301
|
Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, Shultz LD. Humanized Mouse Models of Clinical Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 12:187-215. [PMID: 27959627 DOI: 10.1146/annurev-pathol-052016-100332] [Citation(s) in RCA: 380] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunodeficient mice engrafted with functional human cells and tissues, that is, humanized mice, have become increasingly important as small, preclinical animal models for the study of human diseases. Since the description of immunodeficient mice bearing mutations in the IL2 receptor common gamma chain (IL2rgnull) in the early 2000s, investigators have been able to engraft murine recipients with human hematopoietic stem cells that develop into functional human immune systems. These mice can also be engrafted with human tissues such as islets, liver, skin, and most solid and hematologic cancers. Humanized mice are permitting significant progress in studies of human infectious disease, cancer, regenerative medicine, graft-versus-host disease, allergies, and immunity. Ultimately, use of humanized mice may lead to the implementation of truly personalized medicine in the clinic. This review discusses recent progress in the development and use of humanized mice and highlights their utility for the study of human diseases.
Collapse
Affiliation(s)
- Nicole C Walsh
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Laurie L Kenney
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Sonal Jangalwe
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Ken-Edwin Aryee
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Michael A Brehm
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | | |
Collapse
|
302
|
Rotiroti MC, Arcangeli S, Casucci M, Perriello V, Bondanza A, Biondi A, Tettamanti S, Biagi E. Acute Myeloid Leukemia Targeting by Chimeric Antigen Receptor T Cells: Bridging the Gap from Preclinical Modeling to Human Studies. Hum Gene Ther 2016; 28:231-241. [PMID: 27967241 DOI: 10.1089/hum.2016.092] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Acute myeloid leukemia (AML) still represents an unmet clinical need for adult and pediatric high-risk patients, thus demanding advanced and personalized therapies. In this regard, different targeted immunotherapeutic approaches are available, ranging from naked monoclonal antibodies (mAb) to conjugated and multifunctional mAbs (i.e., BiTEs and DARTs). Recently, researchers have focused their attention on novel techniques of genetic manipulation specifically to redirect cytotoxic T cells endowed with chimeric antigen receptors (CARs) toward selected tumor associated antigens. So far, CAR T cells targeting the CD19 antigen expressed by B-cell origin hematological cancers have gained impressive clinical results, leading to the possibility of translating the CAR platform to treat other hematological malignancies such as AML. However, one of the main concerns in the field of AML CAR immunotherapy is the identification of an ideal target cell surface antigen, being highly expressed on tumor cells but minimally present on healthy tissues, together with the design of an anti-AML CAR appropriately balancing efficacy and safety profiles. The current review focuses mainly on AML target antigens and the related immunotherapeutic approaches developed so far, deeply dissecting methods of CAR T cell safety improvements, when designing novel CARs approaching human studies.
Collapse
Affiliation(s)
- Maria Caterina Rotiroti
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Silvia Arcangeli
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Monica Casucci
- 2 Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Hospital Scientific Institute, Vita-Salute San Raffaele University , Milan, Italy
| | - Vincenzo Perriello
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Attilio Bondanza
- 2 Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Hospital Scientific Institute, Vita-Salute San Raffaele University , Milan, Italy
| | - Andrea Biondi
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Sarah Tettamanti
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Ettore Biagi
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| |
Collapse
|
303
|
Balakrishnan A, Goodpaster T, Randolph-Habecker J, Hoffstrom BG, Jalikis FG, Koch LK, Berger C, Kosasih PL, Rajan A, Sommermeyer D, Porter PL, Riddell SR. Analysis of ROR1 Protein Expression in Human Cancer and Normal Tissues. Clin Cancer Res 2016; 23:3061-3071. [PMID: 27852699 DOI: 10.1158/1078-0432.ccr-16-2083] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/31/2016] [Accepted: 11/02/2016] [Indexed: 11/16/2022]
Abstract
Purpose: This study examines cell surface ROR1 expression in human tumors and normal tissues. ROR1 is considered a promising target for cancer therapy due to putative tumor-specific expression, and multiple groups are developing antibodies and/or chimeric antigen receptor-modified T cells to target ROR1. On-target, off-tumor toxicity is a challenge for most nonmutated tumor antigens; however, prior studies suggest that ROR1 is absent on most normal tissues.Experimental Design: Our studies show that published antibodies lack sensitivity to detect endogenous levels of cell surface ROR1 by immunohistochemistry (IHC) in formalin-fixed, paraffin-embedded tissues. We developed a ROR1-specific monoclonal antibody (mAb) targeting the carboxy-terminus of ROR1 and evaluated its specificity and sensitivity in IHC.Results: The 6D4 mAb is a sensitive and specific reagent to detect cell surface ROR1 by IHC. The data show that ROR1 is homogenously expressed on a subset of ovarian cancer, triple-negative breast cancer, and lung adenocarcinomas. Contrary to previous findings, we found ROR1 is expressed on several normal tissues, including parathyroid; pancreatic islets; and regions of the esophagus, stomach, and duodenum. The 6D4 mAb recognizes rhesus ROR1, and ROR1 expression was similar in human and macaque tissues, suggesting that the macaque is a suitable model to evaluate safety of ROR1-targeted therapies.Conclusions: ROR1 is a promising immunotherapeutic target in many epithelial tumors; however, high cell surface ROR1 expression in multiple normal tissues raises concerns for on-target off-tumor toxicities. Clinical translation of ROR1-targeted therapies warrants careful monitoring of toxicities to normal organs and may require strategies to ensure patient safety. Clin Cancer Res; 23(12); 3061-71. ©2016 AACR.
Collapse
Affiliation(s)
- Ashwini Balakrishnan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Tracy Goodpaster
- Experimental Histopathology Shared Resource, Fred Hutchinson Cancer Research Center, Seattle Washington
| | - Julie Randolph-Habecker
- Experimental Histopathology Shared Resource, Fred Hutchinson Cancer Research Center, Seattle Washington
| | - Benjamin G Hoffstrom
- Antibody Development Shared Resource, Fred Hutchinson Cancer Research Center, Seattle Washington
| | - Florencia G Jalikis
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Lisa K Koch
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Carolina Berger
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Paula L Kosasih
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Anusha Rajan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Daniel Sommermeyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Peggy L Porter
- Department of Pathology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Stanley R Riddell
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Department of Medicine, University of Washington, Seattle, Washington.,Institute for Advanced Study, Technical University of Munich, Munich, Germany
| |
Collapse
|
304
|
Inoo K, Inagaki R, Fujiwara K, Sasawatari S, Kamigaki T, Nakagawa S, Okada N. Immunological quality and performance of tumor vessel-targeting CAR-T cells prepared by mRNA-EP for clinical research. MOLECULAR THERAPY-ONCOLYTICS 2016; 3:16024. [PMID: 27909701 PMCID: PMC5111575 DOI: 10.1038/mto.2016.24] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/14/2016] [Accepted: 08/27/2016] [Indexed: 01/03/2023]
Abstract
We previously reported that tumor vessel-redirected T cells, which were genetically engineered with chimeric antigen receptor (CAR) specific for vascular endothelial growth factor receptor 2 (VEGFR2), demonstrated significant antitumor effects in various murine solid tumor models. In the present study, we prepared anti-VEGFR2 CAR-T cells by CAR-coding mRNA electroporation (mRNA-EP) and analyzed their immunological characteristics and functions for use in clinical research. The expression of anti-VEGFR2 CAR on murine and human T cells was detected with approximately 100% efficiency for a few days, after peaking 6-12 hours after mRNA-EP. Triple transfer of murine anti-VEGFR2 CAR-T cells into B16BL6 tumor-bearing mice demonstrated an antitumor effect comparable to that for the single transfer of CAR-T cells engineered with retroviral vector. The mRNA-EP did not cause any damage or defects to human T-cell characteristics, as determined by viability, growth, and phenotypic parameters. Additionally, two kinds of human anti-VEGFR2 CAR-T cells, which expressed different CAR construction, differentiated to effector phase with cytokine secretion and cytotoxic activity in antigen-specific manner. These results indicate that our anti-VEGFR2 CAR-T cells prepared by mRNA-EP have the potential in terms of quality and performance to offer the prospect of safety and efficacy in clinical research as cellular medicine.
Collapse
Affiliation(s)
- Kanako Inoo
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University , Osaka, Japan
| | - Ryo Inagaki
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University , Osaka, Japan
| | - Kento Fujiwara
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University , Osaka, Japan
| | | | - Takashi Kamigaki
- MEDINET Co., Ltd., Kanagawa, Japan; Seta Clinic Group, Tokyo, Japan
| | - Shinsaku Nakagawa
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University , Osaka, Japan
| | - Naoki Okada
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University , Osaka, Japan
| |
Collapse
|
305
|
ROR1 is a novel prognostic biomarker in patients with lung adenocarcinoma. Sci Rep 2016; 6:36447. [PMID: 27830754 PMCID: PMC5103212 DOI: 10.1038/srep36447] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/13/2016] [Indexed: 02/05/2023] Open
Abstract
Currently, there is no reliable biomarker to clinically predict the prognosis of lung adenocarcinoma (ADC). The receptor-tyrosine-kinase like orphan receptor 1 (ROR1) is reported to be overexpressed and associated with poor prognosis in several tumors. This study aimed to examine the expression of ROR1 and evaluate its prognostic significance in human lung ADC patients. In this present study, Western blot analysis and immunohistochemistry were performed to characterize expression of ROR1 protein in lung ADC patients. The results revealed that ROR1 protein expression was significantly higher in lung ADC tissues than that in their adjacent non-tumor tissues. Patients at advanced stages and those with positive lymph node metastasis expressed higher level of ROR1 (P < 0.001). Moreover, Chi-square test showed that ROR1 expression was correlated to gender (P = 0.028), the 7th edition of the American Joint Committee on Cancer tumor-node-metastasis (AJCC TNM) staging system and lymph node metastasis (P < 0.001). Kaplan-Meier survival analysis indicated an association of high ROR1 expression with worse overall survival (OS) in lung ADC patients (P < 0.001). Multivariate COX regression analysis further confirmed that ROR1 is an independent prognostic predictor (P < 0.001, HR = 4.114, 95% CI: 2.513–6.375) for OS. Therefore, ROR1 expression significantly correlates with malignant attributes of lung ADC and it may serve as a novel prognostic marker in lung ADC patients.
Collapse
|
306
|
Watanabe N, Bajgain P, Sukumaran S, Ansari S, Heslop HE, Rooney CM, Brenner MK, Leen AM, Vera JF. Fine-tuning the CAR spacer improves T-cell potency. Oncoimmunology 2016; 5:e1253656. [PMID: 28180032 DOI: 10.1080/2162402x.2016.1253656] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/20/2016] [Accepted: 10/22/2016] [Indexed: 10/20/2022] Open
Abstract
The adoptive transfer of genetically engineered T cells expressing chimeric antigen receptors (CARs) has emerged as a transformative cancer therapy with curative potential, precipitating a wave of preclinical and clinical studies in academic centers and the private sector. Indeed, significant effort has been devoted to improving clinical benefit by incorporating accessory genes/CAR endodomains designed to enhance cellular migration, promote in vivo expansion/persistence or enhance safety by genetic programming to enable the recognition of a tumor signature. However, our efforts centered on exploring whether CAR T-cell potency could be enhanced by modifying pre-existing CAR components. We now demonstrate how molecular refinements to the CAR spacer can impact multiple biological processes including tonic signaling, cell aging, tumor localization, and antigen recognition, culminating in superior in vivo antitumor activity.
Collapse
Affiliation(s)
- Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| | - Pradip Bajgain
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| | - Sujita Sukumaran
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| | - Salma Ansari
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| | - Juan F Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital , Houston, Texas, USA
| |
Collapse
|
307
|
The Promise of Chimeric Antigen Receptor Engineered T Cells in the Treatment of Hematologic Malignancies. Cancer J 2016; 22:27-33. [PMID: 26841014 DOI: 10.1097/ppo.0000000000000166] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Relapsed and refractory hematologic malignancies have a very poor prognosis. Chimeric antigen receptor T cells are emerging as a powerful therapy in this setting. Early clinical trials of genetically modified T cells for the treatment of non-Hodgkin lymphoma, chronic lymphocytic leukemia, and acute lymphoblastic leukemia have shown high complete response rates in patients with few therapeutic options. Exploration is ongoing for other hematologic malignancies including multiple myeloma, acute myeloid leukemia, and Hodgkin lymphoma (HL). At the same time, the design and production of chimeric antigen receptor T cells are being advanced so that this therapy can be more widely utilized. Cytokine release syndrome and neurotoxicity are common, but they are treatable and fully reversible. This review will review available data as well as future developments and challenges in the field.
Collapse
|
308
|
Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: Summary and perspective. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jocit.2016.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
309
|
Gross G, Eshhar Z. Therapeutic Potential of T Cell Chimeric Antigen Receptors (CARs) in Cancer Treatment: Counteracting Off-Tumor Toxicities for Safe CAR T Cell Therapy. Annu Rev Pharmacol Toxicol 2016; 56:59-83. [PMID: 26738472 DOI: 10.1146/annurev-pharmtox-010814-124844] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A chimeric antigen receptor (CAR) is a recombinant fusion protein combining an antibody-derived targeting fragment with signaling domains capable of activating T cells. Recent early-phase clinical trials have demonstrated the remarkable ability of CAR-modified T cells to eliminate B cell malignancies. This review describes the choice of target antigens and CAR manipulations to maximize antitumor specificity. Benefits and current limitations of CAR-modified T cells are discussed, with a special focus on the distribution of tumor antigens on normal tissues and the risk of on-target, off-tumor toxicities in the clinical setting. We present current methodologies for pre-evaluating these risks and review the strategies for counteracting potential off-tumor effects. Successful implementation of these approaches will improve the safety and efficacy of CAR T cell therapy and extend the range of cancer patients who may be treated.
Collapse
Affiliation(s)
- Gideon Gross
- Laboratory of Immunology, MIGAL, Galilee Research Institute, Kiryat Shmona 11016, Israel; .,Department of Biotechnology, Tel-Hai College, Upper Galilee 12210, Israel.,Center of Cancer Research, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
| | - Zelig Eshhar
- Center of Cancer Research, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel.,Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
310
|
Zhang XY, Zhang PY. Receptor tyrosine kinases in carcinogenesis. Oncol Lett 2016; 12:3679-3682. [PMID: 27900053 PMCID: PMC5104145 DOI: 10.3892/ol.2016.5200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/12/2016] [Indexed: 12/23/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are cell surface glycoproteins with enzymatic activity involved in the regulation of various important functions. In all-important physiological functions including differentiation, cell-cell interactions, survival, proliferation, metabolism, migration and signaling these receptors are the key players of regulation. Additionally, mutations of RTKs or their overexpression have been described in many human cancers and are being explored as a novel avenue for a new therapeutic approach. Some of the deregulated RTKs observed to be significantly affected in cancers included vascular endothelial growth factor receptor, epidermal growth factor receptor, fibroblast growth factor receptor, RTK-like orphan receptor 1 (ROR1) and the platelet-derived growth factor receptor. These deregulated RTKs offer attractive possibilities for the new anticancer therapeutic approach involving specific targeting by monoclonal antibodies as well as kinase. The present review aimed to highlight recent perspectives of RTK ROR1 in cancer.
Collapse
Affiliation(s)
- Xiao-Ying Zhang
- Nanjing University of Chinese Medicine, Information Institute, Nanjing, Jiangsu 221009, P.R. China
| | - Pei-Ying Zhang
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
311
|
Wu Y, Jiang S, Ying T. From therapeutic antibodies to chimeric antigen receptors (CARs): making better CARs based on antigen-binding domain. Expert Opin Biol Ther 2016; 16:1469-1478. [PMID: 27618260 DOI: 10.1080/14712598.2016.1235148] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION A variety of approaches are being pursued to improve the safety and antitumor potency of chimeric antigen receptor (CAR) T-cell therapy. However, most engineering efforts have thus far been focused on its intracellular signaling domain, while its extracellular antigen-binding domain has received less attention. Areas covered: Herein, the authors summarize the current knowledge of CAR T-cell therapy. Accordingly, they focus on its antigen-binding domain, discuss key considerations for selecting an optimal single-chain variable fragment (scFv) when designing a CAR, and suggest potential directions aimed at developing the next-generation CARs. Expert opinion: The extracellular region of CARs can play a decisive role in their safety and efficacy. Instead of directly translating an available therapeutic mAb to a scFv-based CAR construct, the authors suggest that various CAR-displayed scFvs with different affinity, specificity and binding epitopes against an individual target molecule should be generated and evaluated side-by-side. Incorporating new antibody formats that possess characteristics superior to those of scFvs may be one way to engineer safer and more effective CARs. The authors expect that further CAR engineering will enable us to target more antigens involved in hematological and solid malignancies with minimal side effects to serve unmet clinical needs.
Collapse
Affiliation(s)
- Yanling Wu
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Shibo Jiang
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Tianlei Ying
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences , Fudan University , Shanghai , China
| |
Collapse
|
312
|
Abstract
Adoptive T-cell therapies have shown exceptional promise in the treatment of cancer, especially B-cell malignancies. Two distinct strategies have been used to redirect the activity of ex vivo engineered T cells. In one case, the well-known ability of the T-cell receptor (TCR) to recognize a specific peptide bound to a major histocompatibility complex molecule has been exploited by introducing a TCR against a cancer-associated peptide/human leukocyte antigen complex. In the other strategy, synthetic constructs called chimeric antigen receptors (CARs) that contain antibody variable domains (single-chain fragments variable) and signaling domains have been introduced into T cells. Whereas many reviews have described these two approaches, this review focuses on a few recent advances of significant interest. The early success of CARs has been followed by questions about optimal configurations of these synthetic constructs, especially for efficacy against solid tumors. Among the many features that are important, the dimensions and stoichiometries of CAR/antigen complexes at the synapse have recently begun to be appreciated. In TCR-mediated approaches, recent evidence that mutated peptides (neoantigens) serve as targets for endogenous T-cell responses suggests that these neoantigens may also provide new opportunities for adoptive T-cell therapies with TCRs.
Collapse
Affiliation(s)
- Preeti Sharma
- Department of Biochemistry, University of Illinois, Urbana, IL, USA
| | - David M Kranz
- Department of Biochemistry, University of Illinois, Urbana, IL, USA
| |
Collapse
|
313
|
Yang F, Jin H, Wang J, Sun Q, Yan C, Wei F, Ren X. Adoptive Cellular Therapy (ACT) for Cancer Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:169-239. [PMID: 27240459 DOI: 10.1007/978-94-017-7555-7_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Adoptive cellular therapy (ACT) with various lymphocytes or antigen-presenting cells is one stone in the pillar of cancer immunotherapy, which relies on the tumor-specific T cell. The transfusion of bulk T-cell population into patients is an effective treatment for regression of cancer. In this chapter, we summarize the development of various strategies in ACT for cancer immunotherapy and discuss some of the latest progress and obstacles in technical, safety, and even regulatory aspects to translate these technologies to the clinic. ACT is becoming a potentially powerful approach to cancer treatment. Further experiments and clinical trials are needed to optimize this strategy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Hao Jin
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Qian Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China. .,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China. .,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
| |
Collapse
|
314
|
Abstract
The immune system evolved to distinguish non-self from self to protect the organism. As cancer is derived from our own cells, immune responses to dysregulated cell growth present a unique challenge. This is compounded by mechanisms of immune evasion and immunosuppression that develop in the tumour microenvironment. The modern genetic toolbox enables the adoptive transfer of engineered T cells to create enhanced anticancer immune functions where natural cancer-specific immune responses have failed. Genetically engineered T cells, so-called 'living drugs', represent a new paradigm in anticancer therapy. Recent clinical trials using T cells engineered to express chimeric antigen receptors (CARs) or engineered T cell receptors (TCRs) have produced stunning results in patients with relapsed or refractory haematological malignancies. In this Review we describe some of the most recent and promising advances in engineered T cell therapy with a particular emphasis on what the next generation of T cell therapy is likely to entail.
Collapse
MESH Headings
- Antigen Presentation
- Antigens, CD19/immunology
- Antigens, Neoplasm/immunology
- Clinical Trials as Topic
- Costimulatory and Inhibitory T-Cell Receptors/genetics
- Costimulatory and Inhibitory T-Cell Receptors/immunology
- Cytokines/metabolism
- Forecasting
- Gene Editing
- Gene Transfer Techniques
- Genetic Engineering
- HLA Antigens/immunology
- Hematologic Neoplasms/immunology
- Hematologic Neoplasms/therapy
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Models, Immunological
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Syndrome
- T-Cell Antigen Receptor Specificity
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/transplantation
- Tumor Escape
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Andrew D Fesnak
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-5156, USA
| | - Carl H June
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-5156, USA
| | - Bruce L Levine
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-5156, USA
| |
Collapse
|
315
|
Thokala R, Olivares S, Mi T, Maiti S, Deniger D, Huls H, Torikai H, Singh H, Champlin RE, Laskowski T, McNamara G, Cooper LJN. Redirecting Specificity of T cells Using the Sleeping Beauty System to Express Chimeric Antigen Receptors by Mix-and-Matching of VL and VH Domains Targeting CD123+ Tumors. PLoS One 2016; 11:e0159477. [PMID: 27548616 PMCID: PMC4993583 DOI: 10.1371/journal.pone.0159477] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/10/2016] [Indexed: 12/20/2022] Open
Abstract
Adoptive immunotherapy infusing T cells with engineered specificity for CD19 expressed on B- cell malignancies is generating enthusiasm to extend this approach to other hematological malignancies, such as acute myelogenous leukemia (AML). CD123, or interleukin 3 receptor alpha, is overexpressed on most AML and some lymphoid malignancies, such as acute lymphocytic leukemia (ALL), and has been an effective target for T cells expressing chimeric antigen receptors (CARs). The prototypical CAR encodes a VH and VL from one monoclonal antibody (mAb), coupled to a transmembrane domain and one or more cytoplasmic signaling domains. Previous studies showed that treatment of an experimental AML model with CD123-specific CAR T cells was therapeutic, but at the cost of impaired myelopoiesis, highlighting the need for systems to define the antigen threshold for CAR recognition. Here, we show that CARs can be engineered using VH and VL chains derived from different CD123-specific mAbs to generate a panel of CAR+ T cells. While all CARs exhibited specificity to CD123, one VH and VL combination had reduced lysis of normal hematopoietic stem cells. This CAR’s in vivo anti-tumor activity was similar whether signaling occurred via chimeric CD28 or CD137, prolonging survival in both AML and ALL models. Co-expression of inducible caspase 9 eliminated CAR+ T cells. These data help support the use of CD123-specific CARs for treatment of CD123+ hematologic malignancies.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- Caspase 9/genetics
- Caspase 9/immunology
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Gene Expression
- Genetic Engineering/methods
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/pathology
- Humans
- Immunotherapy, Adoptive/methods
- Interleukin-3 Receptor alpha Subunit/genetics
- Interleukin-3 Receptor alpha Subunit/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Molecular Targeted Therapy
- Plasmids
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Single-Domain Antibodies/genetics
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transfection
- Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
Collapse
Affiliation(s)
- Radhika Thokala
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, United States of America
| | - Simon Olivares
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Tiejuan Mi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sourindra Maiti
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Drew Deniger
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Helen Huls
- Intrexon Corporation, Germantown, Maryland, United States of America
| | - Hiroki Torikai
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Harjeet Singh
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Richard E. Champlin
- Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Tamara Laskowski
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - George McNamara
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Laurence J. N. Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Ziopharm Oncology Inc., Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
316
|
Hombach AA, Abken H. Shared target antigens on cancer cells and tissue stem cells: go or no-go for CAR T cells? Expert Rev Clin Immunol 2016; 13:151-155. [PMID: 27546707 DOI: 10.1080/1744666x.2016.1221763] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Adoptive therapy with chimeric antigen receptor (CAR) T cells redirected towards CD19 produces remissions of B cell malignancies, however, it also eradicates healthy B cells sharing the target antigen. Such 'on-target off-tumor' toxicity raises serious safety concerns when the target antigen is also expressed by tissue stem cells, with the risk of lasting tissue destruction. Areas covered: We discuss CAR T cell targeting of activation antigens versus lineage associated antigens on the basis of recent experimental and animal data and the literature in the field. Expert commentary: Targeting an activation associated antigen which is transiently expressed by stem cells seems to be safe, like CAR T cells targeting CD30 spare CD30+ hematopoietic stem and progenitor cells while eliminating CD30+ lymphoma cells, whereas targeting lineage associated antigens which increase in expression during cell maturation, like folate receptor-β and CD123, is of risk to destruct tissue stem cells.
Collapse
Affiliation(s)
- Andreas A Hombach
- a Center for Molecular Medicine Cologne , University of Cologne , Cologne , Germany.,b Department I of Internal Medicine , University Hospital Cologne , Cologne , Germany
| | - Hinrich Abken
- a Center for Molecular Medicine Cologne , University of Cologne , Cologne , Germany.,b Department I of Internal Medicine , University Hospital Cologne , Cologne , Germany
| |
Collapse
|
317
|
Chimeric antigen receptor T cells targeting Fc μ receptor selectively eliminate CLL cells while sparing healthy B cells. Blood 2016; 128:1711-22. [PMID: 27535994 DOI: 10.1182/blood-2016-01-692046] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 08/05/2016] [Indexed: 12/28/2022] Open
Abstract
Adoptive cell therapy of chronic lymphocytic leukemia (CLL) with chimeric antigen receptor (CAR)-modified T cells targeting CD19 induced lasting remission of this refractory disease in a number of patients. However, the treatment is associated with prolonged "on-target off-tumor" toxicities due to the targeted elimination of healthy B cells demanding more selectivity in targeting CLL cells. We identified the immunoglobulin M Fc receptor (FcμR), also known as the Fas apoptotic inhibitory molecule-3 or TOSO, as a target for a more selective treatment of CLL by CAR T cells. FcμR is highly and consistently expressed by CLL cells; only minor levels are detected on healthy B cells or other hematopoietic cells. T cells with a CAR specific for FcμR efficiently responded toward CLL cells, released a panel of proinflammatory cytokines and lytic factors, like soluble FasL and granzyme B, and eliminated the leukemic cells. In contrast to CD19 CAR T cells, anti-FcμR CAR T cells did not attack healthy B cells. T cells with anti-FcμR CAR delayed outgrowth of Mec-1-induced leukemia in a xenograft mouse model. T cells from CLL patients in various stages of the disease, modified by the anti-FcμR CAR, purged their autologous CLL cells in vitro without reducing the number of healthy B cells, which is the case with anti-CD19 CAR T cells. Compared with the currently used therapies, the data strongly imply a superior therapeutic index of anti-FcμR CAR T cells for the treatment of CLL.
Collapse
|
318
|
[Anti-CD19 chimeric antigen receptor T cells (CART-19) for hemotological malignancies therapy]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 37:164-8. [PMID: 27014991 PMCID: PMC7348191 DOI: 10.3760/cma.j.issn.0253-2727.2016.02.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
319
|
Ellebrecht CT, Bhoj VG, Nace A, Choi EJ, Mao X, Cho MJ, Di Zenzo G, Lanzavecchia A, Seykora JT, Cotsarelis G, Milone MC, Payne AS. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science 2016; 353:179-84. [PMID: 27365313 PMCID: PMC5343513 DOI: 10.1126/science.aaf6756] [Citation(s) in RCA: 458] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/09/2016] [Indexed: 12/18/2022]
Abstract
Ideally, therapy for autoimmune diseases should eliminate pathogenic autoimmune cells while sparing protective immunity, but feasible strategies for such an approach have been elusive. Here, we show that in the antibody-mediated autoimmune disease pemphigus vulgaris (PV), autoantigen-based chimeric immunoreceptors can direct T cells to kill autoreactive B lymphocytes through the specificity of the B cell receptor (BCR). We engineered human T cells to express a chimeric autoantibody receptor (CAAR), consisting of the PV autoantigen, desmoglein (Dsg) 3, fused to CD137-CD3ζ signaling domains. Dsg3 CAAR-T cells exhibit specific cytotoxicity against cells expressing anti-Dsg3 BCRs in vitro and expand, persist, and specifically eliminate Dsg3-specific B cells in vivo. CAAR-T cells may provide an effective and universal strategy for specific targeting of autoreactive B cells in antibody-mediated autoimmune disease.
Collapse
Affiliation(s)
| | - Vijay G Bhoj
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arben Nace
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eun Jung Choi
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xuming Mao
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Jeffrey Cho
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giovanni Di Zenzo
- Laboratory of Molecular and Cellular Biology, Istituto Dermopatico dell'Immacolata (IDI-IRCCS), 00167 Rome, Italy
| | | | - John T Seykora
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George Cotsarelis
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Aimee S Payne
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
320
|
Abstract
PURPOSE OF REVIEW Chimeric antigen receptors (CARs) are synthetic immunoreceptors, which can redirect T cells to selectively kill tumor cells, and as 'living drugs' have the potential to generate long-term antitumor immunity. Given their recent clinical successes for the treatment of refractory B-cell malignancies, there is a strong push toward advancing this immunotherapy to other hematological diseases and solid cancers. Here, we summarize the current state of the field, highlighting key variables for the optimal application of CAR T cells for cancer immunotherapy. RECENT FINDINGS Advances in CAR T-cell therapy have highlighted intrinsic CAR design and T-cell manufacturing methods as critical components for maximal therapeutic success. Similarly, addressing the unique extrinsic challenges of each tumor type, including overcoming the immunosuppressive tumor microenvironment and tumor heterogeneity, and mitigating potential toxicity, will dominate the next wave of CAR T-cell development. SUMMARY CAR T-cell therapeutic optimization, including intrinsic and extrinsic factors, is critical to developing effective CAR T-cell therapies for cancer. The excitement of CAR T-cell immunotherapy has just begun, and will continue with new insights revealed in laboratory research and in ongoing clinical investigations.
Collapse
|
321
|
Turtle CJ, Maloney DG. Clinical trials of CD19-targeted CAR-modified T cell therapy; a complex and varied landscape. Expert Rev Hematol 2016; 9:719-21. [DOI: 10.1080/17474086.2016.1203251] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
322
|
Beatty GL, O'Hara M. Chimeric antigen receptor-modified T cells for the treatment of solid tumors: Defining the challenges and next steps. Pharmacol Ther 2016; 166:30-9. [PMID: 27373504 DOI: 10.1016/j.pharmthera.2016.06.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2016] [Indexed: 01/07/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown promise in CD19 expressing hematologic malignancies, but how to translate this success to solid malignancies remains elusive. Effective translation of CAR T cells to solid tumors will require an understanding of potential therapeutic barriers, including factors that regulate CAR T cells expansion, persistence, trafficking, and fate within tumors. Herein, we describe the current state of CAR T cells in solid tumors; define key barriers to CAR T cell efficacy and mechanisms underlying these barriers, outline potential avenues for overcoming these therapeutic obstacles, and discuss the future of translating CAR T cells for the treatment of patients with solid malignancies.
Collapse
Affiliation(s)
- Gregory L Beatty
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mark O'Hara
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
323
|
Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors. Leukemia 2016; 31:186-194. [PMID: 27491640 DOI: 10.1038/leu.2016.180] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/28/2022]
Abstract
Immunotherapy with T cell modified with gamma-retroviral or lentiviral (LV) vectors to express a chimeric antigen receptor (CAR) has shown remarkable efficacy in clinical trials. However, the potential for insertional mutagenesis and genotoxicity of viral vectors is a safety concern, and their cost and regulatory demands a roadblock for rapid and broad clinical translation. Here, we demonstrate that CAR T cells can be engineered through non-viral Sleeping Beauty (SB) transposition of CAR genes from minimalistic DNA vectors called minicircles (MCs). We analyzed genomic distribution of SB and LV integrations and show that a significantly higher proportion of MC-derived CAR transposons compared with LV integrants had occurred outside of highly expressed and cancer-related genes into genomic safe harbor loci that are not expected to cause mutagenesis or genotoxicity. CD19-CAR T cells engineered with our enhanced SB approach conferred potent reactivity in vitro and eradicated lymphoma in a xenograft model in vivo. Intriguingly, electroporation of SB MCs is substantially more effective and less toxic compared with conventional plasmids, and enables cost-effective rapid preparation of therapeutic CAR T-cell doses. This approach sets a new standard in advanced cellular and gene therapy and will accelerate and increase the availability of CAR T-cell therapy to treat hematologic malignancies.
Collapse
|
324
|
Sakemura R, Terakura S, Watanabe K, Julamanee J, Takagi E, Miyao K, Koyama D, Goto T, Hanajiri R, Nishida T, Murata M, Kiyoi H. A Tet-On Inducible System for Controlling CD19-Chimeric Antigen Receptor Expression upon Drug Administration. Cancer Immunol Res 2016; 4:658-68. [PMID: 27329987 DOI: 10.1158/2326-6066.cir-16-0043] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/03/2016] [Indexed: 11/16/2022]
Abstract
T cells genetically modified with a CD19 chimeric antigen receptor (CD19CAR) are remarkably effective against B-cell malignancies in clinical trials. However, major concerns remain regarding toxicities, such as hypogammaglobulinemia, due to B-cell aplasia or severe cytokine release syndrome after overactivation of CAR T cells. To resolve these adverse events, we aimed to develop an inducible CAR system by using a tetracycline regulation system that would be activated only in the presence of doxycycline (Dox). In this study, the second-generation CD19CAR was fused into the third-generation Tet-On vector (Tet-CD19CAR) and was retrovirally transduced into primary CD8(+) T cells. Tet-CD19CAR T cells were successfully generated and had minimal background CD19CAR expression without Dox. Tet-CD19CAR T cells in the presence of Dox were equivalently cytotoxic against CD19(+) cell lines and had equivalent cytokine production and proliferation upon CD19 stimulation, compared with conventional CD19CAR T cells. The Dox(+) Tet-CD19CAR T cells also had significant antitumor activity in a xenograft model. However, without Dox, Tet-CD19CAR T cells lost CAR expression and CAR T-cell functions in vitro and in vivo, clearly segregating the "On" and "Off" status of Tet-CD19CAR cells by Dox administration. In addition to suicide-gene technology, controlling the expression and the functions of CAR with an inducible vector is a potential solution for CAR T-cell therapy-related toxicities, and may improve the safety profile of CAR T-cell therapy. This strategy might also open the way to treat other malignancies in combination with other CAR or TCR gene-modified T cells. Cancer Immunol Res; 4(8); 658-68. ©2016 AACRSee related Spotlight by June, p. 643.
Collapse
Affiliation(s)
- Reona Sakemura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seitaro Terakura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Keisuke Watanabe
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jakrawadee Julamanee
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan. Division of Clinical Hematology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Erina Takagi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kotaro Miyao
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Koyama
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tatsunori Goto
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryo Hanajiri
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Nishida
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Makoto Murata
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
325
|
Turtle CJ, Riddell SR, Maloney DG. CD19-Targeted chimeric antigen receptor-modified T-cell immunotherapy for B-cell malignancies. Clin Pharmacol Ther 2016; 100:252-8. [PMID: 27170467 DOI: 10.1002/cpt.392] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/04/2016] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptors (CARs) comprise a tumor-targeting moiety, often in the form of a single chain variable fragment derived from a monoclonal antibody, fused to one or more intracellular T-cell signaling sequences. Lymphodepletion chemotherapy followed by infusion of T cells that are genetically modified to express a CD19-specific CAR is a promising therapy for patients with refractory CD19(+) B-cell malignancies, producing rates of complete remission that are remarkably high in acute lymphoblastic leukemia and encouraging in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Responses are often durable, although additional studies are needed to define the role of CAR-T cell immunotherapy in the context of other treatments. CAR-modified T-cell immunotherapy can be complicated by cytokine release syndrome and neurologic toxicity, which in most cases are manageable and reversible. Here we review recent clinical trial data and discuss issues for the field.
Collapse
Affiliation(s)
- C J Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - S R Riddell
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - D G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
326
|
Gacerez AT, Arellano B, Sentman CL. How Chimeric Antigen Receptor Design Affects Adoptive T Cell Therapy. J Cell Physiol 2016; 231:2590-8. [PMID: 27163336 DOI: 10.1002/jcp.25419] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 01/09/2023]
Abstract
Chimeric antigen receptor (CAR) T cells have been developed to treat tumors and have shown great success against B cell malignancies. Exploiting modular designs and swappable domains, CARs can target an array of cell surface antigens and, upon receptor-ligand interactions, direct signaling cascades, thereby driving T cell effector functions. CARs have been designed using receptors, ligands, or scFv binding domains. Different regions of a CAR have each been found to play a role in determining the overall efficacy of CAR T cells. Therefore, this review provides an overview of CAR construction and common designs. Each CAR region is discussed in the context of its importance to a CAR's function. Additionally, the review explores how various engineering strategies have been applied to CAR T cells in order to regulate CAR T cell function and activity. J. Cell. Physiol. 231: 2590-2598, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Albert T Gacerez
- Department of Microbiology and Immunology, Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, New Hampshire
| | - Benjamine Arellano
- Department of Microbiology and Immunology, Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, New Hampshire
| | - Charles L Sentman
- Department of Microbiology and Immunology, Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, New Hampshire
| |
Collapse
|
327
|
Vu BT, Tan Le D, Van Pham P. Synergistic effect of chimeric antigen receptors and cytokineinduced killer cells: An innovative combination for cancer therapy. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0025-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
328
|
Torikai H, Cooper LJ. Translational Implications for Off-the-shelf Immune Cells Expressing Chimeric Antigen Receptors. Mol Ther 2016; 24:1178-86. [PMID: 27203439 DOI: 10.1038/mt.2016.106] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022] Open
Abstract
Chimeric antigen receptor (CAR) endows specificity to T-cells independent of human leukocyte antigen (HLA). This enables one immunoreceptor to directly target the same surface antigen on different subsets of tumor cells from multiple HLA-disparate recipients. Most approaches manufacture individualized CAR(+)T-cells from the recipient or HLA-compatible donor, which are revealing promising clinical results. This is the impetus to broaden the number of patients eligible to benefit from adoptive immunotherapy such as to infuse third-party donor derived CAR(+)T-cells. This will overcome issues associated with (i) time to manufacture T-cells, (ii) cost to generate one product for one patient, (iii) inability to generate a product from lymphopenic patients or patient's immune cells fail to complete the manufacturing process, and (iv) heterogeneity of T-cell products produced for or from individual recipients. Establishing a biobank of allogeneic genetically modified immune cells from healthy third-party donors, which are cryopreserved and validated in advance of administration, will facilitate the centralizing manufacturing and widespread distribution of CAR(+)T-cells to multiple points-of-care in a timely manner. To achieve this, it is necessary to engineer an effective strategy to avoid deleterious allogeneic immune responses leading to toxicity and rejection. We review the strategies to establish "off-the-shelf" donor-derived biobanks for human application of CAR(+)T-cells as a drug.
Collapse
Affiliation(s)
- Hiroki Torikai
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Laurence Jn Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Ziopharm Oncology Inc., Boston, Massachusetts, USA
| |
Collapse
|
329
|
Rufener GA, Press OW, Olsen P, Lee SY, Jensen MC, Gopal AK, Pender B, Budde LE, Rossow JK, Green DJ, Maloney DG, Riddell SR, Till BG. Preserved Activity of CD20-Specific Chimeric Antigen Receptor–Expressing T Cells in the Presence of Rituximab. Cancer Immunol Res 2016; 4:509-19. [DOI: 10.1158/2326-6066.cir-15-0276] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/09/2016] [Indexed: 11/16/2022]
|
330
|
Zah E, Lin MY, Silva-Benedict A, Jensen MC, Chen YY. T Cells Expressing CD19/CD20 Bispecific Chimeric Antigen Receptors Prevent Antigen Escape by Malignant B Cells. Cancer Immunol Res 2016; 4:498-508. [PMID: 27059623 DOI: 10.1158/2326-6066.cir-15-0231] [Citation(s) in RCA: 412] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/06/2016] [Indexed: 01/29/2023]
Abstract
The adoptive transfer of T cells expressing anti-CD19 chimeric antigen receptors (CARs) has shown remarkable curative potential against advanced B-cell malignancies, but multiple trials have also reported patient relapses due to the emergence of CD19-negative leukemic cells. Here, we report the design and optimization of single-chain, bispecific CARs that trigger robust cytotoxicity against target cells expressing either CD19 or CD20, two clinically validated targets for B-cell malignancies. We determined the structural parameters required for efficient dual-antigen recognition, and we demonstrate that optimized bispecific CARs can control both wild-type B-cell lymphoma and CD19(-) mutants with equal efficiency in vivo To our knowledge, this is the first bispecific CAR capable of preventing antigen escape by performing true OR-gate signal computation on a clinically relevant pair of tumor-associated antigens. The CD19-OR-CD20 CAR is fully compatible with existing T-cell manufacturing procedures and implementable by current clinical protocols. These results present an effective solution to the challenge of antigen escape in CD19 CAR T-cell therapy, and they highlight the utility of structure-based rational design in the development of receptors with higher-level complexity. Cancer Immunol Res; 4(6); 498-508. ©2016 AACR
Collapse
Affiliation(s)
- Eugenia Zah
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California
| | - Meng-Yin Lin
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California
| | - Anne Silva-Benedict
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington. Department of Oncology and Hematology, St. Luke's Regional Cancer Center, Duluth, Minnesota
| | - Michael C Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington. Division of Pediatric Hematology-Oncology, University of Washington School of Medicine, Seattle, Washington. Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
331
|
Thomas S, Straathof K, Himoudi N, Anderson J, Pule M. An Optimized GD2-Targeting Retroviral Cassette for More Potent and Safer Cellular Therapy of Neuroblastoma and Other Cancers. PLoS One 2016; 11:e0152196. [PMID: 27030986 PMCID: PMC4816271 DOI: 10.1371/journal.pone.0152196] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/10/2016] [Indexed: 01/22/2023] Open
Abstract
Neuroblastoma is the commonest extra cranial solid cancer of childhood. Despite escalation of treatment regimens, a significant minority of patients die of their disease. Disialoganglioside (GD2) is consistently expressed at high-levels in neuroblastoma tumors, which have been targeted with some success using therapeutic monoclonal antibodies. GD2 is also expressed in a range of other cancer but with the exception of some peripheral nerves is largely absent from non-transformed tissues. Chimeric Antigen Receptors (CARs) are artificial type I proteins which graft the specificity of a monoclonal antibody onto a T-cell. Clinical data with early CAR designs directed against GD2 have shown some promise in Neuroblastoma. Here, we describe a GD2-targeting CAR retroviral cassette, which has been optimized for CAR T-cell persistence, efficacy and safety.
Collapse
Affiliation(s)
- Simon Thomas
- Cancer Institute, University College London, London, United Kingdom
| | - Karin Straathof
- Institute of Child Health, University College London, London, United Kingdom
| | - Nourredine Himoudi
- Institute of Child Health, University College London, London, United Kingdom
| | - John Anderson
- Institute of Child Health, University College London, London, United Kingdom
| | - Martin Pule
- Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
332
|
Luo C, Wei J, Han W. Spotlight on chimeric antigen receptor engineered T cell research and clinical trials in China. SCIENCE CHINA-LIFE SCIENCES 2016; 59:349-59. [PMID: 27009301 DOI: 10.1007/s11427-016-5034-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/19/2016] [Indexed: 01/22/2023]
Abstract
T cell mediated adoptive immune response has been characterized as the key to anti-tumor immunity. Scientists around the world including in China, have been trying to harness the power of T cells against tumors for decades. Recently, the biosynthetic chimeric antigen receptor engineered T cell (CAR-T) strategy was developed and exhibited encouraging clinical efficacy, especially in hematological malignancies. Chimeric antigen receptor research reports began in 2009 in China according to our PubMed search results. Clinical trials have been ongoing in China since 2013 according to the trial registrations on clinicaltrials. gov.. After years of assiduous efforts, research and clinical scientists in China have made their own achievements in the CAR-T therapy field. In this review, we aim to highlight CAR-T research and clinical trials in China, to provide an informative reference for colleagues in the field.
Collapse
Affiliation(s)
- Can Luo
- Institute of Basic Medicine/Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianshu Wei
- Institute of Basic Medicine/Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Weidong Han
- Institute of Basic Medicine/Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
333
|
Holzapfel BM, Wagner F, Thibaudeau L, Levesque JP, Hutmacher DW. Concise review: humanized models of tumor immunology in the 21st century: convergence of cancer research and tissue engineering. Stem Cells 2016; 33:1696-704. [PMID: 25694194 DOI: 10.1002/stem.1978] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 01/17/2014] [Indexed: 12/13/2022]
Abstract
Despite positive testing in animal studies, more than 80% of novel drug candidates fail to proof their efficacy when tested in humans. This is primarily due to the use of preclinical models that are not able to recapitulate the physiological or pathological processes in humans. Hence, one of the key challenges in the field of translational medicine is to "make the model organism mouse more human." To get answers to questions that would be prognostic of outcomes in human medicine, the mouse's genome can be altered in order to create a more permissive host that allows the engraftment of human cell systems. It has been shown in the past that these strategies can improve our understanding of tumor immunology. However, the translational benefits of these platforms have still to be proven. In the 21st century, several research groups and consortia around the world take up the challenge to improve our understanding of how to humanize the animal's genetic code, its cells and, based on tissue engineering principles, its extracellular microenvironment, its tissues, or entire organs with the ultimate goal to foster the translation of new therapeutic strategies from bench to bedside. This article provides an overview of the state of the art of humanized models of tumor immunology and highlights future developments in the field such as the application of tissue engineering and regenerative medicine strategies to further enhance humanized murine model systems.
Collapse
Affiliation(s)
- Boris Michael Holzapfel
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, Australia.,Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Koenig-Ludwig-Haus, Wuerzburg, Germany
| | - Ferdinand Wagner
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, Australia.,Department of Orthopedics, University of Regensburg, Asklepios Klinikum Bad Abbach, Bad Abbach, Germany
| | - Laure Thibaudeau
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, Australia
| | - Jean-Pierre Levesque
- Stem Cell Biology Group, Blood and Bone Diseases Program, Mater Research Institute, The University of Queensland, Woolloongabba, Brisbane, Queensland, Australia
| | - Dietmar Werner Hutmacher
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, Australia.,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,Institute for Advanced Study, Technical University Munich, Garching, Munich, Germany
| |
Collapse
|
334
|
Liu L, Sommermeyer D, Cabanov A, Kosasih P, Hill T, Riddell SR. Inclusion of Strep-tag II in design of antigen receptors for T-cell immunotherapy. Nat Biotechnol 2016; 34:430-4. [PMID: 26900664 PMCID: PMC4940167 DOI: 10.1038/nbt.3461] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 12/15/2015] [Indexed: 02/06/2023]
Abstract
The tactical introduction of Strep-tag II into synthetic antigen
receptors provides engineered T cells with a marker for identification and rapid
purification, and a functional element for selective antibody coated
microbead-driven large-scale expansion. Such receptor designs can be applied to
chimeric antigen receptors of different ligand specificities and costimulatory
domains, and to T cell receptors to facilitate cGMP manufacturing of adoptive T
cell therapies to treat cancer and other diseases.
Collapse
Affiliation(s)
- Lingfeng Liu
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Daniel Sommermeyer
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Alexandra Cabanov
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Paula Kosasih
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Tyler Hill
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Stanley R Riddell
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Institute for Advanced Study, Technical University of Munich, Munich, Germany
| |
Collapse
|
335
|
Lynn RC, Feng Y, Schutsky K, Poussin M, Kalota A, Dimitrov DS, Powell DJ. High-affinity FRβ-specific CAR T cells eradicate AML and normal myeloid lineage without HSC toxicity. Leukemia 2016; 30:1355-64. [PMID: 26898190 PMCID: PMC4889499 DOI: 10.1038/leu.2016.35] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/18/2015] [Accepted: 02/02/2016] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive malignancy, and development of new treatments to prolong remissions is warranted. Chimeric antigen receptor (CAR) T-cell therapies appear promising but on-target, off-tumor recognition of antigen in healthy tissues remains a concern. Here we isolated a high-affinity (HA) folate receptor beta (FRβ)-specific single-chain variable fragment (2.48 nm KD) for optimization of FRβ-redirected CAR T-cell therapy for AML. T cells stably expressing the HA-FRβ CAR exhibited greatly enhanced antitumor activity against FRβ(+) AML in vitro and in vivo compared with a low-affinity FRβ CAR (54.3 nm KD). Using the HA-FRβ immunoglobulin G, FRβ expression was detectable in myeloid-lineage hematopoietic cells; however, expression in CD34(+) hematopoietic stem cells (HSCs) was nearly undetectable. Accordingly, HA-FRβ CAR T cells lysed mature CD14(+) monocytes, while HSC colony formation was unaffected. Because of the potential for elimination of mature myeloid lineage, mRNA CAR electroporation for transient CAR expression was evaluated. mRNA-electroporated HA-FRβ CAR T cells retained effective antitumor activity in vitro and in vivo. Together, our results highlight the importance of antibody affinity in target protein detection and CAR development and suggest that transient delivery of potent HA-FRβ CAR T cells is highly effective against AML and reduces the risk for long-term myeloid toxicity.
Collapse
Affiliation(s)
- R C Lynn
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Feng
- Protein Interactions Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - K Schutsky
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Poussin
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - A Kalota
- Division of Hematology and Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - D S Dimitrov
- Protein Interactions Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - D J Powell
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
336
|
Kim MG, Kim D, Suh SK, Park Z, Choi MJ, Oh YK. Current status and regulatory perspective of chimeric antigen receptor-modified T cell therapeutics. Arch Pharm Res 2016; 39:437-452. [DOI: 10.1007/s12272-016-0719-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/04/2016] [Indexed: 10/22/2022]
|
337
|
Fraietta JA, Schwab RD, Maus MV. Improving therapy of chronic lymphocytic leukemia with chimeric antigen receptor T cells. Semin Oncol 2016; 43:291-9. [PMID: 27040708 DOI: 10.1053/j.seminoncol.2016.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adoptive cell immunotherapy for the treatment of chronic lymphocytic leukemia (CLL) has heralded a new era of synthetic biology. The infusion of genetically engineered, autologous chimeric antigen receptor (CAR) T cells directed against CD19 expressed by normal and malignant B cells represents a novel approach to cancer therapy. The results of recent clinical trials of CAR T cells in relapsed and refractory CLL have demonstrated long-term disease-free remissions, underscoring the power of harnessing and redirecting the immune system against cancer. This review will briefly summarize T-cell therapies in development for CLL disease. We discuss the role of T-cell function and phenotype, T-cell culture optimization, CAR design, and approaches to potentiate the survival and anti-tumor effects of infused lymphocytes. Future efforts will focus on improving the efficacy of CAR T cells for the treatment of CLL and incorporating adoptive cell immunotherapy into standard medical management of CLL.
Collapse
Affiliation(s)
- Joseph A Fraietta
- Center for Cellular Immunotherapy, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Robert D Schwab
- Center for Cellular Immunotherapy, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
338
|
Ma JSY, Kim JY, Kazane SA, Choi SH, Yun HY, Kim MS, Rodgers DT, Pugh HM, Singer O, Sun SB, Fonslow BR, Kochenderfer JN, Wright TM, Schultz PG, Young TS, Kim CH, Cao Y. Versatile strategy for controlling the specificity and activity of engineered T cells. Proc Natl Acad Sci U S A 2016; 113:E450-E458. [PMID: 26759368 DOI: 10.1073/pnas.1524193113/suppl_file/pnas.1524193113.sapp.pdf] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023] Open
Abstract
The adoptive transfer of autologous T cells engineered to express a chimeric antigen receptor (CAR) has emerged as a promising cancer therapy. Despite impressive clinical efficacy, the general application of current CAR-T--cell therapy is limited by serious treatment-related toxicities. One approach to improve the safety of CAR-T cells involves making their activation and proliferation dependent upon adaptor molecules that mediate formation of the immunological synapse between the target cancer cell and T-cell. Here, we describe the design and synthesis of structurally defined semisynthetic adaptors we refer to as "switch" molecules, in which anti-CD19 and anti-CD22 antibody fragments are site-specifically modified with FITC using genetically encoded noncanonical amino acids. This approach allows the precise control over the geometry and stoichiometry of complex formation between CD19- or CD22-expressing cancer cells and a "universal" anti-FITC-directed CAR-T cell. Optimization of this CAR-switch combination results in potent, dose-dependent in vivo antitumor activity in xenograft models. The advantage of being able to titrate CAR-T-cell in vivo activity was further evidenced by reduced in vivo toxicity and the elimination of persistent B-cell aplasia in immune-competent mice. The ability to control CAR-T cell and cancer cell interactions using intermediate switch molecules may expand the scope of engineered T-cell therapy to solid tumors, as well as indications beyond cancer therapy.
Collapse
MESH Headings
- Animals
- Antigens, CD19/immunology
- Antigens, Neoplasm/immunology
- Azides
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Female
- Fluorescein-5-isothiocyanate
- Genetic Vectors
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Lentivirus/genetics
- Leukemia, B-Cell/therapy
- Lymphocyte Activation
- Lymphopenia/etiology
- Lymphopenia/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Models, Molecular
- Phenylalanine/analogs & derivatives
- Protein Conformation
- Protein Engineering/methods
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins/immunology
- Sialic Acid Binding Ig-like Lectin 2/immunology
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- T-Cell Antigen Receptor Specificity
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transduction, Genetic
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jennifer S Y Ma
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - Ji Young Kim
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - Stephanie A Kazane
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - Sei-Hyun Choi
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Hwa Young Yun
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Min Soo Kim
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - David T Rodgers
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - Holly M Pugh
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - Oded Singer
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - Sophie B Sun
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - Bryan R Fonslow
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037; SCIEX Separations, Brea, CA 92821
| | - James N Kochenderfer
- Experimental Transplantation and Immunology Branch, National Institutes of Health, National Cancer Institute, Bethesda, MD 20892
| | - Timothy M Wright
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037
| | - Peter G Schultz
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037; Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037;
| | - Travis S Young
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037;
| | - Chan Hyuk Kim
- Department of Biology, California Institute for Biomedical Research, La Jolla, CA 92037;
| | - Yu Cao
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
339
|
Versatile strategy for controlling the specificity and activity of engineered T cells. Proc Natl Acad Sci U S A 2016; 113:E450-8. [PMID: 26759368 DOI: 10.1073/pnas.1524193113] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The adoptive transfer of autologous T cells engineered to express a chimeric antigen receptor (CAR) has emerged as a promising cancer therapy. Despite impressive clinical efficacy, the general application of current CAR-T--cell therapy is limited by serious treatment-related toxicities. One approach to improve the safety of CAR-T cells involves making their activation and proliferation dependent upon adaptor molecules that mediate formation of the immunological synapse between the target cancer cell and T-cell. Here, we describe the design and synthesis of structurally defined semisynthetic adaptors we refer to as "switch" molecules, in which anti-CD19 and anti-CD22 antibody fragments are site-specifically modified with FITC using genetically encoded noncanonical amino acids. This approach allows the precise control over the geometry and stoichiometry of complex formation between CD19- or CD22-expressing cancer cells and a "universal" anti-FITC-directed CAR-T cell. Optimization of this CAR-switch combination results in potent, dose-dependent in vivo antitumor activity in xenograft models. The advantage of being able to titrate CAR-T-cell in vivo activity was further evidenced by reduced in vivo toxicity and the elimination of persistent B-cell aplasia in immune-competent mice. The ability to control CAR-T cell and cancer cell interactions using intermediate switch molecules may expand the scope of engineered T-cell therapy to solid tumors, as well as indications beyond cancer therapy.
Collapse
|
340
|
Chimeric antigen receptor-redirected T cells return to the bench. Semin Immunol 2016; 28:3-9. [PMID: 26797495 DOI: 10.1016/j.smim.2015.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/07/2015] [Accepted: 12/07/2015] [Indexed: 12/29/2022]
Abstract
While the clinical progress of chimeric antigen receptor T cell (CAR-T) immunotherapy has garnered attention to the field, our understanding of the biology of these chimeric molecules is still emerging. Our aim within this review is to bring to light the mechanistic understanding of these multi-modular receptors and how these individual components confer particular properties to CAR-Ts. In addition, we will discuss extrinsic factors that can be manipulated to influence CAR-T performance such as choice of cellular population, culturing conditions and additional modifications that enhance their activity particularly in solid tumors. Finally, we will also consider the emerging toxicity associated with CAR-Ts. By breaking apart the CAR and examining the role of each piece, we can build a better functioning cellular vehicle for optimized treatment of cancer patients.
Collapse
|
341
|
Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies. Proc Natl Acad Sci U S A 2016; 113:E459-68. [PMID: 26759369 DOI: 10.1073/pnas.1524155113] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has produced impressive results in clinical trials for B-cell malignancies. However, safety concerns related to the inability to control CAR-T cells once infused into the patient remain a significant challenge. Here we report the engineering of recombinant antibody-based bifunctional switches that consist of a tumor antigen-specific Fab molecule engrafted with a peptide neo-epitope, which is bound exclusively by a peptide-specific switchable CAR-T cell (sCAR-T). The switch redirects the activity of the bio-orthogonal sCAR-T cells through the selective formation of immunological synapses, in which the sCAR-T cell, switch, and target cell interact in a structurally defined and temporally controlled manner. Optimized switches specific for CD19 controlled the activity, tissue-homing, cytokine release, and phenotype of sCAR-T cells in a dose-titratable manner in a Nalm-6 xenograft rodent model of B-cell leukemia. The sCAR-T-cell dosing regimen could be tuned to provide efficacy comparable to the corresponding conventional CART-19, but with lower cytokine levels, thereby offering a method of mitigating cytokine release syndrome in clinical translation. Furthermore, we demonstrate that this methodology is readily adaptable to targeting CD20 on cancer cells using the same sCAR-T cell, suggesting that this approach may be broadly applicable to heterogeneous and resistant tumor populations, as well as other liquid and solid tumor antigens.
Collapse
|
342
|
Yazdanifar M, Zhou R, Mukherjee P. Emerging immunotherapeutics in adenocarcinomas: A focus on CAR-T cells. CURRENT TRENDS IN IMMUNOLOGY 2016; 17:95-115. [PMID: 28659689 PMCID: PMC5484157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
More than 80% of all cancers arise from epithelial cells referred to as carcinomas. Adenocarcinomas are the most common type of carcinomas arising from the specialized epithelial cells that line the ducts of our major organs. Despite many advances in cancer therapies, metastatic and treatment-refractory cancers remain the 2nd leading cause of death. Immunotherapy has offered potential opportunities with specific targeting of tumor cells and inducing remission in many cancer patients. Numerous therapies using antibodies as antagonists or checkpoint inhibitors/immune modulators, peptide or cell vaccines, cytokines, and adoptive T cell therapies have been developed. The most innovative immunotherapy approach so far has been the use of engineered T cell, also referred to as chimeric antigen receptor T cells (CAR-T cells). CAR-T cells are genetically modified naïve T cells that express a chimeric molecule which comprises of the antigen-recognition domains (scFv) of an anti-tumor antibody and one, two, or three intracellular signaling domains of the T cell receptor (TCR). When these engineered T cells recognize and bind to the tumor antigen target via the scFv fragment, a signal is sent to the intracellular TCR domains of the CAR, leading to activation of the T cells to become cytolytic against the tumor cells. CAR-T cell therapy has shown tremendous success for certain hematopoietic malignancies, but this success has not been extrapolated to adenocarcinomas. This is due to multiple factors associated with adenocarcinoma that are different from hematopoietic tumors. Although many advances have been made in targeting multiple cancers by CAR-T cells, clinical trials have shown adverse effects and toxicity related to this treatment. New strategies are yet to be devised to manage side effects associated with CAR-T cell therapies. In this review, we report some of the promising immunotherapeutic strategies being developed for treatment of most common adenocarcinomas with particular emphasis on the future generation of CAR-T cell therapy.
Collapse
Affiliation(s)
| | | | - Pinku Mukherjee
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Boulevard, Charlotte, NC 28223, USA
| |
Collapse
|
343
|
Abstract
Plasmid DNA is being used as a pharmaceutical agent in vaccination, as well as a basic substance and starting material in gene and cell therapy, and viral vector production. Since the uncontrolled expression of backbone sequences present in such plasmids and the dissemination of antibiotic resistance genes may have profound detrimental effects, an important goal in vector development was to produce supercoiled DNA lacking bacterial backbone sequences: Minicircle (MC) DNA. The Sleeping Beauty (SB) transposon system is a non-viral gene delivery platform enabling a close-to-random profile of genomic integration. In combination, the MC platform greatly enhances SB transposition and transgene integration resulting in higher numbers of stably modified target cells. We have recently developed a strategy for MC-based SB transposition of chimeric antigen receptor (CAR) transgenes that enable improved transposition rates compared to conventional plasmids and rapid manufacturing of therapeutic CAR T cell doses (Monjezi et al. 2016). This advance enables manufacturing CAR T cells in a virus-free process that relies on SB-mediated transposition from MC DNA to accomplish gene-transfer. Advantages of this approach include a strong safety profile due to the nature of the MC itself and the genomic insertion pattern of MC-derived CAR transposons. In addition, stable transposition and high-level CAR transgene expression, as well as easy and reproducible handling, make MCs a preferred vector source for gene-transfer in advanced cellular and gene therapy. In this chapter, we will review our experience in MC-based CAR T cell engineering and discuss our recent advances in MC manufacturing to accelerate both pre-clinical and clinical implementation.
Collapse
|
344
|
Norelli M, Casucci M, Bonini C, Bondanza A. Clinical pharmacology of CAR-T cells: Linking cellular pharmacodynamics to pharmacokinetics and antitumor effects. Biochim Biophys Acta Rev Cancer 2015; 1865:90-100. [PMID: 26748354 DOI: 10.1016/j.bbcan.2015.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 12/14/2015] [Accepted: 12/18/2015] [Indexed: 12/01/2022]
Abstract
Adoptive cell transfer of T cells genetically modified with tumor-reactive chimeric antigen receptors (CARs) is a rapidly emerging field in oncology, which in preliminary clinical trials has already shown striking antitumor efficacy. Despite these premises, there are still a number of open issues related to CAR-T cells, spanning from their exact mechanism of action (pharmacodynamics), to the factors associated with their in vivo persistence (pharmacokinetics), and, finally, to the relative contribution of each of the two in determining the antitumor effects and accompanying toxicities. In light of the unprecedented curative potential of CAR-T cells and of their predicted wide availability in the next few years, in this review we will summarize the current knowledge on the clinical pharmacology aspects of what is anticipated to be a brand new class of biopharmaceuticals to join the therapeutic armamentarium of cancer doctors.
Collapse
Affiliation(s)
- M Norelli
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Hospital Scientific Institute, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - M Casucci
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Hospital Scientific Institute, Milano, Italy
| | - C Bonini
- Vita-Salute San Raffaele University, Milano, Italy; Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Hospital Scientific Institute, Milano, Italy
| | - A Bondanza
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Hospital Scientific Institute, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy.
| |
Collapse
|
345
|
Ji Y, Hocker JD, Gattinoni L. Enhancing adoptive T cell immunotherapy with microRNA therapeutics. Semin Immunol 2015; 28:45-53. [PMID: 26710685 DOI: 10.1016/j.smim.2015.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 12/15/2022]
Abstract
Adoptive T cell-based immunotherapies can mediate complete and durable regressions in patients with advanced cancer, but current response rates remain inadequate. Maneuvers to improve the fitness and antitumor efficacy of transferred T cells have been under extensive exploration in the field. Small non-coding microRNAs have emerged as critical modulators of immune system homeostasis and T cell immunity. Here, we summarize recent advances in our understanding of the role of microRNAs in regulating T cell activation, differentiation, and function. We also discuss how microRNA therapeutics could be employed to fine-tune T cell receptor signaling and enhance T cell persistence and effector functions, paving the way for the next generation of adoptive immunotherapies.
Collapse
Affiliation(s)
- Yun Ji
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA.
| | - James D Hocker
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA
| | - Luca Gattinoni
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA.
| |
Collapse
|
346
|
Harris DT, Kranz DM. Adoptive T Cell Therapies: A Comparison of T Cell Receptors and Chimeric Antigen Receptors. Trends Pharmacol Sci 2015; 37:220-230. [PMID: 26705086 DOI: 10.1016/j.tips.2015.11.004] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/23/2015] [Accepted: 11/12/2015] [Indexed: 01/04/2023]
Abstract
The tumor-killing properties of T cells provide tremendous opportunities to treat cancer. Adoptive T cell therapies have begun to harness this potential by endowing a functionally diverse repertoire of T cells with genetically modified, tumor-specific recognition receptors. Normally, this antigen recognition function is mediated by an αβ T cell receptor (TCR), but the dominant therapeutic forms currently in development are synthetic constructs called chimeric antigen receptors (CARs). While CAR-based adoptive cell therapies are already showing great promise, their basic mechanistic properties have been studied in less detail compared with those of αβ TCRs. In this review, we compare and contrast various features of TCRs versus CARs, with a goal of highlighting issues that need to be addressed to fully exploit the therapeutic potential of both.
Collapse
Affiliation(s)
- Daniel T Harris
- Department of Biochemistry, University of Illinois, 600 S. Matthews Avenue, Urbana, IL 61801, USA
| | - David M Kranz
- Department of Biochemistry, University of Illinois, 600 S. Matthews Avenue, Urbana, IL 61801, USA.
| |
Collapse
|
347
|
Chimeric Antigen Receptors for Cancer: Progress and Challenges. CURRENT STEM CELL REPORTS 2015. [DOI: 10.1007/s40778-015-0026-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
348
|
Liu X, Jiang S, Fang C, Yang S, Olalere D, Pequignot EC, Cogdill AP, Li N, Ramones M, Granda B, Zhou L, Loew A, Young RM, June CH, Zhao Y. Affinity-Tuned ErbB2 or EGFR Chimeric Antigen Receptor T Cells Exhibit an Increased Therapeutic Index against Tumors in Mice. Cancer Res 2015; 75:3596-607. [PMID: 26330166 DOI: 10.1158/0008-5472.can-15-0159] [Citation(s) in RCA: 388] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Target-mediated toxicity is a major limitation in the development of chimeric antigen T-cell receptors (CAR) for adoptive cell therapy of solid tumors. In this study, we developed a strategy to adjust the affinities of the scFv component of CAR to discriminate tumors overexpressing the target from normal tissues that express it at physiologic levels. A CAR-expressing T-cell panel was generated with target antigen affinities varying over three orders of magnitude. High-affinity cells recognized target expressed at any level, including at levels in normal cells that were undetectable by flow cytometry. Affinity-tuned cells exhibited robust antitumor efficacy similar to high-affinity cells, but spared normal cells expressing physiologic target levels. The use of affinity-tuned scFvs offers a strategy to empower wider use of CAR T cells against validated targets widely overexpressed on solid tumors, including those considered undruggable by this approach.
Collapse
Affiliation(s)
- Xiaojun Liu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shuguang Jiang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chongyun Fang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shiyu Yang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Devvora Olalere
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward C Pequignot
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Na Li
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Melissa Ramones
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Brian Granda
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Li Zhou
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Andreas Loew
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Regina M Young
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carl H June
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Yangbing Zhao
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
349
|
Caruso HG, Hurton LV, Najjar A, Rushworth D, Ang S, Olivares S, Mi T, Switzer K, Singh H, Huls H, Lee DA, Heimberger AB, Champlin RE, Cooper LJN. Tuning Sensitivity of CAR to EGFR Density Limits Recognition of Normal Tissue While Maintaining Potent Antitumor Activity. Cancer Res 2015; 75:3505-18. [PMID: 26330164 DOI: 10.1158/0008-5472.can-15-0139] [Citation(s) in RCA: 296] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Many tumors overexpress tumor-associated antigens relative to normal tissue, such as EGFR. This limits targeting by human T cells modified to express chimeric antigen receptors (CAR) due to potential for deleterious recognition of normal cells. We sought to generate CAR(+) T cells capable of distinguishing malignant from normal cells based on the disparate density of EGFR expression by generating two CARs from monoclonal antibodies that differ in affinity. T cells with low-affinity nimotuzumab-CAR selectively targeted cells overexpressing EGFR, but exhibited diminished effector function as the density of EGFR decreased. In contrast, the activation of T cells bearing high-affinity cetuximab-CAR was not affected by the density of EGFR. In summary, we describe the generation of CARs able to tune T-cell activity to the level of EGFR expression in which a CAR with reduced affinity enabled T cells to distinguish malignant from nonmalignant cells.
Collapse
Affiliation(s)
- Hillary G Caruso
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Lenka V Hurton
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Amer Najjar
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Rushworth
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Sonny Ang
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simon Olivares
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tiejuan Mi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kirsten Switzer
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Harjeet Singh
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Helen Huls
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dean A Lee
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laurence J N Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas. The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas.
| |
Collapse
|
350
|
Characterization and Functional Analysis of scFv-based Chimeric Antigen Receptors to Redirect T Cells to IL13Rα2-positive Glioma. Mol Ther 2015; 24:354-363. [PMID: 26514825 DOI: 10.1038/mt.2015.199] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/23/2015] [Indexed: 12/30/2022] Open
Abstract
Immunotherapy with T cells expressing chimeric antigen receptors (CARs) is an attractive approach to improve outcomes for patients with glioblastoma (GBM). IL13Rα2 is expressed at a high frequency in GBM but not in normal brain, making it a promising CAR T-cell therapy target. IL13Rα2-specific CARs generated up to date contain mutated forms of IL13 as an antigen-binding domain. While these CARs target IL13Rα2, they also recognize IL13Rα1, which is broadly expressed. To overcome this limitation, we constructed a panel of IL13Rα2-specific CARs that contain the IL13Rα2-specific single-chain variable fragment (scFv) 47 as an antigen binding domain, short or long spacer regions, a transmembrane domain, and endodomains derived from costimulatory molecules and CD3.ζ (IL13Rα2-CARs). IL13Rα2-CAR T cells recognized IL13Rα2-positive target cells in coculture and cytotoxicity assays with no cross-reactivity to IL13Rα1. However, only IL13Rα2-CAR T cells with a short spacer region produced IL2 in an antigen-dependent fashion. In vivo, T cells expressing IL13Rα2-CARs with short spacer regions and CD28.ζ, 41BB.ζ, and CD28.OX40.ζ endodomains had potent anti-glioma activity conferring a significant survival advantage in comparison to mice that received control T cells. Thus, IL13Rα2-CAR T cells hold the promise to improve current IL13Rα2-targeted immunotherapy approaches for GBM and other IL13Rα2-positive malignancies.
Collapse
|