1
|
Monge C, Eldridge L, Pearlman PC, Venkatesh V, Tregear M, Loehrer PJ, Galassi A, Gopal S, Ginsburg O. Global perspectives on patient-centered outcomes: advancing patient-centered cancer clinical trials globally. J Natl Cancer Inst Monogr 2025; 2025:35-41. [PMID: 39989041 PMCID: PMC11848040 DOI: 10.1093/jncimonographs/lgae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 02/25/2025] Open
Abstract
Patient-centered clinical trials prioritize the patient experience and outcomes that matter most to those affected by cancer. By centering on patient values and experiences, patient-centered outcomes research generates evidence to inform policies and practices, facilitating more personalized and effective cancer care. This manuscript explores the importance of patient-centered approaches in the global context, emphasizing challenges and opportunities for substantive patient engagement and the integration of patient-reported measures in clinical therapeutic trials in low- and middle-income countries. Despite important barriers such as limited infrastructure and funding constraints, leveraging innovative strategies and investing in research infrastructure and regulatory harmonization initiatives can enhance the capacity of low- and middle-income countries to conduct high-quality research and address the global burden of cancer more effectively. Through these efforts, patient-centered care and research can be extended to underserved populations, ensuring equitable access to cancer care worldwide.
Collapse
Affiliation(s)
- Cecilia Monge
- Center for Global Health, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Linsey Eldridge
- Center for Global Health, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Paul C Pearlman
- Center for Global Health, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Viji Venkatesh
- The Max Foundation, India and South Asia, Seattle, WA, United States
| | | | - Patrick J Loehrer
- Center for Global Health, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Annette Galassi
- Leidos Biomedical Research, Inc, Frederick, MD, United States
| | - Satish Gopal
- Center for Global Health, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ophira Ginsburg
- Center for Global Health, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
2
|
Michaeli JC, Michaeli T, Trapani D, Albers S, Dannehl D, Würstlein R, Michaeli DT. Breast cancer drugs: FDA approval, development time, efficacy, clinical benefits, innovation, trials, endpoints, quality of life, value, and price. Breast Cancer 2024; 31:1144-1155. [PMID: 39320645 PMCID: PMC11489271 DOI: 10.1007/s12282-024-01634-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
OBJECTIVE This study analyzes the development, benefits, trial evidence, and price of new breast cancer drugs with US Food and Drug Administration (FDA) approval. METHODS We identified 26 drugs with 42 FDA-approved indications for early and metastatic breast cancer (2000-2023). Data were collected from FDA labels, clinicaltrials.gov, and Medicare and Medicaid. Overall survival (OS) and progression-free survival (PFS) hazard ratios (HRs) and tumor response's relative risk (RR) alongside objective response rate (ORR) were meta-analyzed. RESULTS The median development time for breast cancer drugs was 7.8 years (95% CI 6.2-10.8). 26% of treatments were considered innovative ("first-in-indication") with 88% acting via a targeted mechanism. 64% were small molecules, 19% antibodies, and 18% antibody-drug conjugates. 38% were approved for HR + and 31% for HER2 + breast cancer. 6 indications were for early and 36 for metastatic breast cancer. Indications utilized FDA's special programs: orphan (2%), fast track (24%), accelerated approval (19%), priority review (74%), breakthrough therapy (44%). Approval was predominantly supported by phase 3 trials (88%) of randomized controlled design (66%), enrolling a median of 585 patients (IQR 417-752) at 181 centers (IQR 142-223) across 19 countries (IQR 17-20). New drugs' HR were 0.78 for OS (95% CI 0.74-0.82) and 0.59 for PFS (95% CI 0.54-0.64) with a RR for tumor response of 1.61 (95% CI 1.46-1.76). Median improvements of OS were 2.8 months (IQR 1.8-5.8) and PFS were 4.4 months (IQR 2.2-7.1). In single-arm trials, the average ORR was 31% (95% CI 10-53). In meta-regressions, the correlation between OS/PFS was 0.34 (p = 0.031) and OS/response was 0.01 (p = 0.435). 60% of treatments had a 'high-value' ESMO-MCBS score with 14% demonstrating improvements in quality of life. The median price was $16,013 per month (95% CI 13,097-17,617). There was no association between prices and patient benefit. The median value per life year gained was $62,419 (IQR 25,840-86,062). CONCLUSIONS Over the past two decades, the development of innovative and effective drugs transformed the treatment landscape for breast cancer patients. Yet, investigators and regulators must safeguard that highly-priced new drugs demonstrate improvements in patient-centered clinical endpoints: overall survival and quality of life.
Collapse
Affiliation(s)
- Julia Caroline Michaeli
- Department of Obstetrics and Gynaecology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Thomas Michaeli
- Department of Personalized Oncology, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dario Trapani
- European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Sebastian Albers
- Department of Orthopaedics and Sport Orthopaedics, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Dominik Dannehl
- Department of Women's Health, Tuebingen University Hospital, Tübingen, Germany
| | - Rachel Würstlein
- Department of Obstetrics and Gynaecology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Daniel Tobias Michaeli
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany.
| |
Collapse
|
3
|
Kiesewetter B, Dafni U, de Vries EGE, Barriuso J, Curigliano G, González-Calle V, Galotti M, Gyawali B, Huntly BJP, Jäger U, Latino NJ, Malcovati L, Oosting SF, Ossenkoppele G, Piccart M, Raderer M, Scarfò L, Trapani D, Zielinski CC, Wester R, Zygoura P, Macintyre E, Cherny NI. ESMO-Magnitude of Clinical Benefit Scale for haematological malignancies (ESMO-MCBS:H) version 1.0. Ann Oncol 2023; 34:734-771. [PMID: 37343663 DOI: 10.1016/j.annonc.2023.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND The European Society for Medical Oncology (ESMO)-Magnitude of Clinical Benefit Scale (MCBS) has been accepted as a robust tool to evaluate the magnitude of clinical benefit reported in trials for oncological therapies. However, the ESMO-MCBS hitherto has only been validated for solid tumours. With the rapid development of novel therapies for haematological malignancies, we aimed to develop an ESMO-MCBS version that is specifically designed and validated for haematological malignancies. METHODS ESMO and the European Hematology Association (EHA) initiated a collaboration to develop a version for haematological malignancies (ESMO-MCBS:H). The process incorporated five landmarks: field testing of the ESMO-MCBS version 1.1 (v1.1) to identify shortcomings specific to haematological diseases, drafting of the ESMO-MCBS:H forms, peer review and revision of the draft based on re-scoring (resulting in a second draft), assessment of reasonableness of the scores generated, final review and approval by ESMO and EHA including executive boards. RESULTS Based on the field testing results of 80 haematological trials and extensive review for feasibility and reasonableness, five amendments to ESMO-MCBS were incorporated in the ESMO-MCBS:H addressing the identified shortcomings. These concerned mainly clinical trial endpoints that differ in haematology versus solid oncology and the very indolent nature of nevertheless incurable diseases such as follicular lymphoma, which hampers presentation of mature data. In addition, general changes incorporated in the draft version of the ESMO-MCBS v2 were included, and specific forms for haematological malignancies generated. Here we present the final approved forms of the ESMO-MCBS:H, including instructions. CONCLUSION The haematology-specific version ESMO-MCBS:H allows now full applicability of the scale for evaluating the magnitude of clinical benefit derived from clinical studies in haematological malignancies.
Collapse
Affiliation(s)
- B Kiesewetter
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - U Dafni
- Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian University of Athens, Athens; Frontier Science Foundation-Hellas, Athens, Greece
| | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J Barriuso
- The Christie NHS Foundation Trust and Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - G Curigliano
- European Institute of Oncology, IRCCS, Division of Early Drug Development, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - V González-Calle
- Servicio de Hematología, Hospital Universitario de Salamanca-IBSAL, CIBERONC and Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
| | - M Galotti
- ESMO Head Office, Lugano, Switzerland
| | - B Gyawali
- Departments of Oncology, Oncology; Public Health Sciences, Queen's University, Kingston; Division of Cancer Care and Epidemiology, Queen's University, Kingston, Canada
| | - B J P Huntly
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge & Cambridge University Hospitals, Cambridge, UK
| | - U Jäger
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | | | - L Malcovati
- Department of Molecular Medicine, University of Pavia, Pavia; Department of Hematology Oncology, IRCCS S. Matteo Hospital Foundation, Pavia, Italy
| | - S F Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - G Ossenkoppele
- Department of Haematology, VU University Medical Center, Amsterdam, The Netherlands
| | - M Piccart
- Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - M Raderer
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - L Scarfò
- Strategic Research Program on CLL, Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy
| | - D Trapani
- European Institute of Oncology, IRCCS, Division of Early Drug Development, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - C C Zielinski
- Wiener Privatklinik, Central European Academy Cancer Center, Vienna, Austria
| | - R Wester
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - P Zygoura
- Frontier Science Foundation-Hellas, Athens, Greece
| | - E Macintyre
- Onco-hématologie Biologique, AP-HP, Necker-Enfants Malades Hospital, Paris; Université Paris Cité, INSERM, CNRS, INEM F-75015, Paris, France
| | - N I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel.
| |
Collapse
|
4
|
Delgado-Waldo I, Contreras-Romero C, Salazar-Aguilar S, Pessoa J, Mitre-Aguilar I, García-Castillo V, Pérez-Plasencia C, Jacobo-Herrera NJ. A triple-drug combination induces apoptosis in cervical cancer-derived cell lines. Front Oncol 2023; 13:1106667. [PMID: 37223676 PMCID: PMC10200932 DOI: 10.3389/fonc.2023.1106667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/28/2023] [Indexed: 05/25/2023] Open
Abstract
Introduction Cervical cancer is a worldwide health problem due to the number of deaths caused by this neoplasm. In particular, in 2020, 30,000 deaths of this type of tumor were reported in Latin America. Treatments used to manage patients diagnosed in the early stages have excellent results as measured by different clinical outcomes. Existing first-line treatments are not enough to avoid cancer recurrence, progression, or metastasis in locally advanced and advanced stages. Therefore, there is a need to continue with the proposal of new therapies. Drug repositioning is a strategy to explore known medicines as treatments for other diseases. In this scenario, drugs used in other pathologies that have antitumor activity, such as metformin and sodium oxamate, are analyzed. Methods In this research, we combined the drugs metformin and sodium oxamate with doxorubicin (named triple therapy or TT) based on their mechanism of action and previous investigation of our group against three CC cell lines. Results Through flow cytometry, Western blot, and protein microarray experiments, we found TT-induced apoptosis on HeLa, CaSki, and SiHa through the caspase 3 intrinsic pathway, including the critical proapoptotic proteins BAD, BAX, cytochrome-C, and p21. In addition, mTOR and S6K phosphorylated proteins were inhibited in the three cell lines. Also, we show an anti-migratory activity of the TT, suggesting other targets of the drug combination in the late CC stages. Discussion These results, together with our former studies, conclude that TT inhibits the mTOR pathway leading to cell death by apoptosis. Our work provides new evidence of TT against cervical cancer as a promising antineoplastic therapy.
Collapse
Affiliation(s)
- Izamary Delgado-Waldo
- Unidad de Bioquímica Guillermo Soberón Acevedo, Instituto de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México. Copilco Universidad, Coyoacán, Mexico
| | - Carlos Contreras-Romero
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México. Copilco Universidad, Coyoacán, Mexico
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico
| | - Sandra Salazar-Aguilar
- Laboratorio de Hematopoiesis y Leucemia, Unidad de Investigación, Diferenciación Celular y Cáncer, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Iztapalapa, Mexico
| | - João Pessoa
- CNC - Center for Neuroscience and Cell Biology, CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Irma Mitre-Aguilar
- Unidad de Bioquímica Guillermo Soberón Acevedo, Instituto de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico
| | - Verónica García-Castillo
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-IZTACALA, Universidad Nacional Autónoma de México, Tlalnepantla, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, FES-IZTACALA, Universidad Nacional Autónoma de México, Tlalnepantla, Mexico
| | - Nadia Judith Jacobo-Herrera
- Unidad de Bioquímica Guillermo Soberón Acevedo, Instituto de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico
| |
Collapse
|
5
|
Epstein RJ, Gu Y, Lin FPY. Can cancer go green? It's up to us. Front Oncol 2023; 13:1074091. [PMID: 36910667 PMCID: PMC9992733 DOI: 10.3389/fonc.2023.1074091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/17/2023] [Indexed: 02/24/2023] Open
Affiliation(s)
- Richard J Epstein
- New Hope Cancer Center, Beijing United Hospital, Beijing, China.,Department of Medicine, University of New South Wales, Sydney, NSW, Australia.,Cancer Programme, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Yanfei Gu
- New Hope Cancer Center, Beijing United Hospital, Beijing, China
| | - Frank P Y Lin
- Department of Medicine, University of New South Wales, Sydney, NSW, Australia.,Cancer Programme, Garvan Institute of Medical Research, Sydney, NSW, Australia.,National Health & Medical Research Council (NHMRC) Clinical Trials Centre, Sydney University, Sydney, NSW, Australia
| |
Collapse
|
6
|
Wolff L, Kiesewetter B. Applicability of ESMO-MCBS and ESCAT for molecular tumor boards. MEMO - MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2022. [DOI: 10.1007/s12254-022-00800-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SummaryScoring systems for classifying genomic alterations (GAs) with respect to their potential targeted anticancer therapies (TTs) may be useful for rational and evidence-based decision-making, for example in molecular tumor boards. Therefore, a working group of the European Society for Medical Oncology (ESMO) has developed a comprehensive and reproducible classification score that allows the ranking of GAs and TTs according to their level of evidence and clinical relevance. This score is called the ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT). Another score not explicitly developed for TTs but helpful in grading novel TTs is the ESMO-Magnitude of Clinical Benefit Scale (ESMO-MCBS). This tool was designed to objectively quantify the clinical benefit of novel approved therapies. The current review summarizes the status quo of these scores and their applicability for molecular tumor boards.
Collapse
|
7
|
Barth DA, Stanzer S, Spiegelberg JA, Bauernhofer T, Absenger G, Szkandera J, Gerger A, Smolle MA, Hutterer GC, Ahyai SA, Madl T, Posch F, Riedl JM, Klec C, Jost PJ, Kargl J, Stradner MH, Pichler M. Patterns of Peripheral Blood B-Cell Subtypes Are Associated With Treatment Response in Patients Treated With Immune Checkpoint Inhibitors: A Prospective Longitudinal Pan-Cancer Study. Front Immunol 2022; 13:840207. [PMID: 35432362 PMCID: PMC9010871 DOI: 10.3389/fimmu.2022.840207] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/10/2022] [Indexed: 01/18/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have revolutionized systemic anti-tumor treatments across different types of cancer. Nevertheless, predictive biomarkers regarding treatment response are not routinely established yet. Apart from T-lymphocytes, the humoral immunity of B-lymphocytes is studied to a substantially lesser extent in the respective setting. Thus, the aim of this study was to evaluate peripheral blood B-cell subtypes as potential predictors of ICI treatment response. Methods Thirty-nine cancer patients receiving ICI therapy were included into this prospective single-center cohort study. All had a first blood draw at the date before treatment initiation and a second at the time of first response evaluation (after 8-12 weeks). Seven different B-cell subtypes were quantified by fluorescence-activated cell sorting (FACS). Disease control- (DCR) and objective response rate (ORR) were co-primary study endpoints. Results Overall, DCR was 48.7% and ORR was 25.6%, respectively. At baseline, there was no significant association of any B-cell subtype with neither DCR nor ORR. At the first response evaluation, an increase in the frequency of CD21- B-cells was a statistically significant negative predictor of response, both regarding DCR (OR=0.05, 95%CI=0.00-0.67, p=0.024) and ORR (OR=0.09, 95%CI=0.01-0.96, p=0.046). An increase of the frequency of switched memory B-cells was significantly associated with reduced odds for DCR (OR=0.06, 95%CI=0.01-0.70, p=0.025). Patients with an increased frequency of naïve B-cells were more likely to benefit from ICI therapy as indicated by an improved DCR (OR=12.31, 95%CI=1.13-134.22, p=0.039). Conclusion In this study, certain B-cell subpopulations were associated with ICI treatment response in various human cancer types.
Collapse
Affiliation(s)
- Dominik A Barth
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Stefanie Stanzer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Jasmin A Spiegelberg
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Thomas Bauernhofer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Gudrun Absenger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Joanna Szkandera
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Maria A Smolle
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria
| | - Georg C Hutterer
- Department of Urology, Medical University of Graz, Graz, Austria
| | - Sascha A Ahyai
- Department of Urology, Medical University of Graz, Graz, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Florian Posch
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Jakob M Riedl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Christiane Klec
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp J Jost
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Martin H Stradner
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
8
|
Barth DA, Stanzer S, Spiegelberg J, Bauernhofer T, Absenger G, Posch F, Lipp R, Halm M, Szkandera J, Balic M, Gerger A, Smolle MA, Hutterer GC, Klec C, Jost PJ, Kargl J, Stradner M, Pichler M. Evaluation of autoantibodies as predictors of treatment response and immune-related adverse events during the treatment with immune checkpoint inhibitors: A prospective longitudinal pan-cancer study. Cancer Med 2022; 11:3074-3083. [PMID: 35297215 PMCID: PMC9385593 DOI: 10.1002/cam4.4675] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/20/2022] [Accepted: 03/04/2022] [Indexed: 12/19/2022] Open
Abstract
Background The presence of autoantibodies in the serum of cancer patients has been associated with immune‐checkpoint inhibitor (ICI) therapy response and immune‐related adverse events (irAEs). A prospective evaluation of different autoantibodies in different cancer entities is missing. Materials and Methods In this prospective cohort study, we included a pan‐cancer cohort of patients undergoing ICI treatment and measured a comprehensive panel of autoantibodies at treatment start and at the time point of first response evaluation. The presence and induction of autoantibodies (ANA, ENA, myositis, hepatopathy, rheumatoid arthritis) in different cancer entities were assessed and the association between autoantibodies and disease control rate (DCR), objective response rate (ORR), and progression‐free survival (PFS), as well as the development of grade 3 or higher irAEs were evaluated by logistic regression models, cox proportional hazard models, and Kaplan–Meier estimators. Results Of 44 patients with various cancer entities, neither the presence of any positive autoantibody measurement nor the presence of positive antinuclear antibodies (ANA) [≥1:80] at baseline was associated with the examined clinical endpoints (DCR, ORR, PFS) in univariable and multivariable analyses. After 8–12 weeks of ICI treatment, DCR, ORR, and PFS did not significantly differ between patients with and without any positive autoantibody measurement or positive ANA titers. The frequency of irAEs did not differ depending on autoantibody status of the patients. Conclusion Autoantibodies at treatment initiation or induction after 8–12 weeks of ICI treatment are not associated with treatment efficacy as indicated by DCR, ORR, and PFS or higher grade irAEs.
Collapse
Affiliation(s)
- Dominik A Barth
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Stefanie Stanzer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Jasmin Spiegelberg
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Thomas Bauernhofer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Gudrun Absenger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Posch
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.,Division of Haematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Rainer Lipp
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Michael Halm
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Joanna Szkandera
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Marija Balic
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Maria A Smolle
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria
| | - Georg C Hutterer
- Department of Urology, Medical University of Graz, Graz, Austria
| | - Christiane Klec
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp J Jost
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - Martin Stradner
- Division of Rheumatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|