1
|
Zhou Y, Qin X, Hu Q, Qin S, Xu R, Gu K, Lu H. Cross-talk between disulfidptosis and immune check point genes defines the tumor microenvironment for the prediction of prognosis and immunotherapies in glioblastoma. Sci Rep 2024; 14:3901. [PMID: 38365809 PMCID: PMC10873294 DOI: 10.1038/s41598-024-52128-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/14/2024] [Indexed: 02/18/2024] Open
Abstract
Disulfidptosis is a condition where dysregulated NAPDH levels and abnormal accumulation of cystine and other disulfides occur in cells with high SLC7A11 expression under glucose deficiency. This disrupts normal formation of disulfide bonds among cytoskeletal proteins, leading to histone skeleton collapse and triggering cellular apoptosis. However, the correlation between disulfidptosis and immune responses in relation to glioblastoma survival rates and immunotherapy sensitivity remains understudied. Therefore, we utilized The Cancer Genome Atlas and The Chinese Glioma Genome Atlas to identify disulfidptosis-related immune checkpoint genes and established an overall survival (OS) prediction model comprising six genes: CD276, TNFRSF 14, TNFSF14, TNFSF4, CD40, and TNFRSF18, which could also be used for predicting immunotherapy sensitivity. We identified a cohort of glioblastoma patients classified as high-risk, which exhibited an upregulation of angiogenesis, extracellular matrix remodeling, and epithelial-mesenchymal transition as well as an immunosuppressive tumor microenvironment (TME) enriched with tumor associated macrophages, tumor associated neutrophils, CD8 + T-cell exhaustion. Immunohistochemical staining of CD276 in 144 cases further validated its negative correlation with OS in glioma. Disulfidptosis has the potential to induce chronic inflammation and an immunosuppressive TME in glioblastoma.
Collapse
Affiliation(s)
- Yanjun Zhou
- Department of Radiotherapy and Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214000, Jiangsu, China.
| | - Xue Qin
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Qunchao Hu
- Department of Radiation Oncology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China, Shanghai
| | - Shaolei Qin
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Ran Xu
- Department of Neurosurgery, Affiliated Hospital of Jiangnan University, Wuxi, 214125, Jiangsu, China
| | - Ke Gu
- Department of Radiotherapy and Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214000, Jiangsu, China.
| | - Hua Lu
- Department of Neurosurgery, Affiliated Hospital of Jiangnan University, Wuxi, 214125, Jiangsu, China.
| |
Collapse
|
2
|
Chen Y, Xue W, Zhang Y, Gao Y, Wang Y. A novel disulfidptosis-related immune checkpoint genes signature: forecasting the prognosis of hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:12843-12854. [PMID: 37462769 PMCID: PMC10587022 DOI: 10.1007/s00432-023-05076-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/29/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND HCC is an extremely malignant tumor with a very poor prognosis. In 2023, a brand-new kind of cell death known as disulfidptosis was identified. Although, the prognosis as well as expression of immune checkpoints that are closely connected with it in HCC remain unknown. METHODS In this work, we identified 49 genes with abnormal expression in liver cancer and normal liver tissue, with 23 of them being differentially expressed genes. To create a signature, we classified all HCC cases into three subtypes and used the TCGA database to evaluate each relevant gene's prognostic value for survival. RESULTS Five gene signatures were identified using the LASSO Cox regression approach, while those diagnosed with HCC were split into either low- or high-risk groups. Patients having low-risk HCC showed a much greater likelihood of surviving than those with high risk (p < 0.05). Through immune cell infiltration analysis, it was found that immune-related genes were abundant in high-risk groups and had reduced immune status. CONCLUSION In conclusion, immune checkpoint genes highly associated with disulfidptosis contribute to tumor immunity and can be used to evaluate HCC prognosis. When it comes to predicting overall survival (OS) time in HCC, risk score has been set to be a separate predictor. Through immune cell infiltration analysis, it was found that immune-related genes were abundant in high-risk groups and had reduced immune status. It is possible to measure the prognosis of HCC based on immune checkpoints genes strongly linked to disulfidptosis.
Collapse
Affiliation(s)
- Yuxin Chen
- School of Life Science, Bengbu Medical College, Bengbu, Anhui, China
| | - Wanying Xue
- School of Life Science, Bengbu Medical College, Bengbu, Anhui, China
| | - Yuting Zhang
- School of Life Science, Bengbu Medical College, Bengbu, Anhui, China
| | - Yu Gao
- School of Life Science, Bengbu Medical College, Bengbu, Anhui, China
| | - Yuanyuan Wang
- School of Life Science, Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
3
|
Loukinov D, Anderson AL, Mkrtichyan M, Ghochikyan A, Rivero-Hinojosa S, Tucker J, Lobanenkov V, Agadjanyan MG, Nelson EL. A Therapeutic Vaccine Targeting Rat BORIS (CTCFL) for the Treatment of Rat Breast Cancer Tumors. Int J Mol Sci 2023; 24:5976. [PMID: 36983050 PMCID: PMC10058450 DOI: 10.3390/ijms24065976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Cancer testis antigens are ideal for tumor immunotherapy due to their testis-restricted expression. We previously showed that an immunotherapeutic vaccine targeting the germ cell-specific transcription factor BORIS (CTCFL) was highly effective in treating aggressive breast cancer in the 4T1 mouse model. Here, we further tested the therapeutic efficacy of BORIS in a rat 13762 breast cancer model. We generated a recombinant VEE-VRP (Venezuelan Equine Encephalitis-derived replicon particle) vector-expressing modified rat BORIS lacking a DNA-binding domain (VRP-mBORIS). Rats were inoculated with the 13762 cells, immunized with VRP-mBORIS 48 h later, and then, subsequently, boosted at 10-day intervals. The Kaplan-Meier method was used for survival analysis. Cured rats were re-challenged with the same 13762 cells. We demonstrated that BORIS was expressed in a small population of the 13762 cells, called cancer stem cells. Treatment of rats with VRP-BORIS suppressed tumor growth leading to its complete disappearance in up to 50% of the rats and significantly improved their survival. This improvement was associated with the induction of BORIS-specific cellular immune responses measured by T-helper cell proliferation and INFγ secretion. The re-challenging of cured rats with the same 13762 cells indicated that the immune response prevented tumor growth. Thus, a therapeutic vaccine against rat BORIS showed high efficacy in treating the rat 13762 carcinoma. These data suggest that targeting BORIS can lead to the elimination of mammary tumors and cure animals even though BORIS expression is detected only in cancer stem cells.
Collapse
Affiliation(s)
- Dmitri Loukinov
- Molecular Pathology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Amanda Laust Anderson
- Center for Immunology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92868, USA
| | | | | | | | - Jo Tucker
- Center for Immunology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92868, USA
| | - Victor Lobanenkov
- Molecular Pathology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | | | - Edward L. Nelson
- Center for Immunology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92868, USA
| |
Collapse
|
4
|
Radiosensitization-Related Cuproptosis LncRNA Signature in Non-Small Cell Lung Cancer. Genes (Basel) 2022; 13:genes13112080. [PMID: 36360316 PMCID: PMC9690519 DOI: 10.3390/genes13112080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 11/12/2022] Open
Abstract
A new treatment modality targeting cuproptosis is gradually entering the public horizon. Cuproptosis is a new form of regulated cell death distinct from ferroptosis, apoptosis, autophagy, and necrosis. Previous studies have discovered that the copper level varies considerably in various cancers and that an increase in copper content is directly associated with the proliferation and metastasis of cancer cells. In non-small cell lung cancer (NSCLC) after radiation, the potential utility of cuproptosis-related long noncoding RNAs (lncRNAs) is still unclear. This research aimed to develop a prediction signature based on lncRNAs associated with cuproptosis to predict the prognosis of NSCLC patients following radiation. Methods: Expression data of primary tumors and adjacent solid tissues were downloaded from The Cancer Genome Atlas (TCGA) database, along with the corresponding clinical and mutational data. Univariate and multivariate COX analyses and LASSO regression analyses were performed to obtain a predictive signature of lncRNAs associated with cuproptosis. The data were randomly grouped into a training group used for model construction and a test group used for model validation. The model was validated by drawing a survival curve, risk curve, independent prognostic analysis, ROC curve PFS analysis, etc. Results: The lncRNA signature consisting of six cuproptosis-related lncRNAs (AC104088.1, PPP4R3B-DT, AC006042.3, LUCAT1, HHLA3-AS1, and LINC02029) was used to predict the prognosis of patients. Among them, there were three high-risk lncRNAs (LUCAT1, HHLA3-AS1, and LINC02029) with HR > 1 and three protective lncRNAs (AC104088.1, PPP4R3B-DT, and AC006042.3), with an HR < 1. Data analysis demonstrated that the cuproptosis-related lncRNA signatures could well predict the prognosis of NSCLC patients after radiation. Patients in the high-risk category receive a worse prognosis than those in the low-risk group. Cuproptosis-related risk prediction demonstrated better predictive qualities than age, gender, and pathological stage factors. Conclusion: The risk proposed model can independently predict the prognosis of NSCLC patients after radiotherapy, provide a foundation for the role of cuproptosis-related lncRNAs in NSCLC after radiotherapy, and provide a clinical strategy for radiotherapy combined with cuproptosis in NSCLC patients.
Collapse
|
5
|
Wang J, Wang Z, Jia W, Gong W, Dong B, Wang Z, Zhou M, Tian C. The role of costimulatory molecules in glioma biology and immune microenvironment. Front Genet 2022; 13:1024922. [PMID: 36437961 PMCID: PMC9682268 DOI: 10.3389/fgene.2022.1024922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/28/2022] [Indexed: 10/15/2023] Open
Abstract
Background: Extensive research showed costimulatory molecules regulate tumor progression. Nevertheless, a small amount of literature has concentrated on the potential prognostic and therapeutic effects of costimulatory molecules in patients with glioma. Methods: The data were downloaded from The Cancer Genome Atlas (TCGA) database, Chinese Glioma Genome Atlas (CGGA) database, and Gene Expression Omnibus (GEO) database for bioinformatics analysis. R software was applied for statistical analysis. Using the FigureYa and Xiantao online tools (https://www.xiantao.love/) for mapping. Results: The Least absolute shrinkage and selection operator (LASSO) and Cox regression analysis were utilized to identify the signature consisting of five costimulatory molecules. Multivariate regression analysis revealed that the prognosis of glioma could be independently predicted by the riskscore. Furthermore, we explored clinical and genomic feature differences between the two groups. The level of tumor mutational burden (TMB) was higher in the high-risk group, while more mutation of IDH1 was observed in the low-risk group. Results of Tumor Immune Dysfunction and Exclusion (TIDE) analysis showed that high-risk patients were more prone to be responded to immunotherapy. In addition, subclass mapping analysis was performed to validate our findings and the results revealed that a significantly higher percentage of immunotherapy response rate was observed in the high-risk group. Conclusion: A novel signature with a good independent predictive capacity of prognosis was successfully identified. And our findings reveal that patients with high-risk scores were more likely to be responded to immunotherapy.
Collapse
Affiliation(s)
- Ji Wang
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, Institute of Neurology, China Three Gorges University, Yichang, China
| | - Zi Wang
- Department of Emergency, The First People’s Hospital of Yichang, The People’s Hospital of China Three Gorges University, Yichang, China
| | - Wenxue Jia
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, Institute of Neurology, China Three Gorges University, Yichang, China
| | - Wei Gong
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, Institute of Neurology, China Three Gorges University, Yichang, China
| | - Bokai Dong
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, Institute of Neurology, China Three Gorges University, Yichang, China
| | - Zhuangzhuang Wang
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, Institute of Neurology, China Three Gorges University, Yichang, China
| | - Meng Zhou
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, Institute of Neurology, China Three Gorges University, Yichang, China
| | - Chunlei Tian
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, Institute of Neurology, China Three Gorges University, Yichang, China
| |
Collapse
|
6
|
Han M, Sun Y, Zhao W, Xiang G, Wang X, Jiang Z, Xue Z, Zhou W. Comprehensive characterization of TNFSF14/LIGHT with implications in prognosis and immunotherapy of human gliomas. Front Immunol 2022; 13:1025286. [PMID: 36341396 PMCID: PMC9632349 DOI: 10.3389/fimmu.2022.1025286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/26/2022] [Indexed: 07/02/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a common central neural system malignant tumor among adults. Alongside its microscopic spread, immunosuppression in the tumor microenvironment also induces its refractoriness, which makes immunotherapy for GBM particularly important. Unfortunately, traditional immune checkpoint inhibitors (ICIs) often show limited therapeutic effects in GBM clinical trials, and new therapeutic strategies or targets are urgently needed. TNFSF14/LIGHT is a novel immune checkpoint molecule that plays essential roles in both innate and acquired immunity. Despite recent advances in our understanding of the function of TNFSF14/LIGHT in a variety of cancer types, the clinical and immunological importance of TNFSF14/LIGHT in human gliomas has not been fully explained. Here, we employed a comprehensive in silico analysis with publicly available data to analyze the molecular and immune characteristics of TNFSF14/LIGHT to explore its feasibility as an immunotherapy target. Totally, 2215 glioma cases were enrolled in the current study. Immunohistochemistry staining based on patient tissues (n = 34) was performed for the validation. TNFSF14/LIGHT was expressed higher in higher-WHO-grade gliomas and mesenchymal subtypes, and it was sensitive as a prognostic marker in GBM and low-grade glioma (LGG). A nomogram prognostic model was established based on TNFSF14/LIGHT expression together with other risk factors. Additionally, Gene Ontology and pathway analysis revealed that TNFSF14/LIGHT participated in T-cell activities and inflammatory processes. Moreover, analysis based on the structure and interactions of TNFSF14/LIGHT revealed its mutation sites in tumors as well as crucial interacting proteins. Analysis of IMvigor210 indicated the role of TNFSF14/LIGHT in immunotherapy. Altogether, our results reveal an underlying role of TNFSF14/LIGHT as an immunotherapy target in GBM.
Collapse
Affiliation(s)
- Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanfei Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Wenbo Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Guo Xiang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Xu Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Zheng Jiang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Wei Zhou
- Department of Radiation Oncology, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
7
|
Wang Q, Fang Q, Huang Y, Zhou J, Liu M. Identification of a novel prognostic signature for HCC and analysis of costimulatory molecule-related lncRNA AC099850.3. Sci Rep 2022; 12:9954. [PMID: 35705628 PMCID: PMC9200812 DOI: 10.1038/s41598-022-13792-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 05/27/2022] [Indexed: 12/04/2022] Open
Abstract
Costimulatory molecules are involved in initiation of anti-tumor immune responses while long non‐coding RNAs (lncRNAs) regulate the development of various cancers. However, the roles of lncRNA in hepatocellular carcinoma (HCC) have not been fully established. In this study, we aimed at identifying lncRNAs-related costimulatory molecules in HCC and to construct a prognostic signature for predicting the clinical outcomes for HCC patients. Data were downloaded from The Cancer Genome Atlas database for bioinformatics analyses. Costimulatory molecules were obtained from published literature. The R software, SPSS, and GraphPad Prism were used for statistical analyses. A risk model that is based on five costimulatory molecule-related lncRNAs was constructed using lasso and Cox regression analyses. Multivariate regression analysis revealed that the risk score could predict the prognostic outcomes for HCC. Samples in high- and low-risk groups exhibited significant differences in gene set enrichment and immune infiltration levels. Through colony formation and CCK8 assays, we found that AC099850.3 was strongly associated with HCC cell proliferation. We identified and validated a novel costimulatory molecule-related survival model. In addition, AC099850.3 was found to be closely associated with clinical stages and proliferation of HCC cells, making it a potential target for HCC treatment.
Collapse
Affiliation(s)
- Qi Wang
- Department of Histology and Embryology, Anhui Medical College, Hefei, 230601, Anhui, China
| | - Qiong Fang
- Department of Histology and Embryology, Anhui Medical College, Hefei, 230601, Anhui, China
| | - Yanping Huang
- Department of Histology and Embryology, Anhui Medical College, Hefei, 230601, Anhui, China
| | - Jin Zhou
- Department of Histology and Embryology, Anhui Medical College, Hefei, 230601, Anhui, China
| | - Meimei Liu
- Department of Histology and Embryology, Anhui Medical College, Hefei, 230601, Anhui, China.
| |
Collapse
|
8
|
Aye L, Song X, Yang J, Hu L, Sun X, Zhou J, Liu Q, Yu H, Wang D. Identification of a Costimulatory Molecule Gene Signature to Predict Survival and Immunotherapy Response in Head and Neck Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:695533. [PMID: 34434928 PMCID: PMC8381651 DOI: 10.3389/fcell.2021.695533] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/19/2021] [Indexed: 01/04/2023] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignancies worldwide. Checkpoint blockade immunotherapy has made tremendous progress in the treatment of a variety of cancers in recent years. Costimulatory molecules constitute the foundation of cancer immunotherapies and are deemed to be promising targets for cancer treatment. This study attempted to evaluate the potential value of costimulatory molecule genes (CMGs) in HNSCC. Materials and Methods Based on The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) dataset, we identified the prognostic value of CMGs in HNSCC. Subsequently, CMGs-based signature (CMS) to predict overall survival of HNSCC patients was established and validated. The differences of downstream pathways, clinical outcomes, immune cell infiltration, and predictive immunotherapy responses between different CMS subgroups were investigated via bioinformatic algorithms. We also explored the biological functions of TNFRSF12A, one risk factor of CMS, by in vitro experiments. Results Among CMGs, 22 genes were related to prognosis based on clinical survival time in HNSCC. Nine prognosis-related CMGs were selected to establish CMS. CMS was an independent risk factor and could indicate the survival of HNSCC patients, the component of tumor-infiltrating lymphocytes, and the immunotherapy response rate. Functional enrichment analysis confirmed that CMS might involve immune-relevant processes. Additionally, TNFRSF12A was related to poor prognosis and enhanced malignant phenotype of HNSCC. Conclusion Collectively, CMS could accurately indicate prognosis, evaluate the tumor immune microenvironment, and predict possible immunotherapy outcomes for HNSCC patients.
Collapse
Affiliation(s)
- Ling Aye
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xiaole Song
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Jingyi Yang
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Li Hu
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xicai Sun
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Jiaying Zhou
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Quan Liu
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Hongmeng Yu
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Research Units of New Technologies of Endoscopic Surgery in Skull Base Tumor, Chinese Academy of Medical Sciences, Shanghai, China
| | - Dehui Wang
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Li R, Berglund A, Zemp L, Dhillon J, Putney R, Kim Y, Jain RK, Grass GD, Conejo-Garcia J, Mulé JJ. The 12-CK Score: Global Measurement of Tertiary Lymphoid Structures. Front Immunol 2021; 12:694079. [PMID: 34267760 PMCID: PMC8276102 DOI: 10.3389/fimmu.2021.694079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
There is emerging evidence that the adaptive anti-tumor activity may be orchestrated by secondary lymphoid organ-like aggregates residing in the tumor microenvironment. Known as tertiary lymphoid structures, these lymphoid aggregates serve as key outposts for lymphocyte recruitment, priming and activation. They have been linked to favorable outcomes in many tumor types, and more recently, have been shown to be effective predictors of response to immune checkpoint blockade. We have previously described a 12-chemokine (12-CK) transcriptional score which recapitulates an overwhelming enrichment for immune-related and inflammation-related genes in colorectal carcinoma. Subsequently, the 12-CK score was found to prognosticate favorable survival in multiple tumors types including melanoma, breast cancer, and bladder cancer. In the current study, we summarize the discovery and validation of the 12-CK score in various tumor types, its relationship to TLSs found within the tumor microenvironment, and explore its potential role as both a prognostic and predictive marker in the treatment of various cancers.
Collapse
Affiliation(s)
- Roger Li
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States.,Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Anders Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Logan Zemp
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Jasreman Dhillon
- Department of Pathology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Ryan Putney
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Youngchul Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Rohit K Jain
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - G Daniel Grass
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - José Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - James J Mulé
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
10
|
Woodham AW, Zeigler SH, Zeyang EL, Kolifrath SC, Cheloha RW, Rashidian M, Chaparro RJ, Seidel RD, Garforth SJ, Dearling JL, Mesyngier M, Duddempudi PK, Packard AB, Almo SC, Ploegh HL. In vivo detection of antigen-specific CD8 + T cells by immuno-positron emission tomography. Nat Methods 2020; 17:1025-1032. [PMID: 32929269 PMCID: PMC7541633 DOI: 10.1038/s41592-020-0934-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/23/2020] [Indexed: 12/27/2022]
Abstract
The immune system's ability to recognize peptides on major histocompatibility molecules contributes to the eradication of cancers and pathogens. Tracking these responses in vivo could help evaluate the efficacy of immune interventions and improve mechanistic understanding of immune responses. For this purpose, we employ synTacs, which are dimeric major histocompatibility molecule scaffolds of defined composition. SynTacs, when labeled with positron-emitting isotopes, can noninvasively image antigen-specific CD8+ T cells in vivo. Using radiolabeled synTacs loaded with the appropriate peptides, we imaged human papillomavirus-specific CD8+ T cells by positron emission tomography in mice bearing human papillomavirus-positive tumors, as well as influenza A virus-specific CD8+ T cells in the lungs of influenza A virus-infected mice. It is thus possible to visualize antigen-specific CD8+ T-cell populations in vivo, which may serve prognostic and diagnostic roles.
Collapse
Affiliation(s)
- Andrew W Woodham
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stad H Zeigler
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ella L Zeyang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stephen C Kolifrath
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Scott J Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jason L Dearling
- Division of Nuclear Medicine, Department of Radiology, Children's Hospital Boston, Boston, MA, USA
| | - Maia Mesyngier
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Alan B Packard
- Nuclear Medicine and Molecular Imaging, Boston Children's Hospital/Harvard Medical School, Boston, MA, USA
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
The impact of TNFSF14 on prognosis and immune microenvironment in clear cell renal cell carcinoma. Genes Genomics 2020; 42:1055-1066. [PMID: 32725578 DOI: 10.1007/s13258-020-00974-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND TNFSF14 has been proven to play an important role in various types of tumors. However, its function in renal cell carcinoma (RCC) has not yet been fully elucidated. OBJECTIVE In order to explore molecular mechanism of RCC, we evaluated the effect of TNFSF14 on RCC progression, prognosis and immune microenvironment. METHODS Using TCGA database, the differential expression of TNFSF14 and its relationships between clinicopathological features and prognosis were determined. Cox univariate and multivariate analyses were successively performed to identify whether TNFSF14 was an independent prognostic factor. The discriminating ability of TNFSF14 in RCC prognosis analysis was validated under the same clinical subgroups. Tumor mutational burden (TMB) of each RCC samples was calculated and the differential expression of TNFSF14 between high- and low-TMB groups was analyzed. The immune abundances of 22 leukocyte subtypes in each RCC samples were presented through the CIBERSORT algorithm. TIMER database was used to explore the relationships between copy number of TNFSF14 and the infiltration levels of 6 immune cells. RESULTS Overexpression of TNFSF14 implied adverse clinicopathological features and poor prognosis. Meanwhile, TNFSF14 was identified as an independent prognostic factor (HR = 1.047, P = 0.028) and possessed prevalent applicability in RCC prognostic analysis. TNFSF14 was upregulated in high-TMB group than that in low-TMB group (Log2FC = 0.722). Moreover, overexpression of TNFSF14 brought alteration of immune abundance of 8 leukocyte subtypes. Besides, somatic copy number alteration (SCNA) of TNFSF14 was associated with infiltration levels of 6 immune cells. CONCLUSIONS TNFSF14 has crucial impact on progression, prognosis and immune microenvironment in RCC. Besides, TNFSF14 may be a potential biomarker for predicting the efficacy and response rate of RCC immunotherapy.
Collapse
|
12
|
Skeate JG, Otsmaa ME, Prins R, Fernandez DJ, Da Silva DM, Kast WM. TNFSF14: LIGHTing the Way for Effective Cancer Immunotherapy. Front Immunol 2020; 11:922. [PMID: 32499782 PMCID: PMC7243824 DOI: 10.3389/fimmu.2020.00922] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
Tumor necrosis factor superfamily member 14 (LIGHT) has been in pre-clinical development for over a decade and shows promise as a modality of enhancing treatment approaches in the field of cancer immunotherapy. To date, LIGHT has been used to combat cancer in multiple tumor models where it can be combined with other immunotherapy modalities to clear established solid tumors as well as treat metastatic events. When LIGHT molecules are delivered to or expressed within tumors they cause significant changes in the tumor microenvironment that are primarily driven through vascular normalization and generation of tertiary lymphoid structures. These changes can synergize with methods that induce or support anti-tumor immune responses, such as checkpoint inhibitors and/or tumor vaccines, to greatly improve immunotherapeutic strategies against cancer. While investigators have utilized multiple vectors to LIGHT-up tumor tissues, there are still improvements needed and components to be found within a human tumor microenvironment that may impede translational efforts. This review addresses the current state of this field.
Collapse
Affiliation(s)
- Joseph G Skeate
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Mikk E Otsmaa
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ruben Prins
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Daniel J Fernandez
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Diane M Da Silva
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - W Martin Kast
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
13
|
Increased LIGHT expression and activation of non-canonical NF-κB are observed in gastric lesions of MyD88-deficient mice upon Helicobacter felis infection. Sci Rep 2019; 9:7030. [PMID: 31065023 PMCID: PMC6504916 DOI: 10.1038/s41598-019-43417-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori infection induces a number of pro-inflammatory signaling pathways contributing to gastric inflammation and carcinogenesis. Among those, NF-κB signaling plays a pivotal role during infection and malignant transformation of the gastric epithelium. However, deficiency of the adaptor molecule myeloid differentiation primary response 88 (MyD88), which signals through NF-κB, led to an accelerated development of gastric pathology upon H. felis infection, but the mechanisms leading to this phenotype remained elusive. Non-canonical NF-κB signaling was shown to aggravate H. pylori-induced gastric inflammation via activation of the lymphotoxin β receptor (LTβR). In the present study, we explored whether the exacerbated pathology observed in MyD88-deficient (Myd88−/−) mice was associated with aberrant activation of non-canonical NF-κB. Our results indicate that, in the absence of MyD88, H. felis infection enhances the activation of non-canonical NF-κB that is associated with increase in Cxcl9 and Icam1 gene expression and CD3+ lymphocyte recruitment. In addition, activation of signal transducer and activator of transcription 3 (STAT3) signaling was higher in Myd88−/− compared to wild type (WT) mice, indicating a link between MyD88 deficiency and STAT3 activation in response to H. felis infection. Thereby, MyD88 deficiency results in accelerated and aggravated gastric pathology induced by Helicobacter through activation of non-canonical NF-κB.
Collapse
|
14
|
Abstract
Costimulatory signals initiated by the interaction between the tumor necrosis factor (TNF) ligand and cognate TNF receptor (TNFR) superfamilies promote clonal expansion, differentiation, and survival of antigen-primed CD4+ and CD8+ T cells and have a pivotal role in T-cell-mediated adaptive immunity and diseases. Accumulating evidence in recent years indicates that costimulatory signals via the subset of the TNFR superfamily molecules, OX40 (TNFRSF4), 4-1BB (TNFRSF9), CD27, DR3 (TNFRSF25), CD30 (TNFRSF8), GITR (TNFRSF18), TNFR2 (TNFRSF1B), and HVEM (TNFRSF14), which are constitutive or inducible on T cells, play important roles in protective immunity, inflammatory and autoimmune diseases, and tumor immunotherapy. In this chapter, we will summarize the findings of recent studies on these TNFR family of co-signaling molecules regarding their function at various stages of the T-cell response in the context of infection, inflammation, and cancer. We will also discuss how these TNFR co-signals are critical for immune regulation and have therapeutic potential for the treatment of T-cell-mediated diseases.
Collapse
|
15
|
Maracle CX, Jeucken KCM, Helder B, van Gulik TM, Steins A, van Laarhoven HWM, Tas SW. Silencing NIK potentiates anti-VEGF therapy in a novel 3D model of colorectal cancer angiogenesis. Oncotarget 2018; 9:28445-28455. [PMID: 29983872 PMCID: PMC6033358 DOI: 10.18632/oncotarget.25442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/25/2018] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis is essential for colorectal cancer (CRC) progression, as demonstrated by the beneficial clinical effects of therapeutics inhibiting VEGF signaling. However, alternative mechanisms of neovascularization can develop, resulting in treatment failure. Previously we demonstrated NF-κB-inducing kinase (NIK) contributes to pathological angiogenesis. Here, we investigate NIK as a therapeutic target in endothelial cells (EC) in CRC. To determine NIK expression levels in CRC tissues, we immunostained both primary colorectal tumors and tumors metastasized to the liver. Additionally, a 3D tumor-stromal cell interaction model was developed including EC, fibroblasts and CRC cells to study tumor angiogenesis. This model tested efficacy of NIK-targeting siRNA (siNIK) in EC alone or in combination with the anti-VEGF antibody, bevacizumab. Both primary CRC and liver metastases contained blood vessels expressing NIK. In patients receiving chemotherapy plus bevacizumab, immature NIK+ vessels (p < 0.05) were increased as compared to chemotherapy alone. Activation of NIK by lymphotoxin-beta receptor (LTβR) induced increases in pro-angiogenic mediators, including interleukin (IL)-6, IL-8, chemokine (C-X-C motif) ligand (CXCL)1 and CXCL5 in EC and fibroblasts, accompanied by sprouting in the 3D model, which was blocked by siNIK in EC. Treatment with bevacizumab plus siNIK in EC resulted in a synergistic effect and reduced VEGF and bFGF-induced sprouting (p < 0.05). Here, we demonstrate a role for NIK in CRC-associated angiogenesis. Targeting NIK in EC in combination with anti-VEGF antibody bevacizumab may hold therapeutic potential to increase efficiency in blocking tumor neovascularization, either to prevent treatment failure due to activation of accessory pathways such as NF-κB signaling or as a rescue treatment.
Collapse
Affiliation(s)
- Chrissta X Maracle
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Kim C M Jeucken
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Boy Helder
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Thomas M van Gulik
- Department of Surgery, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Anne Steins
- Department of Medical Oncology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Woodham AW, Cheloha RW, Ling J, Rashidian M, Kolifrath SC, Mesyngier M, Duarte JN, Bader JM, Skeate JG, Da Silva DM, Kast WM, Ploegh HL. Nanobody-Antigen Conjugates Elicit HPV-Specific Antitumor Immune Responses. Cancer Immunol Res 2018; 6:870-880. [PMID: 29792298 DOI: 10.1158/2326-6066.cir-17-0661] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/19/2018] [Accepted: 05/16/2018] [Indexed: 12/29/2022]
Abstract
High-risk human papillomavirus-associated cancers express viral oncoproteins (e.g., E6 and E7) that induce and maintain the malignant phenotype. The viral origin of these proteins makes them attractive targets for development of a therapeutic vaccine. Camelid-derived single-domain antibody fragments (nanobodies or VHHs) that recognize cell surface proteins on antigen-presenting cells (APC) can serve as targeted delivery vehicles for antigens attached to them. Such VHHs were shown to induce CD4+ and CD8+ T-cell responses against model antigens conjugated to them via sortase, but antitumor responses had not yet been investigated. Here, we tested the ability of an anti-CD11b VHH (VHHCD11b) to target APCs and serve as the basis for a therapeutic vaccine to induce CD8+ T-cell responses against HPV+ tumors. Mice immunized with VHHCD11b conjugated to an H-2Db-restricted immunodominant E7 epitope (E749-57) had more E7-specific CD8+ T cells compared with those immunized with E749-57 peptide alone. These CD8+ T cells acted prophylactically and conferred protection against a subsequent challenge with HPV E7-expressing tumor cells. In a therapeutic setting, VHHCD11b-E749-57 vaccination resulted in greater numbers of CD8+ tumor-infiltrating lymphocytes compared with mice receiving E749-57 peptide alone in HPV+ tumor-bearing mice, as measured by in vivo noninvasive VHH-based immune-positron emission tomography (immunoPET), which correlated with tumor regression and survival outcome. Together, these results demonstrate that VHHs can serve as a therapeutic cancer vaccine platform for HPV-induced cancers. Cancer Immunol Res; 6(7); 870-80. ©2018 AACR.
Collapse
Affiliation(s)
- Andrew W Woodham
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts. .,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Jingjing Ling
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Mohammad Rashidian
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Stephen C Kolifrath
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Maia Mesyngier
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Joao N Duarte
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | - Justin M Bader
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Joseph G Skeate
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California
| | - Diane M Da Silva
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - W Martin Kast
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.,Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, California
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.
| |
Collapse
|
17
|
Ward-Kavanagh LK, Lin WW, Šedý JR, Ware CF. The TNF Receptor Superfamily in Co-stimulating and Co-inhibitory Responses. Immunity 2017; 44:1005-19. [PMID: 27192566 DOI: 10.1016/j.immuni.2016.04.019] [Citation(s) in RCA: 301] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Indexed: 02/08/2023]
Abstract
Cytokines related to tumor necrosis factor (TNF) provide a communication network essential for coordinating multiple cell types into an effective host defense system against pathogens and malignant cells. The pathways controlled by the TNF superfamily differentiate both innate and adaptive immune cells and modulate stromal cells into microenvironments conducive to host defenses. Members of the TNF receptor superfamily activate diverse cellular functions from the production of type 1 interferons to the modulation of survival of antigen-activated T cells. Here, we focus attention on the subset of TNF superfamily receptors encoded in the immune response locus in chromosomal region 1p36. Recent studies have revealed that these receptors use diverse mechanisms to either co-stimulate or restrict immune responses. Translation of the fundamental mechanisms of TNF superfamily is leading to the design of therapeutics that can alter pathogenic processes in several autoimmune diseases or promote immunity to tumors.
Collapse
Affiliation(s)
- Lindsay K Ward-Kavanagh
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Wai Wai Lin
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John R Šedý
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Carl F Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
18
|
García-Hernández MDLL, Uribe-Uribe NO, Espinosa-González R, Kast WM, Khader SA, Rangel-Moreno J. A Unique Cellular and Molecular Microenvironment Is Present in Tertiary Lymphoid Organs of Patients with Spontaneous Prostate Cancer Regression. Front Immunol 2017; 8:563. [PMID: 28567040 PMCID: PMC5434117 DOI: 10.3389/fimmu.2017.00563] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 04/27/2017] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Multiple solid cancers contain tertiary lymphoid organs (TLO). However, it is unclear whether they promote tumor rejection, facilitate tumor evasion, or simply whether they are a byproduct of chronic inflammation. We hypothesize that although chronic inflammation induces TLO formation, the tumor milieu can modulate TLO organization and functions in prostate cancer. Therefore, our study seeks to elucidate the cellular and molecular signatures in unique prostatectomy specimens from evanescent carcinoma patients to identify markers of cancer regression, which could be harnessed to modulate local immunosuppression or potentially enhance TLO function. METHODS We used multicolor immunofluorescence to stain prostate tissues, collected at different stages of cancer progression (prostatic intraepithelial neoplasia, intermediate and advanced cancer) or from patients with evanescent prostate carcinoma. Tissues were stained with antibodies specific for pro-inflammatory molecules (cyclooxygenase 2, CXCL10, IL17), tumor-infiltrating immune cells (mature DC-LAMP+ dendritic cells, CD3+ T cells, CD3+Foxp3+ regulatory T cells (Treg), T bet+ Th1 cells, granzyme B+ cytotoxic cells), and stromal cell populations (lymphatic vessels, tumor neovessels, high endothelial venules (HEV), stromal cells), which promote prostate tumor growth or are critical components of tumor-associated TLO. RESULTS Generally, inflammatory cells are located at the margins of tumors. Unexpectedly, we found TLO within prostate tumors from patients at different stages of cancer and in unique samples from patients with spontaneous cancer remission. In evanescent prostate carcinomas, accumulation of Treg was compromised, while Tbet+ T cells and CD8 T cells were abundant in tumor-associated TLO. In addition, we found a global decrease in tumor neovascularization and the coverage by cells positive for cyclooxygenase 2 (COX2). Finally, consistent with tumor regression, prostate stem cell antigen was considerably reduced in TLO and tumor areas from evanescent carcinoma patients. CONCLUSION Collectively, our results suggest that COX2 and Treg are attractive therapeutic targets that can be harnessed to enhance TLO-driven tumor immunity against prostate cancer. Specially, the presence of HEV and lymphatics indicate that TLO can be used as a platform for delivery of cell-based and/or COX2 blocking therapies to improve control of tumor growth in prostate cancer.
Collapse
Affiliation(s)
| | - Norma Ofelia Uribe-Uribe
- Department of Anatomy and Anatomical Pathology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Ricardo Espinosa-González
- Department of Anatomy and Anatomical Pathology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - W. Martin Kast
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Urology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University in Saint Louis, St. Louis, MO, USA
| | | |
Collapse
|
19
|
Abstract
As calculated by the meta-analysis of Korn et al., the prognosis of metastatic melanoma in the pretarget and immunological therapy era was poor, with a median survival of 6.2 and a 1-year life expectancy of 25.5%. Nowadays, significant advances in melanoma treatment have been gained, and immunotherapy is one of the promising approaches to get to durable responses and survival improvement. The aim of the present review is to highlight the recent innovations in melanoma immunotherapy and to propose a critical perspective of the future directions of this enthralling oncology subspecialty.
Collapse
Affiliation(s)
- Sara Valpione
- Christie Hospital NHS Foundation Trust, 550 Wilmslow Rd, Manchester, M20 4BX, UK
| | - Luca G Campana
- Department of Surgery, Oncology & Gastroenterology, University of Padova, via Gattamelata 64, 35128 Padova, Italy
| |
Collapse
|
20
|
Kroeger DR, Milne K, Nelson BH. Tumor-Infiltrating Plasma Cells Are Associated with Tertiary Lymphoid Structures, Cytolytic T-Cell Responses, and Superior Prognosis in Ovarian Cancer. Clin Cancer Res 2016; 22:3005-15. [PMID: 26763251 DOI: 10.1158/1078-0432.ccr-15-2762] [Citation(s) in RCA: 399] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/07/2016] [Indexed: 01/15/2023]
Abstract
PURPOSE CD8(+) tumor-infiltrating lymphocytes (TIL) are key mediators of antitumor immunity and are strongly associated with survival in virtually all solid tumors. However, the prognostic effect of CD8(+) TIL is markedly higher in the presence of CD20(+) B cells, suggesting that cooperative interactions between these lymphocyte subsets lead to more potent antitumor immunity. EXPERIMENTAL DESIGN We assessed the colocalization patterns, phenotypes, and gene expression profiles of tumor-associated T- and B-lineage cells in high-grade serous ovarian cancer (HGSC) by multicolor IHC, flow cytometry, and bioinformatic analysis of gene expression data from The Cancer Genome Atlas. RESULTS T cells and B cells colocalized in four types of lymphoid aggregate, ranging from small, diffuse clusters to large, well-organized tertiary lymphoid structures (TLS) resembling activated lymph nodes. TLS were frequently surrounded by dense infiltrates of plasma cells (PC), which comprised up to 90% of tumor stroma. PCs expressed mature, oligoclonal IgG transcripts, indicative of antigen-specific responses. PCs were associated with the highest levels of CD8(+), CD4(+), and CD20(+) TIL, as well as numerous cytotoxicity-related gene products. CD8(+) TIL carried prognostic benefit only in the presence of PCs and these other TIL subsets. PCs were independent of mutation load, BRCA1/2 status, and differentiation antigens but positively associated with cancer-testis antigens. CONCLUSIONS PCs are associated with the most robust, prognostically favorable CD8(+) TIL responses in HGSC. We propose that TLS facilitate coordinated antitumor responses involving the combined actions of cytolytic T cells and antibody-producing PCs. Clin Cancer Res; 22(12); 3005-15. ©2016 AACR.
Collapse
Affiliation(s)
- David R Kroeger
- Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | - Katy Milne
- Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | - Brad H Nelson
- Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada. Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada. Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
21
|
Johansson-Percival A, Li ZJ, Lakhiani D, He B, Wang X, Hamzah J, Ganss R. Intratumoral LIGHT Restores Pericyte Contractile Properties and Vessel Integrity. Cell Rep 2015; 13:2687-98. [DOI: 10.1016/j.celrep.2015.12.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/12/2015] [Accepted: 11/19/2015] [Indexed: 12/11/2022] Open
|
22
|
Abstract
The inflammatory status of the tumor microenvironment (TME) has been heavily investigated in recent years. Chemokine- and cytokine-signaling pathways such as CCR7, CXCR5, lymphotoxin, and IL-36, which are involved in the generation of secondary lymphoid organs and effector immune responses, are now recognized as having value both as prognostic factors and as immunomodulatory therapeutics in the context of cancer. Furthermore, when produced in the TME, these mediators have been shown to promote the recruitment of immune cells, including T cells, B cells, dendritic cells (DCs), and other specialized immune cell subsets such as follicular DCs and T follicular helper cells, in association with the formation of "tertiary" lymphoid structures (TLSs) within or adjacent to sites of disease. Although TLSs are composed of a heterogeneous collection of immune cell types, whose composition differs based on cancer subtype, the qualitative presence of TLSs has been shown to represent a biomarker of good prognosis for cancer patients. A comprehensive understanding of the role each of these pathways plays within the TME may support the rational design of future immunotherapies to selectively promote/bolster TLS formation and function, leading to improved clinical outcomes across the vast range of solid cancer types.
Collapse
|
23
|
Yan L, Da Silva DM, Verma B, Gray A, Brand HE, Skeate JG, Porras TB, Kanodia S, Kast WM. Forced LIGHT expression in prostate tumors overcomes Treg mediated immunosuppression and synergizes with a prostate tumor therapeutic vaccine by recruiting effector T lymphocytes. Prostate 2015; 75:280-91. [PMID: 25399517 PMCID: PMC4306455 DOI: 10.1002/pros.22914] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/05/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND LIGHT, a ligand for lymphotoxin-β receptor (LTβR) and herpes virus entry mediator, is predominantly expressed on activated immune cells and LTβR signaling leads to the recruitment of lymphocytes. The interaction between LIGHT and LTβR has been previously shown to activate immune cells and result in tumor regression in a virally-induced tumor model, but the role of LIGHT in tumor immunosuppression or in a prostate cancer setting, where self antigens exist, has not been explored. We hypothesized that forced expression of LIGHT in prostate tumors would shift the pattern of immune cell infiltration toward an anti-tumoral milieu, would inhibit T regulatory cells (Tregs) and would induce prostate cancer tumor associated antigen (TAA) specific T cells that would eradicate tumors. METHODS Real Time PCR was used to evaluate expression of forced LIGHT and other immunoregulatory genes in prostate tumors samples. For in vivo studies, adenovirus encoding murine LIGHT was injected intratumorally into TRAMP-C2 prostate cancer cell tumor bearing mice. Chemokine and cytokine concentrations were determined by multiplex ELISA. Flow cytometry was used to phenotype tumor infiltrating lymphocytes and expression of LIGHT on the tumor cell surface. Tumor-specific lymphocytes were quantified via ELISpot assay. Treg induction and Treg suppression assays determined Treg functionality after LIGHT treatment. RESULTS LIGHT in combination with a therapeutic vaccine, PSCA TriVax, reduced tumor burden. LIGHT expression peaked within 48 hr of infection, recruited effector T cells that recognized mouse prostate stem cell antigen (PSCA) into the tumor microenvironment, and inhibited infiltration of Tregs. Tregs isolated from tumor draining lymph nodes had impaired suppressive capability after LIGHT treatment. CONCLUSION Forced LIGHT treatment combined with PSCA TriVax therapeutic vaccination delays prostate cancer progression in mice by recruiting effector T lymphocytes to the tumor and inhibiting Treg mediated immunosuppression. Prostate 75:280-291, 2015. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lisa Yan
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, California, United States of America
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Diane M. Da Silva
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
- Department of Obstetrics & Gynecology, University of Southern California, Los Angeles, California, United States of America
| | - Bhavna Verma
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Andrew Gray
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Heike E. Brand
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Joseph G. Skeate
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, California, United States of America
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Tania B. Porras
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, California, United States of America
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Shreya Kanodia
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
- Samuel Oschin Comprehensive Cancer Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - W. Martin Kast
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, California, United States of America
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
- Department of Obstetrics & Gynecology, University of Southern California, Los Angeles, California, United States of America
- Department of Urology, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
24
|
Abstract
T cells are a crucial component of the immune response to infection and cancer. In addition to coordinating immunity in lymphoid tissue, T cells play a vital role at the disease site, which relies on their efficient and specific trafficking capabilities. The process of T-cell trafficking is highly dynamic, involving a series of distinct processes, which include rolling, adhesion, extravasation, and chemotaxis. Trafficking of T cells to the tumor microenvironment is critical for the success of cancer immunotherapies such as adoptive cellular transfer. Although this approach has achieved some remarkable responses in patients with advanced melanoma and hematologic malignancy, the success against solid cancers has been more moderate. One of the major challenges for adoptive immunotherapy is to be able to effectively target a higher frequency of T cells to the tumor microenvironment, overcoming hurdles associated with immunosuppression and aberrant vasculature. This review summarizes recent advances in our understanding of T-cell migration in solid cancer and immunotherapy based on the adoptive transfer of natural or genetically engineered tumor-specific T cells and discusses new strategies that may enhance the trafficking of these cells, leading to effective eradication of solid cancer and metastases.
Collapse
Affiliation(s)
- Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Parkville, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia.
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Parkville, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia. Department of Immunology, Monash University, Clayton, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Parkville, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia. Department of Immunology, Monash University, Clayton, Australia.
| |
Collapse
|
25
|
Chen L, Fabian KL, Taylor JL, Storkus WJ. Therapeutic use of dendritic cells to promote the extranodal priming of anti-tumor immunity. Front Immunol 2013; 4:388. [PMID: 24348473 PMCID: PMC3843121 DOI: 10.3389/fimmu.2013.00388] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/05/2013] [Indexed: 12/17/2022] Open
Abstract
Ectopic lymphoid tissue, also known as tertiary lymphoid organs (TLO) develop adaptively within sites of chronic tissue inflammation, thereby allowing the host to efficiently crossprime specific immune effector cells within sites of disease. Recent evidence suggests that the presence of TLO in the tumor microenvironment (TME) predicts better overall survival. We will discuss the relevance of extranodal T cell priming within the TME as a means to effectively promote anti-tumor immunity and the strategic use of dendritic cell (DC)-based therapies to reinforce this clinically preferred process in the cancer-bearing host.
Collapse
Affiliation(s)
- Lu Chen
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA ; Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA ; University of Pittsburgh Cancer Institute , Pittsburgh, PA , USA
| |
Collapse
|
26
|
Qin JZ, Upadhyay V, Prabhakar B, Maker AV. Shedding LIGHT (TNFSF14) on the tumor microenvironment of colorectal cancer liver metastases. J Transl Med 2013; 11:70. [PMID: 23514280 PMCID: PMC3623860 DOI: 10.1186/1479-5876-11-70] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 03/12/2013] [Indexed: 12/15/2022] Open
Abstract
Background T-cell infiltration in primary colon tumors is associated with improved patient survival. Preliminary data supports a similar association in colorectal liver metastases (CRLM), and we previously identified increased CRLM expression of the immunostimulatory cytokine LIGHT (TNFSF14) to be related to improved patient prognosis. Therefore, mechanisms to augment the T-cell response in CRLM may be a promising treatment modality, however, the tumor immune microenvironment and LIGHT expression in CRLM remains to be characterized. Methods Utilizing a syngeneic and immunocompetent model of CRLM, the immune microenvironment was characterized for lymphocyte phenotype, function, and location utilizing flow cytometry, immunoassays, and immunofluorescence microscopy. Results CD3+ and CD4+ lymphocytes were decreased, and CD8+ cells were increased in CRLM compared to control liver. When present, greater populations of tumor infiltrating lymphocytes (TIL) were found peritumoral than intratumoral. The TIL expressed significantly higher levels of CD69 and CD107a, but lower levels of LIGHT. Cytokine expression profiles revealed increased levels of the T-helper 1 (Th1) cytokines IFN gamma, IL-12, IL-1b, and IL-8 in CRLM compared to control liver tissue. There was no difference in T-helper 2 (Th2) cytokines between the groups. Conclusions Characterization of the tumor microenvironment of CRLM revealed that although a limited number of activated T-cells infiltrate the tumor and initiate an immune response, the number of LIGHT + T cells infiltrating the tumor were very low. Techniques to decrease suppressive influences or augment the cytotoxic T-cell response are needed and may be possible through mechanisms that can increase intratumoral TIL LIGHT expression.
Collapse
Affiliation(s)
- Jian Zhong Qin
- Department of Surgery, University of Illinois at Chicago, Division of Surgical Oncology, 835 S, Wolcott MC790, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
27
|
A novel series of conferences tackling the hurdles confronting the translation of novel cancer immunotherapies. J Transl Med 2012; 10:218. [PMID: 23127127 PMCID: PMC3551774 DOI: 10.1186/1479-5876-10-218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 10/23/2012] [Indexed: 01/22/2023] Open
Abstract
While there has been significant progress in advancing novel immune therapies to the bedside, much more needs to be done to fully tap into the potential of the immune system. It has become increasingly clear that besides practical and operational challenges, the heterogeneity of cancer and the limited efficacy profile of current immunotherapy platforms are the two main hurdles. Nevertheless, the promising clinical data of several approaches point to a roadmap that carries the promise to significantly advance cancer immunotherapy. A new annual series sponsored by Arrowhead Publishers and Conferences aims at bringing together scientific and business leadership from academia and industry, to identify, share and discuss most current priorities in research and translation of novel immune interventions. This Editorial provides highlights of the first event held earlier this year and outlines the focus of the second meeting to be held in 2013 that will be dedicated to stem cells and immunotherapy.
Collapse
|
28
|
Joh J, Jenson AB, Proctor M, Ingle A, Silva KA, Potter CS, Sundberg JP, Ghim SJ. Molecular diagnosis of a laboratory mouse papillomavirus (MusPV). Exp Mol Pathol 2012; 93:416-21. [PMID: 22796029 DOI: 10.1016/j.yexmp.2012.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 07/03/2012] [Indexed: 11/29/2022]
Abstract
MusPV, a novel papillomavirus (PV) that naturally infects laboratory mice, was isolated and characterized from a colony of NMRI-Foxn1(nu)/Foxn1(nu) (nude) mice in India. Because MusPV may have been missed during routine pathogen screening of mice in colonies worldwide, a variety of detection methods are described to detect MusPV. The clinical and histologic lesions of productive MusPV infections fit PV-associated features, including papillomas, koilocytes within the stratum granulosum of the hyperplastic/acanthotic papillomatous epithelium, and the presence of intranuclear virus particles in koilocytotic cells visualized by electron microscopy. Antiserum against disrupted PV virions, isolated from another species (canine), identified conserved viral antigens in productively infected cells by immunohistochemistry. A rolling circle technique was used to amplify viral circular DNAs followed by endonuclease restriction enzyme digestion to determine the correct size of PV DNA. Consensus PV degenerative primers, My09/11, commonly used to detect many different types of PVs by polymerase chain reaction (PCR), particularly mucosotropic HPVs, also identified MusPV and all rodent PVs tested. Since there was one nucleotide mismatch between the My09/11 primer set and the MusPV template, a new primer set, MusPV-My09/11, was designed to specifically detect MusPV in latent infections and spontaneous MusPV-induced papillomas. Southern blot analysis verified the presence of full size PV DNA in infected tissues. Virus-like particles (VLPs), generated from MusPV L1 genes, provided a substrate for serological testing of naturally and experimentally infected mice. In summary, a series of diagnostic assays were developed and validated to detect MusPV infection in skin tumors and serological response in laboratory mice.
Collapse
Affiliation(s)
- Joongho Joh
- Department of Medicine, James Graham Brown Cancer Center (JGBCC), USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Jenkins M, Chiriva-Internati M, Mirandola L, Tonroy C, Tedjarati SS, Davis N, D'Cunha N, Tijani L, Hardwick F, Nguyen D, Kast WM, Cobos E. Perspective for prophylaxis and treatment of cervical cancer: an immunological approach. Int Rev Immunol 2012; 31:3-21. [PMID: 22251005 DOI: 10.3109/08830185.2011.637254] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
As the second most common cause of cancer-related death in women, human papilloma virus (HPV) vaccines have been a major step in decreasing the morbidity and mortality associated with cervical cancer. An estimated 490,000 women are diagnosed with cervical cancer each year. Increasing knowledge of the HPV role in the etiology of cervical cancer has led to the development and introduction of HPV-based vaccines for active immunotherapy of cervical cancer. Immunotherapies directed at preventing HPV-persistent infections. These vaccines are already accessible for prophylaxis and in the near future, they will be available for the treatment of preexisting HPV-related neoplastic lesions.
Collapse
Affiliation(s)
- Marjorie Jenkins
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, Texas 79430, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bergot AS, Kassianos A, Frazer IH, Mittal D. New Approaches to Immunotherapy for HPV Associated Cancers. Cancers (Basel) 2011; 3:3461-95. [PMID: 24212964 PMCID: PMC3759206 DOI: 10.3390/cancers3033461] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 08/26/2011] [Accepted: 08/29/2011] [Indexed: 02/08/2023] Open
Abstract
Cervical cancer is the second most common cancer of women worldwide and is the first cancer shown to be entirely induced by a virus, the human papillomavirus (HPV, major oncogenic genotypes HPV-16 and -18). Two recently developed prophylactic cervical cancer vaccines, using virus-like particles (VLP) technology, have the potential to prevent a large proportion of cervical cancer associated with HPV infection and to ensure long-term protection. However, prophylactic HPV vaccines do not have therapeutic effects against pre-existing HPV infections and do not prevent their progression to HPV-associated malignancy. In animal models, therapeutic vaccines for persisting HPV infection can eliminate transplantable tumors expressing HPV antigens, but are of limited efficacy in inducing rejection of skin grafts expressing the same antigens. In humans, clinical trials have reported successful immunotherapy of HPV lesions, providing hope and further interest. This review discusses possible new approaches to immunotherapy for HPV associated cancer, based on recent advances in our knowledge of the immunobiology of HPV infection, of epithelial immunology and of immunoregulation, with a brief overview on previous and current HPV vaccine clinical trials.
Collapse
Affiliation(s)
- Anne-Sophie Bergot
- Author to whom correspondence should be addressed; E-Mails: (A.-S.B); (D.M.); Tel.: +61 (07) 3176 2769; Fax: +61 7 3176 5946
| | | | | | - Deepak Mittal
- Author to whom correspondence should be addressed; E-Mails: (A.-S.B); (D.M.); Tel.: +61 (07) 3176 2769; Fax: +61 7 3176 5946
| |
Collapse
|
31
|
Park JJ, Anand S, Zhao Y, Matsumura Y, Sakoda Y, Kuramasu A, Strome SE, Chen L, Tamada K. Expression of anti-HVEM single-chain antibody on tumor cells induces tumor-specific immunity with long-term memory. Cancer Immunol Immunother 2011; 61:203-214. [PMID: 21877247 DOI: 10.1007/s00262-011-1101-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 08/16/2011] [Indexed: 01/01/2023]
Abstract
Genetic engineering of tumor cells to express immune-stimulatory molecules, including cytokines and co-stimulatory ligands, is a promising approach to generate highly efficient cancer vaccines. The co-signaling molecule, LIGHT, is particularly well suited for use in vaccine development as it delivers a potent co-stimulatory signal through the Herpes virus entry mediator (HVEM) receptor on T cells and facilitates tumor-specific T cell immunity. However, because LIGHT binds two additional receptors, lymphotoxin β receptor and Decoy receptor 3, there are significant concerns that tumor-associated LIGHT results in both unexpected adverse events and interference with the ability of the vaccine to enhance antitumor immunity. In order to overcome these problems, we generated tumor cells expressing the single-chain variable fragment (scFv) of anti-HVEM agonistic mAb on the cell surface. Tumor cells expressing anti-HVEM scFv induce a potent proliferation and cytokine production of co-cultured T cells. Inoculation of anti-HVEM scFv-expressing tumor results in a spontaneous tumor regression in CD4+ and CD8+ T cell-dependent fashion, associated with the induction of tumor-specific long-term memory. Stimulation of HVEM and 4-1BB co-stimulatory signals by anti-HVEM scFv-expressing tumor vaccine combined with anti-4-1BB mAb shows synergistic effects which achieve regression of pre-established tumor and T cell memory specific to parental tumor. Taken in concert, our data suggest that genetic engineering of tumor cells to selectively potentiate the HVEM signaling pathway is a promising antitumor vaccine therapy.
Collapse
Affiliation(s)
- Jang-June Park
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA
| | - Sudarshan Anand
- Department of Pathology and Moores UCSD Cancer Center, University of California, San Diego, CA, USA
| | - Yuming Zhao
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA
| | - Yumiko Matsumura
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA
| | - Yukimi Sakoda
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA
| | - Atsuo Kuramasu
- Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Scott E Strome
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA.,Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lieping Chen
- Department of Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Koji Tamada
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA. .,Yamaguchi University Graduate School of Medicine, Ube, Japan. .,Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
32
|
LTβR signaling in dendritic cells induces a type I IFN response that is required for optimal clonal expansion of CD8+ T cells. Proc Natl Acad Sci U S A 2011; 108:2046-51. [PMID: 21245292 DOI: 10.1073/pnas.1014188108] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
During an immune response, antigen-bearing dendritic cells (DCs) migrate to the local draining lymph node and present antigen to CD4(+) helper T cells. Antigen-activated CD4(+) T cells then up-regulate TNF superfamily members including CD40 ligand and lymphotoxin (LT)αβ. Although it is well-accepted that CD40 stimulation on DCs is required for DC licensing and cross-priming of CD8(+) T-cell responses, it is likely that other signals are integrated into a comprehensive DC activation program. Here we show that a cognate interaction between LTαβ on CD4(+) helper T cells and LTβ receptor on DCs results in unique signals that are necessary for optimal CD8(+) T-cell expansion via a type I IFN-dependent mechanism. In contrast, CD40 signaling appears to be more critical for CD8(+) T-cell IFNγ production. Therefore, different TNF family members provide integrative signals that shape the licensing potential of antigen-presenting DCs.
Collapse
|