1
|
Xu Z, Xiao ZX, Wang J, Qiu HW, Cao F, Zhang SQ, Xu YD, Lei HQ, Xia H, He YR, Zha GF, Pang J. Novel mRNA adjuvant ImmunER enhances prostate cancer tumor-associated antigen mRNA therapy via augmenting T cell activity. Oncoimmunology 2024; 13:2373526. [PMID: 38948931 PMCID: PMC11212567 DOI: 10.1080/2162402x.2024.2373526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024] Open
Abstract
Prostate cancer (PCa) is characterized as a "cold tumor" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.
Collapse
Affiliation(s)
- Zhen Xu
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ze-Xiu Xiao
- Drug Discovery Center, Shenzhen MagicRNA Biotech, Shenzhen, Guangdong, China
| | - Jing Wang
- Drug Discovery Center, Shenzhen MagicRNA Biotech, Shenzhen, Guangdong, China
| | - Hao-Wei Qiu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Fei Cao
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Shi-Qiang Zhang
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuan-Dong Xu
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Han-Qi Lei
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Heng Xia
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yun-Ru He
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Gao-Feng Zha
- Drug Discovery Center, Shenzhen MagicRNA Biotech, Shenzhen, Guangdong, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jun Pang
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Pugh KW, Alnaed M, Brackett CM, Blagg BSJ. The biology and inhibition of glucose-regulated protein 94/gp96. Med Res Rev 2022; 42:2007-2024. [PMID: 35861260 PMCID: PMC10003671 DOI: 10.1002/med.21915] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/10/2022]
Abstract
The 94 kDa molecular chaperone, glucose-regulated protein 94 (Grp94), has garnered interest during the last decade due to its direct association with endoplasmic reticulum (ER) stress and disease. Grp94 belongs to the Hsp90 family of molecular chaperones and is a master regulator of ER homeostasis due to its ability to fold and stabilize proteins/receptors, and to chaperone misfolded proteins for degradation. Multiple studies have demonstrated that Grp94 knockdown or inhibition leads to the degradation of client protein substrates, which leads to disruption of disease-dependent signaling pathways. As a result, small molecule inhibitors of Grp94 have become a promising therapeutic approach to target a variety of disease states. Specifically, Grp94 has proven to be a promising target for cancer, glaucoma, immune-mediated inflammation, and viral infection. Moreover, Grp94-peptide complexes have been utilized effectively as adjuvants for vaccines against a variety of disease states. This work highlights the significance of Grp94 biology and the development of therapeutics that target this molecular chaperone in multiple disease states.
Collapse
Affiliation(s)
- Kyler W. Pugh
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Marim Alnaed
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Christopher M. Brackett
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Brian S. J. Blagg
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana 46556, USA
| |
Collapse
|
3
|
Advanced Acral Melanoma Therapies: Current Status and Future Directions. Curr Treat Options Oncol 2022; 23:1405-1427. [PMID: 36125617 PMCID: PMC9526689 DOI: 10.1007/s11864-022-01007-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/17/2022]
Abstract
Melanoma is one of the deadliest malignancies. Its incidence has been significantly increasing in most countries in recent decades. Acral melanoma (AM), a peculiar subgroup of melanoma occurring on the palms, soles, and nails, is the main subtype of melanoma in people of color and is extremely rare in Caucasians. Although great progress has been made in melanoma treatment in recent years, patients with AM have shown limited benefit from current therapies and thus consequently have worse overall survival rates. Achieving durable therapeutic responses in this high-risk melanoma subtype represents one of the greatest challenges in the field. The frequency of BRAF mutations in AM is much lower than that in cutaneous melanoma, which prevents most AM patients from receiving treatment with BRAF inhibitors. However, AM has more frequent mutations such as KIT and CDK4/6, so targeted therapy may still improve the survival of some AM patients in the future. AM may be less susceptible to immune checkpoint inhibitors because of the poor immunogenicity. Therefore, how to enhance the immune response to the tumor cells may be the key to the application of immune checkpoint inhibitors in advanced AM. Anti-angiogenic drugs, albumin paclitaxel, or interferons are thought to enhance the effectiveness of immune checkpoint inhibitors. Combination therapies based on the backbone of PD-1 are more likely to provide greater clinical benefits. Understanding the molecular landscapes and immune microenvironment of AM will help optimize our combinatory strategies.
Collapse
|
4
|
Duan X, Iwanowycz S, Ngoi S, Hill M, Zhao Q, Liu B. Molecular Chaperone GRP94/GP96 in Cancers: Oncogenesis and Therapeutic Target. Front Oncol 2021; 11:629846. [PMID: 33898309 PMCID: PMC8062746 DOI: 10.3389/fonc.2021.629846] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/10/2021] [Indexed: 12/16/2022] Open
Abstract
During tumor development and progression, intrinsic and extrinsic factors trigger endoplasmic reticulum (ER) stress and the unfolded protein response, resulting in the increased expression of molecular chaperones to cope with the stress and maintain tumor cell survival. Heat shock protein (HSP) GRP94, also known as GP96, is an ER paralog of HSP90 and has been shown to promote survival signaling during tumor-induced stress and modulate the immune response through its multiple clients, including TLRs, integrins, LRP6, GARP, IGF, and HER2. Clinically, elevated expression of GRP94 correlates with an aggressive phenotype and poor clinical outcome in a variety of cancers. Thus, GRP94 is a potential molecular marker and therapeutic target in malignancies. In this review, we will undergo deep molecular profiling of GRP94 in tumor development and summarize the individual roles of GRP94 in common cancers, including breast cancer, colon cancer, lung cancer, liver cancer, multiple myeloma, and others. Finally, we will briefly review the therapeutic potential of selectively targeting GRP94 for the treatment of cancers.
Collapse
Affiliation(s)
- Xiaofeng Duan
- Department of Microbiology & Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Stephen Iwanowycz
- Department of Microbiology & Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Soo Ngoi
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Megan Hill
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Clinical Research Center for Cancer, Tianjin, China
| | - Bei Liu
- Department of Microbiology & Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- The Pelotonia Institute for Immuno-Oncology at The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
5
|
Ma S, Yang X, Zhou H, Zhang C, Kang J, Sun D. Combination of CpG Oligodeoxynucleotide and Anti-4-1BB Antibody in the Treatment of Multiple Hepatocellular Carcinoma in Mice. Onco Targets Ther 2020; 13:6997-7005. [PMID: 32764990 PMCID: PMC7381816 DOI: 10.2147/ott.s260353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
Background To investigate the effect of topical application of CpG oligodeoxynucleotide (CpG-ODN) combined with anti-4-1BB antibodies on mouse HCC multiple tumor-bearing models and the degree of improvement of anti-tumor immune response in mice. Materials and Methods We inoculated each BALB/c male mouse subcutaneously with one tumor in the axillae of the four limbs and divided them into four groups. We only selected the tumor-bearing part of the left lower limb for drug treatment. We measured the tumor-bearing volume of mice in each group. Then, we tested the organ coefficients of mice, the concentrations of IL-12 and IFN-γ in peripheral blood, the ratio of spleen Tregs and CD8+T cells, the spleen CTL killing activity, and the survival time of mice. Results We found that the tumor-bearing volume decreased significantly after the combination of CpG-ODN and anti-4-1BB antibody (P<0.001). The organ coefficients of treated mice were not significantly different from normal mice (P>0.05). The concentration of IL-12 and IFN-in serum and the ratio of CD8+T cells in spleen were increased, while the ratio of spleen Tregs was decreased. CTL activity of spleen was increased. The survival time of mice was significantly prolonged (P<0.001). Conclusion The treatment programme combining CpG-ODN with an anti-4-1BB antibody can significantly reduce tumor growth at the treatment site, slow the growth rate of metastases and improve host prognosis.
Collapse
Affiliation(s)
- Shizhao Ma
- The Liver Disease Center of PLA, The 980th Hospital of PLA Joint Logistics Support Force, Shijiazhuang 050082, People's Republic of China
| | - Xinying Yang
- The Liver Disease Center of PLA, The 980th Hospital of PLA Joint Logistics Support Force, Shijiazhuang 050082, People's Republic of China
| | - Huifang Zhou
- The Liver Disease Center of PLA, The 980th Hospital of PLA Joint Logistics Support Force, Shijiazhuang 050082, People's Republic of China
| | - Chaoqun Zhang
- The Liver Disease Center of PLA, The 980th Hospital of PLA Joint Logistics Support Force, Shijiazhuang 050082, People's Republic of China
| | - Jiwen Kang
- The Liver Disease Center of PLA, The 980th Hospital of PLA Joint Logistics Support Force, Shijiazhuang 050082, People's Republic of China
| | - Dianxing Sun
- The Liver Disease Center of PLA, The 980th Hospital of PLA Joint Logistics Support Force, Shijiazhuang 050082, People's Republic of China
| |
Collapse
|
6
|
Huang R, Li X, He Y, Zhu W, Gao L, Liu Y, Gao L, Wen Q, Zhong JF, Zhang C, Zhang X. Recent advances in CAR-T cell engineering. J Hematol Oncol 2020; 13:86. [PMID: 32616000 PMCID: PMC7333410 DOI: 10.1186/s13045-020-00910-5] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy is regarded as an effective solution for relapsed or refractory tumors, particularly for hematological malignancies. Although the initially approved anti-CD19 CAR-T therapy has produced impressive outcomes, setbacks such as high relapse rates and resistance were experienced, driving the need to discover engineered CAR-T cells that are more effective for therapeutic use. Innovations in the structure and manufacturing of CAR-T cells have resulted in significant improvements in efficacy and persistence, particularly with the development of fourth-generation CAR-T cells. Paired with an immune modifier, the use of fourth-generation and next-generation CAR-T cells will not be limited because of cytotoxic effects and will be an efficient tool for overcoming the tumor microenvironment. In this review, we summarize the recent transformations in the ectodomain, transmembrane domain, and endodomain of the CAR structure, which, together with innovative manufacturing technology and improved cell sources, improve the prospects for the future development of CAR-T cell therapy.
Collapse
MESH Headings
- Antigens, CD19/genetics
- Antigens, CD19/immunology
- Antigens, Neoplasm/immunology
- CD28 Antigens/chemistry
- CD28 Antigens/immunology
- Cell Engineering/trends
- Chemotaxis, Leukocyte
- Clinical Trials as Topic
- Cytokines/metabolism
- Genetic Vectors/genetics
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Lentivirus/genetics
- Lymphoma, Large B-Cell, Diffuse/therapy
- Neoplasms/therapy
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Protein Binding
- Protein Domains
- Protein Engineering
- Receptors, Chemokine/immunology
- Receptors, Chimeric Antigen/agonists
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Cell Antigen Receptor Specificity
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transduction, Genetic
- Tumor Microenvironment
Collapse
Affiliation(s)
- Ruihao Huang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Xiaoping Li
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Yundi He
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Wen Zhu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Lei Gao
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Yao Liu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Li Gao
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Qin Wen
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Jiang F Zhong
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Cheng Zhang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
7
|
Schirrmacher V. Cancer Vaccines and Oncolytic Viruses Exert Profoundly Lower Side Effects in Cancer Patients than Other Systemic Therapies: A Comparative Analysis. Biomedicines 2020; 8:biomedicines8030061. [PMID: 32188078 PMCID: PMC7148513 DOI: 10.3390/biomedicines8030061] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/05/2020] [Accepted: 03/08/2020] [Indexed: 12/29/2022] Open
Abstract
This review compares cytotoxic drugs, targeted therapies, and immunotherapies with regard to mechanisms and side effects. Targeted therapies relate to small molecule inhibitors. Immunotherapies include checkpoint inhibitory antibodies, chimeric antigen receptor (CAR) T-cells, cancer vaccines, and oncolytic viruses. All these therapeutic approaches fight systemic disease, be it micro-metastatic or metastatic. The analysis includes only studies with a proven therapeutic effect. A clear-cut difference is observed with regard to major adverse events (WHO grades 3-4). Such severe side effects are not observed with cancer vaccines/oncolytic viruses while they are seen with all the other systemic therapies. Reasons for this difference are discussed.
Collapse
|
8
|
Roszik J, Markovits E, Dobosz P, Layani A, Slabodnik-Kaner K, Baruch EN, Ben-Betzalel G, Grimm E, Berger R, Sidi Y, Schachter J, Shapira-Frommer R, Avni D, Markel G, Leibowitz-Amit R. TNFSF4 (OX40L) expression and survival in locally advanced and metastatic melanoma. Cancer Immunol Immunother 2019; 68:1493-1500. [PMID: 31501955 DOI: 10.1007/s00262-019-02382-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/20/2019] [Indexed: 01/16/2023]
Abstract
Immunotherapy with checkpoint inhibitors revolutionized melanoma treatment in both the adjuvant and metastatic setting, yet not all metastatic patients respond, and metastatic disease still often recurs among immunotherapy-treated patients with locally advanced disease. TNFSF4 is a co-stimulatory checkpoint protein expressed by several types of immune and non-immune cells, and was shown in the past to enhance the anti-neoplastic activity of T cells. Here, we assessed its expression in melanoma and its association with outcome in locally advanced and metastatic disease. We used publicly available data from The Cancer Genome Atlas (TCGA) and the Cancer Cell Line Encyclopedia (CCLE), and RNA sequencing data from anti-PD1-treated patients at Sheba medical center. TNFSF4 mRNA is expressed in melanoma cell lines and melanoma samples, including those with low lymphocytic infiltrates, and is not associated with the ulceration status of the primary tumor. Low expression of TNFSF4 mRNA is associated with worse prognosis in all melanoma patients and in the cohorts of stage III and stage IIIc-IV patients. Low expression of TNFSF4 mRNAs is also associated with worse prognosis in the subgroup of patients with low lymphocytic infiltrates, suggesting that tumoral TNFSF4 is associated with outcome. TNFSF4 expression was not correlated with the expression of other known checkpoint mRNAs. Last, metastatic patients with TNFSF4 mRNA expression within the lowest quartile have significantly worse outcome on anti-PD1 treatment, and a significantly lower response rate to these agents. Our current work points to TNFSF4 expression in melanoma as a potential determinant of prognosis, and warrants further translational and clinical research.
Collapse
Affiliation(s)
- Jason Roszik
- Departments of Melanoma Medical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Ettai Markovits
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Paula Dobosz
- Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Adi Layani
- Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Keren Slabodnik-Kaner
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Erez N Baruch
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Guy Ben-Betzalel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Elizabeth Grimm
- Departments of Melanoma Medical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Raanan Berger
- Department of Oncology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Division of Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.,Oncology Institute and Cancer Research Center, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Yehezkel Sidi
- Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Jacob Schachter
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.,Department of Oncology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Ronnie Shapira-Frommer
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Dror Avni
- Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Gal Markel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel. .,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.
| | - Raya Leibowitz-Amit
- Department of Oncology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel. .,Division of Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel. .,Oncology Institute and Cancer Research Center, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.
| |
Collapse
|
9
|
Lehmann JS, Rughwani P, Kolenovic M, Ji S, Sun B. LEGENDplex™: Bead-assisted multiplex cytokine profiling by flow cytometry. Methods Enzymol 2019; 629:151-176. [PMID: 31727238 DOI: 10.1016/bs.mie.2019.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past two decades there have been tremendous advances in our understanding of tumor immunology, which have in turn led to new and exciting immunology-based therapeutics. However, further research is needed into the dynamics and regulation of the immune response in the tumor microenvironment in order to achieve the full potential of these agents in treating all cancer patients. Defining the role of cytokines, chemokines, and other soluble mediators will be essential to this endeavor. This chapter describes, in detail, the technical protocol and applicability of LEGENDplex™ bead-based multiplex assays in quantifying these critical signaling molecules.
Collapse
|
10
|
Mukhopadhyay A, Wright J, Shirley S, Canton DA, Burkart C, Connolly RJ, Campbell JS, Pierce RH. Characterization of abscopal effects of intratumoral electroporation-mediated IL-12 gene therapy. Gene Ther 2019; 26:1-15. [PMID: 30323352 PMCID: PMC6514882 DOI: 10.1038/s41434-018-0044-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 12/16/2022]
Abstract
Intratumoral electroporation-mediated IL-12 gene therapy (IT-pIL12/EP) has been shown to be safe and effective in clinical trials, demonstrating systemic antitumor effects with local delivery of this potent cytokine. We recently optimized our IL-12 gene delivery platform to increase transgene expression and efficacy in preclinical models. Here we analyze the immunological changes induced with the new IT-pIL12/EP platform in both electroporated and distant, non-electroporated lesions. IT-pIL12/EP-treated tumors demonstrated rapid induction of IL-12-regulated pathways, as well as other cytokines and chemokines pathways, and upregulation of antigen presentation machinery. The distant tumors showed an increase in infiltrating lymphocytes and gene expression changes indicative of a de novo immune response in these untreated lesions. Flow cytometric analyses revealed a KLRG1hi CD8+ effector T-cell population uniquely present in mice treated with IT-pIL12/EP. Despite being highly activated, this population expressed diminished levels of PD-1 when re-exposed to antigen in the PD-L1-rich tumor. Other T-cell exhaustion markers appeared to be downregulated in concert, suggesting an orchestrated "armoring" of these effector T cells against T-cell checkpoints when primed in the presence of IL-12 in situ. These cells may represent an important mechanism by which local IL-12 gene therapy can induce a systemic antitumor immune response without the associated toxicity of systemic IL-12 exposure.
Collapse
Affiliation(s)
| | - Jocelyn Wright
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
| | - Shawna Shirley
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
| | - David A Canton
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
| | - Christoph Burkart
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
| | - Richard J Connolly
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, 98109, USA
| | - Jean S Campbell
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, 98109, USA
| | - Robert H Pierce
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, 98109, USA.
| |
Collapse
|
11
|
Fromm G, de Silva S, Johannes K, Patel A, Hornblower JC, Schreiber TH. Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy. J Immunother Cancer 2018; 6:149. [PMID: 30563566 PMCID: PMC6299665 DOI: 10.1186/s40425-018-0454-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022] Open
Abstract
Simultaneous blockade of immune checkpoint molecules and co-stimulation of the TNF receptor superfamily (TNFRSF) is predicted to improve overall survival in human cancer. TNFRSF co-stimulation depends upon coordinated antigen recognition through the T cell receptor followed by homotrimerization of the TNFRSF, and is most effective when these functions occur simultaneously. To address this mechanism, we developed a two-sided human fusion protein incorporating the extracellular domains (ECD) of PD-1 and OX40L, adjoined by a central Fc domain, termed PD1-Fc-OX40L. The PD-1 end of the fusion protein binds PD-L1 and PD-L2 with affinities of 2.08 and 1.76 nM, respectively, and the OX40L end binds OX40 with an affinity of 246 pM. High binding affinity on both sides of the construct translated to potent stimulation of OX40 signaling and PD1:PD-L1/L2 blockade, in multiple in vitro assays, including improved potency as compared to pembrolizumab, nivolumab, tavolixizumab and combinations of those antibodies. Furthermore, when activated human T cells were co-cultured with PD-L1 positive human tumor cells, PD1-Fc-OX40L was observed to concentrate to the immune synapse, which enhanced proliferation of T cells and production of IL-2, IFNγ and TNFα, and led to efficient killing of tumor cells. The therapeutic activity of PD1-Fc-OX40L in established murine tumors was significantly superior to either PD1 blocking, OX40 agonist, or combination antibody therapy; and required CD4+ T cells for maximum response. Importantly, all agonist functions of PD1-Fc-OX40L are independent of Fc receptor cross-linking. Collectively, these data demonstrate a highly potent fusion protein that is part of a platform, capable of providing checkpoint blockade and TNFRSF costimulation in a single molecule, which uniquely localizes TNFRSF costimulation to checkpoint ligand positive tumor cells.
Collapse
Affiliation(s)
- George Fromm
- Shattuck Labs, Inc, 21 Parmer Way, Suite 200, Durham, NC, 27703, USA
| | - Suresh de Silva
- Shattuck Labs, Inc, 21 Parmer Way, Suite 200, Durham, NC, 27703, USA
| | - Kellsey Johannes
- Shattuck Labs, Inc, 21 Parmer Way, Suite 200, Durham, NC, 27703, USA
| | - Arpita Patel
- Shattuck Labs, Inc, 21 Parmer Way, Suite 200, Durham, NC, 27703, USA
| | | | | |
Collapse
|
12
|
Overman MJ, Ernstoff MS, Morse MA. Where We Stand With Immunotherapy in Colorectal Cancer: Deficient Mismatch Repair, Proficient Mismatch Repair, and Toxicity Management. Am Soc Clin Oncol Educ Book 2018; 38:239-247. [PMID: 30231358 DOI: 10.1200/edbk_200821] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
With the recent U.S. Food and Drug Administration approvals of pembrolizumab and nivolumab for refractory deficient mismatch repair metastatic colorectal cancer, immune checkpoint inhibitors have now entered into clinical care for gastrointestinal cancers. Extensive ongoing efforts are exploring additional combinations of therapy in both deficient and proficient mismatch repair colorectal cancer. This review will outline the current status of such efforts and discuss the critical aspects of recognition and management of immune-related toxicities from checkpoint inhibitors.
Collapse
Affiliation(s)
- Michael J Overman
- From the Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Roswell Park Cancer Center, Buffalo, NY, Division of Hematology Oncology, Department of Medicine, University of Buffalo, Buffalo, NY; Department of Medicine, Duke University Medical Center, Durham, NC
| | - Marc S Ernstoff
- From the Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Roswell Park Cancer Center, Buffalo, NY, Division of Hematology Oncology, Department of Medicine, University of Buffalo, Buffalo, NY; Department of Medicine, Duke University Medical Center, Durham, NC
| | - Michael A Morse
- From the Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Roswell Park Cancer Center, Buffalo, NY, Division of Hematology Oncology, Department of Medicine, University of Buffalo, Buffalo, NY; Department of Medicine, Duke University Medical Center, Durham, NC
| |
Collapse
|
13
|
Manrique-Rincón AJ, de Carvalho AC, Ribeiro de Camargo ME, Franchini KG, Bajgelman MC. Development of a flow cytometry assay which allows to evaluate the efficiency of immunomodulatory vaccines to enhance T cell-mediated antitumor response. J Biotechnol 2018; 284:11-16. [PMID: 30053502 DOI: 10.1016/j.jbiotec.2018.07.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 11/18/2022]
Abstract
Immunotherapy has revolutionized the treatment of cancer. Since tumor cells exhibit low immunogenicity and can induce several mechanisms of tolerance, the use of monoclonal antibodies or other immunomodulators, targeting costimulation of T cells may mediate the inhibition of immunosuppressive mechanisms, favouring immune surveillance and enhancing the detection and elimination of tumor cells. We developed a new in vitro assay, based on flow cytometry, which allows exploring the therapeutic potential of tumor-derived immunomodulatory lineages, enhancing anti-tumor response. We generated tumor-derived cells that simultaneously co-express eGFP and one immunomodulatory molecule (OX40L, 4-1BBL or GM-CSF). These genetically modified tumor-derived cells are irradiated and then incubated with primary T cells to evaluate the killing activity, which can be estimated by a decrease in the eGFP positive cells. The results have shown correlation with in vivo experiments. This model may contribute to the development of high-throughput assays for the screening of immunomodulators and a reduction in the use of experimental animals.
Collapse
Affiliation(s)
- Andrea J Manrique-Rincón
- Brazilian Biosciences National Laboratory, Center for Research in Energy and Materials, Campinas-SP, Brazil; Medical School, University of Campinas, Campinas-SP, Brazil.
| | - Anna C de Carvalho
- Brazilian Biosciences National Laboratory, Center for Research in Energy and Materials, Campinas-SP, Brazil.
| | | | - Kleber G Franchini
- Brazilian Biosciences National Laboratory, Center for Research in Energy and Materials, Campinas-SP, Brazil; Medical School, University of Campinas, Campinas-SP, Brazil.
| | - Marcio C Bajgelman
- Brazilian Biosciences National Laboratory, Center for Research in Energy and Materials, Campinas-SP, Brazil; Medical School, University of Campinas, Campinas-SP, Brazil; Institute of Biology, University of Campinas, Campinas-SP, Brazil.
| |
Collapse
|
14
|
Qu X, Tang Y, Hua S. Immunological Approaches Towards Cancer and Inflammation: A Cross Talk. Front Immunol 2018; 9:563. [PMID: 29662489 PMCID: PMC5890100 DOI: 10.3389/fimmu.2018.00563] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
The inflammation is the protective response of the body against various harmful stimuli; however, the aberrant and inappropriate activation tends to become harmful. The acute inflammatory response tends to resolved once the offending agent is subside but this acute response becomes chronic in nature when the body is unable to successfully neutralized the noxious stimuli. This chronic inflammatory microenvironment is associated with the release of various pro-inflammatory and oncogenic mediators such as nitric oxide (NO), cytokines [IL-1β, IL-2, interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α)], growth factor, and chemokines. These mediators make the inflammatory microenvironment more vulnerable toward tumorigenesis. The pro-inflammatory mediators released during the chronic inflammation tends to induce several molecular signaling cascades such as nuclear factor kappa B, MAPKinase, nuclear factor erythroid 2-related factor 2, phosphoinositide-3-kinase, Janus kinases/STAT, Wnt/B-catenin, and cyclic AMP response element binding protein. The immune system and its components have a pleiotropic effect on inflammation and cancer progression. Immune components such as T cells, natural killer cells, macrophages, and neutrophils either inhibit or enhance tumor initiation depending on the type of tumor and immune cells involved. Tumor-associated macrophages and tumor-associated neutrophils are pro-tumorigenic cells highly prevalent in inflammation-mediated tumors. Similarly, presence of T regulatory (Treg) cells in an inflammatory and tumor setting suppresses the immune system, thus paving the way for oncogenesis. However, Treg cells also inhibit autoimmune inflammation. By contrast, cytotoxic T cells and T helper cells confer antitumor immunity and are associated with better prognosis in patients with cancer. Cytotoxic T cells inflict a direct cytotoxic effect on cells expressing oncogenic markers. Currently, several anti-inflammatory and antitumor therapies are under trials in which these immune cells are exploited. Adoptive cell transfer composed of tumor-infiltrating lymphocytes has been tried for the treatment of tumors after their ex vivo expansion. Mediators released by cells in a tumorigenic and inflammatory microenvironment cross talk with nearby cells, either promoting or inhibiting inflammation and cancer. Recently, several cytokine-based therapies are either being developed or are under trial to treat such types of manifestations. Monoclonal antibodies directed against TNF-α, VEGF, and IL-6 has shown promising results to ameliorate inflammation and cancer, while direct administration of IL-2 has been shown to cause tumor regression.
Collapse
Affiliation(s)
- Xinglong Qu
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| | - Ying Tang
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| | - Shucheng Hua
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Cabo M, Offringa R, Zitvogel L, Kroemer G, Muntasell A, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology 2017; 6:e1371896. [PMID: 29209572 PMCID: PMC5706611 DOI: 10.1080/2162402x.2017.1371896] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
The goal of cancer immunotherapy is to establish new or boost pre-existing anticancer immune responses that eradicate malignant cells while generating immunological memory to prevent disease relapse. Over the past few years, immunomodulatory monoclonal antibodies (mAbs) that block co-inhibitory receptors on immune effectors cells - such as cytotoxic T lymphocyte-associated protein 4 (CTLA4), programmed cell death 1 (PDCD1, best known as PD-1) - or their ligands - such as CD274 (best known as PD-L1) - have proven very successful in this sense. As a consequence, many of such immune checkpoint blockers (ICBs) have already entered the clinical practice for various oncological indications. Considerable attention is currently being attracted by a second group of immunomodulatory mAbs, which are conceived to activate co-stimulatory receptors on immune effector cells. Here, we discuss the mechanisms of action of these immunostimulatory mAbs and summarize recent progress in their preclinical and clinical development.
Collapse
Affiliation(s)
- Mariona Cabo
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
- DKFZ-Bayer Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France
- Université Pierre et Marie Curie/Paris VI, Paris
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
16
|
Malamas AS, Hammond SA, Schlom J, Hodge JW. Combination therapy with an OX40L fusion protein and a vaccine targeting the transcription factor twist inhibits metastasis in a murine model of breast cancer. Oncotarget 2017; 8:90825-90841. [PMID: 29207606 PMCID: PMC5710887 DOI: 10.18632/oncotarget.19967] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 07/23/2017] [Indexed: 12/22/2022] Open
Abstract
OX40 is a costimulatory receptor that potentiates proliferation, survival, memory formation, and effector function of CD4+ and CD8+ T-cells, while overcoming the suppressive activity of regulatory T-cells (Tregs). Here, we explored the combination of an OX40L fusion protein (OX40L-FP) with a poxvirus-based cancer vaccine (MVA-Twist-TRICOM) to inhibit tumor metastasis in the 4T1 murine breast cancer model. Contrary to the single agent treatments, the combination therapy significantly decreased the number of metastatic colonies per lung and prolonged survival. Depletion studies demonstrated that these effects were mediated by both CD4+ and CD8+ T-cells. The combination therapy a) increased the total number of T-cells in the CD4+Foxp3- population and the CD4+ central and effector memory subsets within the lung, spleen, and draining lymph node, b) enhanced infiltration of CD4+ T-cells into metastatic areas of the lung, and (c) increased the number of functional CD8+ T-cells that produced IFNγ and TNFα. The combination therapy also promoted the development of KLRG1-CD127+ memory precursor CD8+ T-cells, while reducing those with a KLRG1+ terminally differentiated phenotype. Moreover, the combination of OX40L-FP and vaccine induced greater CD4+ and CD8+ Twist-specific responses. In addition, Tregs isolated from mice receiving the combination were also less immunosuppressive in ex-vivo proliferation assays than those from the OX40L-FP and MVA-Twist-TRICOM monotherapy groups. Such results provide the rationale to combine co-stimulatory agonists with cancer vaccines for the treatment of tumor metastasis.
Collapse
Affiliation(s)
- Anthony S Malamas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Nowak EC, Lines JL, Varn FS, Deng J, Sarde A, Mabaera R, Kuta A, Le Mercier I, Cheng C, Noelle RJ. Immunoregulatory functions of VISTA. Immunol Rev 2017; 276:66-79. [PMID: 28258694 PMCID: PMC5702497 DOI: 10.1111/imr.12525] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Utilization of negative checkpoint regulators (NCRs) for cancer immunotherapy has garnered significant interest with the completion of clinical trials demonstrating efficacy. While the results of monotherapy treatments are compelling, there is increasing emphasis on combination treatments in an effort to increase response rates to treatment. One of the most recently discovered NCRs is VISTA (V-domain Ig-containing Suppressor of T cell Activation). In this review, we describe the functions of this molecule in the context of cancer immunotherapy. We also discuss factors that may influence the use of anti-VISTA antibody in combination therapy and how genomic analysis may assist in providing indications for treatment.
Collapse
Affiliation(s)
- Elizabeth C. Nowak
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - J. Louise Lines
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Frederick S. Varn
- Department of Biomedical Data Science and Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Jie Deng
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Aurelien Sarde
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Rodwell Mabaera
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Anna Kuta
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | | | - Chao Cheng
- Department of Biomedical Data Science and Department of Molecular and Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Randolph J. Noelle
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|