1
|
Faiena I, Adhikary S, Schweitzer C, Astrow SH, Grogan T, Funt SA, Bot A, Dorff T, Rosenberg JE, Elashoff DA, Pantuck AJ, Drakaki A. Gene and Protein Expression of MAGE and Associated Immune Landscape Elements in Non-Small-Cell Lung Carcinoma and Urothelial Carcinomas. J Immunother 2024; 47:351-360. [PMID: 39169899 PMCID: PMC11446647 DOI: 10.1097/cji.0000000000000538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/21/2024] [Indexed: 08/23/2024]
Abstract
Melanoma-associated antigen-A (MAGE-A) is expressed in multiple cancers with restricted expression in normal tissue. We sought to assess the MAGE-A3/A6 expression profile as well as immune landscape in urothelial (UC) and non-small cell lung carcinoma (NSCLC). We also assessed co-expression of immune-associated markers, including programmed cell death ligand 1 (PD-L1) in tumor and/or immune cells, and assessed the effect of checkpoint inhibitor treatment on these markers in the context of urothelial carcinoma. We used formalin-fixed paraffin-embedded (FFPE) tissue sections from a variety of tumor types were screened by IHC for MAGE-A and PD-L1 expression. Gene expression analyses by RNA sequencing were performed on RNA extracted from serial tissue sections. UC tumor samples from patients treated with checkpoint inhibitors were assessed by IHC and NanoString gene expression analysis for MAGE-A and immune marker expression before and after treatment. Overall, 84 samples (57%) had any detectable MAGE-A expression. Detectable MAGE-A expression was present at similar frequencies in both tumor tissue types, with 41 (50%) NSCLC and 43 (64%) UC. MAGE-A expression was not significantly changed before and after checkpoint inhibitor therapy by both IHC and NanoString mRNA sequencing. Other immune markers were similarly unchanged post immune checkpoint inhibitor therapy. Stable expression of MAGE-A3/A6 pre and post checkpoint inhibitor treatment indicates that archival specimens harvested after checkpoint therapy are applicable to screening potential candidates for MAGE therapies.
Collapse
Affiliation(s)
- Izak Faiena
- Columbia University Irving Medical Center, New York, NY
| | | | | | | | - Tristan Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Samuel A Funt
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Tanya Dorff
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | | | - David A Elashoff
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Allan J Pantuck
- Institute of Urologic Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Alexandra Drakaki
- Institute of Urologic Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
2
|
Jiang Y, Li H. The effect of smoking on tumor immunoediting: Friend or foe? Tob Induc Dis 2024; 22:TID-22-108. [PMID: 38887597 PMCID: PMC11181014 DOI: 10.18332/tid/189302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 05/15/2024] [Accepted: 05/25/2024] [Indexed: 06/20/2024] Open
Abstract
The recognition of smoking as an independent risk factor for lung cancer has become a widely accepted within the realm of respiratory medicine. The emergence of tumor immunotherapy has notably enhanced the prognosis for numerous late-stage cancer patients. Nevertheless, some studies have noted a tendency for lung cancer patients who smoke to derive greater benefit from immunotherapy. This observation has sparked increased interest in the interaction between smoking and the immune response to tumors in lung cancer. The concept of cancer immunoediting has shed light on the intricate and nuanced relationship between the immune system and tumors. Starting from the perspectives of immune surveillance, immune equilibrium, and immune evasion, this narrative review explores how smoking undermines the immune response against tumor cells and induces the generation of tumor neoantigens, and examines other behaviors that trigger tumor immune evasion. By elucidating these aspects, the review concludes that smoking is not conducive to tumor immunoediting.
Collapse
Affiliation(s)
- Yixia Jiang
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hequan Li
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Jia Q, Xian X, Li Y, Mu J, DU Z. Research progress in effects of MAGE-A family on gastric cancer. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:260-267. [PMID: 36999473 PMCID: PMC10930336 DOI: 10.11817/j.issn.1672-7347.2023.220042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Indexed: 04/01/2023]
Abstract
Gastric cancer (GC) is one of the most common malignant tumors worldwide, and most of the patients are diagnosed at the advanced stage. Most of the treating options are comprehensive treatment, in which immunotherapy plays more and more important role. Melanoma antigen-associated gene-A (MAGE-A) family is a kind of cancer testis antigens. Except in germ cells of testis and trophoblast cells of placenta, MAGE-A family is highly expressed in cancerous tissues and participates in a variety of biological processes, such as cancer cell proliferation, differentiation and metastasis. In addition, cancer testis antigen also possesses good immunogenicity, which can induce humoral and cellular immune responses, is a good target for immunotherapy, and has good application value in the diagnosis, treatment and prognosis of GC. A variety of targeted therapeutic drugs based on MAGE-A are in phase I or II clinical trials, it has good safety and potential clinical application value. With the continuous progress of clinical trials and basic research on MAGE-A targets in GC, it is expected to provide a theoretical basis for clinical transformation and immunotherapy of MAGE-A in the future.
Collapse
Affiliation(s)
- Qi Jia
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730000, China.
| | - Xiaohong Xian
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Yangrun Li
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Jiaxin Mu
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Zhixing DU
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730000, China.
| |
Collapse
|
4
|
Chen X, Pan X, Zhang W, Guo H, Cheng S, He Q, Yang B, Ding L. Epigenetic strategies synergize with PD-L1/PD-1 targeted cancer immunotherapies to enhance antitumor responses. Acta Pharm Sin B 2020; 10:723-733. [PMID: 32528824 PMCID: PMC7276686 DOI: 10.1016/j.apsb.2019.09.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/26/2019] [Accepted: 08/06/2019] [Indexed: 12/31/2022] Open
Abstract
Immunotherapy strategies targeting the programmed cell death ligand 1 (PD-L1)/programmed cell death 1 (PD-1) pathway in clinical treatments have achieved remarkable success in treating multiple types of cancer. However, owing to the heterogeneity of tumors and individual immune systems, PD-L1/PD-1 blockade still shows slow response rates in controlling malignancies in many patients. Accumulating evidence has shown that an effective response to anti-PD-L1/anti-PD-1 therapy requires establishing an integrated immune cycle. Damage in any step of the immune cycle is one of the most important causes of immunotherapy failure. Impairments in the immune cycle can be restored by epigenetic modification, including reprogramming the environment of tumor-associated immunity, eliciting an immune response by increasing the presentation of tumor antigens, and by regulating T cell trafficking and reactivation. Thus, a rational combination of PD-L1/PD-1 blockade and epigenetic agents may offer great potential to retrain the immune system and to improve clinical outcomes of checkpoint blockade therapy.
Collapse
Key Words
- 5-AzaC, 5-azacitidine
- ACE1, angiotensin converting enzyme
- ACP1, human red cell acid phosphatase
- APC, antigen-presenting cell
- BETi, bromodomain and extra-terminal motif inhibitors
- CCL22 (MDC), macrophage-derived chemokine
- CLL, chronic lymphocytic leukemia
- CTA, cancer testis antigen
- CTLA-4, cytotoxic T lymphocyte antigen 4
- CTLs, cytotoxic T lymphocytes
- CX3CL1, C-X3-C motif chemokine ligand 1
- CXCL, CXC chemokine ligand
- Cancer
- DC, dendritic cell
- DNMT1, DNA methyltransferase 1
- DNMTi, DNA methyltransferase inhibitors
- EZH2, enhancer of zeste homolog 2
- Epigenetic regulation
- FDA, U. S. Food and Drug Administration
- FOXP3, forkhead box P3
- H3K27me3, tri-methylation of lysine 27 on histone H3
- HDACi, histone deacetylase inhibitor
- IDO, indoleamine 2,3-dioxygenase
- IFN-γ, interferon-gamma
- Immune cycle
- Immunotherapy
- LAG-3, lymphocyte activation gene-3
- MDSCs, myeloid-derived suppressor cells
- MHC, major histocompatibility complex
- OS, overall survival
- PD-1, programmed cell death 1
- PD-L1, programmed cell death ligand 1
- PD-L1/PD-1 blockade
- PRC2, polycomb repressive complex 2
- TAA, tumor-associated antigen
- TET2, ten-eleven translocation 2
- TH-1, T helper type 1
- TIL, tumor infiltrating lymphocytes
- TIM-3, T cell immunoglobulin and mucin domain 3
- Tregs, regulatory T cells
- UHRF1, ubiquitin-like PHD and RING finger domain-containing 1
Collapse
|
5
|
Chen L, Qiao D, Wang J, Tian G, Wang M. Cancer immunotherapy with lymphocytes genetically engineered with T cell receptors for solid cancers. Immunol Lett 2019; 216:51-62. [PMID: 31597088 DOI: 10.1016/j.imlet.2019.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/18/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
Abstract
Adoptive transfer of T cells genetically engineered with chimeric antigen receptors (CAR-T cells) have proven to be highly effective for treating CD19+ B cell-derived hematologic malignancies. However, due to the lack of ideal tumor surface antigens, CAR-T cell therapy has limited success in treating solid tumors. T cells genetically engineered with T cell receptors (TCR-T cells) recognize intracellular and cell-surface antigens in the context of major histocompatibility complex (MHC) presentation and thus have the potential to access much more target antigens than CAR-T cells, providing great promise in treating solid tumors. There is an increasing interest in the application of TCR-T cell therapy for solid tumors, and fifty-six clinical trials are undergoing worldwide to confirm its validity. In this review, we summarize the recent progress in clinical studies of TCR-T cell therapy, describe strategies in the preparation and characterization of TCR-T cells, focusing on antigen selection, TCR isolation and methods to further enhance the potency of adoptively transferred cells.
Collapse
Affiliation(s)
- Lei Chen
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China
| | - Dongjuan Qiao
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China
| | - Juntao Wang
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China
| | - Geng Tian
- Department of Oncology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Mingjun Wang
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China.
| |
Collapse
|
6
|
Chen L, Tian Y, Zhan K, Chen A, Weng Z, Huang J, Li Y, Sun Y, Zheng H, Li Y. A humanized TCR retaining authentic specificity and affinity conferred potent anti-tumour cytotoxicity. Immunology 2018; 155:123-136. [PMID: 29645087 DOI: 10.1111/imm.12935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/05/2018] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
The affinity of T-cell receptor (TCR) determines the efficacy of TCR-based immunotherapy. By using human leucocyte antigen (HLA)-A*02 transgenic mice, a TCR was generated previously specific for human tumour testis antigen peptide MAGE-A3112-120 (KVAELVHFL) HLA-A*02 complex. We developed an approach to humanize the murine TCR by replacing the mouse framework with sequences of folding optimized human TCR variable domains for retaining binding affinity. The resultant humanized TCR exhibited higher affinity and conferred better anti-tumour activity than its parent murine MAGE-A3 TCR (SRm1). In addition, the affinity of humanized TCR was enhanced further to achieve improved T-cell activation. Our studies demonstrated that the human TCR variable domain frameworks could provide support for complementarity-determining regions from a murine TCR, and retain the original binding activity. It could be used as a generic approach of TCR humanization.
Collapse
Affiliation(s)
- Lin Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ye Tian
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Kai Zhan
- XiangXue Life Sciences Research Center, XiangXue Pharmaceutical Co. Ltd, Guangzhou, China
| | - Anan Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhiming Weng
- XiangXue Life Sciences Research Center, XiangXue Pharmaceutical Co. Ltd, Guangzhou, China
| | - Jiao Huang
- XiangXue Life Sciences Research Center, XiangXue Pharmaceutical Co. Ltd, Guangzhou, China
| | - Yanyan Li
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yongjie Sun
- Institute of Health Sciences, Anhui University, Hefei, China
| | - Hongjun Zheng
- XiangXue Life Sciences Research Center, XiangXue Pharmaceutical Co. Ltd, Guangzhou, China
| | - Yi Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,XiangXue Life Sciences Research Center, XiangXue Pharmaceutical Co. Ltd, Guangzhou, China
| |
Collapse
|
7
|
Isolation and Characterization of an HLA-DPB1*04: 01-restricted MAGE-A3 T-Cell Receptor for Cancer Immunotherapy. J Immunother 2018; 39:191-201. [PMID: 27163739 DOI: 10.1097/cji.0000000000000123] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Long-term tumor regressions have been observed in patients following the adoptive transfer of autologous tumor-infiltrating lymphocytes or genetically modified T cells expressing MHC class I-restricted T-cell receptors (TCRs), but clinical trials have not evaluated responses to genetically modified T cells expressing antitumor MHC class II-restricted TCRs. As studies carried out in a murine tumor model system have demonstrated that the adoptive transfer of CD4 T cells could lead to the regression of established tumors, we plan to test the hypothesis that CD4 T cells can also induce tumor regressions in cancer patients. In this study, 2 MAGE-A3-specific TCRs were isolated from a regulatory T-cell clone (6F9) and an effector clone (R12C9), generated from the peripheral blood of 2 melanoma patients after MAGE-A3 vaccination. The results indicated that T cells transduced with 6F9 TCR mediated stronger effector functions than R12C9 TCR. The 6F9 TCR specifically recognized MAGE-A3 and the closely related MAGE-A6 gene product, but not other members of the MAGE-A family in the context of HLA-DPB1*04:01. To test the feasibility of a potential clinical trial using this TCR, a clinical-scale procedure was developed to obtain a large number of purified CD4 T cells transduced with 6F9 TCR. Because HLA-DPB1*04:01 is present in ∼60% of the Caucasian population and MAGE-A3 is frequently expressed in a variety of cancer types, this TCR immunotherapy could potentially be applicable for a significant portion of cancer patients.
Collapse
|
8
|
Lu YC, Parker LL, Lu T, Zheng Z, Toomey MA, White DE, Yao X, Li YF, Robbins PF, Feldman SA, van der Bruggen P, Klebanoff CA, Goff SL, Sherry RM, Kammula US, Yang JC, Rosenberg SA. Treatment of Patients With Metastatic Cancer Using a Major Histocompatibility Complex Class II-Restricted T-Cell Receptor Targeting the Cancer Germline Antigen MAGE-A3. J Clin Oncol 2017; 35:3322-3329. [PMID: 28809608 DOI: 10.1200/jco.2017.74.5463] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Purpose Adoptive transfer of genetically modified T cells is being explored as a treatment for patients with metastatic cancer. Most current strategies use genes that encode major histocompatibility complex (MHC) class I-restricted T-cell receptors (TCRs) or chimeric antigen receptors to genetically modify CD8+ T cells or bulk T cells for treatment. Here, we evaluated the safety and efficacy of an adoptive CD4+ T-cell therapy using an MHC class II-restricted, HLA-DPB1*0401-restricted TCR that recognized the cancer germline antigen, MAGE-A3 (melanoma-associated antigen-A3). Patients and Methods Patients received a lymphodepleting preparative regimen, followed by adoptive transfer of purified CD4+ T cells, retrovirally transduced with MAGE-A3 TCR plus systemic high-dose IL-2. A cell dose escalation was conducted, starting at 107 total cells and escalating at half-log increments to approximately 1011 cells. Nine patients were treated at the highest dose level (0.78 to 1.23 × 1011 cells). Results Seventeen patients were treated. During the cell dose-escalation phase, an objective complete response was observed in a patient with metastatic cervical cancer who received 2.7 × 109 cells (ongoing at ≥ 29 months). Among nine patients who were treated at the highest dose level, objective partial responses were observed in a patient with esophageal cancer (duration, 4 months), a patient with urothelial cancer (ongoing at ≥ 19 months), and a patient with osteosarcoma (duration, 4 months). Most patients experienced transient fevers and the expected hematologic toxicities from lymphodepletion pretreatment. Two patients experienced transient grade 3 and 4 transaminase elevations. There were no treatment-related deaths. Conclusion These results demonstrate the safety and efficacy of administering autologous CD4+ T cells that are genetically engineered to express an MHC class II-restricted antitumor TCR that targets MAGE-A3. This clinical trial extends the reach of TCR gene therapy for patients with metastatic cancer.
Collapse
Affiliation(s)
- Yong-Chen Lu
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Linda L Parker
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Tangying Lu
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Zhili Zheng
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Mary Ann Toomey
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Donald E White
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Xin Yao
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Yong F Li
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Paul F Robbins
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Steven A Feldman
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Pierre van der Bruggen
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Christopher A Klebanoff
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Stephanie L Goff
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Richard M Sherry
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Udai S Kammula
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - James C Yang
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| | - Steven A Rosenberg
- Yong-Chen Lu, Linda L. Parker, Tangying Lu, Zhili Zheng, Mary Ann Toomey, Donald E. White, Xin Yao, Yong F. Li, Paul F. Robbins, Steven A. Feldman, Christopher A. Klebanoff, Stephanie L. Goff, Richard M. Sherry, Udai S. Kammula, James C. Yang, and Steven A. Rosenberg, National Cancer Institute, Bethesda, MD; Pierre van der Bruggen, Ludwig Institute for Cancer Research; De Duve Institute, Université Catholique de Louvain, Brussels; and Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium; Christopher A. Klebanoff, Memorial Sloan Kettering Cancer Center, Parker Institute for Cancer Immunotherapy, New York, NY
| |
Collapse
|
9
|
Lee AK, Potts PR. A Comprehensive Guide to the MAGE Family of Ubiquitin Ligases. J Mol Biol 2017; 429:1114-1142. [PMID: 28300603 DOI: 10.1016/j.jmb.2017.03.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/07/2017] [Accepted: 03/07/2017] [Indexed: 12/28/2022]
Abstract
Melanoma antigen (MAGE) genes are conserved in all eukaryotes and encode for proteins sharing a common MAGE homology domain. Although only a single MAGE gene exists in lower eukaryotes, the MAGE family rapidly expanded in eutherians and consists of more than 50 highly conserved genes in humans. A subset of MAGEs initially garnered interest as cancer biomarkers and immunotherapeutic targets due to their antigenic properties and unique expression pattern that is primary restricted to germ cells and aberrantly reactivated in various cancers. However, further investigation revealed that MAGEs not only drive tumorigenesis but also regulate pathways essential for diverse cellular and developmental processes. Therefore, MAGEs are implicated in a broad range of diseases including neurodevelopmental, renal, and lung disorders, and cancer. Recent biochemical and biophysical studies indicate that MAGEs assemble with E3 RING ubiquitin ligases to form MAGE-RING ligases (MRLs) and act as regulators of ubiquitination by modulating ligase activity, substrate specification, and subcellular localization. Here, we present a comprehensive guide to MAGEs highlighting the molecular mechanisms of MRLs and their physiological roles in germ cell and neural development, oncogenic functions in cancer, and potential as therapeutic targets in disease.
Collapse
Affiliation(s)
- Anna K Lee
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Patrick Ryan Potts
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA.
| |
Collapse
|
10
|
Liu M, Zhou J, Chen Z, Cheng ASL. Understanding the epigenetic regulation of tumours and their microenvironments: opportunities and problems for epigenetic therapy. J Pathol 2016; 241:10-24. [PMID: 27770445 DOI: 10.1002/path.4832] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/06/2016] [Accepted: 10/18/2016] [Indexed: 12/13/2022]
Abstract
The tumour microenvironment plays an instrumental role in cancer development, progression and treatment response/resistance. Accumulating evidence is underscoring the fundamental importance of epigenetic regulation in tumour immune evasion. Following many pioneering discoveries demonstrating malignant transformation through epigenetic anomalies ('epimutations'), there is also a growing emphasis on elucidating aberrant epigenetic mechanisms that reprogramme the milieu of tumour-associated immune and stromal cells towards an immunosuppressive state. Pharmacological inhibition of DNA methylation and histone modifications can augment the efficiency of immune checkpoint blockage, and unleash anti-tumour T-cell responses. However, these non-specific agents also represent a 'double-edged sword', as they can also reactivate gene transcription of checkpoint molecules, interrupting immune surveillance programmes. By understanding the impact of epigenetic control on the tumour microenvironment, rational combinatorial epigenetic and checkpoint blockage therapies have the potential to harness the immune system for the treatment of cancer. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Man Liu
- School of Biomedical Sciences and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| | - Jingying Zhou
- School of Biomedical Sciences and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| | - Zhiwei Chen
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Alfred Sze-Lok Cheng
- School of Biomedical Sciences and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| |
Collapse
|
11
|
Hou S, Sang M, Zhao L, Hou R, Shan B. The expression of MAGE-C1 and MAGE-C2 in breast cancer and their clinical significance. Am J Surg 2015; 211:142-51. [PMID: 26321295 DOI: 10.1016/j.amjsurg.2015.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 04/30/2015] [Accepted: 05/26/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Our study aims to analyze the expression pattern, mechanism, and prognostic significance of melanoma-associated antigen MAGE-C1 and MAGE-C2 in breast cancer. METHODS Reverse transcription polymerase chain reaction (RT-PCR) and immunohistochemistry were used to investigate the expressions of MAGE-C1 and MAGE-C2 in breast benign disease specimens, tumor-free breast specimens, and breast cancer specimens; their correlation with clinicopathologic parameters and recurrence-free survival was elucidated. We examined the influence of DNA methylase inhibitor 5-aza-2'-deoxycytidine (5-aza-CdR) together with histone deacetylase inhibitor trichostatin A on the expression of MAGE-C1 and MAGE-C2 in breast cancer cell lines. RESULT Proteins for MAGE-C1 and MAGE-C2 expressions were 38.3% and 58.3% in breast cancer specimens, messenger RNA for MAGE-C1 and MAGE-C2 expressions were 43.3% and 61.7%, respectively. MAGE-C1 and MAGE-C2 expressions were positively associated with high tumor grade and reduced recurrence-free survival; MAGE-C2 expression was also associated with tumor embolus and histologic type. 5-aza-CdR treatment alone could induce expression of MAGE-C2, whereas trichostatin A was able to synergistically enhance 5-aza-CdR-mediated MAGE-C2 transcription. CONCLUSIONS MAGE-C1 and MAGE-C2 maybe potential targets for tumor immunotherapy, and their expressions are associated with advanced breast cancer and poor outcome.
Collapse
Affiliation(s)
- Shuyun Hou
- Department of Immunology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China; Research Center, The Fourth Hospital of Hebei Medical University, Jiankang Road 12th, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Meixiang Sang
- Research Center, The Fourth Hospital of Hebei Medical University, Jiankang Road 12th, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Jiankang Road 12th, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Ran Hou
- Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Baoen Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Jiankang Road 12th, Shijiazhuang 050017, Hebei, People's Republic of China.
| |
Collapse
|
12
|
Esfandiary A, Ghafouri-Fard S. MAGE-A3: an immunogenic target used in clinical practice. Immunotherapy 2015; 7:683-704. [PMID: 26100270 DOI: 10.2217/imt.15.29] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Melanoma antigen family A, 3 (MAGE-A3) is a cancer-testis antigen whose expression has been demonstrated in a wide array of malignancies including melanoma, brain, breast, lung and ovarian cancer. In addition, its ability to elicit spontaneous humoral and cellular immune responses has been shown in cancer patients. As antigen-specific immune responses can be stimulated by immunization with MAGE-A3, several clinical trials have used MAGE-A3 vaccines to observe clinical responses. The frequent expressions of this antigen in various tumors and its immunogenicity in cancer patients have led to application of this antigen in cancer immunotherapy. However, the results of recent clinical trials indicate that there is a need for research in the vaccine design, adjuvant selection as well as patient selection criteria.
Collapse
Affiliation(s)
- Ali Esfandiary
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| |
Collapse
|
13
|
Li X, Mei Q, Nie J, Fu X, Han W. Decitabine: a promising epi-immunotherapeutic agent in solid tumors. Expert Rev Clin Immunol 2015; 11:363-75. [DOI: 10.1586/1744666x.2015.1002397] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
14
|
Zhao L, Zhang M, Cong H. Advances in the study of HLA-restricted epitope vaccines. Hum Vaccin Immunother 2013; 9:2566-77. [PMID: 23955319 DOI: 10.4161/hv.26088] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Vaccination is a proven strategy for protection from disease. An ideal vaccine would include antigens that elicit a safe and effective protective immune response. HLA-restricted epitope vaccines, which include T-lymphocyte epitopes restricted by HLA alleles, represent a new and promising immunization approach. In recent years, research in HLA-restricted epitope vaccines for the treatment of tumors and for the prevention of viral, bacterial, and parasite-induced infectious diseases have achieved substantial progress. Approaches for the improvement of the immunogenicity of epitope vaccines include (1) improving the accuracy of the methods used for the prediction of epitopes, (2) making use of additional HLA-restricted CD8(+) T-cell epitopes, (3) the inclusion of specific CD4(+) T-cell epitopes, (4) adding B-cell epitopes to the vaccine construction, (5) finding more effective adjuvants and delivery systems, (6) using immunogenic carrier proteins, and (7) using multiple proteins as epitopes sources. In this manuscript, we review recent research into HLA-restricted epitope vaccines.
Collapse
Affiliation(s)
- Lingxiao Zhao
- Department of Human Parasitology; Shandong University School of Medicine; Shandong, P.R. China
| | - Min Zhang
- Department of Human Parasitology; Shandong University School of Medicine; Shandong, P.R. China
| | - Hua Cong
- Department of Human Parasitology; Shandong University School of Medicine; Shandong, P.R. China
| |
Collapse
|
15
|
Srivastava RM, Lee SC, Andrade Filho PA, Lord CA, Jie HB, Davidson HC, López-Albaitero A, Gibson SP, Gooding WE, Ferrone S, Ferris RL. Cetuximab-activated natural killer and dendritic cells collaborate to trigger tumor antigen-specific T-cell immunity in head and neck cancer patients. Clin Cancer Res 2013; 19:1858-72. [PMID: 23444227 DOI: 10.1158/1078-0432.ccr-12-2426] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Tumor antigen-specific monoclonal antibodies (mAb) block oncogenic signaling and induce Fcγ receptor (FcγR)-mediated cytotoxicity. However, the role of CD8(+) CTL and FcγR in initiating innate and adaptive immune responses in mAb-treated human patients with cancer is still emerging. EXPERIMENTAL DESIGN FcγRIIIa codon 158 polymorphism was correlated with survival in 107 cetuximab-treated patients with head and neck cancer (HNC). Flow cytometry was carried out to quantify EGF receptor (EGFR)-specific T cells in cetuximab-treated patients with HNC. The effect of cetuximab on natural killer (NK) cell, dendritic cell (DC), and T-cell activation was measured using IFN-γ release assays and flow cytometry. RESULTS FcγRIIIa polymorphism did not predict clinical outcome in cetuximab-treated patients with HNC; however, elevated circulating EGFR(853-861)-specific CD8(+) T cells were found in cetuximab-treated patients with HNC (P < 0.005). Cetuximab promoted EGFR-specific cellular immunity through the interaction of EGFR(+) tumor cells and FcγRIIIa on NK cells but not on the polymorphism per se. Cetuximab-activated NK cells induced IFN-γ-dependent expression of DC maturation markers, antigen processing machinery components such as TAP-1/2 and T-helper cell (T(H)1) chemokines through NKG2D/MICA binding. Cetuximab initiated adaptive immune responses via NK cell-induced DC maturation, which enhanced cross-presentation to CTL specific for EGFR as well as another tumor antigen, MAGE-3. CONCLUSION Cetuximab-activated NK cells promote DC maturation and CD8(+) T-cell priming, leading to tumor antigen spreading and TH1 cytokine release through "NK-DC cross-talk." FcγRIIIa polymorphism did not predict clinical response to cetuximab but was necessary for NK-DC interaction and mAb-induced cross-presentation. EGFR-specific T cells in cetuximab-treated patients with HNC may contribute to clinical response.
Collapse
|
16
|
Schuler PJ, Hoffmann TK, Gauler TC, Bergmann C, Brandau S, Lang S. [Immunotherapy of head and neck cancer. Current developments]. HNO 2012; 61:559-72. [PMID: 23247754 DOI: 10.1007/s00106-012-2635-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In order to improve the prognosis for patients with head and neck squamous cell cancer (HNSCC) the introduction of new therapeutic strategies is necessary. The concept of immunotherapy has been applied and improved for several years and recent studies have used tumor-specific antigens which facilitates targeted oncologic therapy. However, immunotherapy is hampered by the fact that immunosuppressive mechanisms are pronounced and relevant effector cells are suppressed, especially in patients with HNSCC. Successful immunotherapy could induce an antitumor immune response by restitution of these cell populations. Current anti-tumor immunotherapy includes unspecific immune stimulation, genetic modification of tumor and immune cells, the use of monoclonal antibodies, e.g. cetuximab, adoptive cell transfer and tumor vaccination. In the future, these biologic therapies alone or in combination with conventional therapeutic regimens could present a valuable therapeutic option for HNSCC patients.
Collapse
Affiliation(s)
- P J Schuler
- Klinik und Poliklinik für Hals-, Nasen- und Ohrenkrankheiten, Universität Duisburg-Essen.
| | | | | | | | | | | |
Collapse
|
17
|
Stiff PJ, Potkul RK, Venkataraman G, Sojitra P, Drakes ML. Immune Surveillance Tissue Antigen Profiling in Advanced Ovarian Cancer. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.cogc.2012.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
18
|
Analysis of compensatory substitution and gene evolution on the MAGEA/CSAG-palindrome of the primate X chromosomes. Comput Biol Chem 2012; 42:18-22. [PMID: 23257410 DOI: 10.1016/j.compbiolchem.2012.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 11/06/2012] [Accepted: 11/13/2012] [Indexed: 11/20/2022]
Abstract
The human X chromosome contains a large number of inverted repeat DNA palindromes. Although arbitrary substitutions destroyed the inverted repeat structure of MAGEA/CSAG-palindrome during the evolutionary process of the primates, most of the substitutions are compensatory. Using maximum parsimony, it is demonstrated that the compensatory substitutions are prone to occur between bases with similar structures on the human, chimpanzee and orangutan MAGEA/CSAG-palindromes. Furthermore, it is found that MAGEA/CSAG genes also exist in orangutan and rhesus monkey palindromes by homologous searching. This suggests that the MAGEA/CSAG-palindrome might predate the divergence of human and other primate lineages. Comparative sequence analysis of the arms and genes on the primate MAGEA/CSAG-palindromes provides possible evidence of subsequently arm to arm gene conversion. These compensatory substitutions on the MAGEA/CSAG-palindrome of the primate X chromosomes play an important role in maintaining their structural symmetry during the process of formation.
Collapse
|
19
|
Ferris RL, Lotze MT, Leong SPL, Hoon DSB, Morton DL. Lymphatics, lymph nodes and the immune system: barriers and gateways for cancer spread. Clin Exp Metastasis 2012; 29:729-36. [PMID: 22851005 PMCID: PMC3485421 DOI: 10.1007/s10585-012-9520-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Accepted: 07/14/2012] [Indexed: 12/23/2022]
Abstract
Metastasis to the regional lymph node is the most important prognostic indicator for the outcomes of patients with sold cancer. In general, it is well recognized that cancer development is genetically determined with progression from the microenvironment of the primary tumor site, oftentimes via the SLN gateway, to the distant sites. In about 20 % of the time, the cancer cells may spread directly through the blood vascular system to the distant sites. Thus, in general, cancer progression is consistent with Hellman's spectrum theory in that development of nodal and systemic metastasis from a localized cancer growth is a progressive process. Cancer proliferation within the tumor microenvironment may give rise to increased tumor heterogeneity, which is further complicated by its continuous change through its evolution within the host in a Darwinian sense. It is crucial to understand the molecular process of lymphangiogenesis and hemangiogenesis in the tumor microenvironment with respect to the initial steps of cancer cells entering into the lymphatic and vascular systems so that rational therapy can be developed to curb the process of specific routes of metastasis. This chapter elucidates the role of lymphatics, nodal metastasis and antitumor immunity. We present novel immune targets in nodal metastases, the importance of the lymph node as a pre-metastatic niche, and immune-related proteins as biomarkers of metastasis.
Collapse
Affiliation(s)
- Robert L Ferris
- Hillman Cancer Center Research, Pavilion 5117 Centre Avenue, Room 2.26b, Pittsburgh, PA 15213, USA.
| | | | | | | | | |
Collapse
|
20
|
Toyoshima T, Kumamaru W, Hayashida JN, Moriyama M, Kitamura R, Tanaka H, Yamada A, Itoh K, Nakamura S. In vitro induction of specific CD8+ T lymphocytes by tumor-associated antigenic peptides in patients with oral squamous cell carcinoma. Cancer Lett 2012; 322:86-91. [DOI: 10.1016/j.canlet.2012.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Revised: 02/15/2012] [Accepted: 02/15/2012] [Indexed: 10/28/2022]
|
21
|
Ferris RL, Stefanika P, Xi L, Gooding W, Seethala RR, Godfrey TE. Rapid molecular detection of metastatic head and neck squamous cell carcinoma as an intraoperative adjunct to sentinel lymph node biopsy. Laryngoscope 2012; 122:1020-30. [PMID: 22447185 DOI: 10.1002/lary.22467] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 06/28/2011] [Indexed: 11/10/2022]
Abstract
OBJECTIVES/HYPOTHESIS Clinical staging of early head and neck squamous cell carcinoma (SCCHN) is often inaccurate, leading to elective neck dissection to detect the 30% of patients with micrometastatic disease. Sentinel node biopsy accurately stages the regional lymphatics, but intraoperative pathology is only moderately sensitive, and final pathology takes several days to complete. To facilitate immediate neck dissection where necessary, we have identified several promising marker genes of SCCHN metastasis and developed a rapid, accurate, and automated quantitative real-time polymerase chain reaction (PCR) (qRT-PCR) assay for intraoperative use. STUDY DESIGN Prospective tissue collection, retrospective pathologic correlation with qRT-PCR. METHODS From a 40-gene marker screen, we quantified expression of 11 potential tumor genes using a test set of primary tumors (n = 32) and metastatic (n = 19) and benign (n = 10) lymph nodes. Eight patients' paired primary tumor and metastatic nodes were included. A validation set of 442 grossly tumor-negative nodes was evaluated for expression of the most promising markers, comparing metastasis detection by qRT-PCR with pathologic analysis (hematoxylin and eosin and immunohistochemistry). A novel multiplexed, automated, single-tube qRT-PCR assay was used to analyze more than 100 lymph nodes using a two-marker, 35-minute assay to determine its negative predictive value. RESULTS Based on expression of 11 tumor-associated genes from the marker screen, the two most promising markers of SCCHN metastasis in the test set, pemphigus vulgaris antigen (PVA) and tumor-associated calcium signal transducer 1 (TACSTD1), also known as epithelial cell adhesion molecule (EpCAM), were selected. Development of a multiplexed qRT-PCR assay for the detection of metastasis compared favorably with pathologic analysis in the additional 442-node set. A rapid, multiplexed assay using PVA and TACSTD1 demonstrated excellent reproducibility, linearity, and accuracy (≈96% negative predictive value) for identifying positive (n = 40) and negative (n = 62) nodes in a validation subset. CONCLUSIONS Detection of metastatic SCCHN using multiplexed qRT-PCR can be rapid, accurate, and automated and may enable sentinel node biopsy to be used for intraoperative decision-making. PCR amplification of tumor marker genes is an effective method of intraoperative molecular staging of SCCHN and could more appropriately guide application of neck dissection in pN+ SCCHN patients, sparing 60% to 70% of pN0 patients from unnecessary neck dissection. This technique may also be used for identifying residual neck disease posttreatment, using outpatient fine-needle aspiration biopsy specimens.
Collapse
Affiliation(s)
- Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Allen CT, Judd NP, Bui JD, Uppaluri R. The clinical implications of antitumor immunity in head and neck cancer. Laryngoscope 2011; 122:144-57. [DOI: 10.1002/lary.21913] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
23
|
Rahma OE, Khleif SN. Therapeutic vaccines for gastrointestinal cancers. Gastroenterol Hepatol (N Y) 2011; 7:517-64. [PMID: 22298988 PMCID: PMC3264936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Despite progress in the management of gastrointestinal malignancies, these diseases remain devastating maladies. Conventional treatment with chemotherapy and radiation is still only partially effective and highly toxic. In the era of increasing knowledge of the molecular biology of tumors and the interaction between the tumor and immune system, the development of targeted agents, including cancer vaccines, has emerged as a promising modality. In this paper, we discuss the principals of vaccine development, and we review most of the published trials on gastrointestinal cancer vaccines that have been conducted over the last decade. Many antigens and various treatment approaches have already been tested in colon, pancreatic, and other cancers. Some of these approaches have already shown some clinical benefit. In this paper, we discuss these different strategies and some of the future directions for targeting gastrointestinal malignancies with vaccines.
Collapse
|
24
|
Fratta E, Coral S, Covre A, Parisi G, Colizzi F, Danielli R, Nicolay HJM, Sigalotti L, Maio M. The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Mol Oncol 2011; 5:164-82. [PMID: 21376678 DOI: 10.1016/j.molonc.2011.02.001] [Citation(s) in RCA: 267] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 01/31/2011] [Accepted: 02/03/2011] [Indexed: 12/14/2022] Open
Abstract
Cancer testis antigens (CTA) are a large family of tumor-associated antigens expressed in human tumors of different histological origin, but not in normal tissues except for testis and placenta. This tumor-restricted pattern of expression, together with their strong in vivo immunogenicity, identified CTA as ideal targets for tumor-specific immunotherapeutic approaches, and prompted the development of several clinical trials of CTA-based vaccine therapy. Driven by this practical clinical interest, a more detailed characterization of CTA biology has been recently undertaken. So far, at least 70 families of CTA, globally accounting for about 140 members, have been identified. Most of these CTA are expressed during spermatogenesis, but their function is still largely unknown. Epigenetic events, particularly DNA methylation, appear to be the primary mechanism regulating CTA expression in both normal and transformed cells, as well as in cancer stem cells. In view of the growing interest in CTA biology, the aim of this review is to provide the most recent information on their expression, regulation and function, together with a brief summary of the major clinical trials involving CTA as therapeutic agents. The pharmacologic modulation of CTA expression profiles on neoplastic cells by DNA hypomethylating drugs will also be discussed as a feasible approach to design new combination therapies potentially able to improve the clinical efficacy of currently adopted CTA-based immunotherapeutic regimens in cancer patients.
Collapse
Affiliation(s)
- Elisabetta Fratta
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, Via Franco Gallini 2, 33081 Aviano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chinnasamy N, Wargo JA, Yu Z, Rao M, Frankel TL, Riley JP, Hong JJ, Parkhurst MR, Feldman SA, Schrump DS, Restifo NP, Robbins PF, Rosenberg SA, Morgan RA. A TCR targeting the HLA-A*0201-restricted epitope of MAGE-A3 recognizes multiple epitopes of the MAGE-A antigen superfamily in several types of cancer. THE JOURNAL OF IMMUNOLOGY 2010; 186:685-96. [PMID: 21149604 DOI: 10.4049/jimmunol.1001775] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Adoptive immunotherapy using TCR-engineered PBLs against melanocyte differentiation Ags mediates objective tumor regression but is associated with on-target toxicity. To avoid toxicity to normal tissues, we targeted cancer testis Ag (CTA) MAGE-A3, which is widely expressed in a range of epithelial malignancies but is not expressed in most normal tissues. To generate high-avidity TCRs against MAGE-A3, we employed a transgenic mouse model that expresses the human HLA-A*0201 molecule. Mice were immunized with two HLA-A*0201-restricted peptides of MAGE-A3: 112-120 (KVAELVHFL) or MAGE-A3: 271-279 (FLWGPRALV), and T cell clones were generated. MAGE-A3-specific TCR α- and β-chains were isolated and cloned into a retroviral vector. Expression of both TCRs in human PBLs demonstrated Ag-specific reactivity against a range of melanoma and nonmelanoma tumor cells. The TCR against MAGE-A3: 112-120 was selected for further development based on superior reactivity against tumor target cells. Interestingly, peptide epitopes from MAGE-A3 and MAGE-A12 (and to a lesser extent, peptides from MAGE-A2 and MAGE-A6) were recognized by PBLs engineered to express this TCR. To further improve TCR function, single amino acid variants of the CDR3 α-chain were generated. Substitution of alanine to threonine at position 118 of the α-chain in the CDR3 region of the TCR improved its functional avidity in CD4 and CD8 cells. On the basis of these results, a clinical trial is planned in which patients bearing a variety of tumor histologies will receive autologous PBLs that have been transduced with this optimized anti-MAGE-A3 TCR.
Collapse
Affiliation(s)
- Nachimuthu Chinnasamy
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Badoual C, Sandoval F, Pere H, Hans S, Gey A, Merillon N, Van Ryswick C, Quintin-Colonna F, Bruneval P, Brasnu D, Fridman WH, Tartour E. Better understanding tumor-host interaction in head and neck cancer to improve the design and development of immunotherapeutic strategies. Head Neck 2010; 32:946-58. [PMID: 20191626 DOI: 10.1002/hed.21346] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Head and neck cancers are heavily infiltrated by immune cells, the significance of which is complex. The natural immune response against head and neck tumors, including anti-human papillomavirus (HPV) T cells, and humoral responses has been clearly documented. However, during the course of tumor progression, co-option of the immune system by tumor cells for their own advantage and increased resistance of tumor cells to immune attack also occur. Inflammation and immune subversion to support angiogenesis are key factors promoting tumor growth. Only a better understanding of this tumor-host interaction will permit a rational design of new immunotherapeutic approaches combining immunostimulation with drugs endowed with the ability to counteract immunoevasion mechanisms.
Collapse
Affiliation(s)
- Cécile Badoual
- EA 4054 Universite Paris Descartes, Ecole Nationale Vétérinaire d'Alfort, 7 Avenue du Général de Gaulle, 94704 Maisons Alfort, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Andrade Filho PA, Ito D, DeLeo AB, Ferris RL. CD8+ T cell recognition of polymorphic wild-type sequence p53(65-73) peptides in squamous cell carcinoma of the head and neck. Cancer Immunol Immunother 2010; 59:1561-8. [PMID: 20577877 PMCID: PMC4109029 DOI: 10.1007/s00262-010-0886-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 06/10/2010] [Indexed: 10/19/2022]
Abstract
The TP53 tumor suppressor gene contains a well-studied polymorphism that encodes either proline (P) or arginine (R) at codon 72, and over half of the world's population is homozygous for R at this codon. The wild-type sequence (wt) p53 peptide, p53(65-73), has been identified as a CD8+ T cell-defined tumor antigen for use in broadly applicable cancer vaccines. However, depending on the TP53 codon 72 polymorphism of the recipient, the induced responses to the peptides incorporating R (p53(72R)) or P (p53(72P)) can be "self" or "non-self." Thus, we sought to determine which wt p53(65-73) peptide should be used in wt p53-based cancer vaccines. Despite similar predicted HLA-A2-binding affinities, the p53(72P) peptide was more efficient than the p53(72R) peptide in HLA-A2 stabilization assays. In vitro stimulation (IVS) of CD8+ T cells obtained from healthy HLA-A2(+) donors with these two peptides led to the generation of CD8+ T cell effectors in one-third of the samples tested, at a frequency similar to the responsiveness to other wt p53 peptides. Interestingly, regardless of their p53 codon 72 genotype, CD8+ T cells stimulated with either p53(72P) or p53(72R) peptide were cross-reactive against T2 cells pulsed with either peptide, as well as HLA-A2(+) head and neck cancer (HNC) cell lines presenting p53(72P) and/or p53(72R) peptides for T cell recognition. Therefore, the cross-reactivity of CD8+ T cells for the polymorphic wt p53(65-73) peptides, irrespective of their p53 codon 72 polymorphism, suggests that employing either peptide in wt p53-based vaccines can result in efficient targeting of this epitope.
Collapse
Affiliation(s)
- Pedro A. Andrade Filho
- Department of Otolaryngology, University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, USA
| | - Daisuke Ito
- Department of Pathology, University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Albert B. DeLeo
- Department of Pathology, University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Robert L. Ferris
- Department of Otolaryngology, University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, USA
- Department of Immunology, University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, USA
- UPCI Research Pavilion, The Hillman Cancer Center, 5117 Centre Avenue, Room 2.26b, Pittsburgh, PA 15213-1863 USA
| |
Collapse
|
28
|
Figueiredo DLA, Mamede RCM, Spagnoli GC, Silva WA, Zago M, Neder L, Jungbluth AA, Saggioro FP. High expression of cancer testis antigens MAGE-A, MAGE-C1/CT7, MAGE-C2/CT10, NY-ESO-1, and gage in advanced squamous cell carcinoma of the larynx. Head Neck 2010; 33:702-7. [PMID: 20886663 DOI: 10.1002/hed.21522] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 05/11/2010] [Accepted: 05/13/2010] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Despite diagnostic and therapeutic advances in head and neck cancer, the 5-year survival of patients with laryngeal cancer has not improved in the last 30 years. Several recent studies indicate that specific targets for immunotherapeutic approaches can be useful in the control of cancer. There is considerable interest in the expression of cancer testis antigens in human cancers since they may serve as the basis for an immunologic approach to therapy. METHODS We evaluated by immunohistochemical analysis the expression of cancer testis antigens MAGE-A4 (57B), MAGE-C1 (CT7-33), MAGE-A1 (MA454), MAGE-A3 (M3H67), MAGE-C2 (CT10.5), NY-ESO-1 (E978), and GAGE (GAGE) in squamous cell carcinoma (SCC) of the larynx. RESULTS A total of 63 cases (57 men and 6 women) of laryngeal SCC were available for this study. The findings were correlated with the clinical course and laboratory data. Expression of at least 1 cancer testis antigen was detected in 42 of 63 of the laryngeal SCCs (67%). In 34 of 42 of the positive cases (81%) there was simultaneous expression of ≥2 cancer testis antigens. There was significant correlation between antigen expression and advanced tumor stage (stage III/IV) in cases with reactivity to only 1 antibody (p = .01) as well as in the cases with reactivity to ≥2 primary antibodies (≥2 mAbs, p = .04). There was no association between survival and expression of any of the analyzed antigens. CONCLUSIONS We find a high incidence of cancer testis antigen expression in SCCs of the larynx, which was correlated with advanced clinical stage. Our data indicate that cancer testis antigens could be valuable vaccine targets in laryngeal tumors, especially in those with a worse prognosis.
Collapse
Affiliation(s)
- David L A Figueiredo
- Department of Ophthalmology, Otorhinolaringology and Head and Neck Surgery, Faculty of Medicine, University of São Paulo, Ribeirão Preto, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Laing M, Cummins R, O’Grady A, O’Kelly P, Kay E, Murphy G. Aberrant DNA methylation associated with MTHFR
C677T genetic polymorphism in cutaneous squamous cell carcinoma in renal transplant patients. Br J Dermatol 2010; 163:345-52. [DOI: 10.1111/j.1365-2133.2010.09774.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|