1
|
Gandhi M, Sharma B, Nair S, Vaidya ADB. Current Insights into CAR T-Cell-Based Therapies for Myelodysplastic Syndrome. Pharm Res 2024; 41:1757-1773. [PMID: 39187686 DOI: 10.1007/s11095-024-03761-8] [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: 03/30/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
Myelodysplastic syndromes (MDS) are due to defective hematopoiesis in bone marrow characterized by cytopenia and dysplasia of blood cells, with a varying degree of risk of acute myeloid leukemia (AML). Currently, the only potentially curative strategy is hematopoietic stem cell transplantation (HSCT). Many patients are ineligible for HSCT, due to late diagnosis, presence of co-morbidities, old age and complications likely due to graft-versus-host disease (GvHD). As a consequence, patients with MDS are often treated conservatively with blood transfusions, chemotherapy, immunotherapy etc. based on the grade and manifestations of MDS. The development of chimeric antigen receptor (CAR)-T cell therapy has revolutionized immunotherapy for hematological malignancies, as evidenced by a large body of literature. However, resistance and toxicity associated with it are also a challenge. Hence, there is an urgent need to develop new strategies for immunological and hematopoetic management of MDS. Herein, we discuss current limitations of CAR T-cell therapy and summarize novel approaches to mitigate this. Further, we discuss the in vivo activation of tumor-specific T cells, immune check inhibitors (ICI) and other approaches to normalize the bone marrow milieu for the management of MDS.
Collapse
Affiliation(s)
- Manav Gandhi
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Bhirisha Sharma
- University of Mumbai, Santa Cruz (East), Mumbai, 400055, India
| | - Sujit Nair
- Viridis Biopharma Pvt. Ltd, Mumbai, 400022, India.
- Phytoveda Pvt. Ltd, Mumbai, 400022, India.
| | - Ashok D B Vaidya
- Kasturba Health Society-Medical Research Centre, Vile Parle (West), Mumbai, 400056, India
| |
Collapse
|
2
|
Rojas-Quintero J, Díaz MP, Palmar J, Galan-Freyle NJ, Morillo V, Escalona D, González-Torres HJ, Torres W, Navarro-Quiroz E, Rivera-Porras D, Bermúdez V. Car T Cells in Solid Tumors: Overcoming Obstacles. Int J Mol Sci 2024; 25:4170. [PMID: 38673757 PMCID: PMC11050550 DOI: 10.3390/ijms25084170] [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: 11/03/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR T cell) therapy has emerged as a prominent adoptive cell therapy and a therapeutic approach of great interest in the fight against cancer. This approach has shown notorious efficacy in refractory hematological neoplasm, which has bolstered its exploration in the field of solid cancers. However, successfully managing solid tumors presents considerable intrinsic challenges, which include the necessity of guiding the modified cells toward the tumoral region, assuring their penetration and survival in adverse microenvironments, and addressing the complexity of identifying the specific antigens for each type of cancer. This review focuses on outlining the challenges faced by CAR T cell therapy when used in the treatment of solid tumors, as well as presenting optimizations and emergent approaches directed at improving its efficacy in this particular context. From precise localization to the modulation of the tumoral microenvironment and the adaptation of antigen recognition strategies, diverse pathways will be examined to overcome the current limitations and buttress the therapeutic potential of CAR T cells in the fight against solid tumors.
Collapse
Affiliation(s)
- Joselyn Rojas-Quintero
- Medicine, Pulmonary, Critical Care, and Sleep Medicine Department, Baylor College of Medicine, Houston, TX 77030, USA;
| | - María P. Díaz
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Jim Palmar
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Nataly J. Galan-Freyle
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
| | - Valery Morillo
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Daniel Escalona
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | | | - Wheeler Torres
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Elkin Navarro-Quiroz
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
- Facultad de Ciencias Básicas y Biomédicas, Barranquilla 080002, Colombia
| | - Diego Rivera-Porras
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540001, Colombia;
| | - Valmore Bermúdez
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080002, Colombia;
| |
Collapse
|
3
|
Liu Y, Peng C, Ahad F, Ali Zaidi SA, Muluh TA, Fu Q. Advanced Strategies of CAR-T Cell Therapy in Solid Tumors and Hematological Malignancies. Recent Pat Anticancer Drug Discov 2024; 19:557-572. [PMID: 38213150 DOI: 10.2174/0115748928277331231218115402] [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: 09/18/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 01/13/2024]
Abstract
Chimeric antigen receptor T-cells, known as CAR-T cells, represent a promising breakthrough in the realm of adoptive cell therapy. These T-cells are genetically engineered to carry chimeric antigen receptors that specifically target tumors. They have achieved notable success in the treatment of blood-related cancers, breathing new life into this field of medical research. However, numerous obstacles limit chimeric antigen receptors T-cell therapy's efficacy, such as it cannot survive in the body long. It is prone to fatigue and exhaustion, leading to difficult tumor elimination and repeated recurrence, affecting solid tumors and hematological malignancies. The challenges posed by solid tumors, especially in the context of the complex solid-tumor microenvironment, require specific strategies. This review outlines recent advancements in improving chimeric antigen receptors T-cell therapy by focusing on the chimeric antigen receptors protein, modifying T-cells, and optimizing the interaction between T-cells and other components within the tumor microenvironment. This article aims to provide an extensive summary of the latest discoveries regarding CAR-T cell therapy, encompassing its application across various types of human cancers. Moreover, it will delve into the obstacles that have emerged in recent times, offering insights into the challenges faced by this innovative approach. Finally, it highlights novel therapeutic options in treating hematological and solid malignancies with chimeric antigen receptors T-cell therapies.
Collapse
Affiliation(s)
- Yangjie Liu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan, PRC China
| | - Cao Peng
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| | - Faiza Ahad
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Syed Aqib Ali Zaidi
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Qiuxia Fu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| |
Collapse
|
4
|
Chen C, Wang Z, Qin Y. CRISPR/Cas9 system: recent applications in immuno-oncology and cancer immunotherapy. Exp Hematol Oncol 2023; 12:95. [PMID: 37964355 PMCID: PMC10647168 DOI: 10.1186/s40164-023-00457-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 11/16/2023] Open
Abstract
Clustered regulatory interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) is essentially an adaptive immunity weapon in prokaryotes against foreign DNA. This system inspires the development of genome-editing technology in eukaryotes. In biomedicine research, CRISPR has offered a powerful platform to establish tumor-bearing models and screen potential targets in the immuno-oncology field, broadening our insights into cancer genomics. In translational medicine, the versatile CRISPR/Cas9 system exhibits immense potential to break the current limitations of cancer immunotherapy, thereby expanding the feasibility of adoptive cell therapy (ACT) in treating solid tumors. Herein, we first explain the principles of CRISPR/Cas9 genome editing technology and introduce CRISPR as a tool in tumor modeling. We next focus on the CRISPR screening for target discovery that reveals tumorigenesis, immune evasion, and drug resistance mechanisms. Moreover, we discuss the recent breakthroughs of genetically modified ACT using CRISPR/Cas9. Finally, we present potential challenges and perspectives in basic research and clinical translation of CRISPR/Cas9. This review provides a comprehensive overview of CRISPR/Cas9 applications that advance our insights into tumor-immune interaction and lay the foundation to optimize cancer immunotherapy.
Collapse
Affiliation(s)
- Chen Chen
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zehua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
5
|
Fernandes Â, Azevedo CM, Silva MC, Faria G, Dantas CS, Vicente MM, Pinho SS. Glycans as shapers of tumour microenvironment: A sweet driver of T-cell-mediated anti-tumour immune response. Immunology 2023; 168:217-232. [PMID: 35574724 DOI: 10.1111/imm.13494] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 01/17/2023] Open
Abstract
Essentially all cells are covered with a dense coat of different glycan structures/sugar chains, giving rise to the so-called glycocalyx. Changes in cellular glycosylation are a hallmark of cancer, affecting most of the pathophysiological processes associated with malignant transformation, including tumour immune responses. Glycans are chief macromolecules that define T-cell development, differentiation, fate, activation and signalling. Thus, the diversity of glycans expressed at the surface of T cells constitutes a fundamental molecular interface with the microenvironment by regulating the bilateral interactions between T-cells and cancer cells, fine-tuning the anti-tumour immune response. In this review, we will introduce the power of glycans as orchestrators of T-cell-mediated immune response in physiological conditions and in cancer. We discuss how glycans modulate the glyco-metabolic landscape in the tumour microenvironment, and whether glycans can synergize with immunotherapy as a way of rewiring T-cell effector functions against cancer cells.
Collapse
Affiliation(s)
- Ângela Fernandes
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Catarina M Azevedo
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Mariana C Silva
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Guilherme Faria
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Carolina S Dantas
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Manuel M Vicente
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Salomé S Pinho
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,School of Medicine and Biological Sciences (ICBAS), University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
6
|
Zheng N, Fang J, Xue G, Wang Z, Li X, Zhou M, Jin G, Rahman MM, McFadden G, Lu Y. Induction of tumor cell autosis by myxoma virus-infected CAR-T and TCR-T cells to overcome primary and acquired resistance. Cancer Cell 2022; 40:973-985.e7. [PMID: 36027915 PMCID: PMC9489043 DOI: 10.1016/j.ccell.2022.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 05/28/2022] [Accepted: 08/01/2022] [Indexed: 11/02/2022]
Abstract
Cytotoxicity of tumor-specific T cells requires tumor cell-to-T cell contact-dependent induction of classic tumor cell apoptosis and pyroptosis. However, this may not trigger sufficient primary responses of solid tumors to adoptive cell therapy or prevent tumor antigen escape-mediated acquired resistance. Here we test myxoma virus (MYXV)-infected tumor-specific T (TMYXV) cells expressing chimeric antigen receptor (CAR) or T cell receptor (TCR), which systemically deliver MYXV into solid tumors to overcome primary resistance. In addition to T cell-induced apoptosis and pyroptosis, tumor eradication by CAR/TCR-TMYXV cells is also attributed to tumor cell autosis induction, a special type of cell death. Mechanistically, T cell-derived interferon γ (IFNγ)-protein kinase B (AKT) signaling synergizes with MYXV-induced M-T5-SKP-1-VPS34 signaling to trigger robust tumor cell autosis. CAR/TCR-TMYXV-elicited autosis functions as a type of potent bystander killing to restrain antigen escape. We uncover an unexpected synergy between T cells and MYXV to bolster solid tumor cell autosis that reinforces tumor clearance.
Collapse
Affiliation(s)
- Ningbo Zheng
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Jing Fang
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Gang Xue
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Ziyu Wang
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Xiaoyin Li
- Department of Mathematics and Statistics, St. Cloud State University, St Cloud, MN 56301, USA
| | - Mengshi Zhou
- Department of Mathematics and Statistics, St. Cloud State University, St Cloud, MN 56301, USA
| | - Guangxu Jin
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Masmudur M Rahman
- Biodesign Center for Immunotherapy Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85287, USA
| | - Grant McFadden
- Biodesign Center for Immunotherapy Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85287, USA.
| | - Yong Lu
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| |
Collapse
|
7
|
Kang CH, Kim Y, Lee DY, Choi SU, Lee HK, Park CH. c-Met-Specific Chimeric Antigen Receptor T Cells Demonstrate Anti-Tumor Effect in c-Met Positive Gastric Cancer. Cancers (Basel) 2021; 13:cancers13225738. [PMID: 34830894 PMCID: PMC8616279 DOI: 10.3390/cancers13225738] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary c-Met is known to be overexpressed in gastric cancers. Here, we developed anti-c-Met CAR T cell and measured its anti-tumor efficacy in vitro and in vivo. Our anti c-Met CAR T cells have shown selective killing of c-Met overexpressed gastric cancer cells. Based on our results, we suggest that anti-c-Met CAR T cell therapy could be effective for gastric cancer patients. Abstract Chimeric antigen receptor (CAR) technology has been highlighted in recent years as a new therapeutic approach for cancer treatment. Although the impressive efficacy of CAR-based T cell adoptive immunotherapy has been observed in hematologic cancers, limited effect has been reported on solid tumors. Approximately 20% of gastric cancer (GC) patients exhibit a high expression of c-Met. We have generated an anti c-Met CAR construct that is composed of a single-chain variable fragment (scFv) of c-Met antibody and signaling domains consisting of CD28 and CD3ζ. To test the CAR construct, we used two cell lines: the Jurkat and KHYG-1 cell lines. These are convenient cell lines, compared to primary T cells, to culture and to test CAR constructs. We transduced CAR constructs into Jurkat cells by electroporation. c-Met CAR Jurkat cells secreted interleukin-2 (IL-2) only when incubated with c-Met positive GC cells. To confirm the lytic function of CAR, the CAR construct was transduced into KHYG-1, a NK/T cell line, using lentiviral particles. c-Met CAR KHYG-1 showed cytotoxic effect on c-Met positive GC cells, while c-Met negative GC cell lines were not eradicated by c-Met CAR KHYG-1. Based on these data, we created c-Met CAR T cells from primary T cells, which showed high IL-2 and IFN-γ secretion when incubated with the c-Met positive cancer cell line. In an in vivo xenograft assay with NSG bearing MKN-45, a c-Met positive GC cell line, c-Met CAR T cells effectively inhibited the tumor growth of MKN-45. Our results show that the c-Met CAR T cell therapy can be effective on GC.
Collapse
Affiliation(s)
- Chung Hyo Kang
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea; (C.H.K.); (Y.K.); (D.Y.L.); (S.U.C.); (H.K.L.)
| | - Yeongrin Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea; (C.H.K.); (Y.K.); (D.Y.L.); (S.U.C.); (H.K.L.)
- Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Da Yeon Lee
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea; (C.H.K.); (Y.K.); (D.Y.L.); (S.U.C.); (H.K.L.)
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea
| | - Sang Un Choi
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea; (C.H.K.); (Y.K.); (D.Y.L.); (S.U.C.); (H.K.L.)
| | - Heung Kyoung Lee
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea; (C.H.K.); (Y.K.); (D.Y.L.); (S.U.C.); (H.K.L.)
| | - Chi Hoon Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea; (C.H.K.); (Y.K.); (D.Y.L.); (S.U.C.); (H.K.L.)
- Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon 34113, Korea
- Correspondence: ; Tel.: +82-42-860-7416; Fax: +82-42-861-4246
| |
Collapse
|
8
|
Noh KE, Lee JH, Choi SY, Jung NC, Nam JH, Oh JS, Song JY, Seo HG, Wang Y, Lee HS, Lim DS. TGF-β/IL-7 Chimeric Switch Receptor-Expressing CAR-T Cells Inhibit Recurrence of CD19-Positive B Cell Lymphoma. Int J Mol Sci 2021; 22:ijms22168706. [PMID: 34445415 PMCID: PMC8395772 DOI: 10.3390/ijms22168706] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/02/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cells are effective in the treatment of hematologic malignancies but have shown limited efficacy against solid tumors. Here, we demonstrated an approach to inhibit recurrence of B cell lymphoma by co-expressing both a human anti-CD19-specific single-chain variable fragment (scFv) CAR (CD19 CAR) and a TGF-β/IL-7 chimeric switch receptor (tTRII-I7R) in T cells (CD19 CAR-tTRII-I7R-T cells). The tTRII-I7R was designed to convert immunosuppressive TGF-β signaling into immune-activating IL-7 signaling. The effect of TGF-β on CD19 CAR-tTRII-I7R-T cells was assessed by western blotting. Target-specific killing by CD19 CAR-tTRII-I7R-T cells was evaluated by Eu-TDA assay. Daudi tumor-bearing NSG (NOD/SCID/IL2Rγ-/-) mice were treated with CD19 CAR-tTRII-I7R-T cells to analyze the in vivo anti-tumor effect. In vitro, CD19 CAR-tTRII-I7R-T cells had a lower level of phosphorylated SMAD2 and a higher level of target-specific cytotoxicity than controls in the presence of rhTGF-β1. In the animal model, the overall survival and recurrence-free survival of mice that received CD19 CAR-tTRII-I7R-T cells were significantly longer than in control mice. These findings strongly suggest that CD19 CAR-tTRII-I7R-T cell therapy provides a new strategy for long-lasting, TGF-β-resistant anti-tumor effects against B cell lymphoma, which may lead ultimately to increased clinical efficacy.
Collapse
MESH Headings
- Animals
- Antigens, CD19/immunology
- Cells, Cultured
- Female
- Humans
- Immunotherapy, Adoptive
- Interleukin-7/genetics
- Interleukin-7/metabolism
- K562 Cells
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/therapy
- Receptors, Chimeric Antigen/metabolism
- Signal Transduction
- Single-Chain Antibodies/metabolism
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kyung-Eun Noh
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea; (K.-E.N.); (S.-Y.C.); (J.-H.N.); (J.-S.O.)
| | - Jun-Ho Lee
- Pharos Vaccine Inc., 14 Galmachiro, 288 Bun-gil, Jungwon-gu, Seongnam 13201, Gyeonggi-do, Korea; (J.-H.L.); (N.-C.J.); (H.S.L.)
| | - So-Yeon Choi
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea; (K.-E.N.); (S.-Y.C.); (J.-H.N.); (J.-S.O.)
| | - Nam-Chul Jung
- Pharos Vaccine Inc., 14 Galmachiro, 288 Bun-gil, Jungwon-gu, Seongnam 13201, Gyeonggi-do, Korea; (J.-H.L.); (N.-C.J.); (H.S.L.)
| | - Ji-Hee Nam
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea; (K.-E.N.); (S.-Y.C.); (J.-H.N.); (J.-S.O.)
| | - Ji-Soo Oh
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea; (K.-E.N.); (S.-Y.C.); (J.-H.N.); (J.-S.O.)
| | - Jie-Young Song
- Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea;
| | - Han Geuk Seo
- Department of Food Science and Biotechnology of Animal Products, Sanghuh College of Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea;
| | - Yu Wang
- Immunotech Applied Science Ltd., Beijing 100176, China;
| | - Hyun Soo Lee
- Pharos Vaccine Inc., 14 Galmachiro, 288 Bun-gil, Jungwon-gu, Seongnam 13201, Gyeonggi-do, Korea; (J.-H.L.); (N.-C.J.); (H.S.L.)
| | - Dae-Seog Lim
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea; (K.-E.N.); (S.-Y.C.); (J.-H.N.); (J.-S.O.)
- Correspondence: ; Tel.: +82-10-2770-4777
| |
Collapse
|
9
|
Meggyes M, Nagy DU, Balassa T, Godony K, Peterfalvi A, Szereday L, Polgar B. Influence of Galectin-9 Treatment on the Phenotype and Function of NK-92MI Cells in the Presence of Different Serum Supplements. Biomolecules 2021; 11:biom11081066. [PMID: 34439744 PMCID: PMC8391477 DOI: 10.3390/biom11081066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/16/2022] Open
Abstract
Galectins are one of the critical players in the tumor microenvironment-tumor crosstalk and the regulation of local immunity. Galectin-9 has been in the limelight in tumor immunology. Galectin-9 possesses its multiplex biological functions both extracellularly and intracellularly, plays a pivotal role in the modulation of adaptive and innate immunity, and induces immune tolerance. NK-92MI cell lines against different malignancies were extensively studied, and recently published trials used genetically chimeric antigen receptor-transfected NK-92MI cells in tumor immunotherapy. Besides the intensive research in tumor immunotherapy, limited information is available on their immune-checkpoint expression and the impact of checkpoint ligands on their effector functions. To uncover the therapeutic potential of modulating Galectin-9-related immunological pathways in NK-cell-based therapy, we investigated the dose-dependent effect of soluble Galectin-9 on the TIM-3 checkpoint receptor and NKG2D, CD69, FasL, and perforin expression of NK-92MI cells. We also examined how their cytotoxicity and cytokine production was altered after Gal-9 treatment and in the presence of different serum supplements using flow cytometric analysis. Our study provides evidence that the Galectin-9/TIM-3 pathway plays an important role in the regulation of NK cell function, and about the modulatory role of Galectin-9 on the cytotoxicity and cytokine production of NK-92MI cells in the presence of different serum supplements. We hope that our results will aid the development of novel NK-cell-based strategies that target Galectin-9/TIM-3 checkpoint in tumors resistant to T-cell-based immunotherapy.
Collapse
Affiliation(s)
- Matyas Meggyes
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary; (T.B.); (L.S.); (B.P.)
- Janos Szentagothai Research Centre, University of Pecs, 20 Ifjusag Street, 7624 Pecs, Hungary
- Correspondence: ; Tel.: +3672-536001/1907
| | - David U Nagy
- Medical Centre, Cochrane Hungary, University of Pecs, 7623 Pecs, Hungary;
| | - Timea Balassa
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary; (T.B.); (L.S.); (B.P.)
| | - Krisztina Godony
- Department of Obstetrics and Gynaecology, Medical School, University of Pecs, 17 Edesanyak Street, 7624 Pecs, Hungary;
| | - Agnes Peterfalvi
- Department of Laboratory Medicine, Medical School, University of Pecs, 13 Ifjusag Street, 7624 Pecs, Hungary;
| | - Laszlo Szereday
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary; (T.B.); (L.S.); (B.P.)
- Janos Szentagothai Research Centre, University of Pecs, 20 Ifjusag Street, 7624 Pecs, Hungary
| | - Beata Polgar
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary; (T.B.); (L.S.); (B.P.)
- Janos Szentagothai Research Centre, University of Pecs, 20 Ifjusag Street, 7624 Pecs, Hungary
| |
Collapse
|
10
|
Park CH. Making Potent CAR T Cells Using Genetic Engineering and Synergistic Agents. Cancers (Basel) 2021; 13:cancers13133236. [PMID: 34209505 PMCID: PMC8269169 DOI: 10.3390/cancers13133236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
Immunotherapies are emerging as powerful weapons for the treatment of malignancies. Chimeric antigen receptor (CAR)-engineered T cells have shown dramatic clinical results in patients with hematological malignancies. However, it is still challenging for CAR T cell therapy to be successful in several types of blood cancer and most solid tumors. Many attempts have been made to enhance the efficacy of CAR T cell therapy by modifying the CAR construct using combination agents, such as compounds, antibodies, or radiation. At present, technology to improve CAR T cell therapy is rapidly developing. In this review, we particularly emphasize the most recent studies utilizing genetic engineering and synergistic agents to improve CAR T cell therapy.
Collapse
Affiliation(s)
- Chi Hoon Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Daejeon 34114, Korea; ; Tel.: +82-42-860-7416; Fax: +82-42-861-4246
- Medicinal & Pharmaceutical Chemistry, Korea University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
11
|
Gotti M, Defrancesco I, D'Angelo M, Basso S, Crotto L, Marinelli A, Maccalli C, Iaconianni V. Cancer Immunotherapy Using Chimeric Antigen Receptor Expressing T-Cells: Present and Future Needs of Clinical Cancer Centers. Front Immunol 2020; 11:565236. [PMID: 33193333 PMCID: PMC7662555 DOI: 10.3389/fimmu.2020.565236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 10/06/2020] [Indexed: 12/30/2022] Open
Abstract
Chimeric Antigen Receptor-T cells (CAR-T) are considered novel biological agents, designed to selectively attack cancer cells expressing specific antigens, with demonstrated clinical activity in patients affected with relapsed/refractory B-cell malignancies. In consideration of their complexity, the use of CAR-T requires dedicated clinical setting and health care practitioners with expertise in the selection, treatment, and management of toxicities and side effects. Such issue appears particularly important when contextualized in the rapid progress of CAR-T cell treatment, translating into a constant need of updating and evolution. Moreover, the clinical grade manufacturing of CAR-T cells is complex and implies articulated regulatory and organizational aspects. The main goal of this review is to summarize and provide an accurate analysis of the clinical, logistic, and regulatory requirements of CAR-T cell centers. Finally, we describe a new occupational figure called “CAR-T specialist” devoted to the establishment and coordination of the required facilities and regulatory landscape in the context of cancer centers.
Collapse
Affiliation(s)
- Manuel Gotti
- Division of Hematology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | | | - Mario D'Angelo
- Department of Onco-hematology, Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù IRCCS, Roma, Italy
| | - Sabrina Basso
- Pediatric Hematology/Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Crotto
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Alfredo Marinelli
- Operative Unit (OU) Neuroncology, University Federico II, Napoli, Italy.,IRCCS Neuromed Istituto Neurologico Mediterraneo Pozzilli (INM), Pozzilli, Italy
| | - Cristina Maccalli
- Laboratory of Immune and Biological Therapy, Research Department, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
12
|
Date V, Nair S. Emerging vistas in CAR T-cell therapy: challenges and opportunities in solid tumors. Expert Opin Biol Ther 2020; 21:145-160. [PMID: 32882159 DOI: 10.1080/14712598.2020.1819978] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Despite advances in modern evidence-based medicine, cancer remains a major cause of global disease-associated mortality. CAR T-cell therapy is a major histocompatibility complex (MHC)-independent immunotherapy involving adoptive cell transfer. Cancer immunotherapy witnessed a major breakthrough with the US FDA approval of the first chimeric antigen receptor (CAR) T-cell therapy KymriahTM (tisagenlecleucel) for relapsed or refractory (R/R) acute lymphoblastic leukemia (ALL) in August 2017 followed by approval of Yescarta® (axicabtagene ciloleucel) for R/R non-Hodgkin's lymphoma (NHL) in October 2017. AREAS COVERED We review the potential of CAR T-cell therapy which, despite showing great promise in hematological malignancies, faces significant challenges in targeting solid tumors. We address these challenges and discuss proposed strategies to overcome them in solid tumors. We highlight the potential of CAR T-cell therapy as cancer precision medicine and briefly discuss the 'financial toxicity' of CAR T-cell therapy. EXPERT OPINION Taken together, we discuss various strategies to circumvent the limitations of CAR T-cell therapy in solid tumors. Despite the rapid advances in CAR NK-cell therapies, there is immense scope for CAR T-cell therapy in solid tumors. We provide a synthetic review of CAR T-cell therapy that will drive future research and harness its full potential in cancer precision medicine for solid tumors.
Collapse
Affiliation(s)
- Varada Date
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS University , Mumbai, India
| | - Sujit Nair
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai , Mumbai, India
| |
Collapse
|
13
|
Tian Y, Li Y, Shao Y, Zhang Y. Gene modification strategies for next-generation CAR T cells against solid cancers. J Hematol Oncol 2020; 13:54. [PMID: 32423475 PMCID: PMC7236186 DOI: 10.1186/s13045-020-00890-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapies have become the backbone of cancer treatment. Among them, chimeric antigen receptor (CAR) T cells have demonstrated great success in the treatment of hematological malignancies. However, CAR T therapy against solid tumors is less effective. Antigen targeting; an immunosuppressive tumor microenvironment (TME); and the infiltration, proliferation, and persistence of CAR T cells are the predominant barriers preventing the extension of CAR T therapy to solid tumors. To circumvent these obstacles, the next-generation CAR T cells will require more potent antitumor properties, which can be achieved by gene-editing technology. In this review, we summarize innovative strategies to enhance CAR T cell function by improving target identification, persistence, trafficking, and overcoming the suppressive TME. The construction of multi-target CAR T cells improves antigen recognition and reduces immune escape. Enhancing CAR T cell proliferation and persistence can be achieved by optimizing costimulatory signals and overexpressing cytokines. CAR T cells equipped with chemokines or chemokine receptors help overcome their poor homing to tumor sites. Strategies like knocking out immune checkpoint molecules, incorporating dominant negative receptors, and chimeric switch receptors can favor the depletion or reversal of negative T cell regulators in the TME.
Collapse
Affiliation(s)
- Yonggui Tian
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, 450052, China
| | - Yilu Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,School of Medicine, Zhengzhou University, Zhengzhou, 450052, China
| | - Yupei Shao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,School of Medicine, Zhengzhou University, Zhengzhou, 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, 450052, China.
| |
Collapse
|
14
|
Pharmacological targeting of immune checkpoint A2aR improves function of anti-CD19 CAR T cells in vitro. Immunol Lett 2020; 223:44-52. [PMID: 32289340 DOI: 10.1016/j.imlet.2020.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 12/18/2022]
Abstract
In spite of impressive results in the treatment of acute lymphoblastic B cell leukemia (B-ALL) with chimeric antigen receptor (CAR) T cells, the clinical outcome of some hematological cancers like follicular lymphoma (FL) and chronic lymphocytic leukemia (CLL) has not been very promising likely due to immunosuppressive networks within tumor microenvironment. Hypoxia in the microenvironment of hematological malignancies and consequently generation of adenosine molecule is appeared to be correlated with immunosuppression, tumor progression, and relapse. Herein, we hypothesized that whether pharmacological targeting of adenosine 2a receptor (A2aR) can enhance antitumor activity of anti-CD19 CAR T cells in vitro. Prior to functional assays, A2aR expression was assessed in CAR-expressing T cells. Our results showed that A2aR was not only up-regulated in the fully human anti-CD19 CAR T cells (hereafter referred to as huCAR19 T cells) but also was further overexpressed following re-stimulation with target cells. Although pharmacological inhibition of A2aR could significantly increase proliferation capacity and cytokine production of huCAR19 T cells following treatment with an adenosine analog, cytotoxic activity of huCAR19 T cells was not significantly improved. Considering A2aR overexpression in huCAR19 T cells in the tumor microenvironment, our results indicated that pharmacological targeting of A2aR could not only improve huCAR19 T cells functionality in a hostile tumor microenvironment but also could have a therapeutic advantage, and sought to assess the possibility in a pre-clinical setting.
Collapse
|
15
|
CAR-T Cell Therapy in Cancer: Tribulations and Road Ahead. J Immunol Res 2020; 2020:1924379. [PMID: 32411789 PMCID: PMC7201836 DOI: 10.1155/2020/1924379] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/11/2019] [Accepted: 12/30/2019] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor- (CAR-) T cell therapy is one of the most recent innovative immunotherapies and is rapidly evolving. Like other technologies, CAR-T cell therapy has undergone a long development process, and persistent explorations of the actions of the intracellular signaling domain and make several improvements have led to the superior efficacy when anti-CD19 CAR-T cell treatments in B cell cancers. At present, CAR-T cell therapy is developing rapidly, and many clinical trials have been established on a global scale, which has great commercial potential. This review mainly describes the toxicity of CAR-T cell therapy and the challenges of CAR-T cells in the treatment of solid tumors, and looks forward to future development and opportunities for immunotherapy and reviews major breakthroughs in CAR-T cell therapy.
Collapse
|
16
|
Jain A, Shah H, Simonsick EM, Metter EJ, Mangold L, Humphreys E, Partin A, Fedarko NS. Angiotensin receptor autoantibodies as exposures that modify disease progression: Cross sectional, longitudinal and in vitro studies of prostate cancer. J Transl Autoimmun 2019; 2:100008. [PMID: 31930191 PMCID: PMC6953913 DOI: 10.1016/j.jtauto.2019.100008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022] Open
Abstract
Circulating angiotensin type I receptor (AT1R) agonistic autoantibodies (AT1RaAbs) that bind and chronically activate the receptor have been associated with a number of diseases suggesting that while the autoantibodies are not necessarily causative they may promote disease progression. The prostate has a local renin angiotensin system. The current study examines associations between AT1RaAbs and prostate cancer (PCA), disease-free survival (DFS), overall survival (OS) and AT1RaAb effects on PCA cell phenotype. In a cross-sectional set of serum obtained from 151 men diagnosed with PCA, nonmalignant prostate disease or no disease, higher serum AT1RaAb levels were associated with PCA and non-organ confined PCA. The odds ratio for PCA was 6.3 (95% confidence interval 2.2 to 18) for a positive 1:1600 titer and 18 (95% confidence interval 6.9 to 45) at AT1RaAb levels > 1.04 μg/ml, (p < 0.0001). In a longitudinal set of pre-diagnosis samples from 109 men, DFS hazard ratios of 2.2 (95% confidence interval 1.4 to 3.5) and 1.6 (95% confidence interval 1.0 to 2.5) for most proximal to diagnosis and most distal to diagnosis samples, respectively, were found for high versus low AT1RaAb groups. Hazard ratios for OS in most proximal and distal samples were 2.4 (95% confidence interval 1.6 to 3.6) and 1.8 (95% confidence interval 1.1 to 2.8), respectively. Accelerated failure modeling of survival indicated that a 1 μg/ml increase in AT1RaAb levels was associated with a reduction of DFS and OS by 20% at the most proximal time point and by 15% at the most distal time points. Adjusting for age, did not affect the association with DFS in proximal samples but changed distal time point DFS and OS to a 10% decrease for every 1 μg/ml increase in AT1RaAb. Additional adjustments for body mass index, systolic blood pressure and prostate-specific antigen did not appreciably alter these associations. AT1RaAb treatment of PC3, DU145, and LNCaP cells significantly increased the maximal growth rate approximately 2-fold and invasiveness approximately 3-fold. Conclusions: These observations provide evidence supporting AT1RaAbs as exposures that may modify prostate cancer progression and indicate they may be predictive markers for risk stratification.
Collapse
Affiliation(s)
- Alka Jain
- Department of Medicine, Johns Hopkins University, Baltimore, MD, 21224, USA
| | - Haikoo Shah
- Department of Medicine, Johns Hopkins University, Baltimore, MD, 21224, USA
- Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Eleanor M. Simonsick
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21225, USA
| | - E. Jeffrey Metter
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21225, USA
- Current Address: Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Leslie Mangold
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Elizabeth Humphreys
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Alan Partin
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Neal S. Fedarko
- Department of Medicine, Johns Hopkins University, Baltimore, MD, 21224, USA
| |
Collapse
|
17
|
Current status and hurdles for CAR-T cell immune therapy. BLOOD SCIENCE 2019; 1:148-155. [PMID: 35402809 PMCID: PMC8974909 DOI: 10.1097/bs9.0000000000000025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/25/2019] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cells have emerged as novel and promising immune therapies for the treatment of multiple types of cancer in patients with hematological malignancies. There are several key components critical for development and application of CAR-T therapy. First, the design of CAR vectors can considerably affect several aspects of the physiological functions of these T cells. Moreover, despite the wide use of γ-retrovirus and lentivirus in mediating gene transfer into T cells, optimal CAR delivery systems are also being developed and evaluated. In addition, several classes of mouse models have been used to evaluate the efficacies of CAR-T cells; however, each model has its own limitations. Clinically, although surprising complete remission (CR) rates were observed in acute lymphoblastic leukemia (ALL), lymphoma, and multiple myeloma (MM), there is still a lack of specific targets for acute myeloid leukemia (AML). Leukemia relapse remains a major challenge, and its mechanism is presently under investigation. Cytokine release syndrome (CRS) and neurotoxicity are life-threatening adverse effects that need to be carefully treated. Several factors that compromise the activities of anti-solid cancer CAR-T cells have been recognized, and further improvements targeting these factors are the focus of the development of novel CAR-T cells. Overcoming the current hurdles will lead to optimal responses of CAR-T cells, thus paving the way for their wide clinical application.
Collapse
|
18
|
da Silva JL, Dos Santos ALS, Nunes NCC, de Moraes Lino da Silva F, Ferreira CGM, de Melo AC. Cancer immunotherapy: the art of targeting the tumor immune microenvironment. Cancer Chemother Pharmacol 2019; 84:227-240. [PMID: 31240384 DOI: 10.1007/s00280-019-03894-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/14/2019] [Indexed: 01/07/2023]
Abstract
For many decades, cancer treatment has been strongly directed toward the development of cytotoxic and cytostatic drugs, quite often leading to disappointing results due to the inter- and intra-tumoral heterogeneity. Lately, this intra-cellular look has given way to the understanding of the tumor microenvironment, thus enabling modification of the immunological dynamics between tumor cells and their host. An era of new drugs aiming to unlock the host immune system against tumor cells is steadily increasing. Strategies involving adoptive cell therapy, therapeutic vaccines, immune checkpoint inhibitors and so on have provided spectacular clinical responses and increased survival in previously refractory settings and "hard-to-treat" cancers. Based on a comprehensive search in the main scientific databases, annals of recent renowned oncology congresses and platforms of ongoing trials, the clinical pharmacology characteristics of the main classes of immunotherapeutic agents, as well as the new treatment strategies related to immunotherapy in solid tumors, are carefully discussed throughout this review.
Collapse
Affiliation(s)
- Jesse Lopes da Silva
- Clinic Oncomed, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil.
- Oncomed Clinic Oncologica, Niterói, Rio de Janeiro, 24220-300, Brazil.
| | | | - Natalia Cristina Cardoso Nunes
- Clinic Oncomed, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Oncoclinicas Institute for Research and Education, Sao Paulo, Brazil
| | | | | | - Andreia Cristina de Melo
- Clinic Oncomed, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Oncoclinicas Institute for Research and Education, Sao Paulo, Brazil
| |
Collapse
|
19
|
Yu C, Liu X, Yang J, Zhang M, Jin H, Ma X, Shi H. Combination of Immunotherapy With Targeted Therapy: Theory and Practice in Metastatic Melanoma. Front Immunol 2019; 10:990. [PMID: 31134073 PMCID: PMC6513976 DOI: 10.3389/fimmu.2019.00990] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/16/2019] [Indexed: 02/05/2023] Open
Abstract
Metastatic melanoma is the most aggressive and obstinate skin cancer with poor prognosis. Variant novel applicable regimens have emerged during the past decades intensively, while the most profound approaches are oncogene-targeted therapy and T-lymphocyte mediated immunotherapy. Although targeted therapies generated remarkable and rapid clinical responses in the majority of patients, acquired resistance was developed promptly within months leading to tumor relapse. By contrast, immunotherapies elicited long-term tumor regression. However, the overall response rate was limited. In view of the above, either targeted therapy or immunotherapy cannot elicit durable clinical responses in large range of patients. Interestingly, the advantages and limitations of these regimens happened to be complementary. An increasing number of preclinical studies and clinical trials proved a synergistic antitumor effect with the combination of targeted therapy and immunotherapy, implying a promising prospect for the treatment of metastatic melanoma. In order to achieve a better therapeutic effectiveness and reduce toxicity in patients, great efforts need to be made to illuminate multifaceted interplay between targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Chune Yu
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowei Liu
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiqiao Yang
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Min Zhang
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongyu Jin
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hubing Shi
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Tanna JG, Ulrey R, Williams KM, Hanley PJ. Critical testing and parameters for consideration when manufacturing and evaluating tumor-associated antigen-specific T cells. Cytotherapy 2019; 21:278-288. [PMID: 30929992 DOI: 10.1016/j.jcyt.2019.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 02/07/2019] [Accepted: 02/07/2019] [Indexed: 12/12/2022]
Abstract
The past year has seen remarkable translation of cellular and gene therapies, with U.S. Food and Drug Administration (FDA) approval of three chimeric antigen receptor (CAR) T-cell products, multiple gene therapy products, and the initiation of countless other pivotal clinical trials. What makes these new drugs most remarkable is their path to commercialization: they have unique requirements compared with traditional pharmaceutical drugs and require different potency assays, critical quality attributes and parameters, pharmacological and toxicological data, and in vivo efficacy testing. What's more, each biologic requires its own unique set of tests and parameters. Here we describe the unique tests associated with ex vivo-expanded tumor-associated antigen T cells (TAA-T). These tests include functional assays to determine potency, specificity, and identity; tests for pathogenic contaminants, such as bacteria and fungus as well as other contaminants such as Mycoplasma and endotoxin; tests for product characterization, tests to evaluate T-cell persistence and product efficacy; and finally, recommendations for critical quality attributes and parameters associated with the expansion of TAA-Ts.
Collapse
Affiliation(s)
- Jay G Tanna
- Program for Cell Enhancement and Technologies for Immunotherapy, Center for Cancer and Immunology Research
| | - Robert Ulrey
- Program for Cell Enhancement and Technologies for Immunotherapy, Center for Cancer and Immunology Research
| | - Kirsten M Williams
- Program for Cell Enhancement and Technologies for Immunotherapy, Center for Cancer and Immunology Research; Center for Cancer and Blood Disorders, and the Division of Blood and Marrow Transplantation; Children's National Health System and The George Washington University, Washington, DC, USA
| | - Patrick J Hanley
- Program for Cell Enhancement and Technologies for Immunotherapy, Center for Cancer and Immunology Research; Center for Cancer and Blood Disorders, and the Division of Blood and Marrow Transplantation; Children's National Health System and The George Washington University, Washington, DC, USA.
| |
Collapse
|
21
|
Zhang Z, Jiang J, Wu X, Zhang M, Luo D, Zhang R, Li S, He Y, Bian H, Chen Z. Chimeric antigen receptor T cell targeting EGFRvIII for metastatic lung cancer therapy. Front Med 2019; 13:57-68. [DOI: 10.1007/s11684-019-0683-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/20/2018] [Indexed: 11/24/2022]
|
22
|
DeSelm C, Palomba ML, Yahalom J, Hamieh M, Eyquem J, Rajasekhar VK, Sadelain M. Low-Dose Radiation Conditioning Enables CAR T Cells to Mitigate Antigen Escape. Mol Ther 2018; 26:2542-2552. [PMID: 30415658 DOI: 10.1016/j.ymthe.2018.09.008] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 09/07/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
CD19 chimeric antigen receptors (CARs) have demonstrated great efficacy against a range of B cell malignancies. However, antigen escape and, more generally, heterogeneous antigen expression pose a challenge to applying CAR therapy to a wide range of cancers. We find that low-dose radiation sensitizes tumor cells to immune rejection by locally activated CAR T cells. In a model of pancreatic adenocarcinoma heterogeneously expressing sialyl Lewis-A (sLeA), we show that not only sLeA+ but also sLeA- tumor cells exposed to low-dose radiation become susceptible to CAR therapy, reducing antigen-negative tumor relapse. RNA sequencing analysis of low-dose radiation-exposed tumors reveals the transcriptional signature of cells highly sensitive to TRAIL-mediated death. We find that sLeA-targeted CAR T cells produce TRAIL upon engaging sLeA+ tumor cells, and eliminate sLeA- tumor cells previously exposed to systemic or local low-dose radiation in a TRAIL-dependent manner. These findings enhance the prospects for successfully applying CAR therapy to heterogeneous solid tumors. Local radiation is integral to many tumors' standard of care and can be easily implemented as a CAR conditioning regimen.
Collapse
Affiliation(s)
- Carl DeSelm
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Lia Palomba
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joachim Yahalom
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohamad Hamieh
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin Eyquem
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology Program, Sloan Kettering Institute, New York, NY, USA.
| |
Collapse
|
23
|
Idorn M, Thor Straten P. Chemokine Receptors and Exercise to Tackle the Inadequacy of T Cell Homing to the Tumor Site. Cells 2018; 7:E108. [PMID: 30126117 PMCID: PMC6115859 DOI: 10.3390/cells7080108] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 01/05/2023] Open
Abstract
While cancer immune therapy has revolutionized the treatment of metastatic disease across a wide range of cancer diagnoses, a major limiting factor remains with regard to relying on adequate homing of anti-tumor effector cells to the tumor site both prior to and after therapy. Adoptive cell transfer (ACT) of autologous T cells have improved the outlook of patients with metastatic melanoma. Prior to the approval of checkpoint inhibitors, this strategy was the most promising. However, while response rates of up to 50% have been reported, this strategy is still rather crude. Thus, improvements are needed and within reach. A hallmark of the developing tumor is the evasion of immune destruction. Achieved through the recruitment of immune suppressive cell subsets, upregulation of inhibitory receptors and the development of physical and chemical barriers (such as poor vascularization and hypoxia) leaves the microenvironment a hostile destination for anti-tumor T cells. In this paper, we review the emerging strategies of improving the homing of effector T cells (TILs, CARs, TCR engineered T cells, etc.) through genetic engineering with chemokine receptors matching the chemokines of the tumor microenvironment. While this strategy has proven successful in several preclinical models of cancer and the strategy has moved into the first phase I/II clinical trial in humans, most of these studies show a modest (doubling) increase in tumor infiltration of effector cells, which raises the question of whether road blocks must be tackled for efficient homing. We propose a role for physical exercise in modulating the tumor microenvironment and preparing the platform for infiltration of anti-tumor immune cells. In a time of personalized medicine and genetic engineering, this "old tool" may be a way to augment efficacy and the depth of response to immune therapy.
Collapse
Affiliation(s)
- Manja Idorn
- Center for Cancer Immune Therapy, Herlev Gentofte University Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - Per Thor Straten
- Center for Cancer Immune Therapy, Herlev Gentofte University Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| |
Collapse
|
24
|
Shiozawa M, Chang CH, Huang YC, Chen YC, Chi MS, Hao HC, Chang YC, Takeda S, Chi KH, Wang YS. Pharmacologically upregulated carcinoembryonic antigen-expression enhances the cytolytic activity of genetically-modified chimeric antigen receptor NK-92MI against colorectal cancer cells. BMC Immunol 2018; 19:27. [PMID: 30075754 PMCID: PMC6091054 DOI: 10.1186/s12865-018-0262-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 07/11/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The natural killer cell line, NK-92MI, is cytotoxic against various types of cancer. The aim of this study was to develop chimeric antigen receptor-modified (CAR) NK-92MI cells targeting carcinoembryonic antigen-expressing (CEA) tumours and increase killing efficacy by pharmacologically modifying CEA-expression. RESULT We generated anti-CEA-CAR NK-92MI cells by retroviral vector transduction. This genetically-modified cell line recognised and lysed high CEA-expressing tumour cell lines (LS174T) at 47.54 ± 12.60% and moderate CEA-expressing tumour cell lines (WiDr) at 31.14 ± 16.92% at a 5:1 effector: target (E/T) ratio. The cell line did not lyse low CEA-expressing tumour cells (HCT116) as they did their parental cells (NK-92MI cells). The histone deacetylase-inhibitor (HDAC) sodium butyrate (NaB) and the methylation-inhibitor 5-azacytidine (5-AZA), as epigenetic modifiers, induced CEA-expression in HCT116 and WiDr cells. Although the IC50 of 5 fluorouracil (5-FU) increased, both cell lines showed collateral sensitivity to anti-CEA-CAR NK-92MI cells. The cytolytic function of anti-CEA-CAR NK-92MI cells was increased from 22.99 ± 2.04% of lysis background to 69.20 ± 11.92% after NaB treatment, and 69.70 ± 9.93% after 5-AZA treatment, at a 10:1 E/T ratio in HCT116 cells. The WiDr cells showed similar trend, from 22.99 ± 4.01% of lysis background to 70.69 ± 10.19% after NaB treatment, and 59.44 ± 10.92% after 5-AZA treatment, at a 10:1 E/T ratio. CONCLUSIONS This data indicates that the effector-ability of anti-CEA-CAR NK-92MI increased in a CEA-dependent manner. The combination of epigenetic-modifiers like HDAC-inhibitors, methylation-inhibitors, and adoptive-transfer of ex vivo-expanded allogeneic-NK cells may be clinically applicable to patients with in 5-FU resistant condition.
Collapse
Affiliation(s)
- Masayuki Shiozawa
- Department of Obstetrics and Gynecology, Juntendo University Hospital, 3-1-3 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Chuan-Hsin Chang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, No.95, Wenchang Road, Shilin District, Taipei, Taiwan.,Department of Research and Development, Johnpro Biotech Inc., 2F., No.118, Hougang St., Shilin Dist., Taipei City, Taiwan
| | - Yi-Chun Huang
- Department of Research and Development, Johnpro Biotech Inc., 2F., No.118, Hougang St., Shilin Dist., Taipei City, Taiwan
| | - Yi-Ching Chen
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, No.95, Wenchang Road, Shilin District, Taipei, Taiwan.,Department of Research and Development, Johnpro Biotech Inc., 2F., No.118, Hougang St., Shilin Dist., Taipei City, Taiwan
| | - Mau-Shin Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, No.95, Wenchang Road, Shilin District, Taipei, Taiwan.,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Room 117 Lab Building 1, 75 Bo-Ai Street, Hsinchu, Taiwan
| | - Hsu-Chao Hao
- Department of Biotechnology, Hungkuang University, No. 1018, Sec. 6, Taiwan Boulevard, Shalu District, Taichung City, Taiwan
| | - Yue-Cune Chang
- Department of Mathematics, Tamkang University, No.151, Yingzhuan Rd., Tamsui Dist., New Taipei City, Taiwan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University Hospital, 3-1-3 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, No.95, Wenchang Road, Shilin District, Taipei, Taiwan. .,Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.
| | - Yu-Shan Wang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, No.95, Wenchang Road, Shilin District, Taipei, Taiwan. .,Department of Research and Development, Johnpro Biotech Inc., 2F., No.118, Hougang St., Shilin Dist., Taipei City, Taiwan. .,Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Room 117 Lab Building 1, 75 Bo-Ai Street, Hsinchu, Taiwan.
| |
Collapse
|