1
|
Trautmann T, Yakobian N, Nguyen R. CAR T-cells for pediatric solid tumors: where to go from here? Cancer Metastasis Rev 2024; 43:1445-1461. [PMID: 39317919 DOI: 10.1007/s10555-024-10214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Despite the great success that chimeric antigen receptor (CAR) T-cells have had in patients with B-cell malignancies and multiple myeloma, they continue to have limited efficacy against most solid tumors. Especially in the pediatric population, pre- and post-treatment biopsies are rarely performed due to ethical reasons, and thus, our understanding is still very limited regarding the mechanisms in the tumor microenvironment by which tumor cells exclude effectors and attract immune-suppressive cells. Nevertheless, based on the principles that are known, current T-cell engineering has leveraged some of these processes and created more potent CAR T-cells. The recent discovery of new oncofetal antigens and progress made in CAR design have expanded the potential pool of candidate antigens for therapeutic development. The most promising approaches to enhance CAR T-cells are novel CAR gating strategies, creative ways of cytokine delivery to the TME without enhancing systemic toxicity, and hijacking the chemokine axis of tumors for migratory purposes. With these new modifications, the next step in the era of CAR T-cell development will be the clinical validation of these promising preclinical findings.
Collapse
Affiliation(s)
- Tina Trautmann
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Natalia Yakobian
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Rosa Nguyen
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA.
| |
Collapse
|
2
|
Volkmann ER, Varga J, Blazar BR, Pavletic SZ. Challenges and solutions for cellular therapy development in autoimmune diseases. THE LANCET. RHEUMATOLOGY 2024; 6:e740-e743. [PMID: 39332423 DOI: 10.1016/s2665-9913(24)00274-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024]
Affiliation(s)
- Elizabeth R Volkmann
- Department of Medicine, Division of Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - John Varga
- Department of Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN, USA
| | - Steven Z Pavletic
- Immune Deficiency and Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, US National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
Kim TM, Taszner M, Novelli S, Cho SG, Villasboas JC, Merli M, Jiménez-Ubieto A, Tessoulin B, Poon LM, Tucker D, Walewski J, Yi S, Song Y, Chong G, Bachy E, Guidez S, Alonso A, Jagadeesh D, Zhang W, Magnano L, Iskierka-Jażdżewska E, Tani M, Shen B, Uppala A, Zhu M, Shariff S, Brouwer-Visser J, Chaudhry A, Mohamed H, Ambati S, Luminari S. Safety and efficacy of odronextamab in patients with relapsed or refractory follicular lymphoma. Ann Oncol 2024; 35:1039-1047. [PMID: 39147364 DOI: 10.1016/j.annonc.2024.08.2239] [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: 07/05/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Odronextamab, a CD20×CD3 bispecific antibody that engages cytotoxic T cells to destroy malignant B cells, has demonstrated encouraging activity across multiple subtypes of relapsed/refractory (R/R) B-cell non-Hodgkin lymphoma. PATIENTS AND METHODS This phase II study (ELM-2; NCT03888105) evaluated odronextamab in patients with R/R follicular lymphoma after two or more lines of systemic therapy. Patients received intravenous odronextamab in 21-day cycles, with step-up dosing in cycle 1 to help mitigate the risk of cytokine release syndrome, until disease progression or unacceptable toxicity. The primary endpoint was objective response rate by independent central review. RESULTS Among 128 patients evaluated, 95% completed cycle 1, and 85% completed four or more cycles. At 20.1 months' efficacy follow-up, objective response rate was 80.0% and complete response rate was 73.4%. Median duration of complete response was 25.1 months. Median progression-free survival was 20.7 months, and median overall survival was not reached. Discontinuation of odronextamab due to adverse events occurred in 16% of patients. The most common treatment-emergent adverse events were cytokine release syndrome [56%; grade ≥3 1.7% (1/60) with 0.7/4/20 mg step-up], neutropenia (39%), and pyrexia (38%). CONCLUSIONS Odronextamab achieved high complete response rates with generally manageable safety in patients with heavily pretreated R/R follicular lymphoma.
Collapse
MESH Headings
- Humans
- Lymphoma, Follicular/drug therapy
- Lymphoma, Follicular/pathology
- Lymphoma, Follicular/mortality
- Male
- Middle Aged
- Female
- Aged
- Adult
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/adverse effects
- Aged, 80 and over
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Progression-Free Survival
- Drug Resistance, Neoplasm/drug effects
Collapse
Affiliation(s)
- T M Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea.
| | - M Taszner
- Department of Haematology and Transplantology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - S Novelli
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - S-G Cho
- Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | | | - M Merli
- Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - A Jiménez-Ubieto
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - B Tessoulin
- Hematology Department, Nantes University Hospital, Nantes, France
| | - L M Poon
- Hematology Oncology National University Hospital, Singapore, Singapore
| | - D Tucker
- Department of Hematology, Royal Cornwall Hospital, Truro, UK
| | - J Walewski
- Narodowy Instytut Onkologii im. Marii Skłodowskiej-Curie Państwowy Instytut Badawczy, Warsaw, Poland
| | - S Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Y Song
- Department of Lymphoma, Peking University Cancer Hospital & Institute (Beijing Cancer Hospital), Beijing, China
| | - G Chong
- Department of Medical Oncology, and Clinical Haematology, Olivia Newton-John Cancer Centre, Heidelberg, Australia
| | - E Bachy
- Department of Hematology, Lyon Sud Hospital, Lyon, France; Lymphoma Immuno-Biology (LIB) team, Inserm U1111, CIRI, Lyon, France
| | - S Guidez
- Centre Hospitalier Universitaire (CHU) de Poitiers, Poitiers, France
| | - A Alonso
- Hospital Universitario Quiron Salud Madrid, Madrid, Spain
| | - D Jagadeesh
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, USA
| | - W Zhang
- Hematology Department, Peking Union Medical College Hospital, Beijing, China
| | - L Magnano
- Hematology Department, Hospital Clínic of Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Iskierka-Jażdżewska
- Copernicus Memorial Hospital, Department of General Hematology, Medical University of Łódź, Łódź, Poland
| | - M Tani
- Hematology Unit, Santa Maria delle Croci Hospital, Ravenna, Italy
| | - B Shen
- Regeneron Pharmaceuticals, Inc., Tarrytown, USA
| | - A Uppala
- Regeneron Pharmaceuticals, Inc., Tarrytown, USA
| | - M Zhu
- Regeneron Pharmaceuticals, Inc., Tarrytown, USA
| | | | | | - A Chaudhry
- Regeneron Pharmaceuticals, Inc., Tarrytown, USA
| | - H Mohamed
- Regeneron Pharmaceuticals, Inc., Tarrytown, USA
| | - S Ambati
- Regeneron Pharmaceuticals, Inc., Tarrytown, USA
| | - S Luminari
- Division of Hematology, Azienda Unità Sanitaria Locale-IRCCS, Reggio Emilia, Italy
| |
Collapse
|
4
|
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
|
5
|
Phillips TJ, Carlo-Stella C, Morschhauser F, Bachy E, Crump M, Trněný M, Bartlett NL, Zaucha J, Wrobel T, Offner F, Humphrey K, Relf J, Filézac de L'Etang A, Carlile DJ, Byrne B, Qayum N, Lundberg L, Dickinson M. Glofitamab in Relapsed/Refractory Mantle Cell Lymphoma: Results From a Phase I/II Study. J Clin Oncol 2024:JCO2302470. [PMID: 39365960 DOI: 10.1200/jco.23.02470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/25/2024] [Accepted: 08/21/2024] [Indexed: 10/06/2024] Open
Abstract
PURPOSE Patients with relapsed/refractory (R/R) mantle cell lymphoma (MCL) have a poor prognosis. The phase I/II NP30179 study (ClinicalTrials.gov identifier: NCT03075696) evaluated glofitamab monotherapy in patients with R/R B-cell lymphomas, with obinutuzumab pretreatment (Gpt) to mitigate the risk of cytokine release syndrome (CRS) with glofitamab. We present data for patients with R/R MCL. METHODS Eligible patients with R/R MCL (at least one previous therapy) received Gpt (1,000 or 2,000 mg) 7 days before the first glofitamab dose (single dose or split over 2 days if required). Glofitamab step-up dosing was administered once a day on days 8 (2.5 mg) and 15 (10 mg) of cycle 1, with a target dose of 16 or 30 mg once every 3 weeks from cycle 2 day 1 onward, for 12 cycles. Efficacy end points included investigator-assessed complete response (CR) rate, overall response rate (ORR), and duration of CR. RESULTS Of 61 enrolled patients, 60 were evaluable for safety and efficacy. Patients had received a median of two previous therapies (range, 1-5). CR rate and ORR were 78.3% (95% CI, 65.8 to 87.9) and 85.0% (95% CI, 73.4 to 92.9), respectively. In patients who had received previous treatment with a Bruton tyrosine kinase inhibitor (n = 31), CR rate was 71.0% (95% CI, 52.0 to 85.8) and ORR was 74.2% (95% CI, 55.4 to 88.1). CRS after glofitamab administration occurred in 70.0% of patients, with a lower incidence in the 2,000 mg (63.6% [grade ≥2, 22.7%]) versus 1,000 mg (87.5%; grade ≥2, 62.5%) Gpt cohort. Four adverse events led to glofitamab withdrawal (all infections). CONCLUSION Fixed-duration glofitamab induced high CR rates in heavily pretreated patients with R/R MCL; the safety profile was manageable with appropriate support.
Collapse
Affiliation(s)
- Tycel Jovelle Phillips
- University of Michigan Medical School, Ann Arbor, MI
- Current address: City of Hope National Medical Center, Duarte, CA
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University and IRCCS Humanitas Research Hospital, Milano, Italy
| | | | - Emmanuel Bachy
- Hospices Civils de Lyon and Université Claude Bernard, Pierre-Bénite, France
| | - Michael Crump
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Marek Trněný
- First Faculty of Medicine, Charles University, General Hospital, Prague, Czech Republic
| | | | - Jan Zaucha
- Medical University of Gdańsk, Gdańsk, Poland
| | | | - Fritz Offner
- Department of Hematology, Universitair Ziekenhuis, Gent, Belgium
| | | | - James Relf
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | | | | | - Ben Byrne
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | - Naseer Qayum
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | | | - Michael Dickinson
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
6
|
Dandoy CE, Adams J, Artz A, Bredeson C, Dahi PB, Dodd T, Jaglowski S, Lehmann L, LeMaistre CF, Mian A, Neal A, Page K, Rizzo JD, Rotz S, Sorror M, Steinberg A, Viswabandya A, Howard DS. In Pursuit of Optimal Outcomes: A Framework for Quality Standards in Immune Effector Cell Therapy. Transplant Cell Ther 2024; 30:942-954. [PMID: 39067790 DOI: 10.1016/j.jtct.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Immune effector cell (IEC) therapy represents a transformative advancement in oncology, leveraging the immune system to combat various malignancies. This article outlines a comprehensive framework for establishing and maintaining quality standards in IEC therapy amidst rapid scientific and clinical advancements. We emphasize the integration of structured process measures, robust quality assurance, and meticulous outcome evaluation to ensure treatment efficacy and safety. Key components include multidisciplinary expertise, stringent accreditation protocols, and advanced data management systems, which facilitate standardized reporting and continual innovation. The collaborative effort among stakeholders-ranging from patients and healthcare providers to regulatory bodies-is crucial in delivering high-quality IEC therapies. This framework aims to enhance patient outcomes and cement the role of IEC therapy as a cornerstone of modern oncology, promoting continuous improvement and adherence to high standards across the therapeutic spectrum.
Collapse
Affiliation(s)
- Christopher E Dandoy
- Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio.
| | - Joan Adams
- Stephenson Cancer Center, OU Health Science Center The University of Oklahoma, Oklahoma City, Oklahoma
| | - Andrew Artz
- Division of Leukemia, Department of Hematology and HCT, City of Hope, Duarte, California
| | - Christopher Bredeson
- Ottawa Hospital Research Institute, Division of Hematology, University of Ottawa, Ottawa, Canada
| | - Parastoo B Dahi
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Therese Dodd
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, Tennessee
| | - Samantha Jaglowski
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Leslie Lehmann
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Amir Mian
- Department of Pediatric Hematology and Oncology, Department of Pediatrics at Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Alison Neal
- Department of Bone Marrow Transplant and Cellular Therapy, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Kristen Page
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - J Douglas Rizzo
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Seth Rotz
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio
| | - Mohamed Sorror
- Fred Hutchinson Cancer Center and University of Washington, Seattle, Washington
| | - Amir Steinberg
- Adult Stem Cell Transplantation, Westchester Medical Center, New York Medical College, Valhalla, New York
| | - Auro Viswabandya
- Department of Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Dianna S Howard
- Department of Internal Medicine, Section of Hematology and Oncology, Stem Cell Transplant and Cellular Therapy Program, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, North Carolina
| |
Collapse
|
7
|
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
|
8
|
Sedloev D, Chen Q, Unglaub JM, Schanda N, Hao Y, Besiridou E, Neuber B, Schmitt A, Raffel S, Liu Y, Janssen M, Müller-Tidow C, Schmitt M, Sauer T. Proteasome inhibition enhances the anti-leukemic efficacy of chimeric antigen receptor (CAR) expressing NK cells against acute myeloid leukemia. J Hematol Oncol 2024; 17:85. [PMID: 39285441 PMCID: PMC11406742 DOI: 10.1186/s13045-024-01604-y] [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: 04/18/2024] [Accepted: 08/31/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Relapsed and refractory acute myeloid leukemia (AML) carries a dismal prognosis. CAR T cells have shown limited efficacy in AML, partially due to dysfunctional autologous T cells and the extended time for generation of patient specific CAR T cells. Allogeneic NK cell therapy is a promising alternative, but strategies to enhance efficacy and persistence may be necessary. Proteasome inhibitors (PI) induce changes in the surface proteome which may render malignant cells more vulnerable to NK mediated cytotoxicity. Here, we investigated the potential benefit of combining PIs with CAR-expressing allogeneic NK cells against AML. METHODS We established the IC50 concentrations for Bortezomib and Carfilzomib against several AML cell lines. Surface expression of class-I HLA molecules and stress-associated proteins upon treatment with proteasome inhibitors was determined by multiparameter flow cytometry. Using functional in vitro assays, we explored the therapeutic synergy between pre-treatment with PIs and the anti-leukemic efficacy of NK cells with or without expression of AML-specific CAR constructs against AML cell lines and primary patient samples. Also, we investigated the tolerability and efficacy of a single PI application strategy followed by (CAR-) NK cell infusion in two different murine xenograft models of AML. RESULTS AML cell lines and primary AML patient samples were susceptible to Bortezomib and Carfilzomib mediated cytotoxicity. Conditioned resistance to Azacitidine/Venetoclax did not confer primary resistance to PIs. Treating AML cells with PIs reduced the surface expression of class-I HLA molecules on AML cells in a time-and-dose dependent manner. Stress-associated proteins were upregulated on the transcriptional level and on the cell surface. NK cell mediated killing of AML cells was enhanced in a synergistic manner. PI pre-treatment increased effector-target cell conjugate formation and Interferon-γ secretion, resulting in enhanced NK cell activity against AML cell lines and primary samples in vitro. Expression of CD33- and CD70-specific CARs further improved the antileukemic efficacy. In vivo, Bortezomib pre-treatment followed by CAR-NK cell infusion reduced AML growth, leading to prolonged overall survival. CONCLUSIONS PIs enhance the anti-leukemic efficacy of CAR-expressing allogeneic NK cells against AML in vitro and in vivo, warranting further exploration of this combinatorial treatment within early phase clinical trials.
Collapse
MESH Headings
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/drug effects
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/drug therapy
- Proteasome Inhibitors/pharmacology
- Proteasome Inhibitors/therapeutic use
- Receptors, Chimeric Antigen/immunology
- Animals
- Mice
- Cell Line, Tumor
- Bortezomib/pharmacology
- Bortezomib/therapeutic use
- Oligopeptides/pharmacology
- Oligopeptides/therapeutic use
- Immunotherapy, Adoptive/methods
- Xenograft Model Antitumor Assays
- Mice, Inbred NOD
- Mice, SCID
- Female
Collapse
Affiliation(s)
- David Sedloev
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Qian Chen
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Julia M Unglaub
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Nicola Schanda
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Yao Hao
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Eleni Besiridou
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Anita Schmitt
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Simon Raffel
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Yi Liu
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Maike Janssen
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Michael Schmitt
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Tim Sauer
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
9
|
Sureda A, Adam SE, Yang S, Griffin E, Baker J, Johnston K, Navarro FR, Alhasani S, Chhibber A, Wang A, Mutebi A. Logistical challenges of CAR T-cell therapy in non-Hodgkin lymphoma: a survey of healthcare professionals. Future Oncol 2024:1-14. [PMID: 39268892 DOI: 10.1080/14796694.2024.2393566] [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: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Aim: Characterize the logistical challenges faced by healthcare professionals (HCPs), patients and caregivers during the chimeric antigen receptor T-cell (CAR T) treatment process for non-Hodgkin lymphoma patients.Materials & methods: HCPs in the US and UK experienced with CAR T administration participated in interviews and completed a web-based survey.Results: A total of 133 (80 US, 53 UK) HCPs participated. Two or more logistical challenges were identified by ≥60% of respondents across all stages of the CAR T process. Commonly reported challenges were lengthy waiting periods, administrative and payer-related barriers, limited healthcare capacity, caregiver support and (particularly in the US) patient out-of-pocket costs.Conclusion: The CAR T treatment process presents numerous challenges, highlighting an unmet need for more convenient therapies.
Collapse
Affiliation(s)
- Anna Sureda
- Institut Català d'Oncologia, Hospital Duran i Reynals, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Hungria V, Sureda A, Campelo GR, Salvino MA, Ramasamy K. Proceedings from the First Onco Summit: LATAM Chapter, 19-20 May 2023, Rio de Janeiro, Brazil. Cancers (Basel) 2024; 16:3063. [PMID: 39272921 PMCID: PMC11394439 DOI: 10.3390/cancers16173063] [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: 07/08/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
The Onco Summit 2023: The Latin American (LATAM) Chapter took place over two days, from 19-20 May 2023, in Brazil. The event aimed to share the latest updates across various oncology disciplines, address critical clinical challenges, and exchange best practices to ensure optimal patient treatment. More than 30 international and regional speakers and more than 300 oncology specialists participated in the Summit. The Summit discussions centered on common challenges and therapeutic advances in cancer care, with a specific focus on the unique obstacles faced in LATAM and examples of adaptable strategies to address these challenges. The Summit also facilitated the establishment of a network of oncologists, hematologists, and scientists in LATAM, enabling collaboration to improve cancer care, both in this region and globally, through drug development and clinical research. This report summarizes the key discussions from the Summit for the global and LATAM oncology community.
Collapse
Affiliation(s)
- Vania Hungria
- Hematology, Faculty of Medical Sciences of Santa Casa de São Paulo, São Paulo 01224-001, Brazil
| | - Anna Sureda
- Clinical Hematology Department, Catalan Institut Català d'Oncologia-L'Hospitalet, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), University of Barcelona (UB), 08908 Barcelona, Spain
| | - Garcia Rosario Campelo
- Thoracic Tumors Unit, Medical Oncology Department, University Hospital A Coruña Biomedical Research Institute (INIBIC), 15006 A Coruña, Spain
| | - Marco Aurélio Salvino
- Cell Therapy, D'OR Institute Research & Education (IDOR)/PPGMS-Federal University of Bahia (UFBA), Salvador 40110-100, Brazil
| | - Karthik Ramasamy
- Oxford Translational Myeloma Centre, NDORMS, University of Oxford, Oxford OX3 7LD, UK
| |
Collapse
|
11
|
Nicolai CJ, Parker MH, Qin J, Tang W, Ulrich-Lewis JT, Gottschalk RJ, Cooper SE, Hernandez Lopez SA, Parrilla D, Mangio RS, Ericson NG, Brandes AH, Umuhoza S, Michels KR, McDonnell MM, Park LY, Shin S, Leung WH, Scharenberg AM, Kiem HP, Larson RP, Beitz LO, Ryu BY. In vivo CAR T-cell generation in nonhuman primates using lentiviral vectors displaying a multidomain fusion ligand. Blood 2024; 144:977-987. [PMID: 38861668 PMCID: PMC11406189 DOI: 10.1182/blood.2024024523] [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: 03/05/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell therapies have demonstrated transformative efficacy in treating B-cell malignancies. However, high costs and manufacturing complexities hinder their widespread use. To overcome these hurdles, we have developed the VivoVec platform, a lentiviral vector capable of generating CAR T cells in vivo. Here, we describe the incorporation of T-cell activation and costimulatory signals onto the surface of VivoVec particles (VVPs) in the form of a multidomain fusion protein and show enhanced in vivo transduction and improved CAR T-cell antitumor functionality. Furthermore, in the absence of lymphodepleting chemotherapy, administration of VVPs into nonhuman primates resulted in the robust generation of anti-CD20 CAR T cells and the complete depletion of B cells for >10 weeks. These data validate the VivoVec platform in a translationally relevant model and support its transition into human clinical testing, offering a paradigm shift in the field of CAR T-cell therapies.
Collapse
Affiliation(s)
| | | | - Jim Qin
- Umoja Biopharma, Seattle, WA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hansen DK, Lu X, Puglianini OC, Sorensen S, Usmani SZ, Zhang E, Huo S, Zhang Y, Qureshi ZP, Jagannath S. Cost-per-responder analysis of patients with lenalidomide-refractory multiple myeloma receiving ciltacabtagene autoleucel in CARTITUDE-4. Front Immunol 2024; 15:1408892. [PMID: 39234256 PMCID: PMC11372240 DOI: 10.3389/fimmu.2024.1408892] [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: 03/29/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Ciltacabtagene autoleucel (cilta-cel) is a chimeric antigen receptor T-cell therapy approved for patients with relapsed/refractory multiple myeloma (RRMM). In the phase 3 trial, CARTITUDE-4 (NCT04181827), cilta-cel demonstrated improved efficacy vs. standard of care (SOC; daratumumab plus pomalidomide and dexamethasone [DPd] or pomalidomide plus bortezomib and dexamethasone [PVd]) with a ≥ complete response (≥CR) rate of 73.1% vs. 21.8%. Methods A cost-per-responder model was developed to assess the value of cilta-cel and SOC (87% DPd and 13% PVd) based on the CARTITUDE-4 trial data from a US mixed payer perspective (76.7% commercial, 23.3% Medicare). The model was developed using progression-free survival (PFS), overall survival (OS), and ≥CR endpoints from CARTITUDE-4 over a period of 25.4 months. Inpatient stays, outpatient visits, drug acquisition, administration, and monitoring costs were included. The base-case model assumed an inpatient setting for each cilta-cel infusion; another scenario included 30% outpatient and 70% inpatient infusions. Costs of managing grade 3-4 adverse events (AEs) and grade 1-4 cytokine release syndrome and neurotoxicity were included. Subsequent therapy costs were incurred after disease progression; terminal care costs were considered upon death events. Outcomes included total cost per treated patient, total cost per complete responder, and cost per month in PFS between cilta-cel and SOC. Costs were adjusted to 2024 US dollars. Results Total cost per treated patient, total cost per complete responder, and total cost per month in PFS were estimated at $704,641, $963,941, and $30,978 for cilta-cel, respectively, and $840,730, $3,856,559, and $42,520 for SOC over the 25.4-month period. Cost drivers included treatment acquisition costs before progression and subsequent treatment costs ($451,318 and $111,637 for cilta-cel; $529,795 and $265,167 for SOC). A scenario analysis in which 30% of patients received an outpatient infusion (assuming the same payer mix) showed a lower cost per complete responder for cilta-cel ($956,523) than those with an infusion in the inpatient setting exclusively. Discussion This analysis estimated that cost per treated patient, cost per complete responder, and cost per month in PFS for cilta-cel were remarkably lower than for DPd or PVd, highlighting the substantial clinical and economic benefit of cilta-cel for patients with RRMM.
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
| | - Xiaoxiao Lu
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, United States
| | - Omar Castaneda Puglianini
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | | | - Saad Z. Usmani
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Stephen Huo
- Janssen Global Services, LLC, Raritan, NJ, United States
| | - Yan Zhang
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, United States
| | - Zaina P. Qureshi
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, United States
| | - Sundar Jagannath
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
13
|
Ramamurthy A, Tommasi A, Saha K. Advances in manufacturing chimeric antigen receptor immune cell therapies. Semin Immunopathol 2024; 46:12. [PMID: 39150566 DOI: 10.1007/s00281-024-01019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/20/2024] [Indexed: 08/17/2024]
Abstract
Biomedical research has witnessed significant strides in manufacturing chimeric antigen receptor T cell (CAR-T) therapies, marking a transformative era in cellular immunotherapy. Nevertheless, existing manufacturing methods for autologous cell therapies still pose several challenges related to cost, immune cell source, safety risks, and scalability. These challenges have motivated recent efforts to optimize process development and manufacturing for cell therapies using automated closed-system bioreactors and models created using artificial intelligence. Simultaneously, non-viral gene transfer methods like mRNA, CRISPR genome editing, and transposons are being applied to engineer T cells and other immune cells like macrophages and natural killer cells. Alternative sources of primary immune cells and stem cells are being developed to generate universal, allogeneic therapies, signaling a shift away from the current autologous paradigm. These multifaceted innovations in manufacturing underscore a collective effort to propel this therapeutic approach toward broader clinical adoption and improved patient outcomes in the evolving landscape of cancer treatment. Here, we review current CAR immune cell manufacturing strategies and highlight recent advancements in cell therapy scale-up, automation, process development, and engineering.
Collapse
Affiliation(s)
- Apoorva Ramamurthy
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Tommasi
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
14
|
Goel U, Mian A, Sauter CS. SOHO State of the Art Updates and Next Questions | Contemporary Role of Autologous Stem Cell Transplantation for the Treatment of Relapsed/Refractory Diffuse Large B-Cell Lymphoma in the Era of Cellular Therapies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024:S2152-2650(24)00289-1. [PMID: 39214752 DOI: 10.1016/j.clml.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Since the 1990s, the standard of care for the treatment of relapsed/refractory diffuse large B-cell lymphoma (DLBCL) had been salvage chemotherapy followed by high-dose chemotherapy and autologous stem cell transplantation (HDT-ASCT) in patients with a chemotherapy-sensitive remission. However, promising results from the recent TRANSFORM and ZUMA-7 trials evaluating the efficacy of CAR T-cell therapy versus HDT-ASCT for second line relapsed/refractory DLBCL have sought to challenge this standard of care. While these studies have established a new standard for the treatment of early relapsed and primary refractory DLBCL, significant differences in the trial design between these studies and limitations with the timing of randomization during the disease course warrant a thoughtful interpretation of the results. Additionally, the financial burden and logistic challenges of CAR T-cell administration and limited access to these therapies continue to be ongoing issues. Despite the encouraging results from these trials, HDT-ASCT continues to have a role in the treatment of DLBCL, especially in disease relapsing ≥12 months after initial therapy, and in chemo sensitive disease with a good response to salvage chemotherapy. Ongoing studies evaluating novel salvage regimens for use prior to HDT-ASCT, and future studies evaluating the role of CAR T-cell therapy in chemo sensitive disease will help determine the continued role of HDT-ASCT for relapsed/refractory DLBCL.
Collapse
Affiliation(s)
- Utkarsh Goel
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH
| | - Agrima Mian
- Department of Hematology and Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
| | - Craig S Sauter
- Department of Hematology and Medical Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
15
|
Mohty R, Lazaryan A. "Off-The-Shelf" allogeneic chimeric antigen receptor T-cell therapy for B-cell malignancies: current clinical evidence and challenges. Front Oncol 2024; 14:1433432. [PMID: 39055556 PMCID: PMC11269961 DOI: 10.3389/fonc.2024.1433432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
Chimeric antigen receptor T-cell therapy (CAR T) has revolutionized the treatment landscape for hematologic malignancies, notably B-cell non-Hodgkin lymphoma (B-NHL) and B-cell acute lymphoblastic leukemia (B-ALL). While autologous CAR T products have shown remarkable efficacy, their complex logistics, lengthy manufacturing process, and high costs impede widespread accessibility and pose therapeutic challenge especially for patients in rapid need for therapy. "Off-the-shelf" allogeneic CAR T-cell therapy (alloCAR T) has emerged as a promising alternative therapy, albeit experimental to date. AlloCARTs are derived from healthy donors, manufactured by batches and stored, making them available off-the-shelf which lowers financial burden. Various gene editing techniques have been employed to mitigate graft-versus-host disease (GVHD) and host-versus-graft (HvG) to enhance alloCAR T persistence. In this review, we summarize available manufacturing techniques, current evidence, and discuss challenges faced with the use of alloCAR Ts.
Collapse
Affiliation(s)
- Razan Mohty
- Division of Hematology Oncology, Department of Medicine, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, United States
| | - Aleksandr Lazaryan
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
16
|
Shih SCM, Bhella S. Review of CAR T-Cell Therapy in Multiple Myeloma: A Canadian Perspective. Curr Oncol 2024; 31:3949-3967. [PMID: 39057164 PMCID: PMC11276237 DOI: 10.3390/curroncol31070292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy. In the context of the current standard of care therapies in Canada, outcomes among patients with relapsed/refractory multiple myeloma (RRMM), particularly those with triple-class (or more) refractory disease remain poor. Immunotherapies have significantly changed the treatment landscape of MM. Since 2021, two BCMA-targeting CAR T-cell therapy products have been approved for RRMM-namely Idecabtagene vicleucel (Ide-cel) (ABECMA®) and Ciltacabtagene autoleucel (Cilta-cel) (CARVYKTI®), both of which are available in the US and Europe. Although they have shown unprecedented efficacy in RRMM, their clinical and logistical limitations must be acknowledged. MM CAR T-cell therapy is likely to be approved in Canada soon. Therefore, it is timely that we review the latest evidence for commercially available CAR T-cell therapy in multiple myeloma, with a focus on its relevance and impact in the Canadian setting. There will be challenges to access and strategies must be in place to ensure equitable care for all Canadians with MM. Alongside haematologists working in the immune effector cell therapy programs, providers in the community will also play a role in the ongoing monitoring and management of long-term side effects including opportunistic infections and late neurotoxicity.
Collapse
|
17
|
Lee NK, Chang JW. Manufacturing Cell and Gene Therapies: Challenges in Clinical Translation. Ann Lab Med 2024; 44:314-323. [PMID: 38361427 PMCID: PMC10961620 DOI: 10.3343/alm.2023.0382] [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: 09/26/2023] [Revised: 11/24/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
The safety and efficacy of both cell and gene therapies have been demonstrated in numerous preclinical and clinical trials. Chimeric antigen receptor T (CAR-T) cell therapy, which leverages the technologies of both cell and gene therapies, has also shown great promise for treating various cancers. Advancements in pertinent fields have also highlighted challenges faced while manufacturing cell and gene therapy products. Potential problems and obstacles must be addressed to ease the clinical translation of individual therapies. Literature reviews of representative cell-based, gene-based, and cell-based gene therapies with regard to their general manufacturing processes, the challenges faced during manufacturing, and QC specifications are limited. We review the general manufacturing processes of cell and gene therapies, including those involving mesenchymal stem cells, viral vectors, and CAR-T cells. The complexities associated with the manufacturing processes and subsequent QC/validation processes may present challenges that could impede the clinical progression of the products. This article addresses these potential challenges. Further, we discuss the use of the manufacturing model and its impact on cell and gene therapy.
Collapse
Affiliation(s)
- Na Kyung Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jong Wook Chang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
| |
Collapse
|
18
|
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
|
19
|
Sin WX, Jagannathan NS, Teo DBL, Kairi F, Fong SY, Tan JHL, Sandikin D, Cheung KW, Luah YH, Wu X, Raymond JJ, Lim FLWI, Lee YH, Seng MSF, Soh SY, Chen Q, Ram RJ, Tucker-Kellogg L, Birnbaum ME. A high-density microfluidic bioreactor for the automated manufacturing of CAR T cells. Nat Biomed Eng 2024:10.1038/s41551-024-01219-1. [PMID: 38834752 DOI: 10.1038/s41551-024-01219-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/20/2024] [Indexed: 06/06/2024]
Abstract
The manufacturing of autologous chimaeric antigen receptor (CAR) T cells largely relies either on fed-batch and manual processes that often lack environmental monitoring and control or on bioreactors that cannot be easily scaled out to meet patient demands. Here we show that human primary T cells can be activated, transduced and expanded to high densities in a 2 ml automated closed-system microfluidic bioreactor to produce viable anti-CD19 CAR T cells (specifically, more than 60 million CAR T cells from donor cells derived from patients with lymphoma and more than 200 million CAR T cells from healthy donors). The in vitro secretion of cytokines, the short-term cytotoxic activity and the long-term persistence and proliferation of the cell products, as well as their in vivo anti-leukaemic activity, were comparable to those of T cells produced in a gas-permeable well. The manufacturing-process intensification enabled by the miniaturized perfusable bioreactor may facilitate the analysis of the growth and metabolic states of CAR T cells during ex vivo culture, the high-throughput optimization of cell-manufacturing processes and the scale out of cell-therapy manufacturing.
Collapse
Affiliation(s)
- Wei-Xiang Sin
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - N Suhas Jagannathan
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Denise Bei Lin Teo
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Faris Kairi
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Shin Yie Fong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Joel Heng Loong Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dedy Sandikin
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Ka-Wai Cheung
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Yen Hoon Luah
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Xiaolin Wu
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Joshua Jebaraj Raymond
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
| | - Francesca Lorraine Wei Inng Lim
- Advanced Cell Therapy and Research Institute, Singapore (ACTRIS), Consortium for Clinical Research and Innovation, Singapore (CRIS), Singapore, Singapore
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Programme, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Yie Hou Lee
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Michaela Su-Fern Seng
- SingHealth Duke-NUS Oncology Academic Clinical Programme, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Department of Paediatric Haematology and Oncology, KK Women's and Children's Hospital, Singapore, Singapore
| | - Shui Yen Soh
- SingHealth Duke-NUS Oncology Academic Clinical Programme, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Department of Paediatric Haematology and Oncology, KK Women's and Children's Hospital, Singapore, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Rajeev J Ram
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Lisa Tucker-Kellogg
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore.
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.
| | - Michael E Birnbaum
- Critical Analytics for Manufacturing Personalized-Medicine (CAMP), Singapore-MIT Alliance for Research and Technology Centre (SMART), Singapore, Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
20
|
Bishop MR, Kay GE. CAR T-cell therapy: A collaboration between authorized treatment centers and community oncologists. Semin Oncol 2024; 51:87-94. [PMID: 38531760 DOI: 10.1053/j.seminoncol.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/28/2024]
Abstract
With the approval of the first CAR T-cell products for hematological malignancies in 2017, these autologous cell therapies have changed the treatment paradigm for patients with relapsed or refractory (r/r) non-Hodgkin lymphoma (NHL), who have a poor prognosis and few effective treatment options. Despite the demonstrated clinical benefit in patients with r/r diffuse large B-cell lymphoma, mantle cell lymphoma, and follicular lymphoma, many patients who are eligible for CAR T-cell therapies do not receive them or are treated with CAR T cells as a later line of therapy at advanced stages of disease. Several barriers exist for referring patients to an authorized treatment center (ATC) for CAR T-cell therapy. Although most patients with NHL are treated by community-based oncologists, educational gaps may exist for some community oncologists about the availability of CAR T-cell therapies in certain indications, the overall treatment process, and how they can access these therapies for their patients. In addition to navigation of the referral process from the community setting to the ATC, other barriers include timely identification of candidates eligible for CAR T-cell therapy and logistical and reimbursement concerns. Here, we examine the patient CAR T-cell experience, which begins and ends in the community setting, and identify and discuss opportunities for improved collaboration between community oncologists and ATC physicians to help address barriers to treatment and enhance patient outcomes. Treatment decisions for a patient's second or third line of therapy for NHL are critically important, owing to declining probabilities for favorable outcomes with each successive line of therapy. For patients who are eligible, CAR T-cell therapies should be considered as early as possible in their treatment course. A better understanding of the CAR T-cell process, the patient's experience, and the collaboration necessary for timely patient identification, better access, and successful outcomes will enable more patients to benefit from CAR T-cell therapies.
Collapse
Affiliation(s)
- Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL.
| | - Gary E Kay
- Northwest Oncology and Hematology, Rolling Meadows, IL
| |
Collapse
|
21
|
Bennett R, Dickinson M. SOHO State of the Art Updates and Next Questions | Current Evidence and Future Directions for Bispecific Antibodies in Large B-Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024:S2152-2650(24)00181-2. [PMID: 38871556 DOI: 10.1016/j.clml.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/15/2024]
Abstract
The CD20xCD3 bispecific antibodies (bsAb) are "off-the-shelf" T-cell re-directing therapies that demonstrate remarkable single-agent clinical activity in B-cell lymphomas. Two agents, epcoritamab (epcor) and glofitamab (glofit) have recent global approvals for patients with relapsed/refractory DLBCL (RR DLBCL) following 2 prior treatment lines. Both agents demonstrate activity in patients with prior exposure to chimeric antigen receptor T-cell (CAR-T) treatment. As multiyear follow-up data become available, it is clear that the majority of patients achieving complete remissions do not relapse and that outcomes are similar between epcor and glofit. CD20xCD3 bsAb have a safety profile that reflect their mechanism of action, with cytokine release syndrome (CRS) the key management issue. Neurotoxicity is far less common than observed with CD19-directed CAR-T. BsAbs are attractive, rapidly available, treatment options for patients with RR DLBCL, without the practical and financial challenges seen with autologous CAR-T therapies. Recent data also demonstrate the feasibility and potential efficacy of bsAb in combination with chemoimmunotherapy with large randomized trials evaluating bsAb-chemotherapy combinations underway. There are open questions about the future role of bsAB for LBCL, the optimal duration of therapy, optimal CRS risk mitigation strategies, and potential resistance mechanisms. In this review we seek to describe the current evidence for bsAb in LBCL, and offer opinion regarding these open questions.
Collapse
Affiliation(s)
- Rory Bennett
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Victoria, Australia
| | - Michael Dickinson
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
22
|
Jurgens E, Usmani SZ. SOHO State of the Art Updates and Next Questions: Will CAR-T Replace ASCT in NDMM. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:277-284. [PMID: 38331676 DOI: 10.1016/j.clml.2024.01.001] [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: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024]
Abstract
The treatment landscape for multiple myeloma (MM) has rapidly evolved over the last 2 decades. The development of triplet and quadruplet regimens including proteasome inhibitors (PI), immunomodulatory agents (IMiDs), and anti-CD38 monoclonal antibodies has dramatically extended overall survival. In addition to effective multidrug regimens, autologous stem cell transplant (ASCT) is a cornerstone of management in newly diagnosed multiple myeloma (NDMM). However, despite these combined treatment modalities, curative therapy for MM remains elusive. Recent, novel immunotherapies including chimeric antigen T-cell (CAR-T) therapy have demonstrated deep and durable responses in relapsed and refractory multiple myeloma (RRMM). Currently 2 CAR-T products, ciltacabtagene autoleucel (cilta-cel) and idecabtagene vicleucel (ide-cel), are approved by the FDA for the treatment of RRMM. The success of CAR-T therapy revolutionized the management of RRMM prompting clinical trials studying CAR-T therapy in the first line setting. The ongoing KarMMa-4, CARTITUDE-5, and CARTITUDE-6 clinical trials may establish CAR-T therapy as a first line option potentially supplanting ASCT in the initial treatment of NDMM. In this review, we discuss the current standard of care management of NDMM, trace the evolution of CAR-T clinical trials in RRMM, and survey ongoing clinical trials studying CAR-T therapy in NDMM.
Collapse
Affiliation(s)
- Eric Jurgens
- Department of Medicine, Hematology Oncology Fellowship Program, MSKCC, New York, NY
| | - Saad Z Usmani
- Department of Medicine, Myeloma Service, MSKCC, New York, NY.
| |
Collapse
|
23
|
Ailawadhi S, Shune L, Wong SW, Lin Y, Patel K, Jagannath S. Optimizing the CAR T-Cell Therapy Experience in Multiple Myeloma: Clinical Pearls From an Expert Roundtable. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:e217-e225. [PMID: 38369437 DOI: 10.1016/j.clml.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/20/2024]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies offer substantial advancement in the treatment of multiple myeloma (MM). However, the CAR-T therapy process involves complex decision-making that is informed by many variables. This review aims to provide an overview of the patient selection and administration process for CAR-T therapy for MM from the perspective of experienced healthcare providers (HCPs), including considerations for each step in the CAR-T therapy process. Referring HCPs should initiate conversations with HCPs at CAR-T capable centers earlier in the treatment journey, even before patients are eligible for CAR-T therapy, particularly for patients from underserved populations and patients with high-risk disease, to ensure adequate time for logistical planning and patient education. Patient selection for CAR-T therapy may be guided by factors such as performance status, rate of disease progression, and logistical considerations. Some anticancer therapies may affect T-cell fitness and therefore impact CAR-T manufacturing and patient outcomes; however, additional research is needed to confirm this in MM. Bridging therapies should be tailored to the needs of the patient and ideally halted 1 week or longer before CAR-T infusion, contingent upon the agent(s) used. Lymphodepletion regimens may need to be modified for patients with renal insufficiency. Collaboration with HCPs at both the treating and referring centers is important to optimize coordinated care of patients. Collaboration with and guidance from experienced HCPs throughout patient selection, referral, and CAR-T administration is instrumental in optimizing patient outcomes as access to CAR-T therapies expands.
Collapse
Affiliation(s)
| | - Leyla Shune
- Division of Hematologic Malignancy and Cellular Therapeutics, University of Kansas Cancer Center, Westwood, Kansas
| | - Sandy W Wong
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Krina Patel
- Department of Lymphoma - Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
24
|
Richter J. Like a bridge over troubled water: keeping the myeloma down en route to CAR-T. Blood Cancer J 2024; 14:64. [PMID: 38609377 PMCID: PMC11015014 DOI: 10.1038/s41408-024-01049-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Affiliation(s)
- Joshua Richter
- Tisch Cancer Institute: Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
25
|
Ghilardi G, Williamson S, Pajarillo R, Paruzzo L, Chen L, Grady C, Doucette A, Nemecek E, Gabrielli G, Barta SK, Svoboda J, Chong EA, Yelton R, Nasta SD, Landsburg DJ, Ugwuanyi O, Chen AI, Schachter L, White G, Ballard HJ, Weber E, Curley C, Porter DL, Garfall AL, Hwang WT, Guerra CE, Maziarz RT, Schuster SJ, Ruella M. CAR T-Cell Immunotherapy in Minority Patients with Lymphoma. NEJM EVIDENCE 2024; 3:EVIDoa2300213. [PMID: 38776868 DOI: 10.1056/evidoa2300213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
BACKGROUND Administration of anti-CD19 chimeric antigen receptor T-cell (CART19) immunotherapy for large B-cell lymphomas (LBCLs), a subset of non-Hodgkin lymphoma (NHL), involves high costs and access to specialized tertiary care centers. We investigated whether minority health populations (MHPs) have equal access to CART19 and whether their outcomes are similar to those of non-MHPs. METHODS We analyzed the prevalence and clinical outcomes of patients treated with commercial CART19 at two geographically and socioeconomically different institutions: the Abramson Cancer Center (ACC, Philadelphia, Pennsylvania) and the Knight Cancer Institute (KCI, Portland, Oregon). RESULTS In the ACC catchment area, 8956 patients were diagnosed with NHL between 2015 and 2019 (latest available data from the state registry), including 17.9% MHPs. In the ACC, between 2018 and 2022 (CART became available in 2018), 1492 patients with LBCL were treated, and 194 received CART19. The proportion of MHPs was 15.7% for the entire LBCL cohort but only 6.7% for the CART19 cohort. During the same time, in the KCI catchment area, 4568 patients were diagnosed with NHL, including 4.2% MHPs. In the KCI, 396 patients with LBCL were treated, and 47 received CART19. The proportion of MHPs was 6.6% for the entire LBCL cohort and 4.2% for the CART19 cohort. The 3-month response, survival, and toxicities after CART19 infusion showed similar results, although the number of patients who were treated was limited. CONCLUSIONS This study shows that the access of MHPs to tertiary centers for LBCL care was preserved but appeared reduced for commercial CART19 immunotherapy. Although clinical outcomes of MHPs seemed similar to those of non-MHPs, the small sample size precludes drawing firm conclusions. Further studies are needed. (Funded by the Laffey McHugh Foundation and others.).
Collapse
Affiliation(s)
- Guido Ghilardi
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Staci Williamson
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Raymone Pajarillo
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Luca Paruzzo
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Linhui Chen
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Connor Grady
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Abigail Doucette
- Division of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia
| | - Eneida Nemecek
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Giulia Gabrielli
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Stefan K Barta
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Jakub Svoboda
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Elise A Chong
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Rebecca Yelton
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Sunita D Nasta
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Daniel J Landsburg
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Ositadimma Ugwuanyi
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Andy I Chen
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Levanto Schachter
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Griffin White
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Hatcher J Ballard
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Elizabeth Weber
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Cara Curley
- Division of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia
| | - David L Porter
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
| | - Alfred L Garfall
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
| | - Wei-Ting Hwang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Carmen E Guerra
- Division of General Medicine, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
| | - Richard T Maziarz
- Adult Blood and Marrow Stem Cell Transplant and Cell Therapy Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Stephen J Schuster
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Marco Ruella
- Division of Hematology and Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| |
Collapse
|
26
|
Choe JH, Yu T, Abramson JS, Abou-el-Enein M. Cost-effectiveness of second-line lisocabtagene maraleucel in relapsed or refractory diffuse large B-cell lymphoma. Blood Adv 2024; 8:484-496. [PMID: 38153350 PMCID: PMC10837180 DOI: 10.1182/bloodadvances.2023011793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023] Open
Abstract
ABSTRACT Lisocabtagene maraleucel (liso-cel), a chimeric antigen receptor (CAR) T-cell therapy, received the US Food and Drug Administration approval in 2022 for second-line treatment of diffuse large B-cell lymphoma (DLBCL) for patients with refractory disease or early relapse after first-line chemoimmunotherapy. This decision was based on the TRANSFORM study demonstrating improvements in event-free survival with liso-cel compared with standard care. Given the high costs of CAR T-cell therapies, particularly as they transition to second-line treatment, a cost-effectiveness analysis is essential to determine their economic viability. The study used a partitioned survival model with standard parametric functions to evaluate the cost-effectiveness of liso-cel aganist platinum-based chemotherapy followed by high-dose chemotherapy and autologous hematopoietic stem cell transplantation over a lifetime horizon The analysis relied on data from the TRANSFORM and TRANSCEND trials, established literature, and public data sets to calculate the incremental cost-effectiveness ratio (ICER). For a representative cohort of US adults aged 60 years, ICER of liso-cel was $99 669 per quality-adjusted life-year (QALY) from a health care sector perspective and $68 212 per QALY from a societal perspective, confirming its cost-effectiveness at the $100 000 per QALY threshold. Nonetheless, under certain scenarios, liso-cel surpasses this benchmark but remains within the US acceptable range of $150 000 per QALY. A key finding underlines the importance of incorporating productivity losses into such analyses to capture the broader societal values of novel therapies. Although these therapies offer substantial clinical benefits, their high acquisition costs and limited long-term data critically challenge their economic sustainability.
Collapse
Affiliation(s)
- Jee H. Choe
- Department of Pharmaceutical and Health Economics, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA
- Leonard D. Schaeffer Center for Health Policy & Economics, University of Southern California, Los Angeles, CA
| | - Tianzhou Yu
- Department of Pharmaceutical and Health Economics, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA
- Leonard D. Schaeffer Center for Health Policy & Economics, University of Southern California, Los Angeles, CA
| | - Jeremy S. Abramson
- Center for Lymphoma, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Mohamed Abou-el-Enein
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California and Children’s Hospital of Los Angeles, Los Angeles, CA
- USC/CHLA Cell Therapy Program, University of Southern California and Children’s Hospital of Los Angeles, Los Angeles, CA
| |
Collapse
|
27
|
Oluwole OO, Patel AR, Vadgama S, Smith NJ, Blissett R, Feng C, Dickinson M, Johnston PB, Perales MA. An updated cost-effectiveness analysis of axicabtagene ciloleucel in second-line large B-cell lymphoma patients in the United States. J Med Econ 2024; 27:77-83. [PMID: 38053517 PMCID: PMC11254511 DOI: 10.1080/13696998.2023.2290832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023]
Abstract
AIMS This economic evaluation of axicabtagene ciloleucel (axi-cel) versus previous standard of care (SOC; salvage chemotherapy followed by high-dose therapy with autologous stem cell rescue) in the second line (2L) large B-cell lymphoma population is an update of previous economic models that contained immature survival data. METHODS This analysis is based on primary overall survival (OS) ZUMA-7 clinical trial data (median follow-up of 47.2 months), from a United States (US) payer perspective, with a model time horizon of 50 years. Mixture cure models were used to extrapolate updated survival data; subsequent treatment data and costs were updated. Patients who remained in the event-free survival state by 5 years were assumed to have achieved long-term remission and not require subsequent treatment. RESULTS Substantial survival and quality of life benefits were observed despite 57% of patients in the SOC arm receiving subsequent cellular therapy: median model-projected (ZUMA-7 trial Kaplan-Meier estimated) OS was 78 months (median not reached) for axi-cel versus 25 months (31 months) for SOC, resulting in incremental quality-adjusted life year (QALY) difference of 1.63 in favor of axi-cel. Incrementally higher subsequent treatment costs were observed in the SOC arm due to substantial crossover to cellular therapies, thus, when considering the generally accepted willingness to pay threshold of $150,000 per QALY in the US, axi-cel was cost-effective with an incremental cost-effectiveness ratio of $98,040 per QALY. CONCLUSIONS Results remained consistent across a wide range of sensitivity and scenario analysis, including a crossover adjusted analysis, suggesting that the mature OS data has significantly reduced the uncertainty of axi-cel's cost-effectiveness in the 2L setting in the US. Deferring treatment with CAR T therapies after attempting a path to transplant may result in excess mortality, lower quality of life and would be an inefficient use of resources relative to 2L axi-cel.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
28
|
Budde LE, Olszewski AJ, Assouline S, Lossos IS, Diefenbach C, Kamdar M, Ghosh N, Modi D, Sabry W, Naik S, Mehta A, Nakhoda SK, Smith SD, Dorritie K, Jia T, Pham S, Huw LY, Jing J, Wu H, Ead WS, To I, Batlevi CL, Wei MC, Chavez JC. Mosunetuzumab with polatuzumab vedotin in relapsed or refractory aggressive large B cell lymphoma: a phase 1b/2 trial. Nat Med 2024; 30:229-239. [PMID: 38072960 PMCID: PMC10803244 DOI: 10.1038/s41591-023-02726-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/16/2023] [Indexed: 01/24/2024]
Abstract
Relapsed/refractory aggressive large B cell lymphoma (LBCL) remains an area of unmet need. Here we report the primary analysis of a phase 1b/2 trial of outpatient mosunetuzumab (a CD20xCD3 T-cell-engaging bispecific antibody) plus polatuzumab vedotin (an anti-CD79B antibody-drug conjugate) in relapsed/refractory LBCL. The phase 2 component is a single arm of an ongoing multi-arm trial. The primary endpoint during dose expansion was independent review committee (IRC)-assessed best overall response rate. Secondary endpoints included investigator-assessed overall response rate, complete response, duration of response, progression-free survival and overall survival. At data cutoff, 120 patients were enrolled (22 dose escalation, 98 dose expansion). The primary endpoint was met during dose expansion, with IRC-assessed best overall response rate and complete response rates of 59.2% (58/98; 95% confidence interval (CI): 48.8-69.0) and 45.9% (45/98; 95% CI: 35.8-56.3), respectively (median follow-up, 23.9 months). Median duration of complete was not reached (95% CI: 20.5-not estimable (NE)). Median progression-free survival was 11.4 months (95% CI: 6.2-18.7). Median overall survival was 23.3 months (95% CI: 14.8-NE). Across dose escalation and expansion, the most common grade 3 or higher adverse events were neutropenia (25.0%, 30/120) and fatigue (6.7%, 8/120). Any-grade cytokine release syndrome occurred in 16.7% of patients. These data demonstrate that mosunetuzumab plus polatuzumab vedotin has a favorable safety profile with highly durable responses suitable as second-line therapy in transplant-ineligible relapsed/refractory LBCL. ClinicalTrials.gov identifier: NCT03671018 .
Collapse
Affiliation(s)
- Lihua E Budde
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | | | - Sarit Assouline
- Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Izidore S Lossos
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| | | | | | - Nilanjan Ghosh
- Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC, USA
| | - Dipenkumar Modi
- Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Waleed Sabry
- Saskatoon Cancer Center, Saskatoon, Saskatchewan, Canada
| | - Seema Naik
- Penn State Cancer Institute, Hershey, PA, USA
| | | | | | | | - Kathleen Dorritie
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ting Jia
- Roche (China) Holding Ltd, Shanghai, China
| | - Song Pham
- F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | | | - Jing Jing
- Genentech, Inc., South San Francisco, CA, USA
| | - Hao Wu
- Genentech, Inc., South San Francisco, CA, USA
| | - Wahib S Ead
- Genentech, Inc., South San Francisco, CA, USA
| | - Iris To
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | |
Collapse
|
29
|
Riedell PA, Downs C, Boehmer L, Ebmeier J, Porter D, Williams A. If They RECUR, You Should Refer: A Community Oncologist Patient ID Roundtable Summary. Transplant Cell Ther 2024; 30:14-16. [PMID: 37918667 DOI: 10.1016/j.jtct.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023]
Affiliation(s)
- Peter A Riedell
- Association of American Cancer Institutes, Pittsburgh, Pennsylvania.
| | - Christian Downs
- Association of Community Cancer Centers, Rockville, Maryland
| | - Leigh Boehmer
- Association of Community Cancer Centers, Rockville, Maryland
| | - Judy Ebmeier
- Association of Community Cancer Centers, Rockville, Maryland
| | - David Porter
- American Society for Transplantation and Cellular Therapy, Chicago, Illinois
| | - Andre Williams
- American Society for Transplantation and Cellular Therapy, Chicago, Illinois
| |
Collapse
|
30
|
Chung H, Cho H. Recent advances in cellular immunotherapy for lymphoid malignancies. Blood Res 2023; 58:166-172. [PMID: 37964655 PMCID: PMC10758636 DOI: 10.5045/br.2023.2023177] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023] Open
Abstract
Cellular immunotherapy with chimeric antigen receptor (CAR) T-cells has revolutionized the treatment of lymphoid malignancies. This review addresses the need for CAR expression in our endogenous T-cells to kill tumor cells with a focus on the basic principles of T-cell receptor recognition of major histocompatibility complex-peptide complexes. We review the factors associated with CAR T-cell outcomes and recent efforts to employ CAR T-cells in earlier lines of therapy. We also discuss the value of bispecific T-cell engagers as off-the-shelf products with better toxicity profiles. Finally, natural killer cells are discussed as an important cellular immunotherapy platform with the potential to broaden immunotherapeutic applications beyond lymphoid malignancies.
Collapse
Affiliation(s)
- Haerim Chung
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyunsoo Cho
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
31
|
Chamorro DF, Somes LK, Hoyos V. Engineered Adoptive T-Cell Therapies for Breast Cancer: Current Progress, Challenges, and Potential. Cancers (Basel) 2023; 16:124. [PMID: 38201551 PMCID: PMC10778447 DOI: 10.3390/cancers16010124] [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: 12/05/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Breast cancer remains a significant health challenge, and novel treatment approaches are critically needed. This review presents an in-depth analysis of engineered adoptive T-cell therapies (E-ACTs), an innovative frontier in cancer immunotherapy, focusing on their application in breast cancer. We explore the evolving landscape of chimeric antigen receptor (CAR) and T-cell receptor (TCR) T-cell therapies, highlighting their potential and challenges in targeting breast cancer. The review addresses key obstacles such as target antigen selection, the complex breast cancer tumor microenvironment, and the persistence of engineered T-cells. We discuss the advances in overcoming these barriers, including strategies to enhance T-cell efficacy. Finally, our comprehensive analysis of the current clinical trials in this area provides insights into the future possibilities and directions of E-ACTs in breast cancer treatment.
Collapse
Affiliation(s)
- Diego F. Chamorro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
| | - Lauren K. Somes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
| | - Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
32
|
Steinbach M, Zitella LJ, Florendo E, Lee E, Riccobono C, DiFilippo H, Aronson E. Nursing Care Throughout the Chimeric Antigen Receptor T-Cell Therapy Process for Multiple Myeloma. Semin Oncol Nurs 2023; 39:151505. [PMID: 37752030 DOI: 10.1016/j.soncn.2023.151505] [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: 06/02/2023] [Revised: 08/03/2023] [Accepted: 08/26/2023] [Indexed: 09/28/2023]
Abstract
OBJECTIVES Approvals of chimeric antigen receptor T-cell (CAR-T) therapies for relapsed/refractory multiple myeloma (RRMM) represent advancements in treatment options for a hard-to-treat population. Nursing care during CAR-T therapy is crucial for patients, their caregivers, and the broader CAR-T therapy care team. This manuscript provides an overview of the CAR-T therapy administration process and describes practical considerations for nursing professionals working with patients who receive CAR-T therapy. DATA SOURCES Current literature describing CAR-T therapies for RRMM and published guidelines on nursing care during CAR-T therapy administration were identified from a PubMed database search. Literature was synthesized with practical considerations from nurses and nurse practitioners with expertise in the administration of CAR-T therapy for MM. A practical overview of the role of nursing professionals throughout all stages of CAR-T therapy administration for RRMM is provided. CONCLUSION Planning, administration, and posttreatment monitoring for CAR-T therapy requires collaboration between nursing professionals and other healthcare providers as patients migrate between community oncology providers and specialized treatment centers. Nurses help with assessment of patient eligibility and patient and caregiver education before CAR-T therapy. They act in diverse roles across various settings involved in CAR-T therapy administration. Finally, nurses contribute to long-term identification and management of CAR-T-associated toxicities. IMPLICATIONS FOR NURSING PRACTICE Nurses are crucial to the CAR-T therapy process and make significant contributions to optimizing patient care and subsequent outcomes.
Collapse
Affiliation(s)
- Mary Steinbach
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Laura J Zitella
- Hematology, Blood & Marrow Transplant, and Cellular Therapies Program, University of California San Francisco, San Francisco, CA
| | | | - Erin Lee
- Janssen Pharmaceuticals, Inc, Titusville, NJ
| | | | - Heather DiFilippo
- Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
33
|
Chen RQ, Joffe B, Casteleiro Costa P, Filan C, Wang B, Balakirsky S, Robles F, Roy K, Li J. Real-time semantic segmentation and anomaly detection of functional images for cell therapy manufacturing. Cytotherapy 2023; 25:1361-1369. [PMID: 37725031 PMCID: PMC10719834 DOI: 10.1016/j.jcyt.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND AIMS Cell therapy is a promising treatment method that uses living cells to address a variety of diseases and conditions, including cardiovascular diseases, neurologic disorders and certain cancers. As interest in cell therapy grows, there is a need to shift to a more efficient, scalable and automated manufacturing process that can produce high-quality products at a lower cost. METHODS One way to achieve this is using non-invasive imaging and real-time image analysis techniques to monitor and control the manufacturing process. This work presents a machine learning-based image analysis pipeline that includes semantic segmentation and anomaly detection capabilities. RESULTS/CONCLUSIONS This method can be easily implemented even when given a limited dataset of annotated images, is able to segment cells and debris and can identify anomalies such as contamination or hardware failure.
Collapse
Affiliation(s)
- Rui Qi Chen
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Benjamin Joffe
- Georgia Tech Research Institute, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Paloma Casteleiro Costa
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, Georgia, USA
| | - Caroline Filan
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, Georgia, USA
| | - Bryan Wang
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, Georgia, USA
| | - Stephen Balakirsky
- Georgia Tech Research Institute, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Francisco Robles
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, Georgia, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, Georgia, USA
| | - Jing Li
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
| |
Collapse
|
34
|
McGough SF, Shamas N, Wang J, Jaber M, Swarup B, Blanchet Zumofen MH, Lautié B, Parreira J, Wei MC, Shewade A. Comparative effectiveness between mosunetuzumab monotherapy clinical trial and real-world data in relapsed/refractory follicular lymphoma in third or subsequent lines of systemic therapy. Leuk Lymphoma 2023; 64:2269-2278. [PMID: 37840271 DOI: 10.1080/10428194.2023.2262066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023]
Abstract
A comparison of clinical outcomes in the third or subsequent line (3 L+) of systemic therapy between a real-world data (RWD) external control cohort and a mosunetuzumab single-arm clinical trial cohort is presented. Data for 3 L + patients with relapsed/refractory follicular lymphoma (FL) were obtained from the mosunetuzumab single-arm trial (n = 90) and a US electronic health records database (n = 158), with patients meeting key eligibility criteria from the trial, balanced on pre-specified prognostic factors. Overall response and complete response rates were 80% and 60% in the mosunetuzumab cohort and 75% and 33% in the RWD cohort, odds ratios of 1.23 (95% CI, 0.52-2.93) and 3.18 (95% CI, 1.41-7.17), respectively. Hazard ratios for progression-free survival and overall survival were 0.82 (95% CI, 0.53-1.27) and 0.43 (95% CI, 0.19-0.94). These findings support a clinically meaningful benefit of mosunetuzumab monotherapy as a chemotherapy-free option for the 3 L + FL population.
Collapse
Affiliation(s)
| | | | - Jue Wang
- Genentech, Inc., South San Francisco, California, USA
| | | | | | | | | | | | - Michael C Wei
- Genentech, Inc., South San Francisco, California, USA
| | | |
Collapse
|
35
|
Bell JAH, Jeffries GA, Chen CI. Mitigating inequity: ethically prioritizing patients for CAR T-cell therapy. Blood 2023; 142:1263-1270. [PMID: 37540818 DOI: 10.1182/blood.2023020703] [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: 05/18/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 08/06/2023] Open
Abstract
Manufacturing capacity and institutional infrastructure to deliver chimeric antigen receptor T-cell therapies (CAR-T) are pressured to keep pace with the growing number of approved products and expanding eligible patient population for this potentially life-saving therapy. Consequently, many cell therapy programs must make difficult decisions about which patient should get the next available treatment slot. This situation requires an ethical framework to ensure fair and equitable decision-making. In this perspective, we discuss the application of Accountability for Reasonableness (A4R), a priority-setting framework grounded in procedural justice, to the problem of limited CAR-T slots at our institution. We formed a multidisciplinary working group spanning several hematological malignancies. Through multiple rounds of partner engagement, we used A4R guiding principles to identify 4 main criteria to prioritize patients for CAR-T: medical benefit, safety/risk of complications, psychosocial factors, and medical urgency. Associated measures/tools and an implementation process were developed. We discuss further how ethical principles of fairness and equity demand a consistent approach within health systems that does not disadvantage medically underserved or underrepresented populations and supports overcoming barriers to care. In our commitment to transparency and collaboration, we make our tools available to others, ideally to be used to engage in their own A4R process, adapting the tools to their unique environments. Our hope is that our preliminary work will support the advancement of further study in this area globally, aiming for justice in resource allocation for all potential CAR-T candidates, wherever they may seek care.
Collapse
Affiliation(s)
- Jennifer A H Bell
- Department of Clinical and Organizational Ethics, University Health Network, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Joint Centre for Bioethics, University of Toronto, Toronto, ON, Canada
- The Institute for Education Research, University Health Network, Toronto, ON, Canada
- Department of Supportive Care, Research Division, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Christine I Chen
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
36
|
Monberg TJ, Borch TH, Svane IM, Donia M. TIL Therapy: Facts and Hopes. Clin Cancer Res 2023; 29:3275-3283. [PMID: 37058256 DOI: 10.1158/1078-0432.ccr-22-2428] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/20/2023] [Accepted: 03/31/2023] [Indexed: 04/15/2023]
Abstract
After a positive phase III trial, it is evident that treatment with tumor-infiltrating lymphocytes (TIL) is a safe, feasible, and effective treatment modality for patients with metastatic melanoma. Further, the treatment is safe and feasible in diverse solid tumors, regardless of the histologic type. Still, TIL treatment has not obtained the regulatory approvals to be implemented on a larger scale. Therefore, its availability is currently restricted to a few centers worldwide. In this review, we present the current knowledge of TIL therapy and discuss the practical, logistic, and economic challenges associated with implementing TIL therapy on a larger scale. Finally, we suggest strategies to facilitate the widespread implementation of TIL therapy and approaches to develop the next generation of TILs.
Collapse
Affiliation(s)
- Tine J Monberg
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark
| | - Troels H Borch
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark
| | - Inge M Svane
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark
| | - Marco Donia
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
37
|
Bixby TJ, Brittle CJ, Mangan PA, Stadtmauer EA, Kallenbach LR. Patient Perceptions of CAR-T Therapy in the USA: Findings from In-Depth Interviews. Oncol Ther 2023; 11:303-312. [PMID: 37210682 PMCID: PMC10200031 DOI: 10.1007/s40487-023-00232-9] [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: 03/24/2023] [Accepted: 05/03/2023] [Indexed: 05/22/2023] Open
Abstract
INTRODUCTION Chimeric antigen receptor-T cell (CAR-T) therapy has revolutionized advanced blood cancer treatment. However, preparation, administration, and recovery from these therapies can be complex and burdensome to patients and their care partners. Utilization of an outpatient setting for CAR-T therapy administration could help improve convenience and quality of life. METHODS In-depth qualitative interviews were conducted with 18 patients in the USA with relapsed/refractory multiple myeloma or relapsed/refractory diffuse large B-cell lymphoma, 10 of whom had completed investigational or commercially approved CAR-T therapy and 8 of whom had discussed it with their physicians. We aimed to better understand inpatient experiences and patient expectations regarding CAR-T therapy and to ascertain patient perspectives on the possibility of outpatient care. RESULTS CAR-T offers unique treatment benefits, particularly high response rates with an extended treatment-free period. All study participants completing CAR-T were very positive about their inpatient recovery experience. Most reported mild-to-moderate side effects; two experienced severe side effects. All said that they would opt to undergo CAR-T therapy again. Participants felt that the primary advantage of inpatient recovery was immediate access to care and on-going monitoring. Perceived advantages of the outpatient setting were comfort and familiarity. Because immediate access to care was seen as crucial, patients recovering in an outpatient setting would seek either a direct contact person or phone line for assistance if needed. CONCLUSION As institutions become more experienced with CAR-T therapies, outpatient care may help reduce financial strain. Patient input can help institutions improve the outpatient experience and ensure safety and effectiveness of CAR-T programs.
Collapse
Affiliation(s)
- Todd J Bixby
- Janssen Scientific Affairs, LLC, 800 Ridgeview Dr, Horsham, PA, 19044, USA.
| | | | - Patricia A Mangan
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Edward A Stadtmauer
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Lisa R Kallenbach
- Janssen Scientific Affairs, LLC, 800 Ridgeview Dr, Horsham, PA, 19044, USA
| |
Collapse
|
38
|
Sauter CS, Hill BT. Rush Hour Update: Roadblocks to CARs on the Expressway. Transplant Cell Ther 2023; 29:408-409. [PMID: 37400191 DOI: 10.1016/j.jtct.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Affiliation(s)
- Craig S Sauter
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
39
|
Cliff ERS, Kelkar AH, Russler-Germain DA, Tessema FA, Raymakers AJN, Feldman WB, Kesselheim AS. High Cost of Chimeric Antigen Receptor T-Cells: Challenges and Solutions. Am Soc Clin Oncol Educ Book 2023; 43:e397912. [PMID: 37433102 DOI: 10.1200/edbk_397912] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Chimeric antigen receptor (CAR) T-cells are a cellular immunotherapy with remarkable efficacy in treating multiple hematologic malignancies but they are associated with extremely high prices that are, for many countries, prohibitively expensive. As their use increases both for hematologic malignancies and other indications, and large numbers of new cellular therapies are developed, novel approaches will be needed both to reduce the cost of therapy, and to pay for them. We review the many factors that lead to the high cost of CAR T-cells and offer proposals for reform.
Collapse
Affiliation(s)
- Edward R Scheffer Cliff
- Program on Regulation, Therapeutics, and Law, Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Amar H Kelkar
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - David A Russler-Germain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO
| | - Frazer A Tessema
- Program on Regulation, Therapeutics, and Law, Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Adam J N Raymakers
- Program on Regulation, Therapeutics, and Law, Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - William B Feldman
- Program on Regulation, Therapeutics, and Law, Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Aaron S Kesselheim
- Program on Regulation, Therapeutics, and Law, Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
40
|
Yusuf RA, Preussler JM, Meyer CL, Schoeppner K, Sees Coles JA, Ruffin A, McCann M, Devine SM, Auletta JJ. Reducing barriers of access and care related to hematopoietic cell transplantation and cellular therapy: The mission-driven role of the national marrow donor program. Best Pract Res Clin Haematol 2023; 36:101480. [PMID: 37353289 DOI: 10.1016/j.beha.2023.101480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/25/2023]
Abstract
The treatment of malignant and nonmalignant hematologic disorders continues to benefit from significant scientific advancement and progress in the use of hematopoietic cell transplantation and cellular therapies. However, barriers associated with receiving these lifesaving treatments and care remain, which necessitate innovative approaches to overcome, so all persons in need can receive these therapies. This article reviews barriers to receiving hematopoietic cell transplantation and cellular therapies, and highlights novel approaches taken by the National Marrow Donor Program in reducing barriers for all patients in need.
Collapse
Affiliation(s)
- Rafeek A Yusuf
- National Marrow Donor Program, Minneapolis, MN, USA; Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA.
| | - Jaime M Preussler
- National Marrow Donor Program, Minneapolis, MN, USA; Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA.
| | - Christa L Meyer
- National Marrow Donor Program, Minneapolis, MN, USA; Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA.
| | | | | | - Amber Ruffin
- National Marrow Donor Program, Minneapolis, MN, USA.
| | - Meggan McCann
- National Marrow Donor Program, Minneapolis, MN, USA.
| | - Steven M Devine
- National Marrow Donor Program, Minneapolis, MN, USA; Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA.
| | - Jeffery J Auletta
- National Marrow Donor Program, Minneapolis, MN, USA; Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA.
| |
Collapse
|
41
|
Zafar A, Huang CY, Lo M, Arora S, Chung A, Wong SW, Wolf J, Martin T, Shah N, Banerjee R. Intensity of cyclophosphamide-based bridging therapy before CAR-T therapy in myeloma. Transplant Cell Ther 2023:S2666-6367(23)01299-X. [PMID: 37244643 DOI: 10.1016/j.jtct.2023.05.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Patients receiving autologous chimeric antigen receptor T-cell (CAR-T) therapies for multiple myeloma (MM) may require bridging therapy (BT) before CAR-T infusion to maintain some level of disease control. Alkylators such as cyclophosphamide (Cy) are often used as parts of BT regimens, either in high-intensity regimens such as modified hyperCVAD or once-weekly regimens such as KCd. However, there is no consensus around the optimal BT alkylator dose intensity in MM. METHODS We performed a single-center analysis of all instances of BT before planned autologous CAR-T for MM during a 5-year period ending April 2022. We classified bridging regimens into three cohorts: (1) hyperfractionated Cy (HyperCy) with inpatient Cy every 12-24 hours or as continuous intravenous infusions, (2) less intensive Cy dosing (WeeklyCy) such as KPd or KCd, and (3) NonCy, where no alkylators were used in BT. Demographic, disease-related, and treatment-related characteristics were collected for all patients. The three BT cohorts were compared using Fisher's exact tests, Kruskal-Wallis tests, and log-rank tests where appropriate. RESULTS We identified 70 discrete BT instances among 64 unique patients: 29 (41%) with HyperCy, 23 (33%) with WeeklyCy, and 18 (26%) with NonCy. Median total levels of Cy dosing during BT were 2100 mg/m2, 615 mg/m2, and 0 mg/m2 respectively. Age, number of prior lines, triple-class refractory status, presence of high-risk cytogenetics, extramedullary disease, bone marrow plasma cell burden, involved free light chain (iFLC) kinetics before collection, and other measures of disease aggressiveness were comparable between cohorts. iFLC levels rose ≥25% and ≥100 mg/L during BT (approximating progressive disease) in comparable proportions (p=0.25) between cohorts: 52% HyperCy, 39% WeeklyCy, and 28% NonCy. All BT instances without subsequent CAR-T were due to manufacturing failures. Among 61 instances of bridging followed by CAR-T, vein-to-vein times were slightly longer (p = 0.03) with HyperCy (45 days) vs WeeklyCy (39 days) and NonCy (46.5 days). Neutrophil recovery timeframes were similar between cohorts, but platelet recovery took longer with HyperCy (64 days) vs WeeklyCy (42 days) and NonCy (12 days). PFS was comparable between cohorts, but median OS was not: 15.3 months with HyperCy versus 30.0 months with WeeklyCy and not reached with NonCy. DISCUSSION In our retrospective analysis of BT before CAR-T therapy in MM, HyperCy did not result in superior disease control than WeeklyCy despite a threefold higher dose of Cy. In contrast, HyperCy was associated with longer post-CAR-T platelet recovery and worsened OS despite comparable measurements of disease aggressiveness and tumor burden. Study limitations include our small sample size as well as confounding from gestalt markers of MM aggressiveness that might have led both to poorer outcomes as well as physicians' decisions to prescribe HyperCy. Given the rarity of objective disease responses to chemotherapy in relapsed/refractory MM, our analysis suggests that hyperfractionated Cy regimens do not outperform once-weekly Cy regimens for most patients who require BT before CAR-T therapy.
Collapse
Affiliation(s)
- Aneeqa Zafar
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA
| | - Chiung-Yu Huang
- Department of Epidemiology and Biostatistics, UCSF San Francisco, CA
| | - Mimi Lo
- Division of Hematology and Oncology, Department of Pharmacy, UCSF, San Francisco, CA
| | - Shagun Arora
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA
| | - Alfred Chung
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA
| | - Sandy W Wong
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA
| | - Jeffrey Wolf
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA
| | - Thomas Martin
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA
| | - Nina Shah
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA
| | - Rahul Banerjee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA.
| |
Collapse
|
42
|
Ahmed N, Mahmoudjafari Z, Hamadani M, Hashmi H. Bridging the gap: Early transition and hybrid models of care to improve access to CAR-T cell therapy. Transplant Cell Ther 2023:S2666-6367(23)01197-1. [PMID: 36990223 DOI: 10.1016/j.jtct.2023.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Affiliation(s)
- Nausheen Ahmed
- Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, University of Kansas Cancer Center, Westwood, Kansas.
| | - Zahra Mahmoudjafari
- Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, University of Kansas Cancer Center, Westwood, Kansas
| | - Mehdi Hamadani
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Hamza Hashmi
- Hematology Oncology, Department of Medicine, Medical University of South Carolina, South Carolina
| |
Collapse
|
43
|
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
|