1
|
Ahmed N, Wesson W, Lutfi F, Porter DL, Bachanova V, Nastoupil LJ, Perales MA, Maziarz RT, Brower J, Shah GL, Chen AI, Oluwole OO, Schuster SJ, Bishop MR, McGuirk JP, Riedell PA. Optimizing the post-CAR T monitoring period in recipients of axicabtagene ciloleucel, tisagenlecleucel, and lisocabtagene maraleucel. Blood Adv 2024; 8:5346-5354. [PMID: 39042880 DOI: 10.1182/bloodadvances.2023012549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/15/2024] [Accepted: 05/17/2024] [Indexed: 07/25/2024] Open
Abstract
ABSTRACT CD19-directed chimeric antigen receptor T-cell (CAR T) therapies, including axicabtagene ciloleucel (axi-cel), tisagenlecleucel (tisa-cel), and lisocabtagene maraleucel (liso-cel), have transformed the treatment landscape for B-cell non-Hodgkin lymphoma, showcasing significant efficacy but also highlighting toxicity risks such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). The US Food and Drug Administration has mandated patients remain close to the treatment center for 4 weeks as part of a Risk Evaluation and Mitigation Strategy to monitor and manage these toxicities, which, although cautious, may add to cost of care, be burdensome for patients and their families, and present challenges related to patient access and socioeconomic disparities. This retrospective study across 9 centers involving 475 patients infused with axi-cel, tisa-cel, and liso-cel from 2018 to 2023 aimed to assess CRS and ICANS onset and duration, as well as causes of nonrelapse mortality (NRM) in real-world CAR T recipients. Although differences were noted in the incidence and duration of CRS and ICANS between CAR T products, new-onset CRS and ICANS are exceedingly rare after 2 weeks after infusion (0% and 0.7% of patients, respectively). No new cases of CRS occurred after 2 weeks and a single case of new-onset ICANS occurred in the third week after infusion. NRM is driven by ICANS in the early follow-up period (1.1% until day 28) and then by infection through 3 months after infusion (1.2%). This study provides valuable insights into optimizing CAR T therapy monitoring, and our findings may provide a framework to reduce physical and financial constraints for patients.
Collapse
Affiliation(s)
- Nausheen Ahmed
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Westwood, KS
| | - William Wesson
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Westwood, KS
| | - Forat Lutfi
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Westwood, KS
| | - David L Porter
- Abramson Cancer Center and Center for Cell Therapy and Transplant, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Veronika Bachanova
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | | | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard T Maziarz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Jamie Brower
- Abramson Cancer Center and Center for Cell Therapy and Transplant, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Gunjan L Shah
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andy I Chen
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Olalekan O Oluwole
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Stephen J Schuster
- Abramson Cancer Center and Center for Cell Therapy and Transplant, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Michael R Bishop
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL
| | - Joseph P McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Westwood, KS
| | - Peter A Riedell
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL
| |
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
|
Furqan F, Bhatlapenumarthi V, Dhakal B, Fenske TS, Farrukh F, Longo W, Akhtar O, D'Souza A, Pasquini M, Guru Murthy GS, Runaas L, Abedin S, Mohan M, Shah NN, Hamadani M. Outpatient administration of CAR T-cell therapies using a strategy of no remote monitoring and early CRS intervention. Blood Adv 2024; 8:4320-4329. [PMID: 38889435 PMCID: PMC11372811 DOI: 10.1182/bloodadvances.2024013239] [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: 03/26/2024] [Revised: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
ABSTRACT Recent studies demonstrating the feasibility of outpatient chimeric antigen receptor (CAR)-modified T-cell therapy administration are either restricted to CARs with 41BB costimulatory domains or use intensive at-home monitoring. We report outcomes of outpatient administration of all commercially available CD19- and B-cell maturation antigen (BCMA)-directed CAR T-cell therapy using a strategy of no remote at-home monitoring and an early cytokine release syndrome (CRS) intervention strategy. Patients with hematologic malignancies who received CAR T-cell therapy in the outpatient setting during 2022 to 2023 were included. Patients were seen daily in the cancer center day hospital for the first 7 to 10 days and then twice weekly through day 30. The primary end point was to determine 3-, 7-, and 30-day post-CAR T-cell infusion hospitalizations. Early CRS intervention involved administering tocilizumab as an outpatient for grade ≥1 CRS. Fifty-eight patients received outpatient CAR T-cell infusion (33 myeloma, 24 lymphoma, and 1 acute lymphoblastic leukemia). Of these, 17 (41%), 16 (38%), and 9 patients (21%) were admitted between days 0 to 3, 4 to 7, and 8 to 30 after CAR T-cell infusion, respectively. The most common reason for admission was CAR T-cell-related toxicities (33/42). Hospitalization was prevented in 15 of 35 patients who received tocilizumab for CRS as an outpatient. The nonrelapse mortality rates were 1.7% at 1 month and 3.4% at 6 months. In conclusion, we demonstrate that the administration of commercial CAR T-cell therapies in an outpatient setting is safe and feasible without intensive remote monitoring using an early CRS intervention strategy.
Collapse
Affiliation(s)
- Fateeha Furqan
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Vineel Bhatlapenumarthi
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Binod Dhakal
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Timothy S Fenske
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Faiqa Farrukh
- Department of Medicine, Jefferson Abington Hospital, Abington, PA
| | - Walter Longo
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Othman Akhtar
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Anita D'Souza
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Marcelo Pasquini
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Guru Subramanian Guru Murthy
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Lyndsey Runaas
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Sameem Abedin
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Meera Mohan
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Nirav N Shah
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Mehdi Hamadani
- Blood and Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
4
|
Hansen DK, Dhakal B, Hamadani M, Dingli D, Jain T, Huff CA, Janakiram M, Liu YH, De Braganca KC, Lodowski N, Sander J, Okorozo P, McFarland L, Perciavalle M, Huo S, Qureshi ZP, Patel KK. Clinician and administrator perspectives on outpatient administration of ciltacabtagene autoleucel in relapsed or refractory multiple myeloma. Front Immunol 2024; 15:1405452. [PMID: 38915401 PMCID: PMC11194690 DOI: 10.3389/fimmu.2024.1405452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/24/2024] [Indexed: 06/26/2024] Open
Abstract
Introduction Chimeric antigen receptor (CAR) T-cell therapy (CAR T therapy) is a treatment option for patients with relapsed or refractory multiple myeloma that has led to unprecedented treatment outcomes. Among CAR T therapies available, ciltacabtagene autoleucel (cilta-cel) is a good candidate for outpatient administration due to its generally predictable safety profile. There are multiple advantages of outpatient administration of cilta-cel, including reduced healthcare burden, expanded access, and patient autonomy. This mixed methods qualitative study aimed to identify key factors for outpatient administration of CAR T and best practice recommendations by combining a targeted literature review with expert interviews and panels. Methods The targeted review (Phase 1) aimed to identify factors for outpatient CAR T administration in the US and determine key topics for the exploratory interviews (Phase 2) and expert panels (Phase 3), which aimed to inform on best practices and challenges of outpatient CAR T administration (focusing on cilta-cel). Participants in clinical and administrative positions based in treatment centers that had experience with real-world outpatient administration of cilta-cel were recruited. Results Seventeen studies were identified in Phase 1. Key factors for outpatient administration included the development of protocols for CAR T complications, education for caregivers, outpatient specialists, hospital staff, and emergency services staff for identification and referral after possible adverse events, the creation of multidisciplinary teams for effective communication and management, straightforward patient intake processes encompassing financial eligibility review and provision of patient education materials, and close patient monitoring throughout the treatment journey. In Phase 2, 5 participants from 2 centers were interviewed. In Phase 3, 14 participants across 6 treatment centers were interviewed. Two 90-minute virtual panel discussions took place. All participants agreed that cilta-cel can be safely and effectively administered in an outpatient setting. Key recommendations included the creation of educational resources for patients and caregivers, the development of standard operating procedures, dedicated outpatient infrastructure and establishment of interdisciplinary teams, outpatient monitoring for toxicity management, and monitoring of the reimbursement landscape. Discussion This study offers a comprehensive understanding of the feasibility of outpatient cilta-cel administration in participating CAR T centers and provides actionable recommendations while acknowledging existing challenges.
Collapse
Affiliation(s)
- Doris K. Hansen
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Binod Dhakal
- Blood and Marrow Transplant (BMT) and Cellular Therapy, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Mehdi Hamadani
- Blood and Marrow Transplant (BMT) and Cellular Therapy, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - David Dingli
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Carol Ann Huff
- Division of Hematological Malignancies and Bone Marrow Transplantation, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Murali Janakiram
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, United States
| | - Yi-Hsuan Liu
- Real-World Value & Evidence, Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, United States
| | - Kevin C. De Braganca
- Medical Affairs, Janssen Research & Development, LLC, a Johnson & Johnson company, Raritan, NJ, United States
| | | | | | | | | | | | - Stephen Huo
- Medical Affairs, Janssen Research & Development, LLC, a Johnson & Johnson company, Raritan, NJ, United States
| | - Zaina P. Qureshi
- Real-World Value & Evidence, Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, United States
| | - Krina K. Patel
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
5
|
Foley R, Kuruvilla J. Identification of a Patient Suitable for CAR-T Cell Therapy in the Outpatient Setting: A Vodcast and Case Example. Oncol Ther 2024; 12:239-245. [PMID: 38662113 PMCID: PMC11187035 DOI: 10.1007/s40487-024-00272-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/05/2024] [Indexed: 04/26/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapies targeting the CD19 antigen have been associated with high and durable response rates in patients with diffuse large B cell lymphoma (DLBCL). CAR-T cell therapies are commonly administered in the inpatient setting due to the average onset of cytokine release syndrome within the first 3 days post infusion, but there has been growing interest in delivering CAR-T cell therapies in the outpatient setting to overcome frequent hospital bed shortages and the high cost of inpatient care. Although this approach could improve access whilst catering to patient preference, it requires a multidisciplinary approach as well as careful patient selection. Herein, Dr. Foley and Dr. Kuruvilla discuss the case of a patient presenting with the ideal profile for CAR-T cell therapy referral whilst also determining the key attributes for eligibility from a clinician's perspective. Solutions for successful outpatient management include proper education, caregiver support, and early referral to ensure a timely infusion. In conclusion, outpatient administration of CAR-T cell therapy in patients with DLBCLs should be assessed on a case-by-case basis.A vodcast feature is available for this article.
Collapse
Affiliation(s)
- Ronan Foley
- Juravinski Hospital and Cancer Centre, Hamilton, ON, Canada.
| | | |
Collapse
|
6
|
Perez A, Al Sagheer T, Nahas GR, Linhares YPL. Outpatient administration of CAR T-cell therapy: a focused review with recommendations for implementation in community based centers. Front Immunol 2024; 15:1412002. [PMID: 38779668 PMCID: PMC11109356 DOI: 10.3389/fimmu.2024.1412002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
Chimeric Antigen Receptor T-cell (CAR-T) therapy has transformed the treatment landscape for hematological malignancies, showing high efficacy in patients with relapsed or refractory (R/R) disease and otherwise poor prognosis in the pre-CAR-T era. These therapies have been usually administered in the inpatient setting due to the risk of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). However, there is a growing interest in the transition to outpatient administration due to multiple reasons. We review available evidence regarding safety and feasibility of outpatient administration of CD19 targeted and BCMA targeted CAR T-cell therapy with an emphasis on the implementation of outpatient CAR-T programs in community-based centers.
Collapse
|
7
|
Romon I, Gonzalez-Barrera S, Coello de Portugal C, Ocio E, Sampedro I. Brave new world: expanding home care in stem cell transplantation and advanced therapies with new technologies. Front Immunol 2024; 15:1366962. [PMID: 38736880 PMCID: PMC11082320 DOI: 10.3389/fimmu.2024.1366962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Hematopoietic stem cell transplantation and cell therapies like CAR-T are costly, complex therapeutic procedures. Outpatient models, including at-home transplantation, have been developed, resulting in similar survival results, reduced costs, and increased patient satisfaction. The complexity and safety of the process can be addressed with various emerging technologies (artificial intelligence, wearable sensors, point-of-care analytical devices, drones, virtual assistants) that allow continuous patient monitoring and improved decision-making processes. Patients, caregivers, and staff can also benefit from improved training with simulation or virtual reality. However, many technical, operational, and above all, ethical concerns need to be addressed. Finally, outpatient or at-home hematopoietic transplantation or CAR-T therapy creates a different, integrated operative system that must be planned, designed, and carefully adapted to the patient's characteristics and distance from the hospital. Patients, clinicians, and their clinical environments can benefit from technically improved at-home transplantation.
Collapse
Affiliation(s)
- Iñigo Romon
- Transfusion Section, Hematology Department, University Hospital “Marques de Valdecilla”, Santander, Spain
| | - Soledad Gonzalez-Barrera
- Home Hospitalization Department, University Hospital “Marques de Valdecilla” - Instituto de Investigación Valdecilla (IDIVAL), Santander, Spain
| | | | - Enrique Ocio
- Hematology Department, University Hospital “Marques de Valdecilla” - IDIVAL, Santander, Spain
| | - Isabel Sampedro
- Home Hospitalization Department, University Hospital “Marques de Valdecilla” - Instituto de Investigación Valdecilla (IDIVAL), Santander, Spain
| |
Collapse
|
8
|
Gatwood K, Mahmoudjafari Z, Baer B, Pak S, Lee B, Kim H, Abernathy K, Dholaria B, Oluwole O. Outpatient CAR T-Cell Therapy as Standard of Care: Current Perspectives and Considerations. Clin Hematol Int 2024; 6:11-20. [PMID: 38817307 PMCID: PMC11086991 DOI: 10.46989/001c.115793] [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: 01/11/2024] [Accepted: 02/13/2024] [Indexed: 06/01/2024] Open
Abstract
Chimeric antigen receptor T-cell therapy (CAR-T) has altered the treatment landscape of several hematologic malignancies. Until recently, most CAR-T infusions have been administered in the inpatient setting, due to their toxicity profile. However, the advent of new product constructs, as well as improved detection and management of adverse effects, have greatly increased the safety in administering these therapies. CAR-T indications continue to expand, and inpatient administration is associated with increased healthcare resource utilization and overall cost. Therefore, transitioning CAR-T administration to the outpatient setting has been of great interest in an effort to improve access, reduce financial burden, and improve patient satisfaction. Establishment of a successful outpatient CAR-T requires several components, including a multidisciplinary cellular therapy team and an outpatient center with appropriate clinical space and personnel. Additionally, clear criteria for outpatient administration eligibility and for inpatient admission with pathways for prompt toxicity evaluation and admission, and toxicity management guidelines should be implemented. Education about CAR-T therapy and its associated toxicities is imperative for all clinical staff, as well as patients and their caregivers. Finally, rigorous financial planning and close collaboration with payers to ensure equitable access, while effectively managing cost, are essential to program success and sustainability. This review provides a summary of currently published experiences, as well as expert opinion regarding implementation of an outpatient CAR-T program.
Collapse
Affiliation(s)
| | | | | | - Stacy Pak
- PharmacyCity Of Hope National Medical Center
| | | | - Hoim Kim
- City Of Hope National Medical Center
| | | | | | - Olalekan Oluwole
- MedicineHematology and oncologyVanderbilt University Medical Center
| |
Collapse
|
9
|
Penack O, Dreger P, Ajib S, Ayuk F, Baermann BN, Bug G, Kriege O, Jentzsch M, Kobbe G, Koenecke C, Lutz M, Martin S, Schlegel PG, Schroers R, von Tresckow B, Vucinic V, Subklewe M, Bethge W, Wolff D. Management of Patients Undergoing CAR-T Cell Therapy in Germany. Oncol Res Treat 2024; 47:65-75. [PMID: 38198763 PMCID: PMC10911269 DOI: 10.1159/000536201] [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: 09/07/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
INTRODUCTION Chimeric antigen receptor positive T cell (CAR-T cell) treatment became standard therapy for relapsed or refractory hematologic malignancies, such as non-Hodgkin's lymphoma and multiple myeloma. Owing to the rapidly progressing field of CAR-T cell therapy and the lack of generally accepted treatment guidelines, we hypothesized significant differences between centers in the prevention, diagnosis, and management of short- and long-term complications. METHODS To capture the current CAR-T cell management among German centers to determine the medical need and specific areas for future clinical research, the DAG-HSZT (Deutsche Arbeitsgemeinschaft für Hämatopoetische Stammzelltransplantation und Zelluläre Therapie; German Working Group for Hematopoietic Stem Cell Transplantation and Cellular Therapy) performed a survey among 26 German CAR-T cell centers. RESULTS We received answers from 17 centers (65%). The survey documents the relevance of evidence in the CAR-T cell field with a homogeneity of practice in areas with existing clinical evidence. In contrast, in areas with no - or low quality - clinical evidence, we identified significant variety in management in between the centers: management of cytokine release syndrome, immune effector cell-related neurotoxicity syndrome, IgG substitution, autologous stem cell backups, anti-infective prophylaxis, and vaccinations. CONCLUSION The results indicate the urgent need for better harmonization of supportive care in CAR-T cell therapies including clinical research to improve clinical outcome.
Collapse
Affiliation(s)
- Olaf Penack
- Medical Clinic, Department for Haematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Dreger
- Medical Clinic, Department for Haematology and Oncology, University of Heidelberg, Heidelberg, Germany
| | - Salem Ajib
- Department of Medicine 2, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - Francis Ayuk
- Clinic for Hematology, Stem Cell Transplantation, University Clinic Hamburg Eppendorf, Hamburg, Germany
| | - Ben-Niklas Baermann
- Department of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Gesine Bug
- Department of Medicine 2, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - Oliver Kriege
- Department of Medicine III, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Madlen Jentzsch
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and Stem-Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Mathias Lutz
- Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Sonja Martin
- Robert-Bosch-Krankenhaus, Department for Hematology, Oncology and Palliative Care, Stuttgart, Germany
| | - Paul-Gerhard Schlegel
- Department of Pediatrics, Section of Pediatric Hematology and Oncology, Stem Cell Transplantation (SCT), University Hospital Würzburg, Würzburg, Germany
| | - Roland Schroers
- Ruhr-University Bochum, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Vladan Vucinic
- Medical Clinic and Policlinic 1, Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Marion Subklewe
- Medical Clinic, Department for Haematology and Oncology, Ludwig-Maximilians-University, Munich, Germany
| | - Wolfgang Bethge
- Inteernal Medicine II, Hematology and Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Daniel Wolff
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
10
|
Winestone LE, Bhojwani D, Ghorashian S, Muffly L, Leahy AB, Chao K, Steineck A, Rössig C, Lamble A, Maude SL, Myers R, Rheingold SR. INSPIRED Symposium Part 4A: Access to CAR T Cell Therapy in Unique Populations with B Cell Acute Lymphoblastic Leukemia. Transplant Cell Ther 2024; 30:56-70. [PMID: 37821078 DOI: 10.1016/j.jtct.2023.10.005] [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: 08/28/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
The approval of tisagenlecleucel (tisa-cel) for use in children with B cell acute lymphoblastic leukemia (B-ALL) was based on the phase 2 ELIANA trial, a global registration study. However, the ELIANA trial excluded specific subsets of patients facing unique challenges and did not include a sufficient number of patients to adequately evaluate outcomes in rare subpopulations. Since the commercialization of tisa-cel, data have become available that support therapeutic indications beyond the specific cohorts previously eligible for chimeric antigen receptor (CAR) T cells targeted to CD19 (CD19 CAR-T) therapy on the registration clinical trial. Substantial real-world data and aggregate clinical trial data have addressed gaps in our understanding of response rates, longer-term efficacy, and toxicities associated with CD19 CAR-T in special populations and rare clinical scenarios. These include patients with central nervous system relapsed disease, who were excluded from ELIANA and other early CAR-T trials owing to concerns about risk of neurotoxicity that have not been born out. There is also interest in the use of CD19 CAR-T for very-high-risk patients earlier in the course of therapy, such as patients with persistent minimal residual disease after 2 cycles of upfront chemotherapy and patients with first relapse of B-ALL. However, these indications are not specified on the label for tisa-cel and historically were not included in eligibility criteria for most clinical trials; data addressing these populations are needed. Populations at high risk of relapse, including patients with high-risk cytogenetic lesions, infants with B-ALL, patients with trisomy 21, and young adults with B-ALL, also may benefit from earlier treatment with CD19 CAR-T. It is important to prospectively study patient-reported outcomes given the differential toxicity expected between CD19 CAR-T and the historic standard of care, hematopoietic cell transplantation. Now that CD19 CAR-T therapy is commercially available, studies evaluating potential access disparities created by this very expensive novel therapy are increasingly pressing.
Collapse
Affiliation(s)
- Lena E Winestone
- Division of Allergy, Immunology, and BMT, Department of Pediatrics, UCSF Benioff Children's Hospitals, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.
| | - Deepa Bhojwani
- Division of Pediatric Hematology-Oncology, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center and Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Sara Ghorashian
- Haematology Department, Great Ormond Street Hospital, London UK, Developmental Biology and Cancer, UCL-Great Ormond Street Institute of Child Health, University College London, London United Kingdom
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, California
| | - Allison Barz Leahy
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Karen Chao
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Angela Steineck
- MACC Fund Center for Cancer and Blood Disorders, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Claudia Rössig
- University Children's Hospital Muenster, Pediatric Hematology and Oncology, Muenster, Germany; Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Adam Lamble
- Division of Hematology and Oncology, Seattle Children's Hospital, Department of Pediatrics, University of Washington, Seattle, Washington
| | - Shannon L Maude
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Regina Myers
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan R Rheingold
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Motais B, Charvátová S, Walek Z, Hájek R, Bagó JR. NK92 Expressing Anti-BCMA CAR and Secreted TRAIL for the Treatment of Multiple Myeloma: Preliminary In Vitro Assessment. Cells 2023; 12:2748. [PMID: 38067177 PMCID: PMC10706019 DOI: 10.3390/cells12232748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/03/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Multiple myeloma (MM) has witnessed improved patient outcomes through advancements in therapeutic approaches. Notably, allogeneic stem cell transplantation, proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies have contributed to enhanced quality of life. Recently, a promising avenue has emerged with chimeric antigen receptor (CAR) T cells targeting B-cell maturation antigen (BCMA), expressed widely on MM cells. To mitigate risks associated with allogenic T cells, we investigated the potential of BCMA CAR expression in natural killer cells (NKs), known for potent cytotoxicity and minimal side effects. Using the NK-92 cell line, we co-expressed BCMA CAR and soluble tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL) employing the piggyBac transposon system. Engineered NK cells (CAR-NK-92-TRAIL) demonstrated robust cytotoxicity against a panel of MM cell lines and primary patient samples, outperforming unmodified NK-92 cells with a mean difference in viability of 45.1% (±26.1%, depending on the target cell line). Combination therapy was explored with the proteasome inhibitor bortezomib (BZ) and γ-secretase inhibitors (GSIs), leading to a significant synergistic effect in combination with CAR-NK-92-TRAIL cells. This synergy was evident in cytotoxicity assays where a notable decrease in MM cell viability was observed in combinatorial therapy compared to single treatment. In summary, our study demonstrates the therapeutic potential of the CAR-NK-92-TRAIL cells for the treatment of MM. The synergistic impact of combining these engineered NK cells with BZ and GSI supports further development of allogeneic CAR-based products for effective MM therapy.
Collapse
Affiliation(s)
- Benjamin Motais
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
- Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
| | - Sandra Charvátová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
- Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic
| | - Zuzana Walek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic
| | - Juli R. Bagó
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic
| |
Collapse
|
12
|
Strati P, Gregory T, Majhail NS, Jain N. Chimeric Antigen Receptor T-Cell Therapy for Hematologic Malignancies: A Practical Review. JCO Oncol Pract 2023; 19:706-713. [PMID: 37406255 DOI: 10.1200/op.22.00819] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has become an established therapeutic approach for the treatment of hematologic malignancies. The field continues to evolve rapidly and newer-generation constructs are being designed to enhance proliferative capacity, and achieve long-term persistence and greater efficacy with an overall lower incidence of toxicity. Initial clinical application of CAR-T therapies has focused on relapsed and/or refractory hematologic malignancies, and Food and Drug Administration-approved CAR-T products targeting CD19 are available for B-cell acute lymphoblastic leukemia and low- and high-grade B-cell non-Hodgkin lymphoma, and targeting B-cell maturation antigen are available for multiple myeloma. Cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome have been recognized as class specific toxicities associated with these novel therapies. In this review, we focus on the clinical application of CAR-T therapies in adult patients with hematologic malignancies, including access issues, outpatient administration, and appropriate timing for referring a patient to a CAR-T treatment center.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tara Gregory
- Colorado Blood Cancer Institute, Denver, CO
- Sarah Cannon Transplant and Cellular Therapy Program at Presbyterian/St Luke's Medical Center, Denver, CO
| | - Navneet S Majhail
- Sarah Cannon, Nashville, TN
- Sarah Cannon Transplant and Cellular Therapy Program at TriStar Centennial, Nashville, TN
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
13
|
Schneeberger AR, Werthmueller S, Barco S, Heuss SC. Patients' preference regarding inpatient versus outpatient setting - A systematic review. Int J Health Plann Manage 2023; 38:1409-1419. [PMID: 37293696 DOI: 10.1002/hpm.3669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 03/09/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023] Open
Abstract
OBJECTIVES Continuously rising healthcare costs have led to financial pressure on the healthcare systems. One of the trends for the reduction of costs is the shift towards outpatient treatment. However, research has not focused on the patients' preferences regarding inpatient versus outpatient treatment settings. The purpose of this review is to examine existing studies surveying patients' preferences related to inpatient and outpatient treatment methods. The aim is to find out whether patients' wishes were queried and considered in the decision-making process. DESIGN Therefore, the reviewers performed a systematic approach utilizing the PRISMA standards and screened 1'646 articles out of 5'606 articles from the systematic search. RESULTS AND CONCLUSION The screening resulted in 4 studies that analyzed exclusively the patient's choice of treatment setting. The search showed an apparent paucity of current literature and highlights the need for further research. The authors' recommendation includes a better involvement of patients in the decision-making process as well as adding preferred treatment settings to advanced treatment directives and patient satisfaction questionnaires.
Collapse
Affiliation(s)
| | - Sarah Werthmueller
- University of Applied Sciences Northwestern Switzerland FHNW, Olten, Switzerland
| | | | - Sabina C Heuss
- University of Applied Sciences Northwestern Switzerland FHNW, Olten, Switzerland
| |
Collapse
|
14
|
Aparicio C, Acebal C, González-Vallinas M. Current approaches to develop "off-the-shelf" chimeric antigen receptor (CAR)-T cells for cancer treatment: a systematic review. Exp Hematol Oncol 2023; 12:73. [PMID: 37605218 PMCID: PMC10440917 DOI: 10.1186/s40164-023-00435-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is one of the most promising advances in cancer treatment. It is based on genetically modified T cells to express a CAR, which enables the recognition of the specific tumour antigen of interest. To date, CAR-T cell therapies approved for commercialisation are designed to treat haematological malignancies, showing impressive clinical efficacy in patients with relapsed or refractory advanced-stage tumours. However, since they all use the patient´s own T cells as starting material (i.e. autologous use), they have important limitations, including manufacturing delays, high production costs, difficulties in standardising the preparation process, and production failures due to patient T cell dysfunction. Therefore, many efforts are currently being devoted to contribute to the development of safe and effective therapies for allogeneic use, which should be designed to overcome the most important risks they entail: immune rejection and graft-versus-host disease (GvHD). This systematic review brings together the wide range of different approaches that have been studied to achieve the production of allogeneic CAR-T cell therapies and discuss the advantages and disadvantages of every strategy. The methods were classified in two major categories: those involving extra genetic modifications, in addition to CAR integration, and those relying on the selection of alternative cell sources/subpopulations for allogeneic CAR-T cell production (i.e. γδ T cells, induced pluripotent stem cells (iPSCs), umbilical cord blood T cells, memory T cells subpopulations, virus-specific T cells and cytokine-induced killer cells). We have observed that, although genetic modification of T cells is the most widely used approach, new approaches combining both methods have emerged. However, more preclinical and clinical research is needed to determine the most appropriate strategy to bring this promising antitumour therapy to the clinical setting.
Collapse
Affiliation(s)
- Cristina Aparicio
- Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), Universidad de Valladolid (UVa)-CSIC, Valladolid, Spain
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain
| | - Carlos Acebal
- Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), Universidad de Valladolid (UVa)-CSIC, Valladolid, Spain
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain
| | - Margarita González-Vallinas
- Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), Universidad de Valladolid (UVa)-CSIC, Valladolid, Spain.
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain.
| |
Collapse
|
15
|
Wudhikarn K, Tomas AA, Flynn JR, Devlin SM, Brower J, Bachanova V, Nastoupil LJ, McGuirk JP, Maziarz RT, Oluwole OO, Schuster SJ, Porter DL, Bishop MR, Riedell PA, Perales MA. Low toxicity and excellent outcomes in patients with DLBCL without residual lymphoma at the time of CD19 CAR T-cell therapy. Blood Adv 2023; 7:3192-3198. [PMID: 36355838 PMCID: PMC10338201 DOI: 10.1182/bloodadvances.2022008294] [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: 06/29/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/12/2022] Open
Abstract
CD19 chimeric antigen receptor (CAR) T-cell therapy represents a breakthrough for patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL), inducing sustained remissions in these patients. However, CAR T cells can result in significant toxicities. Preinfusion disease burden is associated with toxicities and outcomes after CAR T-cell therapy. We identified 33 patients with R/R DLBCL treated at 8 academic centers who had no detectable disease at the time of CAR T-cell therapy. The median time from leukapheresis to CAR T-cell infusion was 48 (19-193) days. Nine patients received axicabtagene ciloleucel, and 24 received tisagenlecleucel. There was no severe (grade ≥3) cytokine release syndrome, and only 1 patient developed severe neurotoxicity (grade 4). After a median follow-up of 16 months, 13 patients relapsed (39.4%) and 6 died (18.1%). One-year event-free survival and overall survival were 59.6% and 81.3%, respectively. Our findings suggest that, in patients with R/R DLBCL who have an indication for CAR T-cell therapy, treating patients in complete remission at the time of infusion is feasible, safe, and associated with favorable disease control. Further exploration in a larger clinical trial setting is warranted.
Collapse
Affiliation(s)
- Kitsada Wudhikarn
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Division of Hematology and Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ana Alarcon Tomas
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Division of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Jessica R. Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sean M. Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jamie Brower
- Blood and Marrow Transplant and Cellular Therapy Program, Abramson Cancer Center, The University of Pennsylvania, Philadelphia, PA
| | - Veronika Bachanova
- Division of Hematology, Oncology, Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Loretta J. Nastoupil
- Division of Cancer Medicine, Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Joseph P. McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, The University of Kansas, Kansas City, KS
| | - Richard T. Maziarz
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Olalekan O. Oluwole
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Stephen J. Schuster
- Blood and Marrow Transplant and Cellular Therapy Program, Abramson Cancer Center, The University of Pennsylvania, Philadelphia, PA
| | - David L. Porter
- Blood and Marrow Transplant and Cellular Therapy Program, Abramson Cancer Center, The University of Pennsylvania, Philadelphia, PA
| | - Michael R. Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Peter A. Riedell
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
16
|
Ahmed N, Wesson W, Mushtaq MU, Porter DL, Nasta SD, Brower J, Bachanova V, Hu M, Nastoupil LJ, Oluwole OO, Patel VG, Oliai C, Riedell PA, Bishop MR, Shah GL, Perales MA, Schachter L, Maziarz RT, McGuirk JP. Patient Characteristics and Outcomes of Outpatient Tisagenlecleucel Recipients for B Cell Non-Hodgkin Lymphoma. Transplant Cell Ther 2023; 29:449.e1-449.e7. [PMID: 37120134 PMCID: PMC11027185 DOI: 10.1016/j.jtct.2023.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Tisagenlecleucel (tisa-cel) is an approved CD19-directed chimeric antigen receptor T cell (CAR-T) therapy for relapsed/refractory B cell malignancies. Given potentially life-threatening toxicities, including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, inpatient tisa-cel infusion and toxicity monitoring are often considered; however, the toxicity profile of tisa-cel may be conducive to outpatient administration. Here we review the characteristics and outcomes of tisa-cel recipients treated in the outpatient setting. Patients age ≥18 years with B cell non-Hodgkin lymphoma who received tisa-cel between June 25, 2018, and January 22, 2021, at 9 US academic medical centers were included in a retrospective analysis. Six of the 9 representative centers (75%) had an outpatient program in place. A total of 157 patients were evaluable, including 93 (57%) in the outpatient treatment group and 64 (43%) in the inpatient treatment group. Baseline characteristics, toxicity and efficacy, and resource utilization were summarized. The most common lymphodepletion (LD) regimen was bendamustine in the outpatient group (65%) and fludarabine/cyclophosphamide (91%) in the inpatient group. The outpatient group had more patients with a Charlson Comorbidity Index of 0 (51% versus 15%; P < .001), fewer patients with an elevated lactate dehydrogenase (LDH) level above the normal range at the time of LD (32% versus 57%, P = .003) compared to the inpatient group, and a lower Endothelial Activation and Stress Index score (.57 versus 1.4; P < .001). Any-grade CRS and ICANS were lower in the outpatient group (29% versus 56% [P < .001] and 10% versus 16% [P = .051], respectively). Forty-two outpatient tisa-cel recipients (45%) required an unplanned admission, with a median length of stay of 5 days (range, 1 to 27 days), compared to 13 days (range, 4 to 38 days) in the inpatient group. The median number of tocilizumab doses administered was similar in the 2 groups as were the rate of intensive care unit (ICU) transfer (5% versus 8%; P = .5) and median length of ICU stay (6 days versus 5 days; P = .7). There were no toxicity-related deaths in the 30 days post-CAR-T infusion in either group. Progression-free survival and overall survival were similar in the 2 groups. With careful patient selection, outpatient tisa-cel administration is feasible and associated with similar efficacy outcomes as inpatient treatment. Outpatient toxicity monitoring and management may help optimize healthcare resource utilization.
Collapse
Affiliation(s)
- Nausheen Ahmed
- University of Kansas Medical Center, Kansas City, Kansas.
| | - William Wesson
- University of Kansas Medical Center, Kansas City, Kansas
| | | | - David L Porter
- University of Pennsylvania and Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Sunita D Nasta
- University of Pennsylvania and Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Jamie Brower
- University of Pennsylvania and Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Marie Hu
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Loretta J Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Olalekan O Oluwole
- Hematology Oncology and Stem Cell Transplant, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Vivek G Patel
- Hematology Oncology and Stem Cell Transplant, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Caspian Oliai
- Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Peter A Riedell
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, Illinois
| | - Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, Illinois
| | - Gunjan L Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Levanto Schachter
- Adult Blood and Marrow Stem Cell Transplant Program, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Richard T Maziarz
- Adult Blood and Marrow Stem Cell Transplant Program, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
17
|
Jagannath S, Joseph N, Crivera C, Kharat A, Jackson CC, Valluri S, Cost P, Phelps H, Slowik R, Klein T, Smolen L, Yu X, Cohen AD. Component Costs of CAR-T Therapy in Addition to Treatment Acquisition Costs in Patients with Multiple Myeloma. Oncol Ther 2023:10.1007/s40487-023-00228-5. [PMID: 37014590 DOI: 10.1007/s40487-023-00228-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
INTRODUCTION Ciltacabtagene autoleucel (cilta-cel), is a B-cell maturation antigen-directed, genetically modified autologous chimeric antigen receptor T-cell (CAR-T) immunotherapy. It is indicated for treatment for adult patients with relapsed or refractory multiple myeloma (RRMM) after four or more prior lines of therapy, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody. The objective of this study was to estimate the per-patient US commercial healthcare costs related to cilta-cel (CARVYKTI®) CAR-T therapy (i.e., costs separate from cilta-cel therapy acquisition) for patients with RRMM. METHODS US prescribing information for cilta-cel, publicly available data, and published literature were used with clinician input to identify the cost components and unit costs associated with administration of cilta-cel. Cost components included apheresis, bridging therapy, conditioning therapy, administration, and postinfusion monitoring for 1 year of follow-up. Adverse event (AE) management costs for all grades of cytokine release syndrome and neurologic toxicities, and additional AEs grade ≥ 3 occurring in > 5% of patients were included in the analysis. RESULTS The estimated per-patient average costs of cilta-cel CAR-T therapy administered exclusively in an inpatient setting, excluding cilta-cel therapy acquisition costs, totaled US$160,933 over a 12 month period. Costs assuming different proportions of inpatient/outpatient administration (85%/15% and 70%/30%) were US$158,095 and US$155,257, respectively. CONCLUSION Cost estimates from this analysis, which disaggregates CAR-T therapy costs, provide a comprehensive view of the cost components of CAR-T therapy that can help healthcare decision-makers make informed choices regarding the use of cilta-cel. Real-world costs may differ with improved AE prevention and mitigation strategies.
Collapse
Affiliation(s)
| | - Nedra Joseph
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
| | | | | | | | | | | | | | | | - Timothy Klein
- Medical Decision Modeling Inc., Indianapolis, IN, USA
| | - Lee Smolen
- Medical Decision Modeling Inc., Indianapolis, IN, USA
| | - Xueting Yu
- Medical Decision Modeling Inc., Indianapolis, IN, USA
| | - Adam D Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
18
|
Spanjaart AM, Pennings ER, Kos M, Mutsaers PG, Lugtenburg PJ, van Meerten T, van Doesum JA, Minnema MC, Jak M, van Dorp S, Vermaat JS, van der Poel MW, van Oijen MG, Kuipers MT, Nijhof IS, Kersten MJ. Development of a Core Set of Patient- and Caregiver-Reported Signs and Symptoms to Facilitate Early Recognition of Acute Chimeric Antigen Receptor T-Cell Therapy Toxicities. JCO Oncol Pract 2023; 19:e407-e416. [PMID: 36508702 PMCID: PMC10022884 DOI: 10.1200/op.22.00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/29/2022] [Accepted: 09/30/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Prompt recognition of acute chimeric antigen receptor T (CAR T)-cell-mediated toxicities is crucial because adequate and timely management can prevent or reverse potential life-threatening complications. In the outpatient setting, patients and informal caregivers have to recognize and report signs and symptoms marking these acute toxicities. This study provides a core set of patient- and caregiver-reported signs and symptoms (outcomes, P/CROs) and definitions of red flags warranting immediate action to include in a daily checklist for support at home, with the goal to make outpatient post-CAR T-cell care safer, optimize patient and caregiver support, and thereby facilitating an early discharge/hospital visit reduction strategy. METHODS We performed a systematic review of phase II/III trials of US Food and Drug Administration-approved CAR T-cell products and selected all common and severe adverse events that could be translated into a P/CRO for inclusion in a two-round modified Delphi procedure. Eleven CAR T-cell-dedicated hematologists from the Dutch CAR T-cell tumorboard representing all treating centers selected P/CROs for inclusion in the core set and defined red flags. The final core set was evaluated with patients and caregivers. RESULTS From nine clinical trials, 457 adverse events were identified of which 42 could be used as P/CRO. The final core set contains 28 items, including five signs for measurement via wearables and two signs for caregiver-performed assessments. CONCLUSION This study provides a core set of P/CROs that can serve as a framework for (eHealth) tools that aim to enable patients and caregivers to more effectively recognize and report signs and symptoms of acute toxicities after CAR T-cell therapy, which will enhance safe outpatient treatment monitoring.
Collapse
Affiliation(s)
- Anne M. Spanjaart
- Amsterdam UMC Location University of Amsterdam, Hematology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- LYMMCARE, Amsterdam, the Netherlands
| | - Elise R.A. Pennings
- Amsterdam UMC Location University of Amsterdam, Hematology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- LYMMCARE, Amsterdam, the Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Milan Kos
- Amsterdam University Medical Centers, University of Amsterdam, Department of Oncology, Amsterdam, the Netherlands
| | - Pim G.N.J. Mutsaers
- Erasmus MC Cancer Institute, University Medical Center Rotterdam, Department of Hematology, Rotterdam, the Netherlands
| | - Pieternella J. Lugtenburg
- Erasmus MC Cancer Institute, University Medical Center Rotterdam, Department of Hematology, Rotterdam, the Netherlands
| | - Tom van Meerten
- University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands
| | - Jaap A. van Doesum
- University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands
| | - Monique C. Minnema
- University Medical Center Utrecht, Department of Hematology, Utrecht, the Netherlands
| | - Margot Jak
- University Medical Center Utrecht, Department of Hematology, Utrecht, the Netherlands
| | - Suzanne van Dorp
- Radboud University Medical Center, Department of Hematology, Nijmegen, the Netherlands
| | - Joost S.P. Vermaat
- Leiden University Medical Center, Department of Hematology, Leiden, the Netherlands
| | - Marjolein W.M. van der Poel
- Department of Internal Medicine, Division of Hematology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Martijn G.H. van Oijen
- Amsterdam University Medical Centers, University of Amsterdam, Department of Oncology, Amsterdam, the Netherlands
| | - Maria T. Kuipers
- Amsterdam UMC Location University of Amsterdam, Hematology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- LYMMCARE, Amsterdam, the Netherlands
| | - Inger S. Nijhof
- Amsterdam UMC Location University of Amsterdam, Hematology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- LYMMCARE, Amsterdam, the Netherlands
- St Antonius Ziekenhuis Nieuwegein, Hematology, Nieuwegein, the Netherlands
| | - Marie José Kersten
- Amsterdam UMC Location University of Amsterdam, Hematology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- LYMMCARE, Amsterdam, the Netherlands
| |
Collapse
|
19
|
Wang H, Tsao ST, Gu M, Fu C, He F, Li X, Zhang M, Li N, Hu HM. A simple and effective method to purify and activate T cells for successful generation of chimeric antigen receptor T (CAR-T) cells from patients with high monocyte count. J Transl Med 2022; 20:608. [PMID: 36536403 PMCID: PMC9764707 DOI: 10.1186/s12967-022-03833-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor T (CAR-T) cells are genetically modified T cells with redirected specificity and potent T-cell-mediated cytotoxicity toward malignant cells. Despite several CAR-T products being approved and commercialized in the USA, Europe, and China, CAR-T products still require additional optimization to ensure reproducible and cost-effective manufacture. Here, we investigated the critical parameters in the CD3+ T-cell isolation process that significantly impacted CAR-T manufacturing's success. METHODS CAR-T cells were prepared from cryopreserved peripheral blood mononuclear cells (PBMC). The thawed PBMC was rested overnight before the CD3+ T cell isolation process using CTS™ Dynabeads™ CD3/CD28. Different isolation media, cell-bead co-incubation time, and cell density were examined in this study. Activated CD3+ T cells were transduced with a gamma retroviral vector carrying the CD19 or BCMA CAR sequence. The CAR-T cells proliferated in a culture medium supplemented with interleukin 2 (IL-2). RESULTS CD14+ monocytes hindered T-cell isolation when X-VIVO 15 basic medium was used as the selection buffer. The activation of T cells was blocked because monocytes actively engulfed CD3/28 beads. In contrast, when DPBS was the selection medium, the T-cell isolation and activation were no longer blocked, even in patients whose PBMC contained abnormally high CD14+ monocytes and a low level of CD3+ T cells. CONCLUSIONS In this study, we discovered that selecting CD3+ T-cell isolation media is critical for improving T-cell activation, transduction, and CAR-T proliferation. Using DPBS as a CD3+ T cell isolation buffer significantly improved the success rate and shortened the duration of CAR-T production. The optimized process has been successfully applied in our ongoing clinical trials. Trial registration NCT03798509: Human CD19 Targeted T Cells Injection Therapy for Relapsed and Refractory CD19-positive Leukemia. Date of registration: January 10, 2019. NCT03720457: Human CD19 Targeted T Cells Injection (CD19 CAR-T) Therapy for Relapsed and Refractory CD19-positive Lymphoma. Date of registration: October 25, 2018. NCT04003168: Human BCMA Targeted T Cells Injection Therapy for BCMA-positive Relapsed/Refractory Multiple Myeloma. Date of registration: July 1, 2019.
Collapse
Affiliation(s)
- Haiying Wang
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Shih-Ting Tsao
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Mingyuan Gu
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Chengbing Fu
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Feng He
- Department of Manufacturing, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Xiu Li
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Mian Zhang
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Na Li
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| | - Hong-Ming Hu
- Department of Research and Development, Hrain Biotechnology Co., Ltd., 9th Floor, Building 1, 1238 Zhangjiang Road, Pudong New District, Shanghai, China
| |
Collapse
|
20
|
Mikhael J, Fowler J, Shah N. Chimeric Antigen Receptor T-Cell Therapies: Barriers and Solutions to Access. JCO Oncol Pract 2022; 18:800-807. [PMID: 36130152 DOI: 10.1200/op.22.00315] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies are relatively new treatments for patients with heavily pretreated hematologic malignancies. Although these innovative therapies can offer substantial benefit to patients with limited alternative treatment options, patient-access barriers exist. Conventional clinical trials are time-consuming and may be limited by strict patient eligibility criteria, resources, and availability of enrollment slots. Because of the complexity of the CAR-T administration process, treatment delivery can be associated with additional burden for the patient, including requiring patients to reside close to treatment centers and remain with a caregiver after infusion. Manufacturing of CAR-T cells is completed in specialized facilities and depends on the availability of reagents, manufacturing workforce, and timely transportation. CAR-T therapy is costly, and many US health plans restrict coverage of cell and gene therapies. Several of the existing challenges because of these barriers have been exacerbated during the COVID-19 pandemic. This review discusses these barriers and proposes some potential solutions to improving patient access, including innovation in clinical trial design and manufacturing, location of treatment delivery, and key stakeholder opinions regarding treatment and reimbursement. We propose a call to action for key stakeholder groups to address these barriers to CAR-T therapy to expand treatment access for patients. Future collaboration between key stakeholders, including payers, regulatory agencies, and industry/academia, will be critical to continue to address these barriers and enhance patient access to these therapies.
Collapse
Affiliation(s)
- Joseph Mikhael
- Translational Genomics Research Institute (TGen), Applied Cancer Research and Drug Discovery Division, Phoenix, AZ
| | | | - Nina Shah
- Bone Marrow Transplantation and Hematologic Malignancy Unit, Division of Hematology-Oncology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
21
|
Banerjee R, Lee SS, Cowan AJ. Innovation in BCMA CAR-T therapy: Building beyond the Model T. Front Oncol 2022; 12:1070353. [PMID: 36505779 PMCID: PMC9729952 DOI: 10.3389/fonc.2022.1070353] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Autologous chimeric antigen receptor T-cell (CAR-T) therapies targeting B-cell maturation antigen (BCMA) have revolutionized the field of multiple myeloma in the same way that the Ford Model T revolutionized the original CAR world a century ago. However, we are only beginning to understand how to improve the efficacy and usability of these cellular therapies. In this review, we explore three automotive analogies for innovation with BCMA CAR-T therapies: stronger engines, better mileage, and hassle-free delivery. Firstly, we can build stronger engines in terms of BCMA targeting: improved antigen binding, tools to modulate antigen density, and armoring to better reach the antigen itself. Secondly, we can improve "mileage" in terms of response durability through ex vivo CAR design and in vivo immune manipulation. Thirdly, we can implement hassle-free delivery through rapid manufacturing protocols and off-the-shelf products. Just as the Model T set a benchmark for car manufacturing over 100 years ago, idecabtagene vicleucel and ciltacabtagene autoleucel have now set the starting point for BCMA CAR-T therapy with their approvals. As with any emerging technology, whether automotive or cellular, the best in innovation and optimization is yet to come.
Collapse
Affiliation(s)
- Rahul Banerjee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sarah S. Lee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Andrew J. Cowan
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
22
|
Real-World Experience and Optimization of Outpatient Chimeric Antigen Receptor T Cell Therapy. Transplant Cell Ther 2022; 28:583-585. [DOI: 10.1016/j.jtct.2022.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/21/2022]
|
23
|
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
|
24
|
Atrash S. Between two kingdoms (CAR-T and hematopoietic stem cell transplantation). Transplant Cell Ther 2022; 28:413-414. [DOI: 10.1016/j.jtct.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
25
|
Nasta SD, Hughes ME, Namoglu EC, Garfall A, DiFilippo H, Ballard HJ, Barta SK, Chong EA, Frey NV, Gerson JN, Landsburg DJ, Ruella M, Schuster SJ, Svoboda J, Weber E, Porter DL. Outcomes of Tisagenlecleucel in Lymphoma Patients With Predominant Management in an Ambulatory Setting. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e730-e737. [PMID: 35595619 PMCID: PMC10965010 DOI: 10.1016/j.clml.2022.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/18/2022] [Accepted: 04/03/2022] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Chimeric antigen receptor T-cell therapy (CAR T) is a revolutionary adoptive immunotherapy approach in lymphoma; however, substantial resources are necessary for administration and care of these patients. Our institution has administered tisagenlecleucel primarily in an outpatient setting, and here we report our clinical outcomes. PATIENTS AND METHODS We conducted a single institution, retrospective study investigating outcomes of adult lymphoma patients treated with commercial tisagenlecleucel between 10/2017 and 12/2020. We analyzed patient characteristics and outcomes of efficacy and safety including overall response rate, progression-free survival, overall survival and cytokine-release syndrome, neurotoxicity, and hospitalizations. RESULTS Seventy-two patients with relapsed or refractory non-Hodgkin lymphoma (NHL) who received commercial tisagenlecleucel were identified; 68 (94.4%) patients received outpatient tisagenlecleucel. The overall response rate was 43% with a complete response observed in 25 patients (34.7%). At a median follow-up of 9.1 months, the median progression-free survival was 3.3 months. Grade 3-4 cytokine release syndrome was not observed in the study group and two patients had grade 3-4 neurotoxicity. Twenty-six patients (36.1%) were admitted within 30 days after infusion with a median length of stay of 5 days. Fourteen patients (19.4%) were admitted within 72 hours of infusion. No patient died of CAR T cell-related toxicity. CONCLUSION Our experience affirms treatment with tisagenlecleucel in the outpatient setting is safe and feasible with close supervision and adequate institutional experience. After infusion, adverse events were manageable and the majority of patients did not require hospitalization.
Collapse
Affiliation(s)
- Sunita D Nasta
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA.
| | - Mitchell E Hughes
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Esin C Namoglu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Alfred Garfall
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Heather DiFilippo
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Hatcher J Ballard
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Stefan K Barta
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Elise A Chong
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Noelle V Frey
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - James N Gerson
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | | | - Marco Ruella
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | | | - Jakub Svoboda
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth Weber
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - David L Porter
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
26
|
Borogovac A, Keruakous A, Bycko M, Holter Chakrabarty J, Ibrahimi S, Khawandanah M, Selby GB, Yuen C, Schmidt S, Autry MT, Al-Juhaishi T, Wieduwilt MJ, Asch AS. Safety and feasibility of outpatient chimeric antigen receptor (CAR) T-cell therapy: experience from a tertiary care center. Bone Marrow Transplant 2022; 57:1025-1027. [PMID: 35411106 PMCID: PMC8995917 DOI: 10.1038/s41409-022-01664-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/19/2022] [Accepted: 03/25/2022] [Indexed: 12/21/2022]
Affiliation(s)
- Azra Borogovac
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Amany Keruakous
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Hematology-Oncology Section, Department of Medicine, University of Augusta, Augusta, GA, USA
| | - Michelle Bycko
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jennifer Holter Chakrabarty
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sami Ibrahimi
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mohamad Khawandanah
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - George B Selby
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Carrie Yuen
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sarah Schmidt
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Marcus T Autry
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Taha Al-Juhaishi
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matthew J Wieduwilt
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Adam S Asch
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
27
|
Hoda D, Richards R, Faber EA, Deol A, Hunter BD, Weber E, DiFilippo H, Henderson-Clark T, Meaux L, Crivera C, Riccobono C, Garrett A, Jackson CC, Fowler J, Theocharous P, Stewart R, Lorden AL, Porter DL, Berger A. Process, resource and success factors associated with chimeric antigen receptor T-cell therapy for multiple myeloma. Future Oncol 2022; 18:2415-2431. [PMID: 35583358 DOI: 10.2217/fon-2022-0162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Chimeric antigen receptor T-cell (CAR-T) therapy represents a new frontier in multiple myeloma. It is important to understand critical success factors (CSFs) that may optimize its use in this therapeutic area. Methods: We estimated the CAR-T process using time-driven activity-based costing. Information was obtained through interviews at four US oncology centers and with payer representatives, and through publicly available data. Results: The CAR-T process comprises 13 steps which take 177 days; it was estimated to include 46 professionals and ten care settings. CSFs included proactive collaboration, streamlined reimbursement and CAR-T administration in alternative settings when possible. Implementing CSFs may reduce episode time and costs by 14.4 and 13.2%, respectively. Conclusion: Our research provides a blueprint for improving efficiencies in CAR-T therapy, thereby increasing its sustainability for multiple myeloma.
Collapse
Affiliation(s)
- Daanish Hoda
- Intermountain Healthcare, Salt Lake City, UT, USA
| | - Robert Richards
- Cell Therapy & Transplant Program, Division of Hematology-Oncology & Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward A Faber
- Transplant & Cellular Therapy Program, Oncology/Hematology Care, USA.,Adult BMT & Cellular Therapy Program, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, USA
| | - Abhinav Deol
- Karmanos Cancer Center, 4100 John R St, Detroit, MI 48201, USA
| | | | - Elizabeth Weber
- Cell Therapy & Transplant Program, Division of Hematology-Oncology & Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Heather DiFilippo
- Cell Therapy & Transplant Program, Division of Hematology-Oncology & Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Linda Meaux
- Intermountain Healthcare, Salt Lake City, UT, USA
| | - Concetta Crivera
- Janssen Scientific Affairs, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ 08560, USA
| | - Carrie Riccobono
- US Medical Affairs, Legend Biotech, 2101 Cottontail Lane Somerset, NJ 08873, USA
| | - Ashraf Garrett
- US Medical Affairs, Legend Biotech, 2101 Cottontail Lane Somerset, NJ 08873, USA
| | - Carolyn C Jackson
- Janssen Pharmaceutical Research & Development, 1125 Trenton-Harbourton Road, Titusville, NJ 08560, USA
| | - Jessica Fowler
- Janssen Scientific Affairs, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ 08560, USA
| | | | - Raj Stewart
- Evidera
- PPD, 7101 Wisconsin AvenueSuite 1400Bethesda, MD 20814, USA
| | - Andrea L Lorden
- Evidera
- PPD, 7101 Wisconsin AvenueSuite 1400Bethesda, MD 20814, USA
| | - David L Porter
- Cell Therapy & Transplant Program, Division of Hematology-Oncology & Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ariel Berger
- Evidera
- PPD, 7101 Wisconsin AvenueSuite 1400Bethesda, MD 20814, USA
| |
Collapse
|
28
|
Soldierer M, Bister A, Haist C, Thivakaran A, Cengiz SC, Sendker S, Bartels N, Thomitzek A, Smorra D, Hejazi M, Uhrberg M, Scheckenbach K, Monzel C, Wiek C, Reinhardt D, Niktoreh N, Hanenberg H. Genetic Engineering and Enrichment of Human NK Cells for CAR-Enhanced Immunotherapy of Hematological Malignancies. Front Immunol 2022; 13:847008. [PMID: 35464442 PMCID: PMC9022481 DOI: 10.3389/fimmu.2022.847008] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 01/11/2023] Open
Abstract
The great clinical success of chimeric antigen receptor (CAR) T cells has unlocked new levels of immunotherapy for hematological malignancies. Genetically modifying natural killer (NK) cells as alternative CAR immune effector cells is also highly promising, as NK cells can be transplanted across HLA barriers without causing graft-versus-host disease. Therefore, off-the-shelf usage of CAR NK cell products might allow to widely expand the clinical indications and to limit the costs of treatment per patient. However, in contrast to T cells, manufacturing suitable CAR NK cell products is challenging, as standard techniques for genetically engineering NK cells are still being defined. In this study, we have established optimal lentiviral transduction of primary human NK cells by systematically testing different internal promoters for lentiviral CAR vectors and comparing lentiviral pseudotypes and viral entry enhancers. We have additionally modified CAR constructs recognizing standard target antigens for acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) therapy—CD19, CD33, and CD123—to harbor a CD34-derived hinge region that allows efficient detection of transduced NK cells in vitro and in vivo and also facilitates CD34 microbead-assisted selection of CAR NK cell products to >95% purity for potential clinical usage. Importantly, as most leukemic blasts are a priori immunogenic for activated primary human NK cells, we developed an in vitro system that blocks the activating receptors NKG2D, DNAM-1, NKp30, NKp44, NKp46, and NKp80 on these cells and therefore allows systematic testing of the specific killing of CAR NK cells against ALL and AML cell lines and primary AML blasts. Finally, we evaluated in an ALL xenotransplantation model in NOD/SCID-gamma (NSG) mice whether human CD19 CAR NK cells directed against the CD19+ blasts are relying on soluble or membrane-bound IL15 production for NK cell persistence and also in vivo leukemia control. Hence, our study provides important insights into the generation of pure and highly active allogeneic CAR NK cells, thereby advancing adoptive cellular immunotherapy with CAR NK cells for human malignancies further.
Collapse
Affiliation(s)
- Maren Soldierer
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Arthur Bister
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Corinna Haist
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Aniththa Thivakaran
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sevgi Can Cengiz
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephanie Sendker
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nina Bartels
- Department of Experimental Medical Physics, Heinrich Heine University, Düsseldorf, Germany
| | - Antonia Thomitzek
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Denise Smorra
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Maryam Hejazi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Kathrin Scheckenbach
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Cornelia Monzel
- Department of Experimental Medical Physics, Heinrich Heine University, Düsseldorf, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Dirk Reinhardt
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Naghmeh Niktoreh
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Helmut Hanenberg
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
29
|
Fruhwirth GO, Weigelin B, Daldrup-Link HE, Ponomarev V. Editorial to the Special Issue Entitled "Imaging in Immunooncology". Mol Imaging Biol 2022; 24:177-180. [PMID: 35303206 PMCID: PMC8983534 DOI: 10.1007/s11307-022-01719-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Gilbert O. Fruhwirth
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King’s College London, London, UK
| | - Bettina Weigelin
- Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies,”, University of Tuebingen, Tübingen, Germany
| | | | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| |
Collapse
|
30
|
ABCs of ADCs in Management of Relapsed/refractory diffuse large B-cell lymphoma. Blood Rev 2022; 56:100967. [DOI: 10.1016/j.blre.2022.100967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 11/19/2022]
|
31
|
Zhang C, Fang L, Wang X, Yuan S, Li W, Tian W, Chen J, Zhang Q, Zhang Y, Zhang Q, Zheng J. Oncolytic adenovirus-mediated expression of decorin facilitates CAIX-targeting CAR-T therapy against renal cell carcinoma. Mol Ther Oncolytics 2022; 24:14-25. [PMID: 34977339 PMCID: PMC8688951 DOI: 10.1016/j.omto.2021.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/27/2021] [Indexed: 12/12/2022] Open
Abstract
Although chimeric antigen receptor T cell (CAR-T) therapy has been successful for hematological malignancies, it is less effective for solid tumors. The primary reason is that the immune microenvironment restricts CAR-T cells from infiltrating and proliferating in tumors. Oncolytic virotherapy has emerged as a novel immunogenic therapy to augment antitumor immune response. Here we combined an oncolytic adenovirus carrying decorin with a CAR-T targeting carbonic anhydrase IX (CAIX) to perform the antitumor activity for renal cancer cells. We found that OAV-Decorin combined with CAIX-CAR-T exhibited significantly reduced tumor burden, altered the composition of extracellular matrix (ECM) by inhibiting the distribution of collagen fibers, decreased the expression of TGF-β in tumor cells, enhanced IFN-γ secretion, and obtained higher numbers of CAR-T cells. The combination treatment modality showed prolonged mice survival. The intratumoral injection of OAV-Decorin into tumor-bearing immunocompetent mice activated the inflammatory immune status and resulted in tumor regression. These data supported further investigation of the combination of OAV-Decorin and CAIX-CAR-T cells in solid tumors.
Collapse
Affiliation(s)
- Chen Zhang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China.,Department of Oncology, The First People's Hospital of Yancheng, Yancheng 224001 Jiangsu, China
| | - Lin Fang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Xueyan Wang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China
| | - Sen Yuan
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China
| | - Wanjing Li
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China
| | - Weiping Tian
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China
| | - Jing Chen
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Qi Zhang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China
| | - Qing Zhang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Junnian Zheng
- Jiangsu Key Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| |
Collapse
|
32
|
Hong Y, Kim IS. The therapeutic potential of immune cell-derived exosomes as an alternative to adoptive cell transfer. BMB Rep 2022. [PMCID: PMC8810551 DOI: 10.5483/bmbrep.2022.55.1.075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Yeonsun Hong
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|
33
|
Bhaskar ST, Dholaria BR, Sengsayadeth SM, Savani BN, Oluwole OO. Role of bridging therapy during chimeric antigen receptor T cell therapy. EJHAEM 2022; 3:39-45. [PMID: 35844303 PMCID: PMC9175845 DOI: 10.1002/jha2.335] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 06/15/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has been approved for use in several relapsed/refractory hematologic malignancies and has significantly improved outcomes for these diseases. A number of different CAR T products are now being used in clinical practice and have demonstrated excellent outcomes to those in clinical trials. However, increased real-world use of CAR T therapy has uncovered a number of barriers that can lead to significant delays in treatment. As a result, bridging therapy has become a widely used tool to stabilize or debulk disease between leukapheresis and CAR T cell administration. Here we review the available data regarding bridging therapy, with a focus on patient selection, choice of therapy, timing of therapy, and potential pitfalls.
Collapse
Affiliation(s)
- Shakthi T. Bhaskar
- Division of Hematology/OncologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Bhagirathbhai R. Dholaria
- Division of Hematology/OncologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Salyka M. Sengsayadeth
- Division of Hematology/OncologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Bipin N. Savani
- Division of Hematology/OncologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Olalekan O. Oluwole
- Division of Hematology/OncologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
34
|
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
|
35
|
Grunewald CM, Haist C, König C, Petzsch P, Bister A, Nößner E, Wiek C, Scheckenbach K, Köhrer K, Niegisch G, Hanenberg H, Hoffmann MJ. Epigenetic Priming of Bladder Cancer Cells With Decitabine Increases Cytotoxicity of Human EGFR and CD44v6 CAR Engineered T-Cells. Front Immunol 2021; 12:782448. [PMID: 34868059 PMCID: PMC8637820 DOI: 10.3389/fimmu.2021.782448] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/26/2021] [Indexed: 11/18/2022] Open
Abstract
Background Treatment of B-cell malignancies with CD19-directed chimeric antigen receptor (CAR) T-cells marked a new era in immunotherapy, which yet has to be successfully adopted to solid cancers. Epigenetic inhibitors of DNA methyltransferases (DNMTi) and histone deacetylases (HDACi) can induce broad changes in gene expression of malignant cells, thus making these inhibitors interesting combination partners for immunotherapeutic approaches. Methods Urothelial carcinoma cell lines (UCC) and benign uroepithelial HBLAK cells pretreated with the DNMTi decitabine or the HDACi romidepsin were co-incubated with CAR T-cells directed against EGFR or CD44v6, and subsequent cytotoxicity assays were performed. Effects on T-cell cytotoxicity and surface antigen expression on UCC were determined by flow cytometry. We also performed next-generation mRNA sequencing of inhibitor-treated UCC and siRNA-mediated knockdown of potential regulators of CAR T-cell killing. Results Exposure to decitabine but not romidepsin enhanced CAR T-cell cytotoxicity towards all UCC lines, but not towards the benign HBLAK cells. Increased killing could neither be attributed to enhanced target antigen expression (EGFR and CD44v6) nor fully explained by changes in the T-cell ligands PD-L1, PD-L2, ICAM-1, or CD95. Instead, gene expression analysis suggested that regulators of cell survival and apoptosis were differentially induced by the treatment. Decitabine altered the balance between survival and apoptosis factors towards an apoptosis-sensitive state associated with increased CAR T-cell killing, while romidepsin, at least partially, tilted this balance in the opposite direction. Knockdown experiments with siRNA in UCC confirmed BID and BCL2L1/BCLX as two key factors for the altered susceptibility of the UCC. Conclusion Our data suggest that the combination of decitabine with CAR T-cell therapy is an attractive novel therapeutic approach to enhance tumor-specific killing of bladder cancer. Since BID and BCL2L1 are essential determinants for the susceptibility of a wide variety of malignant cells, their targeting might be additionally suitable for combination with immunotherapies, e.g., CAR T-cells or checkpoint inhibitors in other malignancies.
Collapse
Affiliation(s)
- Camilla M Grunewald
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Corinna Haist
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carolin König
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Arthur Bister
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Elfriede Nößner
- Immunoanalytics: Tissue Control of Immunocytes, German Research Center for Environmental Health, Helmholtz Zentrum München, Munich, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Kathrin Scheckenbach
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Günter Niegisch
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Helmut Hanenberg
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michèle J Hoffmann
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
36
|
Abstract
Cellular therapies such as allogeneic hematopoietic stem cell transplantation (HSCT) and immune-effector cell therapy (IECT) continue to have a critical role in the treatment of patients with high risk malignancies and hematologic conditions. These therapies are also associated with inflammatory conditions such as graft-versus-host disease (GVHD) and cytokine release syndrome (CRS) which contribute significantly to the morbidity and mortality associated with these therapies. Recent advances in our understanding of the immunological mechanisms that underly GVHD and CRS highlight an important role for Janus kinases (JAK). JAK pathways are important for the signaling of several cytokines and are involved in the activation and proliferation of several immune cell subsets. In this review, we provide an overview of the preclinical and clinical evidence supporting the use of JAK inhibitors for acute and chronic GVHD and CRS.
Collapse
Affiliation(s)
- Amer Assal
- Department of Medicine, Bone Marrow Transplantation and Cell Therapy Program, Columbia University Irving Medical Center, New York, NY, United States
| | - Markus Y. Mapara
- Department of Medicine, Bone Marrow Transplantation and Cell Therapy Program, Columbia University Irving Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University, New York, NY, United States
| |
Collapse
|
37
|
Woller N, Engelskircher SA, Wirth T, Wedemeyer H. Prospects and Challenges for T Cell-Based Therapies of HCC. Cells 2021; 10:cells10071651. [PMID: 34209393 PMCID: PMC8304292 DOI: 10.3390/cells10071651] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
The scope of therapeutic options for the treatment of hepatocellular carcinoma (HCC) has recently been expanded by immunotherapeutic regimens. T cell-based therapies, especially in combination with other treatments have achieved far better outcomes compared to conventional treatments alone. However, there is an emerging body of evidence that eliciting T cell responses in immunotherapeutic approaches is insufficient for favorable outcomes. Immune responses in HCC are frequently attenuated in the tumor microenvironment (TME) or may even support tumor progress. Hence, therapies with immune checkpoint inhibitors or adoptive cell therapies appear to necessitate additional modification of the TME to unlock their full potential. In this review, we focus on immunotherapeutic strategies, underlying molecular mechanisms of CD8 T cell immunity, and causes of treatment failure in HCC of viral and non-viral origin. Furthermore, we provide an overview of TME features in underlying etiologies of HCC patients that mediate therapy resistance to checkpoint inhibition and discuss strategies from the literature concerning current approaches to these challenges.
Collapse
Affiliation(s)
- Norman Woller
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Sophie Anna Engelskircher
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Thomas Wirth
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Heiner Wedemeyer
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|