1
|
Li J, Tao M, Liu L, Liu C, Ma M, Liu D, Zhang P, Zhang M, Xue R, Gong J, Zhang C, Zhang X, Shen L, Qi C. Peripheral blood neutrophils contribute to Claudin18.2-specific CAR-T cell treatment resistance in advanced gastric cancer. Br J Cancer 2025:10.1038/s41416-025-03015-3. [PMID: 40246985 DOI: 10.1038/s41416-025-03015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Claudin18.2 (CLDN18.2)-specific chimeric antigen receptor (CAR)-T cell treatment holds promise for advanced gastric cancer (GC) but has variable efficacy. This study investigates the prognostic value of the neutrophil-to-lymphocyte ratio (NLR) in CAR-T cell treatment and elucidates the molecular mechanisms of treatment resistance. METHODS GC patients treated with CLDN18.2-specific CAR-T cell treatment were analyzed. Outcomes included objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Survival analyses utilized Kaplan-Meier methods, log-rank tests, and Cox regression. Single-cell RNA sequencing was performed on peripheral blood samples to investigate the mechanisms of pro-tumor circulating neutrophils. RESULTS Elevated NLR was significantly associated with lower ORR (34.2% vs. 55.9%, P < 0.001), shorter median PFS (3.6 vs. 8.0 months, P < 0.001), and OS (5.6 vs. 13.8 months, P < 0.001). Single-cell sequencing identified a circulating neutrophil subcluster (NE-3) linked to disease progression. NE-3 expressed pro-tumoral factors (MMP-9), and was enriched in the IL-17 signaling pathway. The cellular interactions between neutrophils and T cells were more prominent in progression disease (PD) group than in partial response (PR) group. CONCLUSIONS This study highlights NLR as a significant prognostic factor in advanced GC patients receiving CLDN18.2-specific CAR-T cell treatment and provides insights into neutrophil-mediated treatment resistance. Further validation and exploration of strategies to mitigate neutrophil-induced immunosuppression are needed. TRIAL REGISTRATION NCT03874897.
Collapse
Affiliation(s)
- Jiarui Li
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Min Tao
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lian Liu
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chang Liu
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Mingyang Ma
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dan Liu
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Panpan Zhang
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Miao Zhang
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ran Xue
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Cheng Zhang
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Lin Shen
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Changsong Qi
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
2
|
Salazar-Riojas R, Alvarado-Navarro DM, Chávez-Estrada YO, Hernández-Navarro AK, Ake-Uc MB, Moncada-Saucedo NK, Jaime-Pérez JC, Quezada-Ramírez SI, Rodriguez-Zuñiga AC, Gómez-Almaguer D, Gómez-De León A. Decentralized Point-of-Care Manufacturing of CD19 Chimeric Antigen Receptor T Cells in Mexico. JCO Glob Oncol 2025; 11:e2400581. [PMID: 40249888 DOI: 10.1200/go-24-00581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/22/2025] [Accepted: 02/21/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE To validate and replicate an automated decentralized CD19 chimeric antigen receptor T (CAR-T) cell manufacturing process from healthy adult volunteers in an academic institution in a middle-income country. METHODS Healthy volunteers were recruited and underwent leukapheresis with the continuous mononuclear cell (MNC) collection protocol. Clinical-grade CAR-T cell manufacturing was performed in a closed system using a second-generation CD19 vector with 41BB costimulatory domain. Quality control was assessed at different points in the production process with prespecified release criteria including product's aspect, sterility, cell viability, impurity, and quantity. The target dose formulation was 1 × 106/kg viable CAR-T cells per volunteer. RESULTS Five healthy volunteers were recruited, all donated adequate MNC units, and successfully underwent the manufacturing process. After T-cell culture harvest, the products contained a median CAR-T cell concentration of 16.5 × 106/mL (range, 7.7-22.2 × 106/mL), with a median transduction percentage of 44.7% (range, 39.2%-60.5%) and a median CD3+ cell viability of 97.7% (range, 90.4%-98.7%). Sterility was maintained throughout the manufacturing process. The quantity of cells harvested per kilogram of body weight was 24.6 MB-CART19.1 cells × 106/kg (range, 9.3-33.1 × 106/mL). The quality was similar in both fresh and cryopreserved units. Dose formulations were 1.1 CAR-T cells × 106/kg (range, 1.0-1.2 CAR-T cells × 106/kg). CONCLUSION Our study demonstrates an effective methodology with satisfactory and comparable performance to international reports. Point-of-care manufacturing is a feasible alternative to increase access to CAR-T cells in academic centers.
Collapse
Affiliation(s)
- Rosario Salazar-Riojas
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Dalila M Alvarado-Navarro
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Yair O Chávez-Estrada
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Ana K Hernández-Navarro
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Martha B Ake-Uc
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Nidia K Moncada-Saucedo
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - José C Jaime-Pérez
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Sofía I Quezada-Ramírez
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Anna C Rodriguez-Zuñiga
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - David Gómez-Almaguer
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Andrés Gómez-De León
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| |
Collapse
|
3
|
Yoshida S, Hayashi H, Kawahara T, Katsuki S, Kimura M, Hino R, Sun J, Nakamaru R, Tenma A, Toyoura M, Baba S, Shimamura M, Katsuya T, Morishita R, Rakugi H, Matoba T, Nakagami H. A Vaccine Against Fibroblast Activation Protein Improves Murine Cardiac Fibrosis by Preventing the Accumulation of Myofibroblasts. Circ Res 2025; 136:26-40. [PMID: 39629565 DOI: 10.1161/circresaha.124.325017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/06/2024] [Accepted: 11/24/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Myofibroblasts are primary cells involved in chronic response-induced cardiac fibrosis. Fibroblast activation protein (FAP) is a relatively specific marker of activated myofibroblasts and a potential target molecule. This study aimed to clarify whether a vaccine targeting FAP could eliminate myofibroblasts in chronic cardiac stress model mice and reduce cardiac fibrosis. METHODS We coadministered a FAP peptide vaccine with a cytosine-phosphate-guanine (CpG) K3 oligonucleotide adjuvant to male C57/BL6J mice and confirmed an elevation in the anti-FAP antibody titer. After continuous angiotensin II and phenylephrine administration for 28 days, we evaluated the degree of cardiac fibrosis and the number of myofibroblasts in cardiac tissues. RESULTS We found that cardiac fibrosis was significantly decreased in the FAP-vaccinated mice compared with the angiotensin II and phenylephrine control mice (3.45±1.11% versus 8.62±4.79%; P=4.59×10-3) and that the accumulation of FAP-positive cells was also significantly decreased, as indicated by FAP immunohistochemical staining (4077±1746 versus 7327±1741 cells/mm2; FAP vaccine versus angiotensin II and phenylephrine control; P=6.67×10-3). No systemic or organ-specific inflammation due to antibody-dependent cell cytotoxicity induced by the FAP vaccine was observed. Although the transient activation of myofibroblasts has an important role in maintaining the structural robustness in the process of tissue repair, the FAP vaccine showed no adverse effects in myocardial infarction and skin injury models. CONCLUSIONS Our study demonstrates the FAP vaccine can be a therapeutic tool for cardiac fibrosis.
Collapse
Affiliation(s)
- Shota Yoshida
- Department of Geriatric and General Medicine (S.Y., S.B., H.R.), Osaka University Graduate School of Medicine, Japan
- Department of Health Development and Medicine (S.Y., H.H., J.S., S.B., H.N.), Osaka University Graduate School of Medicine, Japan
| | - Hiroki Hayashi
- Department of Health Development and Medicine (S.Y., H.H., J.S., S.B., H.N.), Osaka University Graduate School of Medicine, Japan
| | - Takuro Kawahara
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (T. Kawahara, S.K., M.K., R.H., T.M.)
- Division of Cardiovascular Medicine, Faculty of Medical Sciences, Research Institute of Angiocardiology, Kyushu University, Fukuoka, Japan (T. Kawahara)
| | - Shunsuke Katsuki
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (T. Kawahara, S.K., M.K., R.H., T.M.)
| | - Mitsukuni Kimura
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (T. Kawahara, S.K., M.K., R.H., T.M.)
| | - Rissei Hino
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (T. Kawahara, S.K., M.K., R.H., T.M.)
| | - Jiao Sun
- Department of Health Development and Medicine (S.Y., H.H., J.S., S.B., H.N.), Osaka University Graduate School of Medicine, Japan
| | - Ryo Nakamaru
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Healthcare Quality Assessment, the University of Tokyo, Japan (R.N.)
| | | | | | - Satoshi Baba
- Department of Geriatric and General Medicine (S.Y., S.B., H.R.), Osaka University Graduate School of Medicine, Japan
- Department of Health Development and Medicine (S.Y., H.H., J.S., S.B., H.N.), Osaka University Graduate School of Medicine, Japan
| | - Munehisa Shimamura
- Department of Gene and Stem Cell Regenerative Therapy (M.S.), Osaka University Graduate School of Medicine, Japan
- Department of Neurology (M.S.), Osaka University Graduate School of Medicine, Japan
| | - Tomohiro Katsuya
- Department of Clinical Gene Therapy (T. Katsuya, R.M.), Osaka University Graduate School of Medicine, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy (T. Katsuya, R.M.), Osaka University Graduate School of Medicine, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine (S.Y., S.B., H.R.), Osaka University Graduate School of Medicine, Japan
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (T. Kawahara, S.K., M.K., R.H., T.M.)
| | - Hironori Nakagami
- Department of Health Development and Medicine (S.Y., H.H., J.S., S.B., H.N.), Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
4
|
Sainatham C, Yadav D, Dilli Babu A, Tallapalli JR, Kanagala SG, Filippov E, Murillo Chavez F, Ahmed N, Lutfi F. The current socioeconomic and regulatory landscape of immune effector cell therapies. Front Med (Lausanne) 2024; 11:1462307. [PMID: 39697210 PMCID: PMC11652178 DOI: 10.3389/fmed.2024.1462307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Immune cell effector therapies, including chimeric antigen receptor (CAR)-T cells, T-cell receptor (TCR) T cells, natural killer (NK) cells, and macrophage-based therapies, represent a transformative approach to cancer treatment, harnessing the immune system to target and eradicate malignant cells. CAR-T cell therapy, the most established among these, involves engineering T cells to express CARs specific to cancer cell antigens, showing remarkable efficacy in hematologic malignancies like leukemias, B-cell lymphomas, and multiple myeloma. Similarly, TCR-modified therapies, which reprogram T cells to recognize intracellular tumor antigens presented by major histocompatibility complex (MHC) molecules, offer promise for a range of solid tumors. NK-cell therapies leverage NK cells' innate cytotoxicity, providing an allogeneic approach that avoids some of the immune-related complications associated with T-cell-based therapies. Macrophage-based therapies, still in early stages of the development, focus on reprogramming macrophages to stimulate an immune response against cancer cells in the tumor microenvironment. Despite their promise, socioeconomic and regulatory challenges hinder the accessibility and scalability of immune cell effector therapies. These treatments are costly, with CAR-T therapies currently exceeding $400,000 per patient, creating significant disparities in access based on socioeconomic status and geographic location. The high manufacturing costs stem from the personalized, labor-intensive processes of harvesting, modifying, and expanding patients' cells. Moreover, complex logistics for manufacturing and delivering these therapies limit their reach, particularly in low-resource settings. Regulatory pathways further complicate the landscape. In the United States., the Food and Drug Administrations' (FDA) accelerated approval processes for cell-based therapies facilitate innovation but do not address cost-related barriers. In Europe, the European Medicines Agency (EMA) offers adaptive pathways, yet decentralized reimbursement systems create uneven access across member states. Additionally, differing regulatory standards for manufacturing and quality control worldwide pose hurdles for global harmonization and access. To expand the reach of immune effector cell therapies, a multipronged approach is needed-streamlined regulatory frameworks, policies to reduce treatment costs, and international collaborations to standardize manufacturing. Addressing these socioeconomic and regulatory obstacles is essential to make these life-saving therapies accessible to a broader patient population worldwide. We present a literature review on the current landscape of immune effector cell therapies and barriers of access to currently approved standard of care therapy at various levels.
Collapse
Affiliation(s)
- Chiranjeevi Sainatham
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Devvrat Yadav
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Aravind Dilli Babu
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Jayanth Reddy Tallapalli
- Division of Infectious Diseases, Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Sai Gautham Kanagala
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital Center, New York, NY, United States
| | - Evgenii Filippov
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Franco Murillo Chavez
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Nausheen Ahmed
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Forat Lutfi
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
5
|
Laetsch T, Zhang J, Yang H, Xie Y, Zhang D, Garrison L. Evolving Evidence-Based Value Assessment of One-Time Therapies: Tisagenlecleucel as a Case Study. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2024; 22:749-765. [PMID: 38683438 PMCID: PMC11339080 DOI: 10.1007/s40258-024-00882-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Economic evaluation of one-time therapies during reimbursement decision-making is challenging due to uncertain long-term outcomes. The availability of 5-year outcome data from the ELIANA trial and real-world evidence of tisagenlecleucel, the first chimeric antigen receptor T-cell (CAR-T) therapy, presents an opportunity to re-evaluate the predictions of prior cost-effectiveness analyses (CEAs). OBJECTIVE To conduct a systematic literature review (SLR) of prior CEAs of tisagenlecleucel for pediatric/young adult relapsed or refractory acute lymphoblastic leukemia (r/r ALL) and evaluate the impact of recently available 5-year efficacy data from ELIANA and advances in CAR-T manufacturing in an updated CEA model. METHODS OVID MEDLINE/Embase and health technology assessment (HTA) databases were searched for full-text economic evaluations in English reporting cost-effectiveness results for tisagenlecleucel for r/r ALL. Evaluations with publicly reported incremental cost-effectiveness ratios (ICERs) were included in the SLR. Study screening and data abstraction were conducted following PRISMA guidelines. Data extracted included the country/currency, perspective, clinical trial evidence, model structures, long-term efficacy extrapolation approaches (i.e., overall survival [OS]), time horizon, discount rates, and outcomes (i.e., life years [LY], quality-adjusted LY [QALY], and ICERs). The CEA model reported in Wakase et al. was updated using 5-year OS data from ELIANA and the CAR-T infusion rate informed by real-world practice. RESULTS Sixteen records corresponding to 15 unique studies were included in the SLR (11 publications and 5 HTA reports); all were conducted from the health care system perspective of the respective countries. Most studies found tisagenlecleucel to be cost effective, but all studies' projected 3- and 5-year OS rates for tisagenlecleucel were lower than the observed 3- and 5-year rates, respectively, derived from 5-year ELIANA data. When applying updated OS projections from the most recent ELIANA data cut and higher infusion rates of 92.5% (per the real-world infusion rate)-96.0% (per the manufacturer success rate) to the CEA of Wakase et al., the associated QALYs for tisagenlecleucel increased from 11.6 to 14.6-15.0, and LYs increased from 13.3 to 17.0-17.5. Accordingly, the ICERs for tisagenlecleucel decreased from ¥2,035,071 to ¥1,787,988-¥1,789,048 versus blinatumomab and from ¥2,644,702 to ¥2,257,837-¥2,275,181 versus clofarabine combination therapy in the updated CEA model. CONCLUSIONS AND RELEVANCE Projections at launch of the likely cost effectiveness of tisagenlecleucel appear to have underestimated its ultimate economic value given more recent trial and real-world data. To balance uncertainty in initial valuation with the need to provide access to novel oncology therapies, payers can consider flexible reimbursement policies alongside ongoing assessments as new data emerge.
Collapse
Affiliation(s)
| | - Jie Zhang
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | | | | - Louis Garrison
- School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Thavorn K, Thompson ER, Kumar S, Heiskanen A, Agarwal A, Atkins H, Shorr R, Hawrysh T, Chan KKW, Presseau J, Ollendorf DA, Graham ID, Grimshaw JM, Lalu MM, Nochaiwong S, Fergusson DA, Hutton B, Coyle D, Kekre N. Economic Evaluations of Chimeric Antigen Receptor T-Cell Therapies for Hematologic and Solid Malignancies: A Systematic Review. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:1149-1173. [PMID: 38641057 DOI: 10.1016/j.jval.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/21/2024]
Abstract
OBJECTIVES This study aimed to systematically review evidence on the cost-effectiveness of chimeric antigen receptor T-cell (CAR-T) therapies for patients with cancer. METHODS Electronic databases were searched in October 2022 and updated in September 2023. Systematic reviews, health technology assessments, and economic evaluations that compared costs and effects of CAR-T therapy in patients with cancer were included. Two reviewers independently screened studies, extracted data, synthesized results, and critically appraised studies using the Philips checklist. Cost data were presented in 2022 US dollars. RESULTS Our search yielded 1809 records, 47 of which were included. Most of included studies were cost-utility analysis, published between 2018 and 2023, and conducted in the United States. Tisagenlecleucel, axicabtagene ciloleucel, idecabtagene vicleucel, ciltacabtagene autoleucel, lisocabtagene maraleucel, brexucabtagene autoleucel, and relmacabtagene autoleucel were compared with various standard of care chemotherapies. The incremental cost-effectiveness ratio (ICER) for CAR-T therapies ranged from $9424 to $4 124 105 per quality-adjusted life-year (QALY) in adults and from $20 784 to $243 177 per QALY in pediatric patients. Incremental cost-effectiveness ratios were found to improve over longer time horizons or when an earlier cure point was assumed. Most studies failed to meet the Philips checklist due to a lack of head-to-head comparisons and uncertainty surrounding CAR-T costs and curative effects. CONCLUSIONS CAR-T therapies were more expensive and generated more QALYs than comparators, but their cost-effectiveness was uncertain and dependent on patient population, cancer type, and model assumptions. This highlights the need for more nuanced economic evaluations and continued research to better understand the value of CAR-T therapies in diverse patient populations.
Collapse
Affiliation(s)
- Kednapa Thavorn
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada; Pharmacoepidemiology and Statistics Research Center, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand.
| | - Emily Rose Thompson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada
| | - Srishti Kumar
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada
| | - Aliisa Heiskanen
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Anubhav Agarwal
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Harold Atkins
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Transplant and Cell Therapy Program, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Risa Shorr
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada
| | - Terry Hawrysh
- Patient Partner, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada
| | | | - Justin Presseau
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Daniel A Ollendorf
- Center for the Evaluation of Value and Risk in Health, Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA; Institute for Clinical and Economic Review, Boston, MA, USA
| | - Ian D Graham
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Jeremy M Grimshaw
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Manoj Mathew Lalu
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; Department of Anesthesiology and Pain Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Surapon Nochaiwong
- Pharmacoepidemiology and Statistics Research Center, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Dean A Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Brian Hutton
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Doug Coyle
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Natasha Kekre
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, General Campus, Ottawa, ON, Canada; Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Transplant and Cell Therapy Program, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| |
Collapse
|
7
|
Dixon KJ, Snyder KM, Khaw M, Hullsiek R, Davis ZB, Matson AW, Shirinbak S, Hancock B, Bjordahl R, Hosking M, Miller JS, Valamehr B, Wu J, Walcheck B. iPSC-derived NK cells expressing high-affinity IgG Fc receptor fusion CD64/16A to mediate flexible, multi-tumor antigen targeting for lymphoma. Front Immunol 2024; 15:1407567. [PMID: 39100677 PMCID: PMC11294090 DOI: 10.3389/fimmu.2024.1407567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/21/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction NK cells can mediate tumor cell killing by natural cytotoxicity and by antibody-dependent cell-mediated cytotoxicity (ADCC), an anti-tumor mechanism mediated through the IgG Fc receptor CD16A (FcγRIIIA). CD16A polymorphisms conferring increased affinity for IgG positively correlate with clinical outcomes during monoclonal antibody therapy for lymphoma, linking increased binding affinity with increased therapeutic potential via ADCC. We have previously reported on the FcγR fusion CD64/16A consisting of the extracellular region of CD64 (FcγRI), a high-affinity Fc receptor normally expressed by myeloid cells, and the transmembrane/cytoplasmic regions of CD16A, to create a highly potent and novel activating fusion receptor. Here, we evaluate the therapeutic potential of engineered induced pluripotent stem cell (iPSC)-derived NK (iNK) cells expressing CD64/16A as an "off-the-shelf", antibody-armed cellular therapy product with multi-antigen targeting potential. Methods iNK cells were generated from iPSCs engineered to express CD64/16A and an interleukin (IL)-15/IL-15Rα fusion (IL-15RF) protein for cytokine independence. iNK cells and peripheral blood NK cells were expanded using irradiated K562-mbIL21-41BBL feeder cells to examine in in vitro and in vivo assays using the Raji lymphoma cell line. ADCC was evaluated in real-time by IncuCyte assays and using a xenograft mouse model with high circulating levels of human IgG. Results Our data show that CD64/16A expressing iNK cells can mediate potent anti-tumor activity against human B cell lymphoma. In particular, (i) under suboptimal conditions, including low antibody concentrations and low effector-to-target ratios, iNK-CD64/16A cells mediate ADCC, (ii) iNK-CD64/16A cells can be pre-loaded with tumor-targeting antibodies (arming) to elicit ADCC, (iii) armed iNK-CD64/16A cells can be repurposed with additional antibodies to target new tumor antigens, and (iv) cryopreserved, armed iNK-CD64/16A are capable of sustained ADCC in a tumor xenograft model under saturating levels of human IgG. Discussion iNK-CD64/16A cells allow for a flexible use of antibodies (antibody arming and antibody targeting), and an "off-the-shelf" platform for multi-antigen recognition to overcome limitations of adoptive cell therapies expressing fixed antigen receptors leading to cancer relapse due to antigen escape variants.
Collapse
Affiliation(s)
- Kate J. Dixon
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Kristin M. Snyder
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Melissa Khaw
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Robert Hullsiek
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Zachary B. Davis
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Anders W. Matson
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | | | | | | | | | - Jeffrey S. Miller
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | | | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
8
|
Duffy C, Hunger SP, Bhakta N, Denburg AE, Antillon F, Barr RD. Curing pediatric cancer: A global view. Examples from acute lymphoblastic leukemia. Cancer 2024; 130:2247-2252. [PMID: 38552145 DOI: 10.1002/cncr.35290] [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: 06/09/2024]
Abstract
There is a substantial difference in observed survival among children with acute lymphoblastic leukemia in high‐income countries versus those in low‐ and middle‐income countries. This gap can be reduced considerably by multilevel investing in health systems and services with innovative frameworks such as implementation science.
Collapse
Affiliation(s)
- Caitlyn Duffy
- St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stephen P Hunger
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nickhill Bhakta
- St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Federico Antillon
- Unidad Nacional de Oncología Pediátrica and Francisco Marroquín University, Guatemala City, Guatemala
| | - Ronald D Barr
- McMaster Children's Hospital and McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
9
|
Hadiloo K, Taremi S, Safa SH, Amidifar S, Esmaeilzadeh A. The new era of immunological treatment, last updated and future consideration of CAR T cell-based drugs. Pharmacol Res 2024; 203:107158. [PMID: 38599467 DOI: 10.1016/j.phrs.2024.107158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/12/2024]
Abstract
Cancer treatment is one of the fundamental challenges in clinical setting, especially in relapsed/refractory malignancies. The novel immunotherapy-based treatments bring new hope in cancer therapy and achieve various treatment successes. One of the distinguished ways of cancer immunotherapy is adoptive cell therapy, which utilizes genetically modified immune cells against cancer cells. Between different methods in ACT, the chimeric antigen receptor T cells have more investigation and introduced a promising way to treat cancer patients. This technology progressed until it introduced six US Food and Drug Administration-approved CAR T cell-based drugs. These drugs act against hematological malignancies appropriately and achieve exciting results, so they have been utilized widely in cell therapy clinics. In this review, we introduce all CAR T cells-approved drugs based on their last data and investigate them from all aspects of pharmacology, side effects, and compressional. Also, the efficacy of drugs, pre- and post-treatment steps, and expected side effects are introduced, and the challenges and new solutions in CAR T cell therapy are in the last speech.
Collapse
Affiliation(s)
- Kaveh Hadiloo
- Department of immunology, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran
| | - Siavash Taremi
- Department of immunology, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran
| | - Salar Hozhabri Safa
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran
| | - Sima Amidifar
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, the Islamic Republic of Iran.
| |
Collapse
|
10
|
Passamonti F, Corrao G, Castellani G, Mora B, Maggioni G, Della Porta MG, Gale RP. Using real-world evidence in haematology. Best Pract Res Clin Haematol 2024; 37:101536. [PMID: 38490764 DOI: 10.1016/j.beha.2024.101536] [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: 07/06/2023] [Revised: 12/26/2023] [Accepted: 01/15/2024] [Indexed: 03/17/2024]
Abstract
Most new drug approvals are based on data from large randomized clinical trials (RCTs). However, there are sometimes contradictory conclusions from seemingly similar trials and generalizability of conclusions from these trials is limited. These considerations explain, in part, the gap between conclusions from data of RCTs and those from registries termed real world data (RWD). Recently, real-world evidence (RWE) from RWD processed by artificial intelligence has received increasing attention. We describe the potential of using RWD in haematology concluding RWE from RWD may complement data from RCTs to support regulatory decisions.
Collapse
Affiliation(s)
- Francesco Passamonti
- Università Degli Stu di di Milano, Milan, Italy; Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Giovanni Corrao
- Department of Statistics and Quantitative Methods, Laboratory of Healthcare Research & Pharmacoepidemiology, University of Milano-Bicocca, Milan, Italy
| | - Gastone Castellani
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Barbara Mora
- Hematology, ASST Sette Laghi, Ospedale di Circolo, Varese, Italy
| | - Giulia Maggioni
- Center for Accelerating Leukemia/Lymphoma Research (CALR) - IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Matteo Giovanni Della Porta
- Center for Accelerating Leukemia/Lymphoma Research (CALR) - IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunolgy and Inflammation, Imperial College London, London, UK.
| |
Collapse
|
11
|
Scoleri‐Longo Y, Pechlivanoglou P, Gupta S. Cost and cost-effectiveness of immunotherapy in childhood ALL: A systematic review. EJHAEM 2024; 5:166-177. [PMID: 38406535 PMCID: PMC10887368 DOI: 10.1002/jha2.814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 09/28/2023] [Accepted: 10/16/2023] [Indexed: 02/27/2024]
Abstract
Survival rates for pediatric acute lymphoblastic leukemia (pALL) have improved dramatically; relapsed/refractory (r/r) acute lymphoblastic leukemia (ALL) remains challenging. Immunotherapies are rapidly evolving treatments for r/r ALL with limited cost-effectiveness data. This study identifies existing economic evaluations of immunotherapy in pALL and summarizes cost-effectiveness. Medline, Embase, and other databases were searched from inception to October 2022. Cost-effectiveness analyses evaluating immunotherapy in pALL were included. Costs reported in 2021 USD. Of 2960 studies, 11 met inclusion criteria. Tisagenlecleucel was compared to standard of care, clofarabine monotherapy, clofarabine combination therapy, or blinatumomab. No studies have evaluated blinatumomab or inotuzumab ozogamicin. Six studies found tisagenlecleucel to be cost-effective, five of which were supported by Novartis. Four found that it had the potential to be cost-effective, and one found that it was not cost-effective. The cost-effectiveness of tisagenlecleucel was highly dependent on list price and cure rates. This study can inform the use of tisagenlecleucel in pALL.
Collapse
Affiliation(s)
- Yolanda Scoleri‐Longo
- Department of PaediatricsPost Graduate Medical EducationThe Hospital for Sick ChildrenTorontoOntarioCanada
| | | | - Sumit Gupta
- Cancer Research ProgramInstitute for Clinical Evaluative SciencesTorontoOntarioCanada
- Division of Haematology/OncologyThe Hospital for Sick ChildrenTorontoOntarioCanada
- Institute for Health PolicyEvaluation and Management, University of TorontoTorontoOntarioCanada
- Faculty of MedicineUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
12
|
Choe JH, Yu T, Abramson JS, Abou-el-Enein M. Cost-effectiveness of second-line lisocabtagene maraleucel in relapsed or refractory diffuse large B-cell lymphoma. Blood Adv 2024; 8:484-496. [PMID: 38153350 PMCID: PMC10837180 DOI: 10.1182/bloodadvances.2023011793] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023] Open
Abstract
ABSTRACT Lisocabtagene maraleucel (liso-cel), a chimeric antigen receptor (CAR) T-cell therapy, received the US Food and Drug Administration approval in 2022 for second-line treatment of diffuse large B-cell lymphoma (DLBCL) for patients with refractory disease or early relapse after first-line chemoimmunotherapy. This decision was based on the TRANSFORM study demonstrating improvements in event-free survival with liso-cel compared with standard care. Given the high costs of CAR T-cell therapies, particularly as they transition to second-line treatment, a cost-effectiveness analysis is essential to determine their economic viability. The study used a partitioned survival model with standard parametric functions to evaluate the cost-effectiveness of liso-cel aganist platinum-based chemotherapy followed by high-dose chemotherapy and autologous hematopoietic stem cell transplantation over a lifetime horizon The analysis relied on data from the TRANSFORM and TRANSCEND trials, established literature, and public data sets to calculate the incremental cost-effectiveness ratio (ICER). For a representative cohort of US adults aged 60 years, ICER of liso-cel was $99 669 per quality-adjusted life-year (QALY) from a health care sector perspective and $68 212 per QALY from a societal perspective, confirming its cost-effectiveness at the $100 000 per QALY threshold. Nonetheless, under certain scenarios, liso-cel surpasses this benchmark but remains within the US acceptable range of $150 000 per QALY. A key finding underlines the importance of incorporating productivity losses into such analyses to capture the broader societal values of novel therapies. Although these therapies offer substantial clinical benefits, their high acquisition costs and limited long-term data critically challenge their economic sustainability.
Collapse
Affiliation(s)
- Jee H. Choe
- Department of Pharmaceutical and Health Economics, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA
- Leonard D. Schaeffer Center for Health Policy & Economics, University of Southern California, Los Angeles, CA
| | - Tianzhou Yu
- Department of Pharmaceutical and Health Economics, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA
- Leonard D. Schaeffer Center for Health Policy & Economics, University of Southern California, Los Angeles, CA
| | - Jeremy S. Abramson
- Center for Lymphoma, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Mohamed Abou-el-Enein
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California and Children’s Hospital of Los Angeles, Los Angeles, CA
- USC/CHLA Cell Therapy Program, University of Southern California and Children’s Hospital of Los Angeles, Los Angeles, CA
| |
Collapse
|
13
|
Bustamante-Ogando JC, Hernández-López A, Galván-Díaz C, Rivera-Luna R, Fuentes-Bustos HE, Meneses-Acosta A, Olaya-Vargas A. Childhood leukemias in Mexico: towards implementing CAR-T cell therapy programs. Front Oncol 2024; 13:1304805. [PMID: 38304036 PMCID: PMC10833104 DOI: 10.3389/fonc.2023.1304805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/15/2023] [Indexed: 02/03/2024] Open
Abstract
Leukemias are the most common type of pediatric cancer around the world. Prognosis has improved during the last decades, and many patients are cured with conventional treatment as chemotherapy; however, many patients still present with a refractory disease requiring additional treatments, including hematopoietic stem cell transplantation. Immunotherapy with monoclonal antibodies or cellular therapy is a promising strategy for treating refractory or relapsed hematological malignancies. Particularly, CAR-T cells have shown clinical efficacy in clinical trials, and different products are now commercially approved by regulatory agencies in the USA and Europe. Many challenges still need to be solved to improve and optimize the potential of these therapies worldwide. Global access to cell therapy is a significant concern, and different strategies are being explored in the middle- and low-income countries. In Mexico, leukemias represent around 50% of total cancer diagnosed in pediatric patients, and the rate of relapsed or refractory disease is higher than reported in other countries, a multi-factorial problem. Although significant progress has been made during the last decades in leukemia diagnosis and treatment, making new therapies available to Mexican patients is a priority, and cell and gene therapies are on the horizon. Efforts are ongoing to make CAR-T cell therapy accessible for patients in Mexico. This article summarizes a general landscape of childhood leukemias in Mexico, and we give a perspective about the current strategies, advances, and challenges ahead to make gene and cell therapies for leukemia clinically available.
Collapse
Affiliation(s)
- Juan Carlos Bustamante-Ogando
- Immunodeficiencies Research Laboratory and Clinical Immunology Department, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Alejandrina Hernández-López
- Laboratorio 7 Biotecnología Farmacéutica, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca, Morelos, Mexico
- Consejo Nacional de Humanidades Ciencias y Tecnologías, CONAHCYT, Mexico City, Mexico
| | - César Galván-Díaz
- Oncology Department, Instituto Nacional de Pediatría, Mexico City, Mexico
| | | | - Hugo E. Fuentes-Bustos
- Laboratorio 7 Biotecnología Farmacéutica, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca, Morelos, Mexico
| | - Angélica Meneses-Acosta
- Laboratorio 7 Biotecnología Farmacéutica, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca, Morelos, Mexico
| | - Alberto Olaya-Vargas
- Hematopoietic Stem Cell Transplantation and Cell Therapy Program, Instituto Nacional de Pediatría, Mexico City, Mexico
| |
Collapse
|
14
|
Yamamoto C, Minakata D, Yokoyama D, Furuki S, Noguchi A, Koyama S, Oyama T, Murahashi R, Nakashima H, Ikeda T, Kawaguchi SI, Hyodo K, Toda Y, Ito S, Nagayama T, Umino K, Morita K, Ashizawa M, Ueda M, Hatano K, Sato K, Ohmine K, Fujiwara SI, Kanda Y. Cost-Effectiveness of Anti-BCMA Chimeric Antigen Receptor T Cell Therapy in Relapsed/Refractory Multiple Myeloma. Transplant Cell Ther 2024; 30:118.e1-118.e15. [PMID: 37802181 DOI: 10.1016/j.jtct.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 09/06/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
Despite its promising outcomes, anti-BCMA chimeric antigen receptor T cell therapy (CAR-T) is the most expensive myeloma treatment developed to date, and its cost-effectiveness is an important issue. This study aimed to assess the cost-effectiveness of anti-BCMA CAR-T compared to standard antimyeloma therapy in patients with relapsed/refractory multiple myeloma. The model included myeloma patients in Japan and the United States who have received ≥3 prior lines of antimyeloma therapy, including immunomodulatory drugs, proteasome inhibitors, and anti-CD38 monoclonal antibodies. A Markov model was constructed to compare the CAR-T strategy, in which patients receive either idecabtagene vicleucel (ide-cel) or ciltacabtagene autoleucel (cilta-cel) followed by 3 lines of multiagent chemotherapy after relapse, and the no CAR-T strategy, in which patients receive only chemotherapy. Data from the LocoMMotion, KarMMa, and CARTITUDE-1 trials were extracted. Several assumptions were made regarding long-term progression-free survival (PFS) with CAR-T. Extensive scenario analyses were made regarding regimens for no CAR-T strategies. The outcome was an incremental cost-effectiveness ratio (ICER) with willingness-to-pay thresholds of ¥7,500,000 in Japan and $150,000 in the United States. When a 5-year PFS of 40% with cilta-cel was assumed, the ICER of the CAR-T strategy versus the no CAR-T strategy was ¥7,603,823 per QALY in Japan and $112,191 per QALY in the United States over a 10-year time horizon. When a 5-year PFS of 15% with ide-cel was assumed, the ICER was ¥20,388,711 per QALY in Japan and $261,678 per QALY in the United States over a 10-year time horizon. The results were highly dependent on the PFS assumption with CAR-T and were robust to changes in most other parameters and scenarios. Although anti-BCMA CAR-T can be cost-effective even under current pricing, a high long-term PFS is necessary.
Collapse
Affiliation(s)
- Chihiro Yamamoto
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Daisuke Minakata
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Daizo Yokoyama
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shuka Furuki
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Atsuto Noguchi
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shunsuke Koyama
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takashi Oyama
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Rui Murahashi
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Hirotomo Nakashima
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takashi Ikeda
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shin-Ichiro Kawaguchi
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kazuki Hyodo
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yumiko Toda
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shoko Ito
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takashi Nagayama
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kento Umino
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kaoru Morita
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Masahiro Ashizawa
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Masuzu Ueda
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kaoru Hatano
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kazuya Sato
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Ken Ohmine
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shin-Ichiro Fujiwara
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan; Division of Cell Transplantation and Transfusion, Jichi Medical University, Tochigi, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
15
|
Maziarz RT, Devine S, Garrison LP, Agodoa I, Badaracco J, Gitlin M, Perales MA. Estimating the Lifetime Medical Cost Burden of an Allogeneic Hematopoietic Cell Transplantation Patient. Transplant Cell Ther 2023; 29:637.e1-637.e9. [PMID: 37364775 PMCID: PMC11035010 DOI: 10.1016/j.jtct.2023.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/02/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) has the potential for curative outcomes for a variety of hematologic malignancies. Current allo-HCT studies often describe the outcomes and costs in the near term; however, research on the lifetime economic burden post-allo-HCT remains limited. This study was conducted to estimate the average total lifetime direct medical costs of an allo-HCT patient and the potential net monetary savings from an alternative treatment associated with improved graft-versus-host disease (GVHD)-free, relapse-free survival (GRFS). A disease-state model was constructed using a short-term decision tree and a long-term semi-Markov partitioned survival model to estimate the average per-patient lifetime cost and expected quality-adjusted life years (QALYs) for an allo-HCT patient from a US healthcare system perspective. Key clinical inputs included overall survival, GRFS, incidence of both acute and chronic GVHD, relapse of the primary disease, and infections. Cost results were reported as ranges based on varying the percentage of chronic GVHD patients that remained on treatment after 2 years (15% or 39%). Over a lifetime, the average per-patient medical cost of allo-HCT was estimated to range from $942,373 to $1,247,917. The majority of the costs were for chronic GVHD treatment (37% to 53%), followed by the allo-HCT procedure (15% to 19%). The expected lifetime QALYs of an allo-HCT patient were estimated as 4.7. Lifetime per-patient treatment costs often exceed $1,000,000 for allo-HCT patients. Innovative research efforts focused on the reduction or elimination of late complications, particularly chronic GVHD, may provide the greatest value to improved patient outcomes.
Collapse
Affiliation(s)
- Richard T Maziarz
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.
| | - Steven Devine
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
| | - Louis P Garrison
- The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, University of Washington, Seattle, Washington
| | | | | | | | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
16
|
Chulián S, Stolz BJ, Martínez-Rubio Á, Blázquez Goñi C, Rodríguez Gutiérrez JF, Caballero Velázquez T, Molinos Quintana Á, Ramírez Orellana M, Castillo Robleda A, Fuster Soler JL, Minguela Puras A, Martínez Sánchez MV, Rosa M, Pérez-García VM, Byrne HM. The shape of cancer relapse: Topological data analysis predicts recurrence in paediatric acute lymphoblastic leukaemia. PLoS Comput Biol 2023; 19:e1011329. [PMID: 37578973 PMCID: PMC10468039 DOI: 10.1371/journal.pcbi.1011329] [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: 09/29/2022] [Revised: 08/30/2023] [Accepted: 07/05/2023] [Indexed: 08/16/2023] Open
Abstract
Although children and adolescents with acute lymphoblastic leukaemia (ALL) have high survival rates, approximately 15-20% of patients relapse. Risk of relapse is routinely estimated at diagnosis by biological factors, including flow cytometry data. This high-dimensional data is typically manually assessed by projecting it onto a subset of biomarkers. Cell density and "empty spaces" in 2D projections of the data, i.e. regions devoid of cells, are then used for qualitative assessment. Here, we use topological data analysis (TDA), which quantifies shapes, including empty spaces, in data, to analyse pre-treatment ALL datasets with known patient outcomes. We combine these fully unsupervised analyses with Machine Learning (ML) to identify significant shape characteristics and demonstrate that they accurately predict risk of relapse, particularly for patients previously classified as 'low risk'. We independently confirm the predictive power of CD10, CD20, CD38, and CD45 as biomarkers for ALL diagnosis. Based on our analyses, we propose three increasingly detailed prognostic pipelines for analysing flow cytometry data from ALL patients depending on technical and technological availability: 1. Visual inspection of specific biological features in biparametric projections of the data; 2. Computation of quantitative topological descriptors of such projections; 3. A combined analysis, using TDA and ML, in the four-parameter space defined by CD10, CD20, CD38 and CD45. Our analyses readily extend to other haematological malignancies.
Collapse
Affiliation(s)
- Salvador Chulián
- Department of Mathematics, Universidad de Cádiz, Puerto Real (Cádiz), Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Cádiz, Spain
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Bernadette J. Stolz
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
- Laboratory for Topology and Neuroscience, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Álvaro Martínez-Rubio
- Department of Mathematics, Universidad de Cádiz, Puerto Real (Cádiz), Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Cádiz, Spain
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Cristina Blázquez Goñi
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Cádiz, Spain
- Department of Paediatric Haematology and Oncology, Hospital Universitario de Jerez, Jerez de la Frontera (Cádiz), Spain
- Department of Haematology, Hospital Universitario Vírgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
| | - Juan F. Rodríguez Gutiérrez
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Cádiz, Spain
- Department of Paediatric Haematology and Oncology, Hospital Universitario de Jerez, Jerez de la Frontera (Cádiz), Spain
| | - Teresa Caballero Velázquez
- Department of Haematology, Hospital Universitario Vírgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
- CSIC, University of Sevilla, Sevilla, Spain
| | - Águeda Molinos Quintana
- Department of Haematology, Hospital Universitario Vírgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
- CSIC, University of Sevilla, Sevilla, Spain
| | - Manuel Ramírez Orellana
- Department of Paediatric Haematology and Oncology, Hospital Infantil Universitario Niño Jesús - Instituto Investigación Sanitaria La Princesa, Madrid, Spain
| | - Ana Castillo Robleda
- Department of Paediatric Haematology and Oncology, Hospital Infantil Universitario Niño Jesús - Instituto Investigación Sanitaria La Princesa, Madrid, Spain
| | - José Luis Fuster Soler
- Department of Paediatric Haematology and Oncology, Hospital Clínico Universitario Virgen de la Arrixaca - Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Alfredo Minguela Puras
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca - Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - María V. Martínez Sánchez
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca - Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - María Rosa
- Department of Mathematics, Universidad de Cádiz, Puerto Real (Cádiz), Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Cádiz, Spain
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Víctor M. Pérez-García
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
- Instituto de Matemática Aplicada a la Ciencia y la Ingeniería (IMACI), Universidad de Castilla-La Mancha, Ciudad Real, Spain
- ETSI Industriales, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Helen M. Byrne
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Andrade AM, Teixeira VR, Pogue R, Figueiredo ACMG, Carvalho JL. A systematic review on the cost-effectiveness assessment of tisagenlecleucel for refractory or relapsing B-cell acute lymphoblastic leukemia (R/R B-ALL) treatment in children and young adults. Cytotherapy 2023:S1465-3249(23)00957-X. [PMID: 37341664 DOI: 10.1016/j.jcyt.2023.05.011] [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: 09/09/2022] [Revised: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND AIMS The advanced therapy product tisagenlecleucel is a CD19-directed genetically modified autologous T-cell immunotherapy that has brought hope for children and young adults with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). We sought to evaluate the cost-effectiveness of tisagenlecleucel compared with conventional salvage therapies in pediatric and young adult patients with R/R B-ALL. METHODS This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses parameters as registered in International Prospective Register of Systematic Reviews (CRD42021266998). Literature was searched using the MEDLINE databases via PubMed, EMBASE, Lilacs, the Cochrane Central Register of Controlled Trials and Web of Science in January 2022. Titles were screened independently by two reviewers. Articles deemed to meet the inclusion criteria were screened independently on abstract, and full texts were reviewed. RESULTS In total, 5627 publications were identified, from which six eligible studies were selected. The conventional therapies identified were blinatumomab (Blina), clofarabine monotherapy (Clo-M), clofarabine combined with cyclophosphamide and etoposide (Clo-C) and the combination of fludarabine, cytarabine and idarubicin (FLA-IDA). The discounted incremental cost-effectiveness ratio (ICER) per quality-adjusted life year (QALY) gained for tisagenlecleucel compared with Clo-C and Blina averages was $38 837 and $25 569, respectively. In relation to the cost of the drug, the average of tisagenlecleucel was approximately 4.3 times, 10.8 times or 4.7 times greater than the Clo-M, Clo-C and Blina, respectively. CONCLUSIONS This systematic review highlighted that tisagenlecleucel is a much more expensive therapy than conventional alternatives. However, tisagenlecleucel performed well on the ICER, not exceeding $100 000/QALY. It was also found that the advanced therapy product was more effective than the conventional small molecule and biological drugs, in terms of life years and QALY gained.
Collapse
Affiliation(s)
- Aurélio Matos Andrade
- Medical Sciences Program, University of Brasilia, Brasilia, Distrito Federal, Brazil; Program of Evidence for Health Policy and Technologies, Oswaldo Cruz Brasilia Foundation, Brasilia, Distrito Federal, Brazil; Interdisciplinary Biosciences Laboratory, Faculty of Medicine, University of Brasília, Brasília, Distrito Federal, Brazil
| | | | - Robert Pogue
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, Brasília, Brazil
| | | | - Juliana Lott Carvalho
- Medical Sciences Program, University of Brasilia, Brasilia, Distrito Federal, Brazil; Interdisciplinary Biosciences Laboratory, Faculty of Medicine, University of Brasília, Brasília, Distrito Federal, Brazil; Faculty of Medicine, University of Brasilia, Brasilia, Distrito Federal, Brazil.
| |
Collapse
|
18
|
Gavan SP, Wright SJ, Thistlethwaite F, Payne K. Capturing the Impact of Constraints on the Cost-Effectiveness of Cell and Gene Therapies: A Systematic Review. PHARMACOECONOMICS 2023; 41:675-692. [PMID: 36905571 DOI: 10.1007/s40273-022-01234-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 05/06/2023]
Abstract
OBJECTIVE Decision-makers need to resolve constraints on delivering cell and gene therapies to patients as these treatments move into routine care. This study aimed to investigate if, and how, constraints that affect the expected cost and health consequences of cell and gene therapies have been included in published examples of cost-effectiveness analyses (CEAs). METHOD A systematic review identified CEAs of cell and gene therapies. Studies were identified from previous systematic reviews and by searching Medline and Embase until 21 January 2022. Constraints described qualitatively were categorised by theme and summarised by a narrative synthesis. Constraints evaluated in quantitative scenario analyses were appraised by whether they changed the decision to recommend treatment. RESULTS Thirty-two CEAs of cell (n = 20) and gene therapies (n = 12) were included. Twenty-one studies described constraints qualitatively (70% cell therapy CEAs; 58% gene therapy CEAs). Qualitative constraints were categorised by four themes: single payment models; long-term affordability; delivery by providers; manufacturing capability. Thirteen studies assessed constraints quantitatively (60% cell therapy CEAs; 8% gene therapy CEAs). Two types of constraint were assessed quantitatively across four jurisdictions (USA, Canada, Singapore, The Netherlands): alternatives to single payment models (n = 9 scenario analyses); improving manufacturing (n = 12 scenario analyses). The impact on decision-making was determined by whether the estimated incremental cost-effectiveness ratios crossed a relevant cost-effectiveness threshold for each jurisdiction (outcome-based payment models: n = 25 threshold comparisons made, 28% decisions changed; improving manufacturing: n = 24 threshold comparisons made, 4% decisions changed). CONCLUSION The net health impact of constraints is vital evidence to help decision-makers scale up the delivery of cell and gene therapies as patient volume increases and more advanced therapy medicinal products are launched. CEAs will be essential to quantify how constraints affect the cost-effectiveness of care, prioritise constraints to be resolved, and establish the value of strategies to implement cell and gene therapies by accounting for their health opportunity cost.
Collapse
Affiliation(s)
- Sean P Gavan
- Manchester Centre for Health Economics, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
| | - Stuart J Wright
- Manchester Centre for Health Economics, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Fiona Thistlethwaite
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Katherine Payne
- Manchester Centre for Health Economics, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| |
Collapse
|
19
|
Wu W, Ding S, Mingming Z, Yuping Z, Sun X, Zhao Z, Yang Y, Hu Y, Dong H. Cost effectiveness analysis of CAR-T cell therapy for patients with relapsed/refractory multiple myeloma in China. J Med Econ 2023; 26:701-709. [PMID: 37145966 DOI: 10.1080/13696998.2023.2207742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
BACKGROUND The landscape of treatment strategies for relapsed/refractory multiple myeloma (RRMM) has dramatically changed due to the emergence of chimeric antigen receptor T (CAR-T) cell therapy. The aim of this study was to evaluate the cost-effectiveness of two CAR-T cell treatments for RRMM patients from the perspective of the Chinese healthcare system. METHODS Markov model was used to compare currently available salvage chemotherapy with Idecabtagene vicleucel (Ide-cel) and Ciltacabtagene autoleucel (Cilta-cel) for treatment of patients with RRMM. The model was developed based on data from three studies: CARTITUDE-1, KarMMa and MAMMOTH. The healthcare cost and utility of RRMM patients were collected from a provincial clinical center in China. RESULTS In the base case analysis, 3.4% and 30.6% of RRMM patients were expected to be long-term survivors after 5 years of Ide-cel and Cilta-cel treatment, respectively. Compared to salvage chemotherapy, Ide-cel and Cilta-cel were associated with incremental QALYs of 1.19 and 3.31, and incremental costs of US $140,693 and $119,806, leading to ICERs of $118,229 and $36,195 per QALY, respectively. At an ICER threshold of $37,653/QALY gained, the probability that Ide-cel and Cilta-cel are cost-effective were estimated to be 0% and 72%, respectively. With younger target people entering the model, and partitioned survival model in scenario analysis, the ICERs of Cilta-cel and Ide-cel changed rather mildly and their cost-effectiveness results were same to base analysis. CONCLUSIONS Base on the willingness-to-pay of 3 times China's per capita GDP in 2021, Cilta-cel was considered to be a more cost-effective option compared to salvage chemotherapy for RRMM in China, while Ide-cel was not.
Collapse
Affiliation(s)
- Weijia Wu
- Center for Health Policy Studies, School of Public Health, and Department of Science and Education of the Fourth Affiliated Hospital, Zhejiang University School of Medicine
| | - Shuyi Ding
- Nursing Department, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhang Mingming
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Zhou Yuping
- Nursing Department, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xueshan Sun
- Center for Health Policy Studies, School of Public Health, and Department of Science and Education of the Fourth Affiliated Hospital, Zhejiang University School of Medicine
| | - Zixuan Zhao
- Center for Health Policy Studies, School of Public Health, and Department of Science and Education of the Fourth Affiliated Hospital, Zhejiang University School of Medicine
| | - Yi Yang
- Center for Health Policy Studies, School of Public Health, and Department of Science and Education of the Fourth Affiliated Hospital, Zhejiang University School of Medicine
| | - Yongxian Hu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Hengjin Dong
- Center for Health Policy Studies, School of Public Health, and Department of Science and Education of the Fourth Affiliated Hospital, Zhejiang University School of Medicine
| |
Collapse
|
20
|
Al-Akashi Z, Zujur D, Kamiya D, Kato T, Kondo T, Ikeya M. Selective vulnerability of human-induced pluripotent stem cells to dihydroorotate dehydrogenase inhibition during mesenchymal stem/stromal cell purification. Front Cell Dev Biol 2023; 11:1089945. [PMID: 36814599 PMCID: PMC9939518 DOI: 10.3389/fcell.2023.1089945] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/24/2023] [Indexed: 02/08/2023] Open
Abstract
The use of induced mesenchymal stem/stromal cells (iMSCs) derived from human induced pluripotent stem cells (hiPSCs) in regenerative medicine involves the risk of teratoma formation due to hiPSCs contamination in iMSCs. Therefore, eradicating the remaining undifferentiated hiPSCs is crucial for the effectiveness of the strategy. The present study demonstrates the Brequinar (BRQ)-induced inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme in de novo pyrimidine biosynthesis, selectively induces apoptosis, cell cycle arrest, and differentiation; furthermore, it promotes transcriptional changes and prevents the growth of 3-dimensional hiPSC aggregates. Contrastingly, BRQ-treated iMSCs showed no changes in survival, differentiation potential, or gene expression. The results suggest that BRQ is a potential agent for the effective purification of iMSCs from a mixed population of iMSCs and hiPSCs, which is a crucial step in successful iMSC-based therapy.
Collapse
Affiliation(s)
- Ziadoon Al-Akashi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Denise Zujur
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Daisuke Kamiya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan,Takeda-CiRA Joint Program, Fujisawa, Kanagawa, Japan
| | - Tomohisa Kato
- Medical Research Institute, Kanazawa Medical University, Kanazawa, Japan
| | - Toru Kondo
- Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Makoto Ikeya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan,Takeda-CiRA Joint Program, Fujisawa, Kanagawa, Japan,*Correspondence: Makoto Ikeya,
| |
Collapse
|
21
|
Gye A, Goodall S, De Abreu Lourenco R. Cost-effectiveness Analysis of Tisagenlecleucel Versus Blinatumomab in Children and Young Adults with Acute Lymphoblastic Leukemia: Partitioned Survival Model to Assess the Impact of an Outcome-Based Payment Arrangement. PHARMACOECONOMICS 2023; 41:175-186. [PMID: 36266557 PMCID: PMC9883311 DOI: 10.1007/s40273-022-01188-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/04/2022] [Indexed: 05/30/2023]
Abstract
OBJECTIVE This research assesses the impact of an outcome-based payment arrangement (OBA) linking complete remission (CR) to survival as a means of maintaining cost-effectiveness for a chimeric antigen receptor T cell (CAR-T) therapy in young patients with acute lymphoblastic leukemia (ALL). METHODS A partitioned survival model (PSM) was used to model the cost-effectiveness of tisagenlecleucel versus blinatumomab in ALL from the Australian healthcare system perspective. A decision tree modeled different OBAs by funneling patients into a series of PSMs based on response. Outcomes were informed by individual patient data, while costs followed Australian treatment practices. Costs and quality-adjusted life years (QALYs) were combined to calculate a single incremental cost-effectiveness ratio (ICER), reported in US dollars (2022) at a discount rate of 5% on costs and outcomes. RESULTS For the base case, incremental costs and benefit were $379,595 and 4.27 QALYs, giving an ICER of $88,979. The ICER was most sensitive to discount rate ($57,660-$75,081), "cure point" ($62,718-$116,206) and extrapolation method ($76,018-$94,049). OBAs had a modest effect on the ICER when response rates varied. A responder-only payment was the most effective arrangement for maintaining the ICER ($88,249-$89,434), although this option was associated with the greatest financial uncertainty. A split payment arrangement (payment on infusion followed by payment on response) reduced variability in the ICER ($82,650-$99,154) compared with a single, upfront payment ($77,599-$107,273). CONCLUSION OBAs had a modest impact on reducing cost-effectiveness uncertainty. The value of OBAs should be weighed against the additional resources needed to administer such arrangements, and importantly overall cost to government.
Collapse
Affiliation(s)
- Amy Gye
- Novartis Pharmaceuticals Australia, University of Technology Sydney, Ultimo, NSW, Australia.
| | - Stephen Goodall
- Centre for Health Economics Research and Evaluation (CHERE), University of Technology Sydney, Level 12, Building 10, 235 Jones Street, Ultimo, NSW, 2007, Australia
| | - Richard De Abreu Lourenco
- Centre for Health Economics Research and Evaluation (CHERE), University of Technology Sydney, Level 12, Building 10, 235 Jones Street, Ultimo, NSW, 2007, Australia
| |
Collapse
|
22
|
Frigault M, Rotte A, Ansari A, Gliner B, Heery C, Shah B. Dose fractionation of CAR-T cells. A systematic review of clinical outcomes. J Exp Clin Cancer Res 2023; 42:11. [PMID: 36627710 PMCID: PMC9830795 DOI: 10.1186/s13046-022-02540-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/18/2022] [Indexed: 01/12/2023] Open
Abstract
CAR-T cells are widely recognized for their potential to successfully treat hematologic cancers and provide durable response. However, severe adverse events such as cytokine release syndrome (CRS) and neurotoxicity are concerning. Our goal is to assess CAR-T cell clinical trial publications to address the question of whether administration of CAR-T cells as dose fractions reduces toxicity without adversely affecting efficacy. Systematic literature review of studies published between January 2010 and May 2022 was performed on PubMed and Embase to search clinical studies that evaluated CAR-T cells for hematologic cancers. Studies published in English were considered. Studies in children (age < 18), solid tumors, bispecific CAR-T cells, and CAR-T cell cocktails were excluded. Data was extracted from the studies that met inclusion and exclusion criteria. Review identified a total of 18 studies that used dose fractionation. Six studies used 2-day dosing schemes and 12 studies used 3-day schemes to administer CAR-T cells. Three studies had both single dose and fractionated dose cohorts. Lower incidence of Grade ≥ 3 CRS and neurotoxicity was seen in fractionated dose cohorts in 2 studies, whereas 1 study reported no difference between single and fractionated dose cohorts. Dose fractionation was mainly recommended for high tumor burden patients. Efficacy of CAR-T cells in fractionated dose was comparable to single dose regimen within the same or historical trial of the same agent in all the studies. The findings suggest that administering dose fractions of CAR-T cells over 2-3 days instead of single dose infusion may mitigate the toxicity of CAR-T cell therapy including CRS and neurotoxicity, especially in patients with high tumor burden. However, controlled studies are likely needed to confirm the benefits of dose fractionation.
Collapse
Affiliation(s)
- Matthew Frigault
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Olry de Labry-Lima A, Ponce-Polo A, García-Mochón L, Ortega-Ortega M, Pérez-Troncoso D, Epstein D. Challenges for Economic Evaluations of Advanced Therapy Medicinal Products: A Systematic Review. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:138-150. [PMID: 36031480 DOI: 10.1016/j.jval.2022.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/20/2022] [Accepted: 07/07/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Advanced therapy medicinal products (ATMPs) are drugs for human use for the treatment of chronic, degenerative, or life-threatening diseases that are based on genes, tissues, or cells. This article aimed to identify and critically review published economic analyses of ATMPs. METHODS A systematic review of economic analyses of ATMPs was undertaken. Study characteristics, design, sources of data, resources and unit costs, modeling and extrapolation methods, study results, and sensitivity analyses were assessed. RESULTS A total of 46 economic analyses of ATMP (from 45 articles) were included; 4 were cell therapy medicinal products, 33 gene therapy medicinal products, and 9 tissue-engineered products. 30 therapies had commercial marketing approval; 39 studies were cost-utility analysis, 5 were cost-effectiveness analysis, and 2 were cost only studies. Four studies predicted that the ATMP offered a step change in the management of the condition and 10 studies estimated that the ATMP would offer a lower mean cost. CONCLUSIONS Comparison with historical controls, pooling of data, and use of techniques such as mixture cure fraction models should be used cautiously. Sensitivity analyses should be used across a plausible range of prices. Clinical studies need to be designed to align with health technology assessment requirements, including generic quality of life, and payers should aim for clarity of criteria. Regulators and national payers should aim for compatibility of registers to allow interchange of data. Given the increasing reliance on industry-funded economic analyses, careful critical review is recommended.
Collapse
Affiliation(s)
- Antonio Olry de Labry-Lima
- Escuela Andaluza de Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria Ibs, Granada, Spain; CIBER en Epidemiología and Salud Pública (CIBERESP), Spain
| | - Angela Ponce-Polo
- Andalusian Network for the Design & Translation of Advanced Therapies, Sevilla, Spain.
| | | | | | | | | |
Collapse
|
24
|
Choe JH, Abdel-Azim H, Padula WV, Abou-el-Enein M. Cost-effectiveness of Axicabtagene Ciloleucel and Tisagenlecleucel as Second-line or Later Therapy in Relapsed or Refractory Diffuse Large B-Cell Lymphoma. JAMA Netw Open 2022; 5:e2245956. [PMID: 36520440 PMCID: PMC9856352 DOI: 10.1001/jamanetworkopen.2022.45956] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
IMPORTANCE Chimeric antigen receptor (CAR) T cell therapies are approved as a third-line or later therapy for several hematological malignant neoplasms. Recently, randomized clinical trials have investigated their efficacy as a second-line treatment in high-risk relapsed or refractory diffuse large B-cell lymphoma (DLBCL) compared with salvage chemotherapy followed by hematopoietic stem cell transplantation (HSCT). OBJECTIVE To evaluate the cost-effectiveness of axicabtagene ciloleucel and tisagenlecleucel vs standard care (SC) as second-line or later therapy for relapsed or refractory DLBCL, from both US health care sector and societal perspectives at a cost-effectiveness threshold of $150 000 per quality-adjusted life-year (QALY). DESIGN, SETTING, AND PARTICIPANTS This economic evaluation assessed cost-effectiveness using a partitioned survival model with 2021 US dollars and QALYs over a lifetime horizon. Model inputs were derived from 2 randomized clinical trials (ZUMA-7 and BELINDA) and published literature. In the trials, patients who did not respond to SC received CAR T cells (treatment switching or crossover), either outside the protocol (ZUMA-7) or as part of the protocol (BELINDA). A separate scenario analysis compared second-line axicabtagene ciloleucel with SC alone without treatment crossover to CAR T cell therapy. Data analysis was performed from December 18, 2021, to September 13, 2022. EXPOSURES CAR T cell therapy (axicabtagene ciloleucel and tisagenlecleucel) compared with salvage chemotherapy followed by HSCT. MAIN OUTCOMES AND MEASURES Costs and QALYs were used to derive incremental cost-effectiveness ratios (ICERs) for the health care sector and societal perspectives. Cost and QALYs were discounted at 3.0% annually. Univariate and multivariate probabilistic sensitivity analysis using 10 000 Monte Carlo simulations were applied to test model uncertainty on the ICER. RESULTS Second-line axicabtagene ciloleucel was associated with an ICER of $99 101 per QALY from the health care sector perspective and an ICER of $97 977 per QALY from the societal perspective, while second-line tisagenlecleucel was dominated by SC (incremental costs of $37 803 from the health care sector and $39 480 from the societal perspective with decremental QALY of -0.02). Third-line or later tisagenlecleucel was associated with an ICER of $126 593 per QALY from the health care sector perspective and an ICER of $128 012 per QALY from the societal perspective. Based on the scenario analysis of no treatment switching, second-line axicabtagene ciloleucel yielded an ICER of $216 790 per QALY from the health care sector perspective and an ICER of $218 907 per QALY from the societal perspective, compared with SC. When accounting for patients achieving prolonged progression-free survival who would not incur progression-related costs, in this scenario ICER changed to $125 962 per QALY from the health care sector perspective and $122 931 per QALY from the societal perspective. These results were most sensitive to increased list prices of CAR T cell therapy and QALY losses associated with axicabtagene ciloleucel and tisagenlecleucel. CONCLUSIONS AND RELEVANCE These findings suggest that second-line axicabtagene ciloleucel and third-line or later tisagenlecleucel were cost-effective in treating patients with relapsed or refractory DLBCL at the cost-effectiveness threshold of $150 000 per QALY. However, uncertainty remains regarding the best candidates who would experience value gains from receiving CAR T cell therapy.
Collapse
Affiliation(s)
- Jee H. Choe
- Department of Pharmaceutical and Health Economics, School of Pharmacy, University of Southern California, Los Angeles
- Leonard D. Schaeffer Center for Health Policy & Economics, University of Southern California, Los Angeles
| | - Hisham Abdel-Azim
- Loma Linda University School of Medicine, Cancer Center, Children Hospital and Medical Center, Loma Linda, California
| | - William V. Padula
- Department of Pharmaceutical and Health Economics, School of Pharmacy, University of Southern California, Los Angeles
- Leonard D. Schaeffer Center for Health Policy & Economics, University of Southern California, Los Angeles
| | - Mohamed Abou-el-Enein
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles
- USC/CHLA Cell Therapy Program, University of Southern California and Children’s Hospital of Los Angeles, Los Angeles
| |
Collapse
|
25
|
Choi G, Shin G, Bae S. Price and Prejudice? The Value of Chimeric Antigen Receptor (CAR) T-Cell Therapy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12366. [PMID: 36231661 PMCID: PMC9566791 DOI: 10.3390/ijerph191912366] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 05/23/2023]
Abstract
Although chimeric antigen receptor (CAR) T-cell therapy has shown a high response rate in lymphoma patients, its cost-effectiveness is controversial due to the high price and uncertainty of the clinical evidence. In addition to the high acquisition cost of CAR T-cell therapy, procedure and facility cost increase the financial burden considering the frequency of adverse events such as cytokine release syndrome. In clinical research, relatively short follow-up periods were used compared to traditional cancer agents. In addition, head-to-head comparative effectiveness data are unavailable, which is an important factor when evaluating the cost-effectiveness of a new treatment. Additional evidence that will compensate for the uncertainty of existing clinical data is needed for full evaluation of long-term efficacy, safety, and comparative effectiveness.
Collapse
Affiliation(s)
| | | | - SeungJin Bae
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
26
|
Ragoonanan D, Sheikh IN, Gupta S, Khazal SJ, Tewari P, Petropoulos D, Li S, Mahadeo KM. The Evolution of Chimeric Antigen Receptor T-Cell Therapy in Children, Adolescents and Young Adults with Acute Lymphoblastic Leukemia. Biomedicines 2022; 10:biomedicines10092286. [PMID: 36140387 PMCID: PMC9496125 DOI: 10.3390/biomedicines10092286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 08/05/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor T-cell (CAR T) therapy is a revolutionary treatment for pediatric, adolescent and young adult patients (AYA) with relapsed/refractory B-cell acute lymphoblastic leukemia. While the landscape of immunotherapy continues to rapidly evolve, widespread use of CAR T therapy is limited and many questions remain regarding the durability of CAR T therapy, methods to avoid CAR T therapy resistance and the role of consolidative stem cell transplant. Modified strategies to develop effective and persistent CAR T cells at lower costs and decreased toxicities are warranted. In this review we present current indications, limitations and future directions of CAR T therapy for ALL in the pediatric and AYA population.
Collapse
Affiliation(s)
- Dristhi Ragoonanan
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (D.R.); (I.N.S.)
| | - Irtiza N. Sheikh
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (D.R.); (I.N.S.)
| | - Sumit Gupta
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sajad J. Khazal
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Priti Tewari
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Demetrios Petropoulos
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shulin Li
- Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kris M. Mahadeo
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
27
|
Rognoni C, Barcellona MR, Bargellini I, Bavetta MG, Bellò M, Brunetto M, Carucci P, Cioni R, Crocetti L, D’Amato F, D’Amico M, Deagostini S, Deandreis D, De Simone P, Doriguzzi A, Finessi M, Fonio P, Grimaldi S, Ialuna S, Lagattuta F, Masi G, Moreci A, Scalisi D, Virdone R, Tarricone R. Cost-effectiveness analysis of personalised versus standard dosimetry for selective internal radiation therapy with TheraSphere in patients with hepatocellular carcinoma. Front Oncol 2022; 12:920073. [PMID: 36106105 PMCID: PMC9464985 DOI: 10.3389/fonc.2022.920073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/28/2022] [Indexed: 11/27/2022] Open
Abstract
Aims To perform a cost-effectiveness analysis (CEA) comparing personalised dosimetry with standard dosimetry in the context of selective internal radiation therapy (SIRT) with TheraSphere for the management of adult patients with locally advanced hepatocellular carcinoma (HCC) from the Italian Healthcare Service perspective. Materials and methods A partition survival model was developed to project costs and the quality-adjusted life years (QALYs) over a lifetime horizon. Clinical inputs were retrieved from a published randomised controlled trial. Health resource utilisation inputs were extracted from the questionnaires administered to clinicians in three oncology centres in Italy, respectively. Cost parameters were based on Italian official tariffs. Results Over a lifetime horizon, the model estimated the average QALYs of 1.292 and 0.578, respectively, for patients undergoing personalised and standard dosimetry approaches. The estimated mean costs per patient were €23,487 and €19,877, respectively. The incremental cost-utility ratio (ICUR) of personalised versus standard dosimetry approaches was €5,056/QALY. Conclusions Personalised dosimetry may be considered a cost-effective option compared to standard dosimetry for patients undergoing SIRT for HCC in Italy. These findings provide evidence for clinicians and payers on the value of personalised dosimetry as a treatment option for patients with HCC.
Collapse
Affiliation(s)
- Carla Rognoni
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Bocconi University, Milan, Italy
- *Correspondence: Carla Rognoni,
| | | | | | | | - Marilena Bellò
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy
| | | | - Patrizia Carucci
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy
| | - Roberto Cioni
- Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | | | - Fabio D’Amato
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Mario D’Amico
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Simona Deagostini
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy
| | - Désirée Deandreis
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy
| | | | - Andrea Doriguzzi
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy
| | - Monica Finessi
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy
| | - Paolo Fonio
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy
| | - Serena Grimaldi
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy
| | - Salvatore Ialuna
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Fabio Lagattuta
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Gianluca Masi
- Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Antonio Moreci
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Daniele Scalisi
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Roberto Virdone
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Rosanna Tarricone
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Bocconi University, Milan, Italy
- Department of Policy Analysis and Public Management, Bocconi University, Milan, Italy
| |
Collapse
|
28
|
Abana CZY, Lamptey H, Bonney EY, Kyei GB. HIV cure strategies: which ones are appropriate for Africa? Cell Mol Life Sci 2022; 79:400. [PMID: 35794316 PMCID: PMC9259540 DOI: 10.1007/s00018-022-04421-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/10/2022]
Abstract
Although combination antiretroviral therapy (ART) has reduced mortality and improved lifespan for people living with HIV, it does not provide a cure. Patients must be on ART for the rest of their lives and contend with side effects, unsustainable costs, and the development of drug resistance. A cure for HIV is, therefore, warranted to avoid the limitations of the current therapy and restore full health. However, this cure is difficult to find due to the persistence of latently infected HIV cellular reservoirs during suppressive ART. Approaches to HIV cure being investigated include boosting the host immune system, genetic approaches to disable co-receptors and the viral genome, purging cells harboring latent HIV with latency-reversing latency agents (LRAs) (shock and kill), intensifying ART as a cure, preventing replication of latent proviruses (block and lock) and boosting T cell turnover to reduce HIV-1 reservoirs (rinse and replace). Since most people living with HIV are in Africa, methods being developed for a cure must be amenable to clinical trials and deployment on the continent. This review discusses the current approaches to HIV cure and comments on their appropriateness for Africa.
Collapse
Affiliation(s)
- Christopher Zaab-Yen Abana
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Helena Lamptey
- Department of Immunology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Evelyn Y Bonney
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - George B Kyei
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.
- Departments of Medicine and Molecular Microbiology, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, MO, USA.
- Medical and Scientific Research Center, University of Ghana Medical Centre, Accra, Ghana.
| |
Collapse
|
29
|
Ring A, Grob B, Aerts E, Ritter K, Volbracht J, Schär B, Greiling M, Müller AMS. Resource utilization for chimeric antigen receptor T cell therapy versus autologous hematopoietic cell transplantation in patients with B cell lymphoma. Ann Hematol 2022; 101:1755-1767. [PMID: 35759026 PMCID: PMC9279251 DOI: 10.1007/s00277-022-04881-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022]
Abstract
CD19-directed chimeric antigen receptor T cells (CAR-T) have emerged as a highly efficacious treatment for patients with relapsed/refractory (r/r) B cell lymphoma (BCL). The value of CAR-T for these patients is indisputable, but one-off production costs are high, and little is known about the ancillary resource consumption associated with CAR-T treatment. Here, we compared the resource use and costs of CAR-T treatment with high-dose chemotherapy followed by autologous stem cell transplantation (ASCT) for patients with r/r BCL. Standard operating procedures were used to develop a process model in ClipMedPPM, which comprises all activities and processes to sustain or generate treatment components that together constitute a treatment path. The software allows a graphic representation and the use of standardized linguistic elements for comparison of different treatment paths. Detailed processes involved in CAR-T treatments (n = 1041 processes) and in ASCT (n = 1535) were analyzed for time consumption of treatment phases and personnel. Process costs were calculated using financial controlling data. CAR-T treatment required ~ 30% less staff time than ASCT (primarily nursing staff) due to fewer chemotherapy cycles, less outpatient visits, and shorter hospital stays. For CAR-T, production costs were ~ 8 × higher, but overall treatment time was shorter compared with ASCT (30 vs 48 days), and direct labor and overhead costs were 40% and 10% lower, respectively. Excluding high product costs, CAR-T uses fewer hospital resources than ASCT for r/r BCL. Fewer hospital days for CAR-T compared to ASCT treatment and the conservation of hospital resources are beneficial to patients and the healthcare system.
Collapse
Affiliation(s)
- Alexander Ring
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Björn Grob
- Health Economics Market Access Pricing, Zurich, Switzerland
| | - Erik Aerts
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Katharina Ritter
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Jörk Volbracht
- Division of Controlling and Data Management, University Hospital Zurich, Zurich, Switzerland
| | - Bettina Schär
- Division of Controlling and Data Management, University Hospital Zurich, Zurich, Switzerland
| | - Michael Greiling
- Institute for Workflow-Management in Health Care, European University of Applied Sciences, Berlin, Germany
| | - Antonia M S Müller
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland.
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
30
|
Shah BD, Smith NJ, Feng C, Jeyakumar S, Castaigne JG, Faghmous I, Masouleh BK, Malone DC, Bishop MR. Cost-Effectiveness of KTE-X19 for Adults with Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia in the United States. Adv Ther 2022; 39:3678-3695. [PMID: 35727476 PMCID: PMC9309154 DOI: 10.1007/s12325-022-02201-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/19/2022] [Indexed: 12/03/2022]
Abstract
Introduction Despite currently available treatments for adults with relapsed/refractory acute lymphoblastic leukemia (R/R ALL), survival outcomes remain poor, highlighting the need for new therapeutic strategies. This study estimates the cost-effectiveness of KTE-X19 to treat adults with R/R ALL from a US payer perspective. Methods The model had two components: a decision-tree, where pre-infusion costs for patients who ultimately did not receive KTE-X19 are accounted for, followed by a partitioned survival analysis, where all KTE-X19 infused patients would enter the three-state (pre-progression, progressed disease, death) model. Comparators included current standard of care treatments, i.e., blinatumomab (BLIN), inotuzumab ozogamicin (INO), and salvage chemotherapy (CHEMO). Both standard parametric and mixture cure models were used to model survival. Efficacy, safety, healthcare resource utilization, and health state utility inputs were derived from the ZUMA-3 trial (NCT02614066) and literature. Cost inputs were derived from literature or publicly available sources. Outcomes and costs were discounted 3% annually. Results of KTE-X19 versus comparators are reported as total and incremental life-years (LYs), quality-adjusted life-years (QALYs), costs, and resulting incremental cost-effectiveness ratio (ICER). Deterministic and probabilistic sensitivity analyses (PSA) and key scenario analyses were also performed. Results In the base case, incremental QALYs for KTE-X19 were 2.44, 3.26, and 4.61 versus BLIN, INO, and CHEMO, respectively. Incremental costs were $50,913, $251,532, and $432,027, respectively, resulting in ICERs of $20,843/QALY (versus BLIN), $77,271/QALY (versus INO), and $93,768/QALY (versus CHEMO). Deterministic sensitivity analysis results were most sensitive to subsequent allogeneic stem cell transplant rates and post-progression utilities. PSA found that KTE-X19 is 78.4%, 74.0%, and 75.4% likely to be cost-effective versus BLIN, INO, and CHEMO, respectively. Across most scenarios, at a willingness-to-pay (WTP) threshold of $150,000/QALY, KTE-X19 was cost-effective versus all treatments. Conclusions Compared to current options for adults with R/R ALL, KTE-X19 is cost-effective, driven primarily by improved survival. Supplementary Information The online version contains supplementary material available at 10.1007/s12325-022-02201-6. Several treatments for adults with relapsed/refractory B-cell precursor acute lymphoblastic leukemia (R/R B-ALL) have been approved in the past decade in the US, including blinatumomab (BLIN) and inotuzumab ozogamicin (INO). However, despite the high costs associated with these treatments, survival for patients remains poor. KTE-X19, an autologous anti-CD19 chimeric antigen receptor T-cell (CAR-T) therapy, approved by the Food and Drug Administration in October 2021, has potential to improve survival, but its economic value has not yet been determined. This model comprehensively evaluated the long-term clinical and economic value of KTE-X19 versus current treatments, including BLIN, INO, and salvage chemotherapy (CHEMO). Inputs were derived from key clinical trials, the literature, and other publicly available sources. The model used the perspective of a US third party payer over a patient lifetime. Compared to BLIN, INO and CHEMO, KTE-X19 resulted in improved quality of life as measured with incremental quality-adjusted life years (QALYs) of 2.44 (vs BLIN), 3.26 (vs INO), and 4.61 (vs CHEMO). Treatment with KTE-X19 had incremental costs of $50,913 (vs BLIN), $251,532 (vs INO), and $432,027 (vs CHEMO). KTE-X19 was found to provide good value for money based on incremental cost-effectiveness ratios of $20,843/QALY (vs BLIN), $77,271/QALY (vs INO), and $93,768/QALY (vs CHEMO). These values are well below the commonly accepted thresholds to determine economic value. Results were also found to be robust across sensitivity and scenario analyses.
Collapse
Affiliation(s)
| | - Nathaniel J Smith
- Maple Health Group, LLC, 1740 Broadway, 15th Floor, New York, NY, 10019, USA.
| | | | - Sushanth Jeyakumar
- Maple Health Group, LLC, 1740 Broadway, 15th Floor, New York, NY, 10019, USA
| | | | | | | | | | - Michael R Bishop
- The David Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, USA
| |
Collapse
|
31
|
Simoens S, De Groote K, Boersma C. Critical Reflections on Reimbursement and Access of Advanced Therapies. Front Pharmacol 2022; 13:771966. [PMID: 35662719 PMCID: PMC9157586 DOI: 10.3389/fphar.2022.771966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Background: The health economic literature has questioned the cost-effectiveness and affordability of advanced therapies, proposed adjustments to value assessment frameworks, and discussed the use of outcome-based managed entry agreements and staggered payments in the last few years. The aim of this manuscript is to conduct a critical reflection on assessment criteria and access conditions for reimbursement of advanced therapies. Methods: A narrative review of the peer-reviewed literature and grey literature was conducted in April 2021 by searching PubMed; Google Scholar; policy and legislative documents; websites of health technology assessment agencies, advanced therapy organisations, governmental advanced therapy innovation programmes, consultancy agencies; ISPOR conference abstracts and presentations. Results: Based on the available evidence, this manuscript argues that: a) advanced therapies can be cost-effective at high prices set by manufacturers; b) the economic evaluation framework adopted by many payers under-values these products; c) advanced therapies can be affordable and may not require spread payments; d) outcome-based managed entry agreements are theoretically attractive, but challenging in practice; e) the cost-effectiveness of advanced therapies depends on the outcome-based managed entry agreement and payment approach; f) there is a role for multinational collaborations to manage reimbursement and access of advanced therapies. Conclusions: This manuscript shows that there is no single approach to reimbursement and access of advanced therapies. Instead, we support a more tailored assessment of health economic aspects of advanced therapies, which considers the heterogeneity of these products and their target populations.
Collapse
Affiliation(s)
- Steven Simoens
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | | | - Cornelis Boersma
- Health Ecore, Zeist, Netherlands.,Open Universiteit, Heerlen, Netherlands.,University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
32
|
Barriers to Chimeric Antigen Receptor T-Cell (CAR-T) Therapies in Clinical Practice. Pharmaceut Med 2022; 36:163-171. [PMID: 35672571 PMCID: PMC9217916 DOI: 10.1007/s40290-022-00428-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2022] [Indexed: 11/30/2022]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a revolutionary cancer treatment modality where a patient’s own T cells are collected and engineered ex vivo to express a chimeric antigen receptor (CAR). These reprogrammed CAR-T cells, when reinfused into the same patient, stimulate a T-cell mediated immune response against the antigen-expressing malignant cells leading to cell death. The initial results from pivotal clinical trials of CAR-T agents have been promising, leading to multiple approvals in various hematologic malignancies in the relapsed setting, including acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, follicular lymphoma, and, more recently, multiple myeloma. However, since the initial trials and US Food and Drug Administration approvals, there have been significant barriers to the widespread use of this therapy. The barriers to the use of CAR-T therapy include complex logistics, manufacturing limitations, toxicity concerns, and financial burden. This review discusses potential solutions to overcome these barriers in order to make this life-changing therapy widely accessible.
Collapse
|
33
|
Wang XJ, Wang YH, Ong MJC, Gkitzia C, Soh SY, Hwang WYK. Cost-Effectiveness and Budget Impact Analyses of Tisagenlecleucel in Pediatric and Young Adult Patients with Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia from the Singapore Healthcare System Perspective. CLINICOECONOMICS AND OUTCOMES RESEARCH 2022; 14:333-355. [PMID: 35535300 PMCID: PMC9078873 DOI: 10.2147/ceor.s355557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/07/2022] [Indexed: 12/29/2022] Open
Abstract
Purpose Children and young adults with relapsed or refractory (r/r) acute lymphoblastic leukemia (ALL) have poor survival due to ineffective therapy options. The newly approved chimeric antigen receptor T-cell (CAR-T) therapy, tisagenlecleucel, has demonstrated improved survival but at a high up-front cost. The study aims to evaluate the cost-effectiveness and budget impact of tisagenlecleucel versus salvage chemotherapy regimen (SCR) or blinatumomab (BLN) for the treatment of pediatric and young adult patients with relapsed/refractory B-cell ALL from the Singapore healthcare system perspective. Patients and Methods A three-health state partitioned survival model was constructed to analyze the cost-effectiveness of tisagenlecleucel vs SCR/BLN with/without allogenic hematopoietic stem cell transplantation (allo-HSCT) over a lifetime period. Clinical efficacy for tisagenlecleucel, SCR and BLN were based on pooled data from ELIANA, ENSIGN and B2101J trials, the study by von Stackelberg et al 2011, and MT103-205 respectively. Medical costs from pre-treatment until terminal care, including treatment, side effects, follow-up, subsequent allo-HSCT and relapse, were considered. Incremental cost-effectiveness ratios (ICERs) were estimated as the incremental costs per quality-adjusted life-year (QALY) gain. Additionally, the financial impact of tisagenlecleucel introduction in Singapore was estimated, comparing the present treatment scenario (without tisagenlecleucel) with a future scenario (with tisagenlecleucel), over 5 years. Results In the base-case analysis, tisagenlecleucel treatment demonstrated cost-effectiveness with an ICER of S$45,840 (US$34,762) per QALY (vs SCR) and S$51,978 (US$39,315) per QALY (vs BLN). The estimated budget ranges from S$477,857 (US$361,438) to S$1.4 million (US$1.05 million) annually for the initial 5 years. Conclusion Tisagenlecleucel is likely to be a cost-effective treatment option with limited budget implications while treating r/r ALL patients who have failed at least 2 lines of prior therapies.
Collapse
Affiliation(s)
| | - Yi-Ho Wang
- Novartis Asia Pacific Pharmaceuticals Pte Ltd, Singapore
| | | | | | | | - William Ying Khee Hwang
- National Cancer Centre Singapore, Singapore
- Singapore General Hospital, Singapore
- Duke-NUS Medical School, Singapore
| |
Collapse
|
34
|
Soliman R, Heneghan C, Bolous NS, Sidhom I, Ahmed S, Roberts N, Oke J, Elhaddad A. Systematic review of costs and cost-effectiveness of treatment for relapsed/refractory acute leukaemia in children and young adults. Expert Rev Hematol 2022; 15:345-357. [PMID: 35485262 DOI: 10.1080/17474086.2022.2069096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Survival outcomes of children with relapsed/refractory (r/r) acute leukaemia remain poor. Novel expensive treatments have been developed to improve their outcomes, yet, limited evidence exists about cost-effectiveness of alternative treatment strategies. AREAS COVERED A systematic review was conducted to summarize the health-economic evidence about costs/cost-effectiveness of treating r/r acute leukaemia in children/young adults. We searched Medline, Embase, and Cochrane databases until August 13th, 2021. Eligible articles included peer-reviewed original studies addressing r/r paediatric/young-adult acute lymphoblastic leukaemia (ALL), and acute myeloid leukaemia (AML). Quality assessment was conducted using Consolidated Health Economics Evaluation Reporting Standards (CHEERS) checklist. EXPERT OPINION The majority of papers focused on CAR-T cell therapy, which is still a novel treatment for r/r ALL, and was found to be cost-effective, yet, there remain concerns over its long-term effectiveness, affordability, and equity in access. The next best treatment option is Blinatumomab, followed by Clofarabine therapy, whereas FLA-IDA salvage chemotherapy provides least value for money. The quality of evidence is moderate to high, with limited generalizability of findings due to high variability in outcomes obtained from modelling studies. Limited studies evaluated r/r AML. We provide recommendations to deliver cost-effective treatments in real-world contexts, with implications for healthcare policy and practice.
Collapse
Affiliation(s)
- Ranin Soliman
- Department for Continuing Education, University of Oxford, UK.,Health Economics and Value Unit, Children's Cancer Hospital 57357 Egypt (CCHE), Egypt
| | - Carl Heneghan
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Nancy S Bolous
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, USA
| | - Iman Sidhom
- Department of Pediatric Oncology, Children's Cancer Hospital 57357 Egypt (CCHE), Egypt.,Department of Pediatric Oncology, National Cancer Institute, Cairo University, Egypt
| | - Sonia Ahmed
- Department of Pediatric Oncology, Children's Cancer Hospital 57357 Egypt (CCHE), Egypt.,Department of Pediatric Oncology, National Cancer Institute, Cairo University, Egypt
| | - Nia Roberts
- Oxford Bodleian Health Care Libraries, University of Oxford, Oxford, UK
| | - Jason Oke
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Alaa Elhaddad
- Department of Pediatric Oncology, Children's Cancer Hospital 57357 Egypt (CCHE), Egypt.,Department of Pediatric Oncology, National Cancer Institute, Cairo University, Egypt
| |
Collapse
|
35
|
Boettcher M, Joechner A, Li Z, Yang SF, Schlegel P. Development of CAR T Cell Therapy in Children-A Comprehensive Overview. J Clin Med 2022; 11:2158. [PMID: 35456250 PMCID: PMC9024694 DOI: 10.3390/jcm11082158] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 01/27/2023] Open
Abstract
CAR T cell therapy has revolutionized immunotherapy in the last decade with the successful establishment of chimeric antigen receptor (CAR)-expressing cellular therapies as an alternative treatment in relapsed and refractory CD19-positive leukemias and lymphomas. There are fundamental reasons why CAR T cell therapy has been approved by the Food and Drug administration and the European Medicines Agency for pediatric and young adult patients first. Commonly, novel therapies are developed for adult patients and then adapted for pediatric use, due to regulatory and commercial reasons. Both strategic and biological factors have supported the success of CAR T cell therapy in children. Since there is an urgent need for more potent and specific therapies in childhood malignancies, efforts should also include the development of CAR therapeutics and expand applicability by introducing new technologies. Basic aspects, the evolution and the drawbacks of childhood CAR T cell therapy are discussed as along with the latest clinically relevant information.
Collapse
Affiliation(s)
- Michael Boettcher
- Department of Pediatric Surgery, University Medical Centre Mannheim, University of Heidelberg, 69117 Heidelberg, Germany;
| | - Alexander Joechner
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia;
- Cellular Cancer Therapeutics Unit, Children’s Medical Research Institute, Sydney 2145, Australia; (Z.L.); (S.F.Y.)
| | - Ziduo Li
- Cellular Cancer Therapeutics Unit, Children’s Medical Research Institute, Sydney 2145, Australia; (Z.L.); (S.F.Y.)
| | - Sile Fiona Yang
- Cellular Cancer Therapeutics Unit, Children’s Medical Research Institute, Sydney 2145, Australia; (Z.L.); (S.F.Y.)
| | - Patrick Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia;
- Cellular Cancer Therapeutics Unit, Children’s Medical Research Institute, Sydney 2145, Australia; (Z.L.); (S.F.Y.)
- Department of Pediatric Hematology and Oncology, Westmead Children’s Hospital, Sydney 2145, Australia
| |
Collapse
|
36
|
Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion. Nat Rev Clin Oncol 2022; 19:342-355. [PMID: 35318469 DOI: 10.1038/s41571-022-00607-3] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2022] [Indexed: 12/14/2022]
Abstract
Chimeric antigen receptor (CAR) T cells have emerged as a potent therapeutic approach for patients with certain haematological cancers, with multiple CAR T cell products currently approved by the FDA for those with relapsed and/or refractory B cell malignancies. However, in order to derive the desired level of effectiveness, patients need to successfully receive the CAR T cell infusion in a timely fashion. This process entails apheresis of the patient's T cells, followed by CAR T cell manufacture. While awaiting infusion at an authorized treatment centre, patients may receive interim disease-directed therapy. Most patients will also receive a course of pre-CAR T cell lymphodepletion, which has emerged as an important factor in enabling durable responses. The time between apheresis and CAR T cell infusion is often not a simple journey, with each milestone being a critical step that can have important downstream consequences for the ability to receive the infusion and the strength of clinical responses. In this Review, we provide a summary of the many considerations for preparing patients with B cell non-Hodgkin lymphoma or acute lymphoblastic leukaemia for CAR T cell therapy, and outline current limitations and areas for future research.
Collapse
|
37
|
Petersohn S, Salles G, Wang M, Wu J, Wade SW, Simons CL, Bennison C, Siddiqi R, Peng W, Kloos I, Castaigne G, Hess G. Cost-effectiveness analysis of KTE-X19 CAR T therapy versus real-world standard of care in patients with relapsed/refractory mantle cell lymphoma post BTKi in England. J Med Econ 2022; 25:730-740. [PMID: 35611697 DOI: 10.1080/13696998.2022.2079317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIMS The objective of this study is to estimate the cost-effectiveness of KTE-X19 versus standard of care (SoC) in the treatment of patients with relapsed/refractory (R/R) mantle cell lymphoma (MCL) post-Bruton tyrosine kinase inhibitor (BTKi) treatment from a UK healthcare perspective. MATERIALS AND METHODS A three-state partitioned survival model (pre-progression, post-progression and death) with a cycle length of one month was used to extrapolate progression-free and overall survival over a lifetime horizon. Population inputs along with KTE-X19 (brexucabtagene autoleucel) efficacy and safety data were derived from the single-arm trial ZUMA-2 (NCT02601313). The composition of SoC was informed by a literature-based meta-analysis, SoC efficacy data were obtained from the SCHOLAR-2 real-world study. Survival was modelled using standard parametric curves for SoC and a mixture-cure methodology for KTE-X19. It was assumed that patients whose disease had not progressed after five years experienced long-term remission. Costs, resource use and utility, and adverse event disutility inputs were obtained from published literature and publicly available data sources. An annual discount rate of 3.5% was applied to costs and health outcomes. Modelled outcomes for KTE-X19 and SoC included expected life years (LY), quality-adjusted life years (QALY) and total costs. Deterministic and probabilistic sensitivity analyses and scenario analyses were performed. RESULTS Estimated median survival was 5.96 years for KTE-X19 and 1.38 for SoC. Discounted LYs, QALYs and lifetime costs were 8.27, 5.99 and £385,765 for KTE-X19 versus 1.98, 1.48 and £79,742 for SoC, respectively. The KTE-X19 versus SoC cost per QALY was £67,713 and the cost per LY was £48,645. Influential scenario analyses use alternative KTE-X19 survival curves and discount rates, and shorter time horizons. CONCLUSION Considering the survival and quality of life benefits compared to SoC, KTE-X19 for R/R MCL appears as a cost-effective treatment in the real-world UK setting.
Collapse
Affiliation(s)
| | - Gilles Salles
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Michael Wang
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Jim Wu
- Kite, a Gilead Company, Santa Monica, United States
| | - Sally W Wade
- Wade Outcomes Research and Consulting, Salt Lake City, United States
| | | | | | | | - Weimin Peng
- Kite, a Gilead Company, Santa Monica, United States
| | - Ioana Kloos
- Kite, a Gilead Company, Santa Monica, United States
| | | | - Georg Hess
- Department of Hematology, Oncology and Pneumology University Medical School of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
38
|
Passamonti F, Corrao G, Castellani G, Mora B, Maggioni G, Gale RP, Della Porta MG. The future of research in hematology: Integration of conventional studies with real-world data and artificial intelligence. Blood Rev 2021; 54:100914. [PMID: 34996639 DOI: 10.1016/j.blre.2021.100914] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/26/2022]
Abstract
Most national health-care systems approve new drugs based on data of safety and efficacy from large randomized clinical trials (RCTs). Strict selection biases and study-entry criteria of subjects included in RCTs often do not reflect those of the population where a therapy is intended to be used. Compliance to treatment in RCTs also differs considerably from real world settings and the relatively small size of most RCTs make them unlikely to detect rare but important safety signals. These and other considerations may explain the gap between evidence generated in RCTs and translating conclusions to health-care policies in the real world. Real-world evidence (RWE) derived from real-world data (RWD) is receiving increasing attention from scientists, clinicians, and health-care policy decision-makers - especially when it is processed by artificial intelligence (AI). We describe the potential of using RWD and AI in Hematology to support research and health-care decisions.
Collapse
Affiliation(s)
- Francesco Passamonti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy; Hematology, ASST Sette Laghi, Ospedale di Circolo, Varese, Italy.
| | - Giovanni Corrao
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, University of Milano-Bicocca, Milan, Italy
| | - Gastone Castellani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Barbara Mora
- Department of Medicine and Surgery, University of Insubria, Varese, Italy; Hematology, ASST Sette Laghi, Ospedale di Circolo, Varese, Italy
| | - Giulia Maggioni
- IRCCS Humanitas Clinical and Research Center, Rozzano, Italy
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunolgy and Inflammation, Imperial College London, London, UK
| | - Matteo Giovanni Della Porta
- IRCCS Humanitas Clinical and Research Center, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
39
|
Bangit LEP, Derwich K. Current Molecular Advancements in Chimeric Antigen Receptor (CAR-T) Cells for the Treatment of Leukemia. J Adolesc Young Adult Oncol 2021; 11:346-351. [PMID: 34935501 DOI: 10.1089/jayao.2021.0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) is a relatively new treatment for pediatric leukemia and has been the focused of recent advancements. CAR is manufactured to express T cells through various ways such as using retroviruses, transposons and transposase, electroporation, and CRISPR (clustered regularly interspaced short palindromic repeats). Together, it provides flexibility since it recognizes proteins without the need of antigen processing and presentation, can recognize carbohydrates and lipids, and it has been proven to be cost-effective. Despite these benefits however, problems faced by this therapy include unrecognized tumor proteins possibly escaping the system, CAR T cell expression being transient, and the therapy being one of the most expensive cancer drug ever approved. As a result, recent progress has been ongoing where researchers have combined CAR-T cells with natural killer (NK) cells and different cytokines to maximize its efficacy and potency while limiting potential risks such as cytokine release syndrome. Consequently, these cells gained the ability to be universal-being able to be used to treat multiple patients, maintain viability for a longer period, and prevent relapse.
Collapse
Affiliation(s)
- Lavina Ery Paula Bangit
- Department of Pediatric Oncology, Hematology and Trasnplantology, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Derwich
- Department of Pediatric Oncology, Hematology and Trasnplantology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
40
|
Mazza R, Maher J. Prospects for Development of Induced Pluripotent Stem Cell-Derived CAR-Targeted Immunotherapies. Arch Immunol Ther Exp (Warsz) 2021; 70:2. [PMID: 34897554 PMCID: PMC8666432 DOI: 10.1007/s00005-021-00640-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/24/2021] [Indexed: 12/27/2022]
Abstract
Technologies required to generate induced pluripotent stem cells (iPSC) were first described 15 years ago, providing a strong impetus to the field of regenerative medicine. In parallel, immunotherapy has finally emerged as a clinically meaningful modality of cancer therapy. In particular, impressive efficacy has been achieved in patients with selected haematological malignancies using ex vivo expanded autologous T cells engineered to express chimeric antigen receptors (CARs). While solid tumours account for over 90% of human cancer, they currently are largely refractory to this therapeutic approach. Nonetheless, given the considerable innovation taking place worldwide in the CAR field, it is likely that effective solutions for common solid tumours will emerge in the near future. Such a development will create significant new challenges in the scalable delivery of these complex, costly and individualised therapies. CAR-engineered immune cell products that originate from iPSCs offer the potential to generate unlimited numbers of homogeneous, standardised cell products in which multiple defined gene modification events have been introduced to ensure safety, potency and reproducibility. Here, we review some of the emerging strategies in use to engineer CAR-expressing iPSC-derived drug products.
Collapse
Affiliation(s)
- Roberta Mazza
- Leucid Bio Ltd, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - John Maher
- Leucid Bio Ltd, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Cancer Centre, Great Maze Pond, London, SE1 9RT, UK.
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, BN21 2UD, East Sussex, UK.
| |
Collapse
|
41
|
Antigen receptor therapy in bone metastasis via optimal control for different human life stages. J Math Biol 2021; 83:44. [PMID: 34596800 DOI: 10.1007/s00285-021-01673-4] [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: 09/25/2020] [Revised: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
In this work we propose a bone metastasis model using power law growth functions in order to describe the biochemical interactions between bone cells and cancer cells. Experimental studies indicate that bone remodeling cycles are different for human life stages: childhood, young adulthood, and adulthood. In order to include such differences in our study, we estimate the model parameter values for each human life stage via bifurcation analysis. Results reveal an intrinsic relationship between the active period of remodeling cycles and the proliferation of cancer cells. Subsequently, using optimal control theory we analyze a possible antigen receptor therapy as a new treatment for bone metastasis. Theoretical results such as existence of optimal solutions are proved. Numerical simulations for late stages of bone metastasis are presented and a discussion of our results is carried out.
Collapse
|
42
|
Huguet M, Raimond V, Kaltenbach E, Augusto V, Perrier L. How much does the hospital stay for infusion of anti-CD19 CAR-T cells cost to the French National Health Insurance? Bull Cancer 2021; 108:1170-1180. [PMID: 34561025 DOI: 10.1016/j.bulcan.2021.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/04/2021] [Accepted: 06/21/2021] [Indexed: 11/18/2022]
Abstract
Chimeric antigen receptor T-cells (CAR-T cells) have the potential to be a major innovation as a new type of cancer treatment, but are associated with extremely high prices and a high level of uncertainty. This study aims to assess the cost of the hospital stay for the administration of anti-CD19 CAR-T cells in France. Data were collected from the French Medical Information Systems Program (PMSI) and all hospital stays associated with an administrated drug encoded 9439938 (tisagenlecleucel, Kymriah®) or 9440456 (axicabtagene ciloleucel, Yescarta®) between January 2019 and December 2020 were included. 485 hospital stays associated with an injection of anti-CD19 CAR-T cells were identified, of which 44 (9%), 139 (28.7%), and 302 (62.3%) were for tisagenlecleucel in acute lymphoblastic leukaemia (ALL), tisagenlecleucel in diffuse large B-cell lymphoma (DLBCL), and axicabtagene ciloleucel respectively. The lengths of the stays were 37.9, 23.8, and 25.9 days for tisagenlecleucel in ALL, tisagenlecleucel in DLBCL, and axicabtagene ciloleucel, respectively. The mean costs per hospital stay were € 372,400 for a tisagenlecleucel in ALL, € 342,903 for tisagenlecleucel in DLBCL, and € 366,562 for axicabtagene ciloleucel. CAR T-cells represented more than 80% of these costs. n=13 hospitals performed CAR-T cell injections, with two hospitals accounting for more than 50% of the total number of injections. This study provides original data in a context of limited information regarding the costs of hospitalization for patients undergoing CAR-T cell treatments. In addition to the financial burden, distance may also be an important barrier for accessing CAR T-cell treatment.
Collapse
Affiliation(s)
- Marius Huguet
- Mines Saint-Étienne, université Clermont-Auvergne, CNRS, UMR 6158 LIMOS, Centre CIS, 42023 Saint-Étienne, France; Université Lyon, centre Léon-Bérard, GATE L-SE UMR 5824, 69008 Lyon, France
| | - Véronique Raimond
- Haute Autorité de santé, Department of Economic and Public Health Evaluation, 93200 Saint Denis, France
| | - Emmanuelle Kaltenbach
- Haute Autorité de santé, Department of Economic and Public Health Evaluation, 93200 Saint Denis, France
| | - Vincent Augusto
- Mines Saint-Étienne, université Clermont-Auvergne, CNRS, UMR 6158 LIMOS, Centre CIS, 42023 Saint-Étienne, France
| | - Lionel Perrier
- Université Lyon, centre Léon-Bérard, GATE L-SE UMR 5824, 69008 Lyon, France; Human and Social Science Department, centre Léon-Bérard, 69008 Lyon, France; Haute Autorité de santé, Committee for Economic and Public Health Evaluation, 93200 Saint Denis, France.
| |
Collapse
|
43
|
Ho JK, Borle K, Dragojlovic N, Dhillon M, Kitchin V, Kopac N, Ross C, Lynd LD. Economic Evidence on Potentially Curative Gene Therapy Products: A Systematic Literature Review. PHARMACOECONOMICS 2021; 39:995-1019. [PMID: 34156648 DOI: 10.1007/s40273-021-01051-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 05/22/2023]
Abstract
OBJECTIVE The aim of this review was to summarize all available evidence on the cost effectiveness of potentially curative gene therapies and identify challenges that economic evaluations face in this area. METHODS We conducted a systematic review of four databases (PubMed/MEDLINE, Embase, CINAHL, EconLit) and grey literature sources. We conducted the search on August 23, 2019 and updated it on November 26, 2020. We included all English, French and Spanish language studies that addressed a gene therapy that had received regulatory approval or had entered a phase III trial, and also reported on costs related to the therapy. Critical appraisal was conducted to assess quality of reporting in included studies. RESULTS Fifty-six studies were identified. Of the 42 full economic evaluations, 71% (n = 30) evaluated chimeric antigen receptor T-cell therapies, most used either a Markov model (n = 17, 40%) and/or a partitioned survival model (n = 17, 40%), and 76% (n = 32) adopted a public or private payer perspective. The model characteristics with the greatest impact on cost effectiveness included assumptions about the efficacy of the treatment and the comparators used. CONCLUSION All gene therapies in this review were shown to be more effective than their comparators, although due to high costs not all were considered cost effective at standard cost-effectiveness thresholds. Despite their high cost, some gene therapies have the potential to dominate the alternatives in conditions with high mortality/disability. The choice of comparator and assumptions regarding long-term effectiveness had substantial impacts on cost-effectiveness estimates and need to be carefully considered. Both the quality of inputs and the quality of reporting were highly variable.
Collapse
Affiliation(s)
- Joseph Khoa Ho
- Faculty of Pharmaceutical Sciences, Collaboration for Outcomes Research and Evaluation, University of British Columbia, Vancouver, BC, Canada
| | - Kennedy Borle
- Faculty of Pharmaceutical Sciences, Collaboration for Outcomes Research and Evaluation, University of British Columbia, Vancouver, BC, Canada
| | - Nick Dragojlovic
- Faculty of Pharmaceutical Sciences, Collaboration for Outcomes Research and Evaluation, University of British Columbia, Vancouver, BC, Canada
| | - Manrubby Dhillon
- Faculty of Pharmaceutical Sciences, Collaboration for Outcomes Research and Evaluation, University of British Columbia, Vancouver, BC, Canada
| | - Vanessa Kitchin
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Nicola Kopac
- Faculty of Pharmaceutical Sciences, Collaboration for Outcomes Research and Evaluation, University of British Columbia, Vancouver, BC, Canada
| | - Colin Ross
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Larry D Lynd
- Faculty of Pharmaceutical Sciences, Collaboration for Outcomes Research and Evaluation, University of British Columbia, Vancouver, BC, Canada.
- Centre for Health Evaluation and Outcome Sciences, Providence Health Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
44
|
Salans M, Courtney PT, Yip A, Murphy JD. Cost-effectiveness of ipilimumab versus high-dose interferon as an adjuvant therapy in resected high-risk melanoma. Cancer Med 2021; 10:6618-6626. [PMID: 34402192 PMCID: PMC8495287 DOI: 10.1002/cam4.4194] [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: 06/04/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Adjuvant ipilimumab was found to improve the overall survival and reduce toxicity compared to high-dose interferon (HDI) in patients with resected, high-risk melanoma. However, the cost of ipilimumab is substantially higher than HDI. This study evaluates the cost-effectiveness of ipilimumab as an adjuvant treatment in melanoma from a healthcare perspective. METHODS We designed a Markov model simulating resected, high-risk melanoma patients receiving either ipilimumab or HDI. Transition probabilities, including risks of survival, disease progression, and toxicity, were ascertained from clinical trial data. Costs and quality of life measurements (health utilities) were extracted from the literature. Incremental cost-effectiveness ratios (ICERs), defined as incremental costs divided by incremental quality-adjusted life-years (QALYs), assessed cost-effectiveness. ICERs <$100,000/QALY were deemed cost-effective. We measured model uncertainty with one-way and probabilistic sensitivity analyses. RESULTS In our base case model, ipilimumab increased costs by $107,100 and increased effectiveness by 0.43 QALY, yielding an ICER of $392,600/QALY. Our model was moderately sensitive to the costs of ipilimumab, though the cost of ipilimumab would need to decrease by 44% for ipilimumab to become cost-effective compared to HDI. The model was not sensitive to survival, toxicity, or other costs. Probabilistic sensitivity analysis showed that HDI would remain the cost-effective treatment option 96.2% of the time at a willingness-to-pay threshold of $100,000/QALY. CONCLUSIONS Adjuvant ipilimumab increases the survival and decreases the toxicity compared to HDI in resected, high-risk melanoma patients, though this would not be considered cost-effective due to the high price of ipilimumab.
Collapse
Affiliation(s)
- Mia Salans
- University of California San Diego School of Medicine, La Jolla, California, USA.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | - Patrick Travis Courtney
- University of California San Diego School of Medicine, La Jolla, California, USA.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | - Anthony Yip
- University of California San Diego School of Medicine, La Jolla, California, USA.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | - James D Murphy
- University of California San Diego School of Medicine, La Jolla, California, USA.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
45
|
McCormick M, Lapinski J, Friehling E, Smith K. Premedication prior to PEG-asparaginase is cost-effective in pediatric patients with acute lymphoblastic leukemia. Pediatr Blood Cancer 2021; 68:e29051. [PMID: 33860989 DOI: 10.1002/pbc.29051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND PEG-asparaginase is critical in pediatric acute lymphoblastic leukemia (ALL) therapy but is highly immunogenic. Severe allergic reactions lead to substitution of further PEG-asparaginase with Erwinia. Erwinia is associated with more frequent dosing, increased expense, and limited availability. Premedication may reduce rates of allergic reactions. PROCEDURES This Markov model evaluated the cost-effectiveness of three strategies: premedication plus therapeutic drug monitoring (TDM), TDM alone, and no premedication or TDM. We modeled two scenarios: a standard-risk (SR) B-ALL patient receiving two asparaginase doses and a high-risk (HR) patient receiving seven asparaginase doses. The model incorporated costs of asparaginase, premedication, TDM and clinic visits, and lost parental wages associated with each additional Erwinia dose. We incorporated a five-year time horizon with a societal perspective. Outcomes were Erwinia substitutions avoided and differences in quality-adjusted life years (QALYs). Probabilistic and one-way sensitivity analyses evaluated model uncertainty. RESULTS In both scenarios, premedication was the least costly strategy. In SR and HR scenarios, premedication with monitoring resulted in 8% and 7% fewer changes to Erwinia compared with monitoring alone and 3% and 2% fewer changes compared with no premedication/monitoring, respectively. Premedication resulted in the most QALYs gained in the SR patients. Individual variation of model inputs did not change premedication/monitoring favorability for either scenario. In probabilistic sensitivity analyses, premedication/monitoring was favored in >87% of iterations in both scenarios. CONCLUSION Compared with other strategies, premedication use and asparaginase level monitoring in children with B-ALL is potentially cost-saving.
Collapse
Affiliation(s)
| | - Jillian Lapinski
- C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan
| | | | | |
Collapse
|
46
|
Geethakumari PR, Ramasamy DP, Dholaria B, Berdeja J, Kansagra A. Balancing Quality, Cost, and Access During Delivery of Newer Cellular and Immunotherapy Treatments. Curr Hematol Malig Rep 2021; 16:345-356. [PMID: 34089485 PMCID: PMC8179081 DOI: 10.1007/s11899-021-00635-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW The chimeric antigen receptor (CAR) T-cell therapy is currently changing the landscape of hematologic malignancies with multiple FDA-approved cell therapy products in the USA. The current administration process of the CAR T-cell therapy is complicated, labor-intensive, and expensive. RECENT FINDINGS The chimeric antigen receptor (CAR) T-cell therapy is currently changing the landscape of hematologic malignancies with multiple FDA-approved cell therapy products in the USA. The current administration process of the CAR T-cell therapy is complicated, labor-intensive, and expensive. This review article addresses the present-day challenges and discusses opportunities to optimize the access and affordability of the CAR T-cell therapy. The field of cellular immunotherapy is going to change the future of solid tumors and non-oncological diseases. However, this promising therapy poses challenges in the administration and management of quality in the current field of healthcare. We describe various novel approaches to manage challenges in improving access and improving widescale implementation of cellular therapies.
Collapse
Affiliation(s)
| | - Dheepthi Perumal Ramasamy
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 2201 Inwood Road, Dallas, TX, 76034, USA
| | | | - Jesús Berdeja
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN, USA
| | - Ankit Kansagra
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 2201 Inwood Road, Dallas, TX, 76034, USA.
| |
Collapse
|
47
|
Sussman M, Crivera C, Benner J, Adair N. Applying State-of-the-Art Survival Extrapolation Techniques to the Evaluation of CAR-T Therapies: Evidence from a Systematic Literature Review. Adv Ther 2021; 38:4178-4194. [PMID: 34251651 PMCID: PMC8342396 DOI: 10.1007/s12325-021-01841-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/22/2021] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Traditional statistical techniques for extrapolating short-term survival data for anticancer therapies assume the same mortality rate for noncured and "cured" patients, which is appropriate for projecting survival of non-curative therapies but may lead to an underestimation of the treatment effectiveness for potentially curative therapies. Our objective was to ascertain research trends in survival extrapolation techniques used to project the survival benefits of chimeric antigen receptor T cell (CAR-T) therapies. METHODS A global systematic literature search produced a review of survival analyses of CAR-T therapies, published between January 1, 2015 and December 14, 2020, based on publications sourced from MEDLINE, scientific conferences, and health technology assessment agencies. Trends in survival extrapolation techniques used, and the rationale for selecting advanced techniques, are discussed. RESULTS Twenty publications were included, the majority of which (65%, N = 13) accounted for curative intent of CAR-T therapies through the use of advanced extrapolation techniques, i.e., mixture cure models [MCMs] (N = 10) or spline-based models (N = 3). The authors' rationale for using the MCM approach included (a) better statistical fits to the observed Kaplan-Meier curves (KMs) and (b) visual inspection of the KMs indicated that a proportion of patients experienced long-term remission and survival which is not inherently captured in standard parametric distributions. DISCUSSION Our findings suggest that an advanced extrapolation technique should be considered in base case survival analyses of CAR-T therapies when extrapolating short-term survival data to long-term horizons extending beyond the clinical trial duration. CONCLUSION Advanced extrapolation techniques allow researchers to account for the proportion of patients with an observed plateau in survival from clinical trial data; by only using standard-partitioned modeling, researchers may risk underestimating the survival benefits for the subset of patients with long-term remission. Sensitivity analysis with an alternative advanced extrapolation technique should be implemented and re-assessment using clinical trial extension data and/or real-world data should be conducted as longer-term data become available.
Collapse
Affiliation(s)
- Matthew Sussman
- Panalgo LLC, 265 Franklin Street, Suite 1101, Boston, MA, 02110, USA.
| | | | - Jennifer Benner
- Panalgo LLC, 265 Franklin Street, Suite 1101, Boston, MA, 02110, USA
| | - Nicholas Adair
- Panalgo LLC, 265 Franklin Street, Suite 1101, Boston, MA, 02110, USA
| |
Collapse
|
48
|
Banerjee R, Fakhri B, Shah N. Toci or not toci: innovations in the diagnosis, prevention, and early management of cytokine release syndrome. Leuk Lymphoma 2021; 62:2600-2611. [PMID: 34151714 DOI: 10.1080/10428194.2021.1924370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cytokine release syndrome (CRS) remains a significant toxicity of chimeric antigen receptor T-cell (CAR-T) therapy for hematologic malignancies. While established guidelines exist for the management of Grade 2+ CRS with immunosuppressive agents such as tocilizumab or corticosteroids, the management of early-grade CRS (i.e. Grade 1 CRS with isolated fevers) has no such consensus beyond supportive care. In this review, we discuss early-grade CRS with an emphasis on its diagnosis, management, and prevention. Strategies to target early-grade CRS include immunosuppression preemptively (once CRS develops) or prophylactically (before CRS develops) as well as novel small-molecule inhibitors or fractionated CAR-T dosing. In the near future, next-generation CAR-T therapies may be able to target CRS precisely or obviate CRS entirely. If shown to prevent CRS-associated morbidity while maintaining therapeutic anti-neoplastic efficacy, these innovative strategies will enhance the safety of CAR-T therapy while also improving its operationalization and accessibility in the real-world setting.
Collapse
Affiliation(s)
- Rahul Banerjee
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Bita Fakhri
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nina Shah
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
49
|
Moradi-Lakeh M, Yaghoubi M, Seitz P, Javanbakht M, Brock E. Cost-Effectiveness of Tisagenlecleucel in Paediatric Acute Lymphoblastic Leukaemia (pALL) and Adult Diffuse Large B-Cell Lymphoma (DLBCL) in Switzerland. Adv Ther 2021; 38:3427-3443. [PMID: 34021886 DOI: 10.1007/s12325-021-01767-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/28/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION The purpose of the present analysis was to explore the cost-effectiveness of tisagenlecleucel in relapsed or refractory (r/r) paediatric acute lymphoblastic leukaemia (pALL) and r/r adult diffuse large B-cell lymphoma (DLBCL) in Switzerland against a range of historical standard-of-care treatments. METHODS Two cost-utility models were constructed for the two licensed indications using similar methodologies but indication-specific data. Clinical efficacy data were based on pooled analyses of clinical trials for tisagenlecleucel (pALL: ELIANA, ENSIGN, B2101J; DLBCL: JULIET, NCT02030834) and published data for comparator treatments. Treatment effects were compared based on matching-adjusted indirect comparison (MAIC) analyses. Four clinical lymphoma and leukaemia experts provided Switzerland-specific input regarding comparators, diagnostic and therapeutic procedures, clinical evidence and costs, which were used to inform the models. The base case analysis reflected the perspective of the Swiss mandatory health insurance system. Deterministic, probabilistic and scenario analyses were carried out to explore the robustness of results. RESULTS The base case analysis resulted in incremental costs of CHF 31,961-CHF 36,419 per quality-adjusted life year (QALY) gained for pALL across the different comparators and CHF 113,179 for DLBCL (1 CHF = 1.09 USD). Incremental costs per life-year gained ranged between CHF 33,906-CHF 97,399 across the two indications. Including productivity gains, tisagenlecleucel was shown to be dominant (more effective and less costly) over all the comparators for pALL and to result in incremental costs per life-year gained of CHF 57,324 for DLBCL. CONCLUSION Using hypothetical willingness-to-pay thresholds of CHF 100,000-150,000 per QALY gained, the present analysis has shown tisagenlecleucel to be a cost-effective treatment option in pALL and DLBCL.
Collapse
Affiliation(s)
| | - Mohsen Yaghoubi
- Department of Pharmacy Practice, Mercer University College of Pharmacy, 3001 Mercer University Dr, Atlanta, GA, 30341, USA
- Canada Optimax Access Consulting, 1803-2138 Madison Ave, Burnaby, BC, V5C6T6, Canada
| | - Patrick Seitz
- Novartis Pharma Schweiz AG, Suurstoffi 14, 6343, Rotkreuz, Switzerland
| | - Mehdi Javanbakht
- Optimax Access Ltd., Suite 30 Kenneth Dibben House, Enterprise Road, Southampton Science Park, Chilworth, Southampton, SO16 7NS, UK
| | | |
Collapse
|
50
|
Forsythe A, Sandman K. What Does the Economic Burden of Acute Myeloid Leukemia Treatment Look Like for the Next Decade? An Analysis of Key Findings, Challenges and Recommendations. J Blood Med 2021; 12:245-255. [PMID: 33981169 PMCID: PMC8107010 DOI: 10.2147/jbm.s279736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/13/2021] [Indexed: 12/24/2022] Open
Abstract
Acute myeloid leukemia (AML) is conventionally treated with chemotherapy in eligible patients. Potentially curative regimens are associated with significant toxicity, and the major cost drivers in AML historically have been hospitalization and hematopoietic stem cell transplantation. The past several years have seen a dramatic increase in the number of treatment options, including oral therapies and drugs targeted to biological pathways implicated in AML. Major current and future drivers of cost in AML include hospitalization and medical costs, stem cell transplantation for eligible patients, and medication costs. It is likely that hospitalization and medical costs will decline as more AML treatment moves to the outpatient setting. Stem cell transplantation costs may increase, if more patients are eligible for improved procedures, although the overall cost of transplantation could decrease if new procedures reduce the need for hospitalization. Medication costs are likely to increase, with various branded drugs available and in development. From a broader perspective, another driver of cost is the proportion of patients with AML who can undergo treatment. Patients who may previously have been unable to tolerate chemotherapy are more likely to be treated with the range of less intensive, more tolerable options now available. The effectiveness of newer AML treatment options also suggests that, overall, there may be more patients staying alive and on treatment longer than in the past. While certain advances, such as increased use of oral and outpatient therapies, could potentially reduce costs, the overall economic impact of AML is likely to increase as more patients are eligible for novel therapies across several phases from induction to maintenance to relapsed/refractory disease. While these novel therapies have the potential to deliver value in the form of improved efficacy, safety, and convenience, payers will need to determine how to cover a longer, more complex AML treatment pathway.
Collapse
|