1
|
Pu L, Zhang X, Pu C, Zhou J, Li J, Wang X, Xi C, Zhang C. Genetic association of tertiary lymphoid structure-related gene signatures with HCC based on Mendelian randomization and machine learning and construction of prognosis model. Int Immunopharmacol 2025; 144:113594. [PMID: 39566392 DOI: 10.1016/j.intimp.2024.113594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/06/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Tertiary lymphoid structures (TLS) are formed in numerous cancer types. However, their value and significance in hepatocellular carcinoma (HCC) is unclear. METHODS We performed differential genes expression analysis of TLS-related Genes (TLSG) based on The Cancer Genome Atlas (TCGA) database, and performed Mendelian randomization (MR) analysis using expression quantitative trait loci, and then took their intersecting genes. A TLSG prognostic signature (TLSGPS)-based risk score was constructed using Least Absolute Shrinkage and Selection Operator (LASSO), univariate and multivariate COX regression analysis, and survival analysis was then performed. We used the International Cancer Genome Consortium for outside validation. We also performed biological function, tumor mutational burden, immune infiltration, single-cell analysis, CeRNA and drug sensitivity analysis based on TLSGPS. RESULTS Three TLSGs (HM13, CSTB, CDCA7L) were identified to construct the TLSGPS, which showed good predictive ability and outperformed most prognostic signatures. MR suggested that HM13 (OR = 0.9997, 95 %CI: 0.9994-0.9999, P = 0.014) and CSTB (OR = 0.9997, 95 %CI: 0.9995-0.9999, P = 0.048) were negatively correlated with the risk of HCC onset, while CDCA7L (OR = 1.0004, 1.0001-1.0007, P = 0.0161) was the opposite. The differences in biological functions between the TLSGPS-based high-risk group (HRG) and low-risk group (LRG) involved cell proliferation, differentiation, and drug metabolism. HRG plus high mutations exhibited extremely poor survival. HRG had higher abundance of immune cell-oncogenic phenotypes, higher immune escape ability, and greater sensitivity to Afatinib, Dasatinib, and Gefitinib. CONCLUSION 3 TLSGs identified by machine learning and MR can predict the onset, prognosis and clinical treatment of HCC patients, and had significant genetic association with HCC.
Collapse
Affiliation(s)
- Lei Pu
- The key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai 200241, PR China.
| | - Xiaoyan Zhang
- The key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai 200241, PR China
| | - Cheng Pu
- School of Martial Arts, Shanghai University of Sport, Shanghai 200438, PR China
| | - Jiacheng Zhou
- Department of Interventional Medicine, Liyang Hospital of Chinese Medicine, Jiangsu 213300, PR China
| | - Jianyue Li
- Department of Oncology, Jiangsu Provincial Hospital of Integrated Chinese and Western Medicine, Jiangsu 210046, PR China
| | - Xiaorong Wang
- Department of Traditional Chinese medicine, Taixing People's Hospital, Jiangsu 225400, PR China
| | - Chenpeng Xi
- School of The First Clinical Medical, Shandong University, Shandong 250100, PR China
| | - Chunyuan Zhang
- The key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai 200241, PR China
| |
Collapse
|
2
|
Duan J, Jiang R, Shen H, Xu X, Sun D. Analysis of nitrogen metabolism-related gene expression in hepatocellular carcinoma to establish relevant indicators for prediction of prognosis and guidance of immunotherapy. Comput Methods Biomech Biomed Engin 2024:1-17. [PMID: 39673385 DOI: 10.1080/10255842.2024.2438922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/14/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND The prognosis of cancers is strongly connected with nitrogen metabolism (NM), which plays a critical role in the microenvironment and growth of tumors. It is unsubstantiated, however, how important NM-related genes are for the prognosis of hepatocellular carcinoma (HCC). METHODS Using publicly available data, we examined potential mechanisms of NM-related genes in HCC, created a predictive model, and assessed immune infiltration and medication sensitivity. RESULTS A prognostic model, which included 12 NM genes (COLQ, GNE, ISCU, MSRA, SARS2, SPHK1, CBS, GOT2, CHST1, EXTL2, GCLM, YARS1), was constructed based on regression analysis. The robustness of the model was validated using multiple methods. The high-risk (HR) and low-risk (LR) groups had varying degrees of immune infiltration, according to an immunology-related study. Of these, B cells and Type_II_IFN_Response were greatly infiltrated in the LR group, whereas aCDs, Macrophages, and Treg were heavily infiltrated in the HR group (p < 0.05). Because of higher immunophenoscore, the low-risk group could benefit from immunotherapy more. Drug sensitivity predictions indicated that people with high CBS expression and low GOT2 and ISCU expression may benefit more from treatment with SCH-772984, Pimasertib, Cobimetinib (isomer1), TAK-733, LY-3214996, ARRY-162, Cladribine, Fludarabine, and Hydroxyurea. CONCLUSION This work created a 12-gene signature based on NM, preliminary investigated immune infiltration in two risk categories, and discovered some possible anti-tumor medications. To sum up, our study findings offer fresh perspectives on the roles played by NM-associated genes in HCC development, prognosis, immunological response, and medication screening.
Collapse
Affiliation(s)
- Jianwen Duan
- Department of Hepatobiliary Surgery, Quzhou Hospital Affiliated of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| | - Renya Jiang
- Department of Hepatobiliary Surgery, Quzhou Hospital Affiliated of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| | - Hongbo Shen
- Department of Hepatobiliary Surgery, Quzhou Hospital Affiliated of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| | - Xiaofang Xu
- Department of Oncology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Da Sun
- Department of Hepatobiliary Surgery, Quzhou Hospital Affiliated of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| |
Collapse
|
3
|
Wang X, Yang T, Shi X. NK cell-based immunotherapy in hepatocellular carcinoma: An attractive therapeutic option for the next decade. Cell Signal 2024; 124:111405. [PMID: 39260532 DOI: 10.1016/j.cellsig.2024.111405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/27/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
Hepatocellular carcinoma (HCC), a major subtype of liver cancer, poses significant therapeutic challenges due to its late diagnosis and rapid progression. The evolving landscape of immunotherapy offers a beacon of hope, with natural killer (NK) cells emerging as pivotal players in combating HCC. NK cells are unique cytotoxic lymphocytes that are essential in the fight against infections and malignancies. Phenotypic and functional NK cell abnormalities have been shown in HCC patients, indicating their significance as a component of the innate immune system against cancer. This review elucidates the critical role of NK cells in combating HCC, focusing on their interaction with the tumor microenvironment, the development of NK cell-based therapies, and the innovative strategies to enhance their efficacy in the immunosuppressive milieu of HCC. The review delves into the various therapeutic strategies, including autologous and allogeneic NK cell therapies, genetic engineering to improve NK cell resilience and targeting, and the integration of NK cells with other immunotherapeutic approaches like checkpoint inhibitors and oncolytic virotherapy. By highlighting recent advancements and the ongoing challenges in the field, this review sets the stage for future research directions that could unlock the full potential of NK cell-based immunotherapy for HCC, offering a beacon of hope for patients battling this formidable cancer.
Collapse
Affiliation(s)
- Xinyi Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu Province 210009, China
| | - Tianye Yang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu Province 210009, China
| | - Xiaoli Shi
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu Province 210029, China; Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
4
|
Liu J, Piranlioglu R, Ye F, Shu K, Lei T, Nakashima H. Immunosuppressive cells in oncolytic virotherapy for glioma: challenges and solutions. Front Cell Infect Microbiol 2023; 13:1141034. [PMID: 37234776 PMCID: PMC10206241 DOI: 10.3389/fcimb.2023.1141034] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Glioblastoma is a highly aggressive form of brain cancer characterized by the abundance of myeloid lineage cells in the tumor microenvironment. Tumor-associated macrophages and microglia (TAM) and myeloid-derived suppressor cells (MDSCs), play a pivotal role in promoting immune suppression and tumor progression. Oncolytic viruses (OVs) are self-amplifying cytotoxic agents that can stimulate local anti-tumor immune responses and have the potential to suppress immunosuppressive myeloid cells and recruit tumor-infiltrating T lymphocytes (TILs) to the tumor site, leading to an adaptive immune response against tumors. However, the impact of OV therapy on the tumor-resident myeloid population and the subsequent immune responses are not yet fully understood. This review provides an overview of how TAM and MDSC respond to different types of OVs, and combination therapeutics that target the myeloid population to promote anti-tumor immune responses in the glioma microenvironment.
Collapse
Affiliation(s)
- Junfeng Liu
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Raziye Piranlioglu
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Fei Ye
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hiroshi Nakashima
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
5
|
Su W, Qiu W, Li SJ, Wang S, Xie J, Yang QC, Xu J, Zhang J, Xu Z, Sun ZJ. A Dual-Responsive STAT3 Inhibitor Nanoprodrug Combined with Oncolytic Virus Elicits Synergistic Antitumor Immune Responses by Igniting Pyroptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209379. [PMID: 36545949 DOI: 10.1002/adma.202209379] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Immune checkpoint blockade (ICB) therapy shows excellent efficacy against malignancies; however, insufficient tumor immunogenicity and the immunosuppressive tumor microenvironment (TME) are considered as the two major stumbling blocks to a broad ICB response. Here, a combinational therapeutic strategy is reported, wherein TME-reactive oxygen species/pH dual-responsive signal transducers and activators of transcription 3 inhibitor nanoprodrugs MPNPs are combined with oncolytic herpes simplex virus 1 virotherapy to synergistically ignite pyroptosis for enhancing immunotherapy. MPNPs exhibit a certain level of tumor accumulation, reduce tumor cell stemness, and enhance antitumor immune responses. Furthermore, the simultaneous application of oncolytic viruses (OVs) confers MPNPs with higher tumor penetration capacity and remarkable gasdermin-E-mediated pyroptosis, thereby reshaping the TME and transforming "cold" tumors into "hot" ones. This "fire of immunity" strategy successfully activates robust T-cell-dependent antitumor responses, potentiating ICB effects against local recurrence and pulmonary metastasis in preclinical "cold" murine triple-negative breast cancer and syngeneic oral cancer models. Collectively, this work may pave a new way and offer an unprecedented opportunity for the combination of OVs with nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Wen Su
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Wei Qiu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Shu-Jin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Shuo Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jun Xie
- State Key Laboratory of Virology, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, P. R. China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Qi-Chao Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jiming Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Junjie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
- State Key Laboratory of Virology, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, P. R. China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| |
Collapse
|
6
|
Huang F, Dai C, Zhang Y, Zhao Y, Wang Y, Ru G. Development of Molecular Mechanisms and Their Application on Oncolytic Newcastle Disease Virus in Cancer Therapy. Front Mol Biosci 2022; 9:889403. [PMID: 35860357 PMCID: PMC9289221 DOI: 10.3389/fmolb.2022.889403] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is caused by the destruction or mutation of cellular genetic materials induced by environmental or genetic factors. It is defined by uncontrolled cell proliferation and abnormality of the apoptotic pathways. The majority of human malignancies are characterized by distant metastasis and dissemination. Currently, the most common means of cancer treatment include surgery, radiotherapy, and chemotherapy, which usually damage healthy cells and cause toxicity in patients. Targeted therapy is an effective tumor treatment method with few side effects. At present, some targeted therapeutic drugs have achieved encouraging results in clinical studies, but finding an effective solution to improve the targeting and delivery efficiency of these drugs remains a challenge. In recent years, oncolytic viruses (OVs) have been used to direct the tumor-targeted therapy or immunotherapy. Newcastle disease virus (NDV) is a solid oncolytic agent capable of directly killing tumor cells and increasing tumor antigen exposure. Simultaneously, NDV can trigger the proliferation of tumor-specific immune cells and thus improve the therapeutic efficacy of NDV in cancer. Based on NDV’s inherent oncolytic activity and the stimulation of antitumor immune responses, the combination of NDV and other tumor therapy approaches can improve the antitumor efficacy while reducing drug toxicity, indicating a broad application potential. We discussed the biological properties of NDV, the antitumor molecular mechanisms of oncolytic NDV, and its application in the field of tumor therapy in this review. Furthermore, we presented new insights into the challenges that NDV will confront and suggestions for increasing NDV’s therapeutic efficacy in cancer.
Collapse
Affiliation(s)
- Fang Huang
- Cancer Center, Department of Pathology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Chuanjing Dai
- Cancer Center, Department of Pathology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, China
| | - Youni Zhang
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, China
- Department of Laboratory Medicine, Tiantai People’s Hospital, Taizhou, China
| | - Yuqi Zhao
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yigang Wang
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, China
- *Correspondence: Yigang Wang, ; Guoqing Ru,
| | - Guoqing Ru
- Cancer Center, Department of Pathology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Yigang Wang, ; Guoqing Ru,
| |
Collapse
|
7
|
Li Q, Oduro PK, Guo R, Li R, Leng L, Kong X, Wang Q, Yang L. Oncolytic Viruses: Immunotherapy Drugs for Gastrointestinal Malignant Tumors. Front Cell Infect Microbiol 2022; 12:921534. [PMID: 35719333 PMCID: PMC9203847 DOI: 10.3389/fcimb.2022.921534] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Oncolytic virus therapy has advanced rapidly in recent years. Natural or transgenic viruses can target tumor cells and inhibit tumor growth and metastasis in various ways without interfering with normal cell and tissue function. Oncolytic viruses have a high level of specificity and are relatively safe. Malignant tumors in the digestive system continue to have a high incidence and mortality rate. Although existing treatment methods have achieved some curative effects, they still require further improvement due to side effects and a lack of specificity. Many studies have shown that oncolytic viruses can kill various tumor cells, including malignant tumors in the digestive system. This review discusses how oncolytic virus therapy improves malignant tumors in the digestive system from the point-of-view of basic and clinical studies. Also, the oncolytic virus anti-tumor mechanisms underpinning the therapeutic potential of oncolytic viruses are expounded. In all, we argue that oncolytic viruses might eventually provide therapeutic solutions to malignant tumors in the digestive system.
Collapse
Affiliation(s)
- Qingbo Li
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Patrick Kwabena Oduro
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine & State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
| | - Rui Guo
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruiqiao Li
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine & State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
| | - Ling Leng
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine & State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
| | - Xianbin Kong
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xianbin Kong, ; Qilong Wang, ; Long Yang,
| | - Qilong Wang
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine & State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
- *Correspondence: Xianbin Kong, ; Qilong Wang, ; Long Yang,
| | - Long Yang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xianbin Kong, ; Qilong Wang, ; Long Yang,
| |
Collapse
|
8
|
Chen L, Niu Y, Sun J, Lin H, Liang G, Xiao M, Shi D, Wang J, Zhu H, Guan Y. Oncolytic Activity of Wild-type Newcastle Disease Virus HK84 Against Hepatocellular Carcinoma Associated with Activation of Type I Interferon Signaling. J Clin Transl Hepatol 2022; 10:284-296. [PMID: 35528990 PMCID: PMC9039698 DOI: 10.14218/jcth.2021.00284] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/18/2021] [Accepted: 10/10/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) is listed as one of the most common causes of cancer-related death. Oncolytic therapy has become a promising treatment because of novel immunotherapies and gene editing technology, but biosafety concerns remain the biggest limitation for clinical application. We studied the the antitumor activity and biosafety of the wild-type Newcastle disease virus HK84 strain (NDV/HK84) and 10 other NDV strains. METHODS Cell proliferation and apoptosis were determined by cell counting Kit-8 and fluorescein isothiocyanate Annexin V apoptosis assays. Colony formation, wound healing, and a xenograft mouse model were used to evaluate in vivo and in vitro oncolytic effectiveness. The safety of NDV/HK84 was tested in nude mice by an in vivo luciferase imaging system. The replication kinetics of NDV/HK84 in normal tissues and tumors were evaluated by infectious-dose assays in eggs. RNA sequencing analysis was performed to explore NDV/HK84 activity and was validated by quantitative real-time PCR. RESULTS The cell counting Kit-8 assays of viability found that the oncolytic activity of the NDV strains differed with the multiplicity of infection (MOI). At an MOI of 20, the oncolytic activity of all NDV strains except the DK/JX/21358/08 strain was >80%. The oncolytic activities of the NDV/HK84 and DK/JX/8224/04 strains were >80% at both MOI=20 and MOI=2. Only NDV/HK84 had >80% oncolytic activities at both MOI=20 and MOI=2. We chose NDV/HK84 as the candidate virus to test the oncolytic effect of NDV in HCC in the in vitro and in vivo experiments. NDV/HK84 killed human SK-HEP-1 HCC cells without affecting healthy cells. CONCLUSIONS Intratumor infection with NDV/HK84 strains compared with vehicle controls or positive controls indicated that NDV/HK84 strain specifically inhibited HCC without affecting healthy mice. High-throughput RNA sequencing showed that the oncolytic activity of NDV/HK84 was dependent on the activation of type I interferon signaling.
Collapse
Affiliation(s)
- Liming Chen
- Department of Oncology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| | - Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jiating Sun
- Department of Oncology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| | - Hong Lin
- Department of Oncology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| | - Guoxi Liang
- Department of Oncology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| | - Min Xiao
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| | - Dongmei Shi
- Department of Oncology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| | - Jia Wang
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| | - Huachen Zhu
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| | - Yi Guan
- International Joint Laboratory for Virology and Emerging Infectious Diseases (Ministry of Education), Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology of STU/HKU, Shantou, Guangdong, China
| |
Collapse
|
9
|
Zhang H, Zhang Y, Dong J, Zuo S, Meng G, Wu J, Wei J. Recombinant adenovirus expressing the fusion protein PD1PVR improves CD8 + T cell-mediated antitumor efficacy with long-term tumor-specific immune surveillance in hepatocellular carcinoma. Cell Oncol (Dordr) 2021; 44:1243-1255. [PMID: 34491549 DOI: 10.1007/s13402-021-00633-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2021] [Indexed: 10/20/2022] Open
Abstract
PURPOSE Treatment-associated upregulation of suppressive checkpoints and a lack of costimulatory signals compromise the antitumor efficacy of oncolytic virus immunotherapy. Therefore, we aimed to identify highly effective therapeutic targets to provide a proof-of-principle for immune checkpoint together with oncolytic virus-mediated viro-immunotherapy for cancer. METHODS A fusion protein containing both the extracellular domain of programmed death-1 (PD-1) and the poliovirus receptor (PVR) was designed. Next, the corresponding expression fragment was inserted into the genome of a replication-competent adenovirus to generate Ad5sPD1PVR. The infection, expression, replication and oncolysis of Ad5sPD1PVR were investigated in hepatocellular carcinoma (HCC) cell lines. Immune activation and the antitumor efficacy of Ad5sPD1PVR were examined in HCC tumor models including a humanized immunocompetent mouse model. RESULTS Ad5sPD1PVR effectively infected and replicated in HCC cells and secreted sPD1PVR. In a H22 ascitic HCC mouse model, intraperitoneal injection of Ad5sPD1PVR markedly recruited lymphocytes and activated antitumor immune responses. Ad5sPD1PVR exerted a profound antitumor effect on ascitic HCC. Furthermore, we found that Ad5sPD1PVR-H expressing sPD1PVR of human origin exhibited potent antitumor effects in a HCC humanized mouse model. We also found that CD8+ T cells mediated the antitumor effects and long-term tumor-specific immune surveillance induced by Ad5sPD1PVR. Finally, when combined with fludarabine, the antitumor efficacy of Ad5sPD1PVR was found to be further improved in the ascitic HCC model. CONCLUSIONS From our data we conclude that the newly designed recombinant Ad5sPD1PVR virus significantly enhances CD8+ T cell-mediated antitumor efficacy with long-term tumor-specific immune surveillance in hepatocellular carcinoma, and that fludarabine is a promising therapeutic partner for Ad5sPD1PVR.
Collapse
Affiliation(s)
- Hailin Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Yonghui Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Jie Dong
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Shuguang Zuo
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Gang Meng
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China
| | - Junhua Wu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China.
- National Institute of Healthcare Data Science At Nanjing University, Nanjing University, Nanjing, 210093, China.
| | - Jiwu Wei
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China.
| |
Collapse
|
10
|
Ma YS, Xin R, Yang XL, Shi Y, Zhang DD, Wang HM, Wang PY, Liu JB, Chu KJ, Fu D. Paving the way for small-molecule drug discovery. Am J Transl Res 2021; 13:853-870. [PMID: 33841626 PMCID: PMC8014367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/18/2020] [Indexed: 06/12/2023]
Abstract
Small-molecule drugs are organic compounds affecting molecular pathways by targeting important proteins, which have a low molecular weight, making them penetrate cells easily. Small-molecule drugs can be developed from leads derived from rational drug design or isolated from natural resources. As commonly used medications, small-molecule drugs can be taken orally, which enter cells to act on intracellular targets. These characteristics make small-molecule drugs promising candidates for drug development, and they are increasingly favored in the pharmaceutical market. Despite the advancements in molecular genetics and effective new processes in drug development, the drugs currently used in clinical practice are inadequate due to their poor efficacy or severe side effects. Therefore, developing new safe and efficient drugs is a top priority for disease control and curing.
Collapse
Affiliation(s)
- Yu-Shui Ma
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and TechnologyChangsha 410004, Hunan, China
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Rui Xin
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Xiao-Li Yang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Yi Shi
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Dan-Dan Zhang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Hui-Min Wang
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Pei-Yao Wang
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| | - Ji-Bin Liu
- Cancer Institute, Nantong Tumor HospitalNantong 226631, China
| | - Kai-Jian Chu
- Department of Biliary Tract Surgery I, Third Affiliated Hospital of Second Military Medical UniversityShanghai 200438, China
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of MedicineShanghai 200072, China
| |
Collapse
|
11
|
Marotel M, Hasim MS, Hagerman A, Ardolino M. The two-faces of NK cells in oncolytic virotherapy. Cytokine Growth Factor Rev 2020; 56:59-68. [PMID: 32586674 DOI: 10.1016/j.cytogfr.2020.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/04/2020] [Indexed: 12/17/2022]
Abstract
Oncolytic viruses (OVs) are immunotherapeutics capable of directly killing cancer cells and with potent immunostimulatory properties. OVs exert their antitumor effect, at least partially, by activating the antitumor immune response, of which NK cells are an important component. However, if on the one hand increasing evidence revealed that NK cells are important mediators of oncolytic virotherapy, on the other hand, NK cells have evolved to fight viral infections, and therefore they can have a detrimental effect for the efficacy of OVs. In this review, we will discuss the dichotomy between the antitumor and antiviral functions of NK cells related to oncolytic virotherapy. We will also review NK cell-based and OV-based therapies, engineered OVs aimed at enhancing immune stimulation, and combination therapies involving OVs and NK cells currently used in cancer immunotherapy.
Collapse
Affiliation(s)
- M Marotel
- Ottawa Hospital Research Institute, Cancer Therapeutics Program, Ottawa, Canada; Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada
| | - M S Hasim
- Ottawa Hospital Research Institute, Cancer Therapeutics Program, Ottawa, Canada; Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada
| | - A Hagerman
- Ottawa Hospital Research Institute, Cancer Therapeutics Program, Ottawa, Canada; Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada; University of Ottawa, Department of Biochemistry, Microbiology and Immunology, Ottawa, Canada
| | - M Ardolino
- Ottawa Hospital Research Institute, Cancer Therapeutics Program, Ottawa, Canada; Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada; University of Ottawa, Department of Biochemistry, Microbiology and Immunology, Ottawa, Canada.
| |
Collapse
|
12
|
Yu S, Wang Y, Hou J, Li W, Wang X, Xiang L, Tan D, Wang W, Jiang L, Claret FX, Jiao M, Guo H. Tumor-infiltrating immune cells in hepatocellular carcinoma: Tregs is correlated with poor overall survival. PLoS One 2020; 15:e0231003. [PMID: 32240238 PMCID: PMC7117689 DOI: 10.1371/journal.pone.0231003] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Systematic interrogation of tumor-infiltrating immune cells (TIICs) is key to the prediction of clinical outcome and development of immunotherapies. However, little is known about the TIICs of hepatocellular carcinoma (HCC) and its impact on the prognosis of patients and potential for immunotherapy. We applied CIBERSORT of 1090 tumors to infer the infiltration of 22 subsets of TIICs using gene expression data. Unsupervised clustering analysis by 22 TIICs revealed 4 clusters of tumors, mainly defined by macrophages and T cells, with distinct prognosis and associations with immune checkpoint molecules, including PD-1, CD274, CTLA-4, LAG-3 and IFNG. We found tumors with decreased number of M1 macrophages or increased regulatory T cells were associated with poor prognosis. Based on the multivariate Cox analysis, a nomogram was also established for clinical application. In conclusion, composition of the TIICs in HCC was quite different, which is an important determinant of prognosis with great potential to identify candidates for immunotherapy.
Collapse
Affiliation(s)
- SiZhe Yu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - Yu Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
- Department of Respirology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - Jia Hou
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - WenYuan Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - Xiao Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - LuoChengLing Xiang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - DeLi Tan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - WenJuan Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - LiLi Jiang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
| | - Francois X. Claret
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Min Jiao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
- * E-mail: (MJ); (HG)
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi, PR China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi’an, Shaanxi, PR China
- * E-mail: (MJ); (HG)
| |
Collapse
|
13
|
Meng G, Li B, Chen A, Zheng M, Xu T, Zhang H, Dong J, Wu J, Yu D, Wei J. Targeting aerobic glycolysis by dichloroacetate improves Newcastle disease virus-mediated viro-immunotherapy in hepatocellular carcinoma. Br J Cancer 2019; 122:111-120. [PMID: 31819179 PMCID: PMC6964686 DOI: 10.1038/s41416-019-0639-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/20/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Oncolytic viro-immunotherapy holds promise for cancer treatment. While immune activation can be robustly triggered by oncolytic viruses, negative feedback is often upregulated in the tumour microenvironment (TME). Lactate accumulation, signal transducer and activator of transcription 3 (STAT3) activation, indoleamine 2,3-dioxygenase 1 (IDO1) expression, and myeloid-derived suppressor cell (MDSC) infiltration coordinate to shape the immunosuppressive TME. METHODS Representative hepatocellular carcinoma (HCC) cell lines and HCC-bearing mice were treated with oncolytic Newcastle disease virus (NDV), alone or in combination with dichloroacetate (DCA, a pyruvate dehydrogenase kinase (PDK) inhibitor). RESULTS We found that infection with oncolytic NDV led to significant induction of the aforementioned suppressive factors. Interestingly, DCA significantly reduced lactate release, STAT3 activation, IDO1 upregulation, and MDSC infiltration in NDV-treated HCC. Consequently, DCA significantly enhanced the antitumour immune responses, leading to improved antitumour efficacy and prolonged survival in mouse models of ascitic and subcutaneous HCC. Furthermore, DCA increased NDV replication in a PDK-1-dependent manner in HCC. CONCLUSIONS Targeting aerobic glycolysis by DCA improves NDV-mediated viro-immunotherapy in HCC by mitigating immune negative feedback and promoting viral replication. These findings provide a rationale for targeting reprogrammed metabolism together with oncolytic virus-mediated viro-immunotherapy for HCC treatment.
Collapse
Affiliation(s)
- Gang Meng
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Binghua Li
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Anxian Chen
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Meihong Zheng
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Tiancheng Xu
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Hailin Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Jie Dong
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Junhua Wu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Decai Yu
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China.
| | - Jiwu Wei
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China.
| |
Collapse
|