1
|
Mohammadian Gol T, Zahedipour F, Trosien P, Ureña-Bailén G, Kim M, Antony JS, Mezger M. Gene therapy in pediatrics - Clinical studies and approved drugs (as of 2023). Life Sci 2024; 348:122685. [PMID: 38710276 DOI: 10.1016/j.lfs.2024.122685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Gene therapy in pediatrics represents a cutting-edge therapeutic strategy for treating a range of genetic disorders that manifest in childhood. Gene therapy involves the modification or correction of a mutated gene or the introduction of a functional gene into a patient's cells. In general, it is implemented through two main modalities namely ex vivo gene therapy and in vivo gene therapy. Currently, a noteworthy array of gene therapy products has received valid market authorization, with several others in various stages of the approval process. Additionally, a multitude of clinical trials are actively underway, underscoring the dynamic progress within this field. Pediatric genetic disorders in the fields of hematology, oncology, vision and hearing loss, immunodeficiencies, neurological, and metabolic disorders are areas for gene therapy interventions. This review provides a comprehensive overview of the evolution and current progress of gene therapy-based treatments in the clinic for pediatric patients. It navigates the historical milestones of gene therapies, currently approved gene therapy products by the U.S. Food and Drug Administration (FDA) and/or European Medicines Agency (EMA) for children, and the promising future for genetic disorders. By providing a thorough compilation of approved gene therapy drugs and published results of completed or ongoing clinical trials, this review serves as a guide for pediatric clinicians to get a quick overview of the situation of clinical studies and approved gene therapy products as of 2023.
Collapse
Affiliation(s)
- Tahereh Mohammadian Gol
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Fatemeh Zahedipour
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Paul Trosien
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Guillermo Ureña-Bailén
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Miso Kim
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Justin S Antony
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Markus Mezger
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
2
|
Pang Y, Ghosh N. Novel and multiple targets for chimeric antigen receptor-based therapies in lymphoma. Front Oncol 2024; 14:1396395. [PMID: 38711850 PMCID: PMC11070555 DOI: 10.3389/fonc.2024.1396395] [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: 03/05/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 in B-cell non-Hodgkin lymphoma (NHL) validates the utility of CAR-based therapy for lymphomatous malignancies. Despite the success, treatment failure due to CD19 antigen loss, mutation, or down-regulation remains the main obstacle to cure. On-target, off-tumor effect of CD19-CAR T leads to side effects such as prolonged B-cell aplasia, limiting the application of therapy in indolent diseases such as chronic lymphocytic leukemia (CLL). Alternative CAR targets and multi-specific CAR are potential solutions to improving cellular therapy outcomes in B-NHL. For Hodgkin lymphoma and T-cell lymphoma, several cell surface antigens have been studied as CAR targets, some of which already showed promising results in clinical trials. Some antigens are expressed by different lymphomas and could be used for designing tumor-agnostic CAR. Here, we reviewed the antigens that have been studied for novel CAR-based therapies, as well as CARs designed to target two or more antigens in the treatment of lymphoma.
Collapse
Affiliation(s)
- Yifan Pang
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest School of Medicine, Charlotte, NC, United States
| | | |
Collapse
|
3
|
Goyal A, Foss F. Allogeneic transplantation and cellular therapies in cutaneous T-cell lymphoma. Expert Rev Anticancer Ther 2024; 24:41-58. [PMID: 38224371 DOI: 10.1080/14737140.2024.2305356] [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: 07/21/2023] [Accepted: 01/10/2024] [Indexed: 01/16/2024]
Abstract
INTRODUCTION Mycosis fungoides (MF) and Sezary syndrome (SS) are the most common types of cutaneous T-cell lymphoma. Although many available treatments offer temporary disease control, allogeneic hematopoietic stem cell transplant (allo-HSCT) is the only curative treatment option for advanced stage MF and SS. CAR T-cell therapy is a promising new avenue for treatment. AREAS COVERED In this review, we discuss the evidence supporting the use of allo-HSCT for the treatment of MF/SS, including disease status at the time of transplant, conditioning regimen, total body irradiation (TBI), and donor lymphocyte infusion (DLI). We also address the potential role for CAR T-cell therapy in CTCL. EXPERT OPINION Allo-HSCT is an effective treatment for patients with advanced MF and SS. However, significant research is required to determine optimal treatment protocols. Data support the use of reduced-intensity conditioning regimens and suggests that the use of TBI for debulking of skin disease may result in more durable remissions. Donor lymphocyte infusions (DLI) appear to be particularly effective in inducing complete remission in MF/SS patients with relapsed or residual disease. Challenges with CAR-T therapies in T-cell lymphoma include T-cell fratricide due to shared antigens on malignant and nonmalignant T-cells, penetrance into the skin compartment, and CAR-T cell persistence.
Collapse
Affiliation(s)
- Amrita Goyal
- Department of Dermatology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Francine Foss
- Department of Hematology/Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
4
|
Muhsen IN, Hill LC, Ramos CA. Chimeric Antigen Receptor T Cells in Hodgkin and T-Cell Lymphomas. Hematol Oncol Clin North Am 2023; 37:1107-1124. [PMID: 37357070 PMCID: PMC10697615 DOI: 10.1016/j.hoc.2023.05.017] [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] [Indexed: 06/27/2023]
Abstract
The authors review the current use of chimeric antigen receptor (CAR)-transduced T cells (CAR-T) in Hodgkin lymphoma (HL) and T-cell lymphomas (TCL) and discuss the data on CD30-targeting CAR-T cells, which seem to be safe and effective in HL. In addition, the authors examine the use of CAR-T cells targeting CD30, CD5, or CD7 in TCL, while highlighting the unique challenges of their use in this subset of lymphomas. Furthermore, the authors present future directions and ongoing trials investigating the use of CAR-T cells in TCL and HL.
Collapse
Affiliation(s)
- Ibrahim N Muhsen
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - LaQuisa C Hill
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Carlos A Ramos
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
5
|
Karsten H, Matrisch L, Cichutek S, Fiedler W, Alsdorf W, Block A. Broadening the horizon: potential applications of CAR-T cells beyond current indications. Front Immunol 2023; 14:1285406. [PMID: 38090582 PMCID: PMC10711079 DOI: 10.3389/fimmu.2023.1285406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Engineering immune cells to treat hematological malignancies has been a major focus of research since the first resounding successes of CAR-T-cell therapies in B-ALL. Several diseases can now be treated in highly therapy-refractory or relapsed conditions. Currently, a number of CD19- or BCMA-specific CAR-T-cell therapies are approved for acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), multiple myeloma (MM), and follicular lymphoma (FL). The implementation of these therapies has significantly improved patient outcome and survival even in cases with previously very poor prognosis. In this comprehensive review, we present the current state of research, recent innovations, and the applications of CAR-T-cell therapy in a selected group of hematologic malignancies. We focus on B- and T-cell malignancies, including the entities of cutaneous and peripheral T-cell lymphoma (T-ALL, PTCL, CTCL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), chronic lymphocytic leukemia (CLL), classical Hodgkin-Lymphoma (HL), Burkitt-Lymphoma (BL), hairy cell leukemia (HCL), and Waldenström's macroglobulinemia (WM). While these diseases are highly heterogenous, we highlight several similarly used approaches (combination with established therapeutics, target depletion on healthy cells), targets used in multiple diseases (CD30, CD38, TRBC1/2), and unique features that require individualized approaches. Furthermore, we focus on current limitations of CAR-T-cell therapy in individual diseases and entities such as immunocompromising tumor microenvironment (TME), risk of on-target-off-tumor effects, and differences in the occurrence of adverse events. Finally, we present an outlook into novel innovations in CAR-T-cell engineering like the use of artificial intelligence and the future role of CAR-T cells in therapy regimens in everyday clinical practice.
Collapse
Affiliation(s)
- Hendrik Karsten
- Faculty of Medicine, University of Hamburg, Hamburg, Germany
| | - Ludwig Matrisch
- Department of Rheumatology and Clinical Immunology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Faculty of Medicine, University of Lübeck, Lübeck, Germany
| | - Sophia Cichutek
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Andreas Block
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|
6
|
Yang Z, Wang Y. Clinical development of chimeric antigen receptor-T cell therapy for hematological malignancies. Chin Med J (Engl) 2023; 136:2285-2296. [PMID: 37358555 PMCID: PMC10538902 DOI: 10.1097/cm9.0000000000002549] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Indexed: 06/27/2023] Open
Abstract
ABSTRACT Cellular therapies have revolutionized the treatment of hematological malignancies since their conception and rapid development. Chimeric antigen receptor (CAR)-T cell therapy is the most widely applied cellular therapy. Since the Food and Drug Administration approved two CD19-CAR-T products for clinical treatment of relapsed/refractory acute lymphoblastic leukemia and diffuse large B cell lymphoma in 2017, five more CAR-T cell products were subsequently approved for treating multiple myeloma or B cell malignancies. Moreover, clinical trials of CAR-T cell therapy for treating other hematological malignancies are ongoing. Both China and the United States have contributed significantly to the development of clinical trials. However, CAR-T cell therapy has many limitations such as a high relapse rate, adverse side effects, and restricted availability. Various methods are being implemented in clinical trials to address these issues, some of which have demonstrated promising breakthroughs. This review summarizes developments in CAR-T cell trials and advances in CAR-T cell therapy.
Collapse
Affiliation(s)
- Zhihuan Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Tianjin Key Laboratory of Cell Therapy for Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | | |
Collapse
|
7
|
Faber ML, Oldham RAA, Thakur A, Rademacher MJ, Kubicka E, Dlugi TA, Gifford SA, McKillop WM, Schloemer NJ, Lum LG, Medin JA. Novel anti-CD30/CD3 bispecific antibodies activate human T cells and mediate potent anti-tumor activity. Front Immunol 2023; 14:1225610. [PMID: 37646042 PMCID: PMC10461807 DOI: 10.3389/fimmu.2023.1225610] [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: 05/19/2023] [Accepted: 07/12/2023] [Indexed: 09/01/2023] Open
Abstract
CD30 is expressed on Hodgkin lymphomas (HL), many non-Hodgkin lymphomas (NHLs), and non-lymphoid malignancies in children and adults. Tumor expression, combined with restricted expression in healthy tissues, identifies CD30 as a promising immunotherapy target. An anti-CD30 antibody-drug conjugate (ADC) has been approved by the FDA for HL. While anti-CD30 ADCs and chimeric antigen receptors (CARs) have shown promise, their shortcomings and toxicities suggest that alternative treatments are needed. We developed novel anti-CD30 x anti-CD3 bispecific antibodies (biAbs) to coat activated patient T cells (ATCs) ex vivo prior to autologous re-infusions. Our goal is to harness the dual specificity of the biAb, the power of cellular therapy, and the safety of non-genetically modified autologous T cell infusions. We present a comprehensive characterization of the CD30 binding and tumor cell killing properties of these biAbs. Five unique murine monoclonal antibodies (mAbs) were generated against the extracellular domain of human CD30. Resultant anti-CD30 mAbs were purified and screened for binding specificity, affinity, and epitope recognition. Two lead mAb candidates with unique sequences and CD30 binding clusters that differ from the ADC in clinical use were identified. These mAbs were chemically conjugated with OKT3 (an anti-CD3 mAb). ATCs were armed and evaluated in vitro for binding, cytokine production, and cytotoxicity against tumor lines and then in vivo for tumor cell killing. Our lead mAb was subcloned to make a Master Cell Bank (MCB) and screened for binding against a library of human cell surface proteins. Only huCD30 was bound. These studies support a clinical trial in development employing ex vivo-loading of autologous T cells with this novel biAb.
Collapse
Affiliation(s)
- Mary L. Faber
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Robyn A. A. Oldham
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Archana Thakur
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Mary Jo Rademacher
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Ewa Kubicka
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Theresa A. Dlugi
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Steven A. Gifford
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - William M. McKillop
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Nathan J. Schloemer
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Lawrence G. Lum
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Jeffrey A. Medin
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, MCW, Milwaukee, WI, United States
| |
Collapse
|
8
|
Ma XC, Lv X, Li Y. Development of CD30 CAR-T cells in refractory or relapsed Hodgkin's lymphoma. Expert Rev Hematol 2023; 16:1017-1023. [PMID: 37888882 DOI: 10.1080/17474086.2023.2276210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/24/2023] [Indexed: 10/28/2023]
Abstract
INTRODUCTION After therapy, approximately 15% of individuals with Hodgkin's lymphoma (HL) develop relapsed or drug-resistant Hodgkin's lymphoma (r/rHL). r/rHL has a high fatality rate and poor therapeutic prognosis. CD30 CAR-T-cell therapy has emerged as a new way to treat r/rHL in recent years. However, CD30CAR-T cells are still being explored in clinical trials. To help more patients, this review focuses on current CD30CAR-T-cell advancements as well as clinical breakthroughs in treatment of r/rHL. AREAS COVERED This research examines the mechanism of action of CD30 CAR-T cells, their function in the real-world therapy of r/rHL, and the influence of different treatment regimens on treatment results. EXPERT OPINION There has been much research into CD30 CAR-T cells as a result of their successful use in treatment of r/rHL. This research has helped us to understand CD30 CAR-T-cell safety as well as the management options available before and after its administration to increase patient survival and reduce side effects.
Collapse
Affiliation(s)
- Xiao Chen Ma
- Department of Haematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Xiao Lv
- Department of Haematology, Shan dong Provincial Hospital Affiliated to Shan dong First Medical University; Shan dong Provincial Hospital, Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Ying Li
- Department of Haematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| |
Collapse
|
9
|
The emerging role of the branched chain aminotransferases, BCATc and BCATm, for anti-tumor T-cell immunity. IMMUNOMETABOLISM (COBHAM (SURREY, ENGLAND)) 2023; 5:e00014. [PMID: 36644500 PMCID: PMC9833117 DOI: 10.1097/in9.0000000000000014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/31/2022] [Indexed: 01/17/2023]
Abstract
Challenges regarding successful immunotherapy are associated with the heterogeneity of tumors and the complex interactions within the surrounding tumor microenvironment (TME), particularly those between immune and tumor cells. Of interest, T cells receive a myriad of environmental signals to elicit differentiation to effector subtypes, which is accompanied by metabolic reprogramming needed to satisfy the high energy and biosynthetic demands of their activated state. However, T cells are subjected to immunosuppressive signals and areas of oxygen and nutrient depletion in the TME, which causes T-cell exhaustion and helps tumor cells escape immune detection. The cytosolic and mitochondrial branched chain amino transferases, BCATc and BCATm, respectively, are responsible for the first step of the branched chain amino acid (BCAA) degradation, of which, metabolites are shunted into various metabolic processes. In recent years, BCAT isoenzymes have been investigated for their role in a variety of cancers found throughout the body; however, a gap of knowledge exists regarding the role BCAT isoenzymes play within immune cells of the TME. The aim of this review is to summarize recent findings about BCAAs and their catabolism at the BCAT step during T-cell metabolic reprogramming and to discuss the BCAT putative role in the anti-tumor immunity of T cells. Not only does this review acknowledges gaps pertaining to BCAA metabolism in the TME but it also identifies the practical application of BCAA metabolism in T cells in response to cancer and spotlights a potential target for pharmacological intervention.
Collapse
|
10
|
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
|
11
|
Tischer-Zimmermann S, Bonifacius A, Santamorena MM, Mausberg P, Stoll S, Döring M, Kalinke U, Blasczyk R, Maecker-Kolhoff B, Eiz-Vesper B. Reinforcement of cell-mediated immunity driven by tumor-associated Epstein-Barr virus (EBV)-specific T cells during targeted B-cell therapy with rituximab. Front Immunol 2023; 14:878953. [PMID: 37033971 PMCID: PMC10079996 DOI: 10.3389/fimmu.2023.878953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction In immunocompromised patients, Epstein-Barr virus (EBV) infection or reactivation is associated with increased morbidity and mortality, including the development of B-cell lymphomas. The first-line treatment consists of reduction of immunosuppression and administration of rituximab (anti-CD20 antibody). Furthermore, the presence of EBV-specific T cells against latent EBV proteins is crucial for the control of EBV-associated diseases. Therefore, in addition to effective treatment strategies, appropriate monitoring of T cells of high-risk patients is of great importance for improving clinical outcome. In this study, we hypothesized that rituximab-mediated lysis of malignant EBV-infected B cells leads to the release and presentation of EBV-associated antigens and results in an augmentation of EBV-specific effector memory T-cell responses. Methods EBV-infected B lymphoblastoid cell lines (B-LCLs) were used as a model for EBV-associated lymphomas, which are capable of expressing latency stage II and III EBV proteins present in all known EBV-positive malignant cells. Rituximab was administered to obtain cell lysates containing EBV antigens (ACEBV). Efficiency of cross-presentation of EBV-antigen by B-LCLs compared to cross-presentation by professional antigen presenting cells (APCs) such as dendritic cells (DCs) and B cells was investigated by in vitro T-cell immunoassays. Deep T-cell profiling of the tumor-reactive EBV-specific T cells in terms of activation, exhaustion, target cell killing, and cytokine profile was performed, assessing the expression of T-cell differentiation and activation markers as well as regulatory and cytotoxic molecules by interferon-γ (IFN-γ) EliSpot assay, multicolor flow cytometry, and multiplex analyses. Results By inhibiting parts of the cross-presentation pathway, B-LCLs were shown to cross-present obtained exogenous ACEBV-derived antigens mainly through major histocompatibility complex (MHC) class I molecules. This mechanism is comparable to that for DCs and B cells and resulted in a strong EBV-specific CD8+ cytotoxic T-cell response. Stimulation with ACEBV-loaded APCs also led to the activation of CD4+ T helper cells, suggesting that longer peptide fragments are processed via the classical MHC class II pathway. In addition, B-LCLs were also found to be able to take up exogenous antigens from surrounding cells by endocytosis leading to induction of EBV-specific T-cell responses although to a much lesser extent than cross-presentation of ACEBV-derived antigens. Increased expression of activation markers CD25, CD71 and CD137 were detected on EBV-specific T cells stimulated with ACEBV-loaded APCs, which showed high proliferative and cytotoxic capacity as indicated by enhanced EBV-specific frequencies and increased secretion levels of cytotoxic effector molecules (e.g. IFN-γ, granzyme B, perforin, and granulysin). Expression of the regulatory proteins PD-1 and Tim-3 was induced but had no negative impact on effector T-cell functions. Conclusion In this study, we showed for the first time that rituximab-mediated lysis of EBV-infected tumor cells can efficiently boost EBV-specific endogenous effector memory T-cell responses through cross-presentation of EBV-derived antigens. This promotes the restoration of antiviral cellular immunity and presents an efficient mechanism to improve the treatment of CD20+ EBV-associated malignancies. This effect is also conceivable for other therapeutic antibodies or even for therapeutically applied unmodified or genetically modified T cells, which lead to the release of tumor antigens after specific cell lysis.
Collapse
Affiliation(s)
- Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Maria Michela Santamorena
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Philip Mausberg
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sven Stoll
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Marius Döring
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between The Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between The Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Paediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
- *Correspondence: Britta Eiz-Vesper,
| |
Collapse
|
12
|
Tannoury M, Garnier D, Susin SA, Bauvois B. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What's Coming Next? Cancers (Basel) 2022; 14:6026. [PMID: 36551511 PMCID: PMC9775488 DOI: 10.3390/cancers14246026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Resistance to death is one of the hallmarks of human B cell malignancies and often contributes to the lack of a lasting response to today's commonly used treatments. Drug discovery approaches designed to activate the death machinery have generated a large number of inhibitors of anti-apoptotic proteins from the B-cell lymphoma/leukemia 2 family and the B-cell receptor (BCR) signaling pathway. Orally administered small-molecule inhibitors of Bcl-2 protein and BCR partners (e.g., Bruton's tyrosine kinase and phosphatidylinositol-3 kinase) have already been included (as monotherapies or combination therapies) in the standard of care for selected B cell malignancies. Agonistic monoclonal antibodies and their derivatives (antibody-drug conjugates, antibody-radioisotope conjugates, bispecific T cell engagers, and chimeric antigen receptor-modified T cells) targeting tumor-associated antigens (TAAs, such as CD19, CD20, CD22, and CD38) are indicated for treatment (as monotherapies or combination therapies) of patients with B cell tumors. However, given that some patients are either refractory to current therapies or relapse after treatment, novel therapeutic strategies are needed. Here, we review current strategies for managing B cell malignancies, with a focus on the ongoing clinical development of more effective, selective drugs targeting these molecules, as well as other TAAs and signaling proteins. The observed impact of metabolic reprogramming on B cell pathophysiology highlights the promise of targeting metabolic checkpoints in the treatment of these disorders.
Collapse
Affiliation(s)
| | | | | | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| |
Collapse
|
13
|
Filling the Gap: The Immune Therapeutic Armamentarium for Relapsed/Refractory Hodgkin Lymphoma. J Clin Med 2022; 11:jcm11216574. [PMID: 36362802 PMCID: PMC9656939 DOI: 10.3390/jcm11216574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/30/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Despite years of clinical progress which made Hodgkin lymphoma (HL) one of the most curable malignancies with conventional chemotherapy, refractoriness and recurrence may still affect up to 20–30% of patients. The revolution brought by the advent of immunotherapy in all kinds of neoplastic disorders is more than evident in this disease because anti-CD30 antibodies and checkpoint inhibitors have been able to rescue patients previously remaining without therapeutic options. Autologous hematopoietic cell transplantation still represents a significant step in the treatment algorithm for chemosensitive HL; however, the possibility to induce complete responses after allogeneic transplant procedures in patients receiving reduced-intensity conditioning regimens informs on its sensitivity to immunological control. Furthermore, the investigational application of adoptive T cell transfer therapies paves the way for future indications in this setting. Here, we seek to provide a fresh and up-to-date overview of the new immunotherapeutic agents dominating the scene of relapsed/refractory HL. In this optic, we will also review all the potential molecular mechanisms of tumor resistance, theoretically responsible for treatment failures, and we will discuss the place of allogeneic stem cell transplantation in the era of novel therapies.
Collapse
|
14
|
Zhang P, Yang X, Cao Y, Wang J, Zhou M, Chen L, Wei J, Mao Z, Wang D, Xiao Y, Zhu H, Zhang S, Zhang T, Zhang Y, Zhou J, Huang L. Autologous stem cell transplantation in tandem with Anti-CD30 CAR T-cell infusion in relapsed/refractory CD30 + lymphoma. Exp Hematol Oncol 2022; 11:72. [PMID: 36253833 PMCID: PMC9578248 DOI: 10.1186/s40164-022-00323-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/19/2022] [Indexed: 11/19/2022] Open
Abstract
Background Long-term outcome is unfavourable for relapsed/refractory (r/r) lymphoma patients who are resistant to salvage chemotherapy, even after subsequent autologous stem-cell transplantation (ASCT). Although anti-CD30 chimeric antigen receptor (CAR30) T-cell therapy induces high response rates in these patients, the duration of response is relatively limited. Methods This open-label, single-center and single-arm pilot study investigated the safety and efficacy of ASCT in tandem with CAR30 T-cell infusion in r/r CD30+ lymphoma. The primary endpoint was safety and key secondary endpoint was overall response rate, overall survival, progression-free survival, and duration of response. Results Five classical Hodgkin lymphoma (cHL) patients and 1 anaplastic lymphoma kinase (ALK)-negative anaplastic large cell lymphoma (ALCL) patient were enrolled. The median age was 24 years. No patient had prior ASCT. Three patients (50.0%) relapsed for ≥ 2 times and 3 patients (50.0%) had primary refractory diseases. All had a Deauville score of 4 or 5, and 5 patients (83.3%) had a stable or progressive disease (SD/PD) at enrollment. All patients received myeloablative chemotherapy and infused CD34-positive hematopoietic stem cells (HSCs) and CAR30 T cells in tandem, with a median dose of 3.9 × 106/kg and 7.6 × 106/kg, respectively. Five paitents presented with cytokine release syndrome (CRS), all of which were grade 1. No neurotoxicity was observed. All patients had successful HSCs engraftment and reached an objective response, including 5 (4 cHL and 1 ALCL, 83.3%) with a complete response (CR) and 1 with a partial response (PR). With a median follow-up of 20.4 (range, 12.1–34.4) months, all remained alive and maintained their responses. Conclusion Our work demonstrates the combined administration of ASCT and CAR30 T-cell therapy is well-tolerate and highly effective in r/r cHL and ALCL, even in PET-positive or chemorefractory patients who are expected to have inferior outcome after ASCT, although further large-scaled validation in prospective clinical trial is warranted. Trial registration The trial was registered with the Chinese Clinical Trial Registry (ChiCTR, number ChiCTR2100053662). Supplementary Information The online version contains supplementary material available at 10.1186/s40164-022-00323-9.
Collapse
Affiliation(s)
- Peiling Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Xiuxiu Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Jue Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Mi Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Zekai Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Di Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Yi Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Haichuan Zhu
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, China.,Wuhan Bio-Raid Biotechnology CO., LTD, Wuhan, 430078, Hubei, China
| | - Shangkun Zhang
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, China.,Wuhan Bio-Raid Biotechnology CO., LTD, Wuhan, 430078, Hubei, China
| | - Tongcun Zhang
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, China.,Wuhan Bio-Raid Biotechnology CO., LTD, Wuhan, 430078, Hubei, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| |
Collapse
|
15
|
To V, Evtimov VJ, Jenkin G, Pupovac A, Trounson AO, Boyd RL. CAR-T cell development for Cutaneous T cell Lymphoma: current limitations and potential treatment strategies. Front Immunol 2022; 13:968395. [PMID: 36059451 PMCID: PMC9433932 DOI: 10.3389/fimmu.2022.968395] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T therapy has demonstrated remarkable outcomes for B cell malignancies, however, its application for T cell lymphoma, particularly cutaneous T cell lymphoma (CTCL), has been limited. Barriers to effective CAR-T cell therapy in treating CTCL include T cell aplasia in autologous transplants, CAR-T product contamination with leukemic T cells, CAR-T fratricide (when the target antigen is present on normal T cells), and tumor heterogeneity. To address these critical challenges, innovative CAR engineering by targeting multiple antigens to strike a balance between efficacy and safety of the therapy is necessary. In this review, we discuss the current obstacles to CAR-T cell therapy and highlight potential targets in treating CTCL. Looking forward, we propose strategies to develop more powerful dual CARs that are advancing towards the clinic in CTCL therapy.
Collapse
Affiliation(s)
- Van To
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | | | - Graham Jenkin
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | | | - Alan O. Trounson
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Richard L. Boyd
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- *Correspondence: Richard L. Boyd,
| |
Collapse
|
16
|
Polgárová K, Otáhal P, Šálek C, Pytlík R. Chimeric Antigen Receptor Based Cellular Therapy for Treatment Of T-Cell Malignancies. Front Oncol 2022; 12:876758. [PMID: 35600381 PMCID: PMC9121778 DOI: 10.3389/fonc.2022.876758] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
T-cell malignancies can be divided into precursor (T-acute lymphoblastic leukemia/lymphoblastic lymphoma, T-ALL/LBL) and mature T-cell neoplasms, which are comprised of 28 different entities. Most of these malignancies are aggressive with rather poor prognosis. Prognosis of relapsed/refractory (R/R) disease is especially dismal, with an expected survival only several months after progression. Targeted therapies, such as antiCD30 immunotoxin brentuximab vedotin, antiCD38 antibody daratumumab, and anti-CCR4 antibody mogamulizumab are effective only in subsets of patients with T-cell neoplasms. T-cells equipped with chimeric antigen receptor (CAR-Ts) are routinely used for treatment of R/R B-cell malignancies, however, there are specific obstacles for their use in T-cell leukemias and lymphomas which are fratricide killing, risk of transfection of malignant cells, and T-cell aplasia. The solution for these problems relies on target antigen selection, CRISPR/Cas9 or TALEN gene editing, posttranslational regulation of CAR-T surface antigen expression, and safety switches. Structural chromosomal changes and global changes in gene expression were observed with gene-edited products. We identified 49 studies of CAR-based therapies registered on www.clinicaltrials.gov. Most of them target CD30 or CD7 antigen. Results are available only for a minority of these studies. In general, clinical responses are above 50% but reported follow-up is very short. Specific toxicities of CAR-based therapies, namely cytokine release syndrome (CRS), seem to be connected with the antigen of interest and source of cells for manufacturing. CRS is more frequent in antiCD7 CAR-T cells than in antiCD30 cells, but it is mild in most patients. More severe CRS was observed after gene-edited allogeneic CAR-T cells. Immune effector cell associated neurotoxicity (ICANS) was mild and infrequent. Graft-versus-host disease (GvHD) after allogeneic CAR-T cells from previous hematopoietic stem cell donor was also observed. Most frequent toxicities, similarly to antiCD19 CAR-T cells, are cytopenias. CAR-based cellular therapy seems feasible and effective for T-cell malignancies, however, the optimal design of CAR-based products is still unknown and long-term follow-up is needed for evaluation of their true potential.
Collapse
Affiliation(s)
- Kamila Polgárová
- 1st Department of Medicine, First Faculty of Medicine, Charles University, Prague, Czechia
- 1 Department of Medicine, General University Hospital in Prague, Prague, Czechia
| | - Pavel Otáhal
- Department of Immunotherapy, Institute of Haematology and Blood Transfusion, Prague, Czechia
| | - Cyril Šálek
- Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czechia
- Clinical Department, Institute of Haematology and Blood Transfusion, Prague, Czechia
| | - Robert Pytlík
- 1st Department of Medicine, First Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cell Therapy, Institute of Haematology and Blood Transfusion, Prague, Czechia
- *Correspondence: Robert Pytlík,
| |
Collapse
|
17
|
Sang W, Wang X, Geng H, Li T, Li D, Zhang B, Zhou Y, Song X, Sun C, Yan D, Li D, Li Z, Li C, Xu K. Anti-PD-1 Therapy Enhances the Efficacy of CD30-Directed Chimeric Antigen Receptor T Cell Therapy in Patients With Relapsed/Refractory CD30+ Lymphoma. Front Immunol 2022; 13:858021. [PMID: 35432352 PMCID: PMC9010867 DOI: 10.3389/fimmu.2022.858021] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/10/2022] [Indexed: 12/26/2022] Open
Abstract
Anti-CD30 CAR-T is a potent candidate therapy for relapsed/refractory (r/r) CD30+ lymphomas with therapy limitations, and the efficacy needed to be further improved. Herein a multi-center phase II clinical trial (NCT03196830) of anti-CD30 CAR-T treatment combined with PD-1 inhibitor in r/r CD30+ lymphoma was conducted. After a lymphocyte-depleting chemotherapy with fludarabine and cyclophosphamide, 4 patients in cohort 1 and 3 patients in cohort 2 received 106/kg and 107/kg CAR-T cells, respectively, and 5 patients in cohort 3 received 107/kg CAR-T cells combined with anti-PD-1 antibody. The safety and the efficacy of CAR-T cell therapy were analyzed. Cytokine release syndrome (CRS) was observed in 4 of 12 patients, and only 1 patient (patient 9) experienced grade 3 CRS and was treated with glucocorticoid and tocilizumab. No CAR-T-related encephalopathy syndrome was observed. Only two patients in cohorts 2 and 3 experienced obviously high plasma levels of IL-6 and ferritin after CD30 CAR-T cell infusion. The overall response rate (ORR) was 91.7% (11/12), with 6 patients achieving complete remission (CR) (50%). In cohorts 1 and 2, 6 patients got a response (85.7%), with 2 patients achieving CR (28.6%). In cohort 3, 100% ORR and 80% CR were obtained in 5 patients without ≥3 grade CRS. With a median follow-up of 21.5 months (range: 3-50 months), the progression-free survival and the overall survival rates were 45 and 70%, respectively. Of the 11 patients who got a response after CAR-T therapy, 7 patients (63.6%) maintained their response until the end of follow-up. Three patients died last because of disease progression. Taken together, the combination of anti-PD-1 antibody showed an enhancement effect on CD30 CAR-T therapy in r/r CD30+ lymphoma patients with minimal toxicities.
Collapse
Affiliation(s)
- Wei Sang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xiangmin Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Hongzhi Geng
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tianci Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Dashan Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Bingpei Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yi Zhou
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xuguang Song
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Cai Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Dongmei Yan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Depeng Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Caixia Li
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Kailin Xu, ; Caixia Li,
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
- *Correspondence: Kailin Xu, ; Caixia Li,
| |
Collapse
|
18
|
Meier JA, Savoldo B, Grover NS. The Emerging Role of CAR T Cell Therapy in Relapsed/Refractory Hodgkin Lymphoma. J Pers Med 2022; 12:197. [PMID: 35207685 PMCID: PMC8877886 DOI: 10.3390/jpm12020197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Treatment for Hodgkin lymphoma (HL) has evolved considerably from the time it was originally described in the 19th century with many patients now being cured with frontline therapy. Despite these advances, upwards of 10% of patients experience progressive disease after initial therapy with an even higher percentage relapsing. Until recently there had been limited therapeutic options for relapsed and/or refractory HL outside of highly intensive chemotherapy with stem cell rescue. Improved understanding of the pathophysiology of HL, coupled with the emergence of more targeted therapeutics, has reshaped how we view the treatment of relapsed/refractory HL and its prognosis. With this, there has been an increased focus on immunotherapies that can reprogram the immune system to better overcome the immunosuppressive milieu found in HL for improved cancer cell killing. In particular, chimeric antigen receptor (CAR) T cells are emerging as a valuable therapeutic tool in this area. Building on the success of antibody-drug conjugates directed against CD30, CAR T cells engineered to recognize the same antigen are now reaching patients. Though still in its infancy, CAR T therapy for relapsed/refractory HL has shown exceptional promise in early-stage clinical trials with the potential for durable responses even in patients who had progressed through multiple lines of prior therapy. Here we will review currently available data on the use of CAR T cells in HL, strategies to optimize their effectiveness, and how this therapy may fit into the treatment paradigm of HL going forward.
Collapse
Affiliation(s)
- Jeremy A. Meier
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.A.M.); (B.S.)
- Department of Medicine, Division of Hematology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.A.M.); (B.S.)
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie S. Grover
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.A.M.); (B.S.)
- Department of Medicine, Division of Hematology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
19
|
Guo J, He S, Zhu Y, Yu W, Yang D, Zhao X. Humanized CD30-Targeted Chimeric Antigen Receptor T Cells Exhibit Potent Preclinical Activity Against Hodgkin's Lymphoma Cells. Front Cell Dev Biol 2022; 9:775599. [PMID: 35096811 PMCID: PMC8790321 DOI: 10.3389/fcell.2021.775599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/17/2021] [Indexed: 02/05/2023] Open
Abstract
CD30-directed chimeric antigen receptors (CARs) with single chain antibody fragment (scFv)-binding domains from murine HRS3 show strong cytotoxicity to Hodgkin’s Lymphoma cells and have been used in clinical trials. However, murine scFv in CAR might induce specific rejective immune responses in patients, which compromises the therapeutic effects. The use of human or humanized antibody fragments for CAR construction, rather than those derived from mouse antibodies, can reduce the immunogenicity of the CAR. Importantly, this strategy might simultaneously decrease the risk of cytokine-mediated toxicities and improve CAR T cell persistence. Murine HRS3 antibody has been successfully humanized by grafting the complementarity-determining regions (CDRs) from the mouse antibody framework onto human immunoglobulin consensus sequences, followed by an in vitro evolutionary strategy to select functional Fab fragments with the same affinity as murine sources. In this study, humanized scFvs were utilized to construct a CD30-directed CAR (hHRS3-CAR), and its effectiveness was compared with that of HRS3-CAR. The hHRS3-CAR-T cells specifically kill CD30-positive tumor cell lines in vitro and eliminate lymphoma xenografts in immunodeficient mice with comparable efficiency to HRS3-CAR. The hHRS-CAR-T could be used in clinical trials based on the previously reported advantages of humanized CARs, such as the reduction of immune rejection and better persistence of cells.
Collapse
Affiliation(s)
- Jing Guo
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shuai He
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yongjie Zhu
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Yu
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Dong Yang
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xudong Zhao
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Dave H, Terpilowski M, Mai M, Toner K, Grant M, Stanojevic M, Lazarski C, Shibli A, Bien SA, Maglo P, Hoq F, Schore R, Glenn M, Hu B, Hanley PJ, Ambinder R, Bollard CM. Tumor-associated antigen-specific T cells with nivolumab are safe and persist in vivo in relapsed/refractory Hodgkin lymphoma. Blood Adv 2022; 6:473-485. [PMID: 34495306 PMCID: PMC8791594 DOI: 10.1182/bloodadvances.2021005343] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/25/2021] [Indexed: 11/20/2022] Open
Abstract
Hodgkin lymphoma (HL) Reed Sternberg cells express tumor-associated antigens (TAA) that are potential targets for cellular therapies. We recently demonstrated that TAA-specific T cells (TAA-Ts) targeting WT1, PRAME, and Survivin were safe and associated with prolonged time to progression in solid tumors. Hence, we evaluated whether TAA-Ts when given alone or with nivolumab were safe and could elicit antitumor effects in vivo in patients with relapsed/refractory (r/r) HL. Ten patients were infused with TAA-Ts (8 autologous and 2 allogeneic) for active HL (n = 8) or as adjuvant therapy after hematopoietic stem cell transplant (n = 2). Six patients received nivolumab priming before TAA-Ts and continued until disease progression or unacceptable toxicity. All 10 products recognized 1 or more TAAs and were polyfunctional. Patients were monitored for safety for 6 weeks after the TAA-Ts and for response until disease progression. The infusions were safe with no clear dose-limiting toxicities. Patients receiving TAA-Ts as adjuvant therapy remain in continued remission at 3+ years. Of the 8 patients with active disease, 1 patient had a complete response and 7 had stable disease at 3 months, 3 of whom remain with stable disease at 1 year. Antigen spreading and long-term persistence of TAA-Ts in vivo were observed in responding patients. Nivolumab priming impacted TAA-T recognition and persistence. In conclusion, treatment of patients with r/r HL with TAA-Ts alone or in combination with nivolumab was safe and produced promising results. This trial was registered at www.clinicaltrials.gov as #NCT022039303 and #NCT03843294.
Collapse
Affiliation(s)
- Hema Dave
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
- Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, DC
| | - Madeline Terpilowski
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
| | - Mimi Mai
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
| | - Keri Toner
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
- Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, DC
| | - Melanie Grant
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Maja Stanojevic
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
| | - Christopher Lazarski
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
| | - Abeer Shibli
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
| | | | - Philip Maglo
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
| | - Fahmida Hoq
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
- Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, DC
| | - Reuven Schore
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
- Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, DC
| | - Martha Glenn
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute/University of Utah, Salt Lake City, UT; and
| | - Boyu Hu
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute/University of Utah, Salt Lake City, UT; and
| | - Patrick J. Hanley
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
- Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, DC
| | | | - Catherine M. Bollard
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC
- Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
21
|
Pu Q, Qiao J, Liu Y, Cao X, Tan R, Yan D, Wang X, Li J, Yue B. Differential diagnosis and identification of prognostic markers for peripheral T-cell lymphoma subtypes based on flow cytometry immunophenotype profiles. Front Immunol 2022; 13:1008695. [PMID: 36466894 PMCID: PMC9715969 DOI: 10.3389/fimmu.2022.1008695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022] Open
Abstract
We compared the differential expression of 15 markers in PTCL (Peripheral T-cell lymphoma) subtypes and T-CUS (T-cell clones of uncertain significance), and summarized the specific immunophenotype profiles of each subtype and its impact on prognosis. PD-1 and CD10 are diagnostic markers for AITL (angioimmunoblastic T-cell lymphoma). To avoid confusion with T-CUS of benign clones, it is recommended to define AITL as bounded by PD-1+%>38.01 and/or CD10+%>7.46. T cell-derived ENKTL-N (extranodal NKT cell lymphoma) specifically expresses CD56. ALCL (anaplastic large cell lymphoma) characteristically expresses CD30 and HLA-DR. PTCL-NOS (peripheral T-cell lymphoma unspecified) still lacks a relatively specific phenotype and is prone to loss of basic lineage markers CD3, CD5, and CD7. The determination of T-CUS can be verified by the overall assessment of the bone marrow and a certain period of follow-up. The clustering results showed that the expression of 8 specific markers was significantly different among the 5 groups, suggesting that a combination of related markers can be analyzed in the identification of PTCLs subtypes. The study explores the advantages of TRBC1 combined with CD45RA/CD45RO in detecting T cell clonality, which can efficiently and sensitively analyze multiple target T cell populations at the same time. The sensitivity of PB to replace BM to monitor the tumor burden or MRD (minimal residual disease) of PTCLs is as high as 85.71%, which can relieve the huge pressure of clinical sampling and improve patient compliance. CD7, CD38, and Ki-67 are prognostic indicators for AITL. CD3 and CD8 on PTCL-NOS, and CD56 and HLA-DR on ENKTL-N have prognostic role. This study supports and validates the current classification of PTCL subtypes and establishes an immunophenotypic profile that can be used for precise diagnosis. The important clinical value of PTCLs immunophenotype in routine classification diagnosis, clonality confirmation, prognosis prediction, and treatment target selection was emphasized.
Collapse
Affiliation(s)
- Qiyao Pu
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Jie Qiao
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Yuke Liu
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Xueyan Cao
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ran Tan
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Dongyao Yan
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Xiaoqian Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Jiwei Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Baohong Yue
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China.,Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
22
|
Xu Z, Huang X. Cellular immunotherapy for hematological malignancy: recent progress and future perspectives. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0801. [PMID: 34351724 PMCID: PMC8610149 DOI: 10.20892/j.issn.2095-3941.2020.0801] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/08/2021] [Indexed: 11/14/2022] Open
Abstract
Advancements in the field of cellular immunotherapy have accelerated in recent years and have changed the treatment landscape for a variety of hematologic malignancies. Cellular immunotherapy strategies exploit the patient's immune system to kill cancer cells. The successful use of CD19 chimeric antigen receptor (CAR) T-cells in treating B-cell malignancies is the paradigm of this revolution, and numerous ongoing studies are investigating and extending this approach to other malignancies. However, resistance to CAR-T-cell therapy and non-durable efficacy have prevented CAR-T-cells from becoming the ultimate therapy. Because natural killer (NK) cells play an essential role in antitumor immunity, adoptively transferred allogeneic NK and CAR-modified NK cell therapy has been attempted in certain disease subgroups. Allogenic hematopoietic stem cell transplantation (allo-HSCT) is the oldest form of cellular immunotherapy and the only curative option for hematologic malignancies. Historically, the breadth of application of allo-HSCT has been limited by a lack of identical sibling donors (ISDs). However, great strides have recently been made in the success of haploidentical allografts worldwide, which enable everyone to have a donor. Haploidentical donors can achieve comparable outcomes to those of ISDs and even better outcomes in certain circumstances because of a stronger graft vs. tumor effect. Currently, novel strategies such as CAR-T or NK-based immunotherapy can be applied as a complement to allo-HSCT for curative effects, particularly in refractory cases. Here, we introduce the developments in cellular immunotherapy in hematology.
Collapse
Affiliation(s)
- Zhengli Xu
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiaojun Huang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| |
Collapse
|
23
|
Raffa EH, Branson HM, Ngan B, Alexander S, Abla O. Central nervous system relapse in a child with anaplastic large cell lymphoma: potential for new therapeutic strategies. Cancer Rep (Hoboken) 2021; 4:e1377. [PMID: 33822480 PMCID: PMC8551994 DOI: 10.1002/cnr2.1377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/11/2021] [Accepted: 03/01/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Central nervous system (CNS) relapse is rare in childhood anaplastic large cell lymphoma (ALCL) and is associated with a poor prognosis. CASE We describe an 8-year-old boy with ALCL who developed an early CNS relapse without initial CNS disease. Despite aggressive medical management, the patient's neurological status deteriorated rapidly and he died shortly after. CONCLUSION Optimal treatment for children with relapsed ALCL involving the CNS remains unclear. Novel agents, including ALK inhibitors, that have CNS-penetration might be helpful and pediatric studies are warranted.
Collapse
Affiliation(s)
- Enass H Raffa
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Helen M Branson
- Division of Neuroradiology, Department of Diagnostic Radiology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Bo Ngan
- Division of Pathology, Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Sarah Alexander
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Oussama Abla
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Wu Z, Nicoll M, Ingham RJ. AP-1 family transcription factors: a diverse family of proteins that regulate varied cellular activities in classical hodgkin lymphoma and ALK+ ALCL. Exp Hematol Oncol 2021; 10:4. [PMID: 33413671 PMCID: PMC7792353 DOI: 10.1186/s40164-020-00197-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/07/2023] Open
Abstract
Classical Hodgkin lymphoma (cHL) and anaplastic lymphoma kinase-positive, anaplastic large cell lymphoma (ALK+ ALCL) are B and T cell lymphomas respectively, which express the tumour necrosis factor receptor superfamily member, CD30. Another feature shared by cHL and ALK+ ALCL is the aberrant expression of multiple members of the activator protein-1 (AP-1) family of transcription factors which includes proteins of the Jun, Fos, ATF, and Maf subfamilies. In this review, we highlight the varied roles these proteins play in the pathobiology of these lymphomas including promoting proliferation, suppressing apoptosis, and evading the host immune response. In addition, we discuss factors contributing to the elevated expression of these transcription factors in cHL and ALK+ ALCL. Finally, we examine therapeutic strategies for these lymphomas that exploit AP-1 transcriptional targets or the signalling pathways they regulate.
Collapse
Affiliation(s)
- Zuoqiao Wu
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada ,grid.17063.330000 0001 2157 2938Present Address: Department of Medicine, University of Toronto, Toronto, Canada
| | - Mary Nicoll
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada ,grid.14709.3b0000 0004 1936 8649Present Address: Department of Biology, McGill University, Montreal, Canada
| | - Robert J. Ingham
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| |
Collapse
|
25
|
Emerging Therapies in Relapsed and Refractory Hodgkin Lymphoma: What Comes Next After Brentuximab Vedotin and PD-1 Inhibition? Curr Hematol Malig Rep 2021; 16:1-7. [PMID: 33409966 DOI: 10.1007/s11899-020-00603-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE OF REVIEW The standard of care for relapsed/refractory (r/r) Hodgkin lymphoma (HL) patients is autologous stem cell transplantation (ASCT) for patients in a first or second relapse. However, a significant number of patients with r/r HL are either medically ineligible for ASCT or relapse post-ASCT. In recent years, significant advances have been made in the management of r/r HL with the introduction of the anti-CD30 antibody-drug conjugate (ADC) brentuximab vedotin (BV) and the anti-PD1 checkpoint inhibitors (CPI) nivolumab and pembrolizumab. Nonetheless, despite excellent tolerability and high response rates, the large majority of patients will ultimately progress on these agents. Allogeneic hematopoietic cell transplantation (alloHCT) has offered a potentially curative option for these patients, but high rates of morbidity and mortality have limited its application, and disease relapse is also common post-alloHCT. Thus, effective therapy for HL patients who fail BV and CPI therapy remains an unmet need. This review will cover different treatment strategies for HL patients in this setting with a focus on emerging new therapies. RECENT FINDINGS Investigators have explored methods with the potential to restore sensitivity to BV and CPIs in patients who develop resistance. Additionally, promising new therapeutics are emerging, such as CD25-directed ADC therapy and CD30-directed chimeric antigen receptor (CAR) T cells. While no consensus guidelines exist for the management of HL patients refractory to BV and checkpoint blockade, potential novel strategies and therapeutics are currently under investigation in hopes of expanding the treatment landscape for this challenging patient population.
Collapse
|
26
|
Iżykowska K, Rassek K, Korsak D, Przybylski GK. Novel targeted therapies of T cell lymphomas. J Hematol Oncol 2020; 13:176. [PMID: 33384022 PMCID: PMC7775630 DOI: 10.1186/s13045-020-01006-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
T cell lymphomas (TCL) comprise a heterogeneous group of non-Hodgkin lymphomas (NHL) that often present at an advanced stage at the time of diagnosis and that most commonly have an aggressive clinical course. Treatment in the front-line setting is most often cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or CHOP-like regimens, which are effective in B cell lymphomas, but in TCL are associated with a high failure rate and frequent relapses. Furthermore, in contrast to B cell NHL, in which substantial clinical progress has been made with the introduction of monoclonal antibodies, no comparable advances have been seen in TCL. To change this situation and improve the prognosis in TCL, new gene-targeted therapies must be developed. This is now possible due to enormous progress that has been made in the last years in the understanding of the biology and molecular pathogenesis of TCL, which enables the implementation of the research findings in clinical practice. In this review, we present new therapies and current clinical and preclinical trials on targeted treatments for TCL using histone deacetylase inhibitors (HDACi), antibodies, chimeric antigen receptor T cells (CARTs), phosphatidylinositol 3-kinase inhibitors (PI3Ki), anaplastic lymphoma kinase inhibitors (ALKi), and antibiotics, used alone or in combinations. The recent clinical success of ALKi and conjugated anti-CD30 antibody (brentuximab-vedotin) suggests that novel therapies for TCL can significantly improve outcomes when properly targeted.
Collapse
Affiliation(s)
- Katarzyna Iżykowska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznań, Poland
| | - Karolina Rassek
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznań, Poland
| | - Dorota Korsak
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznań, Poland
| | - Grzegorz K Przybylski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznań, Poland.
| |
Collapse
|
27
|
Rogers AM, Brammer JE. Hematopoietic Cell Transplantation and Adoptive Cell Therapy in Peripheral T Cell Lymphoma. Curr Hematol Malig Rep 2020; 15:316-332. [PMID: 32529515 DOI: 10.1007/s11899-020-00590-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Peripheral T cell lymphomas (PTCLs) are a heterogeneous group of diseases and represent approximately 10-15% of all non-Hodgkin lymphomas. Multiagent chemotherapy with a CHOP (cyclophosphamide, adriamycin, vincristine, prednisone)-like regimen is the current standard of care in the frontline setting, but outcomes for PTCL patients generally remain poor. Strategies used to improve survival and reduce the risk of relapse in PTCL patients include autologous hematopoietic cell transplant (autoHCT) and allogeneic HCT (alloHCT). Due to the relative rarity of these diseases, the evidence supporting the use of autoHCT and alloHCT is based on retrospective and single-arm prospective studies. Novel targeted therapies are now being incorporated into the treatment of PTCL, and they may play important roles in improving upon current standards of care. Herein, we summarize the evidence supporting HCT for the treatment of the most common PTCL histologic subtypes and highlight novel treatment strategies aimed at improving outcomes for these patients, including cutting-edge approaches using chimeric antigen receptor T cells (CAR-T). RECENT FINDINGS Given recent improvements in OS and PFS in CD30+ PTCL using the drug-antibody conjugate brentuximab vedotin (BV), new questions arise regarding the impact of BV on consolidative autoHCT, and its role as a maintenance therapy. Multiple histone deacetylase inhibitors (HDACis) have been approved for the treatment of relapsed/refractory PTCL, and these agents are being incorporated into HCT approaches, both in the frontline and maintenance settings. Early data incorporating these agents into novel conditioning regimens have been reported, and emerging evidence from recent trials suggests that CART cell therapies may prove effective in relapsed/refractory PTCL. The recommended treatment strategy in non-ALK+ PTCL remains induction with a CHOP-like regimen followed by consolidative autoHCT in first remission. In the relapsed/refractory setting, salvage chemotherapy followed by HCT (autoHCT or alloHCT depending on histologic subtype and HCT history) offers the only potential for cure or long-term remission. Ample room for improvement remains in the treatment of patients with PTCL, and novel treatment strategies incorporating targeted agents and CAR-T therapy may help to address the unmet needs of this patient population.
Collapse
Affiliation(s)
- Andrew M Rogers
- Department of Internal Medicine, Division of Hematology, James Comprehensive Cancer Center, The Ohio State University, 320 West Tenth Avenue, Columbus, OH, 43210, USA
| | - Jonathan E Brammer
- Department of Internal Medicine, Division of Hematology, James Comprehensive Cancer Center, The Ohio State University, 320 West Tenth Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
28
|
Xie C, Li X, Zeng H, Qian W. Molecular insights into pathogenesis and targeted therapy of peripheral T cell lymphoma. Exp Hematol Oncol 2020; 9:30. [PMID: 33292562 PMCID: PMC7664070 DOI: 10.1186/s40164-020-00188-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) are biologically and clinically heterogeneous diseases almost all of which are associated with poor outcomes. Recent advances in gene expression profiling that helps in diagnosis and prognostication of different subtypes and next-generation sequencing have given new insights into the pathogenesis and molecular pathway of PTCL. Here, we focus on a broader description of mutational insights into the common subtypes of PTCL including PTCL not other specified type, angioimmunoblastic T-cell lymphoma, anaplastic large cell lymphoma, and extra-nodal NK/T cell lymphoma, nasal type, and also present an overview of new targeted therapies currently in various stages of clinical trials.
Collapse
Affiliation(s)
- Caiqin Xie
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Xian Li
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Hui Zeng
- Department of Hematology, First Affiliated Hospital of Jiaxing University, 1882# Zhonghuan South Road, Jiaxing, 314000, People's Republic of China.
| | - Wenbin Qian
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China.
| |
Collapse
|
29
|
Ramos CA, Grover NS, Beaven AW, Lulla PD, Wu MF, Ivanova A, Wang T, Shea TC, Rooney CM, Dittus C, Park SI, Gee AP, Eldridge PW, McKay KL, Mehta B, Cheng CJ, Buchanan FB, Grilley BJ, Morrison K, Brenner MK, Serody JS, Dotti G, Heslop HE, Savoldo B. Anti-CD30 CAR-T Cell Therapy in Relapsed and Refractory Hodgkin Lymphoma. J Clin Oncol 2020; 38:3794-3804. [PMID: 32701411 PMCID: PMC7655020 DOI: 10.1200/jco.20.01342] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Chimeric antigen receptor (CAR) T-cell therapy of B-cell malignancies has proved to be effective. We show how the same approach of CAR T cells specific for CD30 (CD30.CAR-Ts) can be used to treat Hodgkin lymphoma (HL). METHODS We conducted 2 parallel phase I/II studies (ClinicalTrials.gov identifiers: NCT02690545 and NCT02917083) at 2 independent centers involving patients with relapsed or refractory HL and administered CD30.CAR-Ts after lymphodepletion with either bendamustine alone, bendamustine and fludarabine, or cyclophosphamide and fludarabine. The primary end point was safety. RESULTS Forty-one patients received CD30.CAR-Ts. Treated patients had a median of 7 prior lines of therapy (range, 2-23), including brentuximab vedotin, checkpoint inhibitors, and autologous or allogeneic stem cell transplantation. The most common toxicities were grade 3 or higher hematologic adverse events. Cytokine release syndrome was observed in 10 patients, all of which were grade 1. No neurologic toxicity was observed. The overall response rate in the 32 patients with active disease who received fludarabine-based lymphodepletion was 72%, including 19 patients (59%) with complete response. With a median follow-up of 533 days, the 1-year progression-free survival and overall survival for all evaluable patients were 36% (95% CI, 21% to 51%) and 94% (95% CI, 79% to 99%), respectively. CAR-T cell expansion in vivo was cell dose dependent. CONCLUSION Heavily pretreated patients with relapsed or refractory HL who received fludarabine-based lymphodepletion followed by CD30.CAR-Ts had a high rate of durable responses with an excellent safety profile, highlighting the feasibility of extending CAR-T cell therapies beyond canonical B-cell malignancies.
Collapse
Affiliation(s)
- Carlos A. Ramos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Natalie S. Grover
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Anne W. Beaven
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Premal D. Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Meng-Fen Wu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
- Biostatistics Shared Resource, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Anastasia Ivanova
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Tao Wang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
- Biostatistics Shared Resource, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Thomas C. Shea
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Cliona M. Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Department of Pathology and Immunology, and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Christopher Dittus
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Steven I. Park
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Adrian P. Gee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Paul W. Eldridge
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kathryn L. McKay
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Birju Mehta
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
| | - Catherine J. Cheng
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Faith B. Buchanan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Bambi J. Grilley
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
| | - Kaitlin Morrison
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Malcolm K. Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Jonathan S. Serody
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Helen E. Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital; Dan L. Duncan Cancer, Baylor College of Medicine; Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
30
|
Abstract
Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of rare and aggressive non-Hodgkin’s lymphomas. Clinical staging, prognostic scoring, and initial treatment strategies have historically been based on paradigms developed in B-cell lymphomas. Despite primary treatment protocols that are typically anthracycline-based and frequently involve consolidative autologous stem cell transplantation in first remission, many patients develop disease progression. There remains a high unmet medical need for improved treatment strategies in the relapsed or refractory setting. Salvage chemotherapy and stem cell transplantation in those who are suitable has traditionally been the accepted approach, but this remains a minority of the total patient population. As increasing knowledge is gleaned regarding the biological heterogeneity within the various PTCL subtypes, newer targeted agents have been developed, studied, and approved in this small, heterogeneous population of relapsed or refractory disease. Given its success and tolerability in this pretreated population, brentuximab vedotin, an anti-CD30 antibody drug conjugate, was brought earlier in the disease course and is a model for advances in the targeted treatment of PTCL. As others undergo further development in the relapsed setting and successes are brought earlier in the disease course, the outcome for PTCL patients is likely to improve. However, innovative clinical trial designs are crucial for the assessment of targeted agents in this highly heterogeneous population. This review explores the current treatment environment for patients with relapsed and refractory PTCL, including newer strategies such as targeted agents and immunotherapy.
Collapse
Affiliation(s)
- Cheryl Foster
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 6-424 700 University Avenue, Toronto, ON, M5G 1Z5, Canada.,Department of Medicine, University of Toronto, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| | - John Kuruvilla
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 6-424 700 University Avenue, Toronto, ON, M5G 1Z5, Canada.,Department of Medicine, University of Toronto, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| |
Collapse
|
31
|
A Head Start: CAR-T Cell Therapy for Primary Malignant Brain Tumors. Curr Treat Options Oncol 2020; 21:73. [PMID: 32725495 DOI: 10.1007/s11864-020-00772-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OPINION STATEMENT Oncology is the midst of a therapeutic renaissance. The realization of immunotherapy as an efficacious and expanding treatment option has empowered physicians and patients alike. However, despite these remarkable advances, we have only just broached the potential immunotherapy has to offer and have yet to successfully expand these novel modalities to the field of neuro-oncology. In recent years, exciting results in preclinical studies of immune adjuvants, oncolytic viruses, or cell therapy have been met with only fleeting signs of response when taken to early phase trials. Although many have speculated why these innovative approaches result in impaired outcomes, we are left empty-handed in a field plagued by a drought of new therapies. Herein, we will review the recent advances across cellular therapy for primary malignant brain tumors, an approach that lends itself to overcoming the inherent resistance mechanisms which have impeded the success of prior treatment attempts.
Collapse
|
32
|
Fiore D, Cappelli LV, Broccoli A, Zinzani PL, Chan WC, Inghirami G. Peripheral T cell lymphomas: from the bench to the clinic. Nat Rev Cancer 2020; 20:323-342. [PMID: 32249838 DOI: 10.1038/s41568-020-0247-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/18/2020] [Indexed: 02/07/2023]
Abstract
Peripheral T cell lymphomas (PTCLs) are a heterogeneous group of orphan neoplasms. Despite the introduction of anthracycline-based chemotherapy protocols, with or without autologous haematopoietic transplantation and a plethora of new agents, the progression-free survival of patients with PTCLs needs to be improved. The rarity of these neoplasms, the limited knowledge of their driving defects and the lack of experimental models have impaired clinical successes. This scenario is now rapidly changing with the discovery of a spectrum of genomic defects that hijack essential signalling pathways and foster T cell transformation. This knowledge has led to new genomic-based stratifications, which are being used to establish objective diagnostic criteria, more effective risk assessment and target-based interventions. The integration of genomic and functional data has provided the basis for targeted therapies and immunological approaches that underlie individual tumour vulnerabilities. Fortunately, novel therapeutic strategies can now be rapidly tested in preclinical models and effectively translated to the clinic by means of well-designed clinical trials. We believe that by combining new targeted agents with immune regulators and chimeric antigen receptor-expressing natural killer and T cells, the overall survival of patients with PTCLs will dramatically increase.
Collapse
MESH Headings
- Epigenesis, Genetic/genetics
- Epigenesis, Genetic/physiology
- Humans
- Immunotherapy
- Lymphoma, T-Cell, Peripheral/drug therapy
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/immunology
- Lymphoma, T-Cell, Peripheral/metabolism
- Molecular Targeted Therapy
- Mutation
- Signal Transduction/genetics
- Signal Transduction/physiology
- T-Lymphocytes/physiology
- Transcription Factors/genetics
- Transcription Factors/physiology
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Danilo Fiore
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Luca Vincenzo Cappelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Alessandro Broccoli
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Pier Luigi Zinzani
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy.
| | - Wing C Chan
- Department of Pathology, City of Hope Medical Center, Duarte, CA, USA.
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
33
|
Hansen HP, Paes Leme AF, Hallek M. Role of ADAM10 as a CD30 Sheddase in Classical Hodgkin Lymphoma. Front Immunol 2020; 11:398. [PMID: 32296414 PMCID: PMC7136452 DOI: 10.3389/fimmu.2020.00398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/19/2020] [Indexed: 12/31/2022] Open
Abstract
Cancer cells generally recruit and influence non-malignant immune cells to support the tumor growth. Classical Hodgkin lymphoma (cHL) is a good example because the affected lymphoid tissue contains only a few malignant Hodgkin and Reed-Sternberg (H-RS) cells, which are supported by a massive infiltrate of lymphocytes, fibroblasts, and innate immune cells. The transmembrane receptor CD30, which is selectively expressed on the H-RS cells, plays an important role, not only in cell stimulation and intercellular communication but also in tumor diagnosis and targeted tumor therapy. Different protein processing pathways influence its functionality. Depending on the conditions, the receptor is internalized or released. The release of CD30 occurs either as an intact molecule, embedded in the membrane of extracellular vesicles (EVs), or as a cleaved soluble ectodomain (sCD30). CD30 cleavage is predominantly catalyzed by ADAM10. The enzyme is catalytically active in cells as well as in EVs and gradually releases sCD30. Because the circulation contains no CD30+ donor cells, this mechanism explains that the cleaved ectodomain represents the predominant form of CD30 in the plasma of cHL patients. CD30 processing might influence the impact of CD30 antibody-drug conjugates, such as Brentuximab Vedotin (BV). Whereas, ADAM10-degraded CD30 impedes the BV efficacy, tumor-derived EVs load bystander cells with CD30 and generate new targets among supporter cells. This crossfire effect might contribute to the enormous clinical impact of BV, whereas the ADAM10-dependent cleavage to the mild systemic off-target effects of the treatment with BV.
Collapse
Affiliation(s)
- Hinrich P Hansen
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University Hospital of Cologne, Cologne, Germany
| | - Adriana F Paes Leme
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, Brazil
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
34
|
Petty AJ, Heyman B, Yang Y. Chimeric Antigen Receptor Cell Therapy: Overcoming Obstacles to Battle Cancer. Cancers (Basel) 2020; 12:cancers12040842. [PMID: 32244520 PMCID: PMC7226583 DOI: 10.3390/cancers12040842] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 01/04/2023] Open
Abstract
Chimeric antigen receptors (CAR) are fusion proteins engineered from antigen recognition, signaling, and costimulatory domains that can be used to reprogram T cells to specifically target tumor cells expressing specific antigens. Current CAR-T cell technology utilizes the patient's own T cells to stably express CARs and has achieved exciting clinical success in the past few years. However, current CAR-T cell therapy still faces several challenges, including suboptimal persistence and potency, impaired trafficking to solid tumors, local immunosuppression within the tumor microenvironment and intrinsic toxicity associated with CAR-T cells. This review focuses on recent strategies to improve the clinical efficacy of CAR-T cell therapy and other exciting CAR approaches currently under investigation, including CAR natural killer (NK) and NKT cell therapies.
Collapse
Affiliation(s)
- Amy J. Petty
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Benjamin Heyman
- Division of Regenerative Medicine, Department of Medicine, UC San Diego, La Jolla, CA 92093, USA
- Correspondence: (B.H.); (Y.Y.)
| | - Yiping Yang
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (B.H.); (Y.Y.)
| |
Collapse
|
35
|
Abstract
Pediatric T-cell hematologic malignancies are a diverse group of rare cancers. The most common pediatric T-cell malignancies include T-cell acute lymphoblastic leukemia (T-ALL) and anaplastic large cell lymphoma (ALCL). Although the overall survival rates have improved markedly in recent years, children with relapsed T-ALL and ALCL have very low rates of cure and few salvage therapies exist. Current treatment regimens rely on toxic chemotherapies with significant short- and long-term morbidity. Immunotherapies, including antibodies and adoptive cellular therapies, have revolutionized the treatment of B-cell malignancies in pediatrics. The adaptation of these therapies to T-cell malignancies has been slower because of challenges implicit in the design and implementation of immunotherapies for T-cell malignancies, including the potential risks of fratricide, immunosuppression, and graft versus host disease (GVHD). We present a review of current challenges in the development of immunotherapies for T-cell hematologic malignancies, potential solutions and therapies under investigation. We include a particular focus on T-ALL and ALCL. Immunotherapies offer promising strategies to improve outcomes in children with T-cell malignancies, particularly in the setting of relapse. Optimizing efficacy, mitigating toxicity, and safely integrating with conventional therapies are key considerations as immunotherapies are translated into the clinic.
Collapse
Affiliation(s)
- Caroline Diorio
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Pereleman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David T Teachey
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Pereleman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
36
|
Abstract
Oligonucleotide aptamers are a class of small-molecule ligands. Functionally similar to protein antibodies, aptamers can specifically bind to their targets with high affinity. Biomedical studies have revealed the potential clinical value of aptamer technology for disease diagnosis and targeted therapy. Lymphoma is a group of cancers originating from the lymphatic system. Currently, chemotherapy is the primary treatment for lymphoma, although it may cause serious side effects in patients due to lack of target specificity. Here, we selectively discuss the recent development of potential applications of aptamer technology for precision lymphoma therapy, which are able to not only achieve high therapeutic efficacy but also do not cause off-target side effects.
Collapse
|
37
|
Bobrowicz M, Zagozdzon R, Domagala J, Vasconcelos-Berg R, Guenova E, Winiarska M. Monoclonal Antibodies in Dermatooncology-State of the Art and Future Perspectives. Cancers (Basel) 2019; 11:E1420. [PMID: 31554169 PMCID: PMC6826541 DOI: 10.3390/cancers11101420] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/08/2019] [Accepted: 09/17/2019] [Indexed: 12/21/2022] Open
Abstract
Monoclonal antibodies (mAbs) targeting specific proteins are currently the most popular form of immunotherapy used in the treatment of cancer and other non-malignant diseases. Since the first approval of anti-CD20 mAb rituximab in 1997 for the treatment of B-cell malignancies, the market is continuously booming and the clinically used mAbs have undergone a remarkable evolution. Novel molecular targets are constantly emerging and the development of genetic engineering have facilitated the introduction of modified mAbs with improved safety and increased capabilities to activate the effector mechanisms of the immune system. Next to their remarkable success in hematooncology, mAbs have also an already established role in the treatment of solid malignancies. The recent development of mAbs targeting the immune checkpoints has opened new avenues for the use of this form of immunotherapy, also in the immune-rich milieu of the skin. In this review we aim at presenting a comprehensive view of mAbs' application in the modern treatment of skin cancer. We present the characteristics and efficacy of mAbs currently used in dermatooncology and summarize the recent clinical trials in the field. We discuss the side effects and strategies for their managing.
Collapse
Affiliation(s)
| | - Radoslaw Zagozdzon
- Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland.
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, 02-006 Warsaw, Poland.
| | - Joanna Domagala
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland.
- Postgraduate School of Molecular Medicine, 02-091 Warsaw, Poland.
| | - Roberta Vasconcelos-Berg
- Department of Dermatology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland.
| | - Emmanuella Guenova
- Department of Dermatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland.
- Department of Dermatology, University of Lausanne, 1011 Lausanne, Switzerland.
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland.
| |
Collapse
|
38
|
Adoptive Cell Therapy for Acute Myeloid Leukemia and T-Cell Acute Lymphoblastic Leukemia. ACTA ACUST UNITED AC 2019; 25:199-207. [PMID: 31135527 DOI: 10.1097/ppo.0000000000000376] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Refractory and relapsed acute myeloid leukemia (AML) and T-lineage leukemia have poor prognosis and limited therapeutic options. Adoptive cellular immunotherapies are emerging as an effective treatment for patients with chemotherapy refractory hematological malignancies. Indeed, the use of unselected donor lymphocyte infusions has demonstrated successes in treating patients with AML and T-lineage leukemia post-allogeneic transplantation. The development of ex vivo manipulation techniques such as genetic modification or selection and expansion of individual cellular components has permitted the clinical translation of a wide range of promising cellular therapies for AML and T-cell acute lymphoblastic leukemia. Here, we will review clinical studies to date using adoptive cell therapy approaches and outline the major challenges limiting the development of safe and effective cell therapies for both types of acute leukemia.
Collapse
|