551
|
|
552
|
|
553
|
Hung CF, Xu X, Li L, Ma Y, Jin Q, Viley A, Allen C, Natarajan P, Shivakumar R, Peshwa MV, Emens LA. Development of Anti-Human Mesothelin-Targeted Chimeric Antigen Receptor Messenger RNA-Transfected Peripheral Blood Lymphocytes for Ovarian Cancer Therapy. Hum Gene Ther 2018; 29:614-625. [PMID: 29334771 DOI: 10.1089/hum.2017.080] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD19-targeted chimeric antigen receptor (CAR) engineered T/natural killer (NK)-cell therapies can result in durable clinical responses in B-cell malignancies. However, CAR-based immunotherapies have been much less successful in solid cancers, in part due to "on-target off-tumor" toxicity related to expression of target tumor antigens on normal tissue. Based on preliminary observations of safety and clinical activity in proof-of-concept clinical trials, tumor antigen-specific messenger RNA (mRNA) CAR transfection into selected, activated, and expanded T/NK cells may permit prospective control of "on-target off-tumor" toxicity. To develop a commercial product for solid tumors, mesothelin was selected as an antigen target based on its association with poor prognosis and overexpression in multiple solid cancers. It was hypothesized that selecting, activating, and expanding cells ex vivo prior to mRNA CAR transfection would not be necessary, thus simplifying the complexity and cost of manufacturing. Now, the development of anti-human mesothelin mRNA CAR transfected peripheral blood lymphocytes (CARMA-hMeso) is reported, demonstrating the manufacture and cryopreservation of multiple cell aliquots for repeat administrations from a single human leukapheresis. A rapid, automated, closed system for cGMP-compliant transfection of mRNA CAR in up to 20 × 109 peripheral blood lymphocytes was developed. Here we show that CARMA-hMeso cells recognize and lyse tumor cells in a mesothelin-specific manner. Expression of CAR was detectable over approximately 7 days in vitro, with a progressive decline of CAR expression that appears to correlate with in vitro cell expansion. In a murine ovarian cancer model, a single intraperitoneal injection of CARMA-hMeso resulted in the dose-dependent inhibition of tumor growth and improved survival of mice. Furthermore, repeat weekly intraperitoneal administrations of the optimal CARMA-hMeso dose further prolonged disease control and survival. No significant off-target toxicities were observed. These data support further investigation of CARMA-hMeso as a potential treatment for ovarian cancer and other mesothelin-expressing cancers.
Collapse
Affiliation(s)
- Chien-Fu Hung
- 1 Department of Pathology, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Xuequn Xu
- 1 Department of Pathology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Linhong Li
- 3 MaxCyte, Inc. , Gaithersburg, Maryland
| | - Ying Ma
- 1 Department of Pathology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Qiu Jin
- 1 Department of Pathology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | | | | | | | | | | | - Leisha A Emens
- 2 Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine , Baltimore, Maryland.,4 Johns Hopkins Bloomberg-Kimmel Institute for Cancer Immunotherapy , Baltimore, Maryland
| |
Collapse
|
554
|
Krackhardt AM, Anliker B, Hildebrandt M, Bachmann M, Eichmüller SB, Nettelbeck DM, Renner M, Uharek L, Willimsky G, Schmitt M, Wels WS, Schüssler-Lenz M. Clinical translation and regulatory aspects of CAR/TCR-based adoptive cell therapies-the German Cancer Consortium approach. Cancer Immunol Immunother 2018; 67:513-523. [PMID: 29380009 PMCID: PMC11028374 DOI: 10.1007/s00262-018-2119-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 01/20/2018] [Indexed: 12/17/2022]
Abstract
Adoptive transfer of T cells genetically modified by TCRs or CARs represents a highly attractive novel therapeutic strategy to treat malignant diseases. Various approaches for the development of such gene therapy medicinal products (GTMPs) have been initiated by scientists in recent years. To date, however, the number of clinical trials commenced in Germany and Europe is still low. Several hurdles may contribute to the delay in clinical translation of these therapeutic innovations including the significant complexity of manufacture and non-clinical testing of these novel medicinal products, the limited knowledge about the intricate regulatory requirements of the academic developers as well as limitations of funds for clinical testing. A suitable good manufacturing practice (GMP) environment is a key prerequisite and platform for the development, validation, and manufacture of such cell-based therapies, but may also represent a bottleneck for clinical translation. The German Cancer Consortium (DKTK) and the Paul-Ehrlich-Institut (PEI) have initiated joint efforts of researchers and regulators to facilitate and advance early phase, academia-driven clinical trials. Starting with a workshop held in 2016, stakeholders from academia and regulatory authorities in Germany have entered into continuing discussions on a diversity of scientific, manufacturing, and regulatory aspects, as well as the benefits and risks of clinical application of CAR/TCR-based cell therapies. This review summarizes the current state of discussions of this cooperative approach providing a basis for further policy-making and suitable modification of processes.
Collapse
Affiliation(s)
- Angela M Krackhardt
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, TU München, TUM School of Medicine, Munich, Germany.
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany.
| | - Brigitte Anliker
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| | - Martin Hildebrandt
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- TUMCells (Interdisciplinary Center for Cellular Therapies), TUM School of Medicine, Munich, Germany
| | - Michael Bachmann
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Helmholtz Zentrum Dresden Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Radio and Tumorimmunology, Dresden, Germany
- Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg and Dresden, Germany
| | - Stefan B Eichmüller
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg and Dresden, Germany
- GMP and T Cell Therapy Unit, DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
| | - Dirk M Nettelbeck
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
| | - Matthias Renner
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| | - Lutz Uharek
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Stem Cell Facility, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gerald Willimsky
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Schmitt
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Department of Internal Medicine V, GMP Core Facility, Heidelberg University Hospital, Heidelberg, Germany
| | - Winfried S Wels
- DKTK-Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium) and DKFZ-Deutsches Krebsforschungszentrum (German Cancer Research Center), Heidelberg, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Martina Schüssler-Lenz
- Paul-Ehrlich-Institut (PEI, German Federal Institute for Vaccines and Biomedicines), Langen, Germany
| |
Collapse
|
555
|
|
556
|
Tat T, Li H, Constantinescu CS, Onaciu A, Chira S, Osan C, Pasca S, Petrushev B, Moisoiu V, Micu WT, Berce C, Tranca S, Dima D, Berindan-Neagoe I, Shen J, Tomuleasa C, Qian L. Genetically enhanced T lymphocytes and the intensive care unit. Oncotarget 2018; 9:16557-16572. [PMID: 29662667 PMCID: PMC5893262 DOI: 10.18632/oncotarget.24637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/26/2018] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor-modified T cells (CAR-T cells) and donor lymphocyte infusion (DLI) are important protocols in lymphocyte engineering. CAR-T cells have emerged as a new modality for cancer immunotherapy due to their potential efficacy against hematological malignancies. These genetically modified receptors contain an antigen-binding moiety, a hinge region, a transmembrane domain, and an intracellular costimulatory domain resulting in lymphocyte T cell activation subsequent to antigen binding. In present-day medicine, four generations of CAR-T cells are described depending on the intracellular signaling domain number of T cell receptors. DLI represents a form of adoptive therapy used after hematopoietic stem cell transplant for its anti-tumor and anti-infectious properties. This article covers the current status of CAR-T cells and DLI research in the intensive care unit (ICU) patient, including the efficacy, toxicity, side effects and treatment.
Collapse
Affiliation(s)
- Tiberiu Tat
- Intensive Care Unit, Ion Chiricuta Clinical Cancer Research, Cluj Napoca, Romania
- Department of Anesthesiology-Intensive Care, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Huming Li
- Department of Pulmonary and Critical Care Medicine, Navy General Hospital of PLA, Beijing, China
| | - Catalin-Sorin Constantinescu
- Intensive Care Unit, Ion Chiricuta Clinical Cancer Research, Cluj Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Anca Onaciu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sergiu Chira
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Ciprian Osan
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sergiu Pasca
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Bobe Petrushev
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Vlad Moisoiu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Wilhelm-Thomas Micu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Cristian Berce
- Department of Experimental Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sebastian Tranca
- Department of Anesthesiology-Intensive Care, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Research, Cluj Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Jianliang Shen
- Department of Hematology, Navy General Hospital of PLA, Beijing, China
| | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Clinical Cancer Research, Cluj Napoca, Romania
- Research Center for Functional Genomics and Translational Medicine / Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Liren Qian
- Department of Hematology, Navy General Hospital of PLA, Beijing, China
| |
Collapse
|
557
|
Murad JM, Graber DJ, Sentman CL. Advances in the use of natural receptor- or ligand-based chimeric antigen receptors (CARs) in haematologic malignancies. Best Pract Res Clin Haematol 2018; 31:176-183. [PMID: 29909918 DOI: 10.1016/j.beha.2018.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/12/2018] [Indexed: 12/27/2022]
Abstract
Chimeric antigen receptors (CAR)-T cell therapy has recently made promising advances towards treatment of B-cell malignancies. This approach makes use of an antibody-derived single chain variable fragment (scFv)-based CAR to target the CD19 antigen. Currently scFvs are the most common strategy for creation of CARs, but tumor cells can also be targeted using non-antibody based approaches with designs focused on the interaction between natural receptors and their ligands. This emerging strategy has been used in unique ways to target multiple tumor types, including solid and haematological malignancies. In this review, we will highlight the performance of receptor-ligand combinations as designs for CARs to treat cancer, with a particular focus on haematologic malignancies.
Collapse
Affiliation(s)
- Joana M Murad
- Celdara Medical LLC, Lebanon, NH, 16 Cavendish Ct Suite 240, Lebanon, NH 03766, USA.
| | - David J Graber
- Center for Synthetic Immunity and Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Dr., Lebanon, NH 03765, USA.
| | - Charles L Sentman
- Center for Synthetic Immunity and Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Dr., Lebanon, NH 03765, USA.
| |
Collapse
|
558
|
Stavrou M, Philip B, Traynor-White C, Davis CG, Onuoha S, Cordoba S, Thomas S, Pule M. A Rapamycin-Activated Caspase 9-Based Suicide Gene. Mol Ther 2018; 26:1266-1276. [PMID: 29661681 PMCID: PMC5993966 DOI: 10.1016/j.ymthe.2018.03.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 02/26/2018] [Accepted: 03/03/2018] [Indexed: 01/20/2023] Open
Abstract
Engineered T cell therapies show considerable promise in the treatment of refractory malignancies. Given the ability of engineered T cells to engraft and persist for prolonged periods along with unpredicted toxicities, incorporation of a suicide gene to allow selective depletion after administration is desirable. Rapamycin is a safe and widely available immunosuppressive pharmaceutical that acts by heterodimerization of FKBP12 with the FRB fragment of mTOR. The apical caspase caspase 9 is activated by homodimerization through its CARD domain. We developed a rapamycin-induced caspase 9 suicide gene. First, we showed that caspase 9 could be activated by a two-protein format with replacement of the CARD domain with both FRB and FKBP12. We next identified an optimal compact single-protein rapamycin caspase 9 (rapaCasp9) by fusing both FRB and FKBP12 with the catalytic domain of caspase 9. Functionality of rapaCasp9 when co-expressed with a CD19 CAR was demonstrated in vitro and in vivo.
Collapse
Affiliation(s)
| | - Brian Philip
- Cancer Institute, University College London, London, UK
| | | | | | | | | | - Simon Thomas
- Autolus Ltd., Forest House, White City, London, UK
| | - Martin Pule
- Autolus Ltd., Forest House, White City, London, UK; Cancer Institute, University College London, London, UK.
| |
Collapse
|
559
|
Rosenberg J, Huang J. CD8 + T Cells and NK Cells: Parallel and Complementary Soldiers of Immunotherapy. Curr Opin Chem Eng 2018; 19:9-20. [PMID: 29623254 PMCID: PMC5880541 DOI: 10.1016/j.coche.2017.11.006] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CD8+ T cells and NK cells are both cytotoxic effector cells of the immune system, but the recognition, specificity, sensitivity, and memory mechanisms are drastically different. While many of these topics have been extensively studied in CD8+ T cells, very little is known about NK cells. Current cancer immunotherapies mainly focus on CD8+ T cells, but have many issues of toxicity and efficacy. Given the heterogeneous nature of cancer, personalized cancer immunotherapy that integrates the power of both CD8+ T cells in adaptive immunity and NK cells in innate immunity might be the future direction, along with precision targeting and effective delivery of tumor-specific, memory CD8+ T cells and NK cells.
Collapse
Affiliation(s)
- Jillian Rosenberg
- Committee on Cancer Biology, The University of Chicago, IL 60637, USA
| | - Jun Huang
- Committee on Cancer Biology, The University of Chicago, IL 60637, USA
- Institute for Molecular Engineering, The University of Chicago, IL 60637, USA
| |
Collapse
|
560
|
Abstract
The practice of autoimmune neurology focuses on the diagnosis and treatment of a wide spectrum of neurological conditions driven by abnormal immune responses directed against neural tissues. These include autoimmune, paraneoplastic, postinfectious, and iatrogenic conditions. Symptoms of autoimmune neurologic disorders can be diverse and often difficult to recognize in their early stages, complicating the diagnosis. This review discusses the classification and management of common autoimmune neurological conditions, placing an emphasis on the rapid identification of autoimmune etiology and mechanism of immune dysfunction to allow for the timely institution of appropriate treatment.
Collapse
|
561
|
CAR-T Cells: Next Generation Cancer Therapeutics. J Indian Inst Sci 2018. [DOI: 10.1007/s41745-018-0062-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
562
|
Tomuleasa C, Fuji S, Berce C, Onaciu A, Chira S, Petrushev B, Micu WT, Moisoiu V, Osan C, Constantinescu C, Pasca S, Jurj A, Pop L, Berindan-Neagoe I, Dima D, Kitano S. Chimeric Antigen Receptor T-Cells for the Treatment of B-Cell Acute Lymphoblastic Leukemia. Front Immunol 2018. [PMID: 29515572 PMCID: PMC5825894 DOI: 10.3389/fimmu.2018.00239] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell technology has seen a rapid development over the last decade mostly due to the potential that these cells may have in treating malignant diseases. It is a generally accepted principle that very few therapeutic compounds deliver a clinical response without treatment-related toxicity, and studies have shown that CAR T-cells are not an exception to this rule. While large multinational drug companies are currently investigating the potential role of CAR T-cells in hematological oncology, the potential of such cellular therapies are being recognized worldwide as they are expected to expand in the patient to support the establishment of the immune memory, provide a continuous surveillance to prevent and/or treat a relapse, and keep the targeted malignant cell subpopulation in check. In this article, we present the possible advantages of using CAR T-cells in treating acute lymphoblastic leukemia, presenting the technology and the current knowledge in their preclinical and early clinical trial use. Thus, this article first presents the main present-day knowledge on the standard of care for acute lymphoblastic leukemia. Afterward, current knowledge is presented about the use of CAR T-cells in cancer immunotherapy, describing their design, the molecular constructs, and the preclinical data on murine models to properly explain the background for their clinical use. Last, but certainly not least, this article presents the use of CAR T-cells for the immunotherapy of B-cell acute lymphoblastic leukemia, describing both their potential clinical advantages and the possible side effects.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Department of Hematology, Oncology Institute Prof. Dr. Ion Chiricuta, Cluj Napoca, Romania.,Research Center for Functional Genomics and Translational Medicine, Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Shigeo Fuji
- Department of Stem Cell Transplantation, Osaka International Cancer Institute, Osaka, Japan
| | - Cristian Berce
- Animal Facility, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Anca Onaciu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sergiu Chira
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Bobe Petrushev
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Wilhelm-Thomas Micu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Vlad Moisoiu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Ciprian Osan
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sergiu Pasca
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Laura Pop
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Delia Dima
- Department of Hematology, Oncology Institute Prof. Dr. Ion Chiricuta, Cluj Napoca, Romania
| | - Shigehisa Kitano
- Division of Cancer Immunotherapy, Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
563
|
Vormittag P, Gunn R, Ghorashian S, Veraitch FS. A guide to manufacturing CAR T cell therapies. Curr Opin Biotechnol 2018; 53:164-181. [PMID: 29462761 DOI: 10.1016/j.copbio.2018.01.025] [Citation(s) in RCA: 224] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/10/2018] [Accepted: 01/22/2018] [Indexed: 01/07/2023]
Abstract
In recent years, chimeric antigen receptor (CAR) modified T cells have been used as a treatment for haematological malignancies in several phase I and II trials and with Kymriah of Novartis and Yescarta of KITE Pharma, the first CAR T cell therapy products have been approved. Promising clinical outcomes have yet been tempered by the fact that many therapies may be prohibitively expensive to manufacture. The process is not yet defined, far from being standardised and often requires extensive manual handling steps. For academia, big pharma and contract manufacturers it is difficult to obtain an overview over the process strategies and their respective advantages and disadvantages. This review details current production processes being used for CAR T cells with a particular focus on efficacy, reproducibility, manufacturing costs and release testing. By undertaking a systematic analysis of the manufacture of CAR T cells from reported clinical trial data to date, we have been able to quantify recent trends and track the uptake of new process technology. Delivering new processing options will be key to the success of the CAR-T cells ensuring that excessive manufacturing costs do not disrupt the delivery of exciting new therapies to the wide possible patient cohort.
Collapse
Affiliation(s)
- Philipp Vormittag
- Karlsruhe Institute of Technology, Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Fritz-Haber-Weg 2, 76131 Karlsruhe, Germany
| | - Rebecca Gunn
- University College London, Department of Biochemical Engineering, Gower Street, London WC1E 6BT, United Kingdom
| | - Sara Ghorashian
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1E, United Kingdom
| | - Farlan S Veraitch
- University College London, Department of Biochemical Engineering, Gower Street, London WC1E 6BT, United Kingdom.
| |
Collapse
|
564
|
Abstract
Chimeric antigen receptor (CAR) T-cells are redirected T-cells that can recognize cancer antigens in a major histocompatibility complex (MHC)-independent fashion. A typical CAR is comprised of two main functional domains: an extracellular antigen recognition domain, called a single-chain variable fragment (scFv), and an intracellular signaling domain. Based on the number of intracellular signaling molecules, CARs are categorized into four generations. CAR T-cell therapy has become a promising treatment for hematologic malignancies. However, results of its clinical trials on solid tumors have not been encouraging. Here, we described the structure of CARs and summarized the clinical trials of CD19-targeted CAR T-cells. The side effects, safety management, challenges, and future prospects of CAR T-cells for the treatment of cancer, particularly for solid tumors, were also discussed.
Collapse
Affiliation(s)
- Niaz Muhammad
- a Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences , Shaanxi Normal University , Xi'an , P.R. China
| | - Qinwen Mao
- b Department of Pathology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Haibin Xia
- a Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences , Shaanxi Normal University , Xi'an , P.R. China
| |
Collapse
|
565
|
Badieyan ZS, Hoseini SS. Adverse Effects Associated with Clinical Applications of CAR Engineered T Cells. Arch Immunol Ther Exp (Warsz) 2018; 66:283-288. [PMID: 29427174 DOI: 10.1007/s00005-018-0507-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 10/18/2017] [Indexed: 01/18/2023]
Abstract
Cancer has been ranked as the second leading cause of death in the United States. To reduce cancer mortality, immunotherapy is gaining momentum among other therapeutic modalities, due to its impressive results in clinical trials. The genetically engineered T cells expressing chimeric antigen receptors (CARs) are emerging as a new approach in cancer immunotherapy, with the most successful outcomes in the refractory/relapse hematologic malignancies. However, the widespread clinical applications are limited by adverse effects some of which are life-threatening. Strategies to reduce the chance of side effects as well as close monitoring, rapid diagnosis and proper treatment of side effects are necessary to take the most advantages of this valuable therapy. Here we review the reported toxicities associated with CAR engineered T cells, the strategies to ameliorate the toxicity, and further techniques and designs leading to a safer CAR T-cell therapy.
Collapse
Affiliation(s)
| | - Sayed Shahabuddin Hoseini
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 170, New York, NY, 10065, USA.
| |
Collapse
|
566
|
Piscopo NJ, Mueller KP, Das A, Hematti P, Murphy WL, Palecek SP, Capitini CM, Saha K. Bioengineering Solutions for Manufacturing Challenges in CAR T Cells. Biotechnol J 2018; 13:10.1002/biot.201700095. [PMID: 28840981 PMCID: PMC5796845 DOI: 10.1002/biot.201700095] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/26/2017] [Indexed: 12/13/2022]
Abstract
The next generation of therapeutic products to be approved for the clinic is anticipated to be cell therapies, termed "living drugs" for their capacity to dynamically and temporally respond to changes during their production ex vivo and after their administration in vivo. Genetically engineered chimeric antigen receptor (CAR) T cells have rapidly developed into powerful tools to harness the power of immune system manipulation against cancer. Regulatory agencies are beginning to approve CAR T cell therapies due to their striking efficacy in treating some hematological malignancies. However, the engineering and manufacturing of such cells remains a challenge for widespread adoption of this technology. Bioengineering approaches including biomaterials, synthetic biology, metabolic engineering, process control and automation, and in vitro disease modeling could offer promising methods to overcome some of these challenges. Here, we describe the manufacturing process of CAR T cells, highlighting potential roles for bioengineers to partner with biologists and clinicians to advance the manufacture of these complex cellular products under rigorous regulatory and quality control.
Collapse
Affiliation(s)
- Nicole J Piscopo
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Katherine P Mueller
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Amritava Das
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA
| | - Christian M Capitini
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
567
|
Liu E, Tong Y, Dotti G, Shaim H, Savoldo B, Mukherjee M, Orange J, Wan X, Lu X, Reynolds A, Gagea M, Banerjee P, Cai R, Bdaiwi MH, Basar R, Muftuoglu M, Li L, Marin D, Wierda W, Keating M, Champlin R, Shpall E, Rezvani K. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 2018; 32:520-531. [PMID: 28725044 PMCID: PMC6063081 DOI: 10.1038/leu.2017.226] [Citation(s) in RCA: 523] [Impact Index Per Article: 87.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/29/2022]
Abstract
Chimeric antigen receptors (CARs) have been used to redirect the specificity of autologous T cells against leukemia and lymphoma with promising clinical results. Extending this approach to allogeneic T cells is problematic as they carry a significant risk of graft-versus-host disease (GVHD). Natural killer (NK) cells are highly cytotoxic effectors, killing their targets in a non-antigen-specific manner without causing GVHD. Cord blood (CB) offers an attractive, allogeneic, off-the-self source of NK cells for immunotherapy. We transduced CB-derived NK cells with a retroviral vector incorporating the genes for CAR-CD19, IL-15 and inducible caspase-9-based suicide gene (iC9), and demonstrated efficient killing of CD19-expressing cell lines and primary leukemia cells in vitro, with marked prolongation of survival in a xenograft Raji lymphoma murine model. Interleukin-15 (IL-15) production by the transduced CB-NK cells critically improved their function. Moreover, iC9/CAR.19/IL-15 CB-NK cells were readily eliminated upon pharmacologic activation of the iC9 suicide gene. In conclusion, we have developed a novel approach to immunotherapy using engineered CB-derived NK cells, which are easy to produce, exhibit striking efficacy and incorporate safety measures to limit toxicity. This approach should greatly improve the logistics of delivering this therapy to large numbers of patients, a major limitation to current CAR-T-cell therapies.
Collapse
Affiliation(s)
- Enli Liu
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - Yijiu Tong
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC
| | - Hila Shaim
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - Barbara Savoldo
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC
| | - Malini Mukherjee
- The Center for Human Immunobiology, Baylor College of Medicine, Houston, TX
| | - Jordan Orange
- The Center for Human Immunobiology, Baylor College of Medicine, Houston, TX
| | - Xinhai Wan
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - Xinyan Lu
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX
| | | | - Mihai Gagea
- Department of Veterinary Medicine & Surgery, MD Anderson Cancer Center, Houston, TX
| | - Pinaki Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - Rong Cai
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | | | | | - Muharrem Muftuoglu
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - Li Li
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - William Wierda
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Michael Keating
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
568
|
Lo Presti V, Nierkens S, Boelens JJ, van Til NP. Use of cord blood derived T-cells in cancer immunotherapy: milestones achieved and future perspectives. Expert Rev Hematol 2018; 11:209-218. [PMID: 29359983 DOI: 10.1080/17474086.2018.1431119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Hematopoietic cell transplantation is a potentially lifesaving procedure for patients with hematological malignancies who are refractory to conventional chemotherapy and/or irradiation treatment. Umbilical cord blood (CB) transplantation, as a hematopoietic stem and progenitor cell (HSPC) source, has several advantages over bone marrow transplantation with respect to matching and prompt availability for transplantation. Additionally, CB has some inherent features, such as rapid expansion of T cells, lower prevalence of graft-versus-host disease and higher graft versus tumor efficacy that make this HSPC cell source more favorable over other HSPC sources. Areas covered: This review summarizes the current CB and CB derived T cell applications aiming to better disease control for hematological malignancies and discusses future directions to more effective therapies. Expert commentary: CB transplantation could be used as a platform to extract cord blood derived T cells for ex vivo expansion and/or gene modification to improve cellular immunotherapies. In addition, combining cord blood gene-engineered T cell products with vaccination strategies, such as cord blood derived dendritic cell based vaccines, may provide synergistic immunotherapies with enhanced anti-tumor effects.
Collapse
Affiliation(s)
- Vania Lo Presti
- a Laboratory of Translational Immunology , University Medical Center Utrecht , Utrecht , the Netherlands
| | - Stefan Nierkens
- a Laboratory of Translational Immunology , University Medical Center Utrecht , Utrecht , the Netherlands
| | - Jaap Jan Boelens
- a Laboratory of Translational Immunology , University Medical Center Utrecht , Utrecht , the Netherlands.,b Pediatric Blood and Marrow Transplantation Program , University Medical Center Utrecht , Utrecht , the Netherlands
| | - Niek P van Til
- a Laboratory of Translational Immunology , University Medical Center Utrecht , Utrecht , the Netherlands
| |
Collapse
|
569
|
Kamran N, Alghamri MS, Nunez FJ, Shah D, Asad AS, Candolfi M, Altshuler D, Lowenstein PR, Castro MG. Current state and future prospects of immunotherapy for glioma. Immunotherapy 2018; 10:317-339. [PMID: 29421984 PMCID: PMC5810852 DOI: 10.2217/imt-2017-0122] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/30/2017] [Indexed: 12/14/2022] Open
Abstract
There is a large unmet need for effective therapeutic approaches for glioma, the most malignant brain tumor. Clinical and preclinical studies have enormously expanded our knowledge about the molecular aspects of this deadly disease and its interaction with the host immune system. In this review we highlight the wide array of immunotherapeutic interventions that are currently being tested in glioma patients. Given the molecular heterogeneity, tumor immunoediting and the profound immunosuppression that characterize glioma, it has become clear that combinatorial approaches targeting multiple pathways tailored to the genetic signature of the tumor will be required in order to achieve optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Neha Kamran
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Mahmoud S Alghamri
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Felipe J Nunez
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Diana Shah
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Antonela S Asad
- Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - David Altshuler
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Maria G Castro
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| |
Collapse
|
570
|
Ulrich P, Blaich G, Baumann A, Fagg R, Hey A, Kiessling A, Kronenberg S, Lindecrona RH, Mohl S, Richter WF, Tibbitts J, Crameri F, Weir L. Biotherapeutics in non-clinical development: Strengthening the interface between safety, pharmacokinetics-pharmacodynamics and manufacturing. Regul Toxicol Pharmacol 2018; 94:91-100. [PMID: 29355662 DOI: 10.1016/j.yrtph.2018.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/14/2018] [Indexed: 12/29/2022]
Abstract
Biological drugs comprise a wide field of different modalities with respect to structure, pharmacokinetics and pharmacological function. Considerable non-clinical experience in the development of proteins (e.g. insulin) and antibodies has been accumulated over the past thirty years. In order to improve the efficacy and the safety of these biotherapeutics, Fc modifications (e.g. Fc silent antibody versions), combinations (antibody-drug conjugates, protein-nanoparticle combinations), and new constructs (darpins, fynomers) have been introduced. In the last decade, advanced therapy medicinal products (ATMPs) in research and development have become a considerable and strongly growing part of the biotherapeutic portfolio. ATMPs consisting of gene and cell therapy modalities or even combinations of them, further expand the level of complexity, which already exists in non-clinical development strategies for biological drugs and has thereby led to a further diversification of expertise in safety and PKPD assessment of biological drugs. It is the fundamental rationale of the BioSafe meetings, held yearly in the EU and in the US, to convene experts on a regular basis and foster knowledge exchange and mutual understanding in this fast growing area. In order to reflect at least partially the variety of the biotherapeutics field, the 2016 EU BioSafe meeting addressed the following topics in six sessions: (i) In vitro Meets in vivo to Leverage Biologics Development (ii) New developments and regulatory considerations in the cell and gene therapy field (iii) CMC Challenges with Biologics development (iv) Minipigs in non-clinical safety assessment (v) Opportunities of PKPD Assessment in Less Common Administration Routes In the breakout sessions the following questions were discussed: (i) Cynomolgus monkey as a reprotoxicology Species: Impact of Immunomodulators on Early Pregnancy Maintenance (ii) Safety Risk of Inflammation and Autoimmunity Induced by Immunomodulators (iii) Experience with non-GMP Material in Pivotal Non-clinical Safety Studies to Support First in Man (FiM) Trials (iv) Safety Assessment of Combination Products for Non-oncology.
Collapse
Affiliation(s)
| | | | | | | | - Adam Hey
- Novartis Pharma, Basel, Switzerland
| | | | - Sven Kronenberg
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | - Silke Mohl
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Wolfgang F Richter
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | - Flavio Crameri
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | |
Collapse
|
571
|
Genta S, Ghisoni E, Giannone G, Mittica G, Valabrega G. Reprogramming T-cells for adoptive immunotherapy of ovarian cancer. Expert Opin Biol Ther 2018; 18:359-367. [PMID: 29307234 DOI: 10.1080/14712598.2018.1425679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Epithelial ovarian cancer (EOC) is the most common cause of death among gynecological malignancies. Despite surgical and pharmacological efforts to improve patients' outcome, persistent and recurrent EOC remains an un-eradicable disease. Chimeric associated antigens (CAR) T cells are T lymphocytes expressing an engineered T cell receptor that activate the immune response after an MHC unrestricted recognition of specific antigens, including tumor associated antigens (TAAs). CART cells have been shown to be effective in the treatment of hematologic tumors even if frequently associated with potentially severe toxicity and high production costs. AREAS COVERED In this review, we will focus on preclinical and clinical studies evaluating CART activity in EOC in order to identify possible difficulties and advantages of their use in this particular setting. EXPERT OPINION The pattern of diffusion within the peritoneal cavity, the tumor microenvironment and the high rate of TAAs make EOC a particularly interesting model for CART cells use. Data from preclinical studies indicate a potential activity of CARTs in EOC, but robust clinical data are still awaited. Further studies are needed to determine the best methods of administration and the most effective CAR type to treat EOC patients.
Collapse
Affiliation(s)
- Sofia Genta
- a Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy .,b Department of Oncology , University of Torino , Turin , Italy
| | - Eleonora Ghisoni
- a Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy .,b Department of Oncology , University of Torino , Turin , Italy
| | - Gaia Giannone
- a Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy .,b Department of Oncology , University of Torino , Turin , Italy
| | - Gloria Mittica
- a Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy .,b Department of Oncology , University of Torino , Turin , Italy
| | - Giorgio Valabrega
- a Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy .,b Department of Oncology , University of Torino , Turin , Italy
| |
Collapse
|
572
|
Kumaresan PR, da Silva TA, Kontoyiannis DP. Methods of Controlling Invasive Fungal Infections Using CD8 + T Cells. Front Immunol 2018; 8:1939. [PMID: 29358941 PMCID: PMC5766637 DOI: 10.3389/fimmu.2017.01939] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022] Open
Abstract
Invasive fungal infections (IFIs) cause high rates of morbidity and mortality in immunocompromised patients. Pattern-recognition receptors present on the surfaces of innate immune cells recognize fungal pathogens and activate the first line of defense against fungal infection. The second line of defense is the adaptive immune system which involves mainly CD4+ T cells, while CD8+ T cells also play a role. CD8+ T cell-based vaccines designed to prevent IFIs are currently being investigated in clinical trials, their use could play an especially important role in acquired immune deficiency syndrome patients. So far, none of the vaccines used to treat IFI have been approved by the FDA. Here, we review current and future antifungal immunotherapy strategies involving CD8+ T cells. We highlight recent advances in the use of T cells engineered using a Sleeping Beauty vector to treat IFIs. Recent clinical trials using chimeric antigen receptor (CAR) T-cell therapy to treat patients with leukemia have shown very promising results. We hypothesized that CAR T cells could also be used to control IFI. Therefore, we designed a CAR that targets β-glucan, a sugar molecule found in most of the fungal cell walls, using the extracellular domain of Dectin-1, which binds to β-glucan. Mice treated with D-CAR+ T cells displayed reductions in hyphal growth of Aspergillus compared to the untreated group. Patients suffering from IFIs due to primary immunodeficiency, secondary immunodeficiency (e.g., HIV), or hematopoietic transplant patients may benefit from bioengineered CAR T cell therapy.
Collapse
Affiliation(s)
- Pappanaicken R. Kumaresan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Thiago Aparecido da Silva
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dimitrios P. Kontoyiannis
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
573
|
Zheng P, Li J, Kros JM. Breakthroughs in modern cancer therapy and elusive cardiotoxicity: Critical research-practice gaps, challenges, and insights. Med Res Rev 2018; 38:325-376. [PMID: 28862319 PMCID: PMC5763363 DOI: 10.1002/med.21463] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 12/16/2022]
Abstract
To date, five cancer treatment modalities have been defined. The three traditional modalities of cancer treatment are surgery, radiotherapy, and conventional chemotherapy, and the two modern modalities include molecularly targeted therapy (the fourth modality) and immunotherapy (the fifth modality). The cardiotoxicity associated with conventional chemotherapy and radiotherapy is well known. Similar adverse cardiac events are resurging with the fourth modality. Aside from the conventional and newer targeted agents, even the most newly developed, immune-based therapeutic modalities of anticancer treatment (the fifth modality), e.g., immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapy, have unfortunately led to potentially lethal cardiotoxicity in patients. Cardiac complications represent unresolved and potentially life-threatening conditions in cancer survivors, while effective clinical management remains quite challenging. As a consequence, morbidity and mortality related to cardiac complications now threaten to offset some favorable benefits of modern cancer treatments in cancer-related survival, regardless of the oncologic prognosis. This review focuses on identifying critical research-practice gaps, addressing real-world challenges and pinpointing real-time insights in general terms under the context of clinical cardiotoxicity induced by the fourth and fifth modalities of cancer treatment. The information ranges from basic science to clinical management in the field of cardio-oncology and crosses the interface between oncology and onco-pharmacology. The complexity of the ongoing clinical problem is addressed at different levels. A better understanding of these research-practice gaps may advance research initiatives on the development of mechanism-based diagnoses and treatments for the effective clinical management of cardiotoxicity.
Collapse
Affiliation(s)
- Ping‐Pin Zheng
- Cardio‐Oncology Research GroupErasmus Medical CenterRotterdamthe Netherlands
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| | - Jin Li
- Department of OncologyShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Johan M Kros
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| |
Collapse
|
574
|
Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, Komanduri KV, Lin Y, Jain N, Daver N, Westin J, Gulbis AM, Loghin ME, de Groot JF, Adkins S, Davis SE, Rezvani K, Hwu P, Shpall EJ. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol 2018; 15:47-62. [PMID: 28925994 PMCID: PMC6733403 DOI: 10.1038/nrclinonc.2017.148] [Citation(s) in RCA: 1605] [Impact Index Per Article: 267.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immunotherapy using T cells genetically engineered to express a chimeric antigen receptor (CAR) is rapidly emerging as a promising new treatment for haematological and non-haematological malignancies. CAR-T-cell therapy can induce rapid and durable clinical responses, but is associated with unique acute toxicities, which can be severe or even fatal. Cytokine-release syndrome (CRS), the most commonly observed toxicity, can range in severity from low-grade constitutional symptoms to a high-grade syndrome associated with life-threatening multiorgan dysfunction; rarely, severe CRS can evolve into fulminant haemophagocytic lymphohistiocytosis (HLH). Neurotoxicity, termed CAR-T-cell-related encephalopathy syndrome (CRES), is the second most-common adverse event, and can occur concurrently with or after CRS. Intensive monitoring and prompt management of toxicities is essential to minimize the morbidity and mortality associated with this potentially curative therapeutic approach; however, algorithms for accurate and consistent grading and management of the toxicities are lacking. To address this unmet need, we formed a CAR-T-cell-therapy-associated TOXicity (CARTOX) Working Group, comprising investigators from multiple institutions and medical disciplines who have experience in treating patients with various CAR-T-cell therapy products. Herein, we describe the multidisciplinary approach adopted at our institutions, and provide recommendations for monitoring, grading, and managing the acute toxicities that can occur in patients treated with CAR-T-cell therapy.
Collapse
Affiliation(s)
- Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Sudhakar Tummala
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - William Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Cristina Gutierrez
- Department of Critical Care, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, Florida 33613, USA
| | - Krishna V Komanduri
- Adult Stem Cell Transplant Program, Sylvester Comprehensive Cancer Center, University of Miami, 1475 Northwest 12 th Avenue, Miami, Florida 33136, USA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, 200 First Street South West, Rochester, Minnesota 55905, USA
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Jason Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Alison M Gulbis
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Monica E Loghin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Sherry Adkins
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Suzanne E Davis
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Patrick Hwu
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| |
Collapse
|
575
|
Mikkilineni L, Kochenderfer JN. Chimeric antigen receptor T-cell therapies for multiple myeloma. Blood 2017; 130:2594-2602. [PMID: 28928126 PMCID: PMC5731088 DOI: 10.1182/blood-2017-06-793869] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/30/2017] [Indexed: 12/22/2022] Open
Abstract
Multiple myeloma (MM) is a nearly always incurable malignancy of plasma cells, so new approaches to treatment are needed. T-cell therapies are a promising approach for treating MM, with a mechanism of action different than those of standard MM treatments. Chimeric antigen receptors (CARs) are fusion proteins incorporating antigen-recognition domains and T-cell signaling domains. T cells genetically engineered to express CARs can specifically recognize antigens. Success of CAR-T cells (CAR-Ts) against leukemia and lymphoma has encouraged development of CAR-T therapies for MM. Target antigens for CARs must be expressed on malignant cells, but expression on normal cells must be absent or limited. B-cell maturation antigen is expressed by normal and malignant plasma cells. CAR-Ts targeting B-cell maturation antigen have demonstrated significant antimyeloma activity in early clinical trials. Toxicities in these trials, including cytokine release syndrome, have been similar to toxicities observed in CAR-T trials for leukemia. Targeting postulated CD19+ myeloma stem cells with anti-CD19 CAR-Ts is a novel approach to MM therapy. MM antigens including CD138, CD38, signaling lymphocyte-activating molecule 7, and κ light chain are under investigation as CAR targets. MM is genetically and phenotypically heterogeneous, so targeting of >1 antigen might often be required for effective treatment of MM with CAR-Ts. Integration of CAR-Ts with other myeloma therapies is an important area of future research. CAR-T therapies for MM are at an early stage of development but have great promise to improve MM treatment.
Collapse
Affiliation(s)
| | - James N Kochenderfer
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD
| |
Collapse
|
576
|
Cauwels A, Van Lint S, Garcin G, Bultinck J, Paul F, Gerlo S, Van der Heyden J, Bordat Y, Catteeuw D, De Cauwer L, Rogge E, Verhee A, Uzé G, Tavernier J. A safe and highly efficient tumor-targeted type I interferon immunotherapy depends on the tumor microenvironment. Oncoimmunology 2017; 7:e1398876. [PMID: 29399401 DOI: 10.1080/2162402x.2017.1398876] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/13/2017] [Accepted: 10/25/2017] [Indexed: 01/18/2023] Open
Abstract
Despite approval for the treatment of various malignancies, clinical application of cytokines such as type I interferon (IFN) is severely impeded by their systemic toxicity. AcTakines (Activity-on-Target cytokines) are optimized immunocytokines that, when injected in mice, only reveal their activity upon cell-specific impact. We here show that type I IFN-derived AcTaferon targeted to the tumor displays strong antitumor activity without any associated toxicity, in contrast with wild type IFN. Treatment with CD20-targeted AcTaferon of CD20+ lymphoma tumors or melanoma tumors engineered to be CD20+, drastically reduced tumor growth. This antitumor effect was completely lost in IFNAR- or Batf3-deficient mice, and depended on IFN signaling in conventional dendritic cells. Also the presence of, but not the IFN signaling in, CD8+ T lymphocytes was critical for proficient antitumor effects. When combined with immunogenic chemotherapy, low-dose TNF, or immune checkpoint blockade strategies such as anti-PDL1, anti-CTLA4 or anti-LAG3, complete tumor regressions and subsequent immunity (memory) were observed, still without any concomitant morbidity, again in sharp contrast with wild type IFN. Interestingly, the combination therapy of tumor-targeted AcTaferon with checkpoint inhibiting antibodies indicated its ability to convert nonresponding tumors into responders. Collectively, our findings demonstrate that AcTaferon targeted to tumor-specific surface markers may provide a safe and generic addition to cancer (immuno)therapies.
Collapse
Affiliation(s)
- Anje Cauwels
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Sandra Van Lint
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Geneviève Garcin
- University Montpellier, Place Eugène Bataillon, Montpellier, France
| | - Jennyfer Bultinck
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.,Present address: Oxyrane, Zwijnaarde-Gent, Belgium
| | - Franciane Paul
- University Montpellier, Place Eugène Bataillon, Montpellier, France
| | - Sarah Gerlo
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - José Van der Heyden
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Yann Bordat
- University Montpellier, Place Eugène Bataillon, Montpellier, France
| | - Dominiek Catteeuw
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Lode De Cauwer
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.,Present address: Argenx BVBA, Zwijnaarde-Gent, Belgium
| | - Elke Rogge
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Annick Verhee
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Gilles Uzé
- University Montpellier, Place Eugène Bataillon, Montpellier, France
| | - Jan Tavernier
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
577
|
Precision medicine for urothelial bladder cancer: update on tumour genomics and immunotherapy. Nat Rev Urol 2017; 15:92-111. [PMID: 29133939 DOI: 10.1038/nrurol.2017.179] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Effective management of advanced urothelial bladder cancer is challenging. New discoveries that improve our understanding of molecular bladder cancer subtypes have revealed numerous potentially targetable genomic alterations and demonstrated the efficacy of treatments that harness the immune system. These findings have begun to change paradigms of bladder cancer therapy. For example, DNA repair pathway mutations in genes such as ERCC2, FANCC, ATM, RB1, and others can predict responses to neoadjuvant platinum-based chemotherapies and to targeted therapies on the basis of mutation status. Furthermore, an increasing number of pan-cancer clinical trials (commonly referred to as basket or umbrella trials) are enrolling patients on the basis of molecular and genetic predictors of response. These studies promise to provide improved insight into the true utility of personalized medicine in the treatment of bladder cancer and many other cancer types. Finally, therapies that modulate immune responses have shown great benefit in many cancer types. Several immune checkpoint inhibitors that target programmed cell death protein 1 (PD1), its ligand PDL1, and cytotoxic T lymphocyte-associated protein 4 (CTLA4) have already been approved for use in bladder cancer, representing the most important change to the urological oncologist's tool-kit in over a decade. These advances also provide opportunities for personalization of bladder cancer therapy.
Collapse
|
578
|
Kojima R, Scheller L, Fussenegger M. Nonimmune cells equipped with T-cell-receptor-like signaling for cancer cell ablation. Nat Chem Biol 2017; 14:42-49. [PMID: 29131143 PMCID: PMC5730048 DOI: 10.1038/nchembio.2498] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 09/14/2017] [Indexed: 12/16/2022]
Abstract
The ability to engineer custom cell-contact-sensing output devices into
human non-immune cells would be useful for extending the applicability of
cell-based cancer therapies and avoiding risks associated with engineered immune
cells. Here, we have developed a new class of synthetic T-cell receptor-like
signal-transduction device that functions efficiently in human non-immune cells
and triggers release of output molecules specifically upon sensing contact with
a target cell. This device employs an interleukin signaling cascade, whose
OFF/ON switching is controlled by biophysical segregation of a transmembrane
signal-inhibitory protein from the sensor cell/target cell interface. We further
showed that designer non-immune cells equipped with this device driving
expression of a membrane-penetrator/prodrug-activating enzyme construct could
specifically kill target cells in the presence of the prodrug, indicating its
potential usefulness for target-cell-specific, cell-based enzyme-prodrug cancer
therapy. Our study also contributes to advancement of synthetic biology by
extending available design principles to transmit extracellular information to
cells.
Collapse
Affiliation(s)
- Ryosuke Kojima
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Leo Scheller
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Martin Fussenegger
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland.,Faculty of Life Science, University of Basel, Basel, Switzerland
| |
Collapse
|
579
|
Lonez C, Verma B, Hendlisz A, Aftimos P, Awada A, Van Den Neste E, Catala G, Machiels JPH, Piette F, Brayer JB, Sallman DA, Kerre T, Odunsi K, Davila ML, Gilham DE, Lehmann FF. Study protocol for THINK: a multinational open-label phase I study to assess the safety and clinical activity of multiple administrations of NKR-2 in patients with different metastatic tumour types. BMJ Open 2017; 7:e017075. [PMID: 29133316 PMCID: PMC5695348 DOI: 10.1136/bmjopen-2017-017075] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION NKR-2 are autologous T cells genetically modified to express a chimeric antigen receptor (CAR) comprising a fusion of the natural killer group 2D (NKG2D) receptor with the CD3ζ signalling domain, which associates with the adaptor molecule DNAX-activating protein of 10 kDa (DAP10) to provide co-stimulatory signal upon ligand binding. NKG2D binds eight different ligands expressed on the cell surface of many tumour cells and which are normally absent on non-neoplastic cells. In preclinical studies, NKR-2 demonstrated long-term antitumour activity towards a breadth of tumour indications, with maximum efficacy observed after multiple NKR-2 administrations. Importantly, NKR-2 targeted tumour cells and tumour neovasculature and the local tumour immunosuppressive microenvironment and this mechanism of action of NKR-2 was established in the absence of preconditioning. METHODS AND ANALYSIS This open-label phase I study will assess the safety and clinical activity of NKR-2 treatment administered three times, with a 2-week interval between each administration in different tumour types. The study will contain two consecutive segments: a dose escalation phase followed by an expansion phase. The dose escalation study involves two arms, one in solid tumours (five specific indications) and one in haematological tumours (two specific indications) and will include three dose levels in each arm: 3×108, 1×109 and 3×109 NKR-2 per injection. On the identification of the recommended dose in the first segment, based on dose-limiting toxicity occurrences, the study will expand to seven different cohorts examining the seven different tumour types separately. Clinical responses will be determined according to standard Response Evaluation Criteria In Solid Tumors (RECIST) criteria for solid tumours or international working group response criteria in haematological tumours. ETHICS APPROVAL AND DISSEMINATION Ethical approval has been obtained at all sites. Written informed consent will be taken from all participants. The results of this study will be disseminated through presentation at international scientific conferences and reported in peer-reviewed scientific journals. TRIAL REGISTRATION NUMBER NCT03018405, EudraCT 2016-003312-12; Pre-result.
Collapse
Affiliation(s)
| | | | - Alain Hendlisz
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe Aftimos
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ahmad Awada
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Eric Van Den Neste
- Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Gaetan Catala
- Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | | | - Fanny Piette
- International Drug Development Institute, Louvain-la-Neuve, Belgium
| | | | | | | | - Kunle Odunsi
- Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Marco L Davila
- H. Lee Moffitt Cancer Center, Tampa, Florida, USA
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | | | | |
Collapse
|
580
|
Bull JMC. A review of immune therapy in cancer and a question: can thermal therapy increase tumor response? Int J Hyperthermia 2017; 34:840-852. [PMID: 28974121 DOI: 10.1080/02656736.2017.1387938] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Immune therapy is a successful cancer treatment coming into its own. This is because checkpoint molecules, adoptive specific lymphocyte transfer and chimeric antigen T-cell (CAR-T) therapy are able to induce more durable responses in an increasing number of malignancies compared to chemotherapy. In addition, immune therapies are able to treat bulky disease, whereas standard cytotoxic therapies cannot treat large tumour burdens. Checkpoint inhibitor monoclonal antibodies are becoming widely used in the clinic and although more complex, adoptive lymphocyte transfer and CAR-T therapies show promise. We are learning that there are nuances to predicting the successful use of the checkpoint inhibitors as well as to specific-antigen adoptive and CAR-T therapies. We are also newly aware of a here-to-fore unrealised natural force, the status of the microbiome. However, despite better understanding of mechanisms of action of the new immune therapies, the best responses to the new immune therapies remain 20-30%. Likely the best way to improve this somewhat low response rate for patients is to increase the patient's own immune response. Thermal therapy is a way to do this. All forms of thermal therapy, from fever-range systemic thermal therapy, to high-temperature HIFU and even cryotherapy improve the immune response pre-clinically. It is time to test the immune therapies with thermal therapy in vivo to test for optimal timing of the combinations that will best enhance tumour response and then to begin to test the immune therapies with thermal therapy in the clinic as soon as possible.
Collapse
Affiliation(s)
- Joan M C Bull
- a Division of Oncology, Department of Internal Medicine , The University of Texas Medical School at Houston , Houston , TX , USA
| |
Collapse
|
581
|
Dawson NAJ, Vent-Schmidt J, Levings MK. Engineered Tolerance: Tailoring Development, Function, and Antigen-Specificity of Regulatory T Cells. Front Immunol 2017; 8:1460. [PMID: 29163527 PMCID: PMC5675854 DOI: 10.3389/fimmu.2017.01460] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/18/2017] [Indexed: 12/29/2022] Open
Abstract
Regulatory T cells (Tregs) are potent suppressors of immune responses and are currently being clinically tested for their potential to stop or control undesired immune responses in autoimmunity, hematopoietic stem cell transplantation, and solid organ transplantation. Current clinical approaches aim to boost Tregs in vivo either by using Treg-promoting small molecules/proteins and/or by adoptive transfer of expanded Tregs. However, the applicability of Treg-based immunotherapies continues to be hindered by technical limitations related to cell isolation and expansion of a pure, well-characterized, and targeted Treg product. Efforts to overcome these limitations and improve Treg-directed therapies are now under intense investigation in animal models and pre-clinical studies. Here, we review cell and protein engineering-based approaches that aim to target different aspects of Treg biology including modulation of IL-2 signaling or FOXP3 expression, and targeted antigen-specificity using transgenic T cell receptors or chimeric antigen receptors. With the world-wide interest in engineered T cell therapy, these exciting new approaches have the potential to be rapidly implemented and developed into therapies that can effectively fine-tune immune tolerance.
Collapse
Affiliation(s)
- Nicholas A J Dawson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Jens Vent-Schmidt
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
582
|
Jelinek T, Mihalyova J, Kascak M, Duras J, Hajek R. PD-1/PD-L1 inhibitors in haematological malignancies: update 2017. Immunology 2017; 152:357-371. [PMID: 28685821 PMCID: PMC5629439 DOI: 10.1111/imm.12788] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/26/2017] [Accepted: 06/30/2017] [Indexed: 12/16/2022] Open
Abstract
The introduction of PD-1/PD-L1 pathway inhibitors is an important landmark in solid oncology with unprecedented practice-changing activity in various types of solid tumours. Among haematological malignancies, PD-1/PD-L1 inhibitors have been successful, so far, only in the treatment of classical Hodgkin lymphoma, which typically exhibits an over-expression of PD-1 ligands (PD-L1, PD-L2) due to alterations in chromosome 9p24.1. Such positive outcomes led to the US Food and Drug Administration approval of nivolumab use in relapsed Hodgkin lymphoma in 2016 as the first haematological indication. Although the results in other lymphoid malignancies have not been so striking, blockade of the PD-1/PD-L1 axis has led to meaningful responses in other lymphoma types such as diffuse large B-cell lymphoma, follicular lymphoma or several T-cell lymphomas. Monotherapy with PD-1/PD-L1 inhibitors in chronic lymphocytic leukaemia and multiple myeloma has been unsatisfactory, suggesting that a combinational approach with other synergistic drugs is needed. In the case of multiple myeloma, immunomodulatory agents together with corticosteroids represent the most promising combinations. Among myeloid malignancies, the anti-PD-1 monoclonal antibodies are examined dominantly in acute myeloid leukaemia and myelodysplastic syndromes in combination with potentially synergistic hypomethylating drugs such as 5-azacitidine, resulting in promising outcomes that warrant further investigation. We have described all available clinical results of PD-1/PD-L1 inhibitors in haematological malignancies and discussed related toxicities, as well as highlighted crucial preclinical studies in this review.
Collapse
Affiliation(s)
- Tomas Jelinek
- Department of Haemato‐oncologyUniversity Hospital OstravaOstravaCzech Republic
- Faculty of MedicineUniversity of OstravaOstravaCzech Republic
- Faculty of ScienceUniversity of OstravaOstravaCzech Republic
- Centro de Investigacion Medica Aplicada (CIMA)Clinica Universidad de NavarraIDISNAPamplonaSpain
| | - Jana Mihalyova
- Department of Haemato‐oncologyUniversity Hospital OstravaOstravaCzech Republic
| | - Michal Kascak
- Department of Haemato‐oncologyUniversity Hospital OstravaOstravaCzech Republic
| | - Juraj Duras
- Department of Haemato‐oncologyUniversity Hospital OstravaOstravaCzech Republic
| | - Roman Hajek
- Department of Haemato‐oncologyUniversity Hospital OstravaOstravaCzech Republic
- Faculty of MedicineUniversity of OstravaOstravaCzech Republic
| |
Collapse
|
583
|
Chimeric Antigen Receptor (CAR) T-Cell Therapy for Thoracic Malignancies. J Thorac Oncol 2017; 13:16-26. [PMID: 29107016 DOI: 10.1016/j.jtho.2017.10.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/16/2017] [Accepted: 10/09/2017] [Indexed: 01/03/2023]
Abstract
Chimeric antigen receptor (CAR) T cells are patient T cells that are transduced with genetically engineered synthetic receptors to target a cancer cell surface antigen. The remarkable clinical response rates achieved by adoptive transfer of T cells that target CD19 in patients with leukemia and lymphoma have led to a growing number of clinical trials exploring CAR T-cell therapy for solid tumors. Herein, we review the evolution of adoptive T-cell therapy; highlight advances in CAR T-cell therapy for thoracic malignancies; and summarize the targets being investigated in clinical trials for patients with lung cancer, malignant pleural mesothelioma, and esophageal cancer. We further discuss the barriers to successfully translating CAR T-cell therapy for solid tumors and present strategies that have been investigated to overcome these hurdles.
Collapse
|
584
|
Liu B, Song Y, Liu D. Clinical trials of CAR-T cells in China. J Hematol Oncol 2017; 10:166. [PMID: 29058636 PMCID: PMC5651613 DOI: 10.1186/s13045-017-0535-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022] Open
Abstract
Novel immunotherapeutic agents targeting tumor-site microenvironment are revolutionizing cancer therapy. Chimeric antigen receptor (CAR)-engineered T cells are widely studied for cancer immunotherapy. CD19-specific CAR-T cells, tisagenlecleucel, have been recently approved for clinical application. Ongoing clinical trials are testing CAR designs directed at novel targets involved in hematological and solid malignancies. In addition to trials of single-target CAR-T cells, simultaneous and sequential CAR-T cells are being studied for clinical applications. Multi-target CAR-engineered T cells are also entering clinical trials. T cell receptor-engineered CAR-T and universal CAR-T cells represent new frontiers in CAR-T cell development. In this study, we analyzed the characteristics of CAR constructs and registered clinical trials of CAR-T cells in China and provided a quick glimpse of the landscape of CAR-T studies in China.
Collapse
Affiliation(s)
- Bingshan Liu
- School of Basic Medical Sciences and The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China.,Henan Cancer Hospital and The Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008, China
| | - Yongping Song
- Henan Cancer Hospital and The Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008, China.
| | - Delong Liu
- Henan Cancer Hospital and The Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008, China.
| |
Collapse
|
585
|
Roberts ZJ, Better M, Bot A, Roberts MR, Ribas A. Axicabtagene ciloleucel, a first-in-class CAR T cell therapy for aggressive NHL. Leuk Lymphoma 2017; 59:1785-1796. [PMID: 29058502 DOI: 10.1080/10428194.2017.1387905] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The development of clinically functional chimeric antigen receptor (CAR) T cell therapy is the culmination of multiple advances over the last three decades. Axicabtagene ciloleucel (formerly KTE-C19) is an anti-CD19 CAR T cell therapy in development for patients with refractory diffuse large B cell lymphoma (DLBCL), including transformed follicular lymphoma (TFL) and primary mediastinal B cell lymphoma (PMBCL). Axicabtagene ciloleucel is manufactured from patients' own peripheral blood mononuclear cells (PBMC) during which T cells are engineered to express a CAR that redirects them to recognize CD19-expressing cells. Clinical trials have demonstrated the feasibility of manufacturing axicabtagene ciloleucel in a centralized facility for use in multicenter clinical trials and have demonstrated potent antitumor activity in patients with refractory DLBCL. Main acute toxicities are cytokine release syndrome and neurologic events. Axicabtagene ciloleucel holds promise for the treatment of patients with CD19-positive malignancies, including refractory DLBCL.
Collapse
Affiliation(s)
| | | | | | | | - Antoni Ribas
- b Department of Medicine , University of California at Los Angeles Jonsson Comprehensive Cancer Center , Los Angeles , CA , USA
| |
Collapse
|
586
|
A versatile pretargeting approach for tumour-selective delivery and activation of TNF superfamily members. Sci Rep 2017; 7:13301. [PMID: 29038485 PMCID: PMC5643434 DOI: 10.1038/s41598-017-13530-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/25/2017] [Indexed: 11/08/2022] Open
Abstract
TNFR superfamily (TNFRSF) members have important immunoregulatory functions and are of clear interest for cancer immunotherapy. Various TNFRSF agonists have been clinically evaluated, but have met with limited efficacy and/or toxicity. Recent insights indicate that 'first-generation' TNFRSF agonists lack efficacy as they do not effectively cross-link their corresponding receptor. Reversely, ubiquitous TNFRSF receptor(s) cross-linking by CD40 and Fas agonistic antibodies resulted in dose-limiting liver toxicity. To overcome these issues, we developed a novel pretargeting strategy exploiting recombinant fusion proteins in which a soluble form of TRAIL, FasL or CD40L is genetically fused to a high-affinity anti-fluorescein scFv antibody fragment (scFvFITC). Fusion proteins scFvFITC:sTRAIL and scFvFITC:sFasL induced potent target antigen-restricted apoptosis in a panel of cancer lines and in primary patient-derived cancer cells, but only when pretargeted with a relevant FITC-labelled antitumour antibody. In a similar pretargeting setting, fusion protein scFvFITC:sCD40L promoted tumour-directed maturation of immature monocyte-derived dendritic cells (iDCs). This novel tumour-selective pretargeting approach may be used to improve efficacy and/or reduce possible off-target toxicity of TNFSF ligands for cancer immunotherapy.
Collapse
|
587
|
Horvat TZ, Seddon AN, Ogunniyi A, King AC, Buie LW, Daley RJ. The ABCs of Immunotherapy for Adult Patients With B-Cell Acute Lymphoblastic Leukemia. Ann Pharmacother 2017; 52:268-276. [PMID: 29025266 DOI: 10.1177/1060028017736539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To review the pharmacology, efficacy, and safety of Food and Drug Administration approved and promising immunotherapy agents used in the treatment of acute lymphoblastic leukemia (ALL). DATA SOURCES A literature search was performed of PubMed and MEDLINE databases (1950 to July 2017) and of abstracts from the American Society of Hematology and the American Society of Clinical Oncology. Searches were performed utilizing the following key terms: rituximab, blinatumomab, inotuzumab, ofatumumab, obinutuzumab, Blincyto, Rituxan, Gazyva, Arzerra, CAR T-cell, and chimeric antigen receptor (CAR). STUDY SELECTION/DATA EXTRACTION Studies of pharmacology, clinical efficacy, and safety of rituximab, ofatumumab, obinutuzumab, inotuzumab, blinatumomab, and CAR T-cells in the treatment of adult patients with ALL were identified. DATA SYNTHESIS Conventional chemotherapy has been the mainstay in the treatment of ALL, producing cure rates of approximately 90% in pediatrics, but it remains suboptimal in adult patients. As such, more effective consolidative modalities and novel therapies for relapsed/refractory disease are needed for adult patients with ALL. In recent years, anti-CD20 antibodies, blinatumomab, inotuzumab, and CD19-targeted CAR T-cells have drastically changed the treatment landscape of B-cell ALL. CONCLUSION Outcomes of patients with relapsed disease are improving thanks to new therapies such as blinatumomab, inotuzumab, and CAR T-cells. Although the efficacy of these therapies is impressive, they are not without toxicity, both physical and financial. The optimal sequencing of these therapies still remains a question.
Collapse
Affiliation(s)
- Troy Z Horvat
- 1 Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amanda N Seddon
- 2 Midwestern University Chicago College of Pharmacy, Downers Grove, IL, USA.,3 Rush University Medical Center, Chicago, IL, USA
| | | | - Amber C King
- 1 Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Larry W Buie
- 1 Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan J Daley
- 1 Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
588
|
Bonifant CL, Velasquez MP, Gottschalk S. Advances in immunotherapy for pediatric acute myeloid leukemia. Expert Opin Biol Ther 2017; 18:51-63. [PMID: 28945115 DOI: 10.1080/14712598.2018.1384463] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Achieving better disease control in patients diagnosed with acute myeloid leukemia (AML) has proven challenging. Overall survival has been impacted by addressing treatment related mortality with focused supportive care measures. Despite this improvement, it remains difficult to induce durable leukemia remissions despite aggressive chemotherapeutic regimens. The addition of hematopoietic stem cell transplants (HSCT) has allowed further treatment intensification and provided the benefit of graft-versus-leukemia (GVL) effect. However, HSCT carries the risk of transplant related morbidities, particularly GVHD, and anti-tumor responsiveness is still suboptimal. Thus, there is a need for alternate therapies. Immunotherapy has the potential to address this need. Areas covered: Expert opinion: The elusiveness of an ideal surface antigen target together with an immunosuppressive leukemic microenvironment add to the already difficult challenge in developing AML-targeted immunotherapies. Though many hurdles remain, recent translational discovery and progressive clinical advances anticipate exciting future developments. AREAS COVERED This review highlights promises and challenges to immune-based therapies for AML. It aims to summarize immunotherapeutic strategies trialed in AML patients to date, inclusive of: antibodies, vaccines, and cellular therapy. It emphasizes those being used in the pediatric population, but also includes adult clinical trials and translational science that may ultimately extend to pediatric patients.
Collapse
Affiliation(s)
- Challice L Bonifant
- a Department of Pediatrics and Communicable Diseases , University of Michigan , Ann Arbor , MI , USA
| | - Mireya Paulina Velasquez
- b Department of Bone Marrow Transplantation and Cellular Therapy , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Stephen Gottschalk
- b Department of Bone Marrow Transplantation and Cellular Therapy , St. Jude Children's Research Hospital , Memphis , TN , USA
| |
Collapse
|
589
|
Li S, Siriwon N, Zhang X, Yang S, Jin T, He F, Kim YJ, Mac J, Lu Z, Wang S, Han X, Wang P. Enhanced Cancer Immunotherapy by Chimeric Antigen Receptor-Modified T Cells Engineered to Secrete Checkpoint Inhibitors. Clin Cancer Res 2017; 23:6982-6992. [PMID: 28912137 DOI: 10.1158/1078-0432.ccr-17-0867] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/09/2017] [Accepted: 09/08/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Despite favorable responses of chimeric antigen receptor (CAR)-engineered T-cell therapy in patients with hematologic malignancies, the outcome has been far from satisfactory in the treatment of solid tumors, partially owing to the development of an immunosuppressive tumor microenvironment. To overcome this limitation, we engineered CAR T cells secreting checkpoint inhibitors (CPI) targeting PD-1 (CAR.αPD1-T) and evaluated their efficacy in a human lung carcinoma xenograft mouse model.Experimental Design: To evaluate the effector function and expansion capacity of CAR.αPD1-T cells in vitro, we measured the production of IFNγ and T-cell proliferation following antigen-specific stimulation. Furthermore, the antitumor efficacy of CAR.αPD1-T cells, CAR T cells, and CAR T cells combined with anti-PD-1 antibody was determined using a xenograft mouse model. Finally, the underlying mechanism was investigated by analyzing the expansion and functional capacity of TILs.Results: Human anti-PD-1 CPIs secreted by CAR.αPD1-T cells efficiently bound to PD-1 and reversed the inhibitory effect of PD-1/PD-L1 interaction on T-cell function. PD-1 blockade by continuously secreted anti-PD-1 attenuated the inhibitory T-cell signaling and enhanced T-cell expansion and effector function both in vitro and in vivo In the xenograft mouse model, we demonstrated that the secretion of anti-PD-1 enhanced the antitumor activity of CAR T cells and prolonged overall survival.Conclusions: With constitutive anti-PD-1 secretion, CAR.αPD1-T cells are more functional and expandable, and more efficient at tumor eradication than parental CAR T cells. Collectively, our study presents an important and novel strategy that enables CAR T cells to achieve better antitumor immunity, especially in the treatment of solid tumors. Clin Cancer Res; 23(22); 6982-92. ©2017 AACR.
Collapse
Affiliation(s)
- Si Li
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California
| | - Natnaree Siriwon
- Mork Family Department of Chemical Engineering and Materials Sciences, University of Southern California, Los Angeles, California
| | - Xiaoyang Zhang
- Mork Family Department of Chemical Engineering and Materials Sciences, University of Southern California, Los Angeles, California
| | - Shuai Yang
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California
| | - Tao Jin
- Technology Department, HRAIN Biotechnology Co., Ltd., Shanghai, China
| | - Feng He
- Technology Department, HRAIN Biotechnology Co., Ltd., Shanghai, China
| | - Yu Jeong Kim
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California
| | - John Mac
- Mork Family Department of Chemical Engineering and Materials Sciences, University of Southern California, Los Angeles, California
| | - Zhengfei Lu
- Department of Pathology, University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California.,Department of Biochemistry and Molecular Biology, University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California.,Department of Biological Sciences, University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California.,Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, California
| | - Sijie Wang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Xiaolu Han
- Genetic, Molecular and Cellular Biology Program, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Pin Wang
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California. .,Mork Family Department of Chemical Engineering and Materials Sciences, University of Southern California, Los Angeles, California.,Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| |
Collapse
|
590
|
Cummins KD, Gill S. Anti-CD123 chimeric antigen receptor T-cells (CART): an evolving treatment strategy for hematological malignancies, and a potential ace-in-the-hole against antigen-negative relapse. Leuk Lymphoma 2017; 59:1539-1553. [DOI: 10.1080/10428194.2017.1375107] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Katherine D. Cummins
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
591
|
Abstract
INTRODUCTION Recent breakthrough advances in Multiple Myeloma (MM) immunotherapy have been achieved with the approval of the first two monoclonal antibodies, elotuzumab and daratumumab. Adoptive cell therapy (ACT) represents yet another, maybe the most powerful modality of immunotherapy, in which allogeneic or autologous effector cells are expanded and activated ex vivo followed by their re-infusion back into patients. Infused effector cells belong to two categories: naturally occurring, non-engineered cells (donor lymphocyte infusion, myeloma infiltrating lymphocytes, deltagamma T cells) or genetically- engineered antigen-specific cells (chimeric antigen receptor T or NK cells, TCR-engineered cells). Areas covered: This review article summarizes our up-to-date knowledge on ACT in MM, its promises, and upcoming strategies to both overcome its toxicity and to integrate it into future treatment paradigms. Expert opinion: Early results of clinical studies using CAR T cells or TCR- engineered T cells in relapsed and refractory MM are particularly exciting, indicating the potential of long-term disease control or even cure. Despite several caveats including toxicity, costs and restricted availability in particular, these forms of immunotherapy are likely to once more revolutionize MM therapy.
Collapse
Affiliation(s)
- Sonia Vallet
- a Department of Internal Medicine , Karl Landsteiner University of Health Sciences, University Hospital , Krems an der Donau , Austria
| | - Martin Pecherstorfer
- a Department of Internal Medicine , Karl Landsteiner University of Health Sciences, University Hospital , Krems an der Donau , Austria
| | - Klaus Podar
- a Department of Internal Medicine , Karl Landsteiner University of Health Sciences, University Hospital , Krems an der Donau , Austria
| |
Collapse
|
592
|
Xin G, Schauder DM, Zander R, Cui W. Two is better than one: advances in pathogen-boosted immunotherapy and adoptive T-cell therapy. Immunotherapy 2017; 9:837-849. [PMID: 28877635 PMCID: PMC5941714 DOI: 10.2217/imt-2017-0055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/11/2017] [Indexed: 01/31/2023] Open
Abstract
The recent tremendous successes in clinical trials take cancer immunotherapy into a new era and have attracted major attention from both academia and industry. Among the variety of immunotherapy strategies developed to boost patients' own immune systems to fight against malignant cells, the pathogen-based and adoptive cell transfer therapies have shown the most promise for treating multiple types of cancer. Pathogen-based therapies could either break the immune tolerance to enhance the effectiveness of cancer vaccines or directly infect and kill cancer cells. Adoptive cell transfer can induce a strong durable antitumor response, with recent advances including engineering dual specificity into T cells to recognize multiple antigens and improving the metabolic fitness of transferred cells. In this review, we focus on the recent prospects in these two areas and summarize some ongoing studies that represent potential advancements for anticancer immunotherapy, including testing combinations of these two strategies.
Collapse
Affiliation(s)
- Gang Xin
- Blood Research Institute, Blood Center of Wisconsin, 8727 West Watertown Plank Road, Milwaukee, WI 53213, USA
| | - David M Schauder
- Blood Research Institute, Blood Center of Wisconsin, 8727 West Watertown Plank Road, Milwaukee, WI 53213, USA
- Department of Microbiology & Molecular Genetics, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Ryan Zander
- Blood Research Institute, Blood Center of Wisconsin, 8727 West Watertown Plank Road, Milwaukee, WI 53213, USA
| | - Weiguo Cui
- Blood Research Institute, Blood Center of Wisconsin, 8727 West Watertown Plank Road, Milwaukee, WI 53213, USA
- Department of Microbiology & Molecular Genetics, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
593
|
Chimeric antigen receptor T-cell therapy for glioblastoma. Transl Res 2017; 187:93-102. [PMID: 28755873 DOI: 10.1016/j.trsl.2017.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/25/2017] [Accepted: 07/11/2017] [Indexed: 02/06/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown great promise in the treatment of hematological disease, and its utility for treatment of solid tumors is beginning to unfold. Glioblastoma continues to portend a grim prognosis and immunotherapeutic approaches are being explored as a potential treatment strategy. Identification of appropriate glioma-associated antigens, barriers to cell delivery, and presence of an immunosuppressive microenvironment are factors that make CAR T-cell therapy for glioblastoma particularly challenging. However, insights gained from preclinical studies and ongoing clinical trials indicate that CAR T-cell therapy will continue to evolve and likely become integrated with current therapeutic strategies for malignant glioma.
Collapse
|
594
|
Bollino D, Webb TJ. Chimeric antigen receptor-engineered natural killer and natural killer T cells for cancer immunotherapy. Transl Res 2017; 187. [PMID: 28651074 PMCID: PMC5604792 DOI: 10.1016/j.trsl.2017.06.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Natural killer (NK) cells of the innate immune system and natural killer T (NKT) cells, which have roles in both the innate and adaptive responses, are unique lymphocyte subsets that have similarities in their functions and phenotypes. Both cell types can rapidly respond to the presence of tumor cells and participate in immune surveillance and antitumor immune responses. This has incited interest in the development of novel cancer therapeutics based on NK and NKT cell manipulation. Chimeric antigen receptors (CARs), generated through the fusion of an antigen-binding region of a monoclonal antibody or other ligand to intracellular signaling domains, can enhance lymphocyte targeting and activation toward diverse malignancies. Most of the CAR studies have focused on their expression in T cells; however, the functional heterogeneity of CAR T cells limits their therapeutic potential and is associated with toxicity. CAR-modified NK and NKT cells are becoming more prevalent because they provide a method to direct these cells more specifically to target cancer cells, with less risk of adverse effects. This review will outline current NK and NKT cell CAR constructs and how they compare to conventional CAR T cells, and discuss future modifications that can be explored to advance adoptive cell transfer of NK and NKT cells.
Collapse
Affiliation(s)
- Dominique Bollino
- Department of Microbiology and Immunology, University of Maryland School of Medicine and the Marlene and Stewart Greenebaum Cancer Center, Baltimore, Md
| | - Tonya J Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine and the Marlene and Stewart Greenebaum Cancer Center, Baltimore, Md.
| |
Collapse
|
595
|
Hegde UP, Mukherji B. Current status of chimeric antigen receptor engineered T cell-based and immune checkpoint blockade-based cancer immunotherapies. Cancer Immunol Immunother 2017; 66:1113-1121. [PMID: 28497159 PMCID: PMC5579167 DOI: 10.1007/s00262-017-2007-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 04/22/2017] [Indexed: 12/27/2022]
Abstract
Adoptive cell therapies with chimeric antigen receptor (CAR) engineered T cells (CAR-T) and immune checkpoint inhibition (ICI)-based cancer immunotherapies have lately shown remarkable success in certain tumor types. CAR-T cell-based therapies targeting CD19 can now induce durable remissions as well as prolong disease-free survival of patients with CD19 positive treatment refractory B cell malignancies and ICI-based therapies with humanized monoclonal antibodies against the T cell inhibitory receptors CTLA-4 and PD-1 as well as against the PD-1 ligand, PD-L1, can now achieve durable remissions as well as prolongation of life of a sizeable fraction of patients with melanoma and Hodgkin's lymphoma and non-small cell cancers. Most importantly, these immuno-therapeutic treatment modalities have raised the possibility of achieving long-term "containment" as well as "cures" for certain types of cancer. While this represents major advances in cancer immunotherapy, both modalities come with considerable toxicities, including fatalities. Although more work will be needed to bring CAR-T cell-based therapies to the bedside for most major cancers and a good deal more will be needed to make ICI-alone or in combination with other treatment modalities-work more consistently and across most major cancers, these two treatment modalities stand out as superb examples of successful translation of bench research to the bedside as well as represent real progress in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Upendra P Hegde
- University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Bijay Mukherji
- University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| |
Collapse
|
596
|
Riccardo F, Réal A, Voena C, Chiarle R, Cavallo F, Barutello G. Maternal Immunization: New Perspectives on Its Application Against Non-Infectious Related Diseases in Newborns. Vaccines (Basel) 2017; 5:E20. [PMID: 28763018 PMCID: PMC5620551 DOI: 10.3390/vaccines5030020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022] Open
Abstract
The continuous evolution in preventive medicine has anointed vaccination a versatile, human-health improving tool, which has led to a steady decline in deaths in the developing world. Maternal immunization represents an incisive step forward for the field of vaccination as it provides protection against various life-threatening diseases in pregnant women and their children. A number of studies to improve prevention rates and expand protection against the largest possible number of infections are still in progress. The complex unicity of the mother-infant interaction, both during and after pregnancy and which involves immune system cells and molecules, is an able partner in the success of maternal immunization, as intended thus far. Interestingly, new studies have shed light on the versatility of maternal immunization in protecting infants from non-infectious related diseases, such as allergy, asthma and congenital metabolic disorders. However, barely any attempt at applying maternal immunization to the prevention of childhood cancer has been made. The most promising study reported in this new field is a recent proof of concept on the efficacy of maternal immunization in protecting cancer-prone offspring against mammary tumor progression. New investigations into the possibility of exploiting maternal immunization to prevent the onset and/or progression of neuroblastoma, one of the most common childhood malignancies, are therefore justified. Maternal immunization is presented in a new guise in this review. Attention will be focused on its versatility and potential applications in preventing tumor progression in neuroblastoma-prone offspring.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Aline Réal
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino 10126, Italy.
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino 10126, Italy.
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| |
Collapse
|
597
|
Lynch A, Hawk W, Nylen E, Ober S, Autin P, Barber A. Adoptive transfer of murine T cells expressing a chimeric-PD1-Dap10 receptor as an immunotherapy for lymphoma. Immunology 2017; 152:472-483. [PMID: 28670716 DOI: 10.1111/imm.12784] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/07/2017] [Accepted: 06/21/2017] [Indexed: 12/15/2022] Open
Abstract
Adoptive transfer of T cells is a promising cancer therapy and expression of chimeric antigen receptors can enhance tumour recognition and T-cell effector functions. The programmed death protein 1 (PD1) receptor is a prospective target for a chimeric antigen receptor because PD1 ligands are expressed on many cancer types, including lymphoma. Therefore, we developed a murine chimeric PD1 receptor (chPD1) consisting of the PD1 extracellular domain fused to the cytoplasmic domain of CD3ζ. Additionally, chimeric antigen receptor therapies use various co-stimulatory domains to enhance efficacy. Hence, the inclusion of a Dap10 or CD28 co-stimulatory domain in the chPD1 receptor was compared to determine which domain induced optimal anti-tumour immunity in a mouse model of lymphoma. The chPD1 T cells secreted pro-inflammatory cytokines and lysed RMA lymphoma cells. Adoptive transfer of chPD1 T cells significantly reduced established tumours and led to tumour-free survival in lymphoma-bearing mice. When comparing chPD1 receptors containing a Dap10 or CD28 domain, both receptors induced secretion of pro-inflammatory cytokines; however, chPD1-CD28 T cells also secreted anti-inflammatory cytokines whereas chPD1-Dap10 T cells did not. Additionally, chPD1-Dap10 induced a central memory T-cell phenotype compared with chPD1-CD28, which induced an effector memory phenotype. The chPD1-Dap10 T cells also had enhanced in vivo persistence and anti-tumour efficacy compared with chPD1-CD28 T cells. Therefore, adoptive transfer of chPD1 T cells could be a novel therapy for lymphoma and inclusion of the Dap10 co-stimulatory domain in chimeric antigen receptors may induce a preferential cytokine profile and T-cell differentiation phenotype for anti-tumour therapies.
Collapse
Affiliation(s)
- Adam Lynch
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| | - William Hawk
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| | - Emily Nylen
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| | - Sean Ober
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| | - Pierre Autin
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| | - Amorette Barber
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| |
Collapse
|
598
|
Adnectin-Based Design of Chimeric Antigen Receptor for T Cell Engineering. Mol Ther 2017; 25:2466-2476. [PMID: 28784559 DOI: 10.1016/j.ymthe.2017.07.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/02/2017] [Accepted: 07/16/2017] [Indexed: 12/21/2022] Open
Abstract
Although chimeric antigen receptor (CAR)-engineered T cell therapy has achieved encouraging clinical trial results for treating hematological cancers, further optimization can likely expand this therapeutic success to more patients and other cancer types. Most CAR constructs used in clinical trials incorporate single chain variable fragment (scFv) as the extracellular antigen recognition domain. The immunogenicity of nonhuman scFv could cause host rejection against CAR T cells and compromise their persistence and efficacy. The limited availability of scFvs and slow discovery of new monoclonal antibodies also limit the development of novel CAR constructs. Adnectin, a class of affinity molecules derived from the tenth type III domain of human fibronectin, can be an alternative to scFv as an antigen-binding moiety in the design of CAR molecules. We constructed adnectin-based CARs targeting epithelial growth factor receptor (EGFR) and found that compared to scFv-based CAR, T cells engineered with adnectin-based CARs exhibited equivalent cell-killing activity against target H292 lung cancer cells in vitro and had comparable antitumor efficacy in xenograft tumor-bearing mice in vivo. In addition, with optimal affinity tuning, adnectin-based CAR showed higher selectivity on target cells with high EGFR expression than on those with low expression. This new design of adnectin CARs can potentially facilitate the development of T cell immunotherapy for cancer and other diseases.
Collapse
|
599
|
Bhattacharyya M, Madden P, Henning N, Gregory S, Aid M, Martinot AJ, Barouch DH, Penaloza-MacMaster P. Regulation of CD4 T cells and their effects on immunopathological inflammation following viral infection. Immunology 2017; 152:328-343. [PMID: 28582800 DOI: 10.1111/imm.12771] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/09/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022] Open
Abstract
CD4 T cells help immune responses, but knowledge of how memory CD4 T cells are regulated and how they regulate adaptive immune responses and induce immunopathology is limited. Using adoptive transfer of virus-specific CD4 T cells, we show that naive CD4 T cells undergo substantial expansion following infection, but can induce lethal T helper type 1-driven inflammation. In contrast, memory CD4 T cells exhibit a biased proliferation of T follicular helper cell subsets and were able to improve adaptive immune responses in the context of minimal tissue damage. Our analyses revealed that type I interferon regulates the expansion of primary CD4 T cells, but does not seem to play a critical role in regulating the expansion of secondary CD4 T cells. Strikingly, blockade of type I interferon abrogated lethal inflammation by primary CD4 T cells following viral infection, despite that this treatment increased the numbers of primary CD4 T-cell responses. Altogether, these data demonstrate important aspects of how primary and secondary CD4 T cells are regulated in vivo, and how they contribute to immune protection and immunopathology. These findings are important for rational vaccine design and for improving adoptive T-cell therapies against persistent antigens.
Collapse
Affiliation(s)
- Mitra Bhattacharyya
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Patrick Madden
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Nathan Henning
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Shana Gregory
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA
| | - Amanda J Martinot
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA.,Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
600
|
Abstract
From the application of Coley's toxin in the early 1900s to the present clinical trials using immune checkpoint regulatory inhibitors, the history of cancer immunotherapy has consisted of extremely high levels of enthusiasm after anecdotal case reports of enormous success, followed by decreasing levels of enthusiasm as the results of controlled clinical trials are available. In this review, this pattern will be documented for the various immunotherapeutic approaches over the years. The sole exception being vaccination against cancer causing viruses, which have already prevented thousands of cancers. We can only hope that the present high level of enthusiasm for the use of immune stimulation by removal of blocks to cancer immunity will be more productive than the incremental improvements using previous immunotherapies.
Collapse
Affiliation(s)
- Stewart Sell
- Wadsworth Center, New York State Department of Health and Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| |
Collapse
|