1
|
Harrold EC, Ogle M, O'Brien E, Wilde C, Sinopoli J, Weiss J, Martino L, Casson A, Kang HJ, Postow M, Cercek A. Fertility outcomes post immune checkpoint inhibitor exposure. Cancer Cell 2024:S1535-6108(24)00391-X. [PMID: 39486412 DOI: 10.1016/j.ccell.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/07/2024] [Accepted: 10/07/2024] [Indexed: 11/04/2024]
Abstract
Harrold et al. evaluate the fertility impact of checkpoint inhibitor blockade (ICB), demonstrating that unlike in utero exposure, post-exposure conception appears to result in uncomplicated pregnancies and healthy progeny. They demonstrate contemporaneous monitoring of temporal female hormonal fluctuations before, on, and post ICB exposure and prior to successful embryo implantation.
Collapse
Affiliation(s)
- Emily C Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martinique Ogle
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin O'Brien
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Callahan Wilde
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jenna Sinopoli
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jill Weiss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lauren Martino
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anne Casson
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hey-Joo Kang
- Department of Reproductive Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Michael Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2
|
Ligon JA, Cupit-Link MC, Yu C, Levine J, Foley T, Rotz S, Sharma A, Gomez-Lobo V, Shah NN. Pediatric Cancer Immunotherapy and Potential for Impact on Fertility: A Need for Evidence-Based Guidance. Transplant Cell Ther 2024; 30:737-749. [PMID: 38866240 DOI: 10.1016/j.jtct.2024.06.006] [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: 02/27/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
The use of immunotherapies for the treatment of cancer in children, adolescents, and young adults has become common. As the use of immunotherapy has expanded, including in earlier lines of therapy, it has become evident that several aspects of how these immunotherapies impact longer-term outcomes among survivors are understudied. Traditional cancer therapies like alkylating and platin agents carry the greatest risk of infertility, but little is known about the impact of novel immunotherapies on fertility. This topic is of great interest to patients, patient advocates, and clinicians. In this article, we review immunotherapeutic agents used to treat childhood and young adult cancers and discuss potential mechanisms by which they may impact fertility based on the known interplay between the immune system and reproductive organs. We highlight the relative paucity of high-quality literature examining these late effects. We discuss interventions to optimize fertility preservation (FP) for our patients. Conducting longitudinal, collaborative, and prospective research on the fertility outcomes of pediatric and young adult patients with cancer who receive immunotherapy is critical to learn how to effectively counsel our patients on long-term fertility outcomes and indications for FP procedures. Collection of patient-level data will be necessary to draft evidence-based guidelines on which providers can make therapy recommendations.
Collapse
Affiliation(s)
- John A Ligon
- Department of Pediatrics, Division of Hematology Oncology, University of Florida, Gainesville, Florida; University of Florida Health Cancer Center, Gainesville, Florida.
| | | | - Christine Yu
- Department of Pediatric Medicine, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Jennifer Levine
- Center for Cancer and Blood Disorders, Division of Oncology, Children's National Hospital, Washington District of Columbia
| | - Toni Foley
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Seth Rotz
- Division of Pediatric Hematology and Oncology and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Veronica Gomez-Lobo
- Pediatric and Adolescent Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Pediatric and Adolescent Gynecology, Children's National Hospital, Washington, District of Columbia
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
3
|
Laguna JC, Tagliamento M, Lambertini M, Hiznay J, Mezquita L. Tackling Non-Small Cell Lung Cancer in Young Adults: From Risk Factors and Genetic Susceptibility to Lung Cancer Profile and Outcomes. Am Soc Clin Oncol Educ Book 2024; 44:e432488. [PMID: 38788188 DOI: 10.1200/edbk_432488] [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: 05/26/2024]
Abstract
Lung cancer has traditionally been associated with advanced age; however, its increasing incidence among young adults raises concerning questions regarding its etiology and unique considerations for this population. In contrast to the older population, the onset of lung cancer at younger age may be attributed to a complex interplay of incompletely understood individual susceptibility and prevalent environmental risk factors beyond tobacco smoke exposure, such as radon gas and air pollution, which are widespread globally. Consequently, this leads to distinct clinical and molecular profiles, requiring a tailored approach. Furthermore, a diagnosis of cancer represents a threatening event during the prime years of a young person's life, prompting concern about career development, social aspects, fertility aspirations, and physical independence. This poses significant additional challenges for health care professionals in a field that remains underexplored. This comprehensive review recognizes lung cancer in young adults as a distinct entity, exploring its clinical and molecular characteristics, diverse predisposing factors, and priorities in terms of quality of life, with the aim of providing practical support to oncologists and enhancing our understanding of this under-researched population.
Collapse
Affiliation(s)
- Juan Carlos Laguna
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Zatelli MC, Faggiano A, Argentiero A, Danesi R, D'Oronzo S, Fogli S, Franchina T, Giorgino F, Marrano N, Giuffrida D, Gori S, Marino G, Mazzilli R, Monami M, Montagnani M, Morviducci L, Natalicchio A, Ragni A, Renzelli V, Russo A, Sciacca L, Tuveri E, Aimaretti G, Avogaro A, Candido R, Di Maio M, Silvestris N, Gallo M. Endocrine-metabolic assessment checklist for cancer patients treated with immunotherapy: A proposal by the Italian Association of Medical Oncology (AIOM), Italian Association of Medical Diabetologists (AMD), Italian Society of Diabetology (SID), Italian Society of Endocrinology (SIE) and Italian Society of Pharmacology (SIF) multidisciplinary group. Cancer Treat Rev 2024; 126:102734. [PMID: 38604051 DOI: 10.1016/j.ctrv.2024.102734] [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: 01/24/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024]
Abstract
Immunotherapy with immune checkpoint inhibitors (ICI) is increasingly employed in oncology. National and international endocrine and oncologic scientific societies have provided guidelines for the management of endocrine immune-related adverse events. However, guidelines recommendations differ according to the specific filed, particularly pertaining to recommendations for the timing of endocrine testing. In this position paper, a panel of experts of the Italian Association of Medical Oncology (AIOM), Italian Association of Medical Diabetologists (AMD), Italian Society of Diabetology (SID), Italian Society of Endocrinology (SIE), and Italian Society of Pharmacology (SIF) offers a critical multidisciplinary consensus for a clear, simple, useful, and easily applicable endocrine-metabolic assessment checklist for cancer patients on immunotherapy.
Collapse
Affiliation(s)
- Maria Chiara Zatelli
- Section of Endocrinology, Geriatrics and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy.
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical & Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.
| | | | - Romano Danesi
- Oncology and Haematology Dept, University of Milano "La Statale", Milano, Italy.
| | - Stella D'Oronzo
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari, Italy.
| | - Stefano Fogli
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Tindara Franchina
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy.
| | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy.
| | - Nicola Marrano
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy.
| | - Dario Giuffrida
- Department of Oncology, Istituto Oncologico del Mediterraneo, Viagrande, Catania, Italy.
| | - Stefania Gori
- Oncologia Medica, IRCCS Ospedale Don Calabria-Sacro Cuore di Negrar, Verona, Italy.
| | - Giampiero Marino
- Internal Medicine Department, Ospedale dei Castelli, Asl Roma 6, Ariccia, RM Italy.
| | - Rossella Mazzilli
- Endocrinology Unit, Department of Clinical & Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.
| | - Matteo Monami
- Diabetology, Careggi Hospital and University of Florence, Italy.
| | - Monica Montagnani
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, University of Bari Aldo Moro, Bari, Italy.
| | - Lelio Morviducci
- Diabetology and Nutrition Unit, Department of Medical Specialties, ASL Roma 1 - S. Spirito Hospital, Rome, Italy.
| | - Annalisa Natalicchio
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy.
| | - Alberto Ragni
- Endocrinology and Metabolic Diseases Unit, Azienda Ospedaliero-Universitaria SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria, Italy.
| | - Valerio Renzelli
- Diabetologist and Endocrinologist, Italian Association of Clinical Diabetologists, Rome, Italy.
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy.
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Catania, Italy.
| | - Enzo Tuveri
- Diabetology, Endocrinology and Metabolic Diseases Service, ASL-Sulcis, Italy.
| | - Gianluca Aimaretti
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.
| | - Angelo Avogaro
- Department of Medicine, Section of Diabetes and Metabolic Diseases, University of Padova, Padova, Italy.
| | - Riccardo Candido
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy.
| | | | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy.
| | - Marco Gallo
- Endocrinology and Metabolic Diseases Unit, Azienda Ospedaliero-Universitaria SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria, Italy.
| |
Collapse
|
5
|
Handelsman DJ, Idan A, Sleiman S, Bacha F, Long GV, Menzies AM, Vaishnav T, Litkouhi N, Volckmar X, Ledger W, Anazodo A. Testicular function after non-cytotoxic and immunotherapy drug treatment. Andrology 2024; 12:891-898. [PMID: 37889046 DOI: 10.1111/andr.13546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/21/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND The effects of novel non-cytotoxic and immunotherapy drugs for cancer treatment on human testicular function have not been studied systematically. OBJECTIVES The present study aimed to characterize effects of non-cytotoxic and immunotherapy drugs in patients with cancers who had not been previously treated with gonadotoxic chemo- or radiotherapy. MATERIALS AND METHODS This study involved 34 men, not previously treated with gonadotoxic regimens, in a mixed longitudinal (Cohort 1: 19 men about to start and approximately 1 year on non-cytotoxic and immunotherapy treatment) and cross-sectional (Cohort 2: 15 men already on non-cytotoxic and immunotherapy treatment) study using data modeling to estimate within-person time-course changes in testicular exocrine and endocrine functions. Cohort 1 provided 45 paired semen and blood samples (34 prior to and nine during treatment) and Cohort 2 provided 45 sets of samples (15 pre-treatment, 30 on treatment), including six men in Cohort 2 who had pre-treatment spermatozoa cryostorage prior to the study. Men on non-cytotoxic and immunotherapy treatment had undergone a median of 33.5 months long-term treatment. RESULTS Spermatozoa output and concentration were reduced by about 50%, with corresponding increases in serum follicle-stimulating hormone and decreases in serum inhibin B. Serum testosterone, luteinizing hormone, and sex hormone-binding globulin were unaffected by non-cytotoxic and immunotherapy treatment. CONCLUSION Within limits of the present study of sample size and duration of on-non-cytotoxic and immunotherapy treatment, non-cytotoxic and immunotherapy drugs have a modest effects on testicular exocrine function (sperm production) or its hormonal correlates (follicle-stimulating hormone, inhibin B), with minimal impact on testicular endocrine (testosterone, luteinizing hormone) function.
Collapse
Affiliation(s)
- David J Handelsman
- Andrology Department and Clinical Andrology Laboratory, Concord Hospital, Sydney, New South Wales, Australia
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Amanda Idan
- Andrology Department and Clinical Andrology Laboratory, Concord Hospital, Sydney, New South Wales, Australia
| | - Sue Sleiman
- Andrology Department and Clinical Andrology Laboratory, Concord Hospital, Sydney, New South Wales, Australia
| | - Fey Bacha
- Andrology Department and Clinical Andrology Laboratory, Concord Hospital, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute of Australia, University of Sydney, Royal North Shore & Mater Hospitals, Sydney, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute of Australia, University of Sydney, Royal North Shore & Mater Hospitals, Sydney, New South Wales, Australia
| | - Tejnei Vaishnav
- School of Women's and Childrens Health, Royal Hospital for Women, Sydney, New South Wales, Australia
| | - Noosha Litkouhi
- School of Women's and Childrens Health, Royal Hospital for Women, Sydney, New South Wales, Australia
| | - Xanthie Volckmar
- School of Women's and Childrens Health, Royal Hospital for Women, Sydney, New South Wales, Australia
| | - William Ledger
- School of Women's and Childrens Health, Royal Hospital for Women, Sydney, New South Wales, Australia
- University of New South Wales, Sydney, New South Wales, Australia
| | - Antoinette Anazodo
- School of Women's and Childrens Health, Royal Hospital for Women, Sydney, New South Wales, Australia
- University of New South Wales, Sydney, New South Wales, Australia
- Sydney Children's Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Grajales V, Martini A, Shore ND. Complications of immuno-oncology care: what urologist should know. BJU Int 2024; 133:524-531. [PMID: 38437876 DOI: 10.1111/bju.16310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
OBJECTIVES To provide a practical review of immune-related adverse events (irAEs) that may be encountered in uro-oncology patients. PATIENTS AND METHODS We conducted a literature review of studies reporting irAEs including articles published through September 2023 for uro-oncology patients and the potential relevancy for the practicing urologist. RESULTS Immunotherapy has revolutionised cancer treatment, extending its impact to urological malignancies including for patients with urothelial, kidney, and prostate cancers. Immuno-oncology (IO) compounds have achieved measurable and durable responses in these cancers. Urologists, choosing to administer or co-manage IO patient care, should be prepared to understand, evaluate, and treat irAEs. This review discusses the spectrum of irAEs that can be encountered. Ongoing trials are exploring the use of immunotherapy at earlier stages of uro-oncological diseases, thus underscoring the evolving landscape of urological cancer treatment. Paradoxically, some data suggests that the occurrence of irAEs is associated with improved oncological outcomes. CONCLUSIONS Immune-related AEs, while manageable, may be life-threatening and require lifelong therapy. A thorough understanding of AEs and toxicity of a novel drug class is imperative.
Collapse
Affiliation(s)
| | - Alberto Martini
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| |
Collapse
|
7
|
Silvestris E, D’Oronzo S, Petracca EA, D’Addario C, Cormio G, Loizzi V, Canosa S, Corrado G. Fertility Preservation in the Era of Immuno-Oncology: Lights and Shadows. J Pers Med 2024; 14:431. [PMID: 38673058 PMCID: PMC11050999 DOI: 10.3390/jpm14040431] [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: 03/16/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
In recent years, immuno-oncology has revolutionized the cancer treatment field by harnessing the immune system's power to counteract cancer cells. While this innovative approach holds great promise for improving cancer outcomes, it also raises important considerations related to fertility and reproductive toxicity. In fact, most young females receiving gonadotoxic anti-cancer treatments undergo iatrogenic ovarian exhaustion, resulting in a permanent illness that precludes the vocation of motherhood as a natural female sexual identity. Although commonly used, oocyte cryopreservation for future in vitro fertilization and even ovarian cortex transplantation are considered unsafe procedures in cancer patients due to their oncogenic risks; whereas, ovarian stem cells might support neo-oogenesis, providing a novel stemness model of regenerative medicine for future fertility preservation programs in oncology. Recent scientific evidence has postulated that immune checkpoint inhibitors (ICIs) might in some way reduce fertility by inducing either primary or secondary hypogonadism, whose incidence and mechanisms are not yet known. Therefore, considering the lack of data, it is currently not possible to define the most suitable FP procedure for young patients who are candidates for ICIs. In this report, we will investigate the few available data concerning the molecular regulation of ICI therapy and their resulting gonadal toxicity, to hypothesize the most suitable fertility preservation strategy for patients receiving these drugs.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (E.A.P.); (G.C.); (V.L.)
| | - Stella D’Oronzo
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy;
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, 70124 Bari, Italy
| | - Easter Anna Petracca
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (E.A.P.); (G.C.); (V.L.)
| | - Claudia D’Addario
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy;
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, 70124 Bari, Italy
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (E.A.P.); (G.C.); (V.L.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy;
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (E.A.P.); (G.C.); (V.L.)
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Stefano Canosa
- IVIRMA, Global Research Alliance, LIVET, 10126 Turin, Italy;
| | - Giacomo Corrado
- Gynecologic Oncology Unit, Department of Woman, Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00136 Roma, Italy;
| |
Collapse
|
8
|
Suijkerbuijk KPM, van Eijs MJM, van Wijk F, Eggermont AMM. Clinical and translational attributes of immune-related adverse events. NATURE CANCER 2024; 5:557-571. [PMID: 38360861 DOI: 10.1038/s43018-024-00730-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/12/2024] [Indexed: 02/17/2024]
Abstract
With immune checkpoint inhibitors (ICIs) becoming the mainstay of treatment for many cancers, managing their immune-related adverse events (irAEs) has become an important part of oncological care. This Review covers the clinical presentation of irAEs and crucial aspects of reversibility, fatality and long-term sequelae, with special attention to irAEs in specific patient populations, such as those with autoimmune diseases. In addition, the genetic basis of irAEs, along with cellular and humoral responses to ICI therapy, are discussed. Detrimental effects of empirically used high-dose steroids and second-line immunosuppression, including impaired ICI effectiveness, call for more tailored irAE-treatment strategies. We discuss open therapeutic challenges and propose potential avenues to accelerate personalized management strategies and optimize outcomes.
Collapse
Affiliation(s)
- Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Mick J M van Eijs
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Alexander M M Eggermont
- University Medical Center Utrecht and Princess Máxima Center, Utrecht, the Netherlands
- Comprehensive Cancer Center Munich of the Technical University of Munich and the Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
9
|
Kazemzadeh K, Behrouzieh S, Rezaei N. Shedding light on the side effects of immunotherapies used for leukemia: an updated review of the literature. Expert Rev Anticancer Ther 2023; 23:1193-1204. [PMID: 37812581 DOI: 10.1080/14737140.2023.2267760] [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: 06/26/2023] [Accepted: 10/03/2023] [Indexed: 10/11/2023]
Abstract
INTRODUCTION Leukemia is an abnormal clonal development of leukemic cells originating from the bone marrow, which is widely known for its significant prevalence and mortality rate. Chemotherapy, surgery, radiation, and combination therapies have been its routine therapeutic methods; however, the advent of cancer immunotherapy is known as revolutionary for its higher efficacy and lesser toxicity. AREAS COVERED Immunotherapy boosts the body's immune system by using components from other living organisms. Although immunotherapy seems to be safer than chemotherapy, many studies have noticed different immune-related side effects in various body systems (e.g. cardiovascular, neurologic) which we have reviewed in this investigation as the main goal. We tried to describe immunotherapy-related side effects in human body systems in detail. EXPERT OPINION Being aware of these side effects leads to better clinical decision-making for each individual, and a one-step-ahead management in case of occurrence. We also briefly discussed the role of immunotherapy in treating leukemia as one of the most prevalent cancers in children and tried to emphasize that it is crucial to monitor adverse events as they may remain obscure until adolescence.
Collapse
Affiliation(s)
- Kimia Kazemzadeh
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Neurosurgery and Artificial Intelligence (NONAI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sadra Behrouzieh
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Neurosurgery and Artificial Intelligence (NONAI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Nelson M, Levine J. Current Issues in Fertility Preservation Among Pediatric and Adolescent Cancer Patients. Curr Oncol Rep 2023; 25:793-802. [PMID: 37036623 DOI: 10.1007/s11912-023-01401-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2023] [Indexed: 04/11/2023]
Abstract
PURPOSE OF REVIEW Although fertility preservation is not yet fully integrated into the comprehensive cancer care of pediatric and adolescent patients, advances continue to take place. This review summarizes recent updates and trends for health care professionals caring for these patients. RECENT FINDINGS The creation of standardized infertility risk assessment guidelines offers the opportunity to provide greater consistency in clinical care and to provide a current baseline for future research studies seeking to refine risk stratification for individual patients. New agents are being introduced into cancer care; as their use increases, information about their impact on fertility is being studied. Ovarian tissue cryopreservation offers a new standard of care option for fertility preservation, but additional studies are needed to further assess efficacy and impact on ovarian reserve among pediatric and adolescent patients. Standardization of fertility preservation recommendations among certain sub-populations may also make it easier to provide greater consistency in clinical care. Advances continue to be made in the field of fertility preservation, but dissemination of this information is critical to moving toward fertility preservation truly being a part of comprehensive cancer care.
Collapse
Affiliation(s)
- Marie Nelson
- Children's National Medical Center, Center for Cancer and Blood Disorders, 111 Michigan Avenue NW, Room W4-604, Washington, D.C., 20010, USA
| | - Jennifer Levine
- Children's National Medical Center, Center for Cancer and Blood Disorders, 111 Michigan Avenue NW, Room W4-604, Washington, D.C., 20010, USA.
| |
Collapse
|
11
|
Tuerxun H, Zhao Y, Li Y, Liu X, Wen S, Cao J, Cui J, Zhao Y. Immune Checkpoint Inhibitors as A Threat to Reproductive Function: A Systematic Review. Crit Rev Oncol Hematol 2023:104064. [PMID: 37379960 DOI: 10.1016/j.critrevonc.2023.104064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/08/2023] [Accepted: 06/25/2023] [Indexed: 06/30/2023] Open
Abstract
In recent years, the indications for immunotherapy in cancer treatment have been expanding. The increased risk of cancer in young people, coupled with the fact that many women or men choose to delay childbearing, has made an increasing number of patients of childbearing age eligible for immunotherapy. Furthermore, with the improvements of various treatments, more young people and children are able to survive cancer. As a result, long-term sequelae of cancer treatments, such as reproductive dysfunction, are increasingly important for survivors. While many anti-cancer drugs are known to cause reproduction dysfunction, the effects of immune checkpoint inhibitors (ICIs) on reproduction function remain largely unknown. Through a retrospective analysis of previous reports and literature, this article aims to elucidate the causes of reproductive dysfunction induced by ICIs and focus on their specific mechanisms, in order to providing some guidance to clinicians and patients.
Collapse
Affiliation(s)
- Halahati Tuerxun
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yawen Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xingyu Liu
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Shuhui Wen
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jingjing Cao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
12
|
Hoffmann I, Greither T, Behre HM. [Fertility and fertility preservation in men]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2023:10.1007/s00105-023-05167-w. [PMID: 37286872 DOI: 10.1007/s00105-023-05167-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 06/09/2023]
Abstract
Fertility preservation is of high importance for patients prior to treatment that can impair fertility. The individual risk of becoming infertile after a fertility-reducing therapy depends on the type and duration of therapy, surgical technique, dose and combination of gonadotoxic drugs or radiation applied, and individual predisposition. Cryopreservation of ejaculated sperm is the standard procedure for creating a fertility reserve in men. In cases of azoospermia or inability to obtain semen by masturbation, testicular sperm can be obtained by (micro-)testicular sperm extraction (TESE) and cryopreserved. In case of retrograde ejaculation, sperm collection can be attempted by rectal electrostimulation or after off-label administration of imipramine from postmasturbatory urine. The cryopreserved sperm can be stored permanently in the gaseous phase of liquid nitrogen before being used in fertility therapy. In Germany, approval according to § 20b of the German Medicines Act (AMG) is a mandatory requirement for performing cryopreservation of sperm and testicular tissue; approval according to § 20c of the AMG must be obtained for use. For prepubertal boys, it is possible to cryopreserve dormant spermatogonial stem cells as part of an experimental procedure.
Collapse
Affiliation(s)
- Ivan Hoffmann
- Zentrum für Reproduktionsmedizin und Andrologie, Universitätsklinikum Halle (Saale), Ernst-Grube-Str. 40, 06120, Halle (Saale), Deutschland.
| | - Thomas Greither
- Zentrum für Reproduktionsmedizin und Andrologie, Universitätsklinikum Halle (Saale), Ernst-Grube-Str. 40, 06120, Halle (Saale), Deutschland
| | - Hermann M Behre
- Zentrum für Reproduktionsmedizin und Andrologie, Universitätsklinikum Halle (Saale), Ernst-Grube-Str. 40, 06120, Halle (Saale), Deutschland
| |
Collapse
|
13
|
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: 5.0] [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
|
14
|
Chen Z, Huang J, Kwak-Kim J, Wang W. Immune checkpoint inhibitors and reproductive failures. J Reprod Immunol 2023; 156:103799. [PMID: 36724630 DOI: 10.1016/j.jri.2023.103799] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/15/2023]
Abstract
The human conceptus is a semi-allograft, which is antigenically foreign to the mother. Hence, the implantation process needs mechanisms to prevent allograft rejection during successful pregnancy. Immune checkpoints are a group of inhibitory pathways expressed on the surface of various immune cells in the form of ligand receptors. Immune cells possess these pathways to regulate the magnitude of immune responses and induce maternal-fetal tolerance. Briefly, 1) CTLA-4 can weaken T cell receptor (TCR) signals and inhibit T cell response; 2) The PD-1/PD-L1 pathway can reduce T cell proliferation, enhance T cell anergy and fatigue, reduce cytokine production, and increase T regulatory cell activity to complete the immunosuppression; 3) TIM3 interacts with T cells by binding Gal-9, weakening Th1 cell-mediated immunity and T cell apoptosis; 4) The LAG-3 binding to MHC II can inhibit T cell activation by interfering with the binding of CD4 to MHC II, and; 5) TIGIT can release inhibitory signals to NK and T cells through the ITIM structure of its cytoplasmic tail. Therefore, dysregulated immune checkpoints or the application of immune checkpoint inhibitors may impair human reproduction. This review intends to deliver a comprehensive overview of immune checkpoints in pregnancy, including CTLA-4, PD-1/PD-L1, TIM-3, LAG-3, TIGIT, and their inhibitors, reviewing their roles in normal and pathological human pregnancies.
Collapse
Affiliation(s)
- Zeyang Chen
- School of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao 266000, PR China; Reproduction Medical Center, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Jinxia Huang
- Reproduction Medical Center, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China; Department of Gynecology, Weihai Central Hospital Affiliated to Qingdao University, 3 Mishan East Road, Weihai 264400, PR China
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA; Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| | - Wenjuan Wang
- Reproduction Medical Center, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China.
| |
Collapse
|
15
|
Ligon JA, Hayashi M, Ciampa D, Kramer C, Guastella A, Fuchs RJ, Herati AS, Christianson MS, Chen AR. A multidisciplinary pediatric oncofertility team improves fertility preservation and counseling across 7 years. Cancer Rep (Hoboken) 2023; 6:e1753. [PMID: 36346013 PMCID: PMC9939996 DOI: 10.1002/cnr2.1753] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/20/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Oncofertility is a developing field of increasing importance, particularly in pediatric oncology, where most patients are likely to survive long-term and have not yet had the opportunity to have children. AIMS We performed a quality improvement initiative to increase our rates of fertility preservation counseling and referral through the implementation of a pediatric oncofertility team, and we report outcomes 7 years following implementation of our initiative. METHODS AND RESULTS We compare our baseline oncofertility survey to 44 post-intervention survey respondents and electronic medical record documentation for 149 patients treated in 2019. Ninety-five percent of post-intervention survey respondents recalled fertility counseling (baseline 70%, p = .004) and 89.3% were appropriately referred for fertility preservation (baseline 50%, p = .017). Counseling was documented in 60.4% of charts; 81% of patients analyzed by chart review were appropriately referred for fertility preservation. Fertility preservation outcomes differed by sex assigned at birth. CONCLUSION Creation of an oncofertility team produced improvements in fertility counseling and fertility preservation referral across an extended period of time.
Collapse
Affiliation(s)
- John A. Ligon
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University (JHU)BaltimoreMarylandUSA
- Department of Pediatrics, Division of Hematology/OncologyUniversity of FloridaGainesvilleFloridaUSA
| | - Masanori Hayashi
- Department of Pediatrics—Hematology/oncology and Bone Marrow TransplantationUniversity of ColoradoAuroraColoradoUSA
| | - Devon Ciampa
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University (JHU)BaltimoreMarylandUSA
| | - Cara Kramer
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University (JHU)BaltimoreMarylandUSA
| | - Alfredo Guastella
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University (JHU)BaltimoreMarylandUSA
| | - Robert J. Fuchs
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University (JHU)BaltimoreMarylandUSA
| | - Amin S. Herati
- James Buchanan Brady Urological Institute and Department of UrologyJHUBaltimoreMarylandUSA
| | | | - Allen R. Chen
- Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins University (JHU)BaltimoreMarylandUSA
| |
Collapse
|
16
|
Cosci I, Grande G, Di Nisio A, Rocca MS, Del Fiore P, Benna C, Mocellin S, Ferlin A. Cutaneous Melanoma and Hormones: Focus on Sex Differences and the Testis. Int J Mol Sci 2022; 24:ijms24010599. [PMID: 36614041 PMCID: PMC9820190 DOI: 10.3390/ijms24010599] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/24/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022] Open
Abstract
Cutaneous melanoma, the most aggressive type of skin cancer, remains one the most represented forms of cancer in the United States and European countries, representing, in Australia, the primary cause of cancer-related deaths. Recently, many studies have shown that sex disparities previously observed in most cancers are particularly accentuated in melanoma, where male sex is consistently associated with an increased risk of disease progression and a higher mortality rate. The causes of these sex differences rely on biological mechanisms related to sex hormones, immune homeostasis and oxidative processes. The development of newer therapies, such as immune checkpoint inhibitors (ICIs) (i.e., anti-PD-1 and anti-CTLA-4 monoclonal antibodies) has dramatically changed the treatment landscape of metastatic melanoma patients, though ICIs can interfere with the immune response and lead to inflammatory immune-related adverse events (irAEs). Recently, some studies have shown a potential adverse influence of this immunotherapy treatment also on male fertility and testicular function. However, while many anticancer drugs are known to cause defects in spermatogenesis, the effects of ICIs therapy remain largely unknown. Notwithstanding the scarce and conflicting information available on this topic, the American Society of Clinical Oncology guidelines recommend sperm cryopreservation in males undergoing ICIs. As investigations regarding the long-term outcomes of anticancer immunotherapy on the male reproductive system are still in their infancy, this review aims to support and spur future research in order to understand a potential gonadotoxic effect of ICIs on testicular function, spermatogenesis and male fertility.
Collapse
Affiliation(s)
- Ilaria Cosci
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Giuseppe Grande
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy
| | - Andrea Di Nisio
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Maria Santa Rocca
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy
| | - Paolo Del Fiore
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | - Clara Benna
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, 35128 Padova, Italy
| | - Simone Mocellin
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, 35128 Padova, Italy
| | - Alberto Ferlin
- Unit of Andrology and Reproductive Medicine, University Hospital of Padova, 35128 Padova, Italy
- Department of Medicine, University of Padova, 35128 Padova, Italy
- Correspondence: ; Tel.: +39-049-8212723
| |
Collapse
|
17
|
Fertility and CAR T-cells: Current practice and future directions. Transplant Cell Ther 2022; 28:605.e1-605.e8. [DOI: 10.1016/j.jtct.2022.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/18/2022] [Accepted: 06/08/2022] [Indexed: 01/09/2023]
|
18
|
Immune-Related Uncommon Adverse Events in Patients with Cancer Treated with Immunotherapy. Diagnostics (Basel) 2022; 12:diagnostics12092091. [PMID: 36140493 PMCID: PMC9498261 DOI: 10.3390/diagnostics12092091] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/06/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Immunotherapy has dramatically changed the therapeutic landscape of oncology, and has become standard of care in multiple cancer types in front or late lines of therapy, with some longstanding responses and outstanding results. Notwithstanding, its use has brought a totally unique spectrum of adverse events, characterized by a myriad of diverse manifestations affecting nearly every organ and system of the body, including the endocrine, nervous, cardiac, respiratory and gastrointestinal systems. Uncommon adverse events, defined as those occurring in less than 1% of patients, comprise an even more heterogeneous group of diseases that are being seen more recurrently as the use of immune check-point inhibitors increases and indications spread in different tumor types and stages. Here, we comprehensively review some uncommon, but exceedingly important, immune-related adverse events, with special emphasis in the clinical approach and diagnostic workup, aiming to reunite the evidence published previously, allowing an increase in awareness and knowledge from all specialists implicated in the diagnosis, treatment, and care of cancer patients treated with immunotherapy.
Collapse
|
19
|
Fertility preservation for patients with melanoma. Melanoma Res 2022; 32:303-308. [PMID: 35855660 DOI: 10.1097/cmr.0000000000000840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The advent of immunotherapy and targeted therapy has outstandingly improved the prognosis in subjects with melanoma. Their use is now advocated also in earlier stages as an adjuvant therapy, and some neoadjuvant clinical trials are ongoing. Consequently, survivors free of disease are increasing, as well as those exposed to these new agents. Parenthood in survivors is, therefore, receiving growing interest. Evidence on the effects of immunotherapy and targeted therapy on future fertility is limited, but not entirely reassuring, in particular for immunotherapy. The necessity of delaying pregnancy seeking up to the end of treatments and follow-up (iatrogenic aging) is an additional albeit neglected source of concern, in particular for women in their late 30s. Subjects with melanoma should be informed on the multifaceted issue of future fertility at the time of cancer diagnosis. Available options of fertility preservations, including sperm and oocytes storage, should also be discussed, especially considering that at the age 0-39, melanoma represents the second most frequent neoplasia. In the decision-making process, most attention should be given to sex, age, and exposure to immunotherapy.
Collapse
|
20
|
Yuen KCJ, Samson SL, Bancos I, Gosmanov AR, Jasim S, Fecher LA, Weber JS. AMERICAN ASSOCIATION OF CLINICAL ENDOCRINOLOGY (AACE) DISEASE STATE CLINICAL REVIEW EVALUATION AND MANAGEMENT OF IMMUNE CHECKPOINT INHIBITOR-MEDIATED ENDOCRINOPATHIES: A PRACTICAL CASE-BASED CLINICAL APPROACH. Endocr Pract 2022; 28:719-731. [PMID: 35477029 DOI: 10.1016/j.eprac.2022.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The aim of this case-based clinical review is to provide a practical approach for clinicians regarding the management of patients with immune checkpoint inhibitor (ICI)-mediated endocrinopathies. METHODS A literature search was conducted using PubMed, Embase and Scopus, and appropriate keywords. The discussions and strategies for diagnosis and management of ICI-mediated endocrinopathies are based on evidence available from prospective randomized clinical studies, cohort studies, cross-sectional studies, case-based studies, and expert consensus. RESULTS Immunotherapy with ICIs has transformed the treatment landscape of diverse cancer types, but frequently results in immune-mediated endocrinopathies that can cause acute and persistent morbidity, and rarely, death. The patterns of endocrinopathies differ between inhibitors of the CTLA-4 and PD-1/PD-L1 pathways, but most often involve the thyroid and pituitary glands. Less common but important presentations include insulin-deficient diabetes mellitus, primary adrenal insufficiency, primary hypoparathyroidism, central diabetes insipidus, primary hypogonadism, and pancreatitis with or without subsequent progression to diabetes or exocrine insufficiency. CONCLUSION In recent years, with increasing numbers of cancer patients being treated with ICIs, more clinicians in a variety of specialties are called upon to diagnose and treat ICI-mediated endocrinopathies. Herein, we review case scenarios of various clinical manifestations, and emphasize the need for a high index of clinical suspicion by all clinicians caring for these patients including endocrinologists, oncologists, primary care providers, and emergency department physicians. We also provide diagnostic and therapeutic approaches for ICI-induced endocrinopathies, and we propose that patients on ICI-therapy be evaluated and treated in a multidisciplinary team in collaboration with endocrinologists.
Collapse
Affiliation(s)
- Kevin C J Yuen
- Co-Chair of Task Force; Professor of Medicine, Department of Medicine, University of Arizona College of Medicine and Creighton School of Medicine, Phoenix, Arizona.
| | - Susan L Samson
- Co-Chair of Task Force; Senior Associate Consultant, Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Jacksonville, Florida
| | - Irina Bancos
- Associate Professor of Medicine; Associate Program Director, Endocrinology Fellowship Program, Division of Endocrinology, Metabolism and Nutrition, Mayo Clinic, Rochester, MN
| | - Aidar R Gosmanov
- Professor of Medicine, Division of Endocrinology, Albany Medical College; Chief, Endocrinology Section, Stratton VAMC, Albany, NY
| | - Sina Jasim
- Associate Professor of Medicine, Washington University in St. Louis, School of Medicine, Division of Endocrinology, Metabolism and Lipid Research, St. Louis, Missouri
| | - Leslie A Fecher
- ASCO Representative, Associate Professor of Medicine and Dermatology, University of Michigan, Rogel Cancer Center, Ann Arbor, Michigan
| | - Jeffrey S Weber
- ASCO Representative, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| |
Collapse
|
21
|
Johnson DB, Nebhan CA, Moslehi JJ, Balko JM. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat Rev Clin Oncol 2022; 19:254-267. [PMID: 35082367 PMCID: PMC8790946 DOI: 10.1038/s41571-022-00600-w] [Citation(s) in RCA: 447] [Impact Index Per Article: 223.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 12/15/2022]
Abstract
The development of immune-checkpoint inhibitors (ICIs) has heralded a new era in cancer treatment, enabling the possibility of long-term survival in patients with metastatic disease, and providing new therapeutic indications in earlier-stage settings. As such, characterizing the long-term implications of receiving ICIs has grown in importance. An abundance of evidence exists describing the acute clinical toxicities of these agents, although chronic effects have not been as well catalogued. Nonetheless, emerging evidence indicates that persistent toxicities might be more common than initially suggested. While generally low-grade, these chronic sequelae can affect the endocrine, rheumatological, pulmonary, neurological and other organ systems. Fatal toxicities also comprise a diverse set of clinical manifestations and can occur in 0.4-1.2% of patients. This risk is a particularly relevant consideration in light of the possibility of long-term survival. Finally, the effects of immune-checkpoint blockade on a diverse range of immune processes, including atherosclerosis, heart failure, neuroinflammation, obesity and hypertension, have not been characterized but remain an important area of research with potential relevance to cancer survivors. In this Review, we describe the current evidence for chronic immune toxicities and the long-term implications of these effects for patients receiving ICIs.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA.
| | - Caroline A Nebhan
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Javid J Moslehi
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| |
Collapse
|
22
|
Prescription Patterns, Recurrence, and Toxicity Rates of Adjuvant Treatment for Stage III/IV Melanoma—A Real World Single-Center Analysis. BIOLOGY 2022; 11:biology11030422. [PMID: 35336796 PMCID: PMC8945449 DOI: 10.3390/biology11030422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 11/20/2022]
Abstract
Simple Summary Adjuvant treatment with the immune checkpoint inhibitors (ICI) pembrolizumab or nivolumab, or the targeted therapies dabrafenib and trametinib is recommended for patients with completely resected stage III melanoma and significantly decreases recurrence risk. Currently, limited data are available on physicians’ prescription preferences regarding ICI and targeted therapies and patient outcome in clinical practice. This study investigates the real-world situation of 109 patients from the Cancer Center of the University Hospital Bern, Switzerland, with an indication for adjuvant treatment since 2018. We describe treatment patterns, recurrence, and toxicity rates under immune checkpoint inhibitors, and targeted therapies. Abstract Approved adjuvant treatment options for stage III melanoma are the immune checkpoint inhibitors (ICI) pembrolizumab and nivolumab, and in presence of a BRAF V600E/K mutation additionally dabrafenib in combination with trametinib (BRAFi/MEKi). This study aims to describe prescription patterns and recurrence and toxicity rates of adjuvant-treated melanoma patients from the Cancer Center of the University Hospital Bern, Switzerland. One hundred and nine patients with an indication for adjuvant treatment were identified. Five (4.6%) had contraindications and, as such, were not proposed any adjuvant treatment, while 10 patients (9.2%) declined treatment. BRAF status was known for 91 (83.5%) patients. Of 40 (36.7%) patients with BRAF V600E/K melanoma, pembrolizumab was prescribed to 18 (45.0%), nivolumab to 16 (40.0%), and dabrafenib/trametinib to three (7.5%) patients. Grade 3–4 toxicity was reported in 18.9% and 16.7% of all the patients treated with pembrolizumab and nivolumab, respectively. No toxicities were observed for dabrafenib/trametinib. Thirty-eight percent of the patients treated with pembrolizumab and 40.0% of those treated with nivolumab relapsed. No relapses were reported for dabrafenib/trametinib. Prescription patterns indicate a clear preference for adjuvant ICI treatment.
Collapse
|
23
|
Sena LA, Sedhom R, Scott S, Kagan A, Marple AH, Canzoniero JV, Hsu M, Qasim Hussaini SM, Herati AS, Reschke L, Antero MF, Christianson MS, Binder AF, Chen AR, Donehower RC, Marrone KA, Gupta A. Trainee-Led Quality Improvement Project to Improve Fertility Preservation Counseling for Patients With Cancer. JCO Oncol Pract 2022; 18:e403-e411. [PMID: 34565170 PMCID: PMC8932487 DOI: 10.1200/op.21.00479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/10/2021] [Accepted: 08/25/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Oncofertility counseling regarding the reproductive risks associated with cancer therapy is essential for quality cancer care. We aimed to increase the rate of oncofertility counseling for patients of reproductive age (18-40 years) with cancer who were initiating systemic therapy at the Johns Hopkins Cancer Center from a baseline rate of 37% (25 of 68, June 2019-January 2020) to 70% by February 2021. METHODS We formed an interprofessional, multidisciplinary team as part of the ASCO Quality Training Program. We obtained data from the electronic medical record and verified data with patients by phone. We surveyed patients, oncologists, and fertility specialists to identify barriers. After considering a prioritization matrix, we implemented Plan-Do-Study-Act (PDSA) cycles. RESULTS We identified the following improvement opportunities: (1) oncologist self-reported lack of knowledge about counseling and local fertility preservation options and (2) lack of a standardized referral mechanism to fertility services. During the first PDSA cycle (February 2020-August 2020, disrupted by COVID-19), we introduced the initiative to increase oncofertility counseling at faculty meetings. From September 2020 to November 2020, we implemented a second PDSA cycle: (1) educating and presenting the initiative at Oncology Grand Rounds, (2) distributing informative pamphlets to oncologists and patients, and (3) implementing an electronic medical record order set. In the third PDSA cycle (December 2020-February 2021), we redesigned the order set to add information (eg, contact information for fertility coordinator) to the patient after-visit summary. Postimplementation (September 2020-February 2021), counseling rates increased from 37% to 81% (38 of 47). CONCLUSION We demonstrate how a trainee-led, patient-centered initiative improved oncofertility care. Ongoing work focuses on ensuring sustainability and assessing the quality of counseling.
Collapse
Affiliation(s)
- Laura A. Sena
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Ramy Sedhom
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Penn Center for Cancer Care Innovation, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Susan Scott
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Amanda Kagan
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Andrew H. Marple
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Jenna V. Canzoniero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Melinda Hsu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | | | - Amin S. Herati
- Department of Urology, Johns Hopkins University, Baltimore, MD
| | - Lauren Reschke
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, MD
| | - Maria Facadio Antero
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, MD
| | | | - Adam F. Binder
- Department of Oncology, Thomas Jefferson University, Philadelphia, PA
| | - Allen R. Chen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Ross C. Donehower
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Kristen A. Marrone
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Arjun Gupta
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
- Division of Hematology, Oncology, Transplantation, University of Minnesota, Minneapolis, MN
| |
Collapse
|
24
|
Wong SK, Nebhan CA, Johnson DB. Impact of Patient Age on Clinical Efficacy and Toxicity of Checkpoint Inhibitor Therapy. Front Immunol 2021; 12:786046. [PMID: 34868071 PMCID: PMC8635107 DOI: 10.3389/fimmu.2021.786046] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/29/2021] [Indexed: 12/19/2022] Open
Abstract
The addition of immune checkpoint inhibitors (ICIs) to the therapeutic armamentarium for solid malignancies has resulted in unprecedented improvements in patient outcomes in many cancers. The landscape of ICIs continues to evolve with novel approaches such as dual immune checkpoint blockade and combination therapies with other anticancer agents including cytotoxic chemotherapies and/or antiangiogenics. However, there is significant heterogeneity seen in antitumor responses, with certain patients deriving durable benefit, others experiencing initial benefit followed by acquired resistance necessitating change in therapy, and still others who are primarily refractory to ICIs. While generally better tolerated than traditional cytotoxic chemotherapy, ICIs are associated with unique toxicities, termed immune-related adverse events (irAEs), which can be severe or even lethal. As a disease of aging, older individuals make up a large proportion of patients diagnosed with cancer, yet this population is often underrepresented in clinical trials. Because ICIs indirectly target malignant cells through T cell activation, it has been hypothesized that age-related changes to the immune system may impact the efficacy and toxicity of these drugs. In this review, we discuss differences in the clinical efficacy and toxicity of ICIs in patients at the extremes of age.
Collapse
Affiliation(s)
- Selina K Wong
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, United States
| | - Caroline A Nebhan
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, United States
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, United States
| |
Collapse
|
25
|
Xu PC, Luan Y, Yu SY, Xu J, Coulter DW, Kim SY. Effects of PD-1 blockade on ovarian follicles in a prepubertal female mouse. J Endocrinol 2021; 252:15-30. [PMID: 34647523 PMCID: PMC8630981 DOI: 10.1530/joe-21-0209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022]
Abstract
Immunotherapy has emerged at the forefront of cancer treatment. Checkpoint inhibitor pembrolizumab (KEYTRUDA), a chimeric antibody which targets programmed cell death protein 1 (PD-1), has been approved by the Food and Drug Administration (FDA) for use in pediatric patients with relapsed or refractory classical Hodgkin's lymphoma. However, there is currently no published data regarding the effects of pembrolizumab on the ovary of female pediatric patients. In this study, prepubertal immunocompetent and immunodeficient female mice were injected with pembrolizumab or anti-mouse PD-1 antibody. The number of primordial follicles significantly decreased post-injection of both pembrolizumab and anti-mouse PD-1 antibody in immunocompetent mice. However, no changes in follicle numbers were observed in immunodeficient nude mice. Superovulation test and vaginal opening experiments suggest that there is no difference in the number of cumulus-oocyte complexes (COCs) and the timing of puberty onset between the control and anti-mouse PD-1 antibody treatment groups, indicating that there is no effect on short-term fertility. Elevation of pro-inflammatory cytokine TNF-α following COX-2 upregulation was observed in the ovary. CD3+ T-cell infiltration was detected within some ovarian follicles and between stromal cells of the ovaries in mice following treatment with anti-mouse PD-1 antibody. Thus, PD-1 immune checkpoint blockade affects the ovarian reserve through a mechanism possibly involving inflammation following CD3+ T-cell infiltration.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacology
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/pharmacology
- Cell Count
- Female
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/pharmacology
- Infertility, Female/chemically induced
- Infertility, Female/pathology
- Mice
- Mice, Nude
- Oocytes/cytology
- Oocytes/drug effects
- Ovarian Follicle/drug effects
- Ovarian Reserve/drug effects
- Ovary/drug effects
- Ovary/physiology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Sexual Maturation/drug effects
Collapse
Affiliation(s)
- Pauline C. Xu
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yi Luan
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Seok-Yeong Yu
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jing Xu
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon; Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Donald W. Coulter
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - So-Youn Kim
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
26
|
Garutti M, Lambertini M, Puglisi F. Checkpoint inhibitors, fertility, pregnancy, and sexual life: a systematic review. ESMO Open 2021; 6:100276. [PMID: 34597942 PMCID: PMC8487000 DOI: 10.1016/j.esmoop.2021.100276] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/09/2021] [Indexed: 01/12/2023] Open
Abstract
Immune checkpoint inhibitors (i.e. anti-PD1, anti-PDL1, and anti-CTLA4) have revolutionized the therapeutic approach of several cancer types. In a subset of metastatic patients, the duration of the response is so long that a cure might be hypothesized, and a treatment discontinuation strategy could be proposed. Considering that long-term efficacy, some patients could also plan to have a child. Moreover, immunotherapy is moving to the early setting in several diseases including melanoma and breast cancer that are common cancers in young patients. However, there is a paucity of data about their potential detrimental effect on fertility, pregnancy, or sexuality. Herein, we conducted a systematic review with the aim to comprehensively collect the available evidence about fertility, pregnancy, and sexual adverse effects of checkpoint inhibitors in order to help clinicians in daily practice and trialists to develop future studies.
Collapse
Affiliation(s)
- M Garutti
- CRO Aviano National Cancer Institute IRCCS, Aviano, Italy.
| | - M Lambertini
- Department of Medical Oncology, Breast Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - F Puglisi
- CRO Aviano National Cancer Institute IRCCS, Aviano, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy; Department of Medicine (DAME), University of Udine, Udine, Italy
| |
Collapse
|
27
|
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
|
28
|
Nagamitsu R, Shiraishi K, Tabara M, Matsuyama H. Effect of tyrosine kinase inhibitors and immune checkpoint inhibitors on spermatogenesis in humans. Int J Urol 2021; 28:1077-1078. [PMID: 34258780 DOI: 10.1111/iju.14646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ryoko Nagamitsu
- Department of Urology, Yamaguchi University School of Medicine, Ube, Yamaguchi, Japan
| | - Koji Shiraishi
- Department of Urology, Yamaguchi University School of Medicine, Ube, Yamaguchi, Japan
| | - Masanori Tabara
- Department of Urology, Yamaguchi University School of Medicine, Ube, Yamaguchi, Japan
| | - Hideyasu Matsuyama
- Department of Urology, Yamaguchi University School of Medicine, Ube, Yamaguchi, Japan
| |
Collapse
|
29
|
Male fertility during and after immune checkpoint inhibitor therapy: A cross-sectional pilot study. Eur J Cancer 2021; 152:41-48. [PMID: 34062486 DOI: 10.1016/j.ejca.2021.04.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/08/2021] [Accepted: 04/22/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are widely used and may induce long-term survival in various types of cancer. Yet, there is scarce evidence on potential effects on patient fertility and the necessity of cryopreservation before treatment onset. The aim of our study was to assess the prevalence of male infertility after initiation of ICI treatment. METHODS This is a monocenter, cross-sectional pilot study. Fertility was investigated by spermiogram, analysis of sexual hormones and questionnaires on sexual function and sexual activity. Male patients under the age of 60 years previously or currently treated with ICI for cutaneous malignancies or uveal melanoma were included. RESULTS Twenty-five patients were included, with a median age of 49 years. Eighteen of 22 (82%) available spermiograms showed no pathologies, all patients reported a normal sexual function and sexual activity. Of four patients with pathological spermiogram, three patients were diagnosed with azoospermia and one with oligoasthenoteratozoospermia. Three patients had significant confounding factors (previous inguinal radiotherapy, chemotherapy and chronic alcohol abuse, and bacterial orchitis). One patient with normal spermiogram before ICI treatment presented 1 year after initiation with azoospermia, showing an asymptomatic, inflammatory infiltrate with predominantly neutrophil granulocytes, macrophages and T-lymphocytes in the ejaculate. Infectious causes were ruled out; andrological examination was unremarkable. A second case with reduced sperm counts during treatment may be ICI-induced also. CONCLUSIONS Most patients had no restrictions in fertility, yet an inflammatory loss of spermatogenesis seems possible. Cryopreservation should be discussed with all patients with potential future desire for children before treatment.
Collapse
|
30
|
Brannigan RE, Fantus RJ, Halpern JA. Fertility preservation in men: a contemporary overview and a look toward emerging technologies. Fertil Steril 2021; 115:1126-1139. [PMID: 33933174 DOI: 10.1016/j.fertnstert.2021.03.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022]
Abstract
Cancer and oncologic therapies can have significant adverse effects on male reproductive potential, leaving many men permanently infertile. Fertility preservation has emerged as a key survivorship issue over the past 20 years, and numerous professional societies have published guidelines calling for fertility preservation to become a routine component of oncologic care. Most males with cancer are able to produce a semen specimen for fertility preservation, but numerous other methods of sperm procurement are available for patients who cannot provide a sufficient sample. Despite these options, fertility preservation will remain a challenge for prepubertal boys and men without sperm production. For these patients, experimental and investigational approaches offer the hope that one day they will translate to the clinical arena, offering additional pathways for successful fertility preservation care.
Collapse
Affiliation(s)
- Robert E Brannigan
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Richard J Fantus
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joshua A Halpern
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
31
|
Özdemir BC. Immune checkpoint inhibitor-related hypogonadism and infertility: a neglected issue in immuno-oncology. J Immunother Cancer 2021; 9:jitc-2020-002220. [PMID: 33589529 PMCID: PMC7887354 DOI: 10.1136/jitc-2020-002220] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2021] [Indexed: 11/08/2022] Open
Abstract
Despite a significant amount of data on incidence and therapy of immune-related adverse events affecting virtually all organ systems, the potential impact of immune checkpoint inhibitors (ICIs) on gonadal function has not been sufficiently studied. The limited evidence available suggests that ICI-related primary hypogonadism due to orchitis as well as secondary hypogonadism due to hypophysitis are a potential risk for infertility. A systematic investigation of gonadal function under ICIs is warranted given the increasing application of ICIs in the adjuvant setting, among young adults and children and the possible influence of sex hormone levels on the efficacy and toxicity of ICIs.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland .,International Cancer Prevention Institute, Lausanne, Switzerland
| |
Collapse
|