1
|
Dandoy CE, Adams J, Artz A, Bredeson C, Dahi PB, Dodd T, Jaglowski S, Lehmann L, LeMaistre CF, Mian A, Neal A, Page K, Rizzo JD, Rotz S, Sorror M, Steinberg A, Viswabandya A, Howard DS. In Pursuit of Optimal Outcomes: A Framework for Quality Standards in Immune Effector Cell Therapy. Transplant Cell Ther 2024; 30:942-954. [PMID: 39067790 DOI: 10.1016/j.jtct.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Immune effector cell (IEC) therapy represents a transformative advancement in oncology, leveraging the immune system to combat various malignancies. This article outlines a comprehensive framework for establishing and maintaining quality standards in IEC therapy amidst rapid scientific and clinical advancements. We emphasize the integration of structured process measures, robust quality assurance, and meticulous outcome evaluation to ensure treatment efficacy and safety. Key components include multidisciplinary expertise, stringent accreditation protocols, and advanced data management systems, which facilitate standardized reporting and continual innovation. The collaborative effort among stakeholders-ranging from patients and healthcare providers to regulatory bodies-is crucial in delivering high-quality IEC therapies. This framework aims to enhance patient outcomes and cement the role of IEC therapy as a cornerstone of modern oncology, promoting continuous improvement and adherence to high standards across the therapeutic spectrum.
Collapse
Affiliation(s)
- Christopher E Dandoy
- Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio.
| | - Joan Adams
- Stephenson Cancer Center, OU Health Science Center The University of Oklahoma, Oklahoma City, Oklahoma
| | - Andrew Artz
- Division of Leukemia, Department of Hematology and HCT, City of Hope, Duarte, California
| | - Christopher Bredeson
- Ottawa Hospital Research Institute, Division of Hematology, University of Ottawa, Ottawa, Canada
| | - Parastoo B Dahi
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Therese Dodd
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, Tennessee
| | - Samantha Jaglowski
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Leslie Lehmann
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Amir Mian
- Department of Pediatric Hematology and Oncology, Department of Pediatrics at Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Alison Neal
- Department of Bone Marrow Transplant and Cellular Therapy, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Kristen Page
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - J Douglas Rizzo
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Seth Rotz
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio
| | - Mohamed Sorror
- Fred Hutchinson Cancer Center and University of Washington, Seattle, Washington
| | - Amir Steinberg
- Adult Stem Cell Transplantation, Westchester Medical Center, New York Medical College, Valhalla, New York
| | - Auro Viswabandya
- Department of Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Dianna S Howard
- Department of Internal Medicine, Section of Hematology and Oncology, Stem Cell Transplant and Cellular Therapy Program, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, North Carolina
| |
Collapse
|
2
|
Voorhees TJ, Bezerra E, Denlinger N, Jaglowski S, de Lima M. SOHO State of the Art Updates and Next Questions Updates on Building Your CAR-T Cell Program. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:649-652. [PMID: 38643029 DOI: 10.1016/j.clml.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 04/22/2024]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has significantly impacted treatment algorithms and clinical outcomes for a variety of patients with hematologic malignancies over the past decade. The field of cellular immunotherapy is currently experiencing a rapid expansion of the number of patients eligible for CAR-T therapies as approvals are being seen in earlier lines of therapy. With the expanded patients eligible for these therapies, more treatment centers will be necessary to keep up with demand. Building a cellular therapy program can be a daunting task, and therefore, we present our experience with building a clinical cellular therapy program.
Collapse
Affiliation(s)
- Timothy J Voorhees
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH.
| | - Evandro Bezerra
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | - Nathan Denlinger
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | - Samantha Jaglowski
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | - Marcos de Lima
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
3
|
Ailawadhi S, Shune L, Wong SW, Lin Y, Patel K, Jagannath S. Optimizing the CAR T-Cell Therapy Experience in Multiple Myeloma: Clinical Pearls From an Expert Roundtable. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:e217-e225. [PMID: 38369437 DOI: 10.1016/j.clml.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/20/2024]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies offer substantial advancement in the treatment of multiple myeloma (MM). However, the CAR-T therapy process involves complex decision-making that is informed by many variables. This review aims to provide an overview of the patient selection and administration process for CAR-T therapy for MM from the perspective of experienced healthcare providers (HCPs), including considerations for each step in the CAR-T therapy process. Referring HCPs should initiate conversations with HCPs at CAR-T capable centers earlier in the treatment journey, even before patients are eligible for CAR-T therapy, particularly for patients from underserved populations and patients with high-risk disease, to ensure adequate time for logistical planning and patient education. Patient selection for CAR-T therapy may be guided by factors such as performance status, rate of disease progression, and logistical considerations. Some anticancer therapies may affect T-cell fitness and therefore impact CAR-T manufacturing and patient outcomes; however, additional research is needed to confirm this in MM. Bridging therapies should be tailored to the needs of the patient and ideally halted 1 week or longer before CAR-T infusion, contingent upon the agent(s) used. Lymphodepletion regimens may need to be modified for patients with renal insufficiency. Collaboration with HCPs at both the treating and referring centers is important to optimize coordinated care of patients. Collaboration with and guidance from experienced HCPs throughout patient selection, referral, and CAR-T administration is instrumental in optimizing patient outcomes as access to CAR-T therapies expands.
Collapse
Affiliation(s)
| | - Leyla Shune
- Division of Hematologic Malignancy and Cellular Therapeutics, University of Kansas Cancer Center, Westwood, Kansas
| | - Sandy W Wong
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Krina Patel
- Department of Lymphoma - Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
4
|
Curran KJ, Nikiforow S, Bachier C, Hsu YM, Maloney D, Maus MV, McCarthy P, Porter D, Shi P, Shpall EJ, William B, Wacker K, Warkentin P, Heslop HE. A robust quality infrastructure is key to safe and effective delivery of immune effector cells: how FACT-finding can help. Blood Adv 2024; 8:1053-1061. [PMID: 37467016 PMCID: PMC10920101 DOI: 10.1182/bloodadvances.2023010401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023] Open
Abstract
ABSTRACT Immune effector cells (IECs) include a broad range of immune cells capable of modulating several disease states, including malignant and nonmalignant conditions. The growth in the use of IECs as both investigational and commercially available products requires medical institutions to develop workflows/processes to safely implement and deliver transformative therapy. Adding to the complexity of this therapy are the variety of targets, diseases, sources, and unique toxicities that a patient experiences following IEC therapy. For over 25 years, the Foundation for the Accreditation of Cellular Therapy (FACT) has established a standard for the use of cellular therapy, initially with hematopoietic cell transplantation (HCT), and more recently, with the development of standards to encompass IEC products such as chimeric antigen receptor (CAR)-T cells. To date, IEC therapy has challenged the bandwidth and infrastructure of the institutions offering this therapy. To address these challenges, FACT has established a programmatic framework to improve the delivery of IEC therapy. In this study, we outline the current state of IEC program development, accreditation, and solutions to the challenges that programs face as they expand their application to novel IEC therapy.
Collapse
Affiliation(s)
- Kevin J. Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sarah Nikiforow
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Carlos Bachier
- Sarah Cannon Transplant and Cellular Therapy Program, St. David's Austin Medical Center, Austin, TX
| | - Yen-Michael Hsu
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA
| | - David Maloney
- Division of Hematology and Oncology Fred Hutchinson Cancer Research Center, Seattle WA
| | - Marcela V. Maus
- Cellular Immunotherapy Program Massachusetts General Hospital, Boston MA
| | - Philip McCarthy
- Department of Medicine, Transplant and Cellular Therapy Program Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - David Porter
- Division of Hematology/Oncology, Department of Medicine and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Patricia Shi
- New York Blood Center Clinical Apheresis and Cellular Therapy Laboratory, New York, NY
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Basem William
- OhioHealth Blood and Marrow Transplant Program, OhioHealth, Columbus, OH
| | - Kara Wacker
- Foundation for the Accreditation of Cellular Therapy, Omaha, NE
| | - Phyllis Warkentin
- Foundation for the Accreditation of Cellular Therapy, Omaha, NE
- Pathology/Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Helen E. Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX
| |
Collapse
|
5
|
Odstrcil MS, Lee CJ, Sobieski C, Weisdorf D, Couriel D. Access to CAR T-cell therapy: Focus on diversity, equity and inclusion. Blood Rev 2024; 63:101136. [PMID: 37863793 DOI: 10.1016/j.blre.2023.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023]
Abstract
Chimeric antigen receptor T-cell (CAR T-cell) therapy has revolutionized the treatment of hematologic malignancies in patients with relapsed or refractory disease without other treatment options. However, only a very small proportion of patients with an indication for CAR T-cell can access the treatment. The imbalance between supply and demand is magnified in minority and vulnerable populations. Limited access is multifactorial and in part a result of factors directly related to the cellular product such as cost, complex logistics and manufacturing limitations. On the other hand, the impact of diversity, equity, and inclusion (DEI) and their social and structural context are also key to understanding access barriers in cellular therapy and health care in general. CAR T-cell therapy provides us with a new opportunity to better understand and prioritize this gap, a key step towards proactively and strategically addressing access. The aim of this review is to provide an analysis of the current state of access to CAR T therapy with a focus on the influence of DEI. We will cover aspects related to the cellular product and the inseparable context of social and structural determinants. Identifying and addressing barriers is necessary to ensure equitable access to this and all future novel therapies.
Collapse
Affiliation(s)
- Maria S Odstrcil
- Huntsman Cancer Institute, University of Utah, Division of Hematology and Hematologic Malignancies, Salt Lake City, UT, USA
| | - Catherine J Lee
- Huntsman Cancer Institute, University of Utah, Division of Hematology and Hematologic Malignancies, Salt Lake City, UT, USA; Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA
| | - Catherine Sobieski
- Huntsman Cancer Institute, University of Utah, Division of Hematology and Hematologic Malignancies, Salt Lake City, UT, USA
| | - Daniel Weisdorf
- University of Minnesota Medical School, Division of Hematology, Oncology and Transplantation, Minneapolis, MN, USA
| | - Daniel Couriel
- Huntsman Cancer Institute, University of Utah, Division of Hematology and Hematologic Malignancies, Salt Lake City, UT, USA.
| |
Collapse
|
6
|
Nze C. Barriers to accessing cellular therapy for patients receiving care in community practices. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:382-385. [PMID: 38066850 PMCID: PMC10727008 DOI: 10.1182/hematology.2023000518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Affiliation(s)
- Chijioke Nze
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
7
|
Elsallab M, Maus MV. Expanding access to CAR T cell therapies through local manufacturing. Nat Biotechnol 2023; 41:1698-1708. [PMID: 37884746 DOI: 10.1038/s41587-023-01981-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/05/2023] [Indexed: 10/28/2023]
Abstract
Chimeric antigen receptor (CAR) T cells are changing the therapeutic landscape for hematological malignancies. To date, all six CAR T cell products approved by the US Food and Drug Administration (FDA) are autologous and centrally manufactured. As the numbers of approved products and indications continue to grow, new strategies to increase cell-manufacturing capacity are urgently needed to ensure patient access. Distributed manufacturing at the point of care or at other local manufacturing sites would go a long way toward meeting the rising demand. To ensure successful implementation, it is imperative to harness novel technologies to achieve uniform product quality across geographically dispersed facilities. This includes the use of automated cell-production systems, in-line sensors and process simulation for enhanced quality control and efficient supply chain management. A comprehensive effort to understand the critical quality attributes of CAR T cells would enable better definition of widely attainable release criteria. To supplement oversight by national regulatory agencies, we recommend expansion of the role of accreditation bodies. Moreover, regulatory standards may need to be amended to accommodate the unique characteristics of distributed manufacturing models.
Collapse
Affiliation(s)
- Magdi Elsallab
- Harvard-MIT Center for Regulatory Science, Harvard Medical School, Boston, MA, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
8
|
Mathews D, Abernethy A, Chaikof E, Charo RA, Daley GQ, Enriquez J, Gottlieb S, Kahn J, Klausner RD, Tavazoie S, Fabi R, Offodile Ii AC, Sherkow JS, Sullenger RD, Freiling E, Balatbat C. Regenerative Medicine: Case Study for Understanding and Anticipating Emerging Science and Technology. NAM Perspect 2023; 2023:202311d. [PMID: 38855738 PMCID: PMC11157685 DOI: 10.31478/202311d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
|
9
|
Cappuzzello E, Vigolo E, D’Accardio G, Astori G, Rosato A, Sommaggio R. How can Cytokine-induced killer cells overcome CAR-T cell limits. Front Immunol 2023; 14:1229540. [PMID: 37675107 PMCID: PMC10477668 DOI: 10.3389/fimmu.2023.1229540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
The successful treatment of patients affected by B-cell malignancies with Chimeric Antigen Receptor (CAR)-T cells represented a breakthrough in the field of adoptive cell therapy (ACT). However, CAR-T therapy is not an option for every patient, and several needs remain unmet. In particular, the production of CAR-T cells is expensive, labor-intensive and logistically challenging; additionally, the toxicities deriving from CAR-T cells infusion, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), have been documented extensively. Alternative cellular therapy products such as Cytokine-induced killer (CIK) cells have the potential to overcome some of these obstacles. CIK cells are a heterogeneous population of polyclonal CD3+CD56+ T cells with phenotypic and functional properties of NK cells. CIK cell cytotoxicity is exerted in a major histocompatibility complex (MHC)-unrestricted manner through the engagement of natural killer group 2 member D (NKG2D) molecules, against a wide range of hematological and solid tumors without the need for prior antigen exposure or priming. The foremost potential of CIK cells lies in the very limited ability to induce graft-versus-host disease (GvHD) reactions in the allogeneic setting. CIK cells are produced with a simple and extremely efficient expansion protocol, which leads to a massive expansion of effector cells and requires a lower financial commitment compared to CAR-T cells. Indeed, CAR-T manufacturing involves the engineering with expensive GMP-grade viral vectors in centralized manufacturing facilities, whereas CIK cell production is successfully performed in local academic GMP facilities, and CIK cell treatment is now licensed in many countries. Moreover, the toxicities observed for CAR-T cells are not present in CIK cell-treated patients, thus further reducing the costs associated with hospitalization and post-infusion monitoring of patients, and ultimately encouraging the delivery of cell therapies in the outpatient setting. This review aims to give an overview of the limitations of CAR-T cell therapy and outline how the use of CIK cells could overcome such drawbacks thanks to their unique features. We highlight the undeniable advantages of using CIK cells as a therapeutic product, underlying the opportunity for further research on the topic.
Collapse
Affiliation(s)
- Elisa Cappuzzello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Emilia Vigolo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Giulia D’Accardio
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Hematology, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Roberta Sommaggio
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Perales MA, Anderson LD, Jain T, Kenderian SS, Oluwole OO, Shah GL, Svoboda J, Hamadani M. Role of CD19 Chimeric Antigen Receptor T Cells in Second-Line Large B Cell Lymphoma: Lessons from Phase 3 Trials. An Expert Panel Opinion from the American Society for Transplantation and Cellular Therapy. Transplant Cell Ther 2022; 28:546-559. [PMID: 35768052 PMCID: PMC9427727 DOI: 10.1016/j.jtct.2022.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/25/2022]
Abstract
Since 2017, 3 CD19-directed chimeric antigen receptor (CAR) T cell therapies-axicabtagene ciloleucel, tisagenlecleucel, and lisocabtagene maraleucel-have been approved for relapsed/refractory aggressive diffuse large B cell lymphoma after 2 lines of therapy. Recently, 3 prospective phase 3 randomized clinical trials were conducted to define the optimal second-line treatment by comparing each of the CAR T cell products to the current standard of care: ZUMA-7 for axicabtagene ciloleucel, BELINDA for tisagenlecleucel, and TRANSFORM for lisocabtagene maraleucel. These 3 studies, although largely addressing the same question, had different outcomes, with ZUMA-7 and TRANSFORM demonstrating significant improvement with CD19 CAR T cells in second-line therapy compared with standard of care but BELINDA not showing any benefit. The US Food and Drug Administration has now approved axicabtagene ciloleucel and lisocabtagene maraleucel for LBCL that is refractory to first-line chemoimmunotherapy or relapse occurring within 12 months of first-line chemoimmunotherapy. Following the reporting of these practice changing studies, here a group of experts convened by the American Society for Transplantation and Cellular Therapy provides a comprehensive review of the 3 studies, emphasizing potential differences, and shares perspectives on what these results mean to clinical practice in this new era of treatment of B cell lymphomas.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Larry D Anderson
- Hematologic Malignancies, Transplantation, and Cellular Therapy Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Tania Jain
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Mayo Clinic Graduate School of Biomedical Sciences, Division of Hematology, Department of Immunology and Department of Molecular Medicine, Rochester, Minnesota
| | - Olalekan O Oluwole
- Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Gunjan L Shah
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Jakub Svoboda
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
11
|
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
|
12
|
Vladimira R, Ines B. Role of flow cytometry in evaluation of the cellular therapy products used in haematopoietic stem cell transplantation. Int J Lab Hematol 2022; 44:446-453. [PMID: 35419954 DOI: 10.1111/ijlh.13849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 11/26/2022]
Abstract
Cellular therapy nowadays includes various products from haematopoietic stem cells (HSC) collected from bone marrow, peripheral blood, and umbilical cord blood to more complex adoptive immune therapy for the treatment of malignant diseases, and gene therapy for inherited immune deficiencies. Broader utilization of cellular therapy requires extensive quality testing of these products that should fulfil the same requirements regarding composition, purity, and potency nevertheless they are manufactured in various centres. Technical improvements of the flow cytometers accompanied by the increased number of available reagents and fluorochromes used to conjugate monoclonal antibodies, enable detailed and precise insight into the function of the immune system and other areas of cell biology, and allows cell evaluation based on size, shape, and morphology or assessment of cell surface markers, as well as cell purity and viability, which greatly contributes to the development and progress of the cell therapy. The aim of this paper is to give an overview of the current use and challenges of flow cytometry analysis in quality assessment of cellular therapy products, with regard to basic principles of determining HSC and leukocyte subpopulation, assessment of cells viability and quality of thawed cryopreserved HSC as well as the importance of validation and quality control of flow cytometry methods according to good laboratory practice.
Collapse
Affiliation(s)
- Rimac Vladimira
- Clinical Department of Transfusion Medicine and Transplantation Biology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Bojanić Ines
- Clinical Department of Transfusion Medicine and Transplantation Biology, University Hospital Centre Zagreb, Zagreb, Croatia
| |
Collapse
|
13
|
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: 2.5] [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]
|
14
|
Gatwood KS, Dholaria BR, Lucena M, Baer B, Savani BN, Oluwole OO. Chimeric antigen receptor T-cell therapy: Challenges and framework of outpatient administration. EJHAEM 2022; 3:54-60. [PMID: 35844300 PMCID: PMC9176074 DOI: 10.1002/jha2.333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 10/11/2021] [Indexed: 05/14/2023]
Abstract
Adoptive cellular therapy has made a landmark change within the treatment paradigm of several hematologic malignancies, and novel cellular therapy products, such as chimeric antigen receptor T-cell therapy (CART), have demonstrated impressive efficacy and produced durable responses. However, the CART treatment process is associated with significant toxicities, healthcare resource utilization, and financial burden. Most of these therapies have been administered in the inpatient setting due to their toxicity profile. Improved toxicity management strategies and a better understanding of cellular therapy processes are now established. Therefore, efforts to transition CART to the outpatient setting are warranted with the potential to translate into enhanced patient quality of life and cost savings. A successful launch of outpatient CART requires several components including a multidisciplinary cellular therapy team and an outpatient center with appropriate clinical space and personnel. Telemedicine should be incorporated for closer monitoring. Additionally, clear criteria for admission upon clinical decompensation, a pathway for prompt inpatient transition, and clear toxicity management guidelines should be implemented. Effective education about cellular therapy and toxicity management is imperative, especially for the Emergency Department and Intensive Care Unit teams. Here, we have outlined the various logistical and clinical considerations required for the care of CART patients, which will aid centers to establish an outpatient CART program.
Collapse
Affiliation(s)
- Katie S. Gatwood
- Department of PharmacyVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Bhagirathbhai R. Dholaria
- Division of Hematology‐OncologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | - Brittney Baer
- Department of NursingClinical Trials OfficeVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Bipin N. Savani
- Division of Hematology‐OncologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Division of Hematology‐OncologyDepartment of MedicineTennessee Valley Healthcare SystemNashvilleTennesseeUSA
| | - Olalekan O. Oluwole
- Division of Hematology‐OncologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
15
|
Serra López-Matencio JM, Gómez Garcia de Soria V, Gómez M, Alañón-Plaza E, Muñoz-Calleja C, Castañeda S. Monitoring and safety of CAR-T therapy in clinical practice. Expert Opin Drug Saf 2021; 21:363-371. [PMID: 34519234 DOI: 10.1080/14740338.2021.1979958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION In the last few years, a new T cell therapy, chimeric antigen receptor-T (CAR-T) cells, has been developed. CAR-T cells are highly effective at inhibiting antitumor activity, but they can cause a wide spectrum of unusual side effects. AREAS COVERED The present review provides an overview of the adverse events of CAR-T cell therapy, focusing on cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, increased risk of infections, and other long-term complications. Representative studies addressing the safety and efficacy of CAR-T cell therapy are summarized. EXPERT OPINION In the coming years, we predict a great expansion in the use of CAR-T cell therapy with it applied to a higher number of patients with both malignant neoplasms and immune-mediated diseases. Despite physicians and patient expectations about the potential of this therapy, there are still several barriers that may limit providers' ability to supply quality care. This exciting and powerful new therapy requires the formation of new multidisciplinary teams to carry out a safe treatment administration and to successfully manage the resultant complications. The follow-up of these therapies is important for two aspects: effectiveness in different populations and real-life safety in short and in long-term follow-up.
Collapse
Affiliation(s)
| | | | - Manuel Gómez
- Methodology Unit, Health Research Institute Princesa (IIS-IP), Madrid, Spain
| | - Estefanía Alañón-Plaza
- Hospital Pharmacy Department, Hospital Universitario de La Princesa, IIS-P, Madrid, Spain
| | | | - Santos Castañeda
- Rheumatology Division, Hospital Universitario de La Princesa, IIS-IP, Madrid, Spain.,Catedra UAM-Roche, EPID-Future, Medicine Department, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| |
Collapse
|
16
|
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: 23] [Impact Index Per Article: 7.7] [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
|
17
|
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell therapy is a highly effective new treatment for relapsed and refractory hematological cancers but is associated with the novel treatment-limiting toxicities of cytokine release syndrome and neurotoxicity. Neurotoxicity, now more commonly referred to as immune effector cell-associated neurotoxicity syndrome (ICANS), is a clinical and neuropsychiatric syndrome that can occur in the days to weeks following CAR T-cell and other T-cell-engaging therapies. While the clinical characteristics of ICANS have been well described, its pathophysiology is poorly understood, and best treatment and preventive strategies remain elusive. Clinical trial experience and animal models suggest a central role for endothelial cell dysfunction, myeloid cells, blood-brain barrier disruption, and elevated central nervous system cytokine levels in the development of ICANS. Here we discuss ICANS incidence, clinical features, risk factors, biomarkers, pathophysiology, and grading and management.
Collapse
|
18
|
Marzal-Alfaro MB, Escudero-Vilaplana V, Revuelta-Herrero JL, Collado-Borrell R, Herranz-Alonso A, Sanjurjo-Saez M. Chimeric Antigen Receptor T Cell Therapy Management and Safety: A Practical Tool From a Multidisciplinary Team Perspective. Front Oncol 2021; 11:636068. [PMID: 33777790 PMCID: PMC7992774 DOI: 10.3389/fonc.2021.636068] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 11/17/2022] Open
Abstract
Purpose The use process for chimeric antigen receptor T (CAR-T) cell drugs is complex and has been associated with a number of potentially severe complications, which requires management by a multidisciplinary team. Pharmacists are a key element in the team and have roles and responsibilities. Our objective was to develop a structured and practical guide that supports hospital pharmacist responsibilities and defines specific activities in a CAR-T cell therapy program, specifically in Europe. Methods A literature review was performed, and the recommendations related to pharmacy practice in CAR-T therapy programs were analyzed. A multidisciplinary team was assembled, and meetings were held to address the key tasks in the CAR-T cells’ management process and to create the guide, based on national and international recommendations and in expert’s opinions. Results The multidisciplinary team defined the following key tasks and issued recommendations to improve patient safety, treatment efficacy, and quality: patient selection and evaluation, CAR-T cell drug order to manufacturer, apheresis and material shipment, reception of CAR-T cell drug and storing, CAR-T cell drug prescription and pharmacy verification, CAR-T cell drug thawing and dispensing, CAR-T cell drug administration, patient education, pharmacovigilance and monitoring and outcomes’ record and evaluation. In each task the pharmacist’s role and how it can improve patient care are defined. A checklist was created to guarantee the compliance of standard operating procedures approved in the institution to manage CAR-T cell therapy and as a tool to collect required data for outcomes’ record and evaluation. Conclusion This article provides a consensus set of safety recommendations regarding CAR-T therapy management in clinical practice, easily implementable by other institutions in the European setting. The guide identifies key steps where the involvement of hospital pharmacists would improve the safety and quality of the process and is a support guide to standardize hospital pharmacists’ responsibilities within the multidisciplinary team.
Collapse
Affiliation(s)
- María Belen Marzal-Alfaro
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Vicente Escudero-Vilaplana
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Jose Luis Revuelta-Herrero
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Roberto Collado-Borrell
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ana Herranz-Alonso
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Maria Sanjurjo-Saez
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
19
|
Komanduri KV. Chimeric Antigen Receptor T-Cell Therapy in the Management of Relapsed Non-Hodgkin Lymphoma. J Clin Oncol 2021; 39:476-486. [DOI: 10.1200/jco.20.01749] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
20
|
Scaradavou A. Cord blood beyond transplantation: can we use the experience to advance all cell therapies? Br J Haematol 2021; 194:14-27. [PMID: 33529385 DOI: 10.1111/bjh.17297] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
Unrelated cord blood (CB) units, already manufactured, fully tested and stored, are high-quality products for haematopoietic stem cell transplantation and cell therapies, as well as an optimal starting material for cell expansion, cell engineering or cell re-programming technologies. CB banks have been pioneers in the development and implementation of Current Good Manufacturing Practices for cell-therapy products. Sharing their technological and regulatory experience will help advance all cell therapies, CB-derived or not, particularly as they transition from autologous, individually manufactured products to stored, 'off-the shelf' treatments. Such strategies will allow broader patient access and wide product utilisation.
Collapse
Affiliation(s)
- Andromachi Scaradavou
- Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering (MSK) Kids, MSK Cancer Center, New York, NY, USA
| |
Collapse
|
21
|
Greenbaum U, Yalniz FF, Srour SA, Rezvani K, Singh H, Olson A, Blumenschein G, Hong DS, Shpall EJ, Kebriaei P. Chimeric Antigen Receptor Therapy: How Are We Driving in Solid Tumors? Biol Blood Marrow Transplant 2020; 26:1759-1769. [PMID: 32623078 PMCID: PMC11409837 DOI: 10.1016/j.bbmt.2020.06.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 12/21/2022]
Abstract
Immune effector cell (IEC) therapy is emerging as a promising approach in the field of cancer immunotherapy. Clinical IEC trials, predominantly using chimeric antigen receptor (CAR) T cells, have shown excellent responses in CD19+ B cell malignancies and multiple myeloma. In solid tumors, preclinical data are encouraging, but clinical data are in their infancy, and there are challenges in using CAR T therapy in this setting, including (1) on-target off-tumor toxicity, (2) optimal target identification, (3) effective trafficking into bulky tumor tissue, and (4) resistance to tumor immune evasion mechanisms. Novel techniques and modifications are being explored in both the preclinical and clinical settings, aiming to improve treatment efficacy and address the aforementioned obstacles to successful CAR T therapy in solid tumors. Here we review these challenges in a clinically oriented approach and summarize published clinical trials using CAR T therapy in a variety of solid tumors.
Collapse
Affiliation(s)
- Uri Greenbaum
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fevzi F Yalniz
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samer A Srour
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Harjeet Singh
- Department of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amanda Olson
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George Blumenschein
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S Hong
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
22
|
Mfarrej B, Gaude J, Couquiaud J, Calmels B, Chabannon C, Lemarie C. Validation of a flow cytometry-based method to quantify viable lymphocyte subtypes in fresh and cryopreserved hematopoietic cellular products. Cytotherapy 2020; 23:77-87. [PMID: 32718876 DOI: 10.1016/j.jcyt.2020.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 06/22/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AIMS Adoptive cellular therapy with immune effector cells (IECs) has shown promising efficacy against some neoplastic diseases as well as potential in immune regulation. Both inherent variability in starting material and variations in cell composition produced by the manufacturing process must be thoroughly evaluated with a validated method established to quantify viable lymphocyte subtypes. Currently, commercialized immunophenotyping methods determine cell viability with significant errors in thawed products since they do not include any viability staining. We hereby report on the validation of a flow cytometry-based method for quantifying viable lymphocyte immunophenotypes in fresh and cryopreserved hematopoietic cellular products. METHODS Using fresh or frozen cellular products and stabilized blood, we report on the validation parameters accuracy, uncertainty, precision, sensitivity, robustness and contamination between samples for quantification of viable CD3+, CD4+ T cells, CD8+ T cells, CD3-CD56+CD16+/- NK cells, CD19+ B cells and CD14+ monocytes of relevance to fresh and cryopreserved hematopoietic cellular products using the Cytomics FC500 cytometer (Beckman Coulter). RESULTS The acceptance criteria set in the validation plan were all met. The method is able to accommodate the variability in absolute numbers of cells in starting materials collected or cryopreserved from patients or healthy donors (uncertainty of ≤20% at three different concentrations), stability over time (compliance over 3 years during regular inter-laboratory comparisons) and confidence in meaningful changes during cell processing and manufacturing (intra-assay and intermediate precision of 10% coefficient of variation). Furthermore, the method can accurately report on the efficacy of cell depletion since the lower limit of quantification was established (CD3+, CD4+ and CD8+ cells at 9, 8 and 8 cells/µL, respectively). The method complies with Foundation for the Accreditation of Cellular Therapy (FACT) standards for IEC, FACT-Joint Accreditation Committee of ISCT-EBMT (JACIE) hematopoietic cell therapy standards, International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use Q2(R1) and International Organization for Standardization 15189 standards. Furthermore, it complies with Ligand Binding Assay Bioanalytical Focus Group/American Association of Pharmaceutical Scientists, International Council for Standardization of Hematology/International Clinical Cytometry Society and European Bioanalysis Forum recommendations for validating such methods. CONCLUSIONS The implications of this effort include standardization of viable cell immunophenotyping of starting material for cell manufacturing, cell selection and in-process quality controls or dosing of IECs. This method also complies with all relevant standards, particularly FACT-JACIE standards, in terms of enumerating and reporting on the viability of the "clinically relevant cell populations."
Collapse
Affiliation(s)
- Bechara Mfarrej
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France.
| | - Julie Gaude
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| | - Jerome Couquiaud
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| | - Boris Calmels
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| | | | - Claude Lemarie
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
23
|
Dulan SO, Viers KL, Wagner JR, Clark MC, Chang B, Gorospe GL, Londrc A, Brown AS, Rosenthal J, Smith EP, Forman SJ, Snyder DS, Budde LE. Developing and Monitoring a Standard-of-Care Chimeric Antigen Receptor (CAR) T Cell Clinical Quality and Regulatory Program. Biol Blood Marrow Transplant 2020; 26:1386-1393. [PMID: 32439475 DOI: 10.1016/j.bbmt.2020.03.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/29/2022]
Abstract
As the world of cellular therapy expands to include immune effector cell (IEC) products such as commercial chimeric antigen receptor (CAR) T cells, quality management (QM) professionals are faced with creating either new IEC stand-alone programs or expand existing hematopoietic cell transplantation (HCT) programs to promote patient safety and be aligned with quality, regulatory, and accreditation requirements. The team professionals at City of Hope (COH) recently expanded the quality HCT program to include IEC products and, in doing so, implemented new regulatory infrastructure while maintaining high quality patient care. At COH, we developed the quality structure of our cellular therapy program through collaborations between quality, regulatory, and CAR T patient care committees, which included physicians and nurse coordinators. To ensure the quality of our program, we monitor data collection and reporting, perform quarterly proactive audits of, for example, outcome analysis, and measure selected end-points for benchmarking purposes. QM professionals play a critical role in the monitoring and evaluation processes and provide guidance on how to implement accreditation requirements and what impact the requirements may have on care management. Here we describe the process by which COH expanded our HCT QM program to include IEC therapy. We share examples of how we developed our overall program structure and other key items such as how we addressed patient care management and accreditation to apprise other programs that wish to create and/or expand existing programs.
Collapse
Affiliation(s)
- Sylvia O Dulan
- Department of Quality, Risk, and Regulatory Management, City of Hope National Medical Center, Duarte, California
| | - Kathie L Viers
- Department of Quality, Risk, and Regulatory Management, City of Hope National Medical Center, Duarte, California
| | - Jamie R Wagner
- Department of Hematology T Cell Therapeutics Research Laboratory, City of Hope Medical Center, Duarte, California
| | - Mary C Clark
- Department of Hematology, The Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California
| | - Brenda Chang
- Department of Hematology T Cell Therapeutics Research Laboratory, City of Hope Medical Center, Duarte, California
| | - Gerardo L Gorospe
- Department of Nursing Support Hem/HCT Nurse Coordinator, City of Hope Medical Center, Duarte, California
| | - Adina Londrc
- Department of Hematology, Division of Transplant and Cellular Therapy Reporting, City of Hope Medical Center, Duarte, California
| | - Annette S Brown
- Department of Hematology, Division of Transplant and Cellular Therapy Reporting, City of Hope Medical Center, Duarte, California
| | - Joseph Rosenthal
- Department of Hematology, The Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California
| | - Eileen P Smith
- Department of Hematology, The Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California
| | - Stephen J Forman
- Department of Hematology T Cell Therapeutics Research Laboratory, City of Hope Medical Center, Duarte, California; Department of Hematology, The Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California
| | - David S Snyder
- Department of Hematology, The Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California.
| | - Lihua E Budde
- Department of Hematology T Cell Therapeutics Research Laboratory, City of Hope Medical Center, Duarte, California; Department of Hematology, The Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California.
| |
Collapse
|
24
|
Management of the Critically Ill Adult Chimeric Antigen Receptor-T Cell Therapy Patient: A Critical Care Perspective. Crit Care Med 2019; 46:1402-1410. [PMID: 29939878 DOI: 10.1097/ccm.0000000000003258] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Chimeric antigen receptor T-cell therapy, a type of immune effector therapy for cancer, has demonstrated encouraging results in clinical trials for the treatment of patients with refractory hematologic malignancies. Nevertheless, there are toxicities specific to these treatments that, if not recognized and treated appropriately, can lead to multiple organ failure and death. This article is a comprehensive review of the available literature and provides, from a critical care perspective, recommendations by experienced intensivists in the care of critically ill adult chimeric antigen receptor T-cell patients. DATA SOURCES PubMed and Medline search of articles published from 2006 to date. STUDY SELECTION Clinical studies, reviews, or guidelines were selected and reviewed by the authors. DATA EXTRACTION Not available. DATA SYNTHESIS Not available. CONCLUSIONS Until modifications in chimeric antigen receptor T-cell therapy decrease their toxicities, the intensivist will play a leading role in the management of critically ill chimeric antigen receptor T-cell patients. As this novel immunotherapeutic approach becomes widely available, all critical care clinicians need to be familiar with the recognition and management of complications associated with this treatment.
Collapse
|
25
|
Santomasso B, Bachier C, Westin J, Rezvani K, Shpall EJ. The Other Side of CAR T-Cell Therapy: Cytokine Release Syndrome, Neurologic Toxicity, and Financial Burden. Am Soc Clin Oncol Educ Book 2019; 39:433-444. [PMID: 31099694 DOI: 10.1200/edbk_238691] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Immune effector cells, including T cells and natural killer cells, which are genetically engineered to express a chimeric antigen receptor (CAR), constitute a powerful new class of therapeutic agents to treat patients with hematologic malignancies. Several CAR T-cell trials have shown impressive remission rates in patients with relapsed/refractory hematologic cancers. Although the clinical responses of these agents in hematologic malignancies have been very encouraging, they have also produced substantial morbidity and occasionally mortality resulting from toxicity. With more experience and collaboration, hopefully the toxicities and the costs will come down, increasing the availability of CAR T cells to patients in need.
Collapse
Affiliation(s)
| | | | - Jason Westin
- 3 The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Katayoun Rezvani
- 3 The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
26
|
Kansagra AJ, Frey NV, Bar M, Laetsch TW, Carpenter PA, Savani BN, Heslop HE, Bollard CM, Komanduri KV, Gastineau DA, Chabannon C, Perales MA, Hudecek M, Aljurf M, Andritsos L, Barrett JA, Bachanova V, Bonini C, Ghobadi A, Gill SI, Hill JA, Kenderian S, Kebriaei P, Nagler A, Maloney D, Liu HD, Shah NN, Kharfan-Dabaja MA, Shpall EJ, Mufti GJ, Johnston L, Jacoby E, Bazarbachi A, DiPersio JF, Pavletic SZ, Porter DL, Grupp SA, Sadelain M, Litzow MR, Mohty M, Hashmi SK. Clinical utilization of Chimeric Antigen Receptor T-cells (CAR-T) in B-cell acute lymphoblastic leukemia (ALL)-an expert opinion from the European Society for Blood and Marrow Transplantation (EBMT) and the American Society for Blood and Marrow Transplantation (ASBMT). Bone Marrow Transplant 2019; 54:1868-1880. [PMID: 31092900 PMCID: PMC8268756 DOI: 10.1038/s41409-019-0451-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/24/2018] [Indexed: 01/05/2023]
Abstract
On August 30, 2017, the U.S. Food and Drug Administration (US-FDA) approved tisagenlecleucel (KYMRIAH, Novartis, Basel, Switzerland), a synthetic bioimmune product of anti-CD19 chimeric antigen receptor-T cells (CAR-T), for the treatment of children and young adults with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). With this new era of personalized cancer immunotherapy, multiple challenges are present ranging from implementation of a CAR-T program to safe delivery of the drug, long-term toxicity monitoring and disease assessments. To address these issues, experts representing the American Society for Blood and Marrow Transplant (ASBMT), the European Group for Blood and Marrow Transplantation (EBMT), the International Society of Cell and Gene Therapy (ISCT), and the Foundation for the Accreditation of Cellular Therapy (FACT), formed a global CAR-T task force to identify and address key questions pertinent for hematologists and transplant physicians regarding the clinical use of anti CD19 CAR-T therapy in patients with B-ALL. This article presents an initial roadmap for navigating common clinical practice scenarios that will become more prevalent now that the first commercially available CAR-T product for B-ALL has been approved.
Collapse
Affiliation(s)
- Ankit J. Kansagra
- 0000 0000 9482 7121grid.267313.2Department of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Noelle V. Frey
- 0000 0004 1936 8972grid.25879.31Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA USA
| | - Merav Bar
- 0000000122986657grid.34477.33Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA USA
| | - Theodore W. Laetsch
- 0000 0000 9482 7121grid.267313.2Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center and Children’s Health, Dallas, TX USA
| | - Paul A. Carpenter
- 0000000122986657grid.34477.33Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA USA
| | - Bipin N. Savani
- 0000 0004 1936 9916grid.412807.8Division of Hematology/Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Helen E. Heslop
- 0000 0004 0445 0041grid.63368.38Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston, TX USA
| | - Catherine M. Bollard
- 0000 0004 0482 1586grid.239560.bCenter for Cancer and Immunology Research, Children’s National Health System, Washington, DC USA
| | - Krishna V. Komanduri
- 0000 0004 0414 313Xgrid.418456.aSylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL USA
| | - Dennis A. Gastineau
- 0000 0000 8875 6339grid.417468.8Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ USA
| | - Christian Chabannon
- 0000 0001 2176 4817grid.5399.6Institut Paoli-Calmettes, Centre de Lutte Contre le Cancer; Centre d’Investigations Cliniques en Biothérapie, Université d’Aix-Marseille, Inserm CBT, 1409 Marseille, France
| | - Miguel A. Perales
- 0000 0001 2171 9952grid.51462.34Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY USA
| | - Michael Hudecek
- 0000 0001 1378 7891grid.411760.5Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Mahmoud Aljurf
- 0000 0001 2191 4301grid.415310.2Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Leslie Andritsos
- 0000 0001 1545 0811grid.412332.5Division of Hematology, Ohio State University Wexner Medical Center, Columbus, OH USA
| | - John A. Barrett
- 0000 0004 1936 9510grid.253615.6Stem Cell Transplantation and Cellular Therapy Program, GW Cancer Center, George Washington University, Washington, DC USA
| | - Veronika Bachanova
- 0000000419368657grid.17635.36Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN USA
| | - Chiara Bonini
- grid.15496.3f0000 0001 0439 0892Experimental Hematology Unit, University Vita-Salute San Raffaele and Ospedale San Raffaele, Milano, Italy
| | - Armin Ghobadi
- 0000 0001 2355 7002grid.4367.6Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO USA
| | - Saar I. Gill
- 0000 0004 1936 8972grid.25879.31Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA USA
| | - Joshua A. Hill
- 0000000122986657grid.34477.33Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA USA
| | - Saad Kenderian
- 0000 0004 0459 167Xgrid.66875.3aDivision of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN USA ,0000 0004 0459 167Xgrid.66875.3aDepartment of Immunology, Mayo Clinic, Rochester, MN USA
| | - Partow Kebriaei
- 0000 0001 2291 4776grid.240145.6Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Arnon Nagler
- 0000 0004 1937 0546grid.12136.37The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - David Maloney
- 0000000122986657grid.34477.33Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA USA
| | - Hien D. Liu
- 0000 0000 9891 5233grid.468198.aH. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Nirali N. Shah
- 0000 0001 2297 5165grid.94365.3dPediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | | | - Elizabeth J. Shpall
- 0000 0004 1937 0546grid.12136.37The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ghulam J. Mufti
- 0000 0001 2322 6764grid.13097.3cDepartment of Haematological Medicine, King’s College, London, UK
| | - Laura Johnston
- 0000000419368956grid.168010.eDepartment of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Elad Jacoby
- 0000 0000 9891 5233grid.468198.aH. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Ali Bazarbachi
- 0000 0004 0581 3406grid.411654.3Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - John F. DiPersio
- 0000 0004 0459 167Xgrid.66875.3aDivision of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN USA
| | - Steven Z. Pavletic
- 0000 0001 2297 5165grid.94365.3dExperimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - David L. Porter
- 0000 0004 1936 8972grid.25879.31Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA USA
| | - Stephan A. Grupp
- 0000 0004 1936 8972grid.25879.31Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA USA
| | - Michel Sadelain
- 0000 0001 2171 9952grid.51462.34Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, NY USA
| | - Mark R. Litzow
- 0000 0004 0459 167Xgrid.66875.3aDepartment of Immunology, Mayo Clinic, Rochester, MN USA
| | - Mohamad Mohty
- 0000 0001 2308 1657grid.462844.8Hôpital Saint-Antoine, APHP, Sorbonne Universite, INSERM UMRs 938, Paris, France
| | - Shahrukh K. Hashmi
- 0000 0001 1545 0811grid.412332.5Division of Hematology, Ohio State University Wexner Medical Center, Columbus, OH USA ,0000 0004 0459 167Xgrid.66875.3aDepartment of Immunology, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
27
|
Kansagra AJ, Frey NV, Bar M, Laetsch TW, Carpenter PA, Savani BN, Heslop HE, Bollard CM, Komanduri KV, Gastineau DA, Chabannon C, Perales MA, Hudecek M, Aljurf M, Andritsos L, Barrett JA, Bachanova V, Bonini C, Ghobadi A, Gill SI, Hill J, Kenderian S, Kebriaei P, Nagler A, Maloney D, Liu HD, Shah NN, Kharfan-Dabaja MA, Shpall EJ, Mufti GJ, Johnston L, Jacoby E, Bazarbachi A, DiPersio JF, Pavletic SZ, Porter DL, Grupp SA, Sadelain M, Litzow MR, Mohty M, Hashmi SK. Clinical Utilization of Chimeric Antigen Receptor T Cells in B Cell Acute Lymphoblastic Leukemia: An Expert Opinion from the European Society for Blood and Marrow Transplantation and the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant 2018; 25:e76-e85. [PMID: 30576834 DOI: 10.1016/j.bbmt.2018.12.068] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023]
Abstract
On August 30, 2017 the US Food and Drug Administration approved tisagenlecleucel (Kymriah; Novartis, Basel, Switzerland), a synthetic bioimmune product of anti-CD19 chimeric antigen receptor T cells (CAR-T), for the treatment of children and young adults with relapsed/refractory B cell acute lymphoblastic leukemia (B-ALL). With this new era of personalized cancer immunotherapy, multiple challenges are present, ranging from implementation of a CAR-T program to safe delivery of the drug, long-term toxicity monitoring, and disease assessments. To address these issues experts representing the American Society for Blood and Marrow Transplant, the European Society for Blood and Marrow Transplantation, the International Society of Cell and Gene Therapy, and the Foundation for the Accreditation of Cellular Therapy formed a global CAR-T task force to identify and address key questions pertinent for hematologists and transplant physicians regarding the clinical use of anti CD19 CAR-T therapy in patients with B-ALL. This article presents an initial roadmap for navigating common clinical practice scenarios that will become more prevalent now that the first commercially available CAR-T product for B-ALL has been approved.
Collapse
Affiliation(s)
- Ankit J Kansagra
- Department of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Noelle V Frey
- Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Merav Bar
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Theodore W Laetsch
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center and Children's Health, Dallas, Texas
| | - Paul A Carpenter
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC
| | - Krishna V Komanduri
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, Florida
| | - Dennis A Gastineau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona
| | - Christian Chabannon
- Institut Paoli-Calmettes, Centre de Lutte Contre le Cancer; Centre d'Investigations Cliniques en Biothérapie, Université d'Aix-Marseille, Inserm CBT 1409, Marseille, France
| | - Miguel A Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Mahmoud Aljurf
- Oncology Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Leslie Andritsos
- Division of Hematology, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - John A Barrett
- Stem Cell Transplantation and Cellular Therapy Program, GW Cancer Center, George Washington University, Washington, DC
| | - Veronika Bachanova
- Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Chiara Bonini
- Experimental Hematology Unit, University Vita-Salute San Raffaele and Ospedale San Raffaele, Milano, Italy
| | - Armin Ghobadi
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Saar I Gill
- Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua Hill
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Saad Kenderian
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota; Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arnon Nagler
- The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - David Maloney
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Hien D Liu
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ghulam J Mufti
- Department of Haematological Medicine, King's College, London, United Kingdom
| | - Laura Johnston
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Elad Jacoby
- The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Steven Z Pavletic
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David L Porter
- Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephan A Grupp
- Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, New York
| | - Mark R Litzow
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mohamad Mohty
- Hôpital Saint-Antoine, APHP, Sorbonne Universite, INSERM UMRs 938, Paris, France
| | - Shahrukh K Hashmi
- Oncology Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia; Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
28
|
Ioannidis JPA, Kim BYS, Trounson A. How to design preclinical studies in nanomedicine and cell therapy to maximize the prospects of clinical translation. Nat Biomed Eng 2018; 2:797-809. [PMID: 30931172 PMCID: PMC6436641 DOI: 10.1038/s41551-018-0314-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022]
Abstract
The clinical translation of promising products, technologies and interventions from the disciplines of nanomedicine and cell therapy has been slow and inefficient. In part, translation has been hampered by suboptimal research practices that propagate biases and hinder reproducibility. These include the publication of small and underpowered preclinical studies, suboptimal study design (in particular, biased allocation of experimental groups, experimenter bias and lack of necessary controls), the use of uncharacterized or poorly characterized materials, poor understanding of the relevant biology and mechanisms, poor use of statistics, large between-model heterogeneity, absence of replication, lack of interdisciplinarity, poor scientific training in study design and methods, a culture that does not incentivize transparency and sharing, poor or selective reporting, misaligned incentives and rewards, high costs of materials and protocols, and complexity of the developed products, technologies and interventions. In this Perspective, we discuss special manifestations of these problems in nanomedicine and in cell therapy, and describe mitigating strategies. Progress on reducing bias and enhancing reproducibility early on ought to enhance the translational potential of biomedical findings and technologies.
Collapse
Affiliation(s)
- John P A Ioannidis
- Departments of Medicine, Health Research and Policy, and Biomedical Data Science, Stanford University School of Medicine, and Department of Statistics, Stanford University School of Humanities and Sciences, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA.
| | - Betty Y S Kim
- Departments of Neurosurgery, Cancer Biology, and Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Alan Trounson
- Monash University & Hudson Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
29
|
LYSAGHT TAMRA, MUNSIE MEGAN, CASTRICUM ADAM, HUI JAMESH, OKADA KYOSHI, SATO YOJI, SAWA YOSHIKI, STEWART CAMERON, TAN LIPKUN, TAN LYNNH, SUGII SHIGEKI. A roundtable on responsible innovation with autologous stem cells in Australia, Japan and Singapore. Cytotherapy 2018; 20:1103-1109. [DOI: 10.1016/j.jcyt.2018.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 01/22/2023]
|
30
|
Abstract
Bioreactors have become indispensable tools in the cell-based therapy industry. Various forms of bioreactors are used to maintain well-controlled microenvironments to regulate cell growth, differentiation, and tissue development. They are essential for providing standardized, reproducible cell-based products for regenerative medicine applications or to establish physiologically relevant
in vitro models for testing of pharmacologic agents. In this review, we discuss three main classes of bioreactors: cell expansion bioreactors, tissue engineering bioreactors, and lab-on-a-chip systems. We briefly examine the factors driving concerted research endeavors in each of these areas and describe the major advancements that have been reported in the last three years. Emerging issues that impact the commercialization and clinical use of bioreactors include (i) the need to scale up to greater cell quantities and larger graft sizes, (ii) simplification of
in vivo systems to function without exogenous stem cells or growth factors or both, and (iii) increased control in the manufacture and monitoring of miniaturized systems to better capture complex tissue and organ physiology.
Collapse
Affiliation(s)
- Makeda Stephenson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Warren Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
31
|
|
32
|
Perica K, Curran KJ, Brentjens RJ, Giralt SA. Building a CAR Garage: Preparing for the Delivery of Commercial CAR T Cell Products at Memorial Sloan Kettering Cancer Center. Biol Blood Marrow Transplant 2018; 24:1135-1141. [PMID: 29499327 DOI: 10.1016/j.bbmt.2018.02.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 10/17/2022]
Abstract
Two commercial chimeric antigen receptor (CAR) T cell therapies for CD19-expressing B cell malignancies, Kymriah and Yescarta, have recently been approved by the Food and Drug Administration. The administration of CAR T cells is a complex endeavor involving cell manufacture, tracking and shipping of apheresis products, and management of novel and severe toxicities. At Memorial Sloan Kettering Cancer Center, we have identified 8 essential tasks that define the CAR T cell workflow. In this review, we discuss practical aspects of CAR T cell program development, including clinical, administrative, and regulatory challenges for successful implementation.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kevin J Curran
- Pediatric Bone Marrow Transplant Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York; Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sergio A Giralt
- Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
33
|
Perales MA, Kebriaei P, Kean LS, Sadelain M. Reprint of: Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR. Biol Blood Marrow Transplant 2018; 24:S15-S19. [PMID: 29425516 DOI: 10.1016/j.bbmt.2017.12.789] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 01/14/2023]
Abstract
Therapeutic T cell engineering has recently garnered widespread interest because of the success of CD19 chimeric antigen receptor (CAR) therapy. CARs are synthetic receptors for antigen that redirect the specificity and reprogram the function of the T cells in which they are genetically introduced. CARs targeting CD19, a cell surface molecule found in most leukemias and lymphomas, have yielded high remission rates in patients with chemorefractory, relapsed disease, including acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin lymphoma. The toxicities of this treatment include B cell aplasia, cytokine release syndrome (CRS), and neurotoxicity. Although reversible in most instances, these toxicities may require specific medical interventions, including transfer to intensive care to treat severe CRS. Guidelines for managing these toxicities are emerging. The recent report of a nonhuman primate model for CRS is poised to help advance the management of this syndrome. Finally, new engineering modalities, based on the use of targeted nucleases like CRISPR, may further enhance the efficacy and safety of CAR T cells.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Leslie S Kean
- The Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Center, Seattle, Washington; The Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Pediatrics, University of Washington, Seattle, Washington
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
34
|
Chimeric Antigen Receptor-T Cell Therapy: Practical Considerations for Implementation in Europe. Hemasphere 2018; 2:e18. [PMID: 31723747 PMCID: PMC6745952 DOI: 10.1097/hs9.0000000000000018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is a new class of cellular immunotherapies that involves ex vivo genetic modification of T cells to incorporate an engineered CAR. After infusion into the patient, the CAR-expressing T cells recognize specific tumor targets and induce an immune response against them. The technology utilized is fundamentally different from previously available cancer treatments. Currently, most CAR-T cell therapies use autologous T cells. Tisagenlecleucel (formerly CTL019) is an anti-CD19 CAR-T cell therapy that was recently approved in the United States for the treatment of pediatric and young adult patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). Tisagenlecleucel has shown robust in vivo expansion and long-term persistence, clinically meaningful durable response and remission rates, and overall survival benefit in pediatric and young adult patients with relapsed/refractory B-ALL and in relapsed/refractory diffuse large B-cell lymphoma. Common adverse events (AEs) include cytokine release syndrome, which may require hospitalization and admission to an intensive care unit, neurological toxicities, and B-cell aplasia. These AEs are manageable when treated by an appropriately trained team. Additional research is required to further develop AE management protocols. In this review, we describe regulatory requirements, clinical considerations, and site-level requirements for clinical study implementation of CAR-T cell therapy in Europe. We also provide a case study of the European experience from the first global clinical trial for tisagenlecleucel, which may serve as a useful starting point for investigators and clinicians looking to implement CAR-T cell therapy at their institutions.
Collapse
|
35
|
Wilkes GM. Targeted Therapy: Attacking Cancer with Molecular and Immunological Targeted Agents. Asia Pac J Oncol Nurs 2018; 5:137-155. [PMID: 29607374 PMCID: PMC5863423 DOI: 10.4103/apjon.apjon_79_17] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Today, personalized cancer therapy with targeted agents has taken center stage, and offers individualized treatment to many. As the mysteries of the genes in a cell's DNA and their specific proteins are defined, advances in the understanding of cancer gene mutations and how cancer evades the immune system have been made. This article provides a basic and simplified understanding of the available (Food and Drug Administration- approved) molecularly and immunologically targeted agents in the USA. Other agents may be available in Asia, and throughout the USA and the world, many more agents are being studied. Nursing implications for drug classes are reviewed.
Collapse
|
36
|
Perales MA, Kebriaei P, Kean LS, Sadelain M. Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR. Biol Blood Marrow Transplant 2018; 24:27-31. [PMID: 29032264 PMCID: PMC5901894 DOI: 10.1016/j.bbmt.2017.10.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 12/01/2022]
Abstract
Therapeutic T cell engineering has recently garnered widespread interest because of the success of CD19 chimeric antigen receptor (CAR) therapy. CARs are synthetic receptors for antigen that redirect the specificity and reprogram the function of the T cells in which they are genetically introduced. CARs targeting CD19, a cell surface molecule found in most leukemias and lymphomas, have yielded high remission rates in patients with chemorefractory, relapsed disease, including acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin lymphoma. The toxicities of this treatment include B cell aplasia, cytokine release syndrome (CRS), and neurotoxicity. Although reversible in most instances, these toxicities may require specific medical interventions, including transfer to intensive care to treat severe CRS. Guidelines for managing these toxicities are emerging. The recent report of a nonhuman primate model for CRS is poised to help advance the management of this syndrome. Finally, new engineering modalities, based on the use of targeted nucleases like CRISPR, may further enhance the efficacy and safety of CAR T cells.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Leslie S Kean
- The Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Center, Seattle, Washington; The Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Pediatrics, University of Washington, Seattle, Washington
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
37
|
Locke FL, Anasetti C. Transplanters drive CARs to the clinic by brewing ICE-T: the Moffitt roadmap. J Immunother Cancer 2017; 5:59. [PMID: 28716155 PMCID: PMC5514522 DOI: 10.1186/s40425-017-0265-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/04/2017] [Indexed: 01/02/2023] Open
Abstract
Recent single institution clinical trial successes with anti-CD19 Chimeric Antigen Receptor (CAR) T cell therapy for B cell malignancies attracted significant attention from industry. Our center sought to rapidly and safely bring industry sponsored pivotal trials to our patients and to prepare for additional cell therapy trials in solid and liquid tumors from both industry and our own investigators. We implemented a collaborative cross departmental program to provide clinical care and trial coordination for immune cell therapies. The Moffitt Immune Cell Therapy (ICE-T) program oversees and administers not only CAR T cell therapy for hematologic malignancies, but TIL and TCR therapy for solid tumor patients. Disease specific experts maintain oversight as principal investigators of these key trials yet the coordination and clinical care is centralized to leverage the expertise and infrastructure of our already robust Blood and Marrow Transplantation program.
Collapse
Affiliation(s)
- Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, 12902 Magnolia Dr. FOB-3, Tampa, FL, 33612, USA. .,Department of Oncologic Sciences, University of South Florida, Tampa, USA. .,Immunology Program, Moffitt Cancer Center, Tampa, USA.
| | - Claudio Anasetti
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, 12902 Magnolia Dr. FOB-3, Tampa, FL, 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, USA.,Immunology Program, Moffitt Cancer Center, Tampa, USA
| | | |
Collapse
|
38
|
Locke FL, Davila ML. Regulatory challenges and considerations for the clinical application of CAR-T cell anti-cancer therapy. Expert Opin Biol Ther 2017; 17:659-661. [PMID: 28454503 DOI: 10.1080/14712598.2017.1322953] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Frederick L Locke
- a Department of Blood and Marrow Transplant and Cellular Immunotherapy , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA.,b Department of Oncologic Sciences, Morsani College of Medicine , University of South Florida , Tampa , FL , USA
| | - Marco L Davila
- a Department of Blood and Marrow Transplant and Cellular Immunotherapy , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA.,b Department of Oncologic Sciences, Morsani College of Medicine , University of South Florida , Tampa , FL , USA
| |
Collapse
|