1
|
Taheri MM, Javan F, Poudineh M, Athari SS. CAR-NKT Cells in Asthma: Use of NKT as a Promising Cell for CAR Therapy. Clin Rev Allergy Immunol 2024; 66:328-362. [PMID: 38995478 DOI: 10.1007/s12016-024-08998-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
NKT cells, unique lymphocytes bridging innate and adaptive immunity, offer significant potential for managing inflammatory disorders like asthma. Activating iNKT induces increasing IFN-γ, TGF-β, IL-2, and IL-10 potentially suppressing allergic asthma. However, their immunomodulatory effects, including granzyme-perforin-mediated cytotoxicity, and expression of TIM-3 and TRAIL warrant careful consideration and targeted approaches. Although CAR-T cell therapy has achieved remarkable success in treating certain cancers, its limitations necessitate exploring alternative approaches. In this context, CAR-NKT cells emerge as a promising approach for overcoming these challenges, potentially achieving safer and more effective immunotherapies. Strategies involve targeting distinct IgE-receptors and their interactions with CAR-NKT cells, potentially disrupting allergen-mast cell/basophil interactions and preventing inflammatory cytokine release. Additionally, targeting immune checkpoints like PDL-2, inducible ICOS, FASL, CTLA-4, and CD137 or dectin-1 for fungal asthma could further modulate immune responses. Furthermore, artificial intelligence and machine learning hold immense promise for revolutionizing NKT cell-based asthma therapy. AI can optimize CAR-NKT cell functionalities, design personalized treatment strategies, and unlock a future of precise and effective care. This review discusses various approaches to enhancing CAR-NKT cell efficacy and longevity, along with the challenges and opportunities they present in the treatment of allergic asthma.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyyed Shamsadin Athari
- Cancer Gene therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
2
|
Hadiloo K, Tahmasebi S, Esmaeilzadeh A. CAR-NKT cell therapy: a new promising paradigm of cancer immunotherapy. Cancer Cell Int 2023; 23:86. [PMID: 37158883 PMCID: PMC10165596 DOI: 10.1186/s12935-023-02923-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Today, cancer treatment is one of the fundamental problems facing clinicians and researchers worldwide. Efforts to find an excellent way to treat this illness continue, and new therapeutic strategies are developed quickly. Adoptive cell therapy (ACT) is a practical approach that has been emerged to improve clinical outcomes in cancer patients. In the ACT, one of the best ways to arm the immune cells against tumors is by employing chimeric antigen receptors (CARs) via genetic engineering. CAR equips cells to target specific antigens on tumor cells and selectively eradicate them. Researchers have achieved promising preclinical and clinical outcomes with different cells by using CARs. One of the potent immune cells that seems to be a good candidate for CAR-immune cell therapy is the Natural Killer-T (NKT) cell. NKT cells have multiple features that make them potent cells against tumors and would be a powerful replacement for T cells and natural killer (NK) cells. NKT cells are cytotoxic immune cells with various capabilities and no notable side effects on normal cells. The current study aimed to comprehensively provide the latest advances in CAR-NKT cell therapy for cancers.
Collapse
Affiliation(s)
- Kaveh Hadiloo
- Student Research Committee, Department of immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of immunology, School of Medicine, Shahid beheshti University of Medical Sciences, Tehran, Iran.
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran.
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
3
|
Neumeister P, Schulz E, Pansy K, Szmyra M, Deutsch AJA. Targeting the Microenvironment for Treating Multiple Myeloma. Int J Mol Sci 2022; 23:ijms23147627. [PMID: 35886976 PMCID: PMC9317002 DOI: 10.3390/ijms23147627] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/23/2022] Open
Abstract
Multiple myeloma (MM) is a malignant, incurable disease characterized by the expansion of monoclonal terminally differentiated plasma cells in the bone marrow. MM is consistently preceded by an asymptomatic monoclonal gammopathy of undetermined significance, and in the absence of myeloma defining events followed by a stage termed smoldering multiple myeloma (SMM), which finally progresses to active myeloma if signs of organ damage are present. The reciprocal interaction between tumor cells and the tumor microenvironment plays a crucial role in the development of MM and the establishment of a tumor-promoting stroma facilitates tumor growth and myeloma progression. Since myeloma cells depend on signals from the bone marrow microenvironment (BMME) for their survival, therapeutic interventions targeting the BMME are a novel and successful strategy for myeloma care. Here, we describe the complex interplay between myeloma cells and the cellular components of the BMME that is essential for MM development and progression. Finally, we present BMME modifying treatment options such as anti-CD38 based therapies, immunomodulatory drugs (IMiDs), CAR T-cell therapies, bispecific antibodies, and antibody-drug conjugates which have significantly improved the long-term outcome of myeloma patients, and thus represent novel therapeutic standards.
Collapse
Affiliation(s)
- Peter Neumeister
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (E.S.); (K.P.); (M.S.); (A.J.D.)
- Correspondence:
| | - Eduard Schulz
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (E.S.); (K.P.); (M.S.); (A.J.D.)
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katrin Pansy
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (E.S.); (K.P.); (M.S.); (A.J.D.)
| | - Marta Szmyra
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (E.S.); (K.P.); (M.S.); (A.J.D.)
| | - Alexander JA Deutsch
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (E.S.); (K.P.); (M.S.); (A.J.D.)
| |
Collapse
|
4
|
Chimeric antigen receptor engineered innate immune cells in cancer immunotherapy. SCIENCE CHINA-LIFE SCIENCES 2019; 62:633-639. [DOI: 10.1007/s11427-018-9451-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/13/2018] [Indexed: 12/18/2022]
|
5
|
Fu X, Rivera A, Tao L, Zhang X. An HSV-2 based oncolytic virus can function as an attractant to guide migration of adoptively transferred T cells to tumor sites. Oncotarget 2015; 6:902-14. [PMID: 25460506 PMCID: PMC4359264 DOI: 10.18632/oncotarget.2817] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/24/2014] [Indexed: 11/25/2022] Open
Abstract
Adoptive T-cell therapy has shown promises for cancer treatment. However, for treating solid tumors, there is a need for improving the ability of the adoptively transferred T cells to home to tumor sites. We explored the possibility of using an oncolytic virus derived from HSV-2, which can actively pull T effector cells to the site of infection, as a local attractant for migration of adoptively transferred T cells. Our data show that intratumoral administration of this virus can indeed attract active migration of the adoptively transferred T cells to the treated tumor. Moreover, once attracted to the tumor site by the virus, T cells persisted in there significantly longer than in mock-treated tumor. Chemokine profiling identified significant elevation of CXCL9 and CXCL10, as well as several other chemokines belonging to the inflammatory chemokine family in the virus-treated tumors. These chemokines initially guided the T-cell migration to and then maintained their persistence in the tumor site, leading to a significantly enhanced therapeutic effect. Our data suggests that this virotherapy may be combined with adoptive T-cell therapy to potentiate its therapeutic effect against solid tumors that are otherwise difficult to manage with the treatment alone.
Collapse
Affiliation(s)
- Xinping Fu
- Department of Biology and Biochemistry and Center for Nuclear Receptors and Cell Signaling, University of Houston, Texas, USA
| | - Armando Rivera
- Department of Biology and Biochemistry and Center for Nuclear Receptors and Cell Signaling, University of Houston, Texas, USA
| | - Lihua Tao
- Department of Biology and Biochemistry and Center for Nuclear Receptors and Cell Signaling, University of Houston, Texas, USA
| | - Xiaoliu Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| |
Collapse
|
6
|
Zhou F, Krishnamurthy J, Wei Y, Li M, Hunt K, Johanning GL, Cooper LJ, Wang-Johanning F. Chimeric antigen receptor T cells targeting HERV-K inhibit breast cancer and its metastasis through downregulation of Ras. Oncoimmunology 2015; 4:e1047582. [PMID: 26451325 DOI: 10.1080/2162402x.2015.1047582] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/27/2015] [Accepted: 04/29/2015] [Indexed: 12/15/2022] Open
Abstract
We have previously reported that human endogenous retrovirus-K (HERV-K) envelope (env) protein is a tumor-associated antigen (TAA) for cancer vaccines, and that its antibodies (mAbs) possess antitumor activity against cancer. In this study, a chimeric antigen receptor (CAR) specific for HERV-K env protein (K-CAR) was generated using anti-HERV-K mAb. K-CAR T cells from peripheral blood mononuclear cells (PBMCs) of 9 breast cancer (BC) patients and 12 normal donors were able to inhibit growth of, and to exhibit significant cytotoxicity toward, BC cells but not MCF-10A normal breast cells. The antitumor effects in cancer cells were significantly reduced when control T cells were used, or the expression of HERV-K was knocked down by an shRNA. Secretion of multiple cytokines, including IFNγ, TNF-α, and IL-2, was significantly enhanced in culture media of BC cells treated with K-CARs. Significantly reduced tumor growth and tumor weight was observed in xenograft models bearing MDA-MB-231 or MDA-MB-435.eB1 BC cells. Importantly, the K-CAR prevented tumor metastasis to other organs. Furthermore, downregulation of HERV-K expression in tumors of mice treated with K-CAR correlated with upregulation of p53 and downregulation of MDM2 and p-ERK. Importantly, the expression of HERV-K env protein in metastatic tumor tissues treated with K-CAR T cells correlated with the expression of Ras. Our results indicate that HERV-K env protein is an oncoprotein and may play an important role in tumorigenesis related to p53 and Ras signaling pathways. Anti-HERV-K treatment, including K-CAR treatment, shows potential for immunotherapy of BC.
Collapse
Affiliation(s)
- Fuling Zhou
- Department of Veterinary Sciences; University of Texas MD Anderson Cancer Center ; Houston, TX USA ; Viral Oncology Program; SRI International ; Menlo Park, CA USA ; Department of Clinical Hematology; Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University ; Xi'an, Shannxi, China
| | - Janani Krishnamurthy
- Division of Pediatrics; University of Texas MD Anderson Cancer Center ; Houston, TX USA ; Graduate School of Biomedical Sciences ; Houston, TX USA
| | - Yongchang Wei
- Department of Veterinary Sciences; University of Texas MD Anderson Cancer Center ; Houston, TX USA ; Viral Oncology Program; SRI International ; Menlo Park, CA USA ; Department of Clinical Oncology, First Affiliated Hospital; School of Medicine; Xi'an Jiaotong University , Xi'an, Shannxi, China
| | - Ming Li
- Department of Veterinary Sciences; University of Texas MD Anderson Cancer Center ; Houston, TX USA ; Viral Oncology Program; SRI International ; Menlo Park, CA USA ; Graduate School of Biomedical Sciences ; Houston, TX USA
| | - Kelly Hunt
- Department of Surgical Oncology; University of Texas MD Anderson Cancer Center ; Houston, TX USA
| | - Gary L Johanning
- Department of Veterinary Sciences; University of Texas MD Anderson Cancer Center ; Houston, TX USA ; Viral Oncology Program; SRI International ; Menlo Park, CA USA ; Graduate School of Biomedical Sciences ; Houston, TX USA
| | - Laurence Jn Cooper
- Division of Pediatrics; University of Texas MD Anderson Cancer Center ; Houston, TX USA ; Graduate School of Biomedical Sciences ; Houston, TX USA
| | - Feng Wang-Johanning
- Department of Veterinary Sciences; University of Texas MD Anderson Cancer Center ; Houston, TX USA ; Viral Oncology Program; SRI International ; Menlo Park, CA USA ; Graduate School of Biomedical Sciences ; Houston, TX USA ; Department of Immunology; University of Texas MD Anderson Cancer Center ; Houston, TX USA
| |
Collapse
|
7
|
Clinical Evaluation of ErbB-Targeted CAR T-Cells, Following Intracavity Delivery in Patients with ErbB-Expressing Solid Tumors. Methods Mol Biol 2015; 1317:365-82. [PMID: 26072418 DOI: 10.1007/978-1-4939-2727-2_21] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Adoptive cell therapy using gene-modified T-cells has achieved impressive results in the treatment of B-cell malignancies. However, the development of similar strategies to treat solid tumors raises challenges with respect to tumor antigen selection and the achievement of efficient T-cell homing, survival and sustained effector function within the tumor microenvironment. To address these challenges, we have developed a gene-modified cellular therapy called T4 immunotherapy. To generate T4 immunotherapy, autologous T-cells are engineered by retroviral transduction to co-express two transgenes: (1) a chimeric antigen receptor (CAR), T1E28z, targeted against a range of ErbB homodimers and heterodimers and (2) a chimeric cytokine receptor, 4αβ, that allows the selective ex vivo expansion of engineered cells using interleukin-4. Targeting of the extended ErbB network using CAR T-cells is supported by prevalence of ErbB dysregulation in diverse solid tumors and the clinical impact of monoclonal antibody therapy directed against members of this family. However, the key obstacle to effective clinical translation is risk of on-target toxicity owing to the lower level expression of ErbB family members in many healthy tissues. To de-risk T4 immunotherapy in man, we are undertaking a trial in patients with locally advanced or recurrent head and neck squamous cell carcinoma. In that setting, engineered T-cells are injected directly into the tumor without prior lymphodepletion, an approach that we believe will minimize risk of toxicity. This chapter outlines how we plan to advance the development of T4 immunotherapy thereafter in Phase II clinical testing. In that setting, regional (intracavitary) approaches will be used to administer this therapy to patients with epithelial ovarian cancer and malignant pleural mesothelioma.
Collapse
|
8
|
T cells expressing VHH-directed oligoclonal chimeric HER2 antigen receptors: Towards tumor-directed oligoclonal T cell therapy. Biochim Biophys Acta Gen Subj 2014; 1840:378-86. [DOI: 10.1016/j.bbagen.2013.09.029] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 08/22/2013] [Accepted: 09/20/2013] [Indexed: 01/01/2023]
|
9
|
Fu X, Rivera A, Tao L, Zhang X. Genetically modified T cells targeting neovasculature efficiently destroy tumor blood vessels, shrink established solid tumors and increase nanoparticle delivery. Int J Cancer 2013; 133:2483-92. [PMID: 23661285 DOI: 10.1002/ijc.28269] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 04/23/2013] [Indexed: 02/03/2023]
Abstract
Converting T cells into tumor cell killers by grafting them with a chimeric antigen receptor (CAR) has shown promise as a cancer immunotherapeutic. However, the inability of these cells to actively migrate and extravasate into tumor parenchyma has limited their effectiveness in vivo. Here we report the construction of a CAR containing an echistatin as its targeting moiety (eCAR). As echistatin has high binding affinity to αvβ3 integrin that is highly expressed on the surface of endothelial cells of tumor neovasculature, T cells engrafted with eCAR (T-eCAR) can efficiently lyse human umbilical vein endothelial cells and tumor cells that express αvβ3 integrin when tested in vitro. Systemic administration of T-eCAR led to extensive bleeding in tumor tissues with no evidence of damage to blood vessels in normal tissues. Destruction of tumor blood vessels by T-eCAR significantly inhibited the growth of established bulky tumors. Moreover, when T-eCAR was codelivered with nanoparticles in a strategically designed temporal order, it dramatically increased nanoparticle deposition in tumor tissues, pointing to the possibility that it may be used together with nanocarriers to increase their capability to selectively deliver antineoplastic drugs to tumor tissues.
Collapse
Affiliation(s)
- Xinping Fu
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX
| | | | | | | |
Collapse
|
10
|
Davies DM, Foster J, van der Stegen SJC, Parente-Pereira AC, Chiapero-Stanke L, Delinassios GJ, Burbridge SE, Kao V, Liu Z, Bosshard-Carter L, van Schalkwyk MCI, Box C, Eccles SA, Mather SJ, Wilkie S, Maher J. Flexible targeting of ErbB dimers that drive tumorigenesis by using genetically engineered T cells. Mol Med 2012; 18:565-76. [PMID: 22354215 PMCID: PMC3388141 DOI: 10.2119/molmed.2011.00493] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 02/16/2012] [Indexed: 11/06/2022] Open
Abstract
Pharmacological targeting of individual ErbB receptors elicits antitumor activity, but is frequently compromised by resistance leading to therapeutic failure. Here, we describe an immunotherapeutic approach that exploits prevalent and fundamental mechanisms by which aberrant upregulation of the ErbB network drives tumorigenesis. A chimeric antigen receptor named T1E28z was engineered, in which the promiscuous ErbB ligand, T1E, is fused to a CD28 + CD3ζ endodomain. Using a panel of ErbB-engineered 32D hematopoietic cells, we found that human T1E28z⁺ T cells are selectively activated by all ErbB1-based homodimers and heterodimers and by the potently mitogenic ErbB2/3 heterodimer. Owing to this flexible targeting capability, recognition and destruction of several tumor cell lines was achieved by T1E28⁺ T cells in vitro, comprising a wide diversity of ErbB receptor profiles and tumor origins. Furthermore, compelling antitumor activity was observed in mice bearing established xenografts, characterized either by ErbB1/2 or ErbB2/3 overexpression and representative of insidious or rapidly progressive tumor types. Together, these findings support the clinical development of a broadly applicable immunotherapeutic approach in which the propensity of solid tumors to dysregulate the extended ErbB network is targeted for therapeutic gain.
Collapse
Affiliation(s)
- David M Davies
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Julie Foster
- Centre for Molecular Oncology and Imaging, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Sjoukje J C van der Stegen
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Ana C Parente-Pereira
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Laura Chiapero-Stanke
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - George J Delinassios
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Sophie E Burbridge
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Vincent Kao
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Zhe Liu
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Leticia Bosshard-Carter
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - May C I van Schalkwyk
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Carol Box
- Tumour Biology and Metastasis, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Suzanne A Eccles
- Tumour Biology and Metastasis, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Stephen J Mather
- Centre for Molecular Oncology and Imaging, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Scott Wilkie
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - John Maher
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
- Department of Immunology, Barnet and Chase Farm National Health Service (NHS) Trust, Barnet, Hertfordshire, UK
- Department of Clinical Immunology and Allergy, King’s College Hospital NHS Foundation Trust, Denmark Hill, London, UK
| |
Collapse
|
11
|
Dodson LF, Hawkins WG, Goedegebuure P. Potential targets for pancreatic cancer immunotherapeutics. Immunotherapy 2011; 3:517-37. [PMID: 21463193 DOI: 10.2217/imt.11.10] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic adenocarcinoma is the fourth leading cause of cancer death with an overall 5-year survival of less than 5%. As there is ample evidence that pancreatic adenocarcinomas elicit antitumor immune responses, identification of pancreatic cancer-associated antigens has spurred the development of vaccination-based strategies for treatment. While promising results have been observed in animal tumor models, most clinical studies have found only limited success. As most trials were performed in patients with advanced pancreatic cancer, the contribution of immune suppressor mechanisms should be taken into account. In this article, we detail recent work in tumor antigen vaccination and the recently identified mechanisms of immune suppression in pancreatic cancer. We offer our perspective on how to increase the clinical efficacy of vaccines for pancreatic cancer.
Collapse
Affiliation(s)
- Lindzy F Dodson
- Washington University School of Medicine, Department of Surgery, Saint Louis, MO 63110, USA.
| | | | | |
Collapse
|
12
|
Parente-Pereira AC, Burnet J, Ellison D, Foster J, Davies DM, van der Stegen S, Burbridge S, Chiapero-Stanke L, Wilkie S, Mather S, Maher J. Trafficking of CAR-engineered human T cells following regional or systemic adoptive transfer in SCID beige mice. J Clin Immunol 2011; 31:710-8. [PMID: 21505816 DOI: 10.1007/s10875-011-9532-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 04/03/2011] [Indexed: 02/07/2023]
Abstract
Adoptive immunotherapy using chimeric antigen receptor-engrafted T cells is a promising emerging therapy for cancer. Prior to clinical testing, it is mandatory to evaluate human therapeutic cell products in meaningful in vivo pre-clinical models. Here, we describe the use of fused single-photon emission CT-CT imaging to monitor real-time migration of chimeric antigen receptor-engineered T cells in immune compromised (SCID Beige) mice. Following intravenous administration, human T cells migrate in a highly similar manner to that reported in man, but penetrate poorly into established tumors. By contrast, when delivered via intraperitoneal or subcutaneous routes, T cells remain at the site of inoculation with minimal systemic absorption-irrespective of the presence or absence of tumor. Together, these data support the validity of pre-clinical testing of human T-cell immunotherapy in SCID Beige mice. In light of their established efficacy, regional administration of engineered human T cells represents an attractive therapeutic option to minimize toxicity in the treatment of selected malignancies.
Collapse
Affiliation(s)
- Ana Caterina Parente-Pereira
- The CAR Mechanics Group, King's College London School of Medicine, Guy's Hospital Campus, St Thomas Street, London, SE1 9RT, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|