1
|
Marshall K, Mastro M, Nankervis B, Shahid S, Ciasullo G, Smith T, Loveras M, Smith D, Miller MM, Gibb SL. Rapid manufacture of low-seed CAR-T cells in a GMP-grade hollow-fiber bioreactor platform. Cytotherapy 2024:S1465-3249(24)00927-7. [PMID: 39601749 DOI: 10.1016/j.jcyt.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/23/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024]
Abstract
Both quality of product and rapidity of manufacture are critical parameters if ex vivo manufacturing of autologous chimeric antigen receptor T cell (CAR-T) therapies is to reach its full potential. The Quantum Flex Cell Expansion System from Terumo Blood and Cell Technologies (Terumo BCT), a hollow-fiber bioreactor platform, is one of several cell expansion systems available to cell and gene therapy manufacturers to generate such cells in a GMP-compliant manner. In this study, the dynamic range of the Quantum Flex platform to expand CD19 CAR-T cells from variable quantities of starting material was investigated. Reflecting the industry's utilization of contract development manufacturing organizations (CDMOs) for accelerating clinical timelines, Terumo Blood and Cell Technologies performed a technology transfer of application protocols for study execution. Four different amounts of starting material (1, 3, 6 and 15 million cells) were expanded on Quantum Flex, using a unique donor's cells for each run. In this study, CAR-T cells were created using commercially obtained T cells and an anti-CD19 CAR-T lentiviral construct. The resultant heterogenous cell populations were expanded for 7 days in the functionally closed bioreactor platform. Expansion kinetics for all 4 starting material amounts were remarkedly similar, resulting in a 150- to 200-fold increase in cell numbers. This allowed for a study maximum of 2.6 billion cells from loading 15 million cells. Viability remained high throughout the expansion process with >93% for all 4 donors at harvest. To complete the manufacturing cycle, the automated and functionally closed Finia Fill and Finish System (Terumo Blood and Cell Technologies, Lakewood, CO) was used to formulate the cells for cryopreservation. Postprocedure analysis for potency and cytotoxicity demonstrated the production of efficacious cells. With this range of starting numbers, the platform is relevant to adult, pediatric and compassionate CAR-T expansion dosing. Today, several platforms are available to achieve sufficient cell yields for therapeutic applications of CAR-T, and awareness of the capabilities, pros and cons of each platform is critical to drive progress.
Collapse
Affiliation(s)
- Kurt Marshall
- Terumo Blood and Cell Technologies, Lakewood, Colorado, USA
| | | | | | - Shahid Shahid
- BioCentriq, Inc., Monmouth Junction, New Jersey, USA
| | | | - Trevor Smith
- Terumo Blood and Cell Technologies, Lakewood, Colorado, USA
| | - Mary Loveras
- BioCentriq, Inc., Monmouth Junction, New Jersey, USA
| | - David Smith
- BioCentriq, Inc., Monmouth Junction, New Jersey, USA
| | - Mindy M Miller
- Terumo Blood and Cell Technologies, Lakewood, Colorado, USA.
| | - Stuart L Gibb
- Terumo Blood and Cell Technologies, Lakewood, Colorado, USA.
| |
Collapse
|
2
|
Locatelli F, del Bufalo F, Quintarelli C. Allogeneic chimeric antigen receptor T cells for children with relapsed/refractory B-cell precursor acute lymphoblastic leukemia. Haematologica 2024; 109:1689-1699. [PMID: 38832424 PMCID: PMC11141659 DOI: 10.3324/haematol.2023.284604] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 02/01/2024] [Indexed: 06/05/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as a breakthrough cancer therapy over the past decade. Remarkable outcomes in B-cell lymphoproliferative disorders and multiple myeloma have been reported in both pivotal trials and real-word studies. Traditionally, the use of a patient's own (autologous) T cells to manufacture CAR products has been the standard practice. Nevertheless, this approach has some drawbacks, including manufacturing delays, dependence on the functional fitness of the patient's T cells, which can be compromised by both the disease and prior therapies, and contamination of the product with blasts. A promising alternative is offered by the development of allogeneic CAR-cell products. This approach has the potential to yield more efficient drug products and enables the use of effector cells with negligible alloreactive potential and a significant CAR-independent antitumor activity through their innate receptors (i.e., natural killer cells, γδ T cells and cytokine induced killer cells). In addition, recent advances in genome editing tools offer the potential to overcome the primary challenges associated with allogeneic CAR T-cell products, namely graft-versus-host disease and host allo-rejection, generating universal, off-the-shelf products. In this review, we summarize the current pre-clinical and clinical approaches based on allogeneic CAR T cells, as well as on alternative effector cells, which represent exciting opportunities for multivalent approaches and optimized antitumor activity.
Collapse
Affiliation(s)
- Franco Locatelli
- Department of Hematology/Oncology, Cell and Gene Therapy – IRCCS, Bambino Gesù Children’s Hospital, Rome
- Catholic University of the Sacred Heart, Department of Life Sciences and Public Health, Rome
| | - Francesca del Bufalo
- Department of Hematology/Oncology, Cell and Gene Therapy – IRCCS, Bambino Gesù Children’s Hospital, Rome
| | - Concetta Quintarelli
- Department of Hematology/Oncology, Cell and Gene Therapy – IRCCS, Bambino Gesù Children’s Hospital, Rome
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
3
|
Mishra AK, Burridge S, Espuelas MO, O'Reilly M, Cummins M, Nicholson E, Wheldon S, Bonney D, Shenton G, Marks DI, Amrolia PJ, Hough R, Ghorashian S. Practice guideline: Preparation for CAR T-cell therapy in children and young adults with B-acute lymphoblastic leukaemia. Br J Haematol 2024; 204:1687-1696. [PMID: 38488312 DOI: 10.1111/bjh.19381] [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: 12/05/2023] [Revised: 02/02/2024] [Accepted: 02/21/2024] [Indexed: 05/15/2024]
Abstract
The objective of this guideline, prepared by the ALL subgroup of the Advanced Cell Therapy Sub-Committee of BSBMTCT (British Society of Blood and Marrow Transplantation), is to provide healthcare professionals with practical guidance on the preparation of children and young adults with B-acute lymphoblastic leukaemia from the point of referral to that of admission for CAR T-cell treatment. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) nomenclature was used to evaluate the levels of evidence and to assess the strength of recommendations. The GRADE criteria can be found at http://www.gradeworkinggroup.org.
Collapse
Affiliation(s)
- Avijeet Kumar Mishra
- Great Ormond Street Hospital for Children, London, UK
- University College London, London, UK
| | | | | | | | | | | | | | - Denise Bonney
- Royal Manchester Children's Hospital, Manchester, UK
| | - Geoff Shenton
- Great North Children's Hospital, Newcastle University, Newcastle upon Tyne, UK
| | - David I Marks
- University Hospitals Bristol and Weston NHS Trust, Bristol, UK
| | - Persis J Amrolia
- Great Ormond Street Hospital for Children, London, UK
- University College London, London, UK
| | - Rachael Hough
- University College London, London, UK
- University College London Hospital, London, UK
| | - Sara Ghorashian
- Great Ormond Street Hospital for Children, London, UK
- University College London, London, UK
| |
Collapse
|
4
|
Kitamura W, Urata T, Fujii K, Fukumi T, Ikeuchi K, Seike K, Fujiwara H, Asada N, Ennishi D, Matsuoka KI, Otsuka F, Maeda Y, Fujii N. Collection efficiency and safety of large-volume leukapheresis for the manufacturing of tisagenlecleucel. Transfusion 2024; 64:674-684. [PMID: 38419458 DOI: 10.1111/trf.17765] [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: 12/20/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND In patients with relapsed or refractory B cell acute lymphoblastic leukemia or B cell non-Hodgkin lymphoma (r/r B-ALL/B-NHL) with low CD3+ cells in the peripheral blood (PB), sufficient CD3+ cell yield in a single day may not be obtained with normal-volume leukapheresis (NVL). Large-volume leukapheresis (LVL) refers to the processing of more than three times the total blood volume (TBV) in a single session for PB apheresis; however, the efficiency and safety of LVL for manufacturing of tisagenlecleucel (tisa-cel) remain unclear. This study aimed to investigate the tolerability of LVL. STUDY DESIGN AND METHODS We retrospectively collected data on LVL (≥3-fold TBV) and NVL (<3-fold TBV) performed for patients with r/r B-ALL/B-NHL in our institution during November 2019 and September 2023. All procedures were performed using a continuous mononuclear cell collection (cMNC) protocol with the Spectra Optia. RESULTS Although pre-apheresis CD3+ cells in the PB were significantly lower in LVL procedures (900 vs. 348/μL, p < .01), all patients could obtain sufficient CD3+ cell yield in a single day with a comparably successful rate of final products (including out-of-specification) between the two groups (97.2% vs. 100.0%, p = 1.00). The incidence and severity of citrate toxicity (no patients with grade ≥ 3) during procedures was not significantly different between the two groups (22.2% vs. 26.1%, p = .43) and no patient discontinued leukapheresis due to any complications. CONCLUSION LVL procedures using Spectra Optia cMNC protocol was well tolerated and did not affect the manufacturing of tisa-cel.
Collapse
Affiliation(s)
- Wataru Kitamura
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Division of Blood Transfusion, Okayama University Hospital, Okayama, Japan
| | - Tomohiro Urata
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Division of Blood Transfusion, Okayama University Hospital, Okayama, Japan
| | - Keiko Fujii
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Division of Clinical Laboratory, Okayama University Hospital, Okayama, Japan
| | - Takuya Fukumi
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Division of Blood Transfusion, Okayama University Hospital, Okayama, Japan
| | - Kazuhiro Ikeuchi
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Division of Blood Transfusion, Okayama University Hospital, Okayama, Japan
| | - Keisuke Seike
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Daisuke Ennishi
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Fumio Otsuka
- Division of Clinical Laboratory, Okayama University Hospital, Okayama, Japan
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Nobuharu Fujii
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Division of Blood Transfusion, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
5
|
Fiuza-Luces C, Valenzuela PL, Gálvez BG, Ramírez M, López-Soto A, Simpson RJ, Lucia A. The effect of physical exercise on anticancer immunity. Nat Rev Immunol 2024; 24:282-293. [PMID: 37794239 DOI: 10.1038/s41577-023-00943-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/06/2023]
Abstract
Regular physical activity is associated with lower cancer incidence and mortality, as well as with a lower rate of tumour recurrence. The epidemiological evidence is supported by preclinical studies in animal models showing that regular exercise delays the progression of cancer, including highly aggressive malignancies. Although the mechanisms underlying the antitumorigenic effects of exercise remain to be defined, an improvement in cancer immunosurveillance is likely important, with different immune cell subtypes stimulated by exercise to infiltrate tumours. There is also evidence that immune cells from blood collected after an exercise bout could be used as adoptive cell therapy for cancer. In this Perspective, we address the importance of muscular activity for maintaining a healthy immune system and discuss the effects of a single bout of exercise (that is, 'acute' exercise) and those of 'regular' exercise (that is, repeated bouts) on anticancer immunity, including tumour infiltrates. We also address the postulated mechanisms and the clinical implications of this emerging area of research.
Collapse
Affiliation(s)
- Carmen Fiuza-Luces
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain.
| | - Pedro L Valenzuela
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
- Systems Biology Department, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Beatriz G Gálvez
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Ramírez
- Oncohematology Unit, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Biomedical Research Foundation, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Heah, Madrid, Spain
| | - Alejandro López-Soto
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Asturias, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.
| | - Richard J Simpson
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, USA
- Department of Paediatrics, The University of Arizona, Tucson, AZ, USA
- Department of Immunobiology, The University of Arizona, Tucson, AZ, USA
| | - Alejandro Lucia
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain.
- Faculty of Sport Sciences, Universidad Europea, Madrid, Spain.
| |
Collapse
|
6
|
Knight E T, Oluwole O, Kitko C. The Implementation of Chimeric Antigen Receptor (CAR) T-cell Therapy in Pediatric Patients: Where Did We Come From, Where Are We Now, and Where are We Going? Clin Hematol Int 2024; 6:96-115. [PMID: 38817691 PMCID: PMC11108586 DOI: 10.46989/001c.94386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/13/2024] [Indexed: 06/01/2024] Open
Abstract
CD19-directed Chimeric Antigen Receptor (CAR) T-cell therapy has revolutionized the treatment of patients with B-cell acute lymphoblastic leukemia (B-ALL). Somewhat uniquely among oncologic clinical trials, early clinical development occurred simultaneously in both children and adults. In subsequent years however, the larger number of adult patients with relapsed/refractory (r/r) malignancies has led to accelerated development of multiple CAR T-cell products that target a variety of malignancies, resulting in six currently FDA-approved for adult patients. By comparison, only a single CAR-T cell therapy is approved by the FDA for pediatric patients: tisagenlecleucel, which is approved for patients ≤ 25 years with refractory B-cell precursor ALL, or B-cell ALL in second or later relapse. Tisagenlecleucel is also under evaluation in pediatric patients with relapsed/refractory B-cell non-Hodgkin lymphoma, but is not yet been approved for this indication. All the other FDA-approved CD19-directed CAR-T cell therapies available for adult patients (axicabtagene ciloleucel, brexucabtagene autoleucel, and lisocabtagene maraleucel) are currently under investigations among children, with preliminary results available in some cases. As the volume and complexity of data continue to grow, so too does the necessity of rapid assimilation and implementation of those data. This is particularly true when considering "atypical" situations, e.g. those arising when patients do not precisely conform to the profile of those included in pivotal clinical trials, or when alternative treatment options (e.g. hematopoietic stem cell transplantation (HSCT) or bispecific T-cell engagers (BITEs)) are also available. We have therefore developed a relevant summary of the currently available literature pertaining to the use of CD19-directed CAR-T cell therapies in pediatric patients, and sought to provide guidance for clinicians seeking additional data about specific clinical situations.
Collapse
Affiliation(s)
| | - Olalekan Oluwole
- Medicine Hematology and Oncology, Vanderbilt University Medical Center
| | | |
Collapse
|
7
|
Pessach I, Nagler A. Leukapheresis for CAR-T cell production and therapy. Transfus Apher Sci 2023; 62:103828. [PMID: 37838564 DOI: 10.1016/j.transci.2023.103828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an effective, individualized immunotherapy, and novel treatment for hematologic malignancies. Six commercial CAR-T cell products are currently approved for lymphatic malignancies and multiple myeloma. In addition, an increasing number of clinical centres produce CAR-T cells on-site, which enable the administration of CAR-T cells on site. The CAR-T cell products are either fresh or cryopreserved. Manufacturing CAR-T cells is a complicated process that begins with leukapheresis to obtain T cells from the patient's peripheral blood. An optimal leukapheresis product is crucial step for a successful CAR-T cell therapy; therefore, it is imperative to understand the factors that may affect the quality or T cells. The leukapheresis for CAR-T cell production is well tolerated and safe for both paediatric and adult patients and CAR-Τ cell therapy presents high clinical response rate in many studies. CAR-T cell therapy is under continuous improvement, and it has transformed into an almost standard procedure in clinical haematology and stem cell transplantation facilities that provide both autologous and allogeneic stem cell transplantations. In patients suffering from advanced haematological malignancies, CAR-T cell therapy shows incredible antitumor efficacy. Even after a single infusion of autologous CD19-targeting CAR-T cells in patients with relapsed or refractory diffuse large B cell lymphoma (DLBCL) and acute lymphoblastic leukaemia (ALL), long lasting remission is observed, and a fraction of the patients are being cured. Future novel constructs are being developed with better T cell persistence and better expansion. New next-generation CAR-T cells are currently designed to avoid toxicities such as cytokine release syndrome and neurotoxicity.
Collapse
Affiliation(s)
- Ilias Pessach
- Hematology Department, Athens Medical Center, Athens, Greece
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Israel.
| |
Collapse
|
8
|
O'Reilly MA, Malhi A, Cheok KPL, Ings S, Balsa C, Keane H, Jalowiec K, Neill L, Peggs KS, Roddie C. A novel predictive algorithm to personalize autologous T-cell harvest for chimeric antigen receptor T-cell manufacture. Cytotherapy 2023; 25:323-329. [PMID: 36513573 DOI: 10.1016/j.jcyt.2022.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AIMS The most widely accepted starting materials for chimeric antigen receptor T-cell manufacture are autologous CD3+ T cells obtained via the process of leukapheresis, also known as T-cell harvest. As this treatment modality gains momentum and apheresis units struggle to meet demand for harvest slots, strategies to streamline this critical step are warranted. METHODS This retrospective review of 262 T-cell harvests, with a control cohort of healthy donors, analyzed the parameters impacting CD3+ T-cell yield in adults with B-cell malignancies. The overall aim was to design a novel predictive algorithm to guide the required processed blood volume (PBV) (L) on the apheresis machine to achieve a specific CD3+ target yield. RESULTS Factors associated with CD3+ T-cell yield on multivariate analysis included peripheral blood CD3+ count (natural log, ×109/L), hematocrit (HCT) and PBV with coefficients of 0.86 (95% confidence interval [CI], 0.80-0.92, P < 0.001), 1.30 (95% CI, 0.51-2.08, P = 0.001) and 0.09 (95% CI, 0.07-0.11, P < 0.001), respectively. The authors' model, incorporating CD3+ cell count, HCT and PBV (L), with an adjusted R2 of 0.87 and root-mean-square error of 0.26 in the training dataset, was highly predictive of CD3+ cell yield in the testing dataset. An online application to estimate PBV using this algorithm can be accessed at https://cd3yield.shinyapps.io/cd3yield/. CONCLUSIONS The authors propose a transferrable model that incorporates clinical and laboratory variables accessible pre-harvest for use across the field of T-cell therapy. Pending further validation, such a model may be used to generate an individual leukapheresis plan and streamline the process of cell harvest, a well-recognized bottleneck in the industry.
Collapse
Affiliation(s)
- Maeve A O'Reilly
- University College London Cancer Institute, London, UK; Department of Hematology, University College London Hospital, London, UK. maeve.o'
| | - Aman Malhi
- Cancer Research UK & University College London Cancer Trials Center, University College London, London, UK
| | - Kathleen P L Cheok
- Department of Hematology, University College London Hospital, London, UK
| | - Stuart Ings
- Department of Hematology, University College London Hospital, London, UK
| | - Carmen Balsa
- Department of Hematology, University College London Hospital, London, UK
| | - Helen Keane
- Department of Hematology, University College London Hospital, London, UK
| | - Katarzyna Jalowiec
- Department of Hematology, University College London Hospital, London, UK
| | - Lorna Neill
- Department of Hematology, University College London Hospital, London, UK
| | - Karl S Peggs
- University College London Cancer Institute, London, UK; Department of Hematology, University College London Hospital, London, UK
| | - Claire Roddie
- University College London Cancer Institute, London, UK; Department of Hematology, University College London Hospital, London, UK
| |
Collapse
|
9
|
Piñeyroa JA, Cid J, Vlagea A, Carbassé G, Henao P, Bailo N, Ortiz-Maldonado V, Martínez-Cibrian N, Español M, Delgado J, Urbano-Ispizua Á, Lozano M. Evaluation of cell collection efficiency in non-mobilized adult donors for autologous chimeric antigen receptor T-cell manufacturing. Vox Sang 2023; 118:217-222. [PMID: 36516201 DOI: 10.1111/vox.13394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/08/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES Data about collection efficiency 1 (CE1), which takes into account blood cell counts before and after collection, thus providing a more accurate estimate, in the collection of autologous T lymphocytes by apheresis for chimeric antigen receptor (CAR) T-cells remain scarce. We evaluated donor- and procedure-related characteristics that might influence the CE1 of lymphocytes. MATERIALS AND METHODS We retrospectively reviewed all mononuclear cell (MNC) collections) performed for CAR T-cell manufacturing in our institution from May 2017 to June 2021 in adult patients. Age, gender, weight, total blood volume (TBV), prior haematopoietic cell transplant, diagnosis, days between last treatment and apheresis, pre-collection cell counts, duration of apheresis, TBV processed, vascular access, inlet flow and device type were analysed as potential factors affecting CE1 of lymphocytes. RESULTS A total of 127 autologous MNC collections were performed on 118 patients diagnosed with acute lymphoblastic leukaemia (n = 53, 45%), non-Hodgkin lymphoma (n = 40, 34%), multiple myeloma (n = 19, 16%), and chronic lymphocytic leukaemia (n = 6, 5%). The median CE1 of lymphocytes was 47% (interquartile range: 32%-65%). In multiple regression analysis, Amicus device was associated with higher CE1 of lymphocytes (p = 0.01) and lower CE1 of platelets (p < 0.01) when compared with Optia device. CONCLUSION The knowledge of the MNC and lymphocyte CE1 of each apheresis device used to collect cells for CAR T therapy, together with the goal of the number of cells required, is essential to define the volume to be processed and to ensure the success of the collection.
Collapse
Affiliation(s)
- Juan A Piñeyroa
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Joan Cid
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain.,IDIBAPS, Barcelona, Spain.,University of Barcelona, Barcelona, Spain
| | - Alexandru Vlagea
- Department of Immunology, CDB, Hospital Clínic, Barcelona, Spain
| | - Gloria Carbassé
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Paola Henao
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Noemí Bailo
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain
| | | | | | - Marta Español
- Department of Immunology, CDB, Hospital Clínic, Barcelona, Spain
| | - Julio Delgado
- IDIBAPS, Barcelona, Spain.,University of Barcelona, Barcelona, Spain.,Department of Hematology, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Álvaro Urbano-Ispizua
- IDIBAPS, Barcelona, Spain.,University of Barcelona, Barcelona, Spain.,Department of Hematology, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Miquel Lozano
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain.,IDIBAPS, Barcelona, Spain.,University of Barcelona, Barcelona, Spain
| |
Collapse
|
10
|
Molina JC, Li Y, Otto WR, Miller TP, Getz KD, Mccoubrey C, Ramos M, Krause E, Cao L, Gramatges MM, Rabin K, Scheurer M, Elgarten CW, Myers RM, Seif AE, Fisher BT, Shah NN, Aplenc R. Absolute lymphocyte count recovery following initial acute myelogenous leukemia therapy: Implications for adoptive cell therapy. Pediatr Blood Cancer 2023; 70:e30062. [PMID: 36370087 PMCID: PMC10823592 DOI: 10.1002/pbc.30062] [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: 06/28/2022] [Revised: 09/03/2022] [Accepted: 09/15/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND An adequate absolute lymphocyte count (ALC) is an essential first step in autologous chimeric antigen receptor (CAR) T-cell manufacturing. For patients with acute myelogenous leukemia (AML), the intensity of chemotherapy received may affect adequate ALC recovery required for CAR T-cell production. We sought to analyze ALC following each course of upfront therapy as one metric for CAR T-cell manufacturing feasibility in children and young adults with AML. PROCEDURE ALC data were collected from an observational study of patients with newly diagnosed AML between the ages of 1 month and 21 years who received treatment between the years of 2006 and 2018 at one of three hospitals in the Leukemia Electronic Abstraction of Records Network (LEARN) consortium. RESULTS Among 193 patients with sufficient ALC data for analysis, the median ALC following induction 1 was 1715 cells/μl (interquartile range: 1166-2388), with successive decreases in ALC with each subsequent course. Similarly, the proportion of patients achieving an ALC >400 cells/μl decreased following each course, ranging from 98.4% (190/193) after course 1 to 66.7% (22/33) for patients who received a fifth course of therapy. CONCLUSIONS There is a successive decline of ALC recovery with subsequent courses of chemotherapy. Despite this decline, ALC values are likely sufficient to consider apheresis prior to the initiation of each course of upfront therapy for the majority of newly diagnosed pediatric AML patients, thereby providing a window of opportunity for T-cell collection for those patients identified at high risk of relapse or with refractory disease.
Collapse
Affiliation(s)
- John C. Molina
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Yimei Li
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William R. Otto
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Tamara P. Miller
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Kelly D. Getz
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Carly Mccoubrey
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark Ramos
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Edward Krause
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lusha Cao
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - M. Monica Gramatges
- Pediatric Hematology/Oncology, Baylor College of Medicine/Texas Children’s Hospital, Houston, Texas, USA
- Pediatric Cardio-Oncology Program, Baylor College of Medicine/Texas Children’s Hospital, Houston,Texas, USA
| | - Karen Rabin
- Pediatric Hematology/Oncology, Baylor College of Medicine/Texas Children’s Hospital, Houston, Texas, USA
| | - Michael Scheurer
- Pediatric Hematology/Oncology, Baylor College of Medicine/Texas Children’s Hospital, Houston, Texas, USA
| | - Caitlin W. Elgarten
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Regina M. Myers
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alix E. Seif
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brian T. Fisher
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Richard Aplenc
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Qayed M, McGuirk JP, Myers GD, Parameswaran V, Waller EK, Holman P, Rodrigues M, Clough LF, Willert J. Leukapheresis guidance and best practices for optimal chimeric antigen receptor T-cell manufacturing. Cytotherapy 2022; 24:869-878. [PMID: 35718701 DOI: 10.1016/j.jcyt.2022.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/22/2022] [Accepted: 05/11/2022] [Indexed: 11/03/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an individualized immunotherapy that genetically reprograms a patient's T cells to target and eliminate cancer cells. Tisagenlecleucel is a US Food and Drug Administration-approved CD19-directed CAR T-cell therapy for patients with relapsed/refractory (r/r) B-cell acute lymphoblastic leukemia and r/r diffuse large B-cell lymphoma. Manufacturing CAR T cells is an intricate process that begins with leukapheresis to obtain T cells from the patient's peripheral blood. An optimal leukapheresis product is essential to the success of CAR T-cell therapy; therefore, understanding factors that may affect the quality or T-cell content is imperative. CAR T-cell therapy requires detailed organization throughout the entire multistep process, including appropriate training of a multidisciplinary team in leukapheresis collection, cell processing, timing and coordination with manufacturing and administration to achieve suitable patient care. Consideration of logistical parameters, including leukapheresis timing, location and patient availability, when clinically evaluating the patient and the trajectory of their disease progression must be reflected in the overall collection strategy. Challenges of obtaining optimal leukapheresis product for CAR T-cell manufacturing include vascular access for smaller patients, achieving sufficient T-cell yield, eliminating contaminating cell types in the leukapheresis product, determining appropriate washout periods for medication and managing adverse events at collection. In this review, the authors provide recommendations on navigating CAR T-cell therapy and leukapheresis based on experience and data from tisagenlecleucel manufacturing in clinical trials and the real-world setting.
Collapse
Affiliation(s)
- Muna Qayed
- Blood and Marrow Transplant Program, Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, Georgia, USA.
| | - Joseph P McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - G Doug Myers
- Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Vinod Parameswaran
- Avera Medical Group Hematology, Transplant & Cellular Therapy, Sioux Falls, South Dakota, USA
| | - Edmund K Waller
- Bone Marrow and Stem Cell Transplant Center, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Peter Holman
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - Lee F Clough
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Jennifer Willert
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| |
Collapse
|
12
|
Jo T, Yoshihara S, Hada A, Arai Y, Kitawaki T, Ikemoto J, Onomoto H, Sugiyama H, Yoshihara K, Obi N, Matsui K, Niwa N, Nakagawa Y, Kanda J, Kondo T, Saida S, Kato I, Hiramatsu H, Adachi S, Takita J, Takaori-Kondo A, Nagao M. A clinically applicable prediction model to improve T-cell collection in CAR-T cell therapy. Transplant Cell Ther 2022; 28:365.e1-365.e7. [PMID: 35460928 DOI: 10.1016/j.jtct.2022.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 01/09/2023]
Abstract
As chimeric antigen receptor T (CAR-T) cell therapy targeting CD19 has shown favorable outcomes in patients with relapsed or refractory (r/r) mature B-cell lymphomas and B-cell acute lymphoblastic leukemia (B-ALL), the number of patients who are waiting to receive these treatments is increasing. Optimized protocols for T-cell collection by lymphapheresis for chimeric antigen receptor (CAR) -T cell therapy must be urgently established to provide CAR-T cell therapy for patients with refractory and progressive disease, and/or a low number of lymphocytes due to prior chemotherapies. Predicted collection efficiency of CD3+ cells in apheresis can guide protocols for apheresis, but a clinically applicable model to produce reliable estimates has not been established yet. Therefore, we prospectively analyzed lymphapheresis procedures for tisagenlecleucel therapy at two centers. A total of 108 apheresis procedures included 20 procedures for patients with B-cell acute lymphoblastic leukemia, and 88 for patients with diffuse large B-cell lymphoma, with a median age at apheresis of 58 years (1-71). After lymphapheresis with a median processing blood volume of 10 L (3-16), a median of 3.2 × 109 (0.1-15.0) CD3+ cells were harvested. Collection efficiency 2 (CE2) for CD3+ cells was highly variable (median, 59.3%; range 11.0-199.8). Multivariate analyses revealed that lower Hgb levels, higher circulating CD3+ cell counts, and higher platelet counts before apheresis significantly decreased apheresis CE2. Based on multivariate analyses, we developed a novel formula that estimates CE2 from pre-collection parameters with high accuracy (r = 0.56, p < 0.01), which also suggests the necessary processing blood volume. Our strategy in lymphapheresis should be helpful to reduce collection failure, as well as to achieve efficient utilization of medical resources in clinical practice, thereby delivering CAR-T cell therapy to more patients in a timely manner.
Collapse
Affiliation(s)
- Tomoyasu Jo
- Department of Clinical Laboratory Medicine and Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan; Department of Hematology and Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Satoshi Yoshihara
- Department of Transfusion Medicine and Cell Therapy, Hyogo College of Medicine Hospital, Hyogo, Japan; Department of Hematology, Hyogo College of Medicine Hospital, Hyogo, Japan
| | - Asuka Hada
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuyuki Arai
- Department of Clinical Laboratory Medicine and Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan; Department of Hematology and Oncology, Kyoto University Hospital, Kyoto, Japan.
| | - Toshio Kitawaki
- Department of Hematology and Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Junko Ikemoto
- Department of Transfusion Medicine and Cell Therapy, Hyogo College of Medicine Hospital, Hyogo, Japan
| | - Hitomi Onomoto
- Department of Transfusion Medicine and Cell Therapy, Hyogo College of Medicine Hospital, Hyogo, Japan
| | - Hiroki Sugiyama
- Department of Transfusion Medicine and Cell Therapy, Hyogo College of Medicine Hospital, Hyogo, Japan
| | - Kyoko Yoshihara
- Department of Transfusion Medicine and Cell Therapy, Hyogo College of Medicine Hospital, Hyogo, Japan; Department of Hematology, Hyogo College of Medicine Hospital, Hyogo, Japan
| | - Natsuno Obi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiko Matsui
- Department of Clinical Laboratory Medicine and Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan
| | - Norimi Niwa
- Department of Clinical Laboratory Medicine and Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan
| | - Yoko Nakagawa
- Department of Clinical Laboratory Medicine and Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan
| | - Junya Kanda
- Department of Hematology and Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tadakazu Kondo
- Department of Hematology and Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Satoshi Saida
- Department of Pediatrics, Kyoto University Hospital, Kyoto, Japan
| | - Itaru Kato
- Department of Pediatrics, Kyoto University Hospital, Kyoto, Japan
| | | | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junko Takita
- Department of Pediatrics, Kyoto University Hospital, Kyoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Clinical Laboratory Medicine and Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan; Department of Hematology and Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Miki Nagao
- Department of Clinical Laboratory Medicine and Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
13
|
Considerations for immune effector cell therapy collections: a white paper from the American Society for Apheresis. Cytotherapy 2022; 24:916-922. [DOI: 10.1016/j.jcyt.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/18/2022]
|
14
|
Hayden PJ, Roddie C, Bader P, Basak GW, Bonig H, Bonini C, Chabannon C, Ciceri F, Corbacioglu S, Ellard R, Sanchez-Guijo F, Jäger U, Hildebrandt M, Hudecek M, Kersten MJ, Köhl U, Kuball J, Mielke S, Mohty M, Murray J, Nagler A, Rees J, Rioufol C, Saccardi R, Snowden JA, Styczynski J, Subklewe M, Thieblemont C, Topp M, Ispizua ÁU, Chen D, Vrhovac R, Gribben JG, Kröger N, Einsele H, Yakoub-Agha I. Management of adults and children receiving CAR T-cell therapy: 2021 best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association (EHA). Ann Oncol 2022; 33:259-275. [PMID: 34923107 DOI: 10.1016/j.annonc.2021.12.003] [Citation(s) in RCA: 185] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Several commercial and academic autologous chimeric antigen receptor T-cell (CAR-T) products targeting CD19 have been approved in Europe for relapsed/refractory B-cell acute lymphoblastic leukemia, high-grade B-cell lymphoma and mantle cell lymphoma. Products for other diseases such as multiple myeloma and follicular lymphoma are likely to be approved by the European Medicines Agency in the near future. DESIGN The European Society for Blood and Marrow Transplantation (EBMT)-Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association collaborated to draft best practice recommendations based on the current literature to support health care professionals in delivering consistent, high-quality care in this rapidly moving field. RESULTS Thirty-six CAR-T experts (medical, nursing, pharmacy/laboratory) assembled to draft recommendations to cover all aspects of CAR-T patient care and supply chain management, from patient selection to long-term follow-up, post-authorisation safety surveillance and regulatory issues. CONCLUSIONS We provide practical, clinically relevant recommendations on the use of these high-cost, logistically complex therapies for haematologists/oncologists, nurses and other stakeholders including pharmacists and health sector administrators involved in the delivery of CAR-T in the clinic.
Collapse
Affiliation(s)
- P J Hayden
- Department of Haematology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - C Roddie
- UCL Cancer Institute, London, UK; University College London Hospital NHS Foundation Trust, London, UK.
| | - P Bader
- Clinic for Children and Adolescents, University Children's Hospital, Frankfurt, Germany
| | - G W Basak
- Medical University of Warsaw, Department of Hematology, Transplantation and Internal Medicine, Warsaw, Poland
| | - H Bonig
- Institute for Transfusion Medicine and Immunohematology of Goethe University and German Red Cross Blood Service, Frankfurt, Germany
| | - C Bonini
- Ospedale San Raffaele and Università Vita-Salute San Raffaele, Milan, Italy
| | - C Chabannon
- Aix-Marseille université, Inserm CBT-1409, Institut Paoli-Calmettes, centre de thérapie cellulaire, unité de transplantation et de thérapie cellulaire, département de biologie du cancer, Marseille, France
| | - F Ciceri
- Università Vita-Salute San Raffaele, IRCCS Ospedale San Raffaele, Milan, Italy
| | - S Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Franz-Josef-Strauss-Allee 11, University Hospital of Regensburg, Regensburg, Germany
| | - R Ellard
- Royal Marsden Hospital, Fulham Rd, London, UK
| | - F Sanchez-Guijo
- IBSAL-Hospital Universitario de Salamanca, CIC, Universidad de Salamanca, Salamanca, Spain
| | - U Jäger
- Clinical Department for Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - M Hildebrandt
- Department of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital Grosshadern, Munich
| | - M Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - M J Kersten
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam and LYMMCARE, Amsterdam, the Netherlands
| | - U Köhl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI) and Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany; Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - J Kuball
- Department of Hematology and Centre for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S Mielke
- Karolinska Institutet and University Hospital, Department of Laboratory Medicine/Department of Cell Therapy and Allogeneic Stem Cell Transplantation (CAST), Stockholm, Sweden
| | - M Mohty
- Hôpital Saint-Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - J Murray
- Christie Hospital NHS Trust, Manchester, UK
| | - A Nagler
- The Chaim Sheba Medical Center, Tel-Hashomer, affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - J Rees
- University College London Hospital NHS Foundation Trust, London, UK; UCL Institute of Neurology, University College of London Hospitals NHS Foundation Trust, London, UK
| | - C Rioufol
- Hospices Civils de Lyon, UCBL1, EMR 3738 CICLY, Lyon, France
| | - R Saccardi
- Cell Therapy and Transfusion Medicine Department, Careggi University Hospital, Florence, Italy
| | - J A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - J Styczynski
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University Torun, Bydgoszcz, Poland
| | - M Subklewe
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Thieblemont
- AP-HP, Saint-Louis Hospital, Hemato-oncology, University of Paris, Paris, France
| | - M Topp
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Á U Ispizua
- Department of Hematology, ICMHO, Hospital Clínic de Barcelona, Barcelona, Spain
| | - D Chen
- University College London Hospital NHS Foundation Trust, London, UK; Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - R Vrhovac
- Department of Haematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - J G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - N Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg, Germany
| | - H Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - I Yakoub-Agha
- CHU de Lille, Univ Lille, INSERM U1286, Infinite, Lille, France
| |
Collapse
|
15
|
Thibodeaux SR, Aqui NA, Park YA, Schneiderman J, Su LL, Winters JL, Zubair AC, Schwartz J, Liu HD. Lack of defined apheresis collection criteria in publicly available CAR-T cell clinical trial descriptions: Comprehensive review of over 600 studies. J Clin Apher 2022; 37:223-236. [PMID: 35085413 DOI: 10.1002/jca.21964] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 11/20/2021] [Accepted: 12/16/2021] [Indexed: 01/28/2023]
Abstract
BACKGROUND Chimeric antigen receptor T (CAR-T) cell successes have encouraged continued clinical study. Apheresis collection of starting material for CAR-T cell therapy product manufacturing is critical but described approaches suggest variability and clinical guidelines are currently lacking. The goal of this study was to gather and assess variability in apheresis collection descriptions in publicly available CAR T-cell therapy clinical trials. STUDY DESIGN We searched clinicaltrials.gov (a publicly available clinical trial database) for "chimeric antigen receptor T cells" on July 01, 2020 and studies accessed July 30, 2020-August 15, 2020. Data collected included date posted, study characteristics, apheresis mentions (number, location, and context), laboratory parameters and transfusion allowances. Apheresis context was analyzed using a qualitative inductive approach of grounded theory method with open coding. Text was classified into 37 context codes, grouped into 12 categories, and then consolidated into patient, procedure, product, and miscellaneous themes. RESULTS Apheresis was mentioned 1044 times in 322 (51.9%) of 621 total studies. Laboratory parameters mentioned included white blood cells (100 studies), absolute neutrophil count (220 studies), absolute lymphocyte count (102 studies), CD3+ cell (38 studies), hemoglobin (233 studies, 54 studies specified transfusion allowance), and platelet (269 studies, 48 studies specified transfusion allowance). CONCLUSIONS Apheresis collection of CAR-T cell products is not well-defined in clinical study descriptions and the context is inconsistent. Laboratory parameters useful for apheresis collection are variably present and do not consistently align with current practices. Further exploration, and clinical guideline development will encourage alignment of apheresis collections for CAR-T cell products.
Collapse
Affiliation(s)
- Suzanne R Thibodeaux
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicole A Aqui
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yara A Park
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jennifer Schneiderman
- Department of Pediatric Hematology/Oncology/Neuro-oncology/Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leon L Su
- Department of Pathology and Laboratory Medicine, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Jeffrey L Winters
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Abba C Zubair
- Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Joseph Schwartz
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine, New York, New York, USA
| | - Hien D Liu
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | | |
Collapse
|
16
|
Yamanaka I, Yamauchi T, Henzan T, Sakoda T, Miyamoto K, Mishima H, Ono H, Koga Y, Nakashima Y, Kato K, Miyamoto T, Mizuno S, Ogawa Y, Ohga S, Akashi K, Maeda T, Kunisaki Y. Optimization of lymphapheresis for manufacturing autologous CAR-T cells. Int J Hematol 2021; 114:449-458. [PMID: 34275066 DOI: 10.1007/s12185-021-03191-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 01/28/2023]
Abstract
Collection of CD3+ lymphocytes via lymphapheresis is essential for manufacturing autologous chimeric antigen receptor (CAR) T cells. Optimization of timing and procedures for lymphapheresis for each patient is critical because patients often have progressive diseases and receive medications that could reduce T cell counts. We conducted a retrospective study of clinical data from 28 patients who underwent lymphapheresis for CD19-directed CAR-T therapy with tisagenlecleucel to identify factors that could affect CD3+ lymphocyte yields. The numbers of CD3+ cells in peripheral blood were significantly correlated with CD3+ cell yields (correlation coefficient r = 0.84), which enabled us to estimate the volume of blood to process before apheresis. We also found that small cell ratio (SCR) at the apheresis site precisely reflected the proportion of lymphocytes, especially in patients without circulating blasts (coefficient of determination: r2 = 0.9). We were able to predict the CD3+ cell yield and prevent excessive apheresis by measuring pre-apheresis circulating CD3+ cell counts and monitoring SCR. Collectively, these results will help us to establish a strategy for optimization of lymphapheresis procedures for CAR-T cell production on a patient-by-patient basis.
Collapse
Affiliation(s)
- Ikumi Yamanaka
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takuji Yamauchi
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoko Henzan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Teppei Sakoda
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kyoko Miyamoto
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroyuki Mishima
- Department of Medical Technology, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Hiroaki Ono
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yuhki Koga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yasuhiro Nakashima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koji Kato
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Toshihiro Miyamoto
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shinichi Mizuno
- Molecular and Cell Processing Center, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takahiro Maeda
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Division of Precision Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yuya Kunisaki
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
17
|
Gustafson MP, Wheatley-Guy CM, Rosenthal AC, Gastineau DA, Katsanis E, Johnson BD, Simpson RJ. Exercise and the immune system: taking steps to improve responses to cancer immunotherapy. J Immunother Cancer 2021; 9:e001872. [PMID: 34215686 PMCID: PMC8256759 DOI: 10.1136/jitc-2020-001872] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2021] [Indexed: 12/18/2022] Open
Abstract
The remarkable success of cancer immunotherapies has provided new hope to cancer patients. Unfortunately, a significant proportion of patients remain unable to respond to immunotherapy or maintain durable clinical responses. The lack of objective responses likely results from profound immune dysfunction often observed in patients with cancer. There is substantial evidence that exercise and physical activity can reduce incidence and improve outcomes in cancer patients. As the immune system is highly responsive to exercise, one potential avenue to improve immune function is through exercise and physical activity. A single event of dynamic exercise results in the substantial mobilization of leukocytes with increased functional capacities into the circulation. Chronic, or long-term, exercise leads to higher physical fitness in terms of greater cardiorespiratory function and/or muscle strength and endurance. High aerobic capacity, as measured by maximal oxygen uptake, has been associated with the reduction of dysfunctional T cells and improvements in the abundance of some T cell populations. To be sure, however, the mechanisms of exercise-mediated immune changes are both extensive and diverse. Here, we examine the evidence and theorize how acute and chronic exercise could be used to improve responses to cancer immunotherapies including immune checkpoint inhibitors, dendritic cell vaccines, natural killer cell therapies, and adoptive T cell therapies such as chimeric antigen receptor (CAR) T cells. Although the parameters of optimal exercise to yield defined outcomes remain to be determined, the available current data provide a compelling justification for additional human studies and clinical trials investigating the adjuvant use of exercise in immuno-oncology.
Collapse
Affiliation(s)
- Michael P Gustafson
- Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | | | | | - Dennis A Gastineau
- Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Emmanuel Katsanis
- Pediatrics, Immunobiology, University of Arizona Medical Center - University Campus, Tucson, Arizona, USA
| | - Bruce D Johnson
- Department of Cardiovascular Diseases, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Richard J Simpson
- Pediatrics, Immunobiology, and Nutritional Sciences, University of Arizona Medical Center - University Campus, Tucson, Arizona, USA
| |
Collapse
|