1
|
Junkuhn C, Schiele P, Walter AL, Hamm F, Obermayer B, Busch D, Stroux A, Frick M, Penack O, Damm F, Polansky J, Bullinger L, Künkele A, Frentsch M, Na IK. Prior chemotherapy deteriorates T-cell quality for CAR T-cell therapy in B-cell non-Hodgkin's lymphoma. J Immunother Cancer 2025; 13:e010709. [PMID: 40210237 PMCID: PMC11987159 DOI: 10.1136/jitc-2024-010709] [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: 10/01/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy depends on T cells that are genetically modified to recognize and attack cancer cells. Their effectiveness thus hinges on the functionality of a patient's own T cells. Since CAR T-cell therapy is currently only approved for advanced cancers after at least one line of chemotherapy, we evaluated the potential negative effects of prior exposure to chemotherapy on T-cell functionality. METHODS We studied T cells of two B-cell non-Hodgkin's lymphoma patient cohorts, one collected before treatment (pre-therapy) and the other after one or more (median 3) lines of chemotherapy (post-therapy). Leveraging advanced multiparameter flow cytometry, single-cell RNA sequencing (scRNA-seq), whole-genome DNA methylation arrays and in vitro functionality testing of generated CAR T cells, we compared patient samples in their suitability for effective CAR T-cell therapy. RESULTS We discovered significant modifications in T-cell subsets and their transcriptional profiles secondary to chemotherapy exposure. Our analysis revealed a discernible shift towards phenotypically more differentiated T cells and an upregulation of markers indicative of T-cell exhaustion. Additionally, scRNA-seq and DNA methylation analyses revealed gene expression and epigenetic changes associated with diminished functionality in post-therapy T cells. Cytotoxicity assays demonstrated superior killing efficacy of CAR T cells derived from treatment-naïve patients compared with those with chemotherapy history. CONCLUSIONS These findings corroborate that employing T cells collected prior to frontline chemotherapy could enhance the effectiveness of CAR T-cell therapy and improve patient outcomes.
Collapse
Affiliation(s)
- Charlotte Junkuhn
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- BSIO Berlin School of Integrative Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
| | - Phillip Schiele
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
| | - Anna Luzie Walter
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- BSIO Berlin School of Integrative Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
| | - Frederik Hamm
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Benedikt Obermayer
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - David Busch
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
| | - Andrea Stroux
- Institute for Biometrie and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mareike Frick
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
| | - Olaf Penack
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
| | - Frederik Damm
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Berlin, Germany
| | - Julia Polansky
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Rheumatism Research Center (DRFZ), Immuno-Epigenetics, Berlin, Germany
| | - Lars Bullinger
- BSIO Berlin School of Integrative Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Berlin, Germany
| | - Annette Künkele
- BSIO Berlin School of Integrative Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Berlin, Germany
- Pediatric Oncology and Hematology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Frentsch
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
| | - Il-Kang Na
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- BSIO Berlin School of Integrative Oncology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Berlin, Germany
| |
Collapse
|
2
|
Dreyzin A, Shao L, Cai Y, Han KL, Prochazkova M, Gertz M, Yates B, Shi R, Martin K, Taylor N, Highfill S, O'Neill M, Andresson T, Stroncek D, Jin P, Shah NN. Immunophenotype of CAR T cells and apheresis products predicts response in CD22 CAR T cell trial for B cell acute lymphoblastic leukemia. Mol Ther 2025:S1525-0016(25)00193-5. [PMID: 40087865 DOI: 10.1016/j.ymthe.2025.03.019] [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: 11/19/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025] Open
Abstract
Although CAR T cell therapy is increasingly used to treat relapsed B cell acute lymphoblastic leukemia (ALL), 20%-30% of patients do not respond, and few clinical predictors of response have been established, especially in the pediatric population. A deeper analysis of CAR T cell infusion products, along with the apheresis product used as the starting material for CAR T cell manufacturing, provides valuable insights for predicting clinical outcomes. We analyzed infusion products and CD4/8-selected T cell starting materials from pediatric and young adult patients on a single-center study with relapsed/refractory B cell ALL who were undergoing treatment with CD22 CAR T cells and evaluated differences between T cells from responders and non-responders (NCT023215612). We found that CAR T cells from non-responders had a more differentiated T cell phenotype and overexpressed genes associated with cytotoxicity and exhaustion compared with those of responders. Furthermore, we found that these differences could be tracked back to the apheresis materials prior to CAR T cell manufacturing. Using flow cytometry-based immunophenotypic markers, we developed a scoring system that distinguished non-responders based on T cell phenotype at the time of apheresis. These findings can help inform outcomes for patients and providers as well as provide insights into targeted manufacturing changes to optimize CAR T cell efficacy.
Collapse
Affiliation(s)
- Alexandra Dreyzin
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA; Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Lipei Shao
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Yihua Cai
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Kyu Lee Han
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Michaela Prochazkova
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Michael Gertz
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bonnie Yates
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rongye Shi
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Kathryn Martin
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven Highfill
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Maura O'Neill
- Center for Cancer Research Protein Characterization Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Thorkell Andresson
- Center for Cancer Research Protein Characterization Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - David Stroncek
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Ping Jin
- Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Ram R, Amit O, Perry C, Herishanu Y, Avivi I, Sarid N, Apel A, Preis M, Aviv A, Shapira S, Shragai T, Joffe E, Shargian L, Herzog-Tsarfati K, Eylati N, Acria L, Fridberg G, Gold R, Glait-Santar C, Kay S, Gal-Rabinovich K, Rosenberg D, Setter-Marco N, Beyar-Katz O. Addition of Nivolumab Tailored by Expansion of CAR-T Cells in Patients with Stable/Progressive Large B Cell Lymphoma at Lymphodepletion-A Phase 2, Prospective Interventional Study. Transplant Cell Ther 2024; 30:1178-1188. [PMID: 39396632 DOI: 10.1016/j.jtct.2024.09.024] [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: 07/31/2024] [Revised: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
Patients with large B-cell lymphoma (LBCL) in stable or progressive disease (SD/PD) at lymphodepletion prior to chimeric antigen receptor T cell (CAR-T) therapy have an inferior outcome. we hypothesized that enhancing in-vivo expansion of CAR-T cells could overcome this grim prognosis leading to improved outcomes. We conducted a phase 2 prospective trial (NCT05385263) investigating the addition of nivolumab to enhance CAR-T cell expansion and response in patients with SD/PD-LBCL. Eligible patients received 1 dose of nivolumab between day +5 and +9 post CAR-T infusion. An additional dose of nivolumab was administered on day +19 only to patients whose CAR-T cell levels in peripheral blood were below 100 cells/µL at day +7. Twenty patients were enrolled and received anti-CD19 CAR-T (Axicabtagene ciloleucel, n = 12; tisagenlecleucel, n = 8). Eight were ineligible to receive nivolumab due to active CAR-T-associated toxicities. Overall, the protocol was safe. One-month PET-CT showed an 84% overall response rate (complete response, 53%). The cumulative incidence of progression-free survival at 6 and 12 months were 50% (95% CI 36%-64%) and 42% (95% CI 26%-58%), respectively. The cumulative incidence of overall survival at 6 and 12 months were 85% (95% CI 72%-98%) and 51% (95% CI 31%-71%), respectively. Nivolumab administration significantly reduced PD-1 expression on all immune cells. CAR-T cell expansion was similar between nivolumab-eligible and noneligible patients. Notably, there was a significant enrichment of CD45RO-CD27+ CD8+ cells and CD45RO-CD27+ CD8+ CAR-T cells in the nivolumab-eligible group compared to those ineligible, suggesting that specific cell enrichment could potentially contribute to an enhanced response rate. We conclude that the addition of nivolumab based on CAR-T cell expansion in patients with SD/PD-LBCL is safe and yields promising early response rates.
Collapse
Affiliation(s)
- Ron Ram
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Odelia Amit
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chava Perry
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yair Herishanu
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irit Avivi
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nadav Sarid
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Hematology Department, Wolfson Medical Center, Holon, Israel
| | - Arie Apel
- Hematology Department, Shamir Medical Center, Be'er Ya'akov, Israel
| | - Meir Preis
- Hematology Department, Carmel Medical Center, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ariel Aviv
- Hematology Department, Emek Medical Center, Afula, Israel
| | - Shirly Shapira
- Hematology Department, Meir Medical Center, Kfar Saba, Israel
| | - Tamir Shragai
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Erel Joffe
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liat Shargian
- Beilinson Medical Center, Hematology Institute, Petah Tikva, Israel
| | | | - Nili Eylati
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Luisa Acria
- Hematology and Blood bank, Galilee Medical Center, Nahariya, Israel
| | - Gil Fridberg
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Gold
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel
| | - Chen Glait-Santar
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel
| | - Sigi Kay
- Tel Aviv Sourasky Medical Center, Hematology Institution, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Dina Rosenberg
- Hematology laboratory, Rambam Health Care Campus, Haifa, Israel
| | - Noga Setter-Marco
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Ofrat Beyar-Katz
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
4
|
Lotze MT, Olejniczak SH, Skokos D. CD28 co-stimulation: novel insights and applications in cancer immunotherapy. Nat Rev Immunol 2024; 24:878-895. [PMID: 39054343 PMCID: PMC11598642 DOI: 10.1038/s41577-024-01061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Substantial progress in understanding T cell signalling, particularly with respect to T cell co-receptors such as the co-stimulatory receptor CD28, has been made in recent years. This knowledge has been instrumental in the development of innovative immunotherapies for patients with cancer, including immune checkpoint blockade antibodies, adoptive cell therapies, tumour-targeted immunostimulatory antibodies, and immunostimulatory small-molecule drugs that regulate T cell activation. Following the failed clinical trial of a CD28 superagonist antibody in 2006, targeted CD28 agonism has re-emerged as a technologically viable and clinically promising strategy for cancer immunotherapy. In this Review, we explore recent insights into the molecular functions and regulation of CD28. We describe how CD28 is central to the success of current cancer immunotherapies and examine how new questions arising from studies of CD28 as a clinical target have enhanced our understanding of its biological role and may guide the development of future therapeutic strategies in oncology.
Collapse
Affiliation(s)
- Michael T Lotze
- Department of Surgery, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Scott H Olejniczak
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | | |
Collapse
|
5
|
Maneechai K, Khopanlert W, Noiperm P, Udomsak P, Viboonjuntra P, Julamanee J. Generation of ex vivo autologous hematopoietic stem cell-derived T lymphocytes for cancer immunotherapy. Heliyon 2024; 10:e38447. [PMID: 39398019 PMCID: PMC11467635 DOI: 10.1016/j.heliyon.2024.e38447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
CD19CAR-T cell therapy demonstrated promising outcomes in relapsed/refractory B-cell malignancies. Nonetheless, the limited T-cell function and ineffective T-cell apheresis for therapeutic purposes are still concern in heavily pretreated patients. We investigated the feasibility of generating hematopoietic stem cell-derived T lymphocytes (HSC-T) for cancer immunotherapy. The patients' autologous peripheral blood HSCs were enriched for CD34+ and CD3+ cells. The CD34+ cells were then cultured following three steps of lymphoid progenitor differentiation, T-cell differentiation, and T-cell maturation processes. HSC-T cells were successfully generated with robust fold expansion of 3735 times. After lymphoid progenitor differentiation, CD5+ and CD7+ cells remarkably increased (65-84 %) while CD34+ cells consequentially declined. The mature CD3+ cells were detected up to 40 % and 90 % on days 42 and 52, respectively. The majority of HSC-T population was naïve phenotype compared to CD3-T cells (73 % vs 34 %) and CD8:CD4 ratio was 2:1. The higher level of cytokine and cytotoxic granule secretion in HSC-T was observed after activation. HSC-T cells were assessed for clinical application and found that CD19CAR-transduced HSC-T cells demonstrated higher cytokine secretion and a trend of superior cytotoxicity against CD19+ target cells compared to control CAR-T cells. A chronic antigen stimulation assay revealed similar T-cell proliferation, stemness, and exhaustion phenotypes among CAR-T cell types. In conclusions, autologous HSC-T was feasible to generate with preserved T-cell efficacy. The HSC-T cells are potentially utilized as an alternative option for cellular immunotherapy.
Collapse
Affiliation(s)
- Kajornkiat Maneechai
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Wannakorn Khopanlert
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Anatomical Pathology Unit, Division of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Panarat Noiperm
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Phakaporn Udomsak
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| | - Pongtep Viboonjuntra
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Jakrawadee Julamanee
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
- Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Thailand
| |
Collapse
|
6
|
Jallouk AP, Sengsayadeth S, Savani BN, Dholaria B, Oluwole O. Allogeneic and other innovative chimeric antigen receptor platforms. Clin Hematol Int 2024; 6:61-72. [PMID: 39351308 PMCID: PMC11441714 DOI: 10.46989/001c.121404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/14/2023] [Indexed: 10/04/2024] Open
Affiliation(s)
- Andrew P Jallouk
- Medicine, Hematology OncologyVanderbilt University Medical Center
| | | | - Bipin N Savani
- Medicine, Hematology OncologyVanderbilt University Medical Center
| | | | - Olalekan Oluwole
- Medicine, Hematology OncologyVanderbilt University Medical Center
| |
Collapse
|
7
|
Andreu-Saumell I, Rodriguez-Garcia A, Mühlgrabner V, Gimenez-Alejandre M, Marzal B, Castellsagué J, Brasó-Maristany F, Calderon H, Angelats L, Colell S, Nuding M, Soria-Castellano M, Barbao P, Prat A, Urbano-Ispizua A, Huppa JB, Guedan S. CAR affinity modulates the sensitivity of CAR-T cells to PD-1/PD-L1-mediated inhibition. Nat Commun 2024; 15:3552. [PMID: 38670972 PMCID: PMC11053011 DOI: 10.1038/s41467-024-47799-z] [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: 10/06/2023] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy for solid tumors faces significant hurdles, including T-cell inhibition mediated by the PD-1/PD-L1 axis. The effects of disrupting this pathway on T-cells are being actively explored and controversial outcomes have been reported. Here, we hypothesize that CAR-antigen affinity may be a key factor modulating T-cell susceptibility towards the PD-1/PD-L1 axis. We systematically interrogate CAR-T cells targeting HER2 with either low (LA) or high affinity (HA) in various preclinical models. Our results reveal an increased sensitivity of LA CAR-T cells to PD-L1-mediated inhibition when compared to their HA counterparts by using in vitro models of tumor cell lines and supported lipid bilayers modified to display varying PD-L1 densities. CRISPR/Cas9-mediated knockout (KO) of PD-1 enhances LA CAR-T cell cytokine secretion and polyfunctionality in vitro and antitumor effect in vivo and results in the downregulation of gene signatures related to T-cell exhaustion. By contrast, HA CAR-T cell features remain unaffected following PD-1 KO. This behavior holds true for CD28 and ICOS but not 4-1BB co-stimulated CAR-T cells, which are less sensitive to PD-L1 inhibition albeit targeting the antigen with LA. Our findings may inform CAR-T therapies involving disruption of PD-1/PD-L1 pathway tailored in particular for effective treatment of solid tumors.
Collapse
Affiliation(s)
- Irene Andreu-Saumell
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Alba Rodriguez-Garcia
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain.
| | - Vanessa Mühlgrabner
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Marta Gimenez-Alejandre
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Berta Marzal
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Joan Castellsagué
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Fara Brasó-Maristany
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Hugo Calderon
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Laura Angelats
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Salut Colell
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Mara Nuding
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Marta Soria-Castellano
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Paula Barbao
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Aleix Prat
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Alvaro Urbano-Ispizua
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Johannes B Huppa
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Sonia Guedan
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain.
| |
Collapse
|
8
|
Atanackovic D, Iraguha T, Omili D, Avila SV, Fan X, Kocoglu M, Gebru E, Baker JM, Dishanthan N, Dietze KA, Oluwafemi A, Hardy NM, Yared JA, Hankey K, Dahiya S, Rapoport AP, Luetkens T. A novel multicolor fluorescent spot assay for the functional assessment of chimeric antigen receptor (CAR) T-cell products. Cytotherapy 2024; 26:318-324. [PMID: 38340107 DOI: 10.1016/j.jcyt.2024.01.006] [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: 05/03/2023] [Revised: 01/20/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND AIMS Chimeric antigen receptor (CAR) T-cell (CAR-T) therapies have revolutionized the treatment of B-cell lymphomas. Unfortunately, relapses after CD19-targeted CAR-T are relatively common and, therefore, there is a critical need for assays able to assess the function and potency of CAR-T products pre-infusion, which will hopefully help to optimize CAR-T therapies. We developed a novel multicolor fluorescent spot assay (MFSA) for the functional assessment of CAR-T products on a single-cell level, combining the numerical assessment of CAR-T products with their functional characterization. METHODS We first used a standard single-cell interferon (IFN)-γ enzyme-linked immune absorbent spot assay to measure CD19-targeted CAR-T responses to CD19-coated beads. We then developed, optimized and validated an MFSA that simultaneously measures the secretion of combinations of different cytokines on a single CAR-T level. RESULTS We identified IFN-γ/tumor necrosis factor-α/granzyme B as the most relevant cytokine combination, and we used our novel MFSA to functionally and numerically characterize two clinical-grade CAR-T products. CONCLUSIONS In conclusion, we have developed a novel assay for the quantitative and functional potency assessment of CAR-T products. Our optimized MFSA is cost-effective, easy to perform, reliable, can be performed overnight, allowing for a fast delivery of the product to the patient, and requires relatively minimal maintenance and training. The clinical value of our novel assay will be assessed in studies correlating the pre-infusion assessment of CAR-T products with the patients' outcome in a prospective fashion.
Collapse
Affiliation(s)
- Djordje Atanackovic
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA; Department of Microbiology and Immunology, University of Maryland, Baltimore, Maryland, USA.
| | - Thierry Iraguha
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Destiny Omili
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Stephanie V Avila
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Xiaoxuan Fan
- Department of Microbiology and Immunology, University of Maryland, Baltimore, Maryland, USA; University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Mehmet Kocoglu
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Etse Gebru
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Jillian M Baker
- Department of Microbiology and Immunology, University of Maryland, Baltimore, Maryland, USA
| | - Nishanthini Dishanthan
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Kenneth A Dietze
- Department of Microbiology and Immunology, University of Maryland, Baltimore, Maryland, USA
| | - Ayooluwakiitan Oluwafemi
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA; Department of Microbiology and Immunology, University of Maryland, Baltimore, Maryland, USA
| | - Nancy M Hardy
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Jean A Yared
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Kim Hankey
- Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Saurabh Dahiya
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA; Stanford University, Stanford, California, USA
| | - Aaron P Rapoport
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Tim Luetkens
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA; Department of Microbiology and Immunology, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Giraudo MF, Jackson Z, Das I, Abiona OM, Wald DN. Chimeric Antigen Receptor (CAR)-T Cell Therapy for Non-Hodgkin's Lymphoma. Pathog Immun 2024; 9:1-17. [PMID: 38550613 PMCID: PMC10972674 DOI: 10.20411/pai.v9i1.647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/28/2024] [Indexed: 04/15/2024] Open
Abstract
This review focuses on the use of chimeric antigen receptor (CAR)-T cell therapy to treat non-Hodgkin's lymphoma (NHL), a classification of heterogeneous malignant neoplasms of the lymphoid tissue. Despite various conventional and multidrug chemotherapies, the poor prognosis for NHL patients remains and has prompted the utilization of groundbreaking personalized therapies such as CAR-T cells. CAR-T cells are T cells engineered to express a CAR that enables T cells to specifically lyse tumor cells with extracellular expression of a tumor antigen of choice. A CAR is composed of an extracellular antibody fragment or target protein binding domain that is conjugated to activating intracellular signaling motifs common to T cells. In general, CAR-T cell therapies for NHL are designed to recognize cellular markers ubiquitously expressed on B cells such as CD19+, CD20+, and CD22+. Clinical trials using CAR-T cells such as ZUMA-7 and TRANSFORM demonstrated promising results compared to standard of care and ultimately led to FDA approval for the treatment of relapsed/refractory NHL. Despite the success of CAR-T therapy for NHL, challenges include adverse side effects as well as extrinsic and intrinsic mechanisms of tumor resistance that lead to suboptimal outcomes. Overall, CAR-T cell therapies have improved clinical outcomes in NHL patients and generated optimism around their future applications.
Collapse
Affiliation(s)
| | - Zachary Jackson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Indrani Das
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | | | - David N. Wald
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
- Department of Pathology, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio
| |
Collapse
|
10
|
Fischer L, Grieb N, Born P, Weiss R, Seiffert S, Boldt A, Fricke S, Franz P, Heyn S, Kubasch AS, Baber R, Weidner H, Wang SY, Bach E, Hoffmann S, Ussmann J, Kirchberg J, Hell S, Schwind S, Metzeler KH, Herling M, Jentzsch M, Franke GN, Sack U, Reiche K, Köhl U, Platzbecker U, Vucinic V, Merz M. Cellular dynamics following CAR T cell therapy are associated with response and toxicity in relapsed/refractory myeloma. Leukemia 2024; 38:372-382. [PMID: 38184754 PMCID: PMC10844085 DOI: 10.1038/s41375-023-02129-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/17/2023] [Accepted: 12/15/2023] [Indexed: 01/08/2024]
Abstract
B-cell maturation antigen (BCMA)-targeting chimeric antigen receptor (CAR) T cells revolutionized the treatment of relapsed/refractory multiple myeloma (RRMM). However, data on cellular (CAR) T cell dynamics and the association with response, resistance or the occurrence of cytokine release syndrome (CRS) are limited. Therefore, we performed a comprehensive flow cytometry analysis of 27 RRMM patients treated with Idecabtagene vicleucel (Ide-cel) to assess the expansion capacity, persistence and effects on bystander cells of BCMA-targeting CAR T cells. Additionally, we addressed side effects, like cytokine release syndrome (CRS) and cytopenia. Our results show that in vivo expansion of CD8+ CAR T cells is correlated to response, however persistence is not essential for durable remission in RRMM patients. In addition, our data provide evidence, that an increased fraction of CD8+ T cells at day of leukapheresis in combination with successful lymphodepletion positively influence the outcome. We show that patients at risk for higher-grade CRS can be identified already prior to lymphodepletion. Our extensive characterization contributes to a better understanding of the dynamics and effects of BCMA-targeting CAR T cells, in order to predict the response of individual patients as well as side effects, which can be counteracted at an early stage or even prevented.
Collapse
Affiliation(s)
- Luise Fischer
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Nora Grieb
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
- Innovation Center Computer Assisted Surgery (ICCAS), Leipzig, Germany
| | - Patrick Born
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Ronald Weiss
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Sabine Seiffert
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Andreas Boldt
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Paul Franz
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Simone Heyn
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Anne Sophie Kubasch
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Ronny Baber
- Institute for Laboratory Medicine Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- Leipzig Medical Biobank, University Leipzig, Leipzig, Germany
| | - Heike Weidner
- Bone Lab Dresden, University Hospital Dresden, Dresden, Germany
| | - Song Yau Wang
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Enrica Bach
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Sandra Hoffmann
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Jule Ussmann
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Janine Kirchberg
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Saskia Hell
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Sebastian Schwind
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Klaus H Metzeler
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Marco Herling
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Madlen Jentzsch
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Georg-Nikolaus Franke
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Ulrich Sack
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Kristin Reiche
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Ulrike Köhl
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Vladan Vucinic
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Maximilian Merz
- Department of Hematology, Hemostaseology, Cellular Therapy and Infectiology, University Hospital of Leipzig, Leipzig, Germany.
| |
Collapse
|
11
|
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
|
12
|
Benitez Fuentes JD, Mohamed Mohamed K, de Luna Aguilar A, Jiménez García C, Guevara-Hoyer K, Fernandez-Arquero M, Rodríguez de la Peña MA, Garciía Bravo L, Jiménez Ortega AF, Flores Navarro P, Bartolome Arcilla J, Alonso Arenilla B, Baos Muñoz E, Delgado-Iribarren García-Campero A, Montealegre Sanz M, Sanchez-Ramon S, Perez Segura P. Evidence of exhausted lymphocytes after the third anti-SARS-CoV-2 vaccine dose in cancer patients. Front Oncol 2022; 12:975980. [PMID: 36605446 PMCID: PMC9808030 DOI: 10.3389/fonc.2022.975980] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Evidence is scant regarding the long-term humoral and cellular responses Q7 triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccines in cancer patients after repeated booster doses. The possibility of T-cell exhaustion following these booster doses in this population has not yet been fully studied and remains uncertain. Methods In this single-center prospective observational study, we explored the specific humoral and cellular response to S1 antigen in 36 patients with solid malignancies at baseline, and after the second and third doses of the mRNA-1273 vaccine. Results A dual behavior was observed: 24 (66.7%) patients showed partial specific IFN-γ response after the second dose that was further enhanced after the third dose; and 11 (30.5%) already showed an optimal response after the second dose and experienced a marked fall-off of specific IFN-γ production after the third (4 patients negativization), which might suggest T cell exhaustion due to repetitive priming to the same antigen. One (2.8%) patient had persistently negative responses after all three doses. Seroconversion occurred in all patients after the second dose. We then studied circulating exhausted CD8+ T-cells in 4 patients from each of the two response patterns, those with increase and those with decrease in cellular response after the third booster. The patients with decreased cellular response after the booster had a higher expression of PD1+CD8+ and CD57+PD1+CD8+ exhausted T cells compared with those with an increased cellular response both in vivo and in vitro. The proportion of PD1+CD8+ and CD57+PD1+CD8+ exhausted T cells inversely correlated with IFN-γ production. Discussion Our preliminary data show that the two-dose SARS-CoV-2 vaccine regimen was beneficial in all cancer patients of our study. An additional booster seems to be beneficial in suboptimal vaccine seroconverters, in contrast to maximal responders that might develop exhaustion. Our data should be interpreted with caution given the small sample size and highlight the urgent need to validate our results in other independent and larger cohorts. Altogether, our data support the relevance of immunological functional studies to personalize preventive and treatment decisions in cancer patients.
Collapse
Affiliation(s)
- Javier David Benitez Fuentes
- Department of Medical Oncology, Hospital Clinico San Carlos, IdISSC, Calle Profesor Martín Lagos, Madrid, Spain,*Correspondence: Javier David Benitez Fuentes,
| | - Kauzar Mohamed Mohamed
- Department of Immunology, IML and IdISSC, Hospital Cliínico San Carlos, Calle Profesor Martín Lagos, Madrid, Spain
| | - Alicia de Luna Aguilar
- Department of Medical Oncology, Hospital Clinico San Carlos, IdISSC, Calle Profesor Martín Lagos, Madrid, Spain
| | - Carlos Jiménez García
- Department of Immunology, IML and IdISSC, Hospital Cliínico San Carlos, Calle Profesor Martín Lagos, Madrid, Spain
| | - Kissy Guevara-Hoyer
- Department of Immunology, IML and IdISSC, Hospital Cliínico San Carlos, Calle Profesor Martín Lagos, Madrid, Spain,Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | - Miguel Fernandez-Arquero
- Department of Immunology, IML and IdISSC, Hospital Cliínico San Carlos, Calle Profesor Martín Lagos, Madrid, Spain,Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | | | - Laura Garciía Bravo
- Department of Immunology, IML and IdISSC, Hospital Cliínico San Carlos, Calle Profesor Martín Lagos, Madrid, Spain
| | | | - Paloma Flores Navarro
- Department of Medical Oncology, Hospital Clinico San Carlos, IdISSC, Calle Profesor Martín Lagos, Madrid, Spain
| | - Jorge Bartolome Arcilla
- Department of Medical Oncology, Hospital Clinico San Carlos, IdISSC, Calle Profesor Martín Lagos, Madrid, Spain
| | - Bárbara Alonso Arenilla
- Department of Immunology, IML and IdISSC, Hospital Cliínico San Carlos, Calle Profesor Martín Lagos, Madrid, Spain
| | - Elvira Baos Muñoz
- Department of Microbiology, IML and IdISSC, Hospital Cliínico San Carlos, Calle Profesor Martín Lagos, Madrid, Spain
| | | | - María Montealegre Sanz
- Department of Medical Oncology, Hospital Clinico San Carlos, IdISSC, Calle Profesor Martín Lagos, Madrid, Spain
| | - Silvia Sanchez-Ramon
- Department of Immunology, IML and IdISSC, Hospital Cliínico San Carlos, Calle Profesor Martín Lagos, Madrid, Spain,Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | - Pedro Perez Segura
- Department of Medical Oncology, Hospital Clinico San Carlos, IdISSC, Calle Profesor Martín Lagos, Madrid, Spain
| |
Collapse
|
13
|
Paixão EA, Barros LRC, Fassoni AC, Almeida RC. Modeling Patient-Specific CAR-T Cell Dynamics: Multiphasic Kinetics via Phenotypic Differentiation. Cancers (Basel) 2022; 14:cancers14225576. [PMID: 36428671 PMCID: PMC9688514 DOI: 10.3390/cancers14225576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cell immunotherapy revolutionized cancer treatment and consists of the genetic modification of T lymphocytes with a CAR gene, aiming to increase their ability to recognize and kill antigen-specific tumor cells. The dynamics of CAR-T cell responses in patients present multiphasic kinetics with distribution, expansion, contraction, and persistence phases. The characteristics and duration of each phase depend on the tumor type, the infused product, and patient-specific characteristics. We present a mathematical model that describes the multiphasic CAR-T cell dynamics resulting from the interplay between CAR-T and tumor cells, considering patient and product heterogeneities. The CAR-T cell population is divided into functional (distributed and effector), memory, and exhausted CAR-T cell phenotypes. The model is able to describe the diversity of CAR-T cell dynamical behaviors in different patients and hematological cancers as well as their therapy outcomes. Our results indicate that the joint assessment of the area under the concentration-time curve in the first 28 days and the corresponding fraction of non-exhausted CAR-T cells may be considered a potential marker to classify therapy responses. Overall, the analysis of different CAR-T cell phenotypes can be a key aspect for a better understanding of the whole CAR-T cell dynamics.
Collapse
Affiliation(s)
- Emanuelle A. Paixão
- Graduate Program, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil
- Correspondence:
| | - Luciana R. C. Barros
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Artur C. Fassoni
- Institute for Mathematics and Computer Science, Universidade Federal de Itajubá, Itajubá 37500-903, Brazil
| | - Regina C. Almeida
- Computational Modeling Department, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil
| |
Collapse
|