1
|
Boytsov N, Multani JK, Zhou Z, Kotowsky N, Bolgioni-Smith A, Huo T, Paltanwale Q, McNamara S, Chen CC. Impact of social determinants of health on treatment patterns and outcomes in multiple myeloma. Future Oncol 2025:1-11. [PMID: 40351131 DOI: 10.1080/14796694.2025.2498878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
AIMS Evaluate treatment and outcomes by social determinants of health (SDoH) in multiple myeloma (MM), which are important for improving care and outcomes. METHODS This was a retrospective study of real-world patients enrolled in a US insurance claims database (MM diagnosis, July 2018-December 2022) with linkage to a SDoH database, supplemented with mortality, provider affiliation (academic/community), and socioeconomically disadvantaged area databases. Treatment and outcomes were evaluated across SDoH domains: race/ethnicity, education level, transportation access, food insecurity, risky health behaviors, living in disadvantaged areas, healthcare needs, and ease of healthcare-systems engagement. RESULTS The study included 4768 patients (2295 and 2731 with care-setting and treatment data); median follow-up, 584 days. Patients treated in academic versus community settings were less likely to be food insecure and live in disadvantaged areas and had lower healthcare needs. Stem cell transplant was more common in White versus non-White patients, those with low versus high food insecurity and healthcare needs, and high versus low ease of healthcare-systems engagement. In multivariable analysis, high versus low disadvantaged areas (HR = 1.75) and medium versus low food insecurity (HR = 1.80) were associated with shorter overall survival. CONCLUSIONS These findings indicate a need for improved access to care in the broader MM population.
Collapse
Affiliation(s)
- Natalie Boytsov
- Real World Evidence & Health Outcomes Research, GSK, Upper Providence, PA, USA
| | - Jasjit K Multani
- Health Economics and Outcomes Research, IQVIA, Falls Church, VA, USA
| | - Zifan Zhou
- Health Economics and Outcomes Research, IQVIA, Falls Church, VA, USA
| | - Nirali Kotowsky
- Real World Evidence & Health Outcomes Research, GSK, San Francisco, CA, USA
| | | | - Tianyao Huo
- Advanced Analytics, IQVIA, King of Prussia, PA, USA
| | | | - Simon McNamara
- Real World Evidence & Health Outcomes Research, GSK, Stevenage, Hertfordshire, UK
| | - Chi-Chang Chen
- Health Economics and Outcomes Research, IQVIA, King of Prussia, PA, USA
| |
Collapse
|
2
|
Majhail NS, Cox T, Larson S, Battiwalla M, Ramakrishnan A, Shaughnessy P, Tees M, Zahradka N, Wilkes M, Pantin J. Outpatient Administration of Chimeric Antigen Receptor T-Cell Therapy Using Remote Patient Monitoring. JCO Oncol Pract 2025:OP2500062. [PMID: 40249898 DOI: 10.1200/op-25-00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 04/20/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies are standard of care for the treatment of several hematologic malignancies. Although patients receiving CAR-T therapies are frequently hospitalized given risks of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), there is increasing interest and evidence for the safety of their outpatient administration. We review various models of care and provide operational considerations for centers that are interested in developing outpatient CAR-T programs, with a particular emphasis on using remote patient monitoring (RPM) to facilitate outpatient care. Safe and high-quality outpatient care requires involvement of a multidisciplinary team with clinical pathways for rapid triage and evaluation for CRS and ICANS and their management and, if necessary, timely transition of patients to a higher level of acute care. RPM can facilitate scaling an outpatient program in a cost-effective manner, especially across multiple sites of care, and can reduce the time patients spend in an acute care setting. Overall minimizing hospital-based care and an outpatient approach can alleviate capacity challenges treatment centers have faced that have partly impacted access to CAR-T therapies and have the potential to positively impact patient and caregiver experience and quality of life.
Collapse
Affiliation(s)
- Navneet S Majhail
- Sarah Cannon Transplant and Cellular Therapy Network, Sarah Cannon Cancer Network, Nashville, TN
- Sarah Cannon Transplant and Cellular Therapy Network Program at TriStar Centennial Medical Center, Nashville, TN
| | - Tonya Cox
- Sarah Cannon Transplant and Cellular Therapy Network, Sarah Cannon Cancer Network, Nashville, TN
| | - Stephanie Larson
- Sarah Cannon Transplant and Cellular Therapy Network, Sarah Cannon Cancer Network, Nashville, TN
| | - Minoo Battiwalla
- Sarah Cannon Transplant and Cellular Therapy Network, Sarah Cannon Cancer Network, Nashville, TN
- Sarah Cannon Transplant and Cellular Therapy Network Program at TriStar Centennial Medical Center, Nashville, TN
| | - Aravind Ramakrishnan
- Sarah Cannon Transplant and Cellular Therapy Network, Sarah Cannon Cancer Network, Nashville, TN
- Sarah Cannon Transplant and Cellular Therapy Network Program at South Austin Medical Center, Austin, TX
| | - Paul Shaughnessy
- Sarah Cannon Transplant and Cellular Therapy Network, Sarah Cannon Cancer Network, Nashville, TN
- Sarah Cannon Transplant and Cellular Therapy Network Program at Methodist Hospital, San Antonio, TX
| | - Michael Tees
- Sarah Cannon Transplant and Cellular Therapy Network, Sarah Cannon Cancer Network, Nashville, TN
- Colorado Blood Cancer Institute, Denver, CO
- Sarah Cannon Transplant and Cellular Therapy Program at Presbyterian/St Luke's Medical Center, Denver, CO
| | | | | | - Jeremy Pantin
- Sarah Cannon Transplant and Cellular Therapy Network, Sarah Cannon Cancer Network, Nashville, TN
- Sarah Cannon Transplant and Cellular Therapy Network Program at TriStar Centennial Medical Center, Nashville, TN
| |
Collapse
|
3
|
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
|
4
|
Di M, Su CT, Cowan AJ, Gopal AK, Banerjee R. Mitigating time toxicity in lymphoma and multiple myeloma. Leuk Lymphoma 2024; 65:1418-1429. [PMID: 38749027 DOI: 10.1080/10428194.2024.2352086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 04/30/2024] [Indexed: 10/02/2024]
Abstract
The concept of time toxicity in oncology refers to the presence of frequent healthcare-related interactions that can interfere with patient well-being. In this review, we examine several manifestations of time toxicity in non-Hodgkin lymphoma and multiple myeloma and discuss their impact on decision-making with patients. For example, time toxicity may influence the choice of chemoimmunotherapy versus lenalidomide-rituximab in follicular lymphoma. In myeloma, it may inform the optimal dosing schedule for proteasome inhibitors and bisphosphonates. In both malignancies, varying time toxicity profiles are a key distinction between chimeric antigen receptor T-cell therapies and bispecific antibodies. We outline the challenges with measuring time toxicity as a trial endpoint but discuss its importance as a consideration for patient care, both in standard-of-care settings and in clinical trials. Throughout the review, we highlight strategies to lower the time toxicity of therapies in lymphoma and myeloma without compromising their efficacy or patient safety.
Collapse
Affiliation(s)
- Mengyang Di
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology/Oncology, University of Washington, Seattle, WA, USA
- Hutchinson Institute for Cancer Outcomes Research, Seattle, WA, USA
| | - Christopher T Su
- Division of Hematology/Oncology, University of Washington, Seattle, WA, USA
- Hutchinson Institute for Cancer Outcomes Research, Seattle, WA, USA
| | - Andrew J Cowan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology/Oncology, University of Washington, Seattle, WA, USA
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology/Oncology, University of Washington, Seattle, WA, USA
| | - Rahul Banerjee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology/Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Siefen AC, Kurte MS, Jakobs F, Teichert M, von Tresckow B, Reinhardt HC, Holtick U, Atta J, Jehn C, Sala E, Warnecke A, Hänel M, Scheid C, Kron F. How to optimize the CAR-T Cell therapy process? A group concept mapping analysis of preconditions for a frictionless process from a German multistakeholder perspective. Front Oncol 2024; 14:1466803. [PMID: 39376984 PMCID: PMC11456517 DOI: 10.3389/fonc.2024.1466803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/28/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction Treatment with chimeric antigen receptor T (CAR-T) cells involves a large number of interdisciplinary stakeholders and is associated with complex processes ranging from patient-specific production to follow-up care. Due to the complexity, maximum process optimization is required in order to avoid efficiency losses. This study aimed at systematically determining the preconditions for a frictionless flow of the CAR-T process by surveying the stakeholders involved. Methods A Group Concept Mapping (GCM) analysis, a mixed-methods participatory research, was conducted. CAR-T experts from different professional backgrounds went through three steps: 1) Brainstorming relevant aspects (statements) for a frictionless process, 2) Sorting the collected statements based on their similarity, and 3) Rating the importance and feasibility of each statement. A cluster map reflecting the overarching topics was derived, and mean ratings per statement and cluster were calculated. Results Overall, 20 CAR-T experts participated. A total of 80 statements were collected, resulting in a map of the following 10 clusters (mean importance/feasibility): Information for patients and physicians (4.16/3.77), Supportive network (4.03/3.53), Eligibility of patients (4.41/3.63), Evidence, transparency and communication (4.01/3.33), Paperwork (4.1/2.52), Interface with pharmaceutical manufacturer (4.03/2.85), Reimbursement (4.29/2.31), Quality Management (4.17/3.18), Infrastructure of CAR-T clinics (4.1/2.93), and Patient-oriented processes (4.46/3.32). Discussion The 80 statements underlined the complex and manifold nature of the CAR-T treatment process. Our results reflect the first step in overcoming hurdles: identifying potential hurdles and required preconditions. Decision-makers and stakeholders can use the results to derive strategies and measures to further promote a frictionless process.
Collapse
Affiliation(s)
- Ann-Cathrine Siefen
- Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
- VITIS Healthcare Group, Cologne, Germany
| | - Melina Sophie Kurte
- Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
- VITIS Healthcare Group, Cologne, Germany
| | - Florian Jakobs
- Department of Hematology and Stem Cell Transplantation, Faculty of Medicine and University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Marcel Teichert
- Department of Hematology and Stem Cell Transplantation, Faculty of Medicine and University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK Partner Site Essen), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, Faculty of Medicine and University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Johannes Atta
- Department of Hematology, Giessen and Marburg University Hospital (UKGM), Giessen, Germany
| | - Christian Jehn
- Department of Hematology/Oncology and Stem-cell Transplantation, St Georg, Asklepios Hamburg, Hamburg, Germany
| | - Elisa Sala
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Anke Warnecke
- Department of Hematology and Medical Oncology, Division for Stem Cell Transplantation and Cellular Therapy, University Hospital Göttingen, Göttingen, Germany
| | - Mathias Hänel
- Department of Internal Medicine III, Klinikum Chemnitz, Chemnitz, Germany
| | - Christof Scheid
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Kron
- VITIS Healthcare Group, Cologne, Germany
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Fachhochschule für Oekonomie & Management (FOM) University of Applied Sciences, Essen, Germany
| |
Collapse
|
6
|
Srivastava S, Tyagi A, Pawar VA, Khan NH, Arora K, Verma C, Kumar V. Revolutionizing Immunotherapy: Unveiling New Horizons, Confronting Challenges, and Navigating Therapeutic Frontiers in CAR-T Cell-Based Gene Therapies. Immunotargets Ther 2024; 13:413-433. [PMID: 39219644 PMCID: PMC11365499 DOI: 10.2147/itt.s474659] [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: 05/11/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The CAR-T cell therapy has marked the dawn of new era in the cancer therapeutics and cell engineering techniques. The review emphasizes on the challenges that obstruct the therapeutic efficiency caused by cell toxicities, immunosuppressive tumor environment, and decreased T cell infiltration. In the interest of achieving the overall survival (OS) and event-free survival (EFS) of patients, the conceptual background of potential target selection and various CAR-T cell design techniques are described which can minimize the off-target effects, reduce toxicity, and thus increase the resilience of CAR-T cell treatment in the haematological malignancies as well as in solid tumors. Furthermore, it delves into cutting-edge technologies like gene editing and synthetic biology, providing new opportunities to enhance the functionality of CAR-T cells and overcome mechanisms of immune evasion. This review provides a comprehensive understanding of the complex and diverse aspects of CAR-T cell-based gene treatments, including both scientific and clinical aspects. By effectively addressing the obstacles and utilizing the capabilities of cutting-edge technology, CAR-T cell therapy shows potential in fundamentally changing immunotherapy and reshaping the approach to cancer treatment.
Collapse
Affiliation(s)
- Shivani Srivastava
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Anuradha Tyagi
- Department of cBRN, Institute of Nuclear Medicine and Allied Science, Delhi, India
| | | | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Arora
- Advanced Instrumentation Research Facility, Jawaharlal Nehru University, New Delhi, India
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
- Department of Biotechnology, SSET, Sharda University, Greater Noida, 201306, India
| | - Vinay Kumar
- Pennsylvania State University Hershey Medical Center, 500 University Dr, Heshey, PA, USA
| |
Collapse
|
7
|
Fowler NH, Chavez JC, Riedell PA. Moving T-Cell Therapies into the Standard of Care for Patients with Relapsed or Refractory Follicular Lymphoma: A Review. Target Oncol 2024; 19:495-510. [PMID: 38896212 PMCID: PMC11271334 DOI: 10.1007/s11523-024-01070-z] [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] [Accepted: 05/06/2024] [Indexed: 06/21/2024]
Abstract
Patients with follicular lymphoma, an indolent form of non-Hodgkin lymphoma, typically experience multiple relapses over their disease course. Periods of remission become progressively shorter with worse clinical outcomes after each subsequent line of therapy. Currently, no clear standard of care/preferred treatment approach exists for patients with relapsed or refractory follicular lymphoma. As novel agents continue to emerge for treatment in the third-line setting, guidance is needed for selecting the most appropriate therapy for each patient. Several classes of targeted therapeutic agents, including monoclonal antibodies, phosphoinositide 3-kinase inhibitors, enhancer of zeste homolog 2 inhibitors, chimeric antigen receptor (CAR) T-cell therapies, and bispecific antibodies, have been approved by regulatory authorities based on clinical benefit in patients with relapsed or refractory follicular lymphoma. Additionally, antibody-drug conjugates and other immunocellular therapies are being evaluated in this setting. Effective integration of CAR-T cell therapy into the treatment paradigm after two or more prior therapies requires appropriate patient selection based on transformation status following a rebiopsy; a risk evaluation based on age, fitness, and remission length; and eligibility for CAR-T cell therapy. Consideration of important logistical factors (e.g., proximity to the treatment center and caregiver support during key periods of CAR-T cell therapy) is also critical. Overall, an individualized treatment plan that considers patient-related factors (e.g., age, disease status, tumor burden, comorbidities) and prior treatment types is recommended for patients with relapsed or refractory follicular lymphoma. Future analyses of real-world data and a better understanding of mechanisms of relapse are needed to further refine patient selection and identify optimal sequencing of therapies in this setting.
Collapse
Affiliation(s)
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Peter A Riedell
- David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, USA
| |
Collapse
|
8
|
Cui C, Feng C, Rosenthal N, Wade SW, Curry L, Fu C, Shah GL. Hospital healthcare resource utilization and costs for chimeric antigen T-cell therapy and autologous hematopoietic cell transplant in patients with large B-cell lymphoma in the United States. Leuk Lymphoma 2024; 65:922-931. [PMID: 38567630 DOI: 10.1080/10428194.2024.2331084] [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: 03/08/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
The efficacy of chimeric antigen receptor (CAR) T-cell therapy for large B-cell lymphoma (LBCL) is well-established. This study, using the Premier PINC AI Healthcare Database, assessed hospital costs and healthcare resource utilization (HRU) between CAR T-cell therapy and autologous hematopoietic cell transplant (AHCT) for 733 LBCL patients from 01/01/2017-04/30/2021 (166 CAR T and 567 AHCT from 37 US hospital systems. CAR T-cell therapy had higher index costs but lower non-pharmacy costs, shorter hospital stays, lower ICU utilization than AHCT. The CAR T-cell cohort also presented fewer preparatory costs and HRU. At a 180-day follow-up, AHCT had lower hospitalization rates and costs. Overall, despite higher index costs, CAR T-cell therapy has lower non-pharmacy costs and HRU during the index procedure and requires less preparation time with lower preparation HRUs and costs than AHCT. This has important implications for resource management and informed decision-making for stakeholders.
Collapse
Affiliation(s)
- Chendi Cui
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | | | - Ning Rosenthal
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | - Sally W Wade
- Wade Outcomes Research and Consulting, Salt Lake City, UT, USA
| | - Laura Curry
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | | | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
9
|
Ghilardi G, Williamson S, Pajarillo R, Paruzzo L, Chen L, Grady C, Doucette A, Nemecek E, Gabrielli G, Barta SK, Svoboda J, Chong EA, Yelton R, Nasta SD, Landsburg DJ, Ugwuanyi O, Chen AI, Schachter L, White G, Ballard HJ, Weber E, Curley C, Porter DL, Garfall AL, Hwang WT, Guerra CE, Maziarz RT, Schuster SJ, Ruella M. CAR T-Cell Immunotherapy in Minority Patients with Lymphoma. NEJM EVIDENCE 2024; 3:EVIDoa2300213. [PMID: 38776868 DOI: 10.1056/evidoa2300213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
BACKGROUND Administration of anti-CD19 chimeric antigen receptor T-cell (CART19) immunotherapy for large B-cell lymphomas (LBCLs), a subset of non-Hodgkin lymphoma (NHL), involves high costs and access to specialized tertiary care centers. We investigated whether minority health populations (MHPs) have equal access to CART19 and whether their outcomes are similar to those of non-MHPs. METHODS We analyzed the prevalence and clinical outcomes of patients treated with commercial CART19 at two geographically and socioeconomically different institutions: the Abramson Cancer Center (ACC, Philadelphia, Pennsylvania) and the Knight Cancer Institute (KCI, Portland, Oregon). RESULTS In the ACC catchment area, 8956 patients were diagnosed with NHL between 2015 and 2019 (latest available data from the state registry), including 17.9% MHPs. In the ACC, between 2018 and 2022 (CART became available in 2018), 1492 patients with LBCL were treated, and 194 received CART19. The proportion of MHPs was 15.7% for the entire LBCL cohort but only 6.7% for the CART19 cohort. During the same time, in the KCI catchment area, 4568 patients were diagnosed with NHL, including 4.2% MHPs. In the KCI, 396 patients with LBCL were treated, and 47 received CART19. The proportion of MHPs was 6.6% for the entire LBCL cohort and 4.2% for the CART19 cohort. The 3-month response, survival, and toxicities after CART19 infusion showed similar results, although the number of patients who were treated was limited. CONCLUSIONS This study shows that the access of MHPs to tertiary centers for LBCL care was preserved but appeared reduced for commercial CART19 immunotherapy. Although clinical outcomes of MHPs seemed similar to those of non-MHPs, the small sample size precludes drawing firm conclusions. Further studies are needed. (Funded by the Laffey McHugh Foundation and others.).
Collapse
Affiliation(s)
- Guido Ghilardi
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Staci Williamson
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Raymone Pajarillo
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Luca Paruzzo
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Linhui Chen
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Connor Grady
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Abigail Doucette
- Division of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia
| | - Eneida Nemecek
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Giulia Gabrielli
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Stefan K Barta
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Jakub Svoboda
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Elise A Chong
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Rebecca Yelton
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Sunita D Nasta
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Daniel J Landsburg
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Ositadimma Ugwuanyi
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Andy I Chen
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Levanto Schachter
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Griffin White
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Hatcher J Ballard
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Elizabeth Weber
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Cara Curley
- Division of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia
| | - David L Porter
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
| | - Alfred L Garfall
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
| | - Wei-Ting Hwang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Carmen E Guerra
- Division of General Medicine, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
| | - Richard T Maziarz
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Stephen J Schuster
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Marco Ruella
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| |
Collapse
|
10
|
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
|
11
|
Kelkar AH, Cliff ERS, Jacobson CA, Abel GA, Dijk SW, Krijkamp EM, Redd R, Zurko JC, Hamadani M, Hunink MGM, Cutler C. Second-Line Chimeric Antigen Receptor T-Cell Therapy in Diffuse Large B-Cell Lymphoma : A Cost-Effectiveness Analysis. Ann Intern Med 2023; 176:1625-1637. [PMID: 38048587 DOI: 10.7326/m22-2276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND First-line treatment of diffuse large B-cell lymphoma (DLBCL) achieves durable remission in approximately 60% of patients. In relapsed or refractory disease, only about 20% achieve durable remission with salvage chemoimmunotherapy and consolidative autologous stem cell transplantation (ASCT). The ZUMA-7 (axicabtagene ciloleucel [axi-cel]) and TRANSFORM (lisocabtagene maraleucel [liso-cel]) trials demonstrated superior event-free survival (and, in ZUMA-7, overall survival) in primary-refractory or early-relapsed (high-risk) DLBCL with chimeric antigen receptor T-cell therapy (CAR-T) compared with salvage chemoimmunotherapy and consolidative ASCT; however, list prices for CAR-T exceed $400 000 per infusion. OBJECTIVE To determine the cost-effectiveness of second-line CAR-T versus salvage chemoimmunotherapy and consolidative ASCT. DESIGN State-transition microsimulation model. DATA SOURCES ZUMA-7, TRANSFORM, other trials, and observational data. TARGET POPULATION "High-risk" patients with DLBCL. TIME HORIZON Lifetime. PERSPECTIVE Health care sector. INTERVENTION Axi-cel or liso-cel versus ASCT. OUTCOME MEASURES Incremental cost-effectiveness ratio (ICER) and incremental net monetary benefit (iNMB) in 2022 U.S. dollars per quality-adjusted life-year (QALY) for a willingness-to-pay (WTP) threshold of $200 000 per QALY. RESULTS OF BASE-CASE ANALYSIS The increase in median overall survival was 4 months for axi-cel and 1 month for liso-cel. For axi-cel, the ICER was $684 225 per QALY and the iNMB was -$107 642. For liso-cel, the ICER was $1 171 909 per QALY and the iNMB was -$102 477. RESULTS OF SENSITIVITY ANALYSIS To be cost-effective with a WTP of $200 000, the cost of CAR-T would have to be reduced to $321 123 for axi-cel and $313 730 for liso-cel. Implementation in high-risk patients would increase U.S. health care spending by approximately $6.8 billion over a 5-year period. LIMITATION Differences in preinfusion bridging therapies precluded cross-trial comparisons. CONCLUSION Neither second-line axi-cel nor liso-cel was cost-effective at a WTP of $200 000 per QALY. Clinical outcomes improved incrementally, but costs of CAR-T must be lowered substantially to enable cost-effectiveness. PRIMARY FUNDING SOURCE No research-specific funding.
Collapse
Affiliation(s)
- Amar H Kelkar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Harvard Medical School, Boston; and Harvard T.H. Chan School of Public Health, Boston, Massachusetts (A.H.K.)
| | - Edward R Scheffer Cliff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Harvard Medical School, Boston; Harvard T.H. Chan School of Public Health, Boston; and Program on Regulation, Therapeutics and Law, Brigham and Women's Hospital, Boston, Massachusetts (E.R.S.C.)
| | - Caron A Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, and Harvard Medical School, Boston, Massachusetts (C.A.J., G.A.A., C.C.)
| | - Gregory A Abel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, and Harvard Medical School, Boston, Massachusetts (C.A.J., G.A.A., C.C.)
| | - Stijntje W Dijk
- Department of Radiology and Nuclear Medicine and Department of Epidemiology and Biostatistics, Erasmus University Medical Center, Rotterdam, the Netherlands (S.W.D.)
| | - Eline M Krijkamp
- Department of Epidemiology and Biostatistics, Erasmus University Medical Center, Rotterdam, and Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands (E.M.K.)
| | - Robert Redd
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts (R.R.)
| | - Joanna C Zurko
- Division of Hematology & Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin (J.C.Z.)
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin (M.H.)
| | - M G Myriam Hunink
- Harvard T.H. Chan School of Public Health, Boston, and Program on Regulation, Therapeutics and Law, Brigham and Women's Hospital, Boston, Massachusetts; and Department of Epidemiology and Biostatistics, Erasmus University Medical Center, Rotterdam, the Netherlands (M.G.M.H.)
| | - Corey Cutler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, and Harvard Medical School, Boston, Massachusetts (C.A.J., G.A.A., C.C.)
| |
Collapse
|
12
|
Russler-Germain DA, Ghobadi A. T-cell redirecting therapies for B-cell non-Hodgkin lymphoma: recent progress and future directions. Front Oncol 2023; 13:1168622. [PMID: 37465110 PMCID: PMC10351267 DOI: 10.3389/fonc.2023.1168622] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/21/2023] [Indexed: 07/20/2023] Open
Abstract
Several key advances in the treatment of B-cell non-Hodgkin lymphoma (B-NHL) over the past two decades have strategically exploited B-cell lineage markers suitable for targeting by immunotherapies. First, the addition of the anti-CD20 monoclonal antibody (mAb) rituximab to a range of standard therapies conferred remarkable outcomes improvements in diverse settings, perhaps most prominently an overall survival advantage in newly diagnosed diffuse large B-cell lymphoma (DLBCL). Subsequently, multiple chimeric antigen receptor (CAR) T-cell therapies targeting CD19 have revolutionized the treatment of relapsed/refractory (rel/ref) DLBCL and are active in other B-NHL subtypes as well. Most recently, the longstanding aspiration to exploit patients' endogenous T-cells to combat lymphoma has been achieved via T-cell redirecting therapies such as bispecific antibodies (BsAbs) that incorporate dual targeting of a T-cell antigen such as CD3 plus a B-cell antigen such as CD19 or CD20 expressed by the tumor. These novel agents have demonstrated impressive activity as monotherapies in patients with heavily pre-treated, rel/ref B-NHL of a variety of subtypes. Now, myriad clinical trials are exploring combinations of T-cell redirectors with targeted therapies, antibody-drug conjugates, conventional chemotherapy, and even new immunotherapies. Here, we highlight key landmarks in the development of T-cell redirecting therapies for the treatment of B-NHL, emerging evidence and lessons from recent clinical trials, and exciting new directions in this arena.
Collapse
Affiliation(s)
- David A. Russler-Germain
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Armin Ghobadi
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
13
|
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
|
14
|
Li YR, Dunn ZS, Yu Y, Li M, Wang P, Yang L. Advancing cell-based cancer immunotherapy through stem cell engineering. Cell Stem Cell 2023; 30:592-610. [PMID: 36948187 PMCID: PMC10164150 DOI: 10.1016/j.stem.2023.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 01/04/2023] [Accepted: 02/22/2023] [Indexed: 03/24/2023]
Abstract
Advances in cell-based therapy, particularly CAR-T cell therapy, have transformed the treatment of hematological malignancies. Although an important step forward for the field, autologous CAR-T therapies are hindered by high costs, manufacturing challenges, and limited efficacy against solid tumors. With ongoing progress in gene editing and culture techniques, engineered stem cells and their application in cell therapy are poised to address some of these challenges. Here, we review stem cell-based immunotherapy approaches, stem cell sources, gene engineering and manufacturing strategies, therapeutic platforms, and clinical trials, as well as challenges and future directions for the field.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zachary Spencer Dunn
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Yanqi Yu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Miao Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA; Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
15
|
Hoffmann MS, Hunter BD, Cobb PW, Varela JC, Munoz J. Overcoming Barriers to Referral for Chimeric Antigen Receptor T-Cell Therapy in Patients With Relapsed/Refractory Diffuse Large B-Cell Lymphoma. Transplant Cell Ther 2023:S2666-6367(23)01234-4. [PMID: 37031747 DOI: 10.1016/j.jtct.2023.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/18/2023] [Accepted: 04/03/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is the most prevalent subtype of non-Hodgkin lymphoma. Although outcomes to frontline therapy are encouraging, patients who are refractory to or relapse after first-line therapy experience inferior outcomes. A significant proportion of patients treated with additional lines of cytotoxic chemotherapy ultimately succumb to their disease as established in the SCHOLAR-1 study. CHIMERIC ANTIGEN RECEPTOR (CAR)-T CELL THERAPY CAR-T cell therapy is a novel approach to cancer management that reprograms a patient's own T cells to better target and eliminate cancer cells. It was initially approved by the US Food and Drug Administration (FDA) for patients with relapsed/refractory (r/r) DLBCL in the third line of treatment. Based on recently published randomized data, CAR-T cell therapy (axicabtagene ciloleucel and lisocabtagene maraleucel) has also been approved in the second line of treatment for patients who are primary refractory or relapse within 12 months of initiation of first-line therapy. Despite the proven efficacy in treating r/r DLBCL with cluster of differentiation (CD)19-directed CAR-T cell therapy, several barriers exist that may prevent eligible patients from receiving treatment. KEY BARRIERS TO CAR-T CELL TREATMENT Barriers to treatment include cost of therapy, patient hesitancy, required travel to academic treatment centers, nonreferrals, lack of understanding of CAR-T cell therapy, lack of caregiver support, knowledge of resources available, and timely patient selection by referring oncologists. CONCLUSION In this review, an overview of the FDA-approved CD19-directed CAR-T cell therapies (tisagenlecleucel, axicabtagene ciloleucel, and lisocabtagene maraleucel) is provided from pivotal clinical trials and supporting real-world evidence from retrospective studies. In both clinical trials and real-world settings CAR-T cell therapy has been shown to be safe and efficacious for treating patients with r/r DLBCL. However, several barriers prevent eligible patients from accessing these therapies. Barriers to referrals for CAR-T cell therapy are presented with recommendations to improve collaboration between community oncologists and physicians from CAR-T cell therapy treatment centers and subsequent long-term care of patients in community treatment centers.
Collapse
Affiliation(s)
- Marc S Hoffmann
- University of Kansas Cancer Center, Division of Hematologic Malignancies and Cellular Therapeutics, Westwood, KS
| | - Bradley D Hunter
- Blood and Marrow Transplantation, LDS Hospital, Intermountain Healthcare, Salt Lake City, UT
| | | | - Juan C Varela
- Blood and Marrow Transplant Program, AdventHealth Hospital, Orlando, FL; Beth Israel Deaconess Medical Center, Dana Farber/Harvard Cancer Center, Boston, MA
| | - Javier Munoz
- Department of Hematology, Mayo Clinic, Phoenix, AZ.
| |
Collapse
|
16
|
Mikhael J, Fowler J, Shah N. Chimeric Antigen Receptor T-Cell Therapies: Barriers and Solutions to Access. JCO Oncol Pract 2022; 18:800-807. [PMID: 36130152 DOI: 10.1200/op.22.00315] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies are relatively new treatments for patients with heavily pretreated hematologic malignancies. Although these innovative therapies can offer substantial benefit to patients with limited alternative treatment options, patient-access barriers exist. Conventional clinical trials are time-consuming and may be limited by strict patient eligibility criteria, resources, and availability of enrollment slots. Because of the complexity of the CAR-T administration process, treatment delivery can be associated with additional burden for the patient, including requiring patients to reside close to treatment centers and remain with a caregiver after infusion. Manufacturing of CAR-T cells is completed in specialized facilities and depends on the availability of reagents, manufacturing workforce, and timely transportation. CAR-T therapy is costly, and many US health plans restrict coverage of cell and gene therapies. Several of the existing challenges because of these barriers have been exacerbated during the COVID-19 pandemic. This review discusses these barriers and proposes some potential solutions to improving patient access, including innovation in clinical trial design and manufacturing, location of treatment delivery, and key stakeholder opinions regarding treatment and reimbursement. We propose a call to action for key stakeholder groups to address these barriers to CAR-T therapy to expand treatment access for patients. Future collaboration between key stakeholders, including payers, regulatory agencies, and industry/academia, will be critical to continue to address these barriers and enhance patient access to these therapies.
Collapse
Affiliation(s)
- Joseph Mikhael
- Translational Genomics Research Institute (TGen), Applied Cancer Research and Drug Discovery Division, Phoenix, AZ
| | | | - Nina Shah
- Bone Marrow Transplantation and Hematologic Malignancy Unit, Division of Hematology-Oncology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
17
|
Ragoonanan D, Bhar S, Mohan G, Beltramo F, Khazal SJ, Hurley C, Andersen C, Margossian S, Neelapu SS, Shpall E, Gutierrez C, Tewari P, Shoberu B, Talleur A, McCall D, Nunez C, Cuglievan B, Tambaro FP, Petropoulos D, Abdel-Azim H, Mahadeo KM. A multicenter study of ICU resource utilization in pediatric, adolescent and young adult patients post CAR-T therapy. Front Oncol 2022; 12:1022901. [PMID: 36353531 PMCID: PMC9638171 DOI: 10.3389/fonc.2022.1022901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/30/2022] [Indexed: 12/03/2022] Open
Abstract
Tisagenlecleucel is associated with remarkable outcomes in treating patients up to the age of 25 years with refractory B-cell acute lymphoblastic leukemia (ALL). Yet, due to unique and potentially life-threatening complications, access remains limited to higher-resource and certified centers. Reports of inequity and related disparities in care are emerging. In this multicenter study of ALL patients admitted for anti-leukemia therapy, who required pediatric intensive care (ICU) support (n = 205), patients receiving tisagenlecleucel (n = 39) were compared to those receiving conventional chemotherapy (n = 166). The median time to ICU transfer was 6 (0–43) versus 1 (0–116) days, respectively (p < 0.0001). There was no difference in the use of vasopressor, ionotropic, sedating, and/or paralytic agents between groups, but use of dexamethasone was higher among tisagenlecleucel patients. Patients receiving tisagenlecleucel were more likely to have cardiorespiratory toxicity (p = 0.0002), but there were no differences in diagnostic interventions between both groups and/or differences in ICU length of stay and/or overall hospital survival. Toxicities associated with tisagenlecleucel are generally reversible, and our findings suggest that resource utilization once admitted to the ICU may be similar among patients with ALL receiving tisagenlecleucel versus conventional chemotherapy. As centers consider improved access to care and the feasibility of tisagenlecleucel certification, our study may inform strategic planning.
Collapse
Affiliation(s)
- Dristhi Ragoonanan
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Dristhi Ragoonanan,
| | - Saleh Bhar
- Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, United States
| | - Gopi Mohan
- Division of Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA, United States
| | - Fernando Beltramo
- Children’s Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Sajad J. Khazal
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Caitlin Hurley
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children’s Research Hospital, Memphis, TN, United States
| | - Clark Andersen
- Department of Biostatistics, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven Margossian
- Division of Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA, United States
| | - Sattva S. Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cristina Gutierrez
- Department of Critical Care, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Priti Tewari
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Basirat Shoberu
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aimee Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children’s Research Hospital, Memphis, TN, United States
| | - David McCall
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cesar Nunez
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Francesco Paolo Tambaro
- Pediatric Stem Cell Transplantation and Cell Therapy Program, UOC SIT-TMO AORN Santobono-Pausilipon, Napoli, Italy
| | - Demetrios Petropoulos
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hisham Abdel-Azim
- Division of Transplant and Cell Therapy, Loma Linda University Cancer Center, Loma Linda, CA, United States
| | - Kris M. Mahadeo
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
18
|
Maakaron JE, Hu M, El Jurdi N. Chimeric antigen receptor T cell therapy for cancer: clinical applications and practical considerations. BRITISH MEDICAL JOURNAL 2022. [DOI: 10.1136/bmj-2021-068956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Abstract
Chimeric antigen receptor T cells have revolutionized the treatment of hematological malignancies during the past five years, boasting impressive response rates and durable remissions for patients who previously had no viable options. In this review, we provide a brief historical overview of their development. We focus on the practical aspects of a patient’s journey through this treatment and the unique toxicities and current best practices to manage those. We then discuss the key registration trials that have led to approvals for the treatment of relapsed/refractory acute lymphoblastic leukemia (ALL), diffuse large B cell lymphoma (DLBCL), follicular lymphoma, mantle cell lymphoma (MCL), and multiple myeloma. Finally, we consider the future development and research directions of this cutting edge therapy.
Collapse
|
19
|
Yang M, Olaoba OT, Zhang C, Kimchi ET, Staveley-O’Carroll KF, Li G. Cancer Immunotherapy and Delivery System: An Update. Pharmaceutics 2022; 14:1630. [PMID: 36015256 PMCID: PMC9413869 DOI: 10.3390/pharmaceutics14081630] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 02/06/2023] Open
Abstract
With an understanding of immunity in the tumor microenvironment, immunotherapy turns out to be a powerful tool in the clinic to treat many cancers. The strategies applied in cancer immunotherapy mainly include blockade of immune checkpoints, adoptive transfer of engineered cells, such as T cells, natural killer cells, and macrophages, cytokine therapy, cancer vaccines, and oncolytic virotherapy. Many factors, such as product price, off-target side effects, immunosuppressive tumor microenvironment, and cancer cell heterogeneity, affect the treatment efficacy of immunotherapies against cancers. In addition, some treatments, such as chimeric antigen receptor (CAR) T cell therapy, are more effective in treating patients with lymphoma, leukemia, and multiple myeloma rather than solid tumors. To improve the efficacy of targeted immunotherapy and reduce off-target effects, delivery systems for immunotherapies have been developed in past decades using tools such as nanoparticles, hydrogel matrix, and implantable scaffolds. This review first summarizes the currently common immunotherapies and their limitations. It then synopsizes the relative delivery systems that can be applied to improve treatment efficacy and minimize side effects. The challenges, frontiers, and prospects for applying these delivery systems in cancer immunotherapy are also discussed. Finally, the application of these approaches in clinical trials is reviewed.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
| | - Olamide Tosin Olaoba
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65212, USA
| | - Eric T. Kimchi
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
20
|
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
|
21
|
Mei M, Hamadani M, Ahn KW, Chen Y, Kharfan-Dabaja MA, Sauter C, Herrera AF. Autologous hematopoietic cell transplantation in diffuse large B-cell lymphoma after three or more lines of prior therapy: evidence of durable benefit. Haematologica 2022; 107:1214-1217. [PMID: 35112554 PMCID: PMC9052914 DOI: 10.3324/haematol.2021.279999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/24/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- Matthew Mei
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| | - Mehdi Hamadani
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI.
| | - Kwang W Ahn
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI; Center for International Blood and Marrow Transplant Research, Milwaukee, WI
| | - Yue Chen
- Center for International Blood and Marrow Transplant Research, Milwaukee, WI
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - Craig Sauter
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| |
Collapse
|
22
|
Ahmed N, Shahzad M, Shippey E, Bansal R, Mushtaq MU, Mahmoudjafari Z, Faisal MS, Hoffmann M, Abdallah AO, Divine C, Hamadani M, McGuirk J, Shune L. Socioeconomic and Racial Disparity in Chimeric antigen receptor T cell (CAR T) Therapy Access. Transplant Cell Ther 2022; 28:358-364. [DOI: 10.1016/j.jtct.2022.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/22/2022] [Accepted: 04/07/2022] [Indexed: 11/26/2022]
|
23
|
Al Hadidi S, Schinke C, Thanendrarajan S, Zangari M, van Rhee F. Enrollment of Black Participants in Pivotal Clinical Trials Supporting US Food and Drug Administration Approval of Chimeric Antigen Receptor-T Cell Therapy for Hematological Malignant Neoplasms. JAMA Netw Open 2022; 5:e228161. [PMID: 35442451 PMCID: PMC9021907 DOI: 10.1001/jamanetworkopen.2022.8161] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPORTANCE Disparities that affect Black persons with various hematological malignant neoplasms are substantial, yet little is known about disparities related to the use of US Food and Drug Administration (FDA)-approved chimeric antigen receptor-T cell (CAR-T) therapy. OBJECTIVE To examine the enrollment of Black participants in clinical trials that resulted in a subsequent FDA approval of CAR-T products in hematological malignant neoplasms. DESIGN, SETTING, AND PARTICIPANTS A cross-sectional study was performed using publicly available data on drug products and demographic subgroups from Drugs@fda in the period of August 2017 to May 2021. Data analysis included patients with large B cell lymphoma, follicular lymphoma, mantle cell lymphoma, acute lymphoblastic leukemia, and multiple myeloma who were enrolled into 7 clinical trials that investigated various CAR-T products. The study was conducted from July 1, 2021, to November 30, 2021. MAIN OUTCOMES AND MEASURES Frequencies of participation of Black participants were calculated with adjustment for disease prevalence. RESULTS Of the 1057 enrolled patients included in the study, CAR-T products were given to 746 patients (71%), and efficacy was reported for 729 enrolled patients (69%) across all the approved CAR-T products and indications. Most patients (1015 patients [96%]) were enrolled in the US. Black participants were included in the racial category other in the study that supported tisagenlecleucel approval in acute lymphoblastic leukemia; otherwise, their enrollment was specified either in the study publication and/or the demographic subgroup information available under the FDA product labeling information. The number of Black participants who received the CAR-T product and had reported efficacy varied between studies (range, 1-12 participants [2%-5%]). Adjusted prevalence measures showed the lowest participation to prevalence ratio of 0.2 for multiple myeloma and 0.6 for large B cell lymphoma. CONCLUSIONS AND RELEVANCE The findings of this study suggest that there are substantial disparities affecting Black patients across all approved CAR-T products used to treat hematological malignant neoplasms with otherwise limited effective treatment options. The study findings might aid policy discussions regarding the immediate need of regulations that enforce certain thresholds of Black patients' enrollment before granting FDA approval.
Collapse
Affiliation(s)
- Samer Al Hadidi
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock
| | - Carolina Schinke
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock
| | - Sharmilan Thanendrarajan
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock
| | - Maurizio Zangari
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock
| | - Frits van Rhee
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock
| |
Collapse
|
24
|
Nath K, James Y, Taylor D, Gardner R, Rai N, Ware RS, Taylor K, Morton J, Durrant S, Irving I, Bashford J. Activity and Outcomes of Autologous Stem Cell Transplantation in the Private Sector in Australia. Intern Med J 2022. [PMID: 35319152 DOI: 10.1111/imj.15754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 03/02/2022] [Accepted: 03/13/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Few Australasian Autologous Stem Cell Transplantation (ASCT) programs perform ASCTs in the private sector. Relatively little is known about ASCT outcomes in the private sector, which varies in care delivery models to the public system. AIMS We investigated transplantation activity and survival outcomes at Icon Cancer Centre's Brisbane-based private clinical and laboratory ASCT program, over a 23-year period. METHODS Retrospective, observational study of all adults who underwent ASCT at Icon between 1996-2018. Main outcome measures were transplant activity, overall survival (OS) and day-100 and 1-year transplant-related mortality (TRM). Outcomes were benchmarked against the Australasian Bone Marrow Transplant Recipient Registry (ABMTRR). RESULTS Between 1996-2018, 1676 ASCTs were performed in 1454 patients. From 2010-2018, ASCTs performed at Icon contributed 40% of all South East Queensland ASCTs. In the last 5-years, 21% of Icon's patients were ≥70-years, compared to 5% across Australasia. For the entire cohort, 100-day, and 1-year TRM was 1.1% and 1.7% respectively, whilst for those aged ≥70-years, it was 2.0% and 3.1%. For ASCTs performed between 2014-2018, 100-day and 1-year TRM was 0.8% and 1.4%, which was half the TRM rates reported by the ABMTRR. The 10-year post-transplant OS at Icon was higher than the ABMTRR data, across all disease subtypes. CONCLUSION Icon is the largest ASCT contributor in Queensland, with excellent OS and low TRM, demonstrating the critical role of the private sector in the administration of this highly complex therapy. The Icon ASCT program is inclusive of patients aged ≥70-years, demonstrating low and acceptable TRM. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Karthik Nath
- Icon Cancer Centre, South Brisbane, QLD, Australia.,Mater Private Hospital, South Brisbane, QLD, Australia
| | - Yvette James
- Icon Institute of Innovation and Research, Brisbane, QLD, Australia
| | - Debra Taylor
- Wesley Cell Therapies Laboratory, Sullivan Nicolaides Pathology, QLD, Australia
| | - Raeina Gardner
- Icon Cancer Centre, South Brisbane, QLD, Australia.,Icon Cancer Centre, Wesley, Brisbane, QLD, Australia
| | - Nicholas Rai
- Menzies Health Institute, Griffith University, Brisbane, QLD, Australia
| | - Robert S Ware
- Menzies Health Institute, Griffith University, Brisbane, QLD, Australia
| | - Kerry Taylor
- Icon Cancer Centre, South Brisbane, QLD, Australia.,Mater Private Hospital, South Brisbane, QLD, Australia
| | - James Morton
- Icon Cancer Centre, South Brisbane, QLD, Australia.,Mater Private Hospital, South Brisbane, QLD, Australia
| | - Simon Durrant
- Icon Institute of Innovation and Research, Brisbane, QLD, Australia.,Icon Cancer Centre, Wesley, Brisbane, QLD, Australia.,The Wesley Hospital, Brisbane, QLD, Australia
| | - Ian Irving
- Icon Cancer Centre, Wesley, Brisbane, QLD, Australia.,The Wesley Hospital, Brisbane, QLD, Australia
| | - John Bashford
- Icon Institute of Innovation and Research, Brisbane, QLD, Australia
| |
Collapse
|
25
|
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: 167] [Impact Index Per Article: 55.7] [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
|
26
|
Santomasso BD, Nastoupil LJ, Adkins S, Lacchetti C, Schneider BJ, Anadkat M, Atkins MB, Brassil KJ, Caterino JM, Chau I, Davies MJ, Ernstoff MS, Fecher L, Funchain P, Jaiyesimi I, Mammen JS, Naidoo J, Naing A, Phillips T, Porter LD, Reichner CA, Seigel C, Song JM, Spira A, Suarez-Almazor M, Swami U, Thompson JA, Vikas P, Wang Y, Weber JS, Bollin K, Ghosh M. Management of Immune-Related Adverse Events in Patients Treated With Chimeric Antigen Receptor T-Cell Therapy: ASCO Guideline. J Clin Oncol 2021; 39:3978-3992. [PMID: 34724386 DOI: 10.1200/jco.21.01992] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To increase awareness, outline strategies, and offer guidance on the recommended management of immune-related adverse events (irAEs) in patients treated with chimeric antigen receptor (CAR) T-cell therapy. METHODS A multidisciplinary panel of medical oncology, neurology, hematology, emergency medicine, nursing, trialists, and advocacy experts was convened to develop the guideline. Guideline development involved a systematic literature review and an informal consensus process. The systematic review focused on evidence published from 2017 to 2021. RESULTS The systematic review identified 35 eligible publications. Because of the paucity of high-quality evidence, recommendations are based on expert consensus. RECOMMENDATIONS The multidisciplinary team issued recommendations to aid in the recognition, workup, evaluation, and management of the most common CAR T-cell-related toxicities, including cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, B-cell aplasia, cytopenias, and infections. Management of short-term toxicities associated with CAR T cells begins with supportive care for most patients, but may require pharmacologic interventions for those without adequate response. Management of patients with prolonged or severe CAR T-cell-associated cytokine release syndrome includes treatment with tocilizumab with or without a corticosteroid. On the basis of the potential for rapid decline, patients with moderate to severe immune effector cell-associated neurotoxicity syndrome should be managed with corticosteroids and supportive care.Additional information is available at www.asco.org/supportive-care-guidelines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ian Chau
- Royal Marsden Hospital and Institute of Cancer Research, London and Surrey, United Kingdom
| | | | | | | | | | | | | | - Jarushka Naidoo
- Beaumont Hospital, Dublin, Ireland and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | | | | | | | | | | | | | | | | | - Umang Swami
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - John A Thompson
- Seattle Cancer Care Alliance, University of Washington/Fred Hutchinson, Seattle, WA
| | | | | | | | | | | |
Collapse
|
27
|
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
|
28
|
Young CM, Quinn C, Trusheim MR. Durable cell and gene therapy potential patient and financial impact: US projections of product approvals, patients treated, and product revenues. Drug Discov Today 2021; 27:17-30. [PMID: 34537333 DOI: 10.1016/j.drudis.2021.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022]
Abstract
Durable cell and gene therapies potentially transform patient lives, but payers fear unsustainable costs arising from the more than 1000 therapies in the development pipeline. A novel multi-module Markov chain Monte Carlo-based model projects product-indication approvals, treated patients, and product revenues. We estimate a mean 63.5 (54-74 5th to 95th percentile range) cumulative US product-indication approvals through 2030, with a mean 93000 patients treated in 2030 generating a mean US$24.4 billion (US$17.0B-35.0B, US$73.0B extreme) list price product revenues not including ancillary medical costs or cost offsets. Thus, the likely dozens of durable cell and gene therapies developed through 2030 are unlikely to threaten US health system financial sustainability.
Collapse
Affiliation(s)
- Colin M Young
- MIT NEWDIGS, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, USA
| | - Casey Quinn
- MIT NEWDIGS, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, USA
| | - Mark R Trusheim
- MIT NEWDIGS, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, USA
| |
Collapse
|
29
|
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
|
30
|
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
|