351
|
Xie YQ, Arik H, Wei L, Zheng Y, Suh H, Irvine DJ, Tang L. Redox-responsive interleukin-2 nanogel specifically and safely promotes the proliferation and memory precursor differentiation of tumor-reactive T-cells. Biomater Sci 2019; 7:1345-1357. [PMID: 30698174 PMCID: PMC6435399 DOI: 10.1039/c8bm01556b] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Interleukin-2 (IL-2) is a potent T-cell mitogen that can adjuvant anti-cancer adoptive T-cell transfer (ACT) immunotherapy by promoting T-cell engraftment. However, the clinical applications of IL-2 in combination with ACT are greatly hindered by the severe adverse effects such as vascular leak syndrome (VLS). Here, we developed a synthetic delivery strategy for IL-2 via backpacking redox-responsive IL-2/Fc nanogels (NGs) to the plasma membrane of adoptively transferred T-cells. The NGs prepared by traceless chemical cross-linking of cytokine proteins selectively released the cargos in response to T-cell receptor activation upon antigen recognition in tumors. We found that IL-2/Fc delivered by T-cell surface-bound NGs expanded transferred tumor-reactive T-cells 80-fold more than the free IL-2/Fc of an equivalent dose administered systemically and showed no effects on tumor-infiltrating regulatory T-cell expansion. Intriguingly, IL-2/Fc NG backpacks that facilitated a sustained and slow release of IL-2/Fc also promoted the CD8+ memory precursor differentiation and induced less T-cell exhaustion in vitro compared to free IL-2/Fc. The controlled responsive delivery of IL-2/Fc enabled the safe administration of repeated doses of the stimulant cytokine with no overt toxicity and improved efficacy against melanoma metastases in a mice model.
Collapse
Affiliation(s)
- Yu-Qing Xie
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, SwitzerlandCH-1015.
| | | | | | | | | | | | | |
Collapse
|
352
|
Kojima R, Fussenegger M. Synthetic Biology: Engineering Mammalian Cells To Control Cell-to-Cell Communication at Will. Chembiochem 2019; 20:994-1002. [PMID: 30589185 DOI: 10.1002/cbic.201800682] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Indexed: 12/12/2022]
Abstract
Cell-to-cell communication plays a key role in the regulation of many natural biological processes. Recent advances in mammalian synthetic biology are making it possible to rationally engineer cell-to-cell communication for therapeutic and other purposes. Here, we review state-of-the-art engineering principles to control cell-to-cell communication, focusing on communication between mammalian cells with diffusible factors (e.g., small molecules or exosomes) or direct cell contact, and on interkingdom communication between mammalian cells and bacteria. Potential applications include construction of artificial tissues able to perform complex computations, sophisticated cell-based cancer therapies, use of mammalian cells as a new class of cargo delivery modality, development of design principles to control pattern formation of cell populations, and treatment of infectious diseases. We also discuss the challenges facing practical applications, and possible enabling technologies to overcome them.
Collapse
Affiliation(s)
- Ryosuke Kojima
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland.,Faculty of Science, University of Basel, Mattenstrasse 26, 4058, Basel, Switzerland
| |
Collapse
|
353
|
Caliendo F, Dukhinova M, Siciliano V. Engineered Cell-Based Therapeutics: Synthetic Biology Meets Immunology. Front Bioeng Biotechnol 2019; 7:43. [PMID: 30937303 PMCID: PMC6431652 DOI: 10.3389/fbioe.2019.00043] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/20/2019] [Indexed: 12/30/2022] Open
Abstract
Synthetic Biology has enabled new approaches to several medical applications including the development of immunotherapies based on bioengineered cells, and most notably the engineering of T-cells with tumor-targeting receptors, the Chimeric Antigen Receptor (CAR)-T cells. CAR-T-cells have successfully treated blood tumors such as large B-cell lymphoma and promise a new scenario of therapeutic interventions also for solid tumors. Learning the lesson from CAR-T cells, we can foster the reprogramming of T lymphocytes with enhanced survival and functional activity in depressing tumor microenvironment, or to challenge diseases such as infections, autoimmune and chronic inflammatory disorders. This review will focus on the most updated bioengineering approaches to increase control, and safety of T-cell activity and to immunomodulate the extracellular microenvironment to augment immune responses. We will also discuss on applications beyond cancer treatment with implications toward the understanding and cure of a broader range of diseases by means of mammalian cells engineering.
Collapse
Affiliation(s)
- Fabio Caliendo
- Istituto Italiano di Tecnologia-IIT, Largo Barsanti e Matteucci, Naples, Italy
| | - Marina Dukhinova
- Istituto Italiano di Tecnologia-IIT, Largo Barsanti e Matteucci, Naples, Italy
| | - Velia Siciliano
- Istituto Italiano di Tecnologia-IIT, Largo Barsanti e Matteucci, Naples, Italy
- Imperial College London, South Kensington, London, United Kingdom
| |
Collapse
|
354
|
Benmebarek MR, Karches CH, Cadilha BL, Lesch S, Endres S, Kobold S. Killing Mechanisms of Chimeric Antigen Receptor (CAR) T Cells. Int J Mol Sci 2019; 20:E1283. [PMID: 30875739 PMCID: PMC6470706 DOI: 10.3390/ijms20061283] [Citation(s) in RCA: 326] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/20/2022] Open
Abstract
Effective adoptive T cell therapy (ACT) comprises the killing of cancer cells through the therapeutic use of transferred T cells. One of the main ACT approaches is chimeric antigen receptor (CAR) T cell therapy. CAR T cells mediate MHC-unrestricted tumor cell killing by enabling T cells to bind target cell surface antigens through a single-chain variable fragment (scFv) recognition domain. Upon engagement, CAR T cells form a non-classical immune synapse (IS), required for their effector function. These cells then mediate their anti-tumoral effects through the perforin and granzyme axis, the Fas and Fas ligand axis, as well as the release of cytokines to sensitize the tumor stroma. Their persistence in the host and functional outputs are tightly dependent on the receptor's individual components-scFv, spacer domain, and costimulatory domains-and how said component functions converge to augment CAR T cell performance. In this review, we bring forth the successes and limitations of CAR T cell therapy. We delve further into the current understanding of how CAR T cells are designed to function, survive, and ultimately mediate their anti-tumoral effects.
Collapse
Affiliation(s)
- Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Clara Helke Karches
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Bruno Loureiro Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| |
Collapse
|
355
|
Eisenberg V, Hoogi S, Shamul A, Barliya T, Cohen CJ. T-cells "à la CAR-T(e)" - Genetically engineering T-cell response against cancer. Adv Drug Deliv Rev 2019; 141:23-40. [PMID: 30653988 DOI: 10.1016/j.addr.2019.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/01/2019] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
The last decade will be remembered as the dawn of the immunotherapy era during which we have witnessed the approval by regulatory agencies of genetically engineered CAR T-cells and of checkpoint inhibitors for cancer treatment. Understandably, T-lymphocytes represent the essential player in these approaches. These cells can mediate impressive tumor regression in terminally-ill cancer patients. Moreover, they are amenable to genetic engineering to improve their function and specificity. In the present review, we will give an overview of the most recent developments in the field of T-cell genetic engineering including TCR-gene transfer and CAR T-cells strategies. We will also elaborate on the development of other types of genetic modifications to enhance their anti-tumor immune response such as the use of co-stimulatory chimeric receptors (CCRs) and unconventional CARs built on non-antibody molecules. Finally, we will discuss recent advances in genome editing and synthetic biology applied to T-cell engineering and comment on the next challenges ahead.
Collapse
|
356
|
A novel luciferase-based assay for the detection of Chimeric Antigen Receptors. Sci Rep 2019; 9:1957. [PMID: 30760795 PMCID: PMC6374361 DOI: 10.1038/s41598-018-38258-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/18/2018] [Indexed: 12/02/2022] Open
Abstract
Chimeric Antigen Receptor-T (CAR-T) cell immunotherapy has produced dramatic responses in hematologic malignancies. One of the challenges in the field is the lack of a simple assay for the detection of CARs on the surface of immune effector cells. In this study, we describe a novel luciferase-based assay, termed Topanga Assay, for the detection of CAR expression. The assay utilizes a recombinant fusion protein, called Topanga reagent, generated by joining the extra-cellular domain of a CAR-target in frame with one of the marine luciferases or their engineered derivatives. The assay involves incubation of CAR expressing cells with the Topanga reagent, a few washes and measurement of luminescence. The assay can detect CARs comprising either immunoglobulin- or non-immunoglobulin-based antigen binding domains. We further demonstrate that addition of epitope tags to the Topanga reagent not only allows its convenient one step purification but also extends its use for detection of CAR cells using flow cytometry. However, crude supernatant containing the secreted Topanga reagent can be directly used in both luminescence and flow-cytometry based assays without prior protein purification. Our results demonstrate that the Topanga assay is a highly sensitive, specific, convenient, economical and versatile assay for the detection of CARs.
Collapse
|
357
|
Lesne J, Chang HJ, De Visch A, Paloni M, Barthe P, Guichou JF, Mayonove P, Barducci A, Labesse G, Bonnet J, Cohen-Gonsaud M. Structural basis for chemically-induced homodimerization of a single domain antibody. Sci Rep 2019; 9:1840. [PMID: 30755682 PMCID: PMC6372657 DOI: 10.1038/s41598-019-38752-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/19/2018] [Indexed: 12/29/2022] Open
Abstract
Chemically-induced dimerization (CID) systems are essential tools to interrogate and control biological systems. AcVHH is a single domain antibody homo-dimerizing upon caffeine binding. AcVHH has a strong potential for clinical applications through caffeine-mediated in vivo control of therapeutic gene networks. Here we provide the structural basis for caffeine-induced homo-dimerization of acVHH.
Collapse
Affiliation(s)
- Jean Lesne
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Hung-Ju Chang
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Angelique De Visch
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Matteo Paloni
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Philippe Barthe
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Jean-François Guichou
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Pauline Mayonove
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Alessandro Barducci
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Gilles Labesse
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France
| | - Jerome Bonnet
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France.
| | - Martin Cohen-Gonsaud
- Centre de Biochimie Structurale, CNRS UMR5048, INSERM U1054, Université de Montpellier, 29 rue de Navacelles, 34090, Montpellier, France.
| |
Collapse
|
358
|
Holzinger A, Abken H. CAR T Cells: A Snapshot on the Growing Options to Design a CAR. Hemasphere 2019; 3:e172. [PMID: 31723811 PMCID: PMC6745938 DOI: 10.1097/hs9.0000000000000172] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/14/2018] [Indexed: 12/27/2022] Open
Abstract
Adoptive cell therapy of malignant diseases with chimeric antigen receptor (CAR) modified T cells rapidly advanced from pre-clinical models to commercial approvals within 2 decades. CARs redirect patient's T cells towards cancer cells and activate the engineered cells for a cytolytic attack resulting in the destruction of the cognate target cell. CAR T cells have demonstrated their powerful capacities in inducing complete and lasting remissions of leukemia/lymphoma in an increasing number of trials worldwide. Since the early 90's, the design of CARs went through various steps of optimization until the very recent developments which include CARs with logic gating in the recognition of antigen patterns on target cells and TRUCKs with a target recognition induced delivery of immune modulating agents. Here we review the generations in CAR design, the impact of specific modifications, the strategies to improve the safety of CAR T cell therapy, and the challenges to adapt the CAR design for broader applications.
Collapse
Affiliation(s)
- Astrid Holzinger
- RCI, Regensburg Center for Interventional Immunology, Chair for Gene-Immune Therapy, University Hospital Regensburg, Regensburg, Germany
| | - Hinrich Abken
- RCI, Regensburg Center for Interventional Immunology, Chair for Gene-Immune Therapy, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
359
|
P Teixeira A, Fussenegger M. Engineering mammalian cells for disease diagnosis and treatment. Curr Opin Biotechnol 2019; 55:87-94. [DOI: 10.1016/j.copbio.2018.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/11/2018] [Accepted: 08/17/2018] [Indexed: 12/11/2022]
|
360
|
Abstract
CAR-T CELLS BIOLOGY, CONCEPTS AND PRINCIPLES: The development of new anti-tumor immunotherapy approaches has recently dramatically increased. Progresses made in molecular biology and the development of various genetic manipulation tools allow the "reprogrammation" of T cells in order to make them express a chimeric receptor including the variable part of an immunoglobulin capable of recognizing a tumor antigen along with the expression of molecules involved in T-lymphocyte activation signaling. Genetically modified T-cells, called "CAR (chimeric antigen receptors) -T cells", have yielded impressive clinical results in the treatment of relapsed or refractory lymphoid hematological malignancies after conventional treatments and are in development in solid tumors. Different generations of CAR-T cells have been developed and technological progress makes it possible to envisage modulations of gene constructs that could further optimize the efficacy and tolerance of CAR-T cells. The first challenge of these approaches concerns the identification of specific tumor antigen targets in order to limit the on-target/off-tumor effects and the loss of expression of the target. Approaches i) targeting several antigens or ii) limiting the duration of expression of CAR in lymphocytes or iii) destroying CAR-T cells by a suicide gene. Interesting approaches are the second objective of improvement concerns the accessibility of CAR-T cells to tumor sites and the control of the immune escape mechanisms of tumor cells to the cytotoxicity of CAR-T cells. This issue is currently under the way of search of innovative strategies that should improve the clinical effectiveness of CAR-T cells, especially in solid tumors. Cet article fait partie du numéro supplément Les cellules CAR-T : une révolution thérapeutique ? réalisé avec le soutien institutionnel des partenaires Gilead : Kite et Celgene.
Collapse
|
361
|
Successful treatment of two relapsed/refractory t(8;21) acute myeloid leukemia patients by CD19-directed chimeric antigen receptor T cells. Bone Marrow Transplant 2019; 54:1138-1140. [DOI: 10.1038/s41409-018-0423-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 11/08/2022]
|
362
|
Sun W, Lee J, Zhang S, Benyshek C, Dokmeci MR, Khademhosseini A. Engineering Precision Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801039. [PMID: 30643715 PMCID: PMC6325626 DOI: 10.1002/advs.201801039] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/10/2018] [Indexed: 05/18/2023]
Abstract
Advances in genomic sequencing and bioinformatics have led to the prospect of precision medicine where therapeutics can be advised by the genetic background of individuals. For example, mapping cancer genomics has revealed numerous genes that affect the therapeutic outcome of a drug. Through materials and cell engineering, many opportunities exist for engineers to contribute to precision medicine, such as engineering biosensors for diagnosis and health status monitoring, developing smart formulations for the controlled release of drugs, programming immune cells for targeted cancer therapy, differentiating pluripotent stem cells into desired lineages, fabricating bioscaffolds that support cell growth, or constructing "organs-on-chips" that can screen the effects of drugs. Collective engineering efforts will help transform precision medicine into a more personalized and effective healthcare approach. As continuous progress is made in engineering techniques, more tools will be available to fully realize precision medicine's potential.
Collapse
Affiliation(s)
- Wujin Sun
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Junmin Lee
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Shiming Zhang
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Cole Benyshek
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Mehmet R. Dokmeci
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
- Department of RadiologyUniversity of California–Los AngelesLos AngelesCA90095USA
| | - Ali Khademhosseini
- Department of BioengineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive Therapeutics (C‐MIT)California NanoSystems InstituteUniversity of California–Los AngelesLos AngelesCA90095USA
- Department of RadiologyUniversity of California–Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California–Los Angeles10833 Le Conte AveLos AngelesCA90024USA
- Department of Chemical and Biomolecular EngineeringUniversity of California–Los AngelesLos AngelesCA90095USA
- Center of NanotechnologyDepartment of PhysicsKing Abdulaziz UniversityJeddah21569Saudi Arabia
- Department of Bioindustrial TechnologiesCollege of Animal Bioscience and TechnologyKonkuk UniversitySeoul05029Republic of Korea
| |
Collapse
|
363
|
Tokarew N, Ogonek J, Endres S, von Bergwelt-Baildon M, Kobold S. Teaching an old dog new tricks: next-generation CAR T cells. Br J Cancer 2019; 120:26-37. [PMID: 30413825 PMCID: PMC6325111 DOI: 10.1038/s41416-018-0325-1] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023] Open
Abstract
Adoptive T cell therapy (ACT) refers to the therapeutic use of T cells. T cells genetically engineered to express chimeric antigen receptors (CAR) constitute the most clinically advanced form of ACT approved to date for the treatment of CD19-positive leukaemias and lymphomas. CARs are synthetic receptors that are able to confer antigen-binding and activating functions on T cells with the aim of therapeutically targeting cancer cells. Several factors are essential for CAR T cell therapy to be effective, such as recruitment, activation, expansion and persistence of bioengineered T cells at the tumour site. Despite the advances made in CAR T cell therapy, however, most tumour entities still escape immune detection and elimination. A number of strategies counteracting these problems will need to be addressed in order to render T cell therapy effective in more situations than currently possible. Non-haematological tumours are also the subject of active investigation, but ACT has so far shown only marginal success rates in these cases. New approaches are needed to enhance the ability of ACT to target solid tumours without increasing toxicity, by improving recognition, infiltration, and persistence within tumours, as well as an enhanced resistance to the suppressive tumour microenvironment.
Collapse
Affiliation(s)
- Nicholas Tokarew
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337, Munich, Germany
| | - Justyna Ogonek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337, Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337, Munich, Germany
| | | | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337, Munich, Germany.
| |
Collapse
|
364
|
Guedan S, Calderon H, Posey AD, Maus MV. Engineering and Design of Chimeric Antigen Receptors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 12:145-156. [PMID: 30666307 PMCID: PMC6330382 DOI: 10.1016/j.omtm.2018.12.009] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
T cells engineered with chimeric antigen receptors (CARs) have emerged as a potent new class of therapeutics for cancer, based on their remarkable potency in blood cancers. Since the first clinical reports of their efficacy emerged 7 years ago, investigators have focused on the mechanisms and properties that make CARs effective or toxic, and their effects on T cell biology. Novel CAR designs coupled with improvements in gene transfer technology, incorporating advances in gene editing, have the potential to increase access to engineered cell therapies, as well as improve their potency in solid tumors.
Collapse
Affiliation(s)
- Sonia Guedan
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Hugo Calderon
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Avery D Posey
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
365
|
Yan L, Liu B. Critical factors in chimeric antigen receptor-modified T-cell (CAR-T) therapy for solid tumors. Onco Targets Ther 2018; 12:193-204. [PMID: 30636882 PMCID: PMC6309774 DOI: 10.2147/ott.s190336] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The success of chimeric antigen receptor-modified T-cell (CAR-T) therapy for B-cell lymphocyte malignancies targeting CD19 places it in a rapidly growing field in cancer immunotherapy for both hematological and solid tumors. However, the two types of tumor are quite different in the following respects. Solid tumors are characterized by complex vasculatures and matrix barriers that significantly affect T-cell functions and migration. Moreover, various immunosuppressive molecules expressed in the tumor microenvironment can impede T-cell activation, and the high metabolic rate of tumors competitively suppresses the metabolism of immune cells. All these factors will exert their influences on the development of a cancer, which is a dynamic balance between the host's immune system and the tumor. At present, solid tumors are treated primarily by surgical resection combined with radiotherapy and chemotherapy, a treatment process that is painful and not always effective. With advantages over traditional treatments, the recently developed CAR-T immunotherapy has been applied and has shown highly promising results. Nevertheless, the complexity of solid tumors presents a great challenge to this technique. This review focuses on elucidating the factors influencing the anti-tumor effects of CAR-T in the specific tumor environment, and hence exploring feasible approaches to overcome them.
Collapse
Affiliation(s)
- Lingli Yan
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou, China,
| | - Bainan Liu
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou, China,
| |
Collapse
|
366
|
Frey N, Porter D. Cytokine Release Syndrome with Chimeric Antigen Receptor T Cell Therapy. Biol Blood Marrow Transplant 2018; 25:e123-e127. [PMID: 30586620 DOI: 10.1016/j.bbmt.2018.12.756] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/17/2018] [Indexed: 12/22/2022]
Abstract
Chimeric antigen receptor (CAR)-modified T cells (CAR-Ts) targeting CD19 have resulted in unprecedented durable remissions for patients with relapsed and refractory B cell malignancies. Cytokine release syndrome (CRS), resulting from rapid immune activation induced by CAR-Ts, is the most significant treatment-related toxicity. CRS initially manifests with fever and can progress to life-threatening capillary leak with hypoxia and hypotension. The clinical signs of CRS correlate with T cell activation and high levels of cytokines including IL-6. Tocilizumab, an anti-IL-6 receptor antagonist, is the standard for CRS management, but optimal timing of administration is unclear. The development of a supportive infrastructure by treatment centers is important to maintain safe administration as access expands. Collaborative efforts are underway to harmonize the definition and grading of CRS to allow for better interpretation of toxicities across CAR-T products and clinical trials and allow for informed management algorithms.
Collapse
Affiliation(s)
- Noelle Frey
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - David Porter
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
367
|
|
368
|
Kim DH, Park S, Kim DK, Jeong MG, Noh J, Kwon Y, Zhou K, Lee NK, Ryu SH. Direct visualization of single-molecule membrane protein interactions in living cells. PLoS Biol 2018; 16:e2006660. [PMID: 30543635 PMCID: PMC6307816 DOI: 10.1371/journal.pbio.2006660] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 12/27/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022] Open
Abstract
Interactions between membrane proteins are poorly understood despite their importance in cell signaling and drug development. Here, we present a co-immunoimmobilization assay (Co-II) enabling the direct observation of membrane protein interactions in single living cells that overcomes the limitations of currently prevalent proximity-based indirect methods. Using Co-II, we investigated the transient homodimerizations of epidermal growth factor receptor (EGFR) and beta-2 adrenergic receptor (β2-AR) in living cells, revealing the differential regulation of these receptors’ dimerizations by molecular conformations and microenvironment in a plasma membrane. Co-II should provide a simple, rapid, and robust platform for visualizing both weak and strong protein interactions in the plasma membrane of living cells. Protein–protein interactions govern cellular processes. The majority of these physical interactions previously identified are strong/permanent interactions, which typically remain unbroken even after purification. The weak/transient interactions between proteins have been implicated in the control of dynamic cellular process that maintain cellular homeostasis and trigger signaling cascades upon environmental changes. However, these interactions are poorly investigated, mainly due to the methodological limitations. Here, we have developed a co-immunoimmobilization assay called Co-II that enables the direct visualization of protein–protein interactions in the membrane of living cells at the single-molecule level. Co-II is based on the intuitive concept that if the protein of interest is immobilized, the interacting protein must be co-immobilized. The use of intrinsic protein diffusivity fundamentally overcomes the limitations of proximity-based methods. Using Co-II, we study the transient homodimerizations of EGFR and β2-AR in living cells, which have been implicated in several types of cancers and heart diseases. We show that the dimerization of these receptors is differently regulated by molecular conformations and the microenvironment in the plasma membrane.
Collapse
Affiliation(s)
- Do-Hyeon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Soyeon Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dong-Kyun Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Min Gyu Jeong
- Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jungeun Noh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yonghoon Kwon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Kai Zhou
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Nam Ki Lee
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
- * E-mail: (SHR); (NKL)
| | - Sung Ho Ryu
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- * E-mail: (SHR); (NKL)
| |
Collapse
|
369
|
Hahn S, Kim MS, Choi SY, Jeong S, Jee J, Kim HK, Jeong SY, Shin H, Kim HS, Park JS, Yoo J. Leucine-rich repeat-containing G-protein coupled receptor 5 enriched organoids under chemically-defined growth conditions. Biochem Biophys Res Commun 2018; 508:430-439. [PMID: 30503340 DOI: 10.1016/j.bbrc.2018.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/02/2018] [Indexed: 12/15/2022]
Abstract
An organoid is a complex, multi-cell three-dimensional (3D) structure that contains tissue-specific cells. Epithelial stem cells, which are marked by leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5), have the potential for self-renewal and expansion as organoids. However, in the case of intestinal organoids from Lgr5-EGFP-IRES-CreERT2 transgenic mice, in vitro expansion of the Lgr5 expression is limited in a culture condition supplemented with essential proteins, such as epidermal growth factor (E), noggin (N), and R-spondin 1 (R). In this study, we hypothesized that self-renewal of Lgr5+ stem cells in a 3D culture system can be stimulated by defined compounds (CHIR99021, Valproic acid, Y-27632, and A83-01). Our results demonstrated that dissociated single cells from organoids were organized into a 3D structure in the four compounds containing the ENR culture medium in a 3D and two-dimensional (2D) culture system. Moreover, the Lgr5 expression level of organoids from the ENR- and compound-containing media increased. Furthermore, the conversion of cultured Lgr5+ stem cells from 2D to 3D was confirmed. Therefore, defined compounds promote the expansion of Lgr5+ stem cells in organoids.
Collapse
Affiliation(s)
- Soojung Hahn
- Department of Microbiology and CHA Organoid Research Center, School of Medicine, CHA University, South Korea
| | - Mi Sun Kim
- Department of Radiation Oncology, Bundang CHA Medical Center, South Korea
| | - Seon Young Choi
- Department of Microbiology and CHA Organoid Research Center, School of Medicine, CHA University, South Korea
| | - Sukin Jeong
- Department of Microbiology and CHA Organoid Research Center, School of Medicine, CHA University, South Korea
| | - JooHyun Jee
- Department of Microbiology and CHA Organoid Research Center, School of Medicine, CHA University, South Korea
| | - Han Kyung Kim
- Department of Microbiology and CHA Organoid Research Center, School of Medicine, CHA University, South Korea
| | - Sang Yun Jeong
- Department of Microbiology and CHA Organoid Research Center, School of Medicine, CHA University, South Korea
| | - Hyunsoo Shin
- Department of Radiation Oncology, Bundang CHA Medical Center, South Korea
| | - Hyoung Sun Kim
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, South Korea
| | - Joon Seong Park
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, South Korea; Department of Surgery, Gangnam Severance Hospital, South Korea
| | - Jongman Yoo
- Department of Microbiology and CHA Organoid Research Center, School of Medicine, CHA University, South Korea.
| |
Collapse
|
370
|
Hell D. Self-Adjusting Cytokine Neutralizer Cells as a Closed-Loop Delivery System of Anti-Inflammatory Biologicals. ACS Synth Biol 2018; 7:2518-2528. [PMID: 30358982 DOI: 10.1021/acssynbio.8b00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The cytokines tumor necrosis factor α (TNFα) and interleukin 1 β (IL-1β) are both strong NF-κB activators and some of the first cytokines to be released in an inflammatory process. TNFα and IL-1β are present in many autoimmune diseases, such as rheumatoid arthritis (RA). TNFα and IL-1β-blocking therapies are quite successful and established in the treatment of RA, but may also be promising in other diseases. For the treatment of recurring autoimmune diseases, strong controlled sensor-effector cells inhibiting TNFα or IL-1β appear highly predestined. Such cells detect a disease biomarker and autonomously react with the dose-dependent production of therapeutic proteins. Hence, we aim to harness and assemble the interactions of TNFα, IL-1β, and NF-κB, which are an ideal match for synthetic biology-based circuits to rewire the transmission to approved TNFα- or IL-1β-blocking biologicals. Considering the high impact of environmental influences on the dynamics of cell-based systems, we established closed-loop controllable cytokine neutralizer cells, monitoring cytokine levels and autonomously delivering powerful biologicals. This real-time processing system may provide dose-dependent drug delivery, which may be tailored for prospective cell and gene therapies against RA, and may offer a more personalized medicine than calculated drug dosing based on body weight.
Collapse
Affiliation(s)
- Dennis Hell
- University Hospital Würzburg, 97070 Würzburg, Germany
| |
Collapse
|
371
|
Rossig C, Kailayangiri S, Jamitzky S, Altvater B. Carbohydrate Targets for CAR T Cells in Solid Childhood Cancers. Front Oncol 2018; 8:513. [PMID: 30483473 PMCID: PMC6240699 DOI: 10.3389/fonc.2018.00513] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/22/2018] [Indexed: 12/23/2022] Open
Abstract
Application of the CAR targeting strategy in solid tumors is challenged by the need for adequate target antigens. As a consequence of their tissue origin, embryonal cancers can aberrantly express membrane-anchored gangliosides. These are carbohydrate molecules consisting of a glycosphingolipid linked to sialic acids residues. The best-known example is the abundant expression of ganglioside GD2 on the cell surface of neuroblastomas which derive from GD2-positive neuroectoderm. Gangliosides are involved in various cellular functions, including signal transduction, cell proliferation, differentiation, adhesion and cell death. In addition, transformation of human cells to cancer cells can be associated with distinct glycosylation profiles which provide advantages for tumor growth and dissemination and can serve as immune targets. Both gangliosides and aberrant glycosylation of proteins escape the direct molecular and proteomic screening strategies currently applied to identify further immune targets in cancers. Due to their highly restricted expression and their functional roles in the malignant behavior, they are attractive targets for immune engineering strategies. GD2-redirected CAR T cells have shown activity in clinical phase I/II trials in neuroblastoma and next-generation studies are ongoing. Further carbohydrate targets for CAR T cells in preclinical development are O-acetyl-GD2, NeuGc-GM3 (N-glycolyl GM3), GD3, SSEA-4, and oncofetal glycosylation variants. This review summarizes knowledge on the role and function of some membrane-expressed non-protein antigens, including gangliosides and abnormal protein glycosylation patterns, and discusses their potential to serve as a CAR targets in pediatric solid cancers.
Collapse
Affiliation(s)
- Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Muenster, Muenster, Germany
| | - Sareetha Kailayangiri
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Silke Jamitzky
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Bianca Altvater
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| |
Collapse
|
372
|
Schmidts A, Maus MV. Making CAR T Cells a Solid Option for Solid Tumors. Front Immunol 2018; 9:2593. [PMID: 30467505 PMCID: PMC6235951 DOI: 10.3389/fimmu.2018.02593] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/22/2018] [Indexed: 01/02/2023] Open
Abstract
Adoptive cell therapy with chimeric antigen receptor (CAR) T cells aims to redirect the patient's own immune system to selectively attack cancer cells. To do so, CAR T cells are endowed with specific antigen recognition moieties fused to signaling and costimulatory domains. While this approach has shown great success for the treatment of B cell malignancies, response rates among patients with solid cancers are less favorable. The major challenges for CAR T cell immunotherapy in solid cancers are the identification of unique tumor target antigens, as well as improving CAR T cell trafficking to and expansion at the tumor site. This review focuses on combinatorial antigen targeting, regional delivery and approaches to improve CAR T cell persistence in the face of a hostile tumor microenvironment.
Collapse
Affiliation(s)
- Andrea Schmidts
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
373
|
Morgan MA, Schambach A. Engineering CAR-T Cells for Improved Function Against Solid Tumors. Front Immunol 2018; 9:2493. [PMID: 30420856 PMCID: PMC6217729 DOI: 10.3389/fimmu.2018.02493] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/09/2018] [Indexed: 12/27/2022] Open
Abstract
Genetic engineering T cells to create clinically applied chimeric antigen receptor (CAR) T cells has led to improved patient outcomes for some forms of hematopoietic malignancies. While this has inspired the biomedical community to develop similar strategies to treat solid tumor patients, challenges such as the immunosuppressive character of the tumor microenvironment, CAR-T cell persistence and trafficking to the tumor seem to limit CAR-T cell efficacy in solid cancers. This review provides an overview of mechanisms that tumors exploit to evade eradication by CAR-T cells as well as emerging approaches that incorporate genetic engineering technologies to improve CAR-T cell activity against solid tumors.
Collapse
Affiliation(s)
- Michael A Morgan
- Hannover Medical School, Institute of Experimental Hematology, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Hannover Medical School, Institute of Experimental Hematology, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
374
|
Hu Q, Sun W, Wang J, Ruan H, Zhang X, Ye Y, Shen S, Wang C, Lu W, Cheng K, Dotti G, Zeidner JF, Wang J, Gu Z. Conjugation of haematopoietic stem cells and platelets decorated with anti-PD-1 antibodies augments anti-leukaemia efficacy. Nat Biomed Eng 2018; 2:831-840. [PMID: 31015615 DOI: 10.1038/s41551-018-0310-2] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 09/07/2018] [Indexed: 12/12/2022]
Abstract
Patients with acute myeloid leukaemia who relapse following therapy have few treatment options and face poor outcomes. Immune checkpoint inhibition, for example, by antibody-mediated programmed death-1 (PD-1) blockade, is a potent therapeutic modality that improves treatment outcomes in acute myeloid leukaemia. Here, we show that systemically delivered blood platelets decorated with anti-PD-1 antibodies (aPD-1) and conjugated to haematopoietic stem cells (HSCs) suppress the growth and recurrence of leukaemia in mice. Following intravenous injection into mice bearing leukaemia cells, the HSC-platelet-aPD-1 conjugate migrated to the bone marrow and locally released aPD-1, significantly enhancing anti-leukaemia immune responses, and increasing the number of active T cells, production of cytokines and chemokines, and survival time of the mice. This cellular conjugate also promoted resistance to re-challenge with leukaemia cells. Taking advantage of the homing capability of HSCs and in situ activation of platelets for the enhanced delivery of a checkpoint inhibitor, this cellular combination-mediated drug delivery strategy can significantly augment the therapeutic efficacy of checkpoint blockade.
Collapse
Affiliation(s)
- Quanyin Hu
- Department of Bioengineering, University of California, Los Angeles, CA, USA.,California NanoSystems Institute, University of California, Los Angeles, CA, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Wujin Sun
- Department of Bioengineering, University of California, Los Angeles, CA, USA.,California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Jinqiang Wang
- Department of Bioengineering, University of California, Los Angeles, CA, USA.,California NanoSystems Institute, University of California, Los Angeles, CA, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Huitong Ruan
- Department of Bioengineering, University of California, Los Angeles, CA, USA.,California NanoSystems Institute, University of California, Los Angeles, CA, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA.,Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
| | - Xudong Zhang
- Department of Bioengineering, University of California, Los Angeles, CA, USA.,California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Song Shen
- National Engineering Research Center for Tissue Restoration and Reconstruction, and School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, China
| | - Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
| | - Ke Cheng
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA.,Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joshua F Zeidner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jun Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, and School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, China
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA, USA. .,California NanoSystems Institute, University of California, Los Angeles, CA, USA. .,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA. .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA. .,Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, USA.
| |
Collapse
|
375
|
Jester BW, Tinberg CE, Rich MS, Baker D, Fields S. Engineered Biosensors from Dimeric Ligand-Binding Domains. ACS Synth Biol 2018; 7:2457-2467. [PMID: 30204430 DOI: 10.1021/acssynbio.8b00242] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biosensors are important components of many synthetic biology and metabolic engineering applications. Here, we report a second generation of Saccharomyces cerevisiae digoxigenin and progesterone biosensors based on destabilized dimeric ligand-binding domains that undergo ligand-induced stabilization. The biosensors, comprising one ligand-binding domain monomer fused to a DNA-binding domain and another fused to a transcriptional activation domain, activate reporter gene expression in response to steroid binding and receptor dimerization. The introduction of a destabilizing mutation to the dimer interface increased biosensor dynamic range by an order of magnitude. Computational redesign of the dimer interface and functional selections were used to create heterodimeric pairs with further improved dynamic range. A heterodimeric biosensor built from the digoxigenin and progesterone ligand-binding domains functioned as a synthetic "AND"-gate, with 20-fold stronger response to the two ligands in combination than to either one alone. We also identified mutations that increase the sensitivity or selectivity of the biosensors to chemically similar ligands. These dimerizing biosensors provide additional flexibility for the construction of logic gates and other applications.
Collapse
Affiliation(s)
- Benjamin W. Jester
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, United States
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Christine E. Tinberg
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Matthew S. Rich
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - David Baker
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, United States
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Stanley Fields
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, United States
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
- Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
376
|
Ward DE, Fay BL, Adejuwon A, Han H, Ma Z. Chimeric Antigen Receptors Based on Low Affinity Mutants of FcεRI Re-direct T Cell Specificity to Cells Expressing Membrane IgE. Front Immunol 2018; 9:2231. [PMID: 30364107 PMCID: PMC6191488 DOI: 10.3389/fimmu.2018.02231] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/07/2018] [Indexed: 12/31/2022] Open
Abstract
IgE is the key mediator of allergic responses. Omalizumab, an IgE-specific monoclonal antibody that depletes IgE, is effective for treating severe allergic asthma. The need for frequent administration of the expensive drug, however, limits its applications. Taking advantage of T cell memory, adoptive T cell therapy (ACT) targeting IgE-producing cells has the potential to achieve long-term suppression of IgE and relief of symptoms for severe allergic diseases. The transmembrane form of IgE (mIgE), which is present on all IgE-producing cells, serves as an excellent molecular target for ACT that employs chimeric antigen receptors (CARs). Here, we designed and tested CARs that use the extracellular domain of high affinity IgE receptor, FcεRIα, for mIgE recognition. When expressed on Jurkat T cells, FcεRIα-based CARs mediated robust responses in terms of CD69 upregulation to U266 myeloma cells expressing low levels of mIgE. FcεRIα-based CARs specifically recognized cells expressing mIgE, but not cells with secreted IgE captured through Fcε receptors. CAR+ Jurkat cells did not respond to LAD2 mast cells with secreted IgE bound through FcεRI or Ramos cells with secreted IgE bound through FcεRII. Co-culture of CAR+ Jurkat cells and LAD2 mast cells with IgE bound did not trigger LAD2 cell degranulation. The activity of CAR using wild type FcεRIα for mIgE binding was inhibited by the presence secreted IgE, which likely blocked CAR-mIgE interaction. The activities of CARs using low affinity mutants of FcεRIα, however, tolerated secreted IgE at relatively high concentrations. Moreover, primary human CD8+ T cells expressing a low affinity mutant CAR responded to U266 cells with INFγ production and cytotoxicity despite the presence of secreted IgE. The potency, specificity, and robustness of our CAR design, combined with repaid advances in the safety of ACT, hold promise for novel and highly effective cell-based therapies against severe allergic diseases.
Collapse
Affiliation(s)
- Dana E Ward
- Department of Biomedical Research, Nemours/A.I. duPont Hospital for Children, Wilmington, DE, United States
| | - Brittany L Fay
- Department of Biomedical Research, Nemours/A.I. duPont Hospital for Children, Wilmington, DE, United States
| | - Adebomi Adejuwon
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Huihui Han
- Department of Biomedical Research, Nemours/A.I. duPont Hospital for Children, Wilmington, DE, United States
| | - Zhengyu Ma
- Department of Biomedical Research, Nemours/A.I. duPont Hospital for Children, Wilmington, DE, United States
| |
Collapse
|
377
|
Abstract
Chemogenetic technologies enable selective pharmacological control of specific cell populations. An increasing number of approaches have been developed that modulate different signaling pathways. Selective pharmacological control over G protein-coupled receptor signaling, ion channel conductances, protein association, protein stability, and small molecule targeting allows modulation of cellular processes in distinct cell types. Here, we review these chemogenetic technologies and instances of their applications in complex tissues in vivo and ex vivo.
Collapse
Affiliation(s)
- Deniz Atasoy
- Department of Physiology, School of Medicine and Regenerative-Restorative Medicine Research Center (REMER), Istanbul Medipol University , Istanbul , Turkey ; and Janelia Research Campus, Howard Hughes Medical Institute , Ashburn, Virginia
| | - Scott M Sternson
- Department of Physiology, School of Medicine and Regenerative-Restorative Medicine Research Center (REMER), Istanbul Medipol University , Istanbul , Turkey ; and Janelia Research Campus, Howard Hughes Medical Institute , Ashburn, Virginia
| |
Collapse
|
378
|
An N, Hou YN, Zhang QX, Li T, Zhang QL, Fang C, Chen H, Lee HC, Zhao YJ, Du X. Anti-Multiple Myeloma Activity of Nanobody-Based Anti-CD38 Chimeric Antigen Receptor T Cells. Mol Pharm 2018; 15:4577-4588. [PMID: 30185037 DOI: 10.1021/acs.molpharmaceut.8b00584] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptor T cells (CAR-Ts) are a promising strategy for the treatment of many cancers, including multiple myeloma (MM), a hematological malignancy characterized by the high expression of CD38. To broaden the applications of using CD38 as a therapeutic target for the disease, we developed a new nanobody against CD38 and constructed a CD38-CAR that was composed of this nanobody as the targeting domain, and 4-1BB and CD3ζ as the costimulatory and activating domains, in a lentiviral vector. CD3+ T cells from healthy individuals were transduced with the CD38-CAR at an efficiency higher than 60%, as determined by CD38-CAR expression using flow cytometry. The CD38-CAR-Ts proliferated efficiently and produced more inflammatory cytokines, such as IL-2, IFN-γ, and TNF-α, when activated. The CD38-CAR-Ts effectively lysed CD38+ MM cell lines, including LP-1, RPMI 8226, OPM2, and MOLP8, and primary MM cells from multiple myeloma patients. The specificity was demonstrated by the fact that CD38-CAR-Ts showed little cytotoxicity on LP-1 cells with CD38 knocked out or on K562 cells, which do not express CD38. CD38-CAR-Ts appeared to have a very slight cytotoxicity against CD38+ fractions of T cells, B cells, and natural killer cells. In addition, the lysis of CD34+ hematopoietic progenitor cells did not completely inhibit the development of colony-forming units. In vivo, CD38-CAR-Ts inhibited tumor growth in NOD/SCID mice that were subcutaneously inoculated with RPMI 8226 cells. These results demonstrate that the CD38-CAR-Ts constructed with the anti-CD38 nanobody are a promising approach for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Na An
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology , Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University , Shenzhen 518035 , China
| | - Yun Nan Hou
- Laboratory of Cytophysiology, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Qiao Xia Zhang
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology , Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University , Shenzhen 518035 , China
| | - Ting Li
- Laboratory of Cytophysiology, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Qiong Li Zhang
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology , Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University , Shenzhen 518035 , China
| | - Cheng Fang
- Laboratory of Cytophysiology, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Huan Chen
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology , Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University , Shenzhen 518035 , China
| | - Hon Cheung Lee
- Laboratory of Cytophysiology, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Yong Juan Zhao
- Laboratory of Cytophysiology, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Xin Du
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology , Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University , Shenzhen 518035 , China
| |
Collapse
|
379
|
Forsberg MH, Das A, Saha K, Capitini CM. The potential of CAR T therapy for relapsed or refractory pediatric and young adult B-cell ALL. Ther Clin Risk Manag 2018; 14:1573-1584. [PMID: 30233192 PMCID: PMC6130274 DOI: 10.2147/tcrm.s146309] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Recent advancements in immunooncology have resulted in the generation of novel therapies such as chimeric antigen receptor (CAR) T cells, which have revolutionized the treatment of pediatric patients with relapsed or refractory B-cell acute lymphoblastic leukemia. The journey of tisagenlecleucel (formerly CTL019) from early preclinical success to the US Food and Drug Administration approval is summarized in this review. Strategies that are currently being investigated to improve the efficacy and safety profile of CAR T-cells are also explored, as well as the factors contributing to the present state of patient access to CAR T therapy.
Collapse
Affiliation(s)
- Matthew H Forsberg
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA,
| | - Amritava Das
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA,
- Morgridge Institute for Research, University of Wisconsin, Madison, WI, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA,
- Department of Medical History & Bioethics, University of Wisconsin, Madison, WI, USA,
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA,
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA,
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, USA,
| |
Collapse
|
380
|
Rotolo A, Karadimitris A, Ruella M. Building upon the success of CART19: chimeric antigen receptor T cells for hematologic malignancies. Leuk Lymphoma 2018; 59:2040-2055. [PMID: 29165008 PMCID: PMC6814196 DOI: 10.1080/10428194.2017.1403024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chimeric antigen receptor T cell (CART) therapy has dramatically changed the therapeutic prospects for B cell malignancies. Over the last decade CD19-redirected CART have demonstrated the ability to induce deep, long-lasting remissions and possibly cure patients with relapsing B cell neoplasms. Such impressive results with CART19 fostered efforts to expand this technology to other incurable malignancies that naturally do not express CD19, such as acute myeloid leukemia (AML), Hodgkin lymphoma (HL) and multiple myeloma (MM). However, to reach this goal, several hurdles have to be overcome, in particular: (i) the apparent lack of suitable targets as effective as CD19; (ii) the immunosuppressive tumor microenvironment; (iii) intra-tumoral heterogeneity and antigen-negative relapses. Therefore, new strategies that allow safer and more potent CART platforms are under development and may provide grounds for new exciting breakthroughs in the field.
Collapse
Affiliation(s)
- Antonia Rotolo
- Centre for Haematology, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Anastasios Karadimitris
- Centre for Haematology, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
381
|
Si W, Li C, Wei P. Synthetic immunology: T-cell engineering and adoptive immunotherapy. Synth Syst Biotechnol 2018; 3:179-185. [PMID: 30345403 PMCID: PMC6190530 DOI: 10.1016/j.synbio.2018.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/28/2018] [Accepted: 08/13/2018] [Indexed: 12/24/2022] Open
Abstract
During the past decades, the rapidly-evolving cancer is hard to be thoroughly eliminated even though the radiotherapy and chemotherapy do exhibit efficacy in some degree. However, a breakthrough appeared when the adoptive cancer therapy [1] was developed, especially T cells armed with chimeric antigen receptors (CARs) showed great potential in tumor clinical trials recently. CAR-T cells successfully elevated the efficiency and specificity of cytotoxicity. In this review, we will talk about the design of CAR and CAR-included combinatory therapeutic applications in the principles of systems and synthetic immunology.
Collapse
Affiliation(s)
- Wen Si
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Cheng Li
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ping Wei
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
382
|
Ajina A, Maher J. Strategies to Address Chimeric Antigen Receptor Tonic Signaling. Mol Cancer Ther 2018; 17:1795-1815. [PMID: 30181329 PMCID: PMC6130819 DOI: 10.1158/1535-7163.mct-17-1097] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/19/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022]
Abstract
Adoptive cell transfer using chimeric antigen receptors (CAR) has emerged as one of the most promising new therapeutic modalities for patients with relapsed or refractory B-cell malignancies. Thus far, results in patients with advanced solid tumors have proven disappointing. Constitutive tonic signaling in the absence of ligand is an increasingly recognized complication when deploying these synthetic fusion receptors and can be a cause of poor antitumor efficacy, impaired survival, and reduced persistence in vivo In parallel, ligand-dependent tonic signaling can mediate toxicity and promote T-cell anergy, exhaustion, and activation-induced cell death. Here, we review the mechanisms underpinning CAR tonic signaling and highlight the wide variety of effects that can emerge after making subtle structural changes or altering the methodology of CAR transduction. We highlight strategies to prevent unconstrained tonic signaling and address its deleterious consequences. We also frame this phenomenon in the context of endogenous TCR tonic signaling, which has been shown to regulate peripheral tolerance, facilitate the targeting of foreign antigens, and suggest opportunities to coopt ligand-dependent CAR tonic signaling to facilitate in vivo persistence and efficacy. Mol Cancer Ther; 17(9); 1795-815. ©2018 AACR.
Collapse
MESH Headings
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Adam Ajina
- CAR Mechanics Group, King's College London, London, United Kingdom.
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
| | - John Maher
- CAR Mechanics Group, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Department of Immunology, Eastbourne Hospital, East Sussex, United Kingdom
| |
Collapse
|
383
|
Srivastava S, Riddell SR. Chimeric Antigen Receptor T Cell Therapy: Challenges to Bench-to-Bedside Efficacy. THE JOURNAL OF IMMUNOLOGY 2018; 200:459-468. [PMID: 29311388 DOI: 10.4049/jimmunol.1701155] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/29/2017] [Indexed: 12/23/2022]
Abstract
Immunotherapy with T cells genetically modified to express chimeric Ag receptors (CARs) that target tumor-associated molecules have impressive efficacy in hematological malignancies. The field has now embraced the challenge of applying this approach to treat common epithelial malignancies, which make up the majority of cancer cases but evade immunologic attack by a variety of subversive mechanisms. In this study, we review the principles that have guided CAR T cell design and the extraordinary clinical results being achieved in B cell malignancies targeting CD19 with a single infusion of engineered T cells. This success has raised expectations that CAR T cells can be applied to solid tumors, but numerous obstacles must be overcome to achieve the success observed in hematologic cancers. Potential solutions driven by advances in genetic engineering, synthetic biology, T cell biology, and improved tumor models that recapitulate the obstacles in human tumors are discussed.
Collapse
Affiliation(s)
- Shivani Srivastava
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Stanley R Riddell
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| |
Collapse
|
384
|
Barros L, Pretti MA, Chicaybam L, Abdo L, Boroni M, Bonamino MH. Immunological-based approaches for cancer therapy. Clinics (Sao Paulo) 2018; 73:e429s. [PMID: 30133560 PMCID: PMC6097086 DOI: 10.6061/clinics/2018/e429s] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/05/2018] [Indexed: 02/06/2023] Open
Abstract
The immunologic landscape of tumors has been continuously unveiled, providing a new look at the interactions between cancer cells and the immune system. Emerging tumor cells are constantly eliminated by the immune system, but some cells establish a long-term equilibrium phase leading to tumor immunoediting and, eventually, evasion. During this process, tumor cells tend to acquire more mutations. Bearing a high mutation burden leads to a greater number of neoantigens with the potential to initiate an immune response. Although many tumors evoke an immune response, tumor clearance by the immune system does not occur due to a suppressive tumor microenvironment. The mechanisms by which tumors achieve the ability to evade immunologic control vary. Understanding these differences is crucial for the improvement and application of new immune-based therapies. Much effort has been placed in developing in silico algorithms to predict tumor immunogenicity and to characterize the microenvironment via high-throughput sequencing and gene expression techniques. Each sequencing source, transcriptomics, and genomics yields a distinct level of data, helping to elucidate the tumor-based immune responses and guiding the fine-tuning of current and upcoming immune-based therapies. In this review, we explore some of the immunological concepts behind the new immunotherapies and the bioinformatic tools to study the immunological aspects of tumors, focusing on neoantigen determination and microenvironment deconvolution. We further discuss the immune-based therapies already in clinical use, those underway for future clinical application, the next steps in immunotherapy, and how the characterization of the tumor immune contexture can impact therapies aiming to promote or unleash immune-based tumor elimination.
Collapse
Affiliation(s)
- Luciana Barros
- Programa de Carcinogenese Molecular, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
| | - Marco Antonio Pretti
- Programa de Carcinogenese Molecular, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
| | | | - Luiza Abdo
- Programa de Carcinogenese Molecular, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
| | - Mariana Boroni
- Laboratorio de Bioinformatica e Biologia Computacional, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
| | - Martin Hernán Bonamino
- Laboratorio de Bioinformatica e Biologia Computacional, Coordenacao de Pesquisa, Instituto Nacional de Cancer (INCA), Rio de Janeiro, RJ, BR
- *Corresponding author. E-mail: /
| |
Collapse
|
385
|
Pantuck M, Palaskas N, Drakaki A. Next generation T-cell therapy for genitourinary malignancies, part B: Overcoming obstacles and future strategies for success. Cancer Treat Res Commun 2018; 17:1-7. [PMID: 30170288 DOI: 10.1016/j.ctarc.2018.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/14/2018] [Indexed: 02/06/2023]
Affiliation(s)
| | - Nicolaos Palaskas
- Division of Hematology and Oncology, David Geffen School of Medicine, UCLA, Los Angeles, USA.
| | - Alexandra Drakaki
- Division of Hematology and Oncology, David Geffen School of Medicine, UCLA, Los Angeles, USA.
| |
Collapse
|
386
|
Jin MY, Han Y, Liu YJ, Gu B, Chen J, Shi SS, Xu MZ, Cai CS, Tang YQ, Wu DP. [Treatment of central nervous system leukemia with CD19-chimeric antigen receptor T-cell immunotherapy: two cases report and literature review]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:650-653. [PMID: 30180466 PMCID: PMC7342845 DOI: 10.3760/cma.j.issn.0253-2727.2018.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Indexed: 11/09/2022]
Abstract
Objective: To explore the efficacy and safety of chimeric antigen receptor T (CAR-T) cells in the treatment of central nervous system leukemia (CNSL). Methods: Two leukemia patients with CNSL were treated with CD19-CAR-T cells. The process and results of the entire treatment is reported and related literature review is conducted. Results: The patients were diagnosed as acute myeloid leukemia (AML)-M(2) with B lymphoid antigen expression and B cell acute lymphoblastic leukemia(B-ALL) by morphology and immunophenotype assay. The immunophenotype was consistent with the abnormal manifestations of AML-M(2) and B-ALL. Their clinical manifestations and laboratory tests met the diagnostic criteria of CNSL. The diagnosis was clear and the two patients were treated with CD19-CAR-T cell immunotherapy. Central nervous system symptoms were relieved. The imaging abnormalities of patient one has disappeared but cytokines release syndrome (CRS) occurred during the treatment. Cerebrospinal fluid of patient two was negative and no obvious CRS reaction was found. Conclusions: CAR-T cell immunotherapy is likely to induce the remission of CNSL and improve the prognosis.
Collapse
Affiliation(s)
- M Y Jin
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Collaborative Innovation Centre of Hematology, Institute of Hematopoietic Stem Cell Transplantation, Key Laboratory of Thrombosis and Haemostasis of Ministry of Health, Suzhou 215006, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
387
|
Zhang E, Yang P, Gu J, Wu H, Chi X, Liu C, Wang Y, Xue J, Qi W, Sun Q, Zhang S, Hu J, Xu H. Recombination of a dual-CAR-modified T lymphocyte to accurately eliminate pancreatic malignancy. J Hematol Oncol 2018; 11:102. [PMID: 30103775 PMCID: PMC6090669 DOI: 10.1186/s13045-018-0646-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/02/2018] [Indexed: 11/08/2022] Open
Abstract
Background The therapeutic application of T cells endowing with chimeric antigen receptors (CARs) is faced with “on-target, off-tumor” toxicity against solid tumors, particularly in the treatment of the pancreatic cancer. To our best knowledge, the pancreatic cancer cell line AsPC-1 often highly expressed some distinct tumor-associated antigens, such as carcino-embryonic antigen (CEA) and mesothelin (MSLN). Therefore, in this research, we have characterized dual-receptor CAR-modified T cells (dCAR-T) that exert effective and safe cytotoxicity against AsPC-1 cells. Methods Based on the dual signaling pathway of wild T cells, we designed a novel dCAR diagram specific for CEA and MSLN, which achieved comparable activity relative to that of conventional CAR-T cells (CEA-CAR T or MSLN-CAR T). In this dCAR, a tandem construct containing two physically separate structures, CEA-CD3ζ and MSLN-4/1BB signaling domains were effectively controlled with tumor antigens CEA and MSLN, respectively. Finally, the activity of dCAR-T cells has been verified via in vitro and in vivo experiments. Results In the presence of cognate tumor cells (AsPC-1) expressing both CEA and MSLN, dCAR-T cells exerted high anti-tumor activity relative to that of other single-receptor CAR-T cells bearing only one signaling pathway (e.g., Cζ-CAR and MBB-CAR). In a xenograft model, dCAR-T cells significantly inhibited the growth of AsPC-1 cells yet no effect on the growth of non-cognate tumor cells. Furthermore, the released cytokines and T cell persistence in mice were comparable with that of conventional CAR-T cells, obtaining specific and controllable cytotoxicity. Conclusions A novel type of CAR-T cells, termed dCAR-T, was designed with specific activities, that is, significant cytotoxicity for two antigen-positive tumor cells yet no cytotoxicity for single antigen-positive tumor cells. Dual-targeted CAR-T cells can be precisely localized at the tumor site and can exert high cytotoxicity against tumor cells, alleviating “on-target, off-tumor” toxicity and enabling accurate application of CAR-T cell therapy. Electronic supplementary material The online version of this article (10.1186/s13045-018-0646-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erhao Zhang
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,Basic Medical Research Center, School of Medicine, Nantong University, Nantong, 226001, People's Republic of China
| | - Peiwei Yang
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Jieyi Gu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Heming Wu
- Jiangsu Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Xiaowei Chi
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Chen Liu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Ying Wang
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Jianpeng Xue
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Weiyan Qi
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Qingbo Sun
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Shengnan Zhang
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Jialiang Hu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China. .,State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Hanmei Xu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, 210009, People's Republic of China. .,State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, People's Republic of China. .,Nanjing Anji Biotechnology Co., Ltd, Nanjing, 210046, People's Republic of China.
| |
Collapse
|
388
|
Cho SF, Anderson KC, Tai YT. Targeting B Cell Maturation Antigen (BCMA) in Multiple Myeloma: Potential Uses of BCMA-Based Immunotherapy. Front Immunol 2018; 9:1821. [PMID: 30147690 PMCID: PMC6095983 DOI: 10.3389/fimmu.2018.01821] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/24/2018] [Indexed: 01/10/2023] Open
Abstract
The approval of the first two monoclonal antibodies targeting CD38 (daratumumab) and SLAMF7 (elotuzumab) in late 2015 for treating relapsed and refractory multiple myeloma (RRMM) was a critical advance for immunotherapies for multiple myeloma (MM). Importantly, the outcome of patients continues to improve with the incorporation of this new class of agents with current MM therapies. However, both antigens are also expressed on other normal tissues including hematopoietic lineages and immune effector cells, which may limit their long-term clinical use. B cell maturation antigen (BCMA), a transmembrane glycoprotein in the tumor necrosis factor receptor superfamily 17 (TNFRSF17), is expressed at significantly higher levels in all patient MM cells but not on other normal tissues except normal plasma cells. Importantly, it is an antigen targeted by chimeric antigen receptor (CAR) T-cells, which have already shown significant clinical activities in patients with RRMM who have undergone at least three prior treatments, including a proteasome inhibitor and an immunomodulatory agent. Moreover, the first anti-BCMA antibody–drug conjugate also has achieved significant clinical responses in patients who failed at least three prior lines of therapy, including an anti-CD38 antibody, a proteasome inhibitor, and an immunomodulatory agent. Both BCMA targeting immunotherapies were granted breakthrough status for patients with RRMM by FDA in Nov 2017. Other promising BCMA-based immunotherapeutic macromolecules including bispecific T-cell engagers, bispecific molecules, bispecific or trispecific antibodies, as well as improved forms of next generation CAR T cells, also demonstrate high anti-MM activity in preclinical and even early clinical studies. Here, we focus on the biology of this promising MM target antigen and then highlight preclinical and clinical data of current BCMA-targeted immunotherapies with various mechanisms of action. These crucial studies will enhance selective anti-MM response, transform the treatment paradigm, and extend disease-free survival in MM.
Collapse
Affiliation(s)
- Shih-Feng Cho
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| |
Collapse
|
389
|
Caruso HG, Heimberger AB, Cooper LJN. Steering CAR T cells to distinguish friend from foe. Oncoimmunology 2018; 8:e1271857. [PMID: 31646067 PMCID: PMC6791456 DOI: 10.1080/2162402x.2016.1271857] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 02/03/2023] Open
Abstract
CD19-specific chimeric antigen receptor (CAR)+ T cells have demonstrated clinical efficacy and long-lasting remissions, concomitant with tolerable normal B-cell aplasia. However, many tumor-associated antigens (TAAs) are expressed on normal tissues, the destruction of which would lead to intolerable toxicity. Thus, there is a need to engineer CAR+ T cells with improved safety profiles to restrict toxicity against TAA-expressing normal tissues. Bioengineering approaches include: (i) targeting CAR+ T cells to the tumor site, (ii) limiting CAR+ T-cell persistence, and (iii) restricting CAR activation. We review and evaluate strategies to engineer CAR+ T cells to reduce the potential of on-target, off-tissue toxicity.
Collapse
Affiliation(s)
- Hillary G Caruso
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy B Heimberger
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laurence J N Cooper
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Ziopharm Oncology, Boston, MA, USA
| |
Collapse
|
390
|
Chen Y, E CY, Gong ZW, Liu S, Wang ZX, Yang YS, Zhang XW. Chimeric antigen receptor-engineered T-cell therapy for liver cancer. Hepatobiliary Pancreat Dis Int 2018; 17:301-309. [PMID: 29861325 DOI: 10.1016/j.hbpd.2018.05.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/09/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Chimeric antigen receptor-engineered T-cell (CAR-T) therapy is a newly developed immunotherapy used in the treatment of cancers. Because CAR-T therapy has shown great success in treating CD19-positive hematological malignancies, its application has been explored in the treatment of solid tumors, such as liver cancer. In this review, we discuss the immune characteristics of liver cancer, the obstacles encountered during the application of CAR-T therapy, and preclinical and clinical progress in the use of CAR-T therapy in patients with liver cancer. DATA SOURCES The data on CAR-T therapy related to liver cancers were collected by searching PubMed and the Web of Science databases prior to December 2017 with the keywords "chimeric antigen receptor", "CAR-T", "liver cancer", "hepatocellular carcinoma", and "solid tumor". Additional articles were identified by manual search of references found in the primary articles. The data for clinical trials were collected by searching ClinicalTrials.gov. RESULTS The liver has a tolerogenic nature in the intrahepatic milieu and its tumor microenvironment significantly affects tumor progression. The obstacles that reduce the efficacy of CAR-T therapy in solid tumors include a lack of specific tumor antigens, limited trafficking and penetration of CAR-T cells to tumor sites, and an immunosuppressive tumor microenvironment. To overcome these obstacles, several strategies have emerged. In addition, several strategies have been developed to manage the side effects of CAR-T, including enhancing the selectivity of CARs and controlling CAR-T activity. To date, no clinical trials of CAR-T therapy against HCC have been completed. However, preclinical studies in vitro and in vivo have shown potent antitumor efficacy. Glypican-3, mucin-1, epithelial cell adhesion molecule, carcinoembryonic antigen, and other targets are currently being studied. CONCLUSIONS The application of CAR-T therapy for liver cancer is just beginning to be explored and more research is needed. However, we are optimistic that CAR-T therapy will offer a new approach for the treatment of liver cancers in the future.
Collapse
Affiliation(s)
- Yang Chen
- Department of Hepatobiliary and Pancreas Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Chang-Yong E
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130021, China
| | - Zhi-Wen Gong
- Department of Hepatobiliary and Pancreas Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Shui Liu
- Department of Hepatobiliary and Pancreas Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Zhen-Xiao Wang
- Department of Hepatobiliary and Pancreas Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Yong-Sheng Yang
- Department of Hepatobiliary and Pancreas Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Xue-Wen Zhang
- Department of Hepatobiliary and Pancreas Surgery, the Second Hospital of Jilin University, Changchun 130041, China.
| |
Collapse
|
391
|
Hickey JW, Kosmides AK, Schneck JP. Engineering Platforms for T Cell Modulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:277-362. [PMID: 30262034 DOI: 10.1016/bs.ircmb.2018.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cells are crucial contributors to mounting an effective immune response and increasingly the focus of therapeutic interventions in cancer, infectious disease, and autoimmunity. Translation of current T cell immunotherapies has been hindered by off-target toxicities, limited efficacy, biological variability, and high costs. As T cell therapeutics continue to develop, the application of engineering concepts to control their delivery and presentation will be critical for their success. Here, we outline the engineer's toolbox and contextualize it with the biology of T cells. We focus on the design principles of T cell modulation platforms regarding size, shape, material, and ligand choice. Furthermore, we review how application of these design principles has already impacted T cell immunotherapies and our understanding of T cell biology. Recent, salient examples from protein engineering, synthetic particles, cellular and genetic engineering, and scaffolds and surfaces are provided to reinforce the importance of design considerations. Our aim is to provide a guide for immunologists, engineers, clinicians, and the pharmaceutical sector for the design of T cell-targeting platforms.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan P Schneck
- Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
392
|
Petersburg J, Shen J, Csizmar CM, Murphy KA, Spanier J, Gabrielse K, Griffith TS, Fife B, Wagner CR. Eradication of Established Tumors by Chemically Self-Assembled Nanoring Labeled T Cells. ACS NANO 2018; 12:6563-6576. [PMID: 29792808 PMCID: PMC6506352 DOI: 10.1021/acsnano.8b01308] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Our laboratory has developed chemically self-assembled nanorings (CSANs) as prosthetic antigen receptors (PARs) for the nongenetic modification of T cell surfaces. PARs have been successfully employed in vitro to activate T cells for the selective killing of leukemia cells. However, PAR efficacy has yet to be evaluated in vivo or against solid tumors. Therefore, we developed bispecific PARs that selectively target the human CD3 receptor and human epithelial cell adhesion molecule (EpCAM), which is overexpressed on multiple carcinomas and cancer stem cells. The αEpCAM/αCD3 PARs were found to stably bind T cells for >4 days, and treating EpCAM+ MCF-7 breast cancer cells with αEpCAM/αCD3 PAR-functionalized T cells resulted in the induction of IL-2, IFN-γ, and MCF-7 cytotoxicity. Furthermore, an orthotopic breast cancer model validated the ability of αEpCAM/αCD3 PAR therapy to direct T cell lytic activity toward EpCAM+ breast cancer cells in vivo, leading to tumor eradication. In vivo biodistribution studies demonstrated that PAR-T cells were formed in vivo and persist for over 48 h with rapid accumulation in tumor tissue. Following PAR treatment, the production of IL-2, IFN-γ, IL-6, and TNF-α could be significantly reduced by an infusion of clinically relevant concentrations of the FDA-approved antibiotic, trimethoprim, signaling pharmacologic PAR deactivation. Importantly, CSANs did not induce naïve T cell activation and thus exhibit a limited potential to induce naïve T cell anergy. In addition, murine immunogenicity studies demonstrated that CSANs do not induce a significant antibody response nor do they activate splenic cells. Collectively, our results demonstrate that bispecific CSANs are able to nongenetically generate reversibly modified T cells that are capable of eradicating targeted solid tumors.
Collapse
Affiliation(s)
- Jacob Petersburg
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Jingjing Shen
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Clifford M Csizmar
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Katherine A Murphy
- Department of Urology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Justin Spanier
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Kari Gabrielse
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Brian Fife
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Carston R. Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Address correspondence to: , University of Minnesota, Department of Medicinal Chemistry, 2231 6th Street S.E., Cancer & Cardiovascular Research Building, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
393
|
Zhao W, Nguyen H, Zeng G, Gao D, Yan H, Liang FS. A chemically induced proximity system engineered from the plant auxin signaling pathway. Chem Sci 2018; 9:5822-5827. [PMID: 30079194 PMCID: PMC6050582 DOI: 10.1039/c8sc02353k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 06/09/2018] [Indexed: 12/16/2022] Open
Abstract
Methods based on chemically induced proximity (CIP) serve as powerful tools to control cellular processes in a temporally specific manner. To expand the repertoire of CIP systems available for studies of cellular processes, we engineered the plant auxin signaling pathway to create a new indole-3-acetic acid (IAA) based CIP method. Auxin-induced protein degradation that occurs in the natural pathway was eliminated in the system. The new IAA based method is both readily inducible and reversible, and used to control the production of therapeutic proteins that induced the apoptosis of cancer cells. The approach is also orthogonal to existing CIP systems and used to construct a biological Boolean logic gate controlling gene expression system. We believe that the new CIP method will be applicable to the artificial control and dissection of complex cellular functions.
Collapse
Affiliation(s)
- Weiye Zhao
- Department of Chemistry and Chemical Biology , University of New Mexico , 300 Terrace Street NE , Albuquerque , New Mexico 87131 , USA .
| | - Huong Nguyen
- Department of Chemistry and Chemical Biology , University of New Mexico , 300 Terrace Street NE , Albuquerque , New Mexico 87131 , USA .
| | - Guihua Zeng
- Department of Chemistry and Chemical Biology , University of New Mexico , 300 Terrace Street NE , Albuquerque , New Mexico 87131 , USA .
| | - Dan Gao
- Department of Chemistry and Chemical Biology , University of New Mexico , 300 Terrace Street NE , Albuquerque , New Mexico 87131 , USA .
| | - Hao Yan
- Department of Chemistry and Chemical Biology , University of New Mexico , 300 Terrace Street NE , Albuquerque , New Mexico 87131 , USA .
| | - Fu-Sen Liang
- Department of Chemistry and Chemical Biology , University of New Mexico , 300 Terrace Street NE , Albuquerque , New Mexico 87131 , USA .
| |
Collapse
|
394
|
Zhai Y, Ran W, Su J, Lang T, Meng J, Wang G, Zhang P, Li Y. Traceable Bioinspired Nanoparticle for the Treatment of Metastatic Breast Cancer via NIR-Trigged Intracellular Delivery of Methylene Blue and Cisplatin. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1802378. [PMID: 29989211 DOI: 10.1002/adma.201802378] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/04/2018] [Indexed: 05/18/2023]
Abstract
Cytotoxic T lymphocyte (CTL) eliminates abnormal cells through target recognition-triggered intracellular toxin delivery. Chimeric antigen receptor T-cell improves cancer cell recognition of CTL, but its effectiveness and safety in solid tumor treatment are still hampered by poor tumor infiltration, suppressive tumor microenvironment, and severe on-target off-tumor toxicity. Given the functionality and challenges of CTL in cancer therapy, herein, a CTL-inspired nanovesicle (MPV) with a cell membrane-derived shell and a methylene blue (MB) and cisplatin (Pt) loaded gelatin nanogel core is created. The MPV generates contrast for tumor photoacoustic imaging, and produces hyperthermia upon laser irradiation, enabling photothermal imaging and deep tumor penetration. Meanwhile, it releases MB and Pt, and then delivers them into the cytosol of cancer cells, which process can be visualized by imaging the recovery of MB-derived fluorescence. The localized hyperthermia, photodynamic therapy, and chemotherapy together kill 4T1 breast cancer cells effectively, resulting in primary tumor regression and 97% inhibition of pulmonary metastasis, without significant toxicity to the animals. Taken together, the MPV shows tumor-specific and stimuli-triggered intracellular toxin delivery with advantages in traceable accumulation and activation, high tumor penetration, and triple combination therapy, and thus can be an effective nanomedicine for combating metastatic breast cancer.
Collapse
Affiliation(s)
- Yihui Zhai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Wei Ran
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Jinghan Su
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Tianqun Lang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Jia Meng
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Guanru Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
- School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai, 264005, China
| |
Collapse
|
395
|
Manipulating the tumor microenvironment by adoptive cell transfer of CAR T-cells. Mamm Genome 2018; 29:739-756. [PMID: 29987406 DOI: 10.1007/s00335-018-9756-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/28/2018] [Indexed: 12/14/2022]
Abstract
T-cells expressing synthetic chimeric antigen receptors (CARs) have revolutionized immuno-oncology and highlighted the use of adoptive cell transfer, for the treatment of cancer. The phenomenal clinical success obtained in the treatment of hematological malignancies with CAR T-cells has not been reproduced in the treatment of solid tumors, mainly due to the suppressive and hostile tumor microenvironment (TME). This review will address the immunosuppressive features of the TME, which include the stroma, cytokine and chemokine milieu, suppressive regulatory cells and hypoxic conditions, which can all pose formidable barriers for the effective anti-tumor function of CAR T-cells. Some of the novel next generation CARs that have been developed and tested against the TME, will be discussed, to highlight the status of current research in CAR T-cell therapy for solid tumors.
Collapse
|
396
|
Tang L, Zheng Y, Melo MB, Mabardi L, Castaño AP, Xie YQ, Li N, Kudchodkar SB, Wong HC, Jeng EK, Maus MV, Irvine DJ. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat Biotechnol 2018; 36:707-716. [PMID: 29985479 PMCID: PMC6078803 DOI: 10.1038/nbt.4181] [Citation(s) in RCA: 466] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 06/07/2018] [Indexed: 12/24/2022]
Abstract
Adoptive cell therapy (ACT) with antigen-specific T cells has shown remarkable clinical success, but approaches to safely and effectively augment T cell function, especially in solid tumors, remain of great interest. Here we describe a strategy to “backpack” large quantities of supporting protein drugs on T cells using protein nanogels (NGs) that selectively release these cargos in response to T cell receptor (TCR) activation. We design cell surface-conjugated NGs that respond to an increase in T cell surface reduction potential upon antigen recognition, limiting drug release to sites of antigen encounter such as the tumor microenvironment. Using NGs carrying an IL-15 superagonist complex, we demonstrate that relative to systemic administration of free cytokines, NG delivery selectively expands T cells 16-fold in tumors, and allows at least 8-fold higher doses of cytokine to be administered without toxicity. The improved therapeutic window enables substantially increased tumor clearance by murine T cell and human CAR-T cell therapy in vivo.
Collapse
Affiliation(s)
- Li Tang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yiran Zheng
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mariane Bandeira Melo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Llian Mabardi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
| | - Ana P Castaño
- Cellular Immunotherapy Program, Massachusetts General Hospital (MGH) Cancer Center, Charlestown, Massachusetts, USA
| | - Yu-Qing Xie
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Na Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Hing C Wong
- Altor BioScience Corporation, Miramar, Florida, USA
| | - Emily K Jeng
- Altor BioScience Corporation, Miramar, Florida, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital (MGH) Cancer Center, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Darrell J Irvine
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
397
|
Lanitis E, Dangaj D, Irving M, Coukos G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann Oncol 2018; 28:xii18-xii32. [PMID: 29045511 DOI: 10.1093/annonc/mdx238] [Citation(s) in RCA: 272] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
T-lymphocytes play a critical role in cancer immunity as evidenced by their presence in resected tumor samples derived from long-surviving patients, and impressive clinical responses to various immunotherapies that reinvigorate them. Indeed, tumors can upregulate a wide array of defense mechanisms, both direct and indirect, to suppress the ability of Tcells to reach the tumor bed and mount curative responses upon infiltration. In addition, patient and tumor genetics, previous antigenic experience, and the microbiome, are all important factors in shaping the T-cell repertoire and sensitivity to immunotherapy. Here, we review the mechanisms that regulate T-cell homing, infiltration, and activity within the solid tumor bed. Finally, we summarize different immunotherapies and combinatorial treatment strategies that enable the immune system to overcome barriers for enhanced tumor control and improved patient outcome.
Collapse
Affiliation(s)
- E Lanitis
- The Ludwig Branch for Cancer Research of the University of Lausanne, Epalinges
| | - D Dangaj
- The Ludwig Branch for Cancer Research of the University of Lausanne, Epalinges
| | - M Irving
- The Ludwig Branch for Cancer Research of the University of Lausanne, Epalinges
| | - G Coukos
- The Ludwig Branch for Cancer Research of the University of Lausanne, Epalinges.,Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| |
Collapse
|
398
|
Tague EP, Dotson HL, Tunney SN, Sloas DC, Ngo JT. Chemogenetic control of gene expression and cell signaling with antiviral drugs. Nat Methods 2018; 15:519-522. [PMID: 29967495 DOI: 10.1038/s41592-018-0042-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 04/24/2018] [Indexed: 02/04/2023]
Abstract
We developed a method in which the NS3 cis-protease from hepatitis C virus can be used as a ligand-inducible connection to control the function and localization of engineered proteins in mammalian cells. To demonstrate the versatility of this approach, we designed drug-sensitive transcription factors and transmembrane signaling proteins, the activities of which can be tightly and reversibly controlled through the use of clinically tested antiviral protease inhibitors.
Collapse
Affiliation(s)
- Elliot P Tague
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, USA
| | - Hannah L Dotson
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, USA
| | - Shannon N Tunney
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, USA
| | - D Christopher Sloas
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, USA
| | - John T Ngo
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, USA.
| |
Collapse
|
399
|
Halim L, Ajina A, Maher J. Pre-clinical development of chimeric antigen receptor T-cell immunotherapy: Implications of design for efficacy and safety. Best Pract Res Clin Haematol 2018; 31:117-125. [PMID: 29909912 DOI: 10.1016/j.beha.2018.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/03/2018] [Accepted: 04/17/2018] [Indexed: 11/18/2022]
Abstract
Following the landmark approvals by the United States Food and Drug Administration, the adoptive transfer of CD19-directed chimeric antigen receptor (CAR) T-cells has now entered mainstream clinical practice for patients with chemotherapy-resistant or refractory B-cell malignancies. These approvals have followed on from a prolonged period of pre-clinical evaluation, informing the design of clinical trials that have demonstrated unprecedented efficacy in this difficult to treat patient population. However, the delivery of autologous CAR-engineered T-cell therapy is complex, costly and not without significant risk. Here we summarize the key themes of CAR T-cell preclinical development and highlight a number of innovative strategies designed to further address toxicity and improve efficacy. In concert with the emerging promise of precision genome editing, it is hoped these next generation products will increase the repertoire of clinical applications of CAR T-cell therapy in malignant and perhaps other disease settings.
Collapse
Affiliation(s)
- Leena Halim
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, UK.
| | - Adam Ajina
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, UK.
| | - John Maher
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, UK; Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, UK; Department of Immunology, Eastbourne Hospital, East Sussex, UK.
| |
Collapse
|
400
|
Chu W, Zhou Y, Tang Q, Wang M, Ji Y, Yan J, Yin D, Zhang S, Lu H, Shen J. Bi-specific ligand-controlled chimeric antigen receptor T-cell therapy for non-small cell lung cancer. Biosci Trends 2018; 12:298-308. [PMID: 29899195 DOI: 10.5582/bst.2018.01048] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Our goal is to develop a switch-controlled approach to enable better control of reactivity and safety of chimeric antigen receptor (CAR)-T therapy for non-small-cell lung cancer (NSCLC). Lentiviral transduction was performed to generate anti-FITC CAR-T cells and target cells stably expressing either isoform of the folate receptor. Colorimetric-based cytotoxic assay, enzyme-linked immunosorbent assay, and multiparametric flow cytometry analysis were used to evaluate the specificity and activity of CAR-T cells in vitro. Human primary T cells stably expressing the fully human anti-FITC CAR were generated. Anti-FITC CAR-T cells displayed antigen-specific and folate-FTIC dependent reactivity against engineered A549-FRα and THP-1-FRβ. The selective activation and proliferation of anti-FITC CAR-T cells in vitro stringently relied on the co-existence of folate-FITC and FR- expressing target cells and was dose-titratable with the folate-FITC switch. The excellent in vitro efficacy and specificity of an adaptor-controlled CAR-T therapy to target both tumor cells and tumor-associated macrophages in NSCLCs were validated.
Collapse
Affiliation(s)
- Wenqi Chu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University
| | - Yixiong Zhou
- Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine
| | - Qi Tang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University
| | - Min Wang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University
| | - Yongjia Ji
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University
| | - Jingjing Yan
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University
| | - Dan Yin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University
| | - Shuye Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University
| | - Hongzhou Lu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University.,Department of Infectious Diseases, Huashan Hospital Affiliated to Fudan University
| | - Jiayin Shen
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University
| |
Collapse
|