1
|
Ali A, Contreras P, Darweesh M, Andersson L, Jin C, Essand M, Yu D. Targeting ZC3H11A elicits immunogenic cancer cell death through augmentation of antigen presentation and interferon response. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102361. [PMID: 39582529 PMCID: PMC11585804 DOI: 10.1016/j.omtn.2024.102361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024]
Abstract
Zinc finger CCCH containing 11A (ZC3H11A) is a stress-induced protein that is upregulated in various conditions such as heat shock and virus infection. It has also been reported to be upregulated in certain cancers. The aim of this study was to evaluate the feasibility of targeting ZC3H11A as a therapeutic approach for cancer treatment, using nuclease-resistant, affinity-enhanced antisense oligonucleotide (ASO). An ASO targeting ZC3H11A was validated and evaluated in vitro and in the B16 melanoma model in vivo. Antigen presentation, interferon response, cell proliferation, and apoptosis were transcriptionally affected. These findings were validated on the protein level by the upregulation of major histocompatibility complex class I (MHC class I), an increased secretion of interferon-β (IFN-β), and induction of apoptosis observed as upregulation of caspases and annexin V. Immunogenic features of the induced apoptosis were evidenced by the surface exposure of calreticulin (CRT) and the secretion of ATP leading to enhanced dendritic cell (DC) phagocytosis, maturation, and activation. Treatment with the ZC3H11A-targeted ASO had limited efficacy in vivo, while constitutive lentiviral shRNA knockdown of ZC3H11A in murine B16 melanoma cells and human HeLa cells led to reduced tumor growth with prolonged survival of mice, validating ZC3H11A as a relevant target for cancer therapy.
Collapse
Affiliation(s)
- Arwa Ali
- Department of Immunology, Genetics, and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Paola Contreras
- Department of Immunology, Genetics, and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Mahmoud Darweesh
- Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23 Uppsala, Sweden
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhr University, Assiut 71526, Egypt
- Immunolgy Laboratory, Natural and Medical Sciences Research Centre (NMSRC), University of Nizwa, PO.Box:33, P.C. 616 Nizwa, Oman
| | - Leif Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23 Uppsala, Sweden
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Chuan Jin
- Department of Immunology, Genetics, and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics, and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Di Yu
- Department of Immunology, Genetics, and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
2
|
Sarén T, Saronio G, Marti Torrell P, Zhu X, Thelander J, Andersson Y, Hofström C, Nestor M, Dimberg A, Persson H, Ramachandran M, Yu D, Essand M. Complementarity-determining region clustering may cause CAR-T cell dysfunction. Nat Commun 2023; 14:4732. [PMID: 37563127 PMCID: PMC10415375 DOI: 10.1038/s41467-023-40303-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is rapidly advancing as cancer treatment, however, designing an optimal CAR remains challenging. A single-chain variable fragment (scFv) is generally used as CAR targeting moiety, wherein the complementarity-determining regions (CDRs) define its specificity. We report here that the CDR loops can cause CAR clustering, leading to antigen-independent tonic signalling and subsequent CAR-T cell dysfunction. We show via CARs incorporating scFvs with identical framework and varying CDR sequences that CARs may cluster on the T cell surface, which leads to antigen-independent CAR-T cell activation, characterized by increased cell size and interferon (IFN)-γ secretion. This results in CAR-T cell exhaustion, activation-induced cell death and reduced responsiveness to target-antigen-expressing tumour cells. CDR mutagenesis confirms that the CAR-clustering is mediated by CDR-loops. In summary, antigen-independent tonic signalling can be induced by CDR-mediated CAR clustering, which could not be predicted from the scFv sequences, but could be tested for by evaluating the activity of unstimulated CAR-T cells.
Collapse
Affiliation(s)
- Tina Sarén
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden
| | - Giulia Saronio
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden
| | - Paula Marti Torrell
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden
| | - Xu Zhu
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden
| | - Josefin Thelander
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden
| | - Yasmin Andersson
- Royal Institute of Technology (KTH), Drug Discovery and Development Platform, Science for Life Laboratory, Solna, Sweden
| | - Camilla Hofström
- Royal Institute of Technology (KTH), Drug Discovery and Development Platform, Science for Life Laboratory, Solna, Sweden
| | - Marika Nestor
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden
| | - Anna Dimberg
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden
| | - Helena Persson
- Royal Institute of Technology (KTH), Drug Discovery and Development Platform, Science for Life Laboratory, Solna, Sweden
| | - Mohanraj Ramachandran
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden
| | - Di Yu
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden.
| | - Magnus Essand
- Uppsala University, Dept Immunology, Genetics, Pathology, Science for Life Laboratory, Uppsala, Sweden.
| |
Collapse
|
3
|
Ramos‑Cardona X, Luo W, Mohammed S. Advances and challenges of CAR T therapy and suitability of animal models (Review). Mol Clin Oncol 2022; 17:134. [PMID: 35949897 PMCID: PMC9353808 DOI: 10.3892/mco.2022.2567] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/06/2022] [Indexed: 11/06/2022] Open
Abstract
Chimeric antigen receptors (CARs) recently gained momentum in cancer treatment due to their ability to promote T-cell mediated responses to a specific tumor-associated antigen. CARs are part of the adoptive cell transfer (ACT) strategies that utilize patients' T lymphocytes, genetically engineered to kill cancer cells. However, despite the therapy's success against blood-related malignancies, treating solid tumors has not reached its fullest potential yet. The reasons include the complex suppressive tumor microenvironment, mutations on cancer cells' target receptors, lethal side-effects, restricted trafficking into the tumor, suboptimal persistence in vivo and the lack of animal models that faithfully resemble human tumor's immunological responses. Currently, rodent models are used to investigate the safety and efficacy of CAR therapies. However, these models are limited in representing the human disease faithfully, fail to predict the adverse treatment events and overestimate the efficacy of the therapy. On the other hand, spontaneously developed tumors in dogs are more suited in CAR research and their efficacy has been demonstrated in a number of diseases, including lymphoma, osteosarcoma and mammary tumors. The present review discusses the design and evolution of CARs, challenges of CAR in solid tumors, human and canine clinical trials and advantages of the canine model.
Collapse
Affiliation(s)
- Xavier Ramos‑Cardona
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA
| | - Weichuan Luo
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sulma Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA
| |
Collapse
|
4
|
CAR T cells expressing a bacterial virulence factor trigger potent bystander antitumour responses in solid cancers. Nat Biomed Eng 2022; 6:830-841. [PMID: 35379957 PMCID: PMC9288934 DOI: 10.1038/s41551-022-00875-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 02/24/2022] [Indexed: 02/05/2023]
Abstract
Chimeric antigen receptor T cells (CAR T cells) are effective against haematologic malignancies. However, in solid tumours, their potency is hampered by local immunosuppression and by the heterogeneous expression of the antigen that the CAR targets. Here we show that CAR T cells expressing a pluripotent pro-inflammatory neutrophil-activating protein (NAP) from Helicobacter pylori trigger endogenous bystander T-cell responses against solid cancers. In mice with subcutaneous murine pancreatic ductal adenocarcinomas, neuroblastomas or colon carcinomas, CAR(NAP) T cells led to slower tumour growth and higher survival rates than conventional mouse CAR T cells, regardless of target antigen, tumour type and host haplotype. In tumours with heterogeneous antigen expression, NAP secretion induced the formation of an immunologically 'hot' microenvironment that supported dendritic cell maturation and bystander responses, as indicated by epitope spreading and infiltration of cytotoxic CD8+ T cells targeting tumour-associated antigens other than the CAR-targeted antigen. CAR T cells armed with NAP neither increased off-tumour toxicity nor hampered the efficacy of CAR T cells, and hence may have advantageous translational potential.
Collapse
|
5
|
Prostate Cancer Biomarkers: From diagnosis to prognosis and precision-guided therapeutics. Pharmacol Ther 2021; 228:107932. [PMID: 34174272 DOI: 10.1016/j.pharmthera.2021.107932] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/23/2022]
Abstract
Prostate cancer (PCa) is one of the most commonly diagnosed malignancies and among the leading causes of cancer-related death worldwide. It is a highly heterogeneous disease, ranging from remarkably slow progression or inertia to highly aggressive and fatal disease. As therapeutic decision-making, clinical trial design and outcome highly depend on the appropriate stratification of patients to risk groups, it is imperative to differentiate between benign versus more aggressive states. The incorporation of clinically valuable prognostic and predictive biomarkers is also potentially amenable in this process, in the timely prevention of metastatic disease and in the decision for therapy selection. This review summarizes the progress that has so far been made in the identification of the genomic events that can be used for the classification, prediction and prognostication of PCa, and as major targets for clinical intervention. We include an extensive list of emerging biomarkers for which there is enough preclinical evidence to suggest that they may constitute crucial targets for achieving significant advances in the management of the disease. Finally, we highlight the main challenges that are associated with the identification of clinically significant PCa biomarkers and recommend possible ways to overcome such limitations.
Collapse
|
6
|
Vanhooren J, Derpoorter C, Depreter B, Deneweth L, Philippé J, De Moerloose B, Lammens T. TARP as antigen in cancer immunotherapy. Cancer Immunol Immunother 2021; 70:3061-3068. [PMID: 34050774 PMCID: PMC8164403 DOI: 10.1007/s00262-021-02972-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022]
Abstract
In recent decades, immunotherapy has become a pivotal element in cancer treatment. A remaining challenge is the identification of cancer-associated antigens suitable as targets for immunotherapeutics with potent on-target and few off-tumor effects. The T-cell receptor gamma (TCRγ) chain alternate reading frame protein (TARP) was first discovered in the human prostate and androgen-sensitive prostate cancer. Thereafter, TARP was also identified in breast and endometrial cancers, salivary gland tumors, and pediatric and adult acute myeloid leukemia. Interestingly, TARP promotes tumor cell proliferation and migration, which is reflected in an association with worse survival. TARP expression in malignant cells, its role in oncogenesis, and its limited expression in normal tissues raised interest in its potential utility as a therapeutic target, and led to development of immunotherapeutic targeting strategies. In this review, we provide an overview of TARP expression, its role in different cancer types, and currently investigated TARP-directed immunotherapeutic options.
Collapse
Affiliation(s)
- Jolien Vanhooren
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium. .,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium. .,Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Charlotte Derpoorter
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Barbara Depreter
- Department of Haematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Larissa Deneweth
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jan Philippé
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Tim Lammens
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
7
|
Comparative analysis of assays to measure CAR T-cell-mediated cytotoxicity. Nat Protoc 2021; 16:1331-1342. [PMID: 33589826 DOI: 10.1038/s41596-020-00467-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023]
Abstract
The antitumor efficacy of genetically engineered 'living drugs', including chimeric antigen receptor and T-cell receptor T cells, is influenced by their activation, proliferation, inhibition, and exhaustion. A sensitive and reproducible cytotoxicity assay that collectively reflects these functions is an essential requirement for translation of these cellular therapeutic agents. Here, we compare various in vitro cytotoxicity assays (including chromium release, bioluminescence, impedance, and flow cytometry) with respect to their experimental setup, appropriate uses, advantages, and disadvantages, and measures to overcome their limitations. We also highlight the US Food and Drug Administration (FDA) directives for a potency assay for release of clinical cell therapy products. In addition, we discuss advanced assays of repeated antigen exposure and simultaneous testing of combinations of immune effector cells, immunomodulatory antibodies, and targets with variable antigen expression. This review article should help to equip investigators with the necessary knowledge to select appropriate cytotoxicity assays to test the efficacy of immunotherapeutic agents alone or in combination.
Collapse
|
8
|
Prokhnevska N, Emerson DA, Kissick HT, Redmond WL. Immunological Complexity of the Prostate Cancer Microenvironment Influences the Response to Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1210:121-147. [PMID: 31900908 DOI: 10.1007/978-3-030-32656-2_7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate cancer is one of the most common cancers in men and a leading cause of cancer-related death. Recent advances in the treatment of advanced prostate cancer, including the use of more potent and selective inhibitors of the androgen signaling pathway, have provided significant clinical benefit for men with metastatic castration-resistant prostate cancer (mCRPC). However, most patients develop progressive lethal disease, highlighting the need for more effective treatments. One such approach is immunotherapy, which harness the power of the patient's immune system to identify and destroy cancer cells through the activation of cytotoxic CD8 T cells specific for tumor antigens. Although immunotherapy, particularly checkpoint blockade, can induce significant clinical responses in patients with solid tumors or hematological malignancies, minimal efficacy has been observed in men with mCRPC. In the current review, we discuss our current understanding of the immunological complexity of the immunosuppressive prostate cancer microenvironment, preclinical models of prostate cancer, and recent advances in immunotherapy clinical trials to improve outcomes for men with mCRPC.
Collapse
Affiliation(s)
| | - Dana A Emerson
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA.,Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA.
| |
Collapse
|
9
|
Čančer M, Hutter S, Holmberg KO, Rosén G, Sundström A, Tailor J, Bergström T, Garancher A, Essand M, Wechsler-Reya RJ, Falk A, Weishaupt H, Swartling FJ. Humanized Stem Cell Models of Pediatric Medulloblastoma Reveal an Oct4/mTOR Axis that Promotes Malignancy. Cell Stem Cell 2019; 25:855-870.e11. [PMID: 31786016 PMCID: PMC6900751 DOI: 10.1016/j.stem.2019.10.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 06/26/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022]
Abstract
Medulloblastoma (MB), the most frequent malignant childhood brain tumor, can arise from cellular malfunctions during hindbrain development. Here we generate humanized models for Sonic Hedgehog (SHH)-subgroup MB via MYCN overexpression in primary human hindbrain-derived neuroepithelial stem (hbNES) cells or iPSC-derived NES cells, which display a range of aggressive phenotypes upon xenografting. iPSC-derived NES tumors develop quickly with leptomeningeal dissemination, whereas hbNES-derived cells exhibit delayed tumor formation with less dissemination. Methylation and expression profiling show that tumors from both origins recapitulate hallmarks of infant SHH MB and reveal that mTOR activation, as a result of increased Oct4, promotes aggressiveness of human SHH tumors. Targeting mTOR decreases cell viability and prolongs survival, showing the utility of these varied models for dissecting mechanisms mediating tumor aggression and demonstrating the value of humanized models for a better understanding of pediatric cancers. Human iPSC-derived or primary neuroepithelial stem cells can be transformed by MYCN MYCN drives infant SHH medulloblastoma with clinically relevant features Epigenetically regulated Oct4 promotes mTOR hyperactivation in infant SHH tumors mTOR inhibition efficiently targets metastatic SHH medulloblastoma models and PDXs
Collapse
Affiliation(s)
- Matko Čančer
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Sonja Hutter
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Karl O Holmberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Gabriela Rosén
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Anders Sundström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Jignesh Tailor
- Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Tobias Bergström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Alexandra Garancher
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 92037 La Jolla, CA, USA
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 92037 La Jolla, CA, USA
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Holger Weishaupt
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| |
Collapse
|
10
|
Depreter B, Weening KE, Vandepoele K, Essand M, De Moerloose B, Themeli M, Cloos J, Hanekamp D, Moors I, D'hont I, Denys B, Uyttebroeck A, Van Damme A, Dedeken L, Snauwaert S, Goetgeluk G, De Munter S, Kerre T, Vandekerckhove B, Lammens T, Philippé J. TARP is an immunotherapeutic target in acute myeloid leukemia expressed in the leukemic stem cell compartment. Haematologica 2019; 105:1306-1316. [PMID: 31371409 PMCID: PMC7193481 DOI: 10.3324/haematol.2019.222612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022] Open
Abstract
Immunotherapeutic strategies targeting the rare leukemic stem cell compartment might provide salvage to the high relapse rates currently observed in acute myeloid leukemia (AML). We applied gene expression profiling for comparison of leukemic blasts and leukemic stem cells with their normal counterparts. Here, we show that the T-cell receptor γ chain alternate reading frame protein (TARP) is over-expressed in de novo pediatric (n=13) and adult (n=17) AML sorted leukemic stem cells and blasts compared to hematopoietic stem cells and normal myeloblasts (15 healthy controls). Moreover, TARP expression was significantly associated with a fms-like tyrosine kinase receptor-3 internal tandem duplication in pediatric AML. TARP overexpression was confirmed in AML cell lines (n=9), and was found to be absent in B-cell acute lymphocytic leukemia (n=5) and chronic myeloid leukemia (n=1). Sequencing revealed that both a classical TARP transcript, as described in breast and prostate adenocarcinoma, and an AML-specific alternative TARP transcript, were present. Protein expression levels mostly matched transcript levels. TARP was shown to reside in the cytoplasmic compartment and showed sporadic endoplasmic reticulum co-localization. TARP-T-cell receptor engineered cytotoxic T-cells in vitro killed AML cell lines and patient leukemic cells co-expressing TARP and HLA-A*0201. In conclusion, TARP qualifies as a relevant target for immunotherapeutic T-cell therapy in AML.
Collapse
Affiliation(s)
- Barbara Depreter
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Karin E Weening
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Karl Vandepoele
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Magnus Essand
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Barbara De Moerloose
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Maria Themeli
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Diana Hanekamp
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ine Moors
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Inge D'hont
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Barbara Denys
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Anne Uyttebroeck
- Department of Pediatrics, University Hospital Gasthuisberg, Louvain, Belgium
| | - An Van Damme
- Department of Pediatric Hematology Oncology, University Hospital Saint-Luc, Brussels, Belgium
| | - Laurence Dedeken
- Department of Pediatric Hematology Oncology, Queen Fabiola Children's University Hospital, Brussels, Belgium
| | - Sylvia Snauwaert
- Department of Hematology, AZ Sint-Jan Hospital Bruges, Bruges, Belgium
| | - Glenn Goetgeluk
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Stijn De Munter
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tessa Kerre
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Bart Vandekerckhove
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tim Lammens
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium .,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Jan Philippé
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
11
|
Papanicolau-Sengos A, Yang Y, Pabla S, Lenzo FL, Kato S, Kurzrock R, DePietro P, Nesline M, Conroy J, Glenn S, Chatta G, Morrison C. Identification of targets for prostate cancer immunotherapy. Prostate 2019; 79:498-505. [PMID: 30614027 DOI: 10.1002/pros.23756] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/29/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND We performed profiling of the immune microenvironment of castration-resistant (CRPC) and castration-sensitive (CSPC) prostate cancer (PC) in order to identify novel targets for immunotherapy. METHODS PD-L1 and CD3/CD8 immunohistochemistry, PD-L1/2 fluorescent in situ hybridization, tumor mutation burden, microsatellite instability, and RNA-seq of 395 immune-related genes were performed in 19 CRPC and CSPC. Targeted genomic sequencing and fusion analysis were performed in 17 of these specimens. RESULTS CD276, PVR, and NECTIN2 were highly expressed in PC. Comparison of CRPC versus CSPC and primary versus metastatic tissue revealed the differential expression of immunostimulatory, immunosuppressive, and epithelial-to-mesenchymal transition (EMT)-related genes. Unsupervised clustering of differentially expressed genes yielded two final clusters best segregated by CRPC and CSPC status. CONCLUSION CD276 and the alternative checkpoint inhibition PVR/NECTIN2/CD226/TIGIT pathway emerged as relevant to PC checkpoint inhibition target development.
Collapse
Affiliation(s)
| | - Yuanquan Yang
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | | | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | | | | | - Jeffrey Conroy
- OmniSeq, Inc., Buffalo, New York
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Sean Glenn
- OmniSeq, Inc., Buffalo, New York
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Carl Morrison
- OmniSeq, Inc., Buffalo, New York
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
12
|
Zhang R, Zhang Y, Hu J, Wu W, Chen X, Lu Z, Yang R, Huang Y, Fan J. Specific T-cell receptor gene transfer enhances immune response: A potential therapeutic strategy for the control of human cytomegalovirus infection in immunocompromised patients. Cell Immunol 2019; 336:58-65. [PMID: 30626494 DOI: 10.1016/j.cellimm.2018.12.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/20/2018] [Accepted: 12/31/2018] [Indexed: 12/24/2022]
Abstract
Human cytomegalovirus (HCMV) infection is a leading cause of morbidity and mortality in immunocompromised patients, but no specific therapeutic strategy is effective clinically, despite recent achievements. HCMV-specific T-cell therapy was thought to be helpful for the management of HCMV infection. To conduct a deep exploration, we investigated the possibility of engineering peripheral blood mononuclear cells (PBMCs) from immunocompetent and immunocompromised subjects with specific T-cell receptor (TCR) genes. CD8-positive T cells that specifically bind to NLV pentamers could be generated by transferring TCR genes to PBMCs from immunocompetent and immunocompromised subjects. The generation of functional T cells varied among transduction of different PBMCs. The numbers of IFN-γ-secreting T cells increased significantly in immunocompetent and immunodeficient PBMCs, but were unchanged in immune-reconstituted PBMCs. TCR gene transfer is a potential therapeutic strategy for controlling HCMV infection in immunocompromised patients. The transfer of TCR genes into immunocompetent and immunodeficient PBMCs would be more meaningful in response to HCMV infection than would the transfer into immune-reconstituted PBMCs.
Collapse
Affiliation(s)
- Runan Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Yanyue Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Jianhua Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Wei Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Xiaoming Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Zhongjie Lu
- Department of Radiotherapy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Rong Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Yaping Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Jun Fan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China.
| |
Collapse
|
13
|
Gallo S, Sangiolo D, Carnevale Schianca F, Aglietta M, Montemurro F. Treating breast cancer with cell-based approaches: an overview. Expert Opin Biol Ther 2017; 17:1255-1264. [PMID: 28728493 DOI: 10.1080/14712598.2017.1356816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Breast cancer is the most common malignancy in women. Despite there being considerable progress in the treatment of this disease, metastatic dissemination is still considered an incurable condition at the present time, causing 500,000 deaths worldwide every year. Although most of the research efforts have been focused on pharmacological approaches, over the last three decades, the use of bone marrow and peripheral blood-derived cell therapy approaches have been attempted and developed. Areas covered: This review will briefly address cell therapy for breast cancer, including autologous stem cell transplantations for overcoming the myelosuppressive effects of high-dose chemotherapy, allogeneic stem cell transplants and adoptive immunotherapy using bone-marrow derived T-cells. Expert opinion: The treatment of breast cancer using bone marrow or peripheral-blood derived cells has evolved from a supportive care approach to allow dose escalation of conventional chemotherapy to a therapeutic strategy aimed at eliciting immune cell mediated anticancer immunity. This latter principle has led to the development of adoptive immunotherapies, either with 'natural' or genetically engineered effectors, which are being intensively investigated for their great potential against several solid tumors, including breast cancer.
Collapse
Affiliation(s)
- Susanna Gallo
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy
| | - Dario Sangiolo
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy.,b Department of Oncology , University of Turin , Turin , Italy
| | | | - Massimo Aglietta
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy.,b Department of Oncology , University of Turin , Turin , Italy
| | - Filippo Montemurro
- c Investigative Clinical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy
| |
Collapse
|
14
|
Yue H, Cai Y, Song Y, Meng L, Chen X, Wang M, Bian Z, Wang R. Elevated TARP promotes proliferation and metastasis of salivary adenoid cystic carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2016; 123:468-476. [PMID: 28153567 DOI: 10.1016/j.oooo.2016.11.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 11/17/2016] [Accepted: 11/29/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study aims to analyze the expression of T-cell receptor γ chain alternate reading frame protein (TARP) in salivary adenoid cystic carcinoma (SACC) and its distant metastases and to investigate its influences on the development and progression of SACC. STUDY DESIGN TARP expression was analyzed in 50 primary SACCs, 13 specimens of metastatic adenoid cystic carcinoma of salivary gland origin, and 20 noncancerous tissues around SACC via immunohistochemistry. Cell Counting Kit-8 tests, wound healing assay, and Transwell experiments were performed to evaluate the effects of lentivirus-mediated TARP overexpression on the proliferation, migration, and invasion of SACC cells. RESULTS TARP expression was significantly increased in primary SACCs compared with adjacent noncancerous tissues, and this increase was further enhanced in metastases compared with primary SACCs. The expression level of TARP correlated significantly with tumor size, tumor-node-metastasis stage, perineural invasion, histologic type, and distant metastasis. Furthermore, TARP overexpression promoted the growth, migration, and invasion of SACC cells. CONCLUSIONS TARP plays an important role in and may be used as a marker to indicate the development and progression of SACC.
Collapse
Affiliation(s)
- Haitang Yue
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, P. R. China
| | - Yu Cai
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, P. R. China
| | - Yaling Song
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, P. R. China
| | - Liuyan Meng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, P. R. China
| | - Xinming Chen
- Oral Histopathology Department, School and Hospital of Stomatology, Wuhan University, Wuhan, P. R. China
| | - Mingwei Wang
- Department of Pathology, Hubei Cancer Hospital, Wuhan, P. R. China
| | - Zhuan Bian
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, P. R. China.
| | - Rong Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, P. R. China.
| |
Collapse
|
15
|
Hillerdal V, Boura VF, Björkelund H, Andersson K, Essand M. Avidity characterization of genetically engineered T-cells with novel and established approaches. BMC Immunol 2016; 17:23. [PMID: 27411667 PMCID: PMC4944473 DOI: 10.1186/s12865-016-0162-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/04/2016] [Indexed: 12/21/2022] Open
Abstract
Background Adoptive transfer of genetically engineered autologous T-cells is becoming a successful therapy for cancer. The avidity of the engineered T-cells is of crucial importance for therapy success. We have in the past cloned a T-cell receptor (TCR) that recognizes an HLA-A2 (MHC class I)-restricted peptide from the prostate and breast cancer- associated antigen TARP. Herein we perform a side-by-side comparison of the TARP-specific TCR (TARP-TCR) with a newly cloned TCR specific for an HLA-A2-restricted peptide from the cytomegalovirus (CMV) pp65 antigen. Results Both CD8+ T-cells and CD4+ T-cells transduced with the HLA-A2-restricted TARP-TCR could readily be detected by multimer analysis, indicating that the binding is rather strong, since binding occured also without the CD8 co-receptor of HLA-A2. Not surprisingly, the TARP-TCR, which is directed against a self-antigen, had weaker binding to the HLA-A2/peptide complex than the CMV pp65-specific TCR (pp65-TCR), which is directed against a viral epitope. Higher peptide concentrations were needed to achieve efficient cytokine release and killing of target cells when the TARP-TCR was used. We further introduce the LigandTracer technology to study cell-cell interactions in real time by evaluating the interaction between TCR-engineered T-cells and peptide-pulsed cancer cells. We were able to successfully detect TCR-engineered T-cell binding kinetics to the target cells. We also used the xCELLigence technology to analyzed cell growth of target cells to assess the killing potency of the TCR-engineered T-cells. T-cells transduced with the pp65 - TCR exhibited more pronounced cytotoxicity, being able to kill their targets at both lower effector to target ratios and lower peptide concentrations. Conclusion The combination of binding assay with functional assays yields data suggesting that TARP-TCR-engineered T-cells bind to their target, but need more antigen stimulation compared to the pp65-TCR to achieve full effector response. Nonetheless, we believe that the TARP-TCR is an attractive candidate for immunotherapy development for prostate and/or breast cancer.
Collapse
Affiliation(s)
- Victoria Hillerdal
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Vanessa F Boura
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hanna Björkelund
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Ridgeview Instruments AB, Vänge, Sweden
| | - Karl Andersson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Ridgeview Instruments AB, Vänge, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden. .,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-75185, Uppsala, Sweden.
| |
Collapse
|
16
|
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
|
17
|
Sun L, Guo H, Jiang R, Lu L, Liu T, He X. Engineered cytotoxic T lymphocytes with AFP-specific TCR gene for adoptive immunotherapy in hepatocellular carcinoma. Tumour Biol 2016; 37:799-806. [PMID: 26250457 DOI: 10.1007/s13277-015-3845-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/24/2015] [Indexed: 12/12/2022] Open
Abstract
Alpha-fetoprotein (AFP) is overexpressed in hepatocellular carcinoma (HCC) and could serve as a tumor-associated antigen (TAA) and potential target for adoptive immunotherapy. However, low frequency and severe functional impairment of AFP-specific T cells in vivo hamper adoptive infusion. TAA-specific T cell receptor (TCR) gene transfer could be an efficient and reliable alternation to generate AFP-specific cytotoxic T lymphocytes (CTLs). Autologous dendritic cells (DC) pulsed with AFP158-166 peptides were used to stimulate AFP-specific CTLs. TCR α/β chain genes of AFP-specific CTLs were cloned and linked by 2A peptide to form full-length TCR coding sequence synthesized into a lentiviral vector. Nonspecific activated T cells were engineered by lentivirus infection. Transgenetic CTLs were evaluated for transfection efficiency, expression of AFP158-166-specific TCR, interferon (IFN)-γ secretion, and specific cytotoxicity toward AFP+ HCC cells in vitro and in vivo. Flow cytometry revealed the AFP158-166-MHC-Pentamer positive transgenetic CTLs was 9.86 %. The number of IFN-γ secretion T cells and the specific cytotoxicity toward HpeG2 in vitro and in tumor-bearing NOD/SCID mice were significantly raised in transgenetic CTLs than that of AFP158-166-specific CTLs obtained by peptide-pulsed DCs or control group. TCR gene transfer is a promising strategy to generate AFP158-166-specific CTLs for the treatment of HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/metabolism
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Expression Regulation, Neoplastic
- Gene Transfer Techniques
- Genes, T-Cell Receptor
- Hep G2 Cells
- Humans
- Immunotherapy, Adoptive/methods
- Interferon-gamma/metabolism
- L-Lactate Dehydrogenase/metabolism
- Liver Neoplasms/metabolism
- MCF-7 Cells
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Monocytes/cytology
- Peptides/chemistry
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes, Cytotoxic/cytology
- Transgenes
- alpha-Fetoproteins/metabolism
Collapse
Affiliation(s)
- Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Hao Guo
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Ruoyu Jiang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Li Lu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road Heping District, Tianjin, 300052, China.
| |
Collapse
|
18
|
Töpfer K, Cartellieri M, Michen S, Wiedemuth R, Müller N, Lindemann D, Bachmann M, Füssel M, Schackert G, Temme A. DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy. THE JOURNAL OF IMMUNOLOGY 2015; 194:3201-12. [PMID: 25740942 DOI: 10.4049/jimmunol.1400330] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NK cells are emerging as new effectors for immunotherapy of cancer. In particular, the genetic engraftment of chimeric Ag receptors (CARs) in NK cells is a promising strategy to redirect NK cells to otherwise NK cell-resistant tumor cells. On the basis of DNAX-activation protein 12 (DAP12), a signaling adaptor molecule involved in signal transduction of activating NK cell receptors, we generated a new type of CAR targeting the prostate stem cell Ag (PSCA). We demonstrate in this article that this CAR, designated anti-PSCA-DAP12, consisting of DAP12 fused to the anti-PSCA single-chain Ab fragment scFv(AM1) confers improved cytotoxicity to the NK cell line YTS against PSCA-positive tumor cells when compared with a CAR containing the CD3ζ signaling chain. Further analyses revealed phosphorylation of the DAP12-associated ZAP-70 kinase and IFN-γ release of CAR-engineered cells after contact with PSCA-positive target cells. YTS cells modified with DAP12 alone or with a CAR bearing a phosphorylation-defective ITAM were not activated. Notably, infused YTS cells armed with anti-PSCA-DAP12 caused delayed tumor xenograft growth and resulted in complete tumor eradication in a significant fraction of treated mice. The feasibility of the DAP12-based CAR was further tested in human primary NK cells and confers specific cytotoxicity against KIR/HLA-matched PSCA-positive tumor cells, which was further enhanced by KIR-HLA mismatches. We conclude that NK cells engineered with DAP12-based CARs are a promising tool for adoptive tumor immunotherapy.
Collapse
Affiliation(s)
- Katrin Töpfer
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Marc Cartellieri
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Susanne Michen
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Ralf Wiedemuth
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Nadja Müller
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Dirk Lindemann
- Institute of Virology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Monika Füssel
- DKMS Life Science Lab, GmbH, 01307 Dresden, Germany; and
| | - Gabriele Schackert
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Achim Temme
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; German Cancer Consortium (DKTK), 01307 Dresden, Germany
| |
Collapse
|
19
|
Dong B, Minze LJ, Xue W, Chen W. Molecular insights into the development of T cell-based immunotherapy for prostate cancer. Expert Rev Clin Immunol 2014; 10:1547-57. [DOI: 10.1586/1744666x.2014.962515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
20
|
Abstract
Recent clinical success has underscored the potential for immunotherapy based on the adoptive cell transfer (ACT) of engineered T lymphocytes to mediate dramatic, potent, and durable clinical responses. This success has led to the broader evaluation of engineered T-lymphocyte-based adoptive cell therapy to treat a broad range of malignancies. In this review, we summarize concepts, successes, and challenges for the broader development of this promising field, focusing principally on lessons gleaned from immunological principles and clinical thought. We present ACT in the context of integrating T-cell and tumor biology and the broader systemic immune response.
Collapse
Affiliation(s)
- Marco Ruella
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
21
|
O'Connor CM, Wilson-Robles H. Developing T Cell Cancer Immunotherapy in the Dog with Lymphoma. ILAR J 2014; 55:169-81. [DOI: 10.1093/ilar/ilu020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
22
|
Le Brocq ML, Fraser AR, Cotton G, Woznica K, McCulloch CV, Hewitt KD, McKimmie CS, Nibbs RJB, Campbell JDM, Graham GJ. Chemokines as novel and versatile reagents for flow cytometry and cell sorting. THE JOURNAL OF IMMUNOLOGY 2014; 192:6120-6130. [PMID: 24850722 DOI: 10.4049/jimmunol.1303371] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cell therapy regimens are frequently compromised by low-efficiency cell homing to therapeutic niches. Improvements in this regard would enhance effectiveness of clinically applicable cell therapy. The major regulators of tissue-specific cellular migration are chemokines, and therefore selection of therapeutic cellular populations for appropriate chemokine receptor expression would enhance tissue-homing competence. A number of practical considerations preclude the use of Abs in this context, and alternative approaches are required. In this study, we demonstrate that appropriately labeled chemokines are at least as effective in detecting their cognate receptors as commercially available Abs. We also demonstrate the utility of biotinylated chemokines as cell-sorting reagents. Specifically, we demonstrate, in the context of CCR7 (essential for lymph node homing of leukocytes), the ability of biotinylated CCL19 with magnetic bead sorting to enrich for CCR7-expressing cells. The sorted cells demonstrate improved CCR7 responsiveness and lymph node-homing capability, and the sorting is effective for both T cells and dendritic cells. Importantly, the ability of chemokines to detect CCR7, and sort for CCR7 positivity, crosses species being effective on murine and human cells. This novel approach to cell sorting is therefore inexpensive, versatile, and applicable to numerous cell therapy contexts. We propose that this represents a significant technological advance with important therapeutic implications.
Collapse
Affiliation(s)
- Michelle L Le Brocq
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Alasdair R Fraser
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Graham Cotton
- Almac Sciences (Scotland) Ltd, Elvingston Science Centre, By Gladsmuir, East Lothian EH33 1EH, UK
| | - Kerry Woznica
- Almac Sciences (Scotland) Ltd, Elvingston Science Centre, By Gladsmuir, East Lothian EH33 1EH, UK
| | - Clare V McCulloch
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Kay D Hewitt
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Clive S McKimmie
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Robert J B Nibbs
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - John D M Campbell
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK.,Clinical Science and Cell Analysis Group, Miltenyi Biotec Ltd
| | - Gerard J Graham
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| |
Collapse
|
23
|
Allogeneic lymphocyte-licensed DCs expand T cells with improved antitumor activity and resistance to oxidative stress and immunosuppressive factors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14001. [PMID: 26015949 PMCID: PMC4362340 DOI: 10.1038/mtm.2014.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/03/2014] [Indexed: 12/17/2022]
Abstract
Adoptive T-cell therapy of cancer is a treatment strategy where T cells are isolated, activated, in some cases engineered, and expanded ex vivo before being reinfused to the patient. The most commonly used T-cell expansion methods are either anti-CD3/CD28 antibody beads or the “rapid expansion protocol” (REP), which utilizes OKT-3, interleukin (IL)-2, and irradiated allogeneic feeder cells. However, REP-expanded or bead-expanded T cells are sensitive to the harsh tumor microenvironment and often short-lived after reinfusion. Here, we demonstrate that when irradiated and preactivated allosensitized allogeneic lymphocytes (ASALs) are used as helper cells to license OKT3-armed allogeneic mature dendritic cells (DCs), together they expand target T cells of high quality. The ASAL/DC combination yields an enriched Th1-polarizing cytokine environment (interferon (IFN)-γ, IL-12, IL-2) and optimal costimulatory signals for T-cell stimulation. When genetically engineered antitumor T cells were expanded by this coculture system, they showed better survival and cytotoxic efficacy under oxidative stress and immunosuppressive environment, as well as superior proliferative response during tumor cell killing compared to the REP protocol. Our result suggests a robust ex vivo method to expand T cells with improved quality for adoptive cancer immunotherapy.
Collapse
|
24
|
Reassessing target antigens for adoptive T-cell therapy. Nat Biotechnol 2013; 31:999-1008. [PMID: 24142051 PMCID: PMC4280065 DOI: 10.1038/nbt.2725] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/24/2013] [Indexed: 12/15/2022]
Abstract
Adoptive T cell therapy can target and kill widespread malignant cells thereby inducing durable clinical responses in melanoma and selected other malignances. However, many commonly targeted tumor antigens are also expressed by healthy tissues, and T cells do not distinguish between benign and malignant tissues if both express the target antigen. As such, autoimmune toxicity from T-cell-mediated destruction of normal tissue has limited the development and adoption of this otherwise promising type of cancer therapy. A review of the unique biology of T-cell therapy and of recent clinical experience compels a reassessment of target antigens that traditionally have been viewed from the perspective of weaker immunotherapeutic modalities. In selecting target antigens for adoptive T-cell therapy, expression by tumors and not by essential healthy tissues is of paramount importance. The risk of autoimmune adverse events can be further mitigated by generating antigen receptors using strategies that reduce the chance of cross-reactivity against epitopes in unintended targets. In general, a circumspect approach to target selection and thoughtful preclinical and clinical studies are pivotal to the ongoing advancement of these promising treatments.
Collapse
|
25
|
Abstract
T cells have the capacity to eradicate diseased cells, but tumours present considerable challenges that render T cells ineffectual. Cancer cells often make themselves almost 'invisible' to the immune system, and they sculpt a microenvironment that suppresses T cell activity, survival and migration. Genetic engineering of T cells can be used therapeutically to overcome these challenges. T cells can be taken from the blood of cancer patients and then modified with genes encoding receptors that recognize cancer-specific antigens. Additional genes can be used to enable resistance to immunosuppression, to extend survival and to facilitate the penetration of engineered T cells into tumours. Using genetic modification, highly active, self-propagating 'slayers' of cancer cells can be generated.
Collapse
Affiliation(s)
- Michael H Kershaw
- Cancer Immunology Research Program, Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia. michael.kershaw@ petermac.org
| | | | | |
Collapse
|
26
|
Abstract
T-cell immunotherapy is a promising approach to treat disseminated cancer. However, it has been limited by the ability to isolate and expand T cells restricted to tumour-associated antigens. Using ex vivo gene transfer, T cells from patients can be genetically engineered to express a novel T cell receptor or chimeric antigen receptor to specifically recognize a tumour-associated antigen and thereby selectively kill tumour cells. Indeed, genetically engineered T cells have recently been successfully used for cancer treatment in a small number of patients. Here we review the recent progress in the field, and summarize the challenges that lie ahead and the strategies being used to overcome them.
Collapse
Affiliation(s)
- M Essand
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|