151
|
Jia Y, Liu L, Shan B. Future of immune checkpoint inhibitors: focus on tumor immune microenvironment. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1095. [PMID: 33145314 PMCID: PMC7575936 DOI: 10.21037/atm-20-3735] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunotherapy has become a powerful clinical strategy in cancer treatment. Immune checkpoint inhibitors (ICIs) have opened a new era for cancer immunotherapy. Nowadays, the number of immunotherapy drug approvals has increased, with numerous treatment options in clinical and preclinical development. However, there remain some obstacles to improve the efficacy of ICIs further. The tumor immune microenvironment (TIME) consists of cancer cell, immune cells and cytokines, et cetera. The dynamics of TIME determine the efficacies of ICIs. Although the ICIs showed manageable toxicity, immune-related adverse effects (irAEs) are still unignorable for clinicians. Since some primary resistance mechanisms exist in TIME, ICIs can only show effects in individual cancer patients. Even for the patients who responded, acquired resistance will occur to neutralize the effect of ICIs. Understanding how to increase the response rates and overcome the resistance to various classes of ICIs is the key to improving clinical efficacy. Besides the novel ICIs in development, there are some approaches to establish combination therapies are underway to improve further the efficacies of ICIs in treating cancer patients. Here, we describe the complicated TIME and state quo of ICIs to prospect the future of ICIs in cancer treatment.
Collapse
Affiliation(s)
- Yunlong Jia
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Baoen Shan
- Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
152
|
Li M, Li W, Yang X, Wang H, Peng Y, Yin J, Lu Y, Liu L, Shang J, Zhao Q. An immune landscape based prognostic signature predicts the immune status and immunotherapeutic responses of patients with colorectal cancer. Life Sci 2020; 261:118368. [PMID: 32888940 DOI: 10.1016/j.lfs.2020.118368] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 12/24/2022]
Abstract
AIMS Colorectal cancer (CRC) is one of the most common cancers with poor prognosis worldwide. The advent of immunotherapy has greatly improved survival in refractory patients of CRC. In this study, we aimed to identify reliable immune classification and biomarkers that predict immunotherapeutic responses in CRC patients. MATERIALS AND METHODS Based on transcriptome profiles of two publicly available CRC datasets, we performed single-sample gene set enrichment analysis (ssGSEA) to calculate the relative abundance of 29 immune-related items of each sample. Unsupervised clustering was used to classify CRC patients. Furthermore, an immune prognostic signature was constructed using the least absolute shrinkage and selection operator (LASSO) Cox regression analysis. KEY FINDINGS The CRC patients were clustered into high, medium, low immune infiltration subtypes based on the immune landscape. There was significant heterogeneity among the three subtypes. The high immune infiltration group showed higher expression of programmed cell death-ligand 1 and better prognosis than the median and low immune infiltration groups. Furthermore, we constructed a 7-immune-related prognostic gene signature and found that the signature had high predictive value and was superior to other clinicopathological parameters. Finally, the correlation analysis of the signature with immune cell infiltration and immune checkpoint molecules suggested that the signature had the potential to assess the immunotherapeutic responses of CRC patients. SIGNIFICANCE The immune landscape and prognostic signature of CRC contribute to a deeper understanding of the tumor microenvironment and guide accurate immunotherapy.
Collapse
Affiliation(s)
- Mengting Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Wenjie Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Xueying Yang
- Department of Medical Records, The Central Hospital of Enshi Autonomous Prefecture, Enshi, China
| | - Haizhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Yanan Peng
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jinwen Yin
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Yuanyuan Lu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jian Shang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China.
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China.
| |
Collapse
|
153
|
Che Y, Luo Z, Zhang C, Sun N, Gao S, He J. Immune signature of tumor-infiltrating immune cells predicts the prognosis and therapeutic effects in squamous cell carcinoma. Int Immunopharmacol 2020; 87:106802. [PMID: 32745903 DOI: 10.1016/j.intimp.2020.106802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/23/2022]
Abstract
Tumor-infiltrating immune cells (TICs) are involved in tumor progression and determine the prognosis. We investigated how TICs affect the prognosis and therapeutic effects of squamous cell carcinoma (SCC), which share common histological features and certain risk factors. The SCC data from The Cancer Genome Atlas (TCGA) and Gene expression Omnibus (GEO) databases were downloaded to evaluate the composition of TICs with the CIBERSORT algorithm. LASSO and Cox multivariate regression analyses were used to build a prognostic risk model. Chemotherapeutic and immunotherapeutic responses were compared between patients with SCC. A Gene set variation analysis (GSVA) was also performed to elucidate the mechanism. Naïve B cells and resting mast cells were selected to construct the prognostic model. According to these two immune cell subtypes, patients with SCC were divided into low- and high-risk groups. The low-risk group with high proportions of naïve B cells and resting mast cells had a better overall survival rate than the high-risk group and might benefit from immunotherapy and chemotherapy due to differences in the immune microenvironment. Activation of the Wnt signaling pathway was observed in the high-risk group. Based on the findings from the present study, the immune signature provides prognostic determinants of SCC and may be a biomarker to guide chemotherapy and immunotherapy. Wnt inhibitors may be attractive candidates for combination treatment in high-risk patients with SCC.
Collapse
Affiliation(s)
- Yun Che
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhiwen Luo
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
154
|
Zhong XF, Sun X. Nanomedicines based on nanoscale metal-organic frameworks for cancer immunotherapy. Acta Pharmacol Sin 2020; 41:928-935. [PMID: 32355277 PMCID: PMC7468577 DOI: 10.1038/s41401-020-0414-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer immunotherapy, with an aim to enhance host immune responses, has been recognized as a promising therapeutic treatment for cancer. A diversity of immunomodulatory agents, including tumor-associated antigens, adjuvants, cytokines and immunomodulators, has been explored for their ability to induce a cascading adaptive immune response. Nanoscale metal-organic frameworks (nMOFs), a class of crystalline-shaped nanomaterials formed by the self-assembly of organic ligands and metal nodes, are attractive for cancer immunotherapy because they feature tunable pore size, high surface area and loading capacity, and intrinsic biodegradability. In this review we summarize recent progress in the development of nMOFs for cancer immunotherapy, including cancer vaccine delivery and combination of in situ vaccination with immunomodulators to reverse immune suppression. Current challenges and future perspectives for rational design of nMOF-based cancer immunotherapy are also discussed.
Collapse
Affiliation(s)
- Xiao-Fang Zhong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
155
|
Sun B, Hyun H, Li LT, Wang AZ. Harnessing nanomedicine to overcome the immunosuppressive tumor microenvironment. Acta Pharmacol Sin 2020; 41:970-985. [PMID: 32424240 PMCID: PMC7470849 DOI: 10.1038/s41401-020-0424-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy has received extensive attention due to its ability to activate the innate or adaptive immune systems of patients to combat tumors. Despite a few clinical successes, further endeavors are still needed to tackle unresolved issues, including limited response rates, development of resistance, and immune-related toxicities. Accumulating evidence has pinpointed the tumor microenvironment (TME) as one of the major obstacles in cancer immunotherapy due to its detrimental impacts on tumor-infiltrating immune cells. Nanomedicine has been battling with the TME in the past several decades, and the experience obtained could be exploited to improve current paradigms of immunotherapy. Here, we discuss the metabolic features of the TME and its influence on different types of immune cells. The recent progress in nanoenabled cancer immunotherapy has been summarized with a highlight on the modulation of immune cells, tumor stroma, cytokines and enzymes to reverse the immunosuppressive TME.
Collapse
Affiliation(s)
- Bo Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Hyesun Hyun
- Laboratory of Nano and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Lian-Tao Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Andrew Z Wang
- Laboratory of Nano and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
156
|
Liu W, Dai E, Liu Z, Ma C, Guo ZS, Bartlett DL. In Situ Therapeutic Cancer Vaccination with an Oncolytic Virus Expressing Membrane-Tethered IL-2. Mol Ther Oncolytics 2020; 17:350-360. [PMID: 32405533 PMCID: PMC7210382 DOI: 10.1016/j.omto.2020.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
Successful in situ therapeutic vaccination would allow locally delivered oncolytic virus (OV) to exert systemic immunologic effects on metastases and improve survival. We have utilized bilateral flank tumor models to determine the most efficacious regimens of in situ vaccination. Intratumoral injection with membrane-tethered interleukin -2-armed OV (vvDD-mIL2) plus a Toll-like receptor 9 ligand (CpG) yielded systemic immunization and decreased tumor growth in a contralateral, noninjected tumor. Our main aims were to study the tumor immune microenvironment (TME) after vaccination and identify additional immune adjuvants that may improve the systemic tumor-specific immunity. Immunological profiles in the spleen showed an increased CD8+ T cell/regulatory T cell (Treg) ratio and increased CD11c+ cells after dual injection in one flank tumor. Concurrently, there was increased infiltration of tumor necrosis factor alpha (TNF-α)+CD8+ T cells and interferon gamma (IFN-γ)+CD4+ T cells and reduced CTLA-4+PD-1+CD8+ T cells in the contralateral, noninjected tumor. The anti-tumoral activity depended on CD8+ T cells and IFN-γ, but not CD4+ T cells. Based on the negative immune components still existing in the untreated tumors, we investigated additional adjuvants: clodronate liposome-mediated depletion of macrophages plus anti-PD-1 therapy. This regimen dramatically reduced the tumor burden in the noninjected tumor and increased median survival by 87%, suggesting that inhibition/elimination of suppressive components in the tumor microenvironment (TME) can improve therapeutic outcomes. This study emphasizes the importance of immune profiling to design rational, combined immunotherapy regimens ultimately to impact patient survival.
Collapse
Affiliation(s)
- Weilin Liu
- The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Enyong Dai
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zuqiang Liu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Congrong Ma
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Zong Sheng Guo
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - David L. Bartlett
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
157
|
Zhao S, Pan W, Jiang H, Zhang R, Jiang H, Liang Z, Hu H. Cerenkov luminescence imaging is an effective preclinical tool for assessing colorectal cancer PD-L1 levels in vivo. EJNMMI Res 2020; 10:64. [PMID: 32542442 PMCID: PMC7295871 DOI: 10.1186/s13550-020-00654-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/03/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Preclinical and clinical studies have demonstrated that immunotherapy has effectively delayed tumor progression, and the clinical outcomes of anti-PD-1/PD-L1 therapy were related to PD-L1 expression level in the tumors. A 131I-labeled anti-PD-L1 monoclonal antibody tracer, 131I-PD-L1-Mab, was developed to study the target ability of noninvasive Cerenkov luminescence imaging in colorectal cancer xenograft mice. METHOD Anti-PD-L1 monoclonal antibody labeled with 131I (131I-PD-L1-Mab), and in vitro binding assays were used to evaluate the affinity of 131I-PD-L1-Mab to PD-L1 and their binding level to different colorectal cancer cells, and compared with flow cytometry, Western blot analysis, and immunofluorescence staining. The clinical application value of 131I-PD-L1-Mab was evaluated through biodistribution and Cerenkov luminescence imaging, and different tumor-bearing models expressing PD-L1 were evaluated. RESULTS 131I-PD-L1-Mab showed high affinity to PD-L1, and the equilibrium dissociation constant was 1.069 × 10-9 M. The competitive inhibition assay further confirmed the specific binding ability of 131I-PD-L1-Mab. In four different tumor-bearing models with different PD-L1 expression, the biodistribution and Cerenkov luminescence imaging showed that the RKO tumors demonstrated the highest uptake of the tracer 131I-PD-L1-Mab, with a maximum uptake of 1.613 ± 0.738% IA/g at 48 h. CONCLUSIONS There is a great potential for 131I-PD-L1-Mab noninvasive Cerenkov luminescence imaging to assess the status of tumor PD-L1 expression and select patients for anti-PD-L1 targeted therapy.
Collapse
Affiliation(s)
- Sheng Zhao
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Pan
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huijie Jiang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | | | - Hao Jiang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zonghui Liang
- Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, China.
| | - Hongbo Hu
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
158
|
Philippou Y, Sjoberg H, Lamb AD, Camilleri P, Bryant RJ. Harnessing the potential of multimodal radiotherapy in prostate cancer. Nat Rev Urol 2020; 17:321-338. [PMID: 32358562 DOI: 10.1038/s41585-020-0310-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
Radiotherapy in combination with androgen deprivation therapy (ADT) is a standard treatment option for men with localized and locally advanced prostate cancer. However, emerging clinical evidence suggests that radiotherapy can be incorporated into multimodality therapy regimens beyond ADT, in combinations that include chemotherapy, radiosensitizing agents, immunotherapy and surgery for the treatment of men with localized and locally advanced prostate cancer, and those with oligometastatic disease, in whom the low metastatic burden in particular might be treatable with these combinations. This multimodal approach is increasingly recognized as offering considerable clinical benefit, such as increased antitumour effects and improved survival. Thus, radiotherapy is becoming a key component of multimodal therapy for many stages of prostate cancer, particularly oligometastatic disease.
Collapse
Affiliation(s)
- Yiannis Philippou
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Hanna Sjoberg
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Philip Camilleri
- Oxford Department of Clinical Oncology, Churchill Hospital Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Headington, Oxford, UK
| | - Richard J Bryant
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK.
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK.
| |
Collapse
|
159
|
Expression Levels of IL-17A, IL-17F, IL-17RA, and IL-17RC in Prostate Cancer with Taking into Account the Histological Grade according to Gleason Scale in Comparison to Benign Prostatic Hyperplasia: In Search of New Therapeutic Options. J Immunol Res 2020; 2020:4910595. [PMID: 32537467 PMCID: PMC7267874 DOI: 10.1155/2020/4910595] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/11/2020] [Indexed: 01/17/2023] Open
Abstract
Prostate cancer (PCa) is the second most commonly diagnosed malignant tumor and the fifth leading cause of cancer death in men in the world. The most common types of tumors are adenocarcinomas. Prostate cancer is a slow-growing cancer. The incidence increases with age. Evaluation of proinflammatory factors such as IL-17A, IL-17F, IL-17RA, and IL-17RC expression makes it possible to assess the impact of inflammatory process on progression of PCa. The aim of the study was to retrospectively assess the histological material of PCa divided into few groups using the Gleason score. Studies were carried out on archival tissue material in the form of paraffin blocks of 40 men with PCa after radical prostatectomy. The control group was composed of 10 men with benign prostatic hyperplasia (BPH). The material was obtained by the transurethral resection of the prostate (TURP). Immunohistochemistry was performed on prepared material using specific primary antibodies against IL-17A, IL-17F, IL-17RA, and IL-17RC. Expression of the antibody to be examined using light microscopy and the Remmele-Stegner score (IRS) in cancer staining was then evaluated. Expression of IL-17 RA was not shown in a group of patients with PCa and in the control group. In the group of patients with Gleason score 8 and 9 PCa, the expression of IL-17A was higher compared to that of IL-17F. In addition, in PCa with an increased grade of Gleason scale, a decrease in the expression of the study inflammatory parameters was found. The inflammatory process has an impact on PCa. A study on IL-17 may become a starting point for further research on an attempt to use, for example, immunotherapy in PCa.
Collapse
|
160
|
Chaib M, Chauhan SC, Makowski L. Friend or Foe? Recent Strategies to Target Myeloid Cells in Cancer. Front Cell Dev Biol 2020; 8:351. [PMID: 32509781 PMCID: PMC7249856 DOI: 10.3389/fcell.2020.00351] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is a complex network of epithelial and stromal cells, wherein stromal components provide support to tumor cells during all stages of tumorigenesis. Among these stromal cell populations are myeloid cells, which are comprised mainly of tumor-associated macrophages (TAM), dendritic cells (DC), myeloid-derived suppressor cells (MDSC), and tumor-associated neutrophils (TAN). Myeloid cells play a major role in tumor growth through nurturing cancer stem cells by providing growth factors and metabolites, increasing angiogenesis, as well as promoting immune evasion through the creation of an immune-suppressive microenvironment. Immunosuppression in the TME is achieved by preventing critical anti-tumor immune responses by natural killer and T cells within the primary tumor and in metastatic niches. Therapeutic success in targeting myeloid cells in malignancies may prove to be an effective strategy to overcome chemotherapy and immunotherapy limitations. Current therapeutic approaches to target myeloid cells in various cancers include inhibition of their recruitment, alteration of function, or functional re-education to an antitumor phenotype to overcome immunosuppression. In this review, we describe strategies to target TAMs and MDSCs, consisting of single agent therapies, nanoparticle-targeted approaches and combination therapies including chemotherapy and immunotherapy. We also summarize recent molecular targets that are specific to myeloid cell populations in the TME, while providing a critical review of the limitations of current strategies aimed at targeting a single subtype of the myeloid cell compartment. The goal of this review is to provide the reader with an understanding of the critical role of myeloid cells in the TME and current therapeutic approaches including ongoing or recently completed clinical trials.
Collapse
Affiliation(s)
- Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Subhash C Chauhan
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, United States.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Liza Makowski
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Hematology Oncology, Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.,Center for Cancer Research, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
161
|
Qiang W, Dai Y, Sun G, Xing X, Sun X. Development of a prognostic index of colon adenocarcinoma based on immunogenomic landscape analysis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:284. [PMID: 32355728 PMCID: PMC7186653 DOI: 10.21037/atm.2020.03.09] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background Colon adenocarcinoma (COAD) is one of the most commonly diagnosed cancers, and it is closely related to the immune microenvironment. Considering that immunotherapy is not effective for all COAD patients, it is necessary to identify the effective population before administering treatment. In this study, we established an independent prognostic index based on immune-related genes (IRGs), in order to evaluate the clinical outcome of COAD. Methods The gene expression profiles and IRGs taken from The Cancer Genome Atlas (TCGA) and Immunology Database and Analysis Portal (ImmPort), respectively, were integrated in order to identify the differentially expressed IRGs. Functional enrichment analysis was conducted and the prognostic value of survival-related IRGs was determined. Based on Cox regression analysis, the IRG-based prognostic index (IRGPI) was established, and the model was evaluated and applied. Results A total of 51 differentially expressed survival-related IRGs were identified. The most significant signaling pathway was "cytokine-cytokine receptor interaction". The index established herein was based on 12 survival-related IRGs, and it was highly accurate in monitoring prognosis. Moreover, the IRGPI was significantly correlated with multiple clinicopathologic factors, as well as with the infiltration of immune cells. Conclusions An independent IRGPI was established in order to assess the immune status and tumor prognosis in COAD patients. This index can serve as a robust biomarker in clinical prognosis applications, including cancer immunotherapy.
Collapse
Affiliation(s)
- Weijie Qiang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Yifei Dai
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xiaoyan Xing
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| |
Collapse
|
162
|
Jiang W, Zhu D, Wang C, Zhu Y. An immune relevant signature for predicting prognoses and immunotherapeutic responses in patients with muscle-invasive bladder cancer (MIBC). Cancer Med 2020; 9:2774-2790. [PMID: 32096345 PMCID: PMC7163112 DOI: 10.1002/cam4.2942] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/15/2020] [Accepted: 02/12/2020] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are novel treatments that significantly improve the survival time of MIBC patients, but immunotherapeutic responses are different among MIBC patients. Therefore, it is urgent to find predictive biomarkers that can accurately identify MIBC patients who are sensitive to ICIs. In this study, we computed the relative abundances of 24 immune cells based on the expression profiles of MIBC patients using single‐sample gene set enrichment analysis (ssGSEA). Unsupervised clustering analysis of the 24 immune cells was performed to classify MIBC patients into different immune‐infiltrating groups. Genome (gene mutation and copy number variation), transcriptome (mRNA, lncRNA, and miRNA), and functional enrichment were found to be heterogeneous among different immune‐infiltrating groups. We identified 282 differentially expressed genes (DEGs) associated with immune infiltration by comparing the expression profiles of patients with different immune infiltration profiles, and 20 core prognostic DEGs were identified by univariate Cox regression analysis. An immune‐relevant gene signature (TIM signature) consisting of nine key prognostic DEGs (CCDC80, CD3D, CIITA, FN1, GBP4, GNLY, SPINK1, UBD, and VIM) was constructed using least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Receiver operating characteristic (ROC) curves and subgroup analysis confirmed that the TIM signature was an ideal biomarker for predicting the prognosis of MIBC patients. Its value in predicting immunotherapeutic responses was also validated in The Cancer Genome Atlas (TCGA) cohort (AUC = 0.69, 95% CI = 0.63‐0.74) and the IMvigor210 cohort (AUC = 0.64, 95% = 0.55‐0.74). The TIM signature demonstrates a powerful ability to distinguish MIBC patients with different prognoses and immunotherapeutic responses, but more prospective studies are needed to assess its reliability in the future.
Collapse
Affiliation(s)
- Wen Jiang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dandan Zhu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenghe Wang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
163
|
Sobierajska K, Ciszewski WM, Sacewicz-Hofman I, Niewiarowska J. Endothelial Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1234:71-86. [PMID: 32040856 DOI: 10.1007/978-3-030-37184-5_6] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Angiogenesis is a critical process required for tumor progression. Newly formed blood vessels provide nutrition and oxygen to the tumor contributing to its growth and development. However, endothelium also plays other functions that promote tumor metastasis. It is involved in intravasation, which allows invasive cancer cells to translocate into the blood vessel lumen. This phenomenon is an important stage for cancer metastasis. Besides direct association with cancer development, endothelial cells are one of the main sources of cancer-associated fibroblasts (CAFs). The heterogeneous group of CAFs is the main inductor of migration and invasion abilities of cancer cells. Therefore, the endothelium is also indirectly responsible for metastasis. Considering the above, the endothelium is one of the important targets of anticancer therapy. In the chapter, we will present mechanisms regulating endothelial function, dependent on cancer and cancer niche cells. We will focus on possibilities of suppressing pro-metastatic endothelial functions, applied in anti-cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Jolanta Niewiarowska
- Department of Molecular Cell Mechanisms, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
164
|
Gray SG, Mutti L. Immunotherapy for mesothelioma: a critical review of current clinical trials and future perspectives. Transl Lung Cancer Res 2020; 9:S100-S119. [PMID: 32206576 PMCID: PMC7082257 DOI: 10.21037/tlcr.2019.11.23] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/14/2019] [Indexed: 12/25/2022]
Abstract
At the clinical level the role of immunotherapy in cancer is currently at a pivotal point. Therapies such as checkpoint inhibitors are being approved at many levels in cancers such as non-small cell lung cancer (NSCLC). Mesothelioma is a rare orphan disease associated with prior exposure to asbestos, with a dismal prognosis. Various clinical trials for checkpoint inhibitors have been conducted in this rare disease, and suggest that such therapies may play a role as a treatment option for a proportion of patients with this cancer. Most recently approved as a salvage therapy in mesothelioma was granted in Japan, regulatory approval for their use in the clinic elsewhere lags. In this article we review the current pertinent clinical trials of immunotherapies in malignant mesothelioma, discuss the current issues that may affect the clinical outcomes of such therapies and further evaluate potential candidate new avenues that may become future targets for immunotherapy in this cancer.
Collapse
Affiliation(s)
- Steven G. Gray
- Thoracic Oncology Research Group, Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James’s Hospital, Dublin, Ireland
| | - Luciano Mutti
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
165
|
Häfner SJ. Tumour travel tours - Why circulating cancer cells value company. Biomed J 2020; 43:1-7. [PMID: 32200951 PMCID: PMC7090313 DOI: 10.1016/j.bj.2020.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/17/2022] Open
Abstract
Welcome to the New Year and a new issue of the Biomedical Journal, where we learn that travelling with company boosts the metastatic potential of circulating tumour cells, as well as that a worm could be an excellent model to study antidiabetic drugs. In addition, we discover another pair of molecular scissors for genetic engineering, how exactly Leptospira wreaks havoc on its run through the host organism, and that hyperparathyroidism brings its own risks, but does not worsen the outcome of papillary thyroid carcinoma. Furthermore, the importance of taking into account differing beauty ideals for aesthetic surgery surveys is discussed, alongside the question how bad isolated local recurrence is in the case of HR + breast cancer. Finally, we find out that virtual colonoscopy deserves more credit, that the first medical experiment in space was all about the H-reflex, and that it is possible to survive advanced necrotising fasciitis of the face and neck.
Collapse
Affiliation(s)
- Sophia Julia Häfner
- University of Copenhagen, BRIC Biotech Research & Innovation Centre, Anders Lund Group, Copenhagen, Denmark.
| |
Collapse
|
166
|
Franco-Luzón L, González-Murillo Á, Alcántara-Sánchez C, García-García L, Tabasi M, Huertas AL, Chesler L, Ramírez M. Systemic oncolytic adenovirus delivered in mesenchymal carrier cells modulate tumor infiltrating immune cells and tumor microenvironment in mice with neuroblastoma. Oncotarget 2020; 11:347-361. [PMID: 32064039 PMCID: PMC6996901 DOI: 10.18632/oncotarget.27401] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/21/2019] [Indexed: 12/25/2022] Open
Abstract
Celyvir (autologous mesenchymal cells -MSCs- that carry an oncolytic adenovirus) is a new therapeutic strategy for metastatic tumors developed by our research group over the last decade. There are limitations for studying the immune effects of human oncolytic adenoviruses in murine models since these viruses do not replicate naturally in these animals. The use of xenografts in immunodeficient mice prevent assessing important clinical aspects of this therapy such as the antiadenoviral immune response or the possible intratumoral immune changes, both of tumor infiltrating leukocytes and of the microenvironment. In our strategy, the presence of MSCs in the medicinal product adds an extra level of complexity. We present here a murine model that overcomes many of these limitations. We found that carrier cells outcompeted intravenous administration of naked particles in delivering the oncolytic virus into the tumor masses. The protection that MSCs could provide to the oncolytic adenovirus did not preclude the development of an antiadenoviral immune response. However, the presence of circulating antiadenoviral antibodies did not prevent changes detected at the tumor masses: increased infiltration and changes in the quality of immune cells per unit of tumor volume, and a less protumoral and more inflammatory profile of the tumor microenvironment. We believe that the model described here will enable the study of crucial events related to the immune responses affecting both the medicinal product and the tumor.
Collapse
Affiliation(s)
| | - África González-Murillo
- Unidad de Terapias Avanzadas, Oncología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | | | - Lorena García-García
- Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Maryam Tabasi
- Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Ana Luis Huertas
- Servicio de Cirugía, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Louis Chesler
- Paediatric Solid Tumour Biology and Therapeutics Team, Division of Clinical Studies and Cancer Therapeutics Division, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Manuel Ramírez
- Unidad de Terapias Avanzadas, Oncología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| |
Collapse
|
167
|
Zhang W, Huang Q, Xiao W, Zhao Y, Pi J, Xu H, Zhao H, Xu J, Evans CE, Jin H. Advances in Anti-Tumor Treatments Targeting the CD47/SIRPα Axis. Front Immunol 2020; 11:18. [PMID: 32082311 PMCID: PMC7003246 DOI: 10.3389/fimmu.2020.00018] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/07/2020] [Indexed: 12/16/2022] Open
Abstract
CD47 is an immunoglobulin that is overexpressed on the surface of many types of cancer cells. CD47 forms a signaling complex with signal-regulatory protein α (SIRPα), enabling the escape of these cancer cells from macrophage-mediated phagocytosis. In recent years, CD47 has been shown to be highly expressed by various types of solid tumors and to be associated with poor patient prognosis in various types of cancer. A growing number of studies have since demonstrated that inhibiting the CD47-SIRPα signaling pathway promotes the adaptive immune response and enhances the phagocytosis of tumor cells by macrophages. Improved understanding in this field of research could lead to the development of novel and effective anti-tumor treatments that act through the inhibition of CD47 signaling in cancer cells. In this review, we describe the structure and function of CD47, provide an overview of studies that have aimed to inhibit CD47-dependent avoidance of macrophage-mediated phagocytosis by tumor cells, and assess the potential and challenges for targeting the CD47-SIRPα signaling pathway in anti-cancer therapy.
Collapse
Affiliation(s)
- Wenting Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The Scientific Research Center of Dongguan, College of Pharmacy, Institute of Clinical Laboratory Medicine, Guangdong Medical University, Dongguan, China.,Marine Medical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Qinghua Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The Scientific Research Center of Dongguan, College of Pharmacy, Institute of Clinical Laboratory Medicine, Guangdong Medical University, Dongguan, China.,Marine Medical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Weiwei Xiao
- Biosafety Level-3 Laboratory, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yue Zhao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The Scientific Research Center of Dongguan, College of Pharmacy, Institute of Clinical Laboratory Medicine, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- Key Laboratory for Tropical Diseases Control of the Ministry of Education, Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Huan Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The Scientific Research Center of Dongguan, College of Pharmacy, Institute of Clinical Laboratory Medicine, Guangdong Medical University, Dongguan, China
| | - Hongxia Zhao
- School of Biomedical and Pharmaceutical Science, Guangdong University of Technology, Guangzhou, China
| | - Junfa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The Scientific Research Center of Dongguan, College of Pharmacy, Institute of Clinical Laboratory Medicine, Guangdong Medical University, Dongguan, China
| | - Colin E Evans
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The Scientific Research Center of Dongguan, College of Pharmacy, Institute of Clinical Laboratory Medicine, Guangdong Medical University, Dongguan, China.,Marine Medical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| |
Collapse
|
168
|
Abdelaziz MO, Ossmann S, Kaufmann AM, Leitner J, Steinberger P, Willimsky G, Raftery MJ, Schönrich G. Development of a Human Cytomegalovirus (HCMV)-Based Therapeutic Cancer Vaccine Uncovers a Previously Unsuspected Viral Block of MHC Class I Antigen Presentation. Front Immunol 2019; 10:1776. [PMID: 31417555 PMCID: PMC6682651 DOI: 10.3389/fimmu.2019.01776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) induces a uniquely high frequency of virus-specific effector/memory CD8+ T-cells, a phenomenon termed “memory inflation”. Thus, HCMV-based vaccines are particularly interesting in order to stimulate a sustained and strong cellular immune response against cancer. Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with high lethality and inevitable relapse. The current standard treatment does not significantly improve the desperate situation underlining the urgent need to develop novel approaches. Although HCMV is highly fastidious with regard to species and cell type, GBM cell lines are susceptible to HCMV. In order to generate HCMV-based therapeutic vaccine candidates, we deleted all HCMV-encoded proteins (immunoevasins) that interfere with MHC class I presentation. The aim being to use the viral vector as an adjuvant for presentation of endogenous tumor antigens, the presentation of high levels of vector-encoded neoantigens and finally the repurposing of bystander HCMV-specific CD8+ T cells to fight the tumor. As neoantigen, we exemplarily used the E6 and E7 proteins of human papillomavirus type 16 (HPV-16) as a non-transforming fusion protein (E6/E7) that covers all relevant antigenic peptides. Surprisingly, GBM cells infected with E6/E7-expressing HCMV-vectors failed to stimulate E6-specific T cells despite high level expression of E6/E7 protein. Further experiments revealed that MHC class I presentation of E6/E7 is impaired by the HCMV-vector although it lacks all known immunoevasins. We also generated HCMV-based vectors that express E6-derived peptide fused to HCMV proteins. GBM cells infected with these vectors efficiently stimulated E6-specific T cells. Thus, fusion of antigenic sequences to HCMV proteins is required for efficient presentation via MHC class I molecules during infection. Taken together, these results provide the preclinical basis for development of HCMV-based vaccines and also reveal a novel HCMV-encoded block of MHC class I presentation.
Collapse
Affiliation(s)
- Mohammed O Abdelaziz
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sophia Ossmann
- Clinic for Gynecology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Andreas M Kaufmann
- Clinic for Gynecology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Judith Leitner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, Partner Site Berlin, Berlin, Germany
| | - Martin J Raftery
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Günther Schönrich
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
169
|
Tumor Microenvironment as A "Game Changer" in Cancer Radiotherapy. Int J Mol Sci 2019; 20:ijms20133212. [PMID: 31261963 PMCID: PMC6650939 DOI: 10.3390/ijms20133212] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy (RT), besides cancer cells, also affects the tumor microenvironment (TME): tumor blood vessels and cells of the immune system. It damages endothelial cells and causes radiation-induced inflammation. Damaged vessels inhibit the infiltration of CD8+ T lymphocytes into tumors, and immunosuppressive pathways are activated. They lead to the accumulation of radioresistant suppressor cells, including tumor-associated macrophages (TAMs) with the M2 phenotype, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs). The area of tumor hypoxia increases. Hypoxia reduces oxygen-dependent DNA damage and weakens the anti-cancer RT effect. It activates the formation of new blood vessels and leads to cancer relapse after irradiation. Irradiation may also activate the immune response through immunogenic cell death induction. This leads to the "in situ" vaccination effect. In this article, we review how changes in the TME affect radiation-induced anticancer efficacy. There is a very delicate balance between the activation of the immune system and the immunosuppression induced by RT. The effects of RT doses on immune system reactions and also on tumor vascularization remain unclear. A better understanding of these interactions will contribute to the optimization of RT treatment, which may prevent the recurrence of cancer.
Collapse
|
170
|
Davola ME, Vito A, Wei J, El-Sayes N, Workenhe S, Mossman KL. Genetic modification of oncolytic viruses to enhance antitumor immunity. Methods Enzymol 2019; 635:231-250. [PMID: 32122548 DOI: 10.1016/bs.mie.2019.05.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Among the many immunotherapies being developed and tested both preclinically and clinically, oncolytic viruses (OVs) are gaining traction as a forerunner in the search for potent new therapeutic agents, with a genetically engineered herpes simplex virus type 1 (HSV-1) recently approved by the FDA for the treatment of melanoma. The great potential of OVs to fight cancer is driving different approaches to improve OV-based therapy, with genetic modification of OVs to enhance host antitumor immunity being one of the most promising approaches. In this chapter we describe possible modifications in the OV genome that could increase its antitumor activity and immunostimulatory capacity, together with different methods to achieve these goals. Finally, we present different analyses to verify the desired genetic modification and evaluate its impact on host antitumor immunity in preliminary stages.
Collapse
Affiliation(s)
- Maria Eugenia Davola
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Alyssa Vito
- Department of Biochemistry and Biomedical Science, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Jiarun Wei
- Department of Biochemistry and Biomedical Science, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Nader El-Sayes
- Department of Biochemistry and Biomedical Science, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Samuel Workenhe
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Karen Louise Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
171
|
Wang H, Jiang H, Corbet C, de Mey S, Law K, Gevaert T, Feron O, De Ridder M. Piperlongumine increases sensitivity of colorectal cancer cells to radiation: Involvement of ROS production via dual inhibition of glutathione and thioredoxin systems. Cancer Lett 2019; 450:42-52. [PMID: 30790679 DOI: 10.1016/j.canlet.2019.02.034] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 01/19/2023]
Abstract
Piperlongumine (PL), naturally synthesized in long pepper, is known to selectively kill tumor cells via perturbation of reactive oxygen species (ROS) homeostasis. ROS are the primary effector molecules of radiation, and increase of ROS production by pharmacological modulation is known to enhance radioresponse. We therefore investigated the radiosensitizing effect of PL in colorectal cancer cells (CT26 and DLD-1) and CT26 tumor-bearing mice. Firstly, we found that PL induced excessive production of ROS due to depletion of glutathione and inhibition of thioredoxin reductase. Secondly, PL enhanced both the intrinsic and hypoxic radiosensitivity of tumor cells, linked to ROS-mediated increase of DNA damage, G2/M cell cycle arrest, and inhibition of cellular respiration. Finally, the radiosensitizing effect of PL was verified in vivo. PL improved the tumor response to both single and fractionated radiation, resulting in a significant increase of survival rate of tumor-bearing mice, while it was ineffective on its own. In line with in vitro findings, enhanced radioresponse is associated with inhibition of antioxidant systems. In conclusion, our results suggest that PL could be a potential radiosensitizer in colorectal cancer.
Collapse
Affiliation(s)
- Hui Wang
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Heng Jiang
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Sven de Mey
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kalun Law
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thierry Gevaert
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Mark De Ridder
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|