology, Drogheda, Ireland
| | - C Berger
- Department of Pediatric Hematology and Oncology, University-Hospital, Saint-Etienne, France
| | - U Dirksen
- Department of Paediatric Haematology and Oncology, University Hospital Muenster, Muenster, Germany
| | - J F Winther
- Danish Cancer Society Research Center, Childhood Cancer Research Group, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University and University Hospital, Aarhus, Denmark
| | - S D Fossa
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - D Grabow
- German Childhood Cancer Registry (GCCR), Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center, Mainz, Germany
| | | | - R Haupt
- Gaslini Children Hospital, Epidemiology and Biostatistics Section, Genova, Italy
| | - M M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Paediatric Oncology, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - M Kaiser
- German Childhood Cancer Registry (GCCR), Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center, Mainz, Germany
| | - T Kepak
- University Hospital Brno, Brno, Czech Republic
- International Clinical Research Center (FNUSA-ICRC), Brno, Czech Republic
| | - A L F van der Kooi
- Department of Obstetrics and Gynecology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - L C M Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - J Kruseova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - C B Lambalk
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam
| | - F E van Leeuwen
- Department of Epidemiology and Biostatistics, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Leiper
- Great Ormond Street Children's Hospital, London, UK
| | - D Modan-Moses
- Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Ramat Gan, Israel
- The Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - C Spix
- German Childhood Cancer Registry (GCCR), Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center, Mainz, Germany
| | - J W R Twisk
- Department of Epidemiology and Biostatistics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, THE Netherlands
| | - C M Ronckers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Institute for Biostatistics and Registry Research, Medical University Brandenburg, Neuruppin, Germany
| | - P Kaatsch
- German Childhood Cancer Registry (GCCR), Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center, Mainz, Germany
| | - E van Dulmen-den Broeder
- Department of Paediatrics, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Li J, Long H, Cong Y, Gao H, Lyu Q, Yu S, Kuang Y. Quercetin prevents primordial follicle loss via suppression of PI3K/Akt/Foxo3a pathway activation in cyclophosphamide-treated mice. Reprod Biol Endocrinol 2021; 19:63. [PMID: 33892727 PMCID: PMC8063466 DOI: 10.1186/s12958-021-00743-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/11/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chemotherapy improves the survival rates of patients with various cancers but often causes some adverse effects, including ovarian damage, characterised by a decrease in primordial follicle stockpiles. Recent studies have revealed that chemotherapy may stimulate the PI3K signalling pathway, thereby resulting in accelerated primordial follicle activation and a decreased ovarian reserve. Quercetin is an inhibitor of the PI3K pathway; however, its protective effects against chemotherapy-induced follicle loss in mice have not been established. In this study, the effects of quercetin in a mouse model of cyclophosphamide-induced ovarian dysfunction were investigated. METHODS C57BL/6 female mice were used for the study. Paraffin sections of mouse ovaries (n = 30 mice) were stained with haematoxylin and eosin for differential follicle counts. Apoptosis (n = 5 mice per group) was evaluated by TUNEL assay. Immunohistochemical staining for ki67 and Foxo3a (n = 5 mice per group) was performed to evaluate the activation of primordial follicles. The role of the PI3K signalling pathway in the ovaries (n = 45 mice) was assessed by western blotting. RESULTS Quercetin attenuated the cyclophosphamide-induced reduction in dormant primordial follicles. Analysis of the PI3K/Akt/Foxo3a pathway showed that quercetin decreased the phosphorylation of proteins that stimulate follicle activation in cyclophosphamide-induced ovaries. Furthermore, quercetin prevented cyclophosphamide-induced apoptosis in early growing follicles and early antral follicles, maintained anti-Müllerian hormone levels secreted by these follicles, and preserved the quiescence of the primordial follicle pool, as determined by intranuclear Foxo3a staining. CONCLUSIONS Quercetin attenuates cyclophosphamide-induced follicle loss by preventing the phosphorylation of PI3K/Akt/Foxo3a pathway members and maintaining the anti-Müllerian hormone level through reduced apoptosis in growing follicles. Accordingly, quercetin is expected to improve fertility preservation and the prevention of endocrine-related side effects of chemotherapy.
Collapse
Affiliation(s)
- Jianghui Li
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Hui Long
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Yanyan Cong
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Hongyuan Gao
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Qifeng Lyu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Sha Yu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China.
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China.
| |
Collapse
|