1
|
Fiorcari S, Strati P, Dondi E. Editorial: Tumor microenvironment and hematological malignancies: new evidences and new questions. Front Immunol 2024; 15:1407981. [PMID: 38690284 PMCID: PMC11059779 DOI: 10.3389/fimmu.2024.1407981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Affiliation(s)
- Stefania Fiorcari
- Department of Oncology and Hematology, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Paolo Strati
- Department of Lymphoma and Myeloma & Department of Translational Molecular Pathology The University of Texas MD (UT MD) Anderson Cancer Center, Houston, TX, United States
| | - Elisabetta Dondi
- U978 Institut National de la Santé et de la Recherche Médicale/Université Sorbonne Paris Nord, Labex INFLAMEX, Bobigny, France
| |
Collapse
|
2
|
Mc Laughlin AM, Milligan PA, Yee C, Bergstrand M. Model-informed drug development of autologous CAR-T cell therapy: Strategies to optimize CAR-T cell exposure leveraging cell kinetic/dynamic modeling. CPT Pharmacometrics Syst Pharmacol 2023; 12:1577-1590. [PMID: 37448343 PMCID: PMC10681459 DOI: 10.1002/psp4.13011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023] Open
Abstract
Autologous Chimeric antigen receptor (CAR-T) cell therapy has been highly successful in the treatment of aggressive hematological malignancies and is also being evaluated for the treatment of solid tumors as well as other therapeutic areas. A challenge, however, is that up to 60% of patients do not sustain a long-term response. Low CAR-T cell exposure has been suggested as an underlying factor for a poor prognosis. CAR-T cell therapy is a novel therapeutic modality with unique kinetic and dynamic properties. Importantly, "clear" dose-exposure relationships do not seem to exist for any of the currently approved CAR-T cell products. In other words, dose increases have not led to a commensurate increase in the measurable in vivo frequency of transferred CAR-T cells. Therefore, alternative approaches beyond dose titration are needed to optimize CAR-T cell exposure. In this paper, we provide examples of actionable variables - design elements in CAR-T cell discovery, development, and clinical practice, which can be modified to optimize autologous CAR-T cell exposure. Most of these actionable variables can be assessed throughout the various stages of discovery and development as part of a well-informed research and development program. Model-informed drug development approaches can enable such study and program design choices from discovery through to clinical practice and can be an important contributor to cell therapy effectiveness and efficiency.
Collapse
Affiliation(s)
| | | | - Cassian Yee
- Department of Melanoma Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
- Department of ImmunologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | |
Collapse
|
3
|
Perrone S, Lopedote P, De Sanctis V, Iamundo De Cumis I, Pulsoni A, Strati P. Novel Drugs and Radiotherapy in Relapsed Lymphomas: Abscopal Response and Beyond. Cancers (Basel) 2023; 15:2751. [PMID: 37345088 DOI: 10.3390/cancers15102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
Combined modality has represented a mainstay of treatment across many lymphoma histologies, given their sensitivity to both multi-agent chemotherapy and intermediate-dose radiotherapy. More recently, several new agents, including immunotherapies, have reshaped the therapeutic panorama of some lymphomas. In parallel, radiotherapy techniques have witnessed substantial improvement, accompanied by a growing understanding that radiation itself comes with an immune-mediated effect. Six decades after a metastatic lesion regression outside the irradiated field was first described, there is increasing evidence that a combination of radiotherapy and immunotherapy could boost an abscopal effect. This review focuses on the mechanisms underlying this interaction in the setting of lymphomas, and on the results of pivotal prospective studies. Furthermore, the available evidence on the concomitant use of radiotherapy and small molecules (i.e., lenalidomide, venetoclax, and ibrutinib), as well as brentuximab vedotin, and chimeric antigen receptor (CAR) T-cell therapy, is summarized. Currently, combining radiotherapy with new agents in patients who are affected by lymphomas appears feasible, particularly as a bridge to anti-CD19 autologous CAR T-cell infusion. However, more studies are required to assess these combinations, and preliminary data suggest only a synergistic rather than a curative effect.
Collapse
Affiliation(s)
- Salvatore Perrone
- Department of Hematology, S.M. Goretti Hospital, Polo Universitario Pontino, 04100 Latina, Italy
| | - Paolo Lopedote
- Department of Medicine, St Elizabeth's Medical Center, Boston University, Boston, MA 02135, USA
| | - Vitaliana De Sanctis
- Department of Radiation Oncology, Faculty of Medicina e Psicologia, Sant'Andrea Hospital, University of Rome "La Sapienza", 00185 Rome, Italy
| | - Ilenia Iamundo De Cumis
- Department of Radiation Oncology, A. Businco Hospital, ARNAS G. Brotzu, 09121 Cagliari, Italy
| | - Alessandro Pulsoni
- Department of Hematology, S.M. Goretti Hospital, Polo Universitario Pontino, 04100 Latina, Italy
| | - Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
4
|
Caballero AC, Escribà-Garcia L, Pujol-Fernández P, Escudero-López E, Ujaldón-Miró C, Montserrat-Torres R, Sierra J, Alvarez-Fernández C, Briones J. High CAR intensity of expression confers enhanced antitumor effect against lymphoma without functional exhaustion. Cancer Gene Ther 2023; 30:51-61. [PMID: 36031661 DOI: 10.1038/s41417-022-00518-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/22/2022] [Accepted: 08/02/2022] [Indexed: 01/19/2023]
Abstract
Identifying factors that ameliorates clinical outcomes following CART therapy represents an unmet need. We hypothesized that CAR expression level would have a significant impact on CART efficacy and tested this with CAR30+ TSCM-LIKE enriched cells. By sorting T-cells according to CAR mean fluorescence intensity in two markedly different populations (CARHI and CARLO), we showed that a high CAR expression enhances antitumor efficacy in vitro, that is sustained after sequential re-exposures to tumor cells and is not associated with T-cell exhaustion or differentiation. Furthermore, we found a correlation between high surface CAR expression and antitumor effect with CAR19+ T-cells, thus validating our findings with CAR30. Definitive proof of CARHI T-cells improved antitumor efficacy was demonstrated in a human Hodgkin's lymphoma xenograft mouse model, where CAR30-TSCM-LIKE enriched products with high intensity of CAR expression achieved superior tumor control in vivo and longer survival than those with a low intensity of CAR expression. Our data suggest that modulation of CAR intensity of expression represents an additional strategy to increase CART therapy clinical efficacy.
Collapse
Affiliation(s)
- Ana Carolina Caballero
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain.,Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Laura Escribà-Garcia
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Paula Pujol-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Eva Escudero-López
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain.,Autonomous University of Barcelona, Barcelona, Spain
| | - Cristina Ujaldón-Miró
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain.,Autonomous University of Barcelona, Barcelona, Spain
| | - Rosanna Montserrat-Torres
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Jorge Sierra
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain.,Autonomous University of Barcelona, Barcelona, Spain
| | - Carmen Alvarez-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain. .,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain. .,Josep Carreras Leukemia Research Institute, Barcelona, Spain.
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain. .,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain. .,Josep Carreras Leukemia Research Institute, Barcelona, Spain. .,Autonomous University of Barcelona, Barcelona, Spain.
| |
Collapse
|
5
|
Asmamaw Dejenie T, Tiruneh G/Medhin M, Dessie Terefe G, Tadele Admasu F, Wale Tesega W, Chekol Abebe E. Current updates on generations, approvals, and clinical trials of CAR T-cell therapy. Hum Vaccin Immunother 2022; 18:2114254. [PMID: 36094837 DOI: 10.1080/21645515.2022.2114254] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a novel, customized immunotherapy that is considered a 'living' and self-replicating drug to treat cancer, sometimes resulting in a complete cure. CAR T-cells are manufactured through genetic engineering of T-cells by equipping them with CARs to detect and target antigen-expressing cancer cells. CAR is designed to have an ectodomain extracellularly, a transmembrane domain spanning the cell membrane, and an endodomain intracellularly. Since its first discovery, the CAR structure has evolved greatly, from the first generation to the fifth generation, to offer new therapeutic alternatives for cancer patients. This treatment has achieved long-term and curative therapeutic efficacy in multiple blood malignancies that nowadays profoundly change the treatment landscape of lymphoma, leukemia, and multiple myeloma. But CART-cell therapy is associated with several hurdles, such as limited therapeutic efficacy, little effect on solid tumors, adverse effects, expensive cost, and feasibility issues, hindering its broader implications.
Collapse
Affiliation(s)
- Tadesse Asmamaw Dejenie
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Markeshaw Tiruneh G/Medhin
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Gashaw Dessie Terefe
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Fitalew Tadele Admasu
- Department of Biochemistry, College of Medicine and Health Science Arbaminch University, Arbaminch, Ethiopia
| | - Wondwossen Wale Tesega
- Department of Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Endeshaw Chekol Abebe
- Department of Biochemistry, College of Medicine and Health Science Arbaminch University, Arbaminch, Ethiopia
| |
Collapse
|
6
|
Chekol Abebe E, Yibeltal Shiferaw M, Tadele Admasu F, Asmamaw Dejenie T. Ciltacabtagene autoleucel: The second anti-BCMA CAR T-cell therapeutic armamentarium of relapsed or refractory multiple myeloma. Front Immunol 2022; 13:991092. [PMID: 36119032 PMCID: PMC9479060 DOI: 10.3389/fimmu.2022.991092] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Ciltacabtagene autoleucel (also known as cilta-cel) is a chimeric antigen receptor (CAR) T-cell therapy that targets B-cell maturation antigen (BCMA) on the surface of cancer cells in B cell malignancies, such as multiple myeloma (MM). It is a second-generation CAR that is outfitted with an ectodomain comprising two BCMA-binding single chain variable fragment (ScFv) domains, a transmembrane domain, and an endodomain possessing CD3ζ and 4-1BB. Cilta-cel is an autologous, gene-edited CAR T-cell that is prepared by collecting and modifying the recipient’s T-cells to create a patient personalized treatment in the laboratory to be infused back. This CAR T-cell product exceptionally entails CARs with two BCMA-targeting single-domain antibodies that detect two epitopes of BCMA expressed on the malignant cells of MM. Cilta-cel is the current addition to the treatment armamentarium of relapsed or refractory (r/r) MM after its approval by the FDA on February 28, 2022, based on the results of the Phase 1b/2 CARTITUDE-1 study. It was the second approved anti-BCMA CAR T-cell product after idecabtagene vicleucel (ide-cel) to treat myeloma patients. It induces early, deep, and long-lasting responses with a tolerable safety profile in r/r MM. Cilta-cel-treated myeloma patients may potentially experience adverse effects ranging from mild to life-threatening, but they are mostly manageable toxicities. Besides, it has a consistent safety profile upon a longer follow-up of patients. Cilta-cel generally outperforms ide cel in terms of efficacy in MM, but shows comparable adverse events. This review highlights the current updates on cilta-cel efficacy, adverse events, comparison with ide-cel, and its future direction in the treatment of MM.
Collapse
Affiliation(s)
- Endeshaw Chekol Abebe
- Department of Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
- *Correspondence: Endeshaw Chekol Abebe,
| | - Mestet Yibeltal Shiferaw
- Department of Medicine, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Fitalew Tadele Admasu
- Department of Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Tadesse Asmamaw Dejenie
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
7
|
Liu J, Xu N, Wang X, Wang Y, Wu Q, Li X, Pan D, Wang L, Xu Y, Yan J, Li X, Yu L, Yang M. Quantitative radio-thin-layer chromatography and positron emission tomography studies for measuring streptavidin transduced chimeric antigen receptor T cells. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1182:122944. [PMID: 34592686 DOI: 10.1016/j.jchromb.2021.122944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/13/2021] [Accepted: 09/15/2021] [Indexed: 11/20/2022]
Abstract
The proliferation of chimeric antigen receptor (CAR) T cells is closely related to their efficacy, but it is still a great challenge to monitor and quantify CAR T cells in vivo. Based on the high affinity (Kd ≈ 10-15 M) of streptavidin (SA) and biotin, radiolabeled biotin may be used to quantify SA-transduced CAR T cells (SA-CAR T cells). Radio-thin-layer chromatography (radio-TLC) and positron emission tomography (PET) are highly sensitive for trace analysis. Our aim was to develop radio-TLC and PET methods to quantify SA-CAR T cells in vitro and in vivo. First, we developed [68Ga]-DOTA-biotin. Commercially available SA was used as a standard, and quantitative standard curves were established in vitro and in vivo by radio-TLC and PET. Furthermore, the feasibility of the method was verified in Raji model mice. The linear range of radio-TLC was 0.02 ∼ 0.15 pmol/μL with R2 = 0.9993 in vitro. The linear range of PET was 0.02 ∼ 0.76 pmol/μL with R2 = 0.9986 in vivo. SA in CAR T cells can also be accurately quantified in a Raji leukemia model according to PET imaging. The radio-TLC/PET method established in this study is promising for using in the dynamic monitoring and analysis of SA-CAR T cells during therapy.
Collapse
Affiliation(s)
- Jingjing Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Nan Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai 200062, China
| | - Xinyu Wang
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yan Wang
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Qiong Wu
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xinxin Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Donghui Pan
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Lizhen Wang
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yuping Xu
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Junjie Yan
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lei Yu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai 200062, China
| | - Min Yang
- Molecular Imaging Center, NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| |
Collapse
|
8
|
Riederer S, Fux R, Lehmann MH, Volz A, Sutter G, Rojas JJ. Activation of interferon regulatory factor 3 by replication-competent vaccinia viruses improves antitumor efficacy mediated by T cell responses. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:399-409. [PMID: 34553028 PMCID: PMC8430050 DOI: 10.1016/j.omto.2021.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/01/2021] [Indexed: 11/16/2022]
Abstract
Recently, oncolytic vaccinia viruses (VACVs) have shown their potential to provide for clinically effective cancer treatments. The reason for this clinical usefulness is not only the direct destruction of infected cancer cells but also activation of immune responses directed against tumor antigens. For eliciting a robust antitumor immunity, a dominant T helper 1 (Th1) cell differentiation of the response is preferred, and such polarization can be achieved by activating the Toll-like receptor 3 (TLR3)-interferon regulatory factor 3 (IRF3) signaling pathway. However, current VACVs used as oncolytic viruses to date still encode several immune evasion proteins involved in the inhibition of this signaling pathway. By inactivating genes of selected regulatory virus proteins, we aimed for a candidate virus with increased potency to activate cellular antitumor immunity but at the same time with a fully maintained replicative capacity in cancer cells. The removal of up to three key genes (C10L, N2L, and C6L) from VACV did not reduce the strength of viral replication, both in vitro and in vivo, but resulted in the rescue of IRF3 phosphorylation upon infection of cancer cells. In syngeneic mouse tumor models, this activation translated to enhanced cytotoxic T lymphocyte (CTL) responses directed against tumor-associated antigens and neo-epitopes and improved antitumor activity.
Collapse
Affiliation(s)
- Stephanie Riederer
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 80539 Munich, Germany
| | - Robert Fux
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 80539 Munich, Germany
| | - Michael H Lehmann
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 80539 Munich, Germany
| | - Asisa Volz
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 80539 Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, 80539 Munich, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 80539 Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, 80539 Munich, Germany
| | - Juan J Rojas
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 80539 Munich, Germany.,Department of Pathology and Experimental Therapies, IDIBELL, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain
| |
Collapse
|
9
|
Pollari M, Leivonen SK, Leppä S. Testicular Diffuse Large B-Cell Lymphoma-Clinical, Molecular, and Immunological Features. Cancers (Basel) 2021; 13:cancers13164049. [PMID: 34439203 PMCID: PMC8392512 DOI: 10.3390/cancers13164049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Testicular diffuse large B-cell lymphoma (T-DLBCL) is a rare and aggressive lymphoma entity that mainly affects elderly men. It has a high relapse rate with especially the relapses of the central nervous system associating with dismal outcome. T-DLBCL has a unique biology with distinct genetic characteristics and clinical presentation, and the increasing knowledge on the tumor microenvironment of T-DLBCL highlights the significance of the host immunity and immune escape in this rare lymphoma, presenting in an immune-privileged site of the testis. This review provides an update on the latest progress made in T-DLBCL research and summarizes the clinical perspectives in T-DLBCL. Abstract Primary testicular lymphoma is a rare lymphoma entity, yet it is the most common testicular malignancy among elderly men. The majority of the cases represent non-germinal center B-cell-like (non-GCB) diffuse large B-cell lymphoma (DLBCL) with aggressive clinical behavior and a relatively high relapse rate. Due to the rareness of the disease, no randomized clinical trials have been conducted and the currently recognized standard of care is based on retrospective analyses and few phase II trials. During recent years, the tumor microenvironment (TME) and tumor-related immunity have been the focus of many tumor biology studies, and the emergence of targeted therapies and checkpoint inhibitors has significantly modulated the field of cancer therapies. Testicular DLBCL (T-DLBCL) is presented in an immune-privileged site of the testis, and the roles of NF-κB pathway signaling, 9p24.1 aberrations, and tumor-infiltrating immune cells, especially immune checkpoint expressing lymphocytes and macrophages, seem to be unique compared to other lymphoma entities. Preliminary data on the use of immune checkpoint inhibitors in the treatment of T-DLBCL are promising and more studies are ongoing.
Collapse
Affiliation(s)
- Marjukka Pollari
- Research Program Unit, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.-K.L.); (S.L.)
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, 33521 Tampere, Finland
- Correspondence:
| | - Suvi-Katri Leivonen
- Research Program Unit, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.-K.L.); (S.L.)
- Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, 00029 Helsinki, Finland
| | - Sirpa Leppä
- Research Program Unit, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.-K.L.); (S.L.)
- Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, 00029 Helsinki, Finland
| |
Collapse
|
10
|
Iovino L, Thur LA, Gnjatic S, Chapuis A, Milano F, Hill JA. Shared inflammatory pathways and therapeutic strategies in COVID-19 and cancer immunotherapy. J Immunother Cancer 2021; 9:e002392. [PMID: 33986127 PMCID: PMC8126446 DOI: 10.1136/jitc-2021-002392] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 01/28/2023] Open
Abstract
COVID-19, the syndrome caused by the infection with SARS-CoV-2 coronavirus, is characterized, in its severe form, by interstitial diffuse pneumonitis and acute respiratory distress syndrome (ARDS). ARDS and systemic manifestations of COVID-19 are mainly due to an exaggerated immune response triggered by the viral infection. Cytokine release syndrome (CRS), an inflammatory syndrome characterized by elevated levels of circulating cytokines, and endothelial dysfunction are systemic manifestations of COVID-19. CRS is also an adverse event of immunotherapy (IMTX), the treatment of diseases using drugs, cells, and antibodies to stimulate or suppress the immune system. Graft-versus-host disease complications after an allogeneic stem cell transplant, toxicity after the infusion of chimeric antigen receptor-T cell therapy and monoclonal antibodies can all lead to CRS. It is hypothesized that anti-inflammatory drugs used for treatment of CRS in IMTX may be useful in reducing the mortality in COVID-19, whereas IMTX itself may help in ameliorating effects of SARS-CoV-2 infection. In this paper, we focused on the potential shared mechanisms and differences between COVID-19 and IMTX-related toxicities. We performed a systematic review of the clinical trials testing anti-inflammatory therapies and of the data published from prospective trials. Preliminary evidence suggests there might be a benefit in targeting the cytokines involved in the pathogenesis of COVID-19, especially by inhibiting the interleukin-6 pathway. Many other approaches based on novel drugs and cell therapies are currently under investigation and may lead to a reduction in hospitalization and mortality due to COVID-19.
Collapse
Affiliation(s)
- Lorenzo Iovino
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Laurel A Thur
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Sacha Gnjatic
- Medicine-Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aude Chapuis
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Filippo Milano
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Joshua A Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
11
|
Coscia M, Bruno B, Neelapu S. Editorial: CAR T-Cell Therapies in Hematologic Tumors. Front Oncol 2020; 10:588134. [PMID: 33178614 PMCID: PMC7596269 DOI: 10.3389/fonc.2020.588134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/13/2020] [Indexed: 01/23/2023] Open
Affiliation(s)
- Marta Coscia
- University Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, and Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Benedetto Bruno
- University Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, and Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Sattva Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
12
|
Strati P, Varma A, Adkins S, Nastoupil LJ, Westin JR, Hagemeister FB, Fowler NH, Lee HJ, Fayad LE, Samaniego F, Ahmed S, Chen Y, Horowitz S, Arafat S, Johncy S, Kebriaei P, Mulanovich VE, Heredia EA, Neelapu SS. Hematopoietic recovery and immune reconstitution after axicabtagene ciloleucel in patients with large B-cell lymphoma. Haematologica 2020; 106:2667-2672. [PMID: 32732355 PMCID: PMC8485681 DOI: 10.3324/haematol.2020.254045] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 may be associated with long-term adverse effects such as cytopenia and immune deficiency. In order to characterize these late events, we analyzed 31 patients with relapsed or refractory large B-cell lymphoma treated with axicabtagene ciloleucel at our institution on two clinical trials, ZUMA-1 (clinicaltrials gov. Identifier: NCT02348216) and ZUMA-9 (clinicaltrials gov. Identifier: NCT03153462). Complete blood counts, lymphocyte subsets, and immunoglobulin levels were measured serially until month 24 or progression. Fifteen (48%) patients had grade 3-4 cytopenia, including anemia (five, 16%), neutropenia (nine, 29%), or thrombocytopenia (13, 42%) at day 30. Cytopenia at day 30 was not significantly associated with later diagnosis of myelodysplasia. Among patients with ongoing remission, grade 3-4 cytopenia was observed in one of nine (11%) at 2 years. While peripheral CD8+ T cells recovered early, CD4+ T-cell recovery was delayed with a count of <200/mL in three of nine (33%) patients at 1 year and two of seven (29%) at 2 years. Immunoglobulin G levels normalized in five of nine (56%) patients at 2 years. Thirteen (42%) patients developed grade 3-4 infectious complications, including herpes zoster and Pneumocystis jiroveci pneumonia. These results suggest the need for prolonged monitoring and prophylaxis against opportunistic infections in these patients, to improve the longterm safety of axicabtagene ciloleucel therapy.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ankur Varma
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
- Division of Hematology, Oncology and Cellular Therapy, Rush University, Chicago, IL, USA
| | - Sherry Adkins
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Loretta J. Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jason R. Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Fredrick B Hagemeister
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nathan H Fowler
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hun J. Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luis E. Fayad
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Felipe Samaniego
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sairah Ahmed
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yiming Chen
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sandra Horowitz
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sara Arafat
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Swapna Johncy
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Partow Kebriaei
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Ella Ariza Heredia
- Department of Infectious Disease, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sattva S. Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
- SATTVA S. NEELAPU
| |
Collapse
|
13
|
Simbaqueba CC, Aponte MP, Kim P, Deswal A, Palaskas NL, Iliescu C, Jahangir E, Yang EH, Steiner RE, Lopez-Mattei J. Cardiovascular Complications of Chimeric Antigen Receptor T-Cell Therapy: The Cytokine Release Syndrome and Associated Arrhythmias. ACTA ACUST UNITED AC 2020; 3:113-120. [DOI: 10.36401/jipo-20-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022]
Abstract
ABSTRACT
In recent years, cancer treatment has evolved, and new therapies have been introduced with significant improvement in prognosis. The immunotherapies stand out owing to their efficacy and remission rate. Chimeric antigen receptor (CAR) T-cell therapy is a part of this new era of therapies. Chimeric antigen receptor T-cell therapy is a form of adoptive cellular therapy that uses a genetically encoded CAR in modified human T cells to target specific tumor antigens in a nonconventional, non-major histocompatibility complex (MHC) protein presentation. Chimeric antigen receptor T-cell therapy successfully identifies tumor antigens and through activation of T cells destroys tumoral cells. It has been found to efficiently induce remission in patients who have been previously treated for B-cell malignancies and have persistent disease. As the use of this novel therapy increases, its potential side effects also have become more evident, including major complications like cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Cytokine release syndrome is a major systemic inflammatory process as a result of massive cytokine production by the proliferating and activated CAR T cells in which multiple interleukins and immune cells contribute to the inflammatory response. Cytokine release syndrome has been associated with cardiovascular life-threatening complications including hypotension, shock, tachycardia, arrhythmias, left ventricular dysfunction, heart failure, and cardiovascular death. Arrhythmias, among its major complications, vary from asymptomatic prolonged corrected QT interval (QTc) to supraventricular tachycardia, atrial fibrillation, flutter, and ventricular arrhythmias like Torsade de pointes. This article focuses on the cardiovascular complications and arrhythmias associated with CRS and CAR T-cell therapy.
Collapse
Affiliation(s)
- Cesar Clavijo Simbaqueba
- Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria Patarroyo Aponte
- Department of Cardiology, University of Texas Health System, Memorial Hermann Hospital, Houston, Texas
| | - Peter Kim
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nicolas L. Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cezar Iliescu
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eiman Jahangir
- Department of Cardiology, Vanderbilt University Medical Center, Vanderbilt, Tennessee
| | - Eric H. Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Raphael E. Steiner
- Department of Lymphoma-Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Juan Lopez-Mattei
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
14
|
Sievers S, Watson G, Johncy S, Adkins S. Recognizing and Grading CAR T-Cell Toxicities: An Advanced Practitioner Perspective. Front Oncol 2020; 10:885. [PMID: 32670871 PMCID: PMC7327099 DOI: 10.3389/fonc.2020.00885] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/05/2020] [Indexed: 12/30/2022] Open
Abstract
Over the past decade, chimeric antigen receptor (CAR) T-cell therapy has significantly improved the outlook for many patients with relapsed and/or refractory B-cell malignancies. The use of CAR T-cell therapy and other therapeutic immune effector cells will likely continue to expand with the development of other targets and use in solid tumors. Although these therapies have shown significant promise in the treatment of some malignancies, they can be associated with unique toxicities including cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome which can be fatal if not identified early and treated appropriately. An understanding of how best to manage the toxicities associated with CAR T-cell therapy is continually evolving. Institutions providing CAR T-cell therapy have undergone changes in infrastructure and staffing models in order to safely care for patients receiving this novel therapy. As members of a multi-disciplinary health care team, advanced practice providers play significant roles in caring for this patient population and must be well-versed in the recognition, grading, and appropriate management of CAR T-cell therapy-related toxicities as these providers care for patients in multiple settings across the continuum of care.
Collapse
Affiliation(s)
- Savannah Sievers
- Department of Physician Assistant Programs, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Grace Watson
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Swapna Johncy
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sherry Adkins
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
15
|
Calabretta E, Carlo-Stella C. The Many Facets of CD38 in Lymphoma: From Tumor-Microenvironment Cell Interactions to Acquired Resistance to Immunotherapy. Cells 2020; 9:E802. [PMID: 32225002 PMCID: PMC7226059 DOI: 10.3390/cells9040802] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
The CD38 antigen is expressed in several hematological malignancies, and the anti-CD38 monoclonal antibodies Daratumumab and Isatuximab have an established role in the therapy of multiple myeloma. However, data on the therapeutic utility of CD38 targeting in other lymphoid malignancies are limited. In chronic lymphocytic leukemia, the prognostic significance of CD38 expression is well accepted, and preclinical studies on the use of Daratumumab in monotherapy or combination therapy have demonstrated considerable efficacy. In other lymphoproliferative disorders, preclinical and clinical data have not been as compelling; however, CD38 overexpression likely contributes to resistance to checkpoint inhibitors, prompting numerous clinical trials in Hodgkin and non-Hodgkin lymphoma to investigate whether blocking CD38 enhances the efficacy of checkpoint inhibitors. Furthermore, due to its widespread expression in hematological tumors, CD38 represents an attractive target for cellular therapies such as CAR-T cells. The present review discusses current knowledge of CD38 expression and its implications in various lymphoid malignancies. Furthermore, it addresses current and future therapeutic perspectives, with a particular emphasis on the significance of CD38 interaction with immune cells of the tumor microenvironment. Lastly, results of ongoing studies using anti-CD38 antibodies will be reviewed.
Collapse
Affiliation(s)
- Eleonora Calabretta
- Department of Oncology and Hematology, Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, 20089 Milano, Italy;
| | - Carmelo Carlo-Stella
- Department of Oncology and Hematology, Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, 20089 Milano, Italy;
- Department of Biomedical Sciences, Humanitas University, Rozzano, 20089 Milano, Italy
| |
Collapse
|
16
|
Enhancement of the Follicular Lymphoma International Prognostic Index (FLIPI) with lymphopenia (FLIPI-L): a predictor for overall survival and histologic transformation. Blood Cancer J 2020; 9:104. [PMID: 31894139 PMCID: PMC6938796 DOI: 10.1038/s41408-019-0269-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 11/08/2022] Open
|
17
|
Kambhampati S, Gray L, Fakhri B, Lo M, Vu K, Arora S, Kaplan L, Ai WZ, Andreadis C. Immune-related Adverse Events Associated With Checkpoint Inhibition in the Setting of CAR T Cell Therapy: A Case Series. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 20:e118-e123. [PMID: 31948859 DOI: 10.1016/j.clml.2019.12.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/03/2019] [Accepted: 12/14/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Swetha Kambhampati
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA; University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA.
| | - Lissa Gray
- University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA
| | - Bita Fakhri
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA; University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA
| | - Mimi Lo
- University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA; Department of Pharmacy, University of California San Francisco, San Francisco, CA
| | - Khoan Vu
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA; University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA
| | - Shagun Arora
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA; University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA
| | - Lawry Kaplan
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA; University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA
| | - Weiyun Z Ai
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA; University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA
| | - Charalambos Andreadis
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA; University of California San Francisco Helen Diller Family Comprehensive Center, San Francisco, CA
| |
Collapse
|
18
|
Current status and hurdles for CAR-T cell immune therapy. BLOOD SCIENCE 2019; 1:148-155. [PMID: 35402809 PMCID: PMC8974909 DOI: 10.1097/bs9.0000000000000025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/25/2019] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cells have emerged as novel and promising immune therapies for the treatment of multiple types of cancer in patients with hematological malignancies. There are several key components critical for development and application of CAR-T therapy. First, the design of CAR vectors can considerably affect several aspects of the physiological functions of these T cells. Moreover, despite the wide use of γ-retrovirus and lentivirus in mediating gene transfer into T cells, optimal CAR delivery systems are also being developed and evaluated. In addition, several classes of mouse models have been used to evaluate the efficacies of CAR-T cells; however, each model has its own limitations. Clinically, although surprising complete remission (CR) rates were observed in acute lymphoblastic leukemia (ALL), lymphoma, and multiple myeloma (MM), there is still a lack of specific targets for acute myeloid leukemia (AML). Leukemia relapse remains a major challenge, and its mechanism is presently under investigation. Cytokine release syndrome (CRS) and neurotoxicity are life-threatening adverse effects that need to be carefully treated. Several factors that compromise the activities of anti-solid cancer CAR-T cells have been recognized, and further improvements targeting these factors are the focus of the development of novel CAR-T cells. Overcoming the current hurdles will lead to optimal responses of CAR-T cells, thus paving the way for their wide clinical application.
Collapse
|
19
|
Singh A, Nandwana V, Rink JS, Ryoo SR, Chen TH, Allen SD, Scott EA, Gordon LI, Thaxton CS, Dravid VP. Biomimetic Magnetic Nanostructures: A Theranostic Platform Targeting Lipid Metabolism and Immune Response in Lymphoma. ACS NANO 2019; 13:10301-10311. [PMID: 31487458 DOI: 10.1021/acsnano.9b03727] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
B-cell lymphoma cells depend upon cholesterol to maintain pro-proliferation and pro-survival signaling via the B-cell receptor. Targeted cholesterol depletion of lymphoma cells is an attractive therapeutic strategy. We report here high-density lipoprotein mimicking magnetic nanostructures (HDL-MNSs) that can bind to the high-affinity HDL receptor, scavenger receptor type B1 (SR-B1), and interfere with cholesterol flux mechanisms in SR-B1 receptor positive lymphoma cells, causing cellular cholesterol depletion. In addition, the MNS core can be utilized for its ability to generate heat under an external radio frequency field. The thermal activation of MNS can lead to both innate and adaptive antitumor immune responses by inducing the expression of heat shock proteins that lead to activation of antigen presenting cells and finally lymphocyte trafficking. In the present study, we demonstrate SR-B1 receptor mediated binding and cellular uptake of HDL-MNS and prevention of phagolysosome formation by transmission electron microscopy, fluorescence microscopy, and ICP-MS analysis. The combinational therapeutics of cholesterol depletion and thermal activation significantly improves therapeutic efficacy in SR-B1 expressing lymphoma cells. HDL-MNS reduces the T2 relaxation time under magnetic resonance imaging (MRI) more effectively compared with a commercially available contrast agent, and the specificity of HDL-MNS toward the SR-B1 receptor leads to differential contrast between SR-B1 positive and negative cells suggesting its utility in diagnostic imaging. Overall, we have demonstrated that HDL-MNSs have cell specific targeting efficiency, can modulate cholesterol efflux, can induce thermal activation mediated antitumor immune response, and possess high contrast under MRI, making it a promising theranostic platform in lymphoma.
Collapse
Affiliation(s)
- Abhalaxmi Singh
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
| | - Vikas Nandwana
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
| | - Jonathan S Rink
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine , Northwestern University , Chicago , Illinois 60611 , United States
- Simpson-Querrey Institute for Bionanotechnology , Northwestern University , Chicago , Illinois 60611 , United States
| | - Soo-Ryoon Ryoo
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
| | - Tzu Hung Chen
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
| | - Sean David Allen
- Interdisciplinary Biological Sciences Program , Northwestern University , Evanston , Illinois 60208 , United States
| | - Evan A Scott
- Simpson-Querrey Institute for Bionanotechnology , Northwestern University , Chicago , Illinois 60611 , United States
- Interdisciplinary Biological Sciences Program , Northwestern University , Evanston , Illinois 60208 , United States
- Department of Biomedical Engineering , Northwestern University , Evanston , Illinois 60208 , United States
| | - Leo I Gordon
- Department of Medicine, Division of Hematology/Oncology, Feinberg School of Medicine , Northwestern University , Chicago , Illinois 60611 , United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University , Chicago , Illinois 60611 , United States
| | - C Shad Thaxton
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University , Chicago , Illinois 60611 , United States
- Department of Urology, Feinberg School of Medicine , Northwestern University , Chicago , Illinois 60611 , United States
| | - Vinayak P Dravid
- Department of Materials Science & Engineering , Northwestern University , Evanston , Illinois 60208 , United States
- International Institute of Nanotechnology , Evanston , Illinois 60208 , United States
| |
Collapse
|