1
|
Liang G, Ma Y, Deng P, Li S, He C, He H, Liu H, Fan Y, Li Z. Role of cell-based therapies in digestive disorders: Obstacles and opportunities. Regen Ther 2025; 29:1-18. [PMID: 40124469 PMCID: PMC11925584 DOI: 10.1016/j.reth.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/01/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Stem cell-based therapies have emerged as a promising frontier in the treatment of gastrointestinal disorders, offering potential solutions for challenges posed by conventional treatments. This review comprehensively examines recent advancements in cell-based therapeutic strategies, particularly focusing on stem cell applications, immunotherapy, and cellular therapies for digestive diseases. It highlights the successful differentiation of enteric neural progenitors from pluripotent stem cells and their application in animal models, such as Hirschsprung disease. Furthermore, the review evaluates clinical trials and experimental studies demonstrating the potential of stem cells in regenerating damaged tissues, modulating immune responses, and promoting healing in conditions like Crohn's disease and liver failure. By addressing challenges, such as scalability, immunogenicity, and ethical considerations, the review underscores the translational opportunities and obstacles in realizing the clinical potential of these therapies. Concluding with an emphasis on future directions, the study provides insights into optimizing therapeutic efficacy and fostering innovations in personalized medicine for digestive disorders.
Collapse
Affiliation(s)
- Guodong Liang
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Yuehan Ma
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Ping Deng
- Medical Department, Jilin Cancer Hospital, Changchun 130012, China
| | - Shufeng Li
- First Department of Gynecological Tumor, Jilin Cancer Hospital, Changchun 130012, China
| | - Chunyan He
- Department of Anaesthesia, Jilin Cancer Hospital, Changchun 130012, China
| | - Haihang He
- Department of Otorhinolaryngology, Oral Maxillofacial, Head and Neck, Jilin Cancer Hospital, Changchun 130012, China
| | - Hairui Liu
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Yunda Fan
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Ze Li
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| |
Collapse
|
2
|
Wu C, Johnson NM, Yu S, Lo AS, Sahu GK, Marx PA, von Laer D, Skowron G, Geleziunas R, Shaw GM, Kaur A, Junghans RP, Braun SE. Persistence of CMV-specific anti-HIV CAR T cells after adoptive immunotherapy. J Virol 2025; 99:e0193324. [PMID: 40207929 PMCID: PMC12090794 DOI: 10.1128/jvi.01933-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/18/2025] [Indexed: 04/11/2025] Open
Abstract
The success of chimeric antigen receptor (CAR)-T cell (Tc) immunotherapy in refractory B-cell acute lymphoblastic leukemia (B-ALL) suggests adaptation of this strategy toward HIV. Because cytomegalovirus (CMV) vaccine vectors generated Tc responses that controlled viral replication, these studies aim to genetically modify CMV-specific Tc with HIV-CAR2 vectors and link HIV immunotherapy to persistent CMV antigen stimulation. To mimic a clinical scenario, rhesus macaques were challenged with the CCR5-tropic simian/human immunodeficiency virus (SHIV-D) prior to antiretroviral therapy (ART). Autologous CMV-specific Tc were transduced with the control CEA-CAR2 or CD4-CAR2/maC46 vectors and reinfused. After stopping ART, the plasma viral load (PVL) in the control rebounded and was sustained above 1.7 × 104 copies/mL; PVL in CD4-CAR2-treated animals was delayed up to 6 weeks and 10-fold lower. The CD4 CAR-Tc frequency peaked at day 7 and was detected in lymphoid tissues at 6 weeks. Both CEA-CAR2 and CD4-CAR2 persisted in PBMCs for about 2 years, which indicates that the CMV-specific CAR Tc were maintained based on their CMV specificity. However, long-term PVL was stable in all animals. Thus, CMV-specific CAR-Tc were active initially, persisted long term, but failed to control viral replication.IMPORTANCEBecause of latent viral reservoirs and a dysfunctional immune response, HIV replication rebounds when antiretroviral therapy is interrupted. Therefore, cytomegalovirus (CMV)-specific Tc were genetically modified with anti-HIV CD4-CAR2 vectors to link the targeting of the HIV envelope to the persistent CMV immune response. In this clinical scenario with simian/human immunodeficiency virus (SHIV) challenge and antiretroviral therapy (ART) suppression, early activity of the CAR Tc delayed rebound in the rhesus macaque/SHIV challenge model. However, even with long-term persistence of CAR Tc in the blood, control of viral replication was not achieved. These data suggest that CAR Tc will require additional interventions to cure HIV infection.
Collapse
Affiliation(s)
- Chengxiang Wu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
| | - Nathan M. Johnson
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
| | - Shan Yu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
- Department of Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Agnes S. Lo
- Tufts Medical School, Boston, Massachusetts, USA
| | - Gautam K. Sahu
- Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Preston A. Marx
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | | | - Gail Skowron
- Roger Williams Medical Center, Providence, Rhode Island, USA
| | | | - George M. Shaw
- Division of Hematology/Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
- Department of Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Richard P. Junghans
- Tufts Medical School, Boston, Massachusetts, USA
- Roger Williams Medical Center, Providence, Rhode Island, USA
- IT Bio, LLC., Boston, Massachusetts, USA
| | - Stephen E. Braun
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
3
|
Bhutani B, Sharma V, Ganguly NK, Rana R. Unravelling the modified T cell receptor through Gen-Next CAR T cell therapy in Glioblastoma: Current status and future challenges. Biomed Pharmacother 2025; 186:117987. [PMID: 40117901 DOI: 10.1016/j.biopha.2025.117987] [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] [Received: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Despite current technological advancements in the treatment of glioma, immediate alleviation of symptoms can be catered by therapeutic modalities, including surgery, chemotherapy, and combinatorial radiotherapy that exploit aberrations of glioma. Additionally, a small number of target antigens, their heterogeneity, and immune evasion are the potential reasons for developing targeted therapies. This oncologic milestone has catalyzed interest in developing immunotherapies against Glioblastoma to improve overall survival and cure patients with high-grade glioma. The next-gen CAR-T Cell therapy is one of the effective immunotherapeutic strategies in which autologous T cells have been modified to express receptors against GBM and it modulates cytotoxicity. METHODS In this review article, we examine preclinical and clinical outcomes, and limitations as well as present cutting-edge techniques to improve the function of CAR-T cell therapy and explore the possibility of combination therapy. FINDINGS To date, several CAR T-cell therapies are being evaluated in clinical trials for GBM and other brain malignancies and multiple preclinical studies have demonstrated encouraging outcomes. IMPLICATIONS CAR-T cell therapy represents a promising therapeutic paradigm in the treatment of solid tumors but a few limitations include, the blood-brain barrier (BBB), antigen escape, tumor microenvironment (TME), tumor heterogeneity, and its plasticity that suppresses immune responses weakens the ability of this therapy. Additional investigation is required that can accurately identify the targets and reflect the similar architecture of glioblastoma, thus optimizing the efficiency of CAR-T cell therapy; allowing for the selection of patients most likely to benefit from immuno-based treatments.
Collapse
Affiliation(s)
- Bhavya Bhutani
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Vyoma Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
4
|
Ahmed EN, Cutmore LC, Marshall JF. Syngeneic Mouse Models for Pre-Clinical Evaluation of CAR T Cells. Cancers (Basel) 2024; 16:3186. [PMID: 39335157 PMCID: PMC11430534 DOI: 10.3390/cancers16183186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cells have revolutionized the treatment of hematological malignancies. Unfortunately, this improvement has yet to be translated into the solid tumor field. Current immunodeficient models used in pre-clinical testing often overestimate the efficacy of CAR T cell therapy as they fail to recapitulate the immunosuppressive tumor microenvironment characteristic of solid tumors. As CAR T cell monotherapy is unlikely to be curative for many solid tumors, combination therapies must be investigated, for example, stromal remodeling agents and immunomodulators. The evaluation of these combination therapies requires a fully immunocompetent mouse model in order to recapitulate the interaction between the host's immune system and the CAR T cells. This review will discuss the need for improved immunocompetent murine models for the pre-clinical evaluation of CAR T cells, the current use of such models and future directions.
Collapse
Affiliation(s)
- Eman N Ahmed
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Lauren C Cutmore
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - John F Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
5
|
Zeng S, Jin N, Yu B, Ren Q, Yan Z, Fu S. Chimeric antigen receptor-T cells targeting epithelial cell adhesion molecule antigens are effective in the treatment of colorectal cancer. BMC Gastroenterol 2024; 24:249. [PMID: 39107717 PMCID: PMC11302356 DOI: 10.1186/s12876-024-03286-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/07/2024] [Indexed: 08/10/2024] Open
Abstract
OBJECTIVE To construct chimeric antigen receptor (CAR)-T cells targeting epithelial cell adhesion molecule (EpCAM) antigen (anti-EpCAM-CAR-T). METHODS A third-generation CAR-T cell construct used a single-chain variable fragment derived from monoclonal antibody against human EpCAM. Peripheral blood mononuclear cells were extracted from volunteers. The proportion of cluster of differentiation 8 positive (CD8+) and CD4 + T cells was measured using flow cytometry. Western blot was used to detect the expression of EpCAM-CAR. The killing efficiency was detected using the MTT assay and transwell assay, and the secretion of killer cytokines tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) was detected using the ELISA. The inhibitory effect of EpCAM-CAR-T on colorectal cancer in vivo was detected using xenografts. RESULTS It was found that T cells expanded greatly, and the proportion of CD3+, CD8 + and CD4 + T cells was more than 60%. Furthermore, EpCAM-CAR-T cells had a higher tumour inhibition rate in the EpCAM expression positive group than in the negative group (P < 0.05). The secretion of killer cytokines TNF-α and IFN-γ in the EpCAM expression positive cell group was higher than that in the negative group (P < 0.05). In the experimental group treated with EpCAM-CAR-T cells, the survival rate of nude mice was higher (P < 0.05), and the tumour was smaller than that in the blank and control groups (P < 0.05). The secretion of serum killer cytokines TNF-α and IFN-γ in tumour-bearing nude mice in the experimental group treated with EpCAM-CAR-T cells was higher than that in the blank and control groups (P < 0.05). CONCLUSION This study successfully constructed EpCAM-CAR cells and found that they can target and recognise EpCAM-positive tumour cells, secrete killer cytokines TNF-α and IFN-γ and better inhibit the growth and metastasis of colorectal cancer in vitro and in vivo than unmodified T cells.
Collapse
Affiliation(s)
- Siheng Zeng
- Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
- Birth Defects and Regenerative Medicine Laboratory, Department of Biochemistry & Molecular Biology, Biomedicine and Health Graduate Education Innovation Center, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan, Shanxi, 030001, China
| | - Ning Jin
- Birth Defects and Regenerative Medicine Laboratory, Department of Biochemistry & Molecular Biology, Biomedicine and Health Graduate Education Innovation Center, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan, Shanxi, 030001, China
| | - Baofeng Yu
- Birth Defects and Regenerative Medicine Laboratory, Department of Biochemistry & Molecular Biology, Biomedicine and Health Graduate Education Innovation Center, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan, Shanxi, 030001, China
| | - Qing Ren
- Department of Gynecology, Hainan West Central Hospital, Danzhou, Hainan, 571700, China
| | - Zhiqiang Yan
- Department of Gynecology, Hainan West Central Hospital, Danzhou, Hainan, 571700, China
| | - Songtao Fu
- Birth Defects and Regenerative Medicine Laboratory, Department of Biochemistry & Molecular Biology, Biomedicine and Health Graduate Education Innovation Center, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan, Shanxi, 030001, China.
- Biomedicine and Health Graduate Education Innovation Center, Taiyuan, Shanxi, 030001, China.
| |
Collapse
|
6
|
Uscanga-Palomeque AC, Chávez-Escamilla AK, Alvizo-Báez CA, Saavedra-Alonso S, Terrazas-Armendáriz LD, Tamez-Guerra RS, Rodríguez-Padilla C, Alcocer-González JM. CAR-T Cell Therapy: From the Shop to Cancer Therapy. Int J Mol Sci 2023; 24:15688. [PMID: 37958672 PMCID: PMC10649325 DOI: 10.3390/ijms242115688] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer is a worldwide health problem. Nevertheless, new technologies in the immunotherapy field have emerged. Chimeric antigen receptor (CAR) technology is a novel biological form to treat cancer; CAR-T cell genetic engineering has positively revolutionized cancer immunotherapy. In this paper, we review the latest developments in CAR-T in cancer treatment. We present the structure of the different generations and variants of CAR-T cells including TRUCK (T cells redirected for universal cytokine killing. We explain the approaches of the CAR-T cells manufactured ex vivo and in vivo. Moreover, we describe the limitations and areas of opportunity for this immunotherapy and the current challenges of treating hematological and solid cancer using CAR-T technology as well as its constraints and engineering approaches. We summarize other immune cells that have been using CAR technology, such as natural killer (NK), macrophages (M), and dendritic cells (DC). We conclude that CAR-T cells have the potential to treat not only cancer but other chronic diseases.
Collapse
Affiliation(s)
- Ashanti Concepción Uscanga-Palomeque
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, Nuevo León, Mexico; (A.K.C.-E.); (C.A.A.-B.); (S.S.-A.); (L.D.T.-A.); (R.S.T.-G.); (C.R.-P.)
| | | | | | | | | | | | | | - Juan Manuel Alcocer-González
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, Nuevo León, Mexico; (A.K.C.-E.); (C.A.A.-B.); (S.S.-A.); (L.D.T.-A.); (R.S.T.-G.); (C.R.-P.)
| |
Collapse
|
7
|
Luksik AS, Yazigi E, Shah P, Jackson CM. CAR T Cell Therapy in Glioblastoma: Overcoming Challenges Related to Antigen Expression. Cancers (Basel) 2023; 15:cancers15051414. [PMID: 36900205 PMCID: PMC10000604 DOI: 10.3390/cancers15051414] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor, yet prognosis remains dismal with current treatment. Immunotherapeutic strategies have had limited effectiveness to date in GBM, but recent advances hold promise. One such immunotherapeutic advance is chimeric antigen receptor (CAR) T cell therapy, where autologous T cells are extracted and engineered to express a specific receptor against a GBM antigen and are then infused back into the patient. There have been numerous preclinical studies showing promising results, and several of these CAR T cell therapies are being tested in clinical trials for GBM and other brain cancers. While results in tumors such as lymphomas and diffuse intrinsic pontine gliomas have been encouraging, early results in GBM have not shown clinical benefit. Potential reasons for this are the limited number of specific antigens in GBM, their heterogenous expression, and their loss after initiating antigen-specific therapy due to immunoediting. Here, we review the current preclinical and clinical experiences with CAR T cell therapy in GBM and potential strategies to develop more effective CAR T cells for this indication.
Collapse
|
8
|
Mao R, Kong W, He Y. The affinity of antigen-binding domain on the antitumor efficacy of CAR T cells: Moderate is better. Front Immunol 2022; 13:1032403. [PMID: 36325345 PMCID: PMC9618871 DOI: 10.3389/fimmu.2022.1032403] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
The overall efficacy of chimeric antigen receptor modified T cells (CARTs) remain limited in solid tumors despite intensive studies that aim at targeting multiple antigens, enhancing migration, reducing tonic signaling, and improving tumor microenvironment. On the other hand, how the affinity and engaging kinetics of antigen-binding domain (ABD) affects the CART's efficacy has not been carefully investigated. In this article, we first analyzed 38 published solid tumor CART trials and correlated the response rate to their ABD affinity. Not surprisingly, majority (25 trials) of the CARTs utilized high-affinity ABDs, but generated merely 5.7% response rate. In contrast, 35% of the patients treated with the CARTs built from moderate-affinity ABDs had clinical responses. Thus, CARTs with moderate-affinity ABDs not only have less off-target toxicity, but also are more effective. We then reviewed the effects of ABD affinity on the biology and function of CARTs, providing further evidence that moderate-affinity ABDs may be better in CART development. In the end, we propose that a fast-on/fast-off (high Kon and Koff ) kinetics of CART-target engagement in solid tumor allow CARTs to generate sufficient signaling to kill tumor cells without being driven to exhaustion. We believe that studying the ABD affinity and the kinetics of CART-tumor interaction may hold a key to designing effective CARTs for solid tumors.
Collapse
Affiliation(s)
- Rui Mao
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Wanqing Kong
- South Carolina Governors School for Science and Math, Hartsville, SC, United States
| | - Yukai He
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
9
|
Karimi-Shahri M, Khorramdel M, Zarei S, Attarian F, Hashemian P, Javid H. Glioblastoma, an opportunity T cell trafficking could bring for the treatment. Mol Biol Rep 2022; 49:9863-9875. [DOI: 10.1007/s11033-022-07510-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/22/2022] [Indexed: 01/22/2023]
|
10
|
Chai LF, Hardaway JC, Heatherton KR, O'Connell KP, LaPorte JP, Guha P, Lopes MC, Rabinowitz BA, Jaroch D, Cox BF, Knight R, Katz SC. Regional Delivery of CAR-T Effectively Controls Tumor Growth in Colorectal Liver Metastasis Model. J Surg Res 2021; 272:37-50. [PMID: 34929499 DOI: 10.1016/j.jss.2021.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 10/01/2021] [Accepted: 11/05/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Effective treatment of solid tumors requires multi-modality approaches. In many patients with stage IV liver disease, current treatments are not curative. Chimeric antigen receptor T cells (CAR-T) are an intriguing option following success in hematological malignancies, but this has not been translated to solid tumors. Limitations include sub-optimal delivery and elevated interstitial fluid pressures. We developed a murine model to test the impact of high-pressure regional delivery (HPRD) on trafficking to liver metastases (LM) and tumor response. MATERIALS AND METHODS CAR-T were generated from CD45.1 mice and adoptively transferred into LM-bearing CD45.2 mice via regional or systemic delivery (RD, SD). Trafficking, tumor growth, and toxicity were evaluated with flow cytometry, tumor bioluminescence (TB, photons/sec log2-foldover baseline), and liver function tests (LFTs). RESULTS RD of CAR-T was more effective at controlling tumor growth versus SD from post-treatment days (PTD) 2-7 (P = 0.002). HPRD resulted in increased CAR-T penetration versus low-pressure RD (LPRD, P = 0.004), suppression of tumor proliferation (P = 0.03), and trended toward improved long-term control at PTD17 (TB=3.7 versus 6.1, P = 0.47). No LFT increase was noted utilizing HPRD versus LPRD (AST/ALT P = 0.65/0.84) while improved LFTs in RD versus SD groups suggested better tumor control (HPRD AST/ALT P = 0.04/0.04, LPRD AST/ALT P = 0.02/0.02). CONCLUSIONS Cellular immunotherapy is an emerging option for solid tumors. Our model suggests RD and HPRD improved CAR-T penetration into solid tumors with improved short-term tumor control. Barriers associated with SD can be overcome using RD techniques to maximize therapeutic delivery and HPRD may further augment efficacy without increased toxicity.
Collapse
Affiliation(s)
- Louis F Chai
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island.
| | - John C Hardaway
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island
| | - Kara R Heatherton
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island
| | - Kyle P O'Connell
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island
| | - Jason P LaPorte
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island
| | - Prajna Guha
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island
| | - Mikayla C Lopes
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island
| | - Benjamin A Rabinowitz
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island
| | - David Jaroch
- TriSalus Life Sciences, Inc, Westminster, Colorado
| | - Bryan F Cox
- TriSalus Life Sciences, Inc, Westminster, Colorado
| | | | - Steven C Katz
- Roger Williams Medical Center, Immuno-oncology Institute and Department of Medicine, Providence Rhode Island; Boston University Medical Center, Department of Surgery, Boston Massachusetts
| |
Collapse
|
11
|
Saif MW. From Screening to Treatment of Pancreatic Cancer: A Comprehensive Review. JOP : JOURNAL OF THE PANCREAS 2021; 22:70-79. [PMID: 34483790 PMCID: PMC8411391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Pancreatic adenocarcinoma is a devastating malignancy, associated with a grim prognosis, due to its silent presentation and lack of diagnostic tests. In addition, treatment options are limited to few agents, such as 5-FU, irinotecan, oxaliplatin, gemcitabine and nab-paclitaxel. METHODS We performed a literature search for relevant published clinical trials, abstracts of trials in progress and ongoing or planned trials for the treatment of APC using Pubmed.com, ClinicalTrials.gov and American Society of Clinical Oncology (ASCO) abstract search as sources. We present an in-depth analysis of the phase I-III clinical trials determining the role and efficacy of different modalities. We also describe rationale for future investigation. DISCUSSION Despite advances in first-line and second-line therapies for APC, median OS remains short of a year. We need collaborative efforts between the cooperative groups, institutions, community practices and industry to work together in enrolling these patients in clinical trials. In addition to use new technologies, such as organoids, we must pay attention to the palliative aspect of care for these patients from the beginning including nutritionist, social worker and supportive care health providers to assist with goals of care, symptom management and end of life discussions.
Collapse
Affiliation(s)
- Muhammad Wasif Saif
- Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine, Feinstein Institute for Medical Research, Lake Success, NY, USA
| |
Collapse
|
12
|
Hull CM, Maher J. Approaches for refining and furthering the development of CAR-based T cell therapies for solid malignancies. Expert Opin Drug Discov 2021; 16:1105-1117. [PMID: 34038292 DOI: 10.1080/17460441.2021.1929920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction: Chimeric antigen receptor-engineered T-cells typically use the binding domains of antibodies to target cytotoxicity toward tumors. This approach has produced great efficacy against selected hematological cancers, but benefit in solid tumors has been limited. Characteristically, the microenvironment in solid tumors restricts CAR T cell function, thereby limiting success. Enhancing efficacy will depend on novel target discovery to refine specificity and reduce toxicity. Additionally, overcoming immunosuppressive mechanisms may be achieved by altering the structure of the CAR itself, together with ancillary gene expression or additional therapeutic interventions.Areas covered: Herein, the authors discuss approaches for refining and further developing CAR T cell therapies specifically for use with solid malignancies. The authors survey the existing literature and provide perspectives for the future.Expert opinion: Pronounced efficacy in solid tumors will likely require combination therapies, targeting both the tumor itself and associated immunosuppressive mechanisms. Future exploration of CAR T cell therapies for solid tumors is likely to incorporate next-generation designs that couple more precise targeting of cancer-associated targets with enhanced potency and resistance to exhaustion.
Collapse
Affiliation(s)
| | - John Maher
- King's College London, Division of Cancer Studies, Guy's Hospital, London, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, UK.,Department of Immunology, Eastbourne Hospital, East Sussex, UK
| |
Collapse
|
13
|
CD28 Co-Stimulus Achieves Superior CAR T Cell Effector Function against Solid Tumors Than 4-1BB Co-Stimulus. Cancers (Basel) 2021; 13:cancers13051050. [PMID: 33801448 PMCID: PMC7958604 DOI: 10.3390/cancers13051050] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/02/2023] Open
Abstract
Spacer or co-stimulatory components in chimeric antigen receptor (CAR) design influence CAR T cell effector function. Few preclinical mouse models optimally support CAR candidate pre-selection for clinical development. Here we use a model in which murine CAR T cells can be exploited with human tumor xenografts. This mouse-in-mouse approach avoids limitations caused by species-specific factors crucial for CAR T cell survival, trafficking and function. We compared trafficking, expansion and tumor control for T cells expressing different CAR construct designs targeting two antigens (L1CAM or HER2), structurally identical except for spacer (long or short) or co-stimulatory (4-1BB or CD28) domains to be evaluated. Using monoclonal, murine-derived L1CAM-specific CAR T cells in Rag-/- mice harboring established xenografted tumors from a human neuroblastoma cell line revealed a clear superiority in CAR T cell trafficking using CD28 co-stimulation. L1CAM-targeting short spacer-CD28/ζ CAR T cells expanded the most at the tumor site and induced initial tumor regression. Treating patient-derived neuroblastoma xenografts with human L1CAM-targeting CAR T cells confirmed the superiority of CD28 co-stimulus. CD28 superiority was also demonstrated with HER2-specific CAR T cells (targeting ovarian carcinoma xenografts). Our findings encourage incorporating CD28 signaling into CAR design for adoptive T cell treatment of solid tumors.
Collapse
|
14
|
Stewart JH, Blazer DG, Calderon MJG, Carter TM, Eckhoff A, Al Efishat MA, Fernando DG, Foster JM, Hayes-Jordan A, Johnston FM, Lautz TB, Levine EA, Maduekwe UN, Mangieri CW, Moaven O, Mogal H, Shen P, Votanopoulos KI. The Evolving Management of Peritoneal Surface Malignancies. Curr Probl Surg 2020; 58:100860. [PMID: 33832580 DOI: 10.1016/j.cpsurg.2020.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023]
Affiliation(s)
| | - Dan G Blazer
- Division of Surgical Oncology, Duke University Medical Center, Durham, NC
| | | | | | | | | | | | - Jason M Foster
- Fred and Pamela Buffet Cancer Center, University of Nebraska, Omaha, NE
| | | | - Fabian M Johnston
- Complex General Surgical Oncology Program, Johns Hopkins University, Baltimore, MD
| | - Timothy B Lautz
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Ugwuji N Maduekwe
- Division of Surgical Oncology and Endocrine Surgery, University of North Carolina, Chapel Hill, NC
| | | | | | | | - Perry Shen
- Wake Forest University School of Medicine, Winston-Salem, NC
| | | |
Collapse
|
15
|
Balkhi MY. Receptor signaling, transcriptional, and metabolic regulation of T cell exhaustion. Oncoimmunology 2020; 9:1747349. [PMID: 32363117 PMCID: PMC7185212 DOI: 10.1080/2162402x.2020.1747349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/11/2020] [Accepted: 02/28/2020] [Indexed: 12/23/2022] Open
Abstract
Exhaustion cripples T cell effector responses against metastatic cancers and chronic infections alike. There has been considerable interest in understanding the molecular and cellular mechanisms driving T cell exhaustion in human cancers fueled by the success of immunotherapy drugs especially the checkpoint receptor blockade (CRB) inhibitory antibodies that reverses T cell functional exhaustion. The current understanding of molecular mechanism of T cell exhaustion has been elucidated from the studies utilizing murine models of chronic viral infections. These studies have formed the basis for much of our understanding of the process of exhaustion and proven vital to developing anti-exhaustion therapies against human cancers. In this review, we discuss the T cell exhaustion differentiation pathway in cancers and chronic viral infections and explore how the transcription factors expression dynamics play role in T cell exhaustion fate choices and maturation. Finally, we summarize the role of some of the most important transcription factors involved in T cell functional exhaustion and construct exhaustion specific signaling pathway maps.
Collapse
Affiliation(s)
- Mumtaz Y Balkhi
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME, USA.,Division of Hematology/Oncology Tufts Medical Center and Tufts University School of Medicine, Boston, MA, USA.,Immune Therapy Bio, Nest.Bio Labs, Vassar St. Cambridge, MA, USA
| |
Collapse
|
16
|
Pancreatic Cancer UK Grand Challenge: Developments and challenges for effective CAR T cell therapy for pancreatic ductal adenocarcinoma. Pancreatology 2020; 20:394-408. [PMID: 32173257 DOI: 10.1016/j.pan.2020.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
Death from pancreatic ductal adenocarcinoma (PDAC) is rising across the world and PDAC is predicted to be the second most common cause of cancer death in the USA by 2030. Development of effective biotherapies for PDAC are hampered by late presentation, a low number of differentially expressed molecular targets and a tumor-promoting microenvironment that forms both a physical, collagen-rich barrier and is also immunosuppressive. In 2017 Pancreatic Cancer UK awarded its first Grand Challenge Programme award to tackle this problem. The team plan to combine the use of novel CAR T cells with strategies to overcome the barriers presented by the tumor microenvironment. In advance of publication of those data this review seeks to highlight the key problems in effective CAR T cell therapy of PDAC and to describe pre-clinical and clinical progress in CAR T bio-therapeutics.
Collapse
|
17
|
Hardy IR, Schamel WW, Baeuerle PA, Getts DR, Hofmeister R. Implications of T cell receptor biology on the development of new T cell therapies for cancer. Immunotherapy 2020; 12:89-103. [PMID: 31902264 DOI: 10.2217/imt-2019-0046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recently, two chimeric antigen receptor (CAR) T cell therapies were approved based on their remarkable efficacy in patients with hematological malignancies. By contrast, CAR-T cell therapies results in solid tumors have been less promising. To develop the next generation of T cell therapies a better understanding of T cell receptor (TCR) biology and its implication for the design of synthetic receptors is critical. Here, we review current and newly developed forms of T cell therapies and how their utilization of different components of the TCR signaling machinery and their requirement for engagement (or not) of human leukocyte antigen impacts their design, efficacy and applicability as cancer drugs. Notably, we highlight the development of human leukocyte antigen-independent T cell platforms that utilize the full TCR complex as having promise to overcome some of the limitations of existing T cell therapies.
Collapse
Affiliation(s)
- Ian R Hardy
- TCR2 Therapeutics, Inc., 100 Binney St. Suite 710, Cambridge, MA 02142, USA
| | - Wolfgang W Schamel
- Department of Immunology, Faculty of Biology, BIOSS Center for Biological Signalling Studies, CIBSS - Centre for Integrative Biological Signalling Studies & Centre for Chronic Immunodeficiency CCI, University of Freiburg, Schänzlestraβe 18,79104 Freiburg, Germany
| | - Patrick A Baeuerle
- TCR Therapeutics, Inc., 100 Binney St. Suite 710, Cambridge, MA 02142, USA.,Institute for Immunology, Ludwig-Maximilians-University Munich, Grosshadernerstr. 9, 82152 Planegg-Martinsried, Germany
| | - Daniel R Getts
- TCR Therapeutics, Inc., 100 Binney St. Suite 710, Cambridge, MA 02142, USA
| | - Robert Hofmeister
- TCR Therapeutics, Inc., 100 Binney St. Suite 710, Cambridge, MA 02142, USA
| |
Collapse
|
18
|
Clinical investigation of CAR T cells for solid tumors: Lessons learned and future directions. Pharmacol Ther 2020; 205:107419. [DOI: 10.1016/j.pharmthera.2019.107419] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022]
|
19
|
Gershovich PM, Karabelskii AV, Ulitin AB, Ivanov RA. The Role of Checkpoint Inhibitors and Cytokines in Adoptive Cell-Based Cancer Immunotherapy with Genetically Modified T Cells. BIOCHEMISTRY (MOSCOW) 2019; 84:695-710. [PMID: 31509722 DOI: 10.1134/s0006297919070022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This review focuses on the structure and molecular action mechanisms of chimeric antigen receptors (CARs) and major aspects of the manufacturing and clinical application of products for the CAR-T (CAR-modified T lymphocyte) therapy of hematological and solid tumors with special emphasis on the strategies for combined use of CAR-T therapy with immuno-oncological monoclonal antibodies (checkpoint inhibitors) and cytokines to boost survival, persistence, and antitumor efficacy of CAR-T therapy. The review also summarizes preclinical and clinical data on the additive effects of the combined use of CAR-T therapy with interleukins and monoclonal antibodies targeting immune checkpoints.
Collapse
Affiliation(s)
- P M Gershovich
- CJSC Biocad, St. Petersburg, 198515, Russia. .,St. Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, 197376, Russia
| | - A V Karabelskii
- CJSC Biocad, St. Petersburg, 198515, Russia.,St. Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, 197376, Russia
| | - A B Ulitin
- CJSC Biocad, St. Petersburg, 198515, Russia
| | - R A Ivanov
- CJSC Biocad, St. Petersburg, 198515, Russia
| |
Collapse
|
20
|
Aranda F, Barajas M, Huarte E. Transgenic Tumor Models for Evaluating CAR T-Cell Immunotherapies. ACTA ACUST UNITED AC 2019; 86:e66. [PMID: 31539924 DOI: 10.1002/cpph.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy against tumor antigens involves a recombinant immunoreceptor that combines an antibody-derived targeting fragment with signaling domains capable of activating T cells and fusion of this receptor domain to a costimulatory domain (typically CD28 or 4-1BB). Clinical trials of CAR T-cell therapeutics targeting CD19 antigens for relapsed or refractory B-cell malignancies have shown unparalleled results and consequently have recently been approved by the U.S. Food and Drug Administration. However, the lack of efficacy beyond B-cell malignancies, the emergence of resistance to CAR T-cell therapy due to loss of the antigenic epitope, and severe cases of cytokine release syndrome and neurotoxicity necessitate further preclinical studies. As it is very complicated to develop a single animal model that would replicate the complexity of the clinical scenario, this article focuses on transgenic models used to study human tumor-associated antigens in an immunocompetent model. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Fernando Aranda
- Group of Immunoreceptors of the Innate and Adaptive System, Institute d'Investigacions Biomédiques August pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Miguel Barajas
- Biochemistry Area, Health Science Department, Faculty of Health Sciences, Public University of Navarra, Pamplona, Spain
| | | |
Collapse
|
21
|
Albert S, Koristka S, Gerbaulet A, Cartellieri M, Arndt C, Feldmann A, Berndt N, Loureiro LR, von Bonin M, Ehninger G, Eugster A, Bonifacio E, Bornhäuser M, Bachmann MP, Ehninger A. Tonic Signaling and Its Effects on Lymphopoiesis of CAR-Armed Hematopoietic Stem and Progenitor Cells. THE JOURNAL OF IMMUNOLOGY 2019; 202:1735-1746. [PMID: 30728213 DOI: 10.4049/jimmunol.1801004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/04/2019] [Indexed: 01/01/2023]
Abstract
Long-term survival of adoptively transferred chimeric Ag receptor (CAR) T cells is often limited. Transplantation of hematopoietic stem cells (HSCs) transduced to express CARs could help to overcome this problem as CAR-armed HSCs can continuously deliver CAR+ multicell lineages (e.g., T cells, NK cells). In dependence on the CAR construct, a variable extent of tonic signaling in CAR T cells was reported; thus, effects of CAR-mediated tonic signaling on the hematopoiesis of CAR-armed HSCs is unclear. To assess the effects of tonic signaling, two CAR constructs were established and analyzed 1) a signaling CAR inducing a solid Ag-independent tonic signaling termed CAR-28/ζ and 2) a nonstimulating control CAR construct lacking intracellular signaling domains termed CAR-Stop. Bone marrow cells from immunocompetent mice were isolated, purified for HSC-containing Lin-cKit+ cells or the Lin-cKit+ Sca-1+ subpopulation (Lin-Sca-1+cKit+), and transduced with both CAR constructs. Subsequently, modified bone marrow cells were transferred into irradiated mice, in which they successfully engrafted and differentiated into hematopoietic progenitors. HSCs expressing the CAR-Stop sustained normal hematopoiesis. In contrast, expression of the CAR-28/ζ led to elimination of mature CAR+ T and B cells, suggesting that the CAR-mediated tonic signaling mimics autorecognition via the newly recombined immune receptors in the developing lymphocytes.
Collapse
Affiliation(s)
- Susann Albert
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.,University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Stefanie Koristka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Alexander Gerbaulet
- Institute of Immunology, Medical Faculty Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | | | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Nicole Berndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Liliana R Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Malte von Bonin
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Gerhard Ehninger
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Anne Eugster
- Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, 01307 Dresden, Germany; and
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, 01307 Dresden, Germany; and
| | - Martin Bornhäuser
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Michael P Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; .,University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Armin Ehninger
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,GEMoaB Monoclonals GmbH, 01307 Dresden, Germany
| |
Collapse
|
22
|
The challenges of solid tumor for designer CAR-T therapies: a 25-year perspective. Cancer Gene Ther 2019; 24:89-99. [PMID: 28392558 DOI: 10.1038/cgt.2016.82] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
23
|
Shirjang S, Alizadeh N, Mansoori B, Mahmoodpoor A, Kafil HS, Hojjat-Farsangi M, Yousefi M. Promising immunotherapy: Highlighting cytokine-induced killer cells. J Cell Biochem 2018; 120:8863-8883. [PMID: 30556298 DOI: 10.1002/jcb.28250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
For many years, cancer therapy has appeared to be a challenging issue for researchers and physicians. By the introduction of novel methods in immunotherapy, the prospect of cancer therapy even more explained than before. Cytokine-induced killer (CIK) cell-based immunotherapy demonstrated to have potentiality in improving clinical outcomes and relieving major side effects of standard treatment options. In addition, given the distinctive features such as high safety, low toxicity effects on healthy cells, numerous clinical trials conducted on CIK cells. Due to the shortcomings that observed in CIK cell immunotherapy alone, arising a tendency to make modifications (combined modality therapy or combination therapy) including the addition of various types of cytokines, genetic engineering, combination with immune checkpoints, and so on. In this review, we have tried to bring forth the latest immunotherapy methods and their overview. We have discussed the combination therapies with CIK cells and the conducted clinical trials. This helps the future studies to use integrated therapies with CIK cells as a promising treatment of many types of cancers.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 2018; 564:268-272. [PMID: 30479382 DOI: 10.1038/s41586-018-0694-x] [Citation(s) in RCA: 856] [Impact Index Per Article: 122.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 10/19/2018] [Indexed: 01/26/2023]
Abstract
T cells are key elements of cancer immunotherapy1 but certain fundamental properties, such as the development and migration of T cells within tumours, remain unknown. The enormous T cell receptor (TCR) repertoire, which is required for the recognition of foreign and self-antigens2, could serve as lineage tags to track these T cells in tumours3. Here we obtained transcriptomes of 11,138 single T cells from 12 patients with colorectal cancer, and developed single T cell analysis by RNA sequencing and TCR tracking (STARTRAC) indices to quantitatively analyse the dynamic relationships among 20 identified T cell subsets with distinct functions and clonalities. Although both CD8+ effector and 'exhausted' T cells exhibited high clonal expansion, they were independently connected with tumour-resident CD8+ effector memory cells, implicating a TCR-based fate decision. Of the CD4+ T cells, most tumour-infiltrating T regulatory (Treg) cells showed clonal exclusivity, whereas certain Treg cell clones were developmentally linked to several T helper (TH) cell clones. Notably, we identified two IFNG+ TH1-like cell clusters in tumours that were associated with distinct IFNγ-regulating transcription factors -the GZMK+ effector memory T cells, which were associated with EOMES and RUNX3, and CXCL13+BHLHE40+ TH1-like cell clusters, which were associated with BHLHE40. Only CXCL13+BHLHE40+ TH1-like cells were preferentially enriched in patients with microsatellite-instable tumours, and this might explain their favourable responses to immune-checkpoint blockade. Furthermore, IGFLR1 was highly expressed in both CXCL13+BHLHE40+ TH1-like cells and CD8+ exhausted T cells and possessed co-stimulatory functions. Our integrated STARTRAC analyses provide a powerful approach to dissect the T cell properties in colorectal cancer comprehensively, and could provide insights into the dynamic relationships of T cells in other cancers.
Collapse
|
25
|
Sengupta S, Katz SC, Sengupta S, Sampath P. Glycogen synthase kinase 3 inhibition lowers PD-1 expression, promotes long-term survival and memory generation in antigen-specific CAR-T cells. Cancer Lett 2018; 433:131-139. [PMID: 29959057 DOI: 10.1016/j.canlet.2018.06.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/07/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022]
Abstract
Successful remission in hematological cancers by CAR-T cell immunotherapy has yet to be replicated in solid tumors like GBM. A significant impediment of CAR-T immunotherapy in solid tumors is poor exposure of T cells to tumor antigens resulting in suboptimal CAR-T cell activation, which ultimately fails to induce a robust anti-tumor immune response. Costimulatory moieties in advanced-generation CARs, along with additional IL2 therapy has been shown to be insufficient to overcome this hurdle and have its cytotoxic limitations. GSK3 is constitutively active in naïve T cells and is transiently inactivated during T cell activation resulting in rapid T cell proliferation. Pharmacologic inhibition of GSK3 in GBM-specific CAR-T cells reduced FasL expression, increased T cell proliferation and reduced exhaustion by lowering PD-1 levels resulting in the development of CAR-T effector memory phenotype. Treatment with GSK3-inhibited CAR-T cells resulted in 100% tumor elimination during the tumor-rechallenge experiment in GBM-bearing animals and increased accumulation of memory CAR-T cells in secondary lymphoid organs. These adjuvant-like effects of GSK3 inhibition on activated CAR-T cells may be a valuable adjunct to a successful implementation of CAR-T immunotherapy against GBM and other solid tumors.
Collapse
Affiliation(s)
- Sadhak Sengupta
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, RI, USA; Department of Neurosurgery, Alpert School of Medicine, Brown University, Providence, RI, USA.
| | - Steven C Katz
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA; Department of Surgery, Boston University School of Medicine, Boston, MA, USA
| | | | - Prakash Sampath
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, RI, USA; Department of Neurosurgery, Alpert School of Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
26
|
Balkhi MY, Wittmann G, Xiong F, Junghans RP. YY1 Upregulates Checkpoint Receptors and Downregulates Type I Cytokines in Exhausted, Chronically Stimulated Human T Cells. iScience 2018; 2:105-122. [PMID: 30428369 PMCID: PMC6136936 DOI: 10.1016/j.isci.2018.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/31/2018] [Accepted: 02/22/2018] [Indexed: 12/26/2022] Open
Abstract
T cells infiltrate affected organs in chronic infections and malignancy, but they may fail to eradicate virus-infected cells or tumor because of exhaustion. This report describes a Yin Yang-1 (YY1)-centered mechanism for diverse components that have been correlated with exhaustion. Utilizing an in vitro reconstruction of chronic T cell activation, YY1 is shown to positively regulate the checkpoint receptors PD1, Lag3, and Tim3 and to negatively regulate the type I cytokines interleukin-2 (IL-2) (in collaboration with Ezh2 histone methyltransferase) and interferon gamma (IFN-?). Other tests suggest that IL-2 failure drives a large component of cytotoxic functional decline rather than solely checkpoint receptor-ligand interactions that have been the focus of current anti-exhaustion therapies. Clinical evaluations confirm elevated YY1 and Ezh2 in melanoma tumor-infiltrating lymphocytes and in PD1+ T cells in patients with HIV. Exhaustion is revealed to be an active process as the culmination of repetitive two-signal stimulation in a feedback loop via CD3/CD28?p38MAPK/JNK?YY1? exhaustion.
Collapse
Affiliation(s)
- Mumtaz Y Balkhi
- Biotherapeutics Development Lab, Division of Hematology/Oncology, Department of Medicine, Tufts University School of Medicine, 800 Washington St, Boston, MA 02111, USA
| | - Gabor Wittmann
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Fang Xiong
- Biotherapeutics Development Lab, Division of Hematology/Oncology, Department of Medicine, Tufts University School of Medicine, 800 Washington St, Boston, MA 02111, USA
| | - Richard P Junghans
- Biotherapeutics Development Lab, Division of Hematology/Oncology, Department of Medicine, Tufts University School of Medicine, 800 Washington St, Boston, MA 02111, USA.
| |
Collapse
|
27
|
Dalal AR, Homsy S, Balkhi MY. Third-Generation Human Epidermal Growth Factor Receptor 2 Chimeric Antigen Receptor Expression on Human T Cells Improves with Two-Signal Activation. Hum Gene Ther 2018; 29:845-852. [PMID: 29373929 DOI: 10.1089/hum.2017.244] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Patient derived T cells activated ex vivo with CD3/CD28 beads show superior expansion. Therefore, CD3/CD28 beads have huge potential to be used in the clinic for immunotherapy applications. Two protocols were devised to evaluate if the expression of third-generation human epidermal growth factor receptor 2 chimeric antigen receptor (CAR) can be improved on human T cells activated with CD3/CD28 beads. In protocol 1, unconcentrated human epidermal growth factor receptor 2 CAR retroviral supernatants were used, and in protocol 2, concentrated virus was used. The results demonstrate that compared to unconcentrated viral supernatants, transduction with the concentrated virus improved the infection rate of bead activated CD4 T cells from ∼40% to ∼70%, and the fluorescent intensity values improved from ∼12,000 to ∼28,000 mean fluorescence intensity units. These results demonstrate the utility of these protocols for CAR immunotherapies.
Collapse
Affiliation(s)
- Al-Rubaye Dalal
- Division of Hematology/Oncology, Tufts University School of Medicine, Boston, Massachusetts.,Division of Biomedical Engineering, Boston University, Boston, Massachusetts.,Biotechnology Department, College of Science, University of Baghdad, Baghdad, Iraq
| | - Sylvester Homsy
- Division of Hematology/Oncology, Tufts University School of Medicine, Boston, Massachusetts
| | - Mumtaz Y Balkhi
- Division of Hematology/Oncology, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
28
|
Yeku O, Li X, Brentjens RJ. Adoptive T-Cell Therapy for Solid Tumors. AMERICAN SOCIETY OF CLINICAL ONCOLOGY EDUCATIONAL BOOK. AMERICAN SOCIETY OF CLINICAL ONCOLOGY. ANNUAL MEETING 2017. [PMID: 28561728 DOI: 10.14694/edbk_180328] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an innovative form of immunotherapy wherein autologous T cells are genetically modified to express chimeric receptors encoding an antigen-specific single-chain variable fragment and various costimulatory molecules. Upon administration, these modified T cells traffic to, and recognize, cancer cells in an HLA-independent manner. CAR T-cell therapy has shown remarkable success in the treatment of CD-19-expressing B-cell acute lymphocytic leukemia. However, clinical gains to the same magnitude have not been reported in solid tumors. Several known obstacles to CAR T-cell therapy for solid tumors include target antigen identification, effective trafficking to the tumor, robust activation, proliferation, and in vivo cytotoxicity. Beyond these T-cell intrinsic properties, a complex and dynamic immunosuppressive tumor microenvironment in solid tumors hinders T-cell efficacy. Notable advancements in CAR design to include multiple costimulatory molecules, ligands, and soluble cytokines have shown promise in preclinical models, and some of these are currently in early-phase clinical trials. In this review, we discuss selected solid tumor malignancies and relevant preclinical data and highlight clinical trial results that are available. Furthermore, we outline some obstacles to CAR T-cell therapy for each tumor and propose strategies to overcome some of these limitations.
Collapse
Affiliation(s)
- Oladapo Yeku
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xinghuo Li
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J Brentjens
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
29
|
Yeku O, Li X, Brentjens RJ. Adoptive T-Cell Therapy for Solid Tumors. Am Soc Clin Oncol Educ Book 2017; 37:193-204. [PMID: 28561728 DOI: 10.1200/edbk_180328] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an innovative form of immunotherapy wherein autologous T cells are genetically modified to express chimeric receptors encoding an antigen-specific single-chain variable fragment and various costimulatory molecules. Upon administration, these modified T cells traffic to, and recognize, cancer cells in an HLA-independent manner. CAR T-cell therapy has shown remarkable success in the treatment of CD-19-expressing B-cell acute lymphocytic leukemia. However, clinical gains to the same magnitude have not been reported in solid tumors. Several known obstacles to CAR T-cell therapy for solid tumors include target antigen identification, effective trafficking to the tumor, robust activation, proliferation, and in vivo cytotoxicity. Beyond these T-cell intrinsic properties, a complex and dynamic immunosuppressive tumor microenvironment in solid tumors hinders T-cell efficacy. Notable advancements in CAR design to include multiple costimulatory molecules, ligands, and soluble cytokines have shown promise in preclinical models, and some of these are currently in early-phase clinical trials. In this review, we discuss selected solid tumor malignancies and relevant preclinical data and highlight clinical trial results that are available. Furthermore, we outline some obstacles to CAR T-cell therapy for each tumor and propose strategies to overcome some of these limitations.
Collapse
Affiliation(s)
- Oladapo Yeku
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xinghuo Li
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J Brentjens
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
30
|
Yakoub-Agha I, Ferrand C, Chalandon Y, Ballot C, Castilla Llorente C, Deschamps M, Gauthier J, Labalette M, Larghero J, Maheux C, Moreau AS, Varlet P, Pétillon MO, Pinturaud M, Rubio MT, Chabannon C. Prérequis nécessaires pour la mise en place de protocoles de recherche clinique évaluant des thérapies cellulaires et géniques par lymphocytes T dotés de récepteur chimérique à l’antigène (CAR T-cells) : recommandations de la Société francophone de greffe de moelle et de thérapie cellulaire (SFGM-TC). Bull Cancer 2017; 104:S43-S58. [DOI: 10.1016/j.bulcan.2017.10.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 11/30/2022]
|
31
|
Huang Y, Li D, Qin DY, Gou HF, Wei W, Wang YS, Wei YQ, Wang W. Interleukin-armed chimeric antigen receptor-modified T cells for cancer immunotherapy. Gene Ther 2017; 25:192-197. [DOI: 10.1038/gt.2017.81] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 04/10/2017] [Accepted: 07/28/2017] [Indexed: 01/01/2023]
|
32
|
Guha P, Cunetta M, Somasundar P, Espat NJ, Junghans RP, Katz SC. Frontline Science: Functionally impaired geriatric CAR-T cells rescued by increased α5β1 integrin expression. J Leukoc Biol 2017; 102:201-208. [PMID: 28546503 DOI: 10.1189/jlb.5hi0716-322rr] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 03/22/2017] [Accepted: 04/12/2017] [Indexed: 12/20/2022] Open
Abstract
Chimeric antigen receptor expressing T cells (CAR-T) are a promising form of immunotherapy, but the influence of age-related immune changes on CAR-T production remains poorly understood. We showed that CAR-T cells from geriatric donors (gCAR-T) are functionally impaired relative to CAR-T from younger donors (yCAR-T). Higher transduction efficiencies and improved cell expansion were observed in yCAR-T cells compared with gCAR-T. yCAR-T demonstrated significantly increased levels of proliferation and signaling activation of phosphorylated (p)Erk, pAkt, pStat3, and pStat5. Furthermore, yCAR-T contained higher proportions of CD4 and CD8 effector memory (EM) cells, which are known to have enhanced cytolytic capabilities. Accordingly, yCAR-T demonstrated higher levels of tumor antigen-specific cytotoxicity compared with gCAR-T. Enhanced tumor killing by yCAR-T correlated with increased levels of perforin and granzyme B. yCAR-T had increased α5β1 integrin expression, a known mediator of retroviral transduction. We found that treatment with M-CSF or TGF-β1 rescued the impaired transduction efficiency of the gCAR-T by increasing the α5β1 integrin expression. Neutralization of α5β1 confirmed that this integrin was indispensable for CAR expression. Our study suggests that the increase of α5β1 integrin expression levels enhances CAR expression and thereby improves tumor killing by gCAR-T.
Collapse
Affiliation(s)
- Prajna Guha
- Division of Surgical Oncology, Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA; and
| | - Marissa Cunetta
- Division of Surgical Oncology, Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA; and
| | - Ponnandai Somasundar
- Division of Surgical Oncology, Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA; and
| | - N Joseph Espat
- Division of Surgical Oncology, Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA; and
| | - Richard P Junghans
- Division of Surgical Oncology, Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA; and
| | - Steven C Katz
- Division of Surgical Oncology, Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA; and .,Department of Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Immunosuppression in liver tumors: opening the portal to effective immunotherapy. Cancer Gene Ther 2016; 24:114-120. [DOI: 10.1038/cgt.2016.54] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 12/11/2022]
|
34
|
Zhang Q, Zhang Z, Peng M, Fu S, Xue Z, Zhang R. CAR-T cell therapy in gastrointestinal tumors and hepatic carcinoma: From bench to bedside. Oncoimmunology 2016; 5:e1251539. [PMID: 28123893 PMCID: PMC5214859 DOI: 10.1080/2162402x.2016.1251539] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
The chimeric antigen receptor (CAR) is a genetically engineered receptor that combines a scFv domain, which specifically recognizes the tumor-specific antigen, with T cell activation domains. CAR-T cell therapies have demonstrated tremendous efficacy against hematologic malignancies in many clinical trials. Recent studies have extended these efforts to the treatment of solid tumors. However, the outcomes of CAR-T cell therapy for solid tumors are not as remarkable as the outcomes have been for hematologic malignancies. A series of hurdles has arisen with respect to CAR-T cell-based immunotherapy, which needs to be overcome to target solid tumors. The major challenge for CAR-T cell therapy in solid tumors is the selection of the appropriate specific antigen to demarcate the tumor from normal tissue. In this review, we discuss the application of CAR-T cells to gastrointestinal and hepatic carcinomas in preclinical and clinical research. Furthermore, we analyze the usefulness of several specific markers in the study of gastrointestinal tumors and hepatic carcinoma.
Collapse
Affiliation(s)
- Qi Zhang
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| | - Zimu Zhang
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| | - Meiyu Peng
- Department of Immunology, Basic Medical College, Weifang Medical University , Weifang, China
| | - Shuyu Fu
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| | - Zhenyi Xue
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| | - Rongxin Zhang
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
35
|
Junghans RP, Ma Q, Rathore R, Gomes EM, Bais AJ, Lo ASY, Abedi M, Davies RA, Cabral HJ, Al-Homsi AS, Cohen SI. Phase I Trial of Anti-PSMA Designer CAR-T Cells in Prostate Cancer: Possible Role for Interacting Interleukin 2-T Cell Pharmacodynamics as a Determinant of Clinical Response. Prostate 2016; 76:1257-70. [PMID: 27324746 DOI: 10.1002/pros.23214] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/17/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-modified "designer" T cells (dTc, CAR-T) against PSMA selectively target antigen-expressing cells in vitro and eliminate tumors in vivo. Interleukin 2 (IL2), widely used in adoptive therapies, was proven essential in animal models for dTc to eradicate established solid tumors. METHODS Patients under-went chemotherapy condi-tion-ing, followed by dTc dosing under a Phase I escalation with continuous infusion low dose IL2 (LDI). A target of dTc escalation was to achieve ≥20% engraftment of infused activated T cells. RESULTS Six patients enrolled with doses prepared of whom five were treated. Patients received 10(9) or 10(10) autologous T cells, achieving expansions of 20-560-fold over 2 weeks and engraftments of 5-56%. Pharmacokinetic and pharmacodynamic analyses established the impact of conditioning to promote expansion and engraftment of the infused T cells. Unexpectedly, administered IL2 was depleted up to 20-fold with high engraftments of activated T cells (aTc) in an inverse correlation (P < 0.01). Clinically, no anti-PSMA toxicities were noted, and no anti-CAR reactivities were detected post-treatment. Two-of-five patients achieved clinical partial responses (PR), with PSA declines of 50% and 70% and PSA delays of 78 and 150 days, plus a minor response in a third patient. Responses were unrelated to dose size (P = 0.6), instead correlating inversely with engraftment (P = 0.06) and directly with plasma IL2 (P = 0.03), suggesting insufficient IL2 with our LDI protocol to support dTc anti-tumor activity under optimal (high) dTc engraftments. CONCLUSIONS Under a Phase I dose escalation in prostate cancer, a 20% engraftment target was met or exceeded in three subjects with adequate safety, leading to study conclusion. Clinical responses were obtained but were suggested to be restrained by low plasma IL2 when depleted by high levels of engrafted activated T cells. This report presents a unique example of how the pharmaco-dynamics of "drug-drug" interactions may have a critical impact on the efficacy of their co-application. A new Pilot/Phase II trial is planned to test moderate dose IL2 (MDI) together with high dTc engraftments for anticipated improved therapeutic efficacy. Prostate 76:1257-1270, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Richard P Junghans
- Division of Hematology-Oncology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
- Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Qiangzhong Ma
- Division of Hematology-Oncology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
- Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Ritesh Rathore
- Division of Hematology-Oncology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | - Erica M Gomes
- Division of Hematology-Oncology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | - Anthony J Bais
- Division of Hematology-Oncology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | - Agnes S Y Lo
- Division of Hematology-Oncology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
- Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Mehrdad Abedi
- Division of Hematology-Oncology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | - Robin A Davies
- Protocol Office, Roger Williams Medical Center, Providence, Rhode Island
| | - Howard J Cabral
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - A Samer Al-Homsi
- Division of Hematologic Malignancies and Blood and Marrow Transplantation, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | - Stephen I Cohen
- Division of Urology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| |
Collapse
|
36
|
Wang L, Ma N, Okamoto S, Amaishi Y, Sato E, Seo N, Mineno J, Takesako K, Kato T, Shiku H. Efficient tumor regression by adoptively transferred CEA-specific CAR-T cells associated with symptoms of mild cytokine release syndrome. Oncoimmunology 2016; 5:e1211218. [PMID: 27757303 PMCID: PMC5048773 DOI: 10.1080/2162402x.2016.1211218] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/29/2016] [Accepted: 07/03/2016] [Indexed: 01/01/2023] Open
Abstract
Carcinoembryonic antigen (CEA) is a cell surface antigen highly expressed in various cancer cell types and in healthy tissues. It has the potential to be a target for chimeric antigen receptor (CAR)-modified T-cell therapy; however, the safety of this approach in terms of on-target/off-tumor effects needs to be determined. To address this issue in a clinically relevant model, we used a mouse model in which the T cells expressing CEA-specific CAR were transferred into tumor-bearing CEA-transgenic (Tg) mice that physiologically expressed CEA as a self-antigen. The adoptive transfer in conjunction with lymphodepleting and myeloablative preconditioning mediated significant tumor regression but caused weight loss in CEA-Tg, but not in wild-type mice. The weight loss was not associated with overt inflammation in the CEA-expressing gastrointestinal tract but was associated with malnutrition, reflected in elevated systemic levels of cytokines linked to anorexia, which could be controlled by the administration of an anti-IL-6 receptor monoclonal antibody without compromising efficacy. The apparent relationship between lymphodepleting and myeloablative preconditioning, efficacy, and off-tumor toxicity of CAR-T cells would necessitate the development of CEA-specific CAR-T cells with improved signaling domains that require less stringent preconditioning for their efficacy. Taken together, these results suggest that CEA-specific CAR-based adoptive T-cell therapy may be effective for patients with CEA+ solid tumors. Distinguishing the fine line between therapeutic efficacy and off-tumor toxicity would involve further modifications of CAR-T cells and preconditioning regimens.
Collapse
Affiliation(s)
- Linan Wang
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine , Tsu, Mie, Japan
| | - Ning Ma
- Faculty of Nursing Science, Suzuka Medical Science University , Suzuka, Mie, Japan
| | | | | | - Eiichi Sato
- Department of Pathology, Institute of Medical Science, Medical Research Center, Tokyo Medical University , Tokyo, Japan
| | - Naohiro Seo
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine , Tsu, Mie, Japan
| | | | - Kazutoh Takesako
- Department of Pathology, Institute of Medical Science, Medical Research Center, Tokyo Medical University , Tokyo, Japan
| | - Takuma Kato
- Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Tsu, Mie, Japan; Center for Comprehensive Cancer Immunotherapy, Mie University, Tsu, Mie, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Mie, Japan; Center for Comprehensive Cancer Immunotherapy, Mie University, Tsu, Mie, Japan
| |
Collapse
|
37
|
Di S, Li Z. Treatment of solid tumors with chimeric antigen receptor-engineered T cells: current status and future prospects. SCIENCE CHINA-LIFE SCIENCES 2016; 59:360-9. [PMID: 26968709 DOI: 10.1007/s11427-016-5025-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/19/2016] [Indexed: 01/01/2023]
Abstract
Chimeric antigen receptors (CARs) are artificial recombinant receptors that generally combine the antigen-recognition domain of a monoclonal antibody with T cell activation domains. Recent years have seen great success in clinical trials employing CD19-specific CAR-T cell therapy for B cell leukemia. Nevertheless, solid tumors remain a major challenge for CAR-T cell therapy. This review summarizes the preclinical and clinical studies on the treatment of solid tumors with CAR-T cells. The major hurdles for the success of CAR-T and the novel strategies to address these hurdles have also been described and discussed.
Collapse
Affiliation(s)
- Shengmeng Di
- State Key Laboratory of Oncogenes & Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes & Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China.
| |
Collapse
|
38
|
Morello A, Sadelain M, Adusumilli PS. Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors. Cancer Discov 2015; 6:133-46. [PMID: 26503962 DOI: 10.1158/2159-8290.cd-15-0583] [Citation(s) in RCA: 372] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/08/2015] [Indexed: 12/19/2022]
Abstract
UNLABELLED Chimeric antigen receptors (CAR) are synthetic receptors that target T cells to cell-surface antigens and augment T-cell function and persistence. Mesothelin is a cell-surface antigen implicated in tumor invasion, which is highly expressed in mesothelioma and lung, pancreas, breast, ovarian, and other cancers. Its low-level expression in mesothelia, however, commands thoughtful therapeutic interventions. Encouragingly, recent clinical trials evaluating active immunization or immunoconjugates in patients with pancreatic adenocarcinoma or mesothelioma have shown responses without toxicity. Altogether, these findings and preclinical CAR therapy models using either systemic or regional T-cell delivery argue favorably for mesothelin CAR therapy in multiple solid tumors. SIGNIFICANCE Recent success obtained with adoptive transfer of CAR T cells targeting CD19 in patients with refractory hematologic malignancies has generated much enthusiasm for T-cell engineering and raises the prospect of implementing similar strategies for solid tumors. Mesothelin is expressed in a wide range and a high percentage of solid tumors, which we review here in detail. Mesothelin CAR therapy has the potential to treat multiple solid malignancies.
Collapse
Affiliation(s)
- Aurore Morello
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Prasad S Adusumilli
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York. Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
39
|
Katz SC, Burga RA, McCormack E, Wang LJ, Mooring W, Point GR, Khare PD, Thorn M, Ma Q, Stainken BF, Assanah EO, Davies R, Espat NJ, Junghans RP. Phase I Hepatic Immunotherapy for Metastases Study of Intra-Arterial Chimeric Antigen Receptor-Modified T-cell Therapy for CEA+ Liver Metastases. Clin Cancer Res 2015; 21:3149-59. [PMID: 25850950 PMCID: PMC4506253 DOI: 10.1158/1078-0432.ccr-14-1421] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 03/23/2015] [Indexed: 12/24/2022]
Abstract
PURPOSE Chimeric antigen receptor-modified T cells (CAR-T) have demonstrated encouraging results in early-phase clinical trials. Successful adaptation of CAR-T technology for CEA-expressing adenocarcinoma liver metastases, a major cause of death in patients with gastrointestinal cancers, has yet to be achieved. We sought to test intrahepatic delivery of anti-CEA CAR-T through percutaneous hepatic artery infusions (HAIs). EXPERIMENTAL DESIGN We conducted a phase I trial to test HAI of CAR-T in patients with CEA(+) liver metastases. Six patients completed the protocol, and 3 received anti-CEA CAR-T HAIs alone in dose-escalation fashion (10(8), 10(9), and 10(10) cells). We treated an additional 3 patients with the maximum planned CAR-T HAI dose (10(10) cells × 3) along with systemic IL2 support. RESULTS Four patients had more than 10 liver metastases, and patients received a mean of 2.5 lines of conventional systemic therapy before enrollment. No patient suffered a grade 3 or 4 adverse event related to the CAR-T HAIs. One patient remains alive with stable disease at 23 months following CAR-T HAI, and 5 patients died of progressive disease. Among the patients in the cohort that received systemic IL2 support, CEA levels decreased 37% (range, 19%-48%) from baseline. Biopsies demonstrated an increase in liver metastasis necrosis or fibrosis in 4 of 6 patients. Elevated serum IFNγ levels correlated with IL2 administration and CEA decreases. CONCLUSIONS We have demonstrated the safety of anti-CEA CAR-T HAIs with encouraging signals of clinical activity in a heavily pretreated population with large tumor burdens. Further clinical testing of CAR-T HAIs for liver metastases is warranted.
Collapse
Affiliation(s)
- Steven C Katz
- Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Rachel A Burga
- Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Elise McCormack
- Department of Medicine, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Li Juan Wang
- Department of Pathology, Roger Williams Medical Center, Providence, Rhode Island
| | - Wesley Mooring
- Department of Pathology, Roger Williams Medical Center, Providence, Rhode Island
| | - Gary R Point
- Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Pranay D Khare
- Roger Williams Medical Center, GMP Core Facility and Clinical Protocol Office, Providence, Rhode Island
| | - Mitchell Thorn
- Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Qiangzhong Ma
- Department of Medicine, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Brian F Stainken
- Department of Radiology, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Earle O Assanah
- Department of Radiology, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Robin Davies
- Roger Williams Medical Center, GMP Core Facility and Clinical Protocol Office, Providence, Rhode Island
| | - N Joseph Espat
- Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts
| | - Richard P Junghans
- Department of Medicine, Roger Williams Medical Center, Providence, Rhode Island/Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
40
|
Burga RA, Thorn M, Point GR, Guha P, Nguyen CT, Licata LA, DeMatteo RP, Ayala A, Joseph Espat N, Junghans RP, Katz SC. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol Immunother 2015; 64:817-29. [PMID: 25850344 DOI: 10.1007/s00262-015-1692-6] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 03/26/2015] [Indexed: 01/08/2023]
Abstract
Chimeric antigen receptor-modified T cell (CAR-T) technology, a promising immunotherapeutic tool, has not been applied specifically to treat liver metastases (LM). While CAR-T delivery to LM can be optimized by regional intrahepatic infusion, we propose that liver CD11b+Gr-1+ myeloid-derived suppressor cells (L-MDSC) will inhibit the efficacy of CAR-T in the intrahepatic space. We studied anti-CEA CAR-T in a murine model of CEA+ LM and identified mechanisms through which L-MDSC expand and inhibit CAR-T function. We established CEA+ LM in mice and studied purified L-MDSC and responses to treatment with intrahepatic anti-CEA CAR-T infusions. L-MDSC expanded threefold in response to LM, and their expansion was dependent on GM-CSF, which was produced by tumor cells. L-MDSC utilized PD-L1 to suppress anti-tumor responses through engagement of PD-1 on CAR-T. GM-CSF, in cooperation with STAT3, promoted L-MDSC PD-L1 expression. CAR-T efficacy was rescued when mice received CAR-T in combination with MDSC depletion, GM-CSF neutralization to prevent MDSC expansion, or PD-L1 blockade. As L-MDSC suppressed anti-CEA CAR-T, infusion of anti-CEA CAR-T in tandem with agents targeting L-MDSC is a rational strategy for future clinical trials.
Collapse
Affiliation(s)
- Rachel A Burga
- Division of Surgical Oncology, Department of Surgery, Roger Williams Medical Center, 825 Chalkstone Avenue, Prior 4, Providence, RI, 02908, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lanitis E, Poussin M, Klattenhoff AW, Song D, Sandaltzopoulos R, June CH, Powell DJ. Chimeric antigen receptor T Cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivo. Cancer Immunol Res 2015; 1:43-53. [PMID: 24409448 DOI: 10.1158/2326-6066.cir-13-0008] [Citation(s) in RCA: 277] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Adoptive immunotherapy using T lymphocytes genetically modified to express a chimeric antigen receptor (CAR-T) holds considerable promise for the treatment of cancer. However, CAR-based therapies may involve on-target toxicity against normal tissues expressing low amounts of the targeted tumor-associated antigen (TAA). To specify T cells for robust effector function that is selective for tumor but not normal tissue, we developed a trans-signaling CAR strategy, whereby T-cell activation signal 1 (CD3z) is physically dissociated from costimulatory signal 2 (CD28) in two CARs of differing antigen specificity: mesothelin and a-folate receptor (FRa). Human T cells were genetically modified to coexpress signal 1 (anti-Meso scFv-CD3z) and signal 2 (anti-FRa scFv-CD28) CARs in trans. Trans-signaling CAR-T cells showed weak cytokine secretion against target cells expressing only one TAA in vitro, similar to first-generation CAR-T cells bearing CD3z only, but showed enhanced cytokine secretion upon encountering natural or engineered tumor cells coexpressing both antigens, equivalent to that of second-generation CAR-T cells with dual signaling in cis. CAR-T cells with dual specificity also showed potent anticancer activity and persistence in vivo, which was superior to first-generation CAR-T cells and equivalent to second-generation CARs. Importantly, second-generation CAR-T cells exhibited potent activity against cells expressing mesothelin alone, recapitulating normal tissue, whereas trans-signaling CAR-T cells did not. Thus, a dual specificity, trans-signaling CAR approach can potentiate the therapeutic efficacy of CAR-T cells against cancer while minimizing parallel reactivity against normal tissues bearing single antigen.
Collapse
|
42
|
Interleukin-13 receptor alpha 2-targeted glioblastoma immunotherapy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:952128. [PMID: 25247196 PMCID: PMC4163479 DOI: 10.1155/2014/952128] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 08/05/2014] [Indexed: 01/23/2023]
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor, and despite several refinements in its multimodal management, generally has very poor prognosis. Targeted immunotherapy is an emerging field of research that shows great promise in the treatment of GBM. One of the most extensively studied targets is the interleukin-13 receptor alpha chain variant 2 (IL13Rα2). Its selective expression on GBM, discovered almost two decades ago, has been a target for therapy ever since. Immunotherapeutic strategies have been developed targeting IL13Rα2, including monoclonal antibodies as well as cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-expressing T cells. Advanced therapeutic development has led to the completion of several clinical trials with promising outcomes. In this review, we will discuss the recent advances in the IL13Rα2-targeted immunotherapy and evaluate the most promising strategy for targeted GBM immunotherapy.
Collapse
|
43
|
Thorn M, Point GR, Burga RA, Nguyen CT, Joseph Espat N, Katz SC. Liver metastases induce reversible hepatic B cell dysfunction mediated by Gr-1+CD11b+ myeloid cells. J Leukoc Biol 2014; 96:883-94. [PMID: 25085111 DOI: 10.1189/jlb.3a0114-012rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
LM escape immune surveillance, in part, as a result of the expansion of CD11b+MC, which alter the intrahepatic microenvironment to promote tumor tolerance. HBC make up a significant proportion of liver lymphocytes and appear to delay tumor progression; however, their significance in the setting of LM is poorly defined. Therefore, we characterized HBC and HBC/CD11b+MC interactions using a murine model of LM. Tumor-bearing livers showed a trend toward elevated absolute numbers of CD19+ HBC. A significant increase in the frequency of IgM(lo)IgD(hi) mature HBC was observed in mice with LM compared with normal mice. HBC derived from tumor-bearing mice demonstrated increased proliferation in response to TLR and BCR stimulation ex vivo compared with HBC from normal livers. HBC from tumor-bearing livers exhibited significant down-regulation of CD80 and were impaired in inducing CD4(+) T cell proliferation ex vivo. We implicated hepatic CD11b+MC as mediators of CD80 down-modulation on HBC ex vivo via a CD11b-dependent mechanism that required cell-to-cell contact and STAT3 activity. Therefore, CD11b+MC may compromise the ability of HBC to promote T cell activation in the setting of LM as a result of diminished expression of CD80. Cross-talk between CD11b+MC and HBC may be an important component of LM-induced immunosuppression.
Collapse
Affiliation(s)
- Mitchell Thorn
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and Boston University School of Medicine, Boston, Massachusetts, USA
| | - Gary R Point
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and
| | - Rachel A Burga
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and
| | - Cang T Nguyen
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and
| | - N Joseph Espat
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and Boston University School of Medicine, Boston, Massachusetts, USA
| | - Steven C Katz
- Roger Williams Medical Center, Department of Surgery, Providence, Rhode Island, USA; and Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
44
|
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
|
45
|
Im EJ, Bais AJ, Yang W, Ma Q, Guo X, Sepe SM, Junghans RP. Recombination-deletion between homologous cassettes in retrovirus is suppressed via a strategy of degenerate codon substitution. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14022. [PMID: 25419532 PMCID: PMC4239131 DOI: 10.1038/mtm.2014.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transduction and expression procedures in gene therapy protocols may optimally transfer more than a single gene to correct a defect and/or transmit new functions to recipient cells or organisms. This may be accomplished by transduction with two (or more) vectors, or, more efficiently, in a single vector. Occasionally, it may be useful to coexpress homologous genes or chimeric proteins with regions of shared homology. Retroviridae include the dominant vector systems for gene transfer (e.g., gamma-retro and lentiviruses) and are capable of such multigene expression. However, these same viruses are known for efficient recombination–deletion when domains are duplicated within the viral genome. This problem can be averted by resorting to two-vector strategies (two-chain two-vector), but at a penalty to cost, convenience, and efficiency. Employing a chimeric antigen receptor system as an example, we confirm that coexpression of two genes with homologous domains in a single gamma-retroviral vector (two-chain single-vector) leads to recombination–deletion between repeated sequences, excising the equivalent of one of the chimeric antigen receptors. Here, we show that a degenerate codon substitution strategy in the two-chain single-vector format efficiently suppressed intravector deletional loss with rescue of balanced gene coexpression by minimizing sequence homology between repeated domains and preserving the final protein sequence.
Collapse
Affiliation(s)
- Eung Jun Im
- Biotherapeutics Development Lab, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA ; Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Anthony J Bais
- Biotherapeutics Development Lab, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA ; Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Wen Yang
- Biotherapeutics Development Lab, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA ; Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Qiangzhong Ma
- Biotherapeutics Development Lab, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA ; Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Xiuyang Guo
- Biotherapeutics Development Lab, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA ; Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Steven M Sepe
- Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Richard P Junghans
- Biotherapeutics Development Lab, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA ; Department of Medicine, Boston University School of Medicine, Roger Williams Medical Center, Providence, Rhode Island, USA
| |
Collapse
|
46
|
Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro Oncol 2014; 16:1304-12. [PMID: 24723564 DOI: 10.1093/neuonc/nou045] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) remains one of the most lethal primary brain tumors despite surgical and therapeutic advancements. Targeted therapies of neoplastic diseases, including GBM, have received a great deal of interest in recent years. A highly studied target of GBM is interleukin-13 receptor α chain variant 2 (IL13Rα2). Targeted therapies against IL13Rα2 in GBM include fusion chimera proteins of IL-13 and bacterial toxins, nanoparticles, and oncolytic viruses. In addition, immunotherapies have been developed using monoclonal antibodies and cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-modified T cells. Advanced therapeutic development has led to the completion of phase I clinical trials for chimeric antigen receptor-modified T cells and phase III clinical trials for IL-13-conjugated bacterial toxin, with promising outcomes. Selective expression of IL13Rα2 on tumor cells, while absent in the surrounding normal brain tissue, has motivated continued study of IL13Rα2 as an important candidate for targeted glioma therapy. Here, we review the preclinical and clinical studies targeting IL13Rα2 in GBM and discuss new advances and promising applications.
Collapse
Affiliation(s)
- Bart Thaci
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Christine E Brown
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Emanuela Binello
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Katherine Werbaneth
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Prakash Sampath
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Sadhak Sengupta
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| |
Collapse
|
47
|
Cheadle EJ, Sheard V, Rothwell DG, Bridgeman JS, Ashton G, Hanson V, Mansoor AW, Hawkins RE, Gilham DE. Differential role of Th1 and Th2 cytokines in autotoxicity driven by CD19-specific second-generation chimeric antigen receptor T cells in a mouse model. THE JOURNAL OF IMMUNOLOGY 2014; 192:3654-65. [PMID: 24623129 DOI: 10.4049/jimmunol.1302148] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
T cells engrafted with chimeric AgRs (CAR) are showing exciting potential for targeting B cell malignancies in early-phase clinical trials. To determine whether the second-generation CAR was essential for optimal antitumor activity, two CD28-based CAR constructs targeting CD19 were tested for their ability to redirect mouse T cell function against established B cell lymphoma in a BALB/c syngeneic model system. T cells armed with either CAR eliminated A20 B cell lymphoma in vivo; however, one construct induced a T cell dose-dependent acute toxicity associated with a raised serum Th1 type cytokine profile on transfer into preconditioned mice. Moreover, a chronic toxicity manifested as granuloma-like formation in spleen, liver, and lymph nodes was observed in animals receiving T cells bearing either CD28 CAR, albeit with different kinetics dependent upon the specific receptor used. This phenotype was associated with an expansion of CD4+ CAR+ T cells and CD11b+ Gr-1(+) myeloid cells and increased serum Th2-type cytokines, including IL-10 and IL-13. Mouse T cells engrafted with a first-generation CAR failed to develop such autotoxicity, whereas toxicity was not apparent when T cells bearing the same receptors were transferred into C57BL/6 or C3H animals. In summary, the adoptive transfer of second-generation CD19-specific CAR T cells can result in a cell dose-dependent acute toxicity, whereas the prolonged secretion of high levels of Th2 cytokines from these CAR T cells in vivo drives a granulomatous reaction resulting in chronic toxicity. Strategies that prevent a prolonged Th2-cytokine biased CAR T cell response are clearly warranted.
Collapse
Affiliation(s)
- Eleanor J Cheadle
- Clinical and Experimental Immunotherapy Group, Department of Medical Oncology, Institute of Cancer Sciences, The University of Manchester, Manchester, M20 4BX, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ma Q, Gomes EM, Lo ASY, Junghans RP. Advanced generation anti-prostate specific membrane antigen designer T cells for prostate cancer immunotherapy. Prostate 2014; 74:286-96. [PMID: 24174378 DOI: 10.1002/pros.22749] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 10/03/2013] [Indexed: 11/11/2022]
Abstract
BACKGROUND Adoptive immunotherapy by infusion of designer T cells (dTc) engineered with chimeric antigen receptors (CARs) for tumoricidal activity represents a potentially highly specific modality for the treatment of cancer. In this study, 2nd generation (gen) anti-prostate specific membrane antigen (PSMA) dTc were developed for improving the efficacy of previously developed 1st gen dTc for prostate cancer immunotherapy. The 1st gen dTc are modified with chimeric immunoglobulin-T cell receptor (IgTCR) while the 2nd gen dTc are engineered with an immunoglobulin-CD28-T cell receptor (IgCD28TCR), which incorporates a CD28 costimulatory signal for optimal T cell activation. METHODS A 2nd gen anti-PSMA IgCD28TCR CAR was constructed by inserting the CD28 signal domain into the 1st gen CAR. 1st and 2nd gen anti-PSMA dTc were created by transducing human T cells with anti-PSMA CARs and their antitumor efficacy was compared for specific activation on PSMA-expressing tumor contact, cytotoxicity against PSMA-expressing tumor cells in vitro, and suppression of tumor growth in an animal model. RESULTS The 2nd gen dTc can be optimally activated to secrete larger amounts of cytokines such as IL2 and IFNγ than 1st gen and to proliferate more vigorously on PSMA-expressing tumor contact. More importantly, the 2nd gen dTc preserve the PSMA-specific cytotoxicity in vitro and suppress tumor growth in animal models with significant higher potency. CONCLUSIONS Our results demonstrate that 2nd gen anti-PSMA designer T cells exhibit superior antitumor functions versus 1st gen, providing a rationale for advancing this improved agent toward clinical application in prostate cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- CD28 Antigens/immunology
- Cell Line, Tumor
- Cell Membrane/immunology
- Cytotoxicity, Immunologic
- Genetic Vectors/genetics
- Humans
- Immunotherapy, Adoptive/methods
- Interferon-gamma/metabolism
- Interleukin-2/metabolism
- Jurkat Cells
- Lymphocyte Activation
- Male
- Mice
- Mice, Nude
- Prostate/immunology
- Prostatic Neoplasms/therapy
- Receptors, Antigen/genetics
- Receptors, Antigen/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins
- Retroviridae/genetics
- T-Lymphocytes/immunology
- Transduction, Genetic
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Qiangzhong Ma
- Department of Medicine, Biotherapeutics Development Lab, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | | | | | | |
Collapse
|
49
|
Cell transfer therapy for cancer: past, present, and future. J Immunol Res 2014; 2014:525913. [PMID: 24741604 PMCID: PMC3987872 DOI: 10.1155/2014/525913] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 12/26/2013] [Indexed: 12/30/2022] Open
Abstract
Cell transfer therapy for cancer has made a rapid progress recently and the immunotherapy has been recognized as the fourth anticancer modality after operation, chemotherapy, and radiotherapy. Lymphocytes used for cell transfer therapy include dendritic cells, natural killer (NK) cells, and T lymphocytes such as tumor-infiltrating lymphocytes (TILs) and cytotoxic T lymphocytes (CTLs). In vitro activated or engineered immune cells can traffic to cancer tissues to elicit persistent antitumor immune response which is very important especially after immunosuppressive treatments such as chemotherapy. In this review, we overviewed recent advances in the exploration of dendritic cells, NK cells, and T cells for the treatment of human cancer cells.
Collapse
|
50
|
Bozhenko VK, Shramova EI, Shishkin AM, Ivanov AV, Khokhlova EV, Lebedin YS, Shkoporov AN. Characteristics of new monomolecular chimeric T-cell receptors to carcinoembryonic antigen. Bull Exp Biol Med 2013; 156:165-71. [PMID: 24319717 DOI: 10.1007/s10517-013-2302-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We described two original genetic constructs encoding chimeric monomolecular T-cell receptors, where the effector T-cell receptor fragment was linked with the antigen-recognizing part consisting of two variable fragments of two different antibodies to carcinoembryonic antigen. Following transfection, these receptors were expressed on the cell surface and bound carcinoembryonic antigen. Human peripheral blood lymphocytes transfected with the above constructs demonstrated high cytotoxic activity against HCT116 cells expressing carcinoembryonic antigen.
Collapse
Affiliation(s)
- V K Bozhenko
- Russian Scientific Center of Roentgenoradiology, Ministry of Health of the Russian Federation; N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation; XEMA Company, Moscow, Russia.
| | | | | | | | | | | | | |
Collapse
|