1
|
Hassanin O, Abdallah F, Mohamed MH, Ahmed MS, Al-Rasheed M, Rashad EM. The kinetics of gene expression related to innate and adaptive immunity in the lung and spleen following Newcastle disease virus (NDV) infection in vaccinated broiler chickens employing different vaccination regimes. Vet Microbiol 2025; 305:110525. [PMID: 40262238 DOI: 10.1016/j.vetmic.2025.110525] [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: 12/30/2024] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Newcastle disease virus (NDV) classified as an avian paramyxovirus-1 poses a significant risk to the global poultry sector. This study explored the kinetics of the innate and adaptive immune responses in chickens exposed to virulent Newcastle Disease Virus (vNDV) genotype VII. A challenge experiment was carried out with both non-vaccinated and NDV-vaccinated chickens utilizing different vaccination strategies commonly used in the industry. The four groups of vaccinated birds were administered either two doses of live NDV vaccines, live vaccines boosted with an inactivated vaccine, the rHVT-NDV-IBDV vaccine alone, or the rHVT-NDV-IBDV vaccine in conjunction with a live vaccine booster. To assess seven cytokines linked to antiviral and proinflammatory innate responses quantitative real-time polymerase chain reaction (qRT-PCR) analyses were implemented. The analyses revealed robust innate immune responses in all cytokines measured in lung and spleen tissues of the group that received both live and inactivated vaccines. Notably these increases were found to correlate with the humoral immune response within that same group. Significant transcriptional activity in the lung and spleen tissues of non-vaccinated chickens at 24 hour post-infection (pi). The most significant upregulations were identified in toll-like receptor 7 (TLR7), TLR5, interleukin 6 (IL6), chicken interferon-alpha (chIFN-α), and myxovirus resistance protein 1 (Mx1), which later exhibited a regression. The administration of both live and inactivated vaccines has proven effective in restoring the suppression or inhibition of vNDV infection across a wide range of cytokines, including TLR7, TLR5, chIFN-α, Mx1, IL6, and MHC-1. Various vaccination strategies have been shown to either mitigate or prevent cytokine storms within 24 hour pi in the lungs, spleen, or both, in certain cytokines such as chIFN-α, Mx1, and IL6. Collectively these results suggest that different vaccination strategies modify the kinetics and pathophysiological responses associated with vNDV infection in chickens.
Collapse
Affiliation(s)
- Ola Hassanin
- Department of Avian and Rabbit Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| | - Fatma Abdallah
- Department of Virology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Mahmoud Ha Mohamed
- Department of Avian and Rabbit Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt; Department of Clinical Studies, Collage of Veterinary Medicine, King Faisal University, 31982, Saudi Arabia
| | - Mohamed S Ahmed
- Department of Clinical Studies, Collage of Veterinary Medicine, King Faisal University, 31982, Saudi Arabia; Department of Poultry Diseases, Faculty of Veterinary Medicine, South Valley University, Egypt
| | - Mohammed Al-Rasheed
- Department of Clinical Studies, Collage of Veterinary Medicine, King Faisal University, 31982, Saudi Arabia
| | - Eman M Rashad
- Animal Health Research Institute (Domietta Branch), Agriculture Research Centre, Egypt
| |
Collapse
|
2
|
Liu L, Song L, Liu T, Hui K, Hu C, Yang J, Pi X, Yan Y, Liu S, Zhang Y, Chen H, Cao Y, Zhou L, Qiao Y, Yu D, Yin C, Li X, Zhang C, Li D, Wang Z, Liu Z, Jiang X. Recombinant oncolytic virus NDV-anti-VEGFR2 enhances radiotherapy sensitivity in NSCLC by targeting VEGF signaling and impairing DNA repair. Gene Ther 2025:10.1038/s41434-025-00540-x. [PMID: 40382521 DOI: 10.1038/s41434-025-00540-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 04/09/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Resistance to radiotherapy is a significant challenge in the clinical management of non-small cell lung cancer (NSCLC). This study investigates a novel multimodal therapeutic strategy that combines oncolytic Newcastle disease virus (NDV) with an anti-VEGFR2 single-chain variable fragment (NDV-anti-VEGFR2) to enhance radiosensitivity in NSCLC. We engineered NDV-anti-VEGFR2 and assessed its efficacy in sensitizing Calu-1 cells to radiation. In vitro results demonstrated that NDV-anti-VEGFR2 significantly inhibited tumor cell proliferation when combined with radiotherapy. In vivo experiments revealed that NDV-anti-VEGFR2, combined with radiation, achieved a tumor growth inhibition rate of 86.48%, surpassing the effects of NDV or radiation alone. Mechanistic investigations indicated that NDV-anti-VEGFR2 mitigated hypoxia by downregulating HIF-1α and impaired DNA repair pathways, as evidenced by reduced levels of RAD51 and γ-H2AX. These findings suggest that NDV-anti-VEGFR2 not only normalizes tumor vasculature but also enhances the cytotoxic effects of radiation by compromising DNA repair mechanisms. Collectively, our results support the clinical potential of NDV-anti-VEGFR2 combined with radiotherapy as a promising strategy to overcome radiotherapy resistance in NSCLC. Future studies in immunocompetent models are warranted to elucidate the immune-mediated effects of this innovative therapeutic approach.
Collapse
Affiliation(s)
- Liang Liu
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Liying Song
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Tianyan Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Kaiyuan Hui
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Chenxi Hu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Jiarui Yang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Xuelei Pi
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yuanyuan Yan
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Shishi Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yating Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Hongna Chen
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yukai Cao
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Lihua Zhou
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Yun Qiao
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Dan Yu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Chengkai Yin
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Xu Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Chenfeng Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Deshan Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Zhihang Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China.
| | - Xiaodong Jiang
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China.
| |
Collapse
|
3
|
Yu Z, Chen Y, Chen S, Ye W, Li R, Fu Y, Chen Y, Fu W, Wei X, Yu Q, Cai Y, Wang L, Zhang Y, Ying H, Dai F, Han W. Oligoadenylate synthetase-like aggravated Newcastle disease virus-induced necroptosis in glioma cells. Front Oncol 2025; 15:1574214. [PMID: 40330823 PMCID: PMC12053153 DOI: 10.3389/fonc.2025.1574214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Background Newcastle disease virus (NDV) has emerged as a tumor-lysing agent in a variety of cancers. Previous studies have shown that NDV has cytolytic activity in gliomas; however, the underlying mechanisms have not been fully elucidated. Methods Comparing the glioma cells LN229 controlled group with the infected group of NDV rLa Sota-GFP strain, we strive to observe the changes in the genome and protein levels as well as the activation of the signalling pathways before and after the infection at the cellular level and at the level of the genes in the transcriptome, to study the molecular mechanism of necroptosis of the NDV-infected lethal LN229. Results We found that NDV infection which inhibited glioma cells LN229 proliferation and promoted apoptosis in a dose-dependent manner involved mitochondrial disruption by a molecular mechanism, whereas the Fe2+ assay didn't change. Additionally, the necroptosis inhibitor Nec-1 alleviated the cellular damage caused by NDV during infection of LN229 cells. Using RNA-seq analysis, the necroptosis pathway was significantly enriched in NDV-infected LN229 cells, and the antiviral gene OASL (Oligoadenylate synthetase-like) was significantly up-regulated in the apoptotic signalling pathway, which could be directly induced by NDV infection. Knockdown of OASL attenuates NDV infection-induced necroptosis in LN229 cells. Conclusion Our study demonstrates that NDV has cytolytic activity on glioma cells by inducing necroptosis. Additionally, targeting upregulation of OASL may provide a novel strategy to enhance necrotic apoptosis in glioma cells after NDV infection.
Collapse
Affiliation(s)
- Zecheng Yu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuxin Chen
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Sisi Chen
- Center of Laboratory Animal, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenjing Ye
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ruirui Li
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yutang Fu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yangkun Chen
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenhao Fu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xianqiao Wei
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qin Yu
- School of Information Engineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yili Cai
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lingyun Wang
- School of Medical Laboratory and Biological Engineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuheng Zhang
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huazhong Ying
- Center of Laboratory Animal, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fangwei Dai
- Center of Laboratory Animal, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wei Han
- Center of Laboratory Animal, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Ngantcha Tatchou E, Milcamps R, Oldenhove G, Lambrecht B, Ingrao F. Generation and characterization of chicken monocyte-derived dendritic cells. Front Immunol 2025; 16:1517697. [PMID: 39967657 PMCID: PMC11832469 DOI: 10.3389/fimmu.2025.1517697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Dendritic cells (DCs) play a crucial role in orchestrating immune responses by bridging innate and adaptive immunity. In vitro generation of DCs from mouse and human tissues such as bone marrow and peripheral blood monocytes, has been widely used to study their immunological functions. In chicken, DCs have mainly been derived from bone marrow cell cultures, with limited characterization from blood monocytes. Methods The present study takes advantage of newly available chicken immunological tools to further characterize chicken monocyte-derived dendritic cells (MoDCs), focusing on their phenotype, and functions, including antigen capture and T-cell stimulation, and response to live Newcastle disease virus (NDV) stimulation. Results Adherent chicken PBMCs were cultured with recombinant chicken granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4), for 5 days, leading to the upregulation of putative CD11c and MHCII, markers of DC differentiation. Subsequent stimulation with lipopolysaccharide (LPS) or 24 h triggered phenotypic maturation of MoDCs, characterized by the increased surface expression of MHCII and co-stimulatory molecules CD80 and CD40, and elevated IL-12p40 secretion. This maturation reduced endocytic capacity but enhanced the allogenic stimulatory activity of the chicken MoDCs. Upon NDV stimulation for 6 h, MoDCs upregulated antiviral pathways, including retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), melanoma differentiation-associated protein 5 (MDA5) and laboratory of genetics and physiology 2 (LGP2), alongside increased production of type I interferons (IFNs), and the pro-inflammatory cytokines tumor necrosis factor-α (TNF-α), IL-1β, and IL-6. However, these responses were downregulated after 24 hours. Conclusion These findings provide a comprehensive characterization of chicken MoDCs and suggest their potential as a model for studying host-pathogen interactions.
Collapse
Affiliation(s)
- Elie Ngantcha Tatchou
- Service of Avian Virology and Immunology, Sciensano, Brussels, Belgium
- Laboratory of Immunobiology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Romane Milcamps
- Service of Avian Virology and Immunology, Sciensano, Brussels, Belgium
- Molecular Virology Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Guillaume Oldenhove
- Laboratory of Immunobiology, Université Libre de Bruxelles, Gosselies, Belgium
| | | | - Fiona Ingrao
- Service of Avian Virology and Immunology, Sciensano, Brussels, Belgium
| |
Collapse
|
5
|
Jafari M, Abdoli S, Asgari M, Moghaddam Pour M, Shokrgozar MA, Sharifzadeh Z. Combination Therapy of Oncolytic Newcastle Virus and Lenalidomide Enhanced Cytotoxicity in Prostate Cancer Cells. IRANIAN BIOMEDICAL JOURNAL 2025; 29:9-19. [PMID: 40223343 PMCID: PMC12040631 DOI: 10.61186/ibj.4367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/10/2024] [Indexed: 04/20/2025]
Abstract
Background Despite existing treatments, advanced solid tumors, such as prostate cancer (PCa), require the development of novel anticancer therapies. Oncolytic viruses (OVs) present a potential treatment option for solid tumors. Newcastle disease virus (NDV) is one of the most promising OVs that can replicate within and destroys human cancer cells. This study aimed to evaluate the cytotoxic and apoptotic effects of the NDV strain on human PCa cells in vitro. Additionally, We examined a novel treatment for PCa by combining Lenalidomide (Len) with NDV. Methods NDV strains La Sota, B1, and I2 were tested for cytotoxicity against several cell lines. A safety assessment was conducted in primary cells using peripheral blood mononuclear cells (PBMCs). Also, apoptosis induction was measured using annexin V/7AAD staining. Finally, the cytotoxic effects of NDV alone and in combination with Len, were assessed using MTT. Results The NDV showed cytotoxic effects on tumor cell lines and induced apoptosis in infected prostate cells compared to control cells. The NDV La Sota strain exhibited significant oncolytic capacity, reducing the viability of LNCaP and DU145 cells to less than 40% at specific concentrations, while showing no cytotoxic effects on primary PBMCs. Also, NDV induced apoptosis in the prostate cell line by 60%. Furthermore, Len enhanced the cytotoxicity of PCa cells when combined with NDV. Conclusion Our study confirms the efficacy of oncolytic NDV treatment for PCa, particularly utilizing the La Sota strain. When combined with Len, NDV indicates an enhanced effectiveness in destroying tumor cells. These findings suggest a prospective treatment approach that needs more preclinical and clinical studies to improve outcomes in PCa treatment.
Collapse
Affiliation(s)
- Mahdie Jafari
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Shahriyar Abdoli
- School of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Asgari
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
- Myeloma Center, Winthrop P, Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Science, Little Rock, Arkansas, USA
| | - Masoud Moghaddam Pour
- Poultry Viral Vaccines Research and Production Department, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran
| | | | | |
Collapse
|
6
|
Jung B, An YH, Jang SH, Ryu G, Jung S, Kim S, Kim C, Jang H. The tumor suppressive effect and apoptotic mechanism of TRAIL gene-containing recombinant NDV in TRAIL-resistant colorectal cancer HT-29 cells and TRAIL-nonresistant HCT116 cells, with each cell bearing a mouse model. Cancer Med 2023; 12:20380-20395. [PMID: 37843231 PMCID: PMC10652305 DOI: 10.1002/cam4.6622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/12/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND TRAIL is an anticancer drug that induces cancer cell apoptosis by interacting with death receptors (DRs). However, owing to low cell-surface expression of DRs, certain colorectal cancer (CRC) cells resist TRAIL-induced apoptosis. Newcastle disease virus (NDV) infection can elevate DR protein expression in cancer cells, potentially influencing their TRAIL sensitivity. However, the precise mechanism by which NDV infection modulates DR expression and impacts TRAIL sensitivity in cancer cells remains unknown. METHODS Herein, we developed nonpathogenic NDV VG/GA strain-based recombinant NDV (rNDV) and TRAIL gene-containing rNDV (rNDV-TRAIL). We observed that viral infections lead to increased DR and TRAIL expressions and activate signaling proteins involved in intrinsic and extrinsic apoptosis pathways. Experiments were conducted in vitro using TRAIL-resistant CRC cells (HT-29) and nonresistant CRC cells (HCT116) and in vivo using relevant mouse models. RESULTS rNDV-TRAIL was found to exhibit better apoptotic efficacy than rNDV in CRC cells. Notably, rNDV-TRAIL had the stronger cancer cell-killing effect in TRAIL-resistant CRC cells. Western blot analyses showed that both rNDV and rNDV-TRAIL infections activate signaling proteins involved in the intrinsic and extrinsic apoptotic pathways. Notably, rNDV-TRAIL promotes concurrent intrinsic and extrinsic signal transduction in both HCT-116 and HT-29 cells. CONCLUSIONS Therefore, rNDV-TRAIL infection effectively enhances DR expression in DR-depressed HT-29 cells. Moreover, the TRAIL protein expressed by rNDV-TRAIL effectively interacts with DR, leading to enhanced apoptosis in TRAIL-resistant HT-29 cells. Therefore, rNDV-TRAIL has potential as a promising therapeutic approach for treating TRAIL-resistant cancers.
Collapse
Affiliation(s)
| | | | - Sung Hoon Jang
- Graduate School of Medical Science, College of medicineYonsei UniversitySeoulRepublic of Korea
| | | | | | - Seonhee Kim
- Department of Physiology & Medical Science, College of MedicineChungnam National UniversityDaejeonRepublic of Korea
| | - Cuk‐Seong Kim
- Department of Physiology & Medical Science, College of MedicineChungnam National UniversityDaejeonRepublic of Korea
| | - Hyun Jang
- Libentech Co. LTDDaejeonRepublic of Korea
| |
Collapse
|
7
|
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: 16] [Impact Index Per Article: 5.3] [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
|
8
|
Javaheri A, Bykov Y, Mena I, García-Sastre A, Cuadrado-Castano S. Avian Paramyxovirus 4 Antitumor Activity Leads to Complete Remissions and Long-term Protective Memory in Preclinical Melanoma and Colon Carcinoma Models. CANCER RESEARCH COMMUNICATIONS 2022; 2:602-615. [PMID: 35937459 PMCID: PMC9351398 DOI: 10.1158/2767-9764.crc-22-0025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/26/2022] [Accepted: 06/17/2022] [Indexed: 06/15/2023]
Abstract
Avulaviruses represent a diverse subfamily of non-segmented negative strand RNA viruses infecting avian species worldwide. To date, 22 different serotypes have been identified in a variety of avian hosts, including wild and domestic birds. APMV-1, also known as Newcastle disease virus (NDV), is the only avulavirus that has been extensively characterized due to its relevance for the poultry industry and, more recently, its inherent oncolytic activity and potential as a cancer therapeutic. An array of both naturally-occurring and recombinant APMV-1 strains has been tested in different preclinical models and clinical trials, highlighting NDV as a promising viral agent for human cancer therapy. To date, the oncolytic potential of other closely related avulaviruses remains unknown. Here, we have examined the in vivo anti-tumor capability of prototype strains of APMV serotypes -2, -3, -4, -6, -7, -8 and -9 in syngeneic murine colon carcinoma and melanoma tumor models. Our studies have identified APMV-4 Duck/Hong Kong/D3/1975 virus as a novel oncolytic agent with greater therapeutic potential than one of the NDV clinical candidate strains, La Sota. Intratumoral administration of the naturally-occurring APMV-4 virus significantly extends survival, promotes complete remission, and confers protection against re-challenge in both murine colon carcinoma and melanoma tumor models. Furthermore, we have designed a plasmid rescue strategy that allows us to develop recombinant APMV-4-based viruses. The infectious clone rAPMV-4 preserves the extraordinary antitumor capacity of its natural counterpart, paving the way to a promising next generation of viral therapeutics.
Collapse
Affiliation(s)
- Aryana Javaheri
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York
| | - Yonina Bykov
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York
| | | |
Collapse
|
9
|
Miguel Cejalvo J, Falato C, Villanueva L, Tolosa P, González X, Pascal M, Canes J, Gavilá J, Manso L, Pascual T, Prat A, Salvador F. Oncolytic Viruses: a new immunotherapeutic approach for breast cancer treatment? Cancer Treat Rev 2022; 106:102392. [DOI: 10.1016/j.ctrv.2022.102392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 12/22/2022]
|
10
|
Fulber JPC, Farnós O, Kiesslich S, Yang Z, Dash S, Susta L, Wootton SK, Kamen AA. Process Development for Newcastle Disease Virus-Vectored Vaccines in Serum-Free Vero Cell Suspension Cultures. Vaccines (Basel) 2021; 9:vaccines9111335. [PMID: 34835266 PMCID: PMC8623276 DOI: 10.3390/vaccines9111335] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023] Open
Abstract
The ongoing COVID-19 pandemic drew global attention to infectious diseases, attracting numerous resources for development of pandemic preparedness plans and vaccine platforms—technologies with robust manufacturing processes that can quickly be pivoted to target emerging diseases. Newcastle Disease Virus (NDV) has been studied as a viral vector for human and veterinary vaccines, but its production relies heavily on embryonated chicken eggs, with very few studies producing NDV in cell culture. Here, NDV is produced in suspension Vero cells, and analytical assays (TCID50 and ddPCR) are developed to quantify infectious and total viral titer. NDV-GFP and NDV-FLS (SARS-CoV-2 full-length spike protein) constructs were adapted to replicate in Vero and HEK293 suspension cultures using serum-free media, while fine-tuning parameters such as MOI, temperature, and trypsin concentration. Shake flask productions with Vero cells resulted in infectious titers of 1.07 × 108 TCID50/mL for NDV-GFP and 1.33 × 108 TCID50/mL for NDV-FLS. Production in 1 L batch bioreactors also resulted in high titers in culture supernatants, reaching 2.37 × 108 TCID50/mL for NDV-GFP and 3.16 × 107 TCID50/mL for NDV-FLS. This shows effective NDV production in cell culture, building the basis for a scalable vectored-vaccine manufacturing process that can be applied to different targets.
Collapse
Affiliation(s)
- Julia Puppin Chaves Fulber
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Omar Farnós
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Sascha Kiesslich
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Zeyu Yang
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Shantoshini Dash
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Leonardo Susta
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.S.); (S.K.W.)
| | - Sarah K. Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.S.); (S.K.W.)
| | - Amine A. Kamen
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
- Correspondence:
| |
Collapse
|
11
|
Meng Q, He J, Zhong L, Zhao Y. Advances in the Study of Antitumour Immunotherapy for Newcastle Disease Virus. Int J Med Sci 2021; 18:2294-2302. [PMID: 33967605 PMCID: PMC8100649 DOI: 10.7150/ijms.59185] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/21/2021] [Indexed: 01/08/2023] Open
Abstract
This article reviews the preclinical research, clinical application and development of Newcastle disease virus (NDV) in the field of cancer therapy. Based on the distinctive antitumour properties of NDV and its positive interaction with the patient's immune system, this biologic could be considered a major breakthrough in cancer treatment. On one hand, NDV infection creates an inflammatory environment in the tumour microenvironment, which can directly activate NK cells, monocytes, macrophages and dendritic cells and promote the recruitment of immune cells. On the other hand, NDV can induce the upregulation of immune checkpoint molecules, which may break immune tolerance and immune checkpoint blockade resistance. In fact, clinical data have shown that NDV combined with immune checkpoint blockade can effectively enhance the antitumour response, leading to the regression of local tumours and distant tumours when injected, and this effect is further enhanced by targeted manipulation and modification of the NDV genome. At present, recombinant NDV and recombinant NDV combined with immune checkpoint blockers have entered different stages of clinical trials. Based on these studies, further research on NDV is warranted.
Collapse
Affiliation(s)
- Qiuxing Meng
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, China
| | - Jian He
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, China
| | - Liping Zhong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, China
| | - Yongxiang Zhao
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
12
|
Burman B, Pesci G, Zamarin D. Newcastle Disease Virus at the Forefront of Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12123552. [PMID: 33260685 PMCID: PMC7761210 DOI: 10.3390/cancers12123552] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/23/2022] Open
Abstract
Preclinical and clinical studies dating back to the 1950s have demonstrated that Newcastle disease virus (NDV) has oncolytic properties and can potently stimulate antitumor immune responses. NDV selectively infects, replicates within, and lyses cancer cells by exploiting defective antiviral defenses in cancer cells. Inflammation within the tumor microenvironment in response to NDV leads to the recruitment of innate and adaptive immune effector cells, presentation of tumor antigens, and induction of immune checkpoints. In animal models, intratumoral injection of NDV results in T cell infiltration of both local and distant non-injected tumors, demonstrating the potential of NDV to activate systemic adaptive antitumor immunity. The combination of intratumoral NDV with systemic immune checkpoint blockade leads to regression of both injected and distant tumors, an effect further potentiated by introduction of immunomodulatory transgenes into the viral genome. Clinical trials with naturally occurring NDV administered intravenously demonstrated durable responses across numerous cancer types. Based on these studies, further exploration of NDV is warranted, and clinical studies using recombinant NDV in combination with immune checkpoint blockade have been initiated.
Collapse
Affiliation(s)
- Bharat Burman
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (B.B.); (G.P.)
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Giulio Pesci
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (B.B.); (G.P.)
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dmitriy Zamarin
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (B.B.); (G.P.)
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Weill-Cornell Medical College, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
13
|
Ginting TE, Christian S, Larasati YO, Suryatenggara J, Suriapranata IM, Mathew G. Antiviral interferons induced by Newcastle disease virus (NDV) drive a tumor-selective apoptosis. Sci Rep 2019; 9:15160. [PMID: 31641164 PMCID: PMC6806003 DOI: 10.1038/s41598-019-51465-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/01/2019] [Indexed: 01/06/2023] Open
Abstract
Newcastle disease virus (NDV) strongly induces both type I and III antiviral interferons (IFNs-α/-β and IFN-λ, respectively) in tumor cells while it induces mainly type III IFN in normal cells. Impairment of antiviral type I IFN signaling in tumor cells is thought to be the reason for effective oncolysis. However, there is lack of clarity why lentogenic strain NDV can also induce oncolysis. NDV infection caused apoptosis in normal and tumor cells as demonstrated with the caspase-3 enzyme activation and annexin-V detection. The apoptosis response was inhibited by B18R protein (a type I IFN inhibitor) in tumor cells i.e. A549 and U87MG, and not in normal cells i.e. NB1RGB and HEK293. Similarly, UV-inactivated medium from NDV infection was shown to induce apoptosis in corresponding cells and the response was inhibited in A549 and U87MG cells with the addition of B18R protein. Treatment with combination of IFNs-α/-β/-λ or IFNs-α/-β or IFN-λ in NB1RGB, HEK293, A549 and U87MG showed that caspase activity in IFNs-α/-β/-λ group was the highest, followed with IFN-α/-β group and IFN-λ group. This suggests that tumor-selectivity of NDV is mainly because of the cumulative effect of type I and III in tumor cells that lead to higher apoptotic effect.
Collapse
Affiliation(s)
- Teridah Ernala Ginting
- Division of Immunology, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Jalan Boulevard Jenderal Sudirman 1688, Lippo Karawaci, Tangerang, Banten, 15811, Indonesia.
| | - Salomo Christian
- Division of Immunology, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Jalan Boulevard Jenderal Sudirman 1688, Lippo Karawaci, Tangerang, Banten, 15811, Indonesia
| | - Young Othiwi Larasati
- Division of Immunology, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Jalan Boulevard Jenderal Sudirman 1688, Lippo Karawaci, Tangerang, Banten, 15811, Indonesia
| | - Jeremiah Suryatenggara
- Division of Immunology, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Jalan Boulevard Jenderal Sudirman 1688, Lippo Karawaci, Tangerang, Banten, 15811, Indonesia
| | - Ivet Marita Suriapranata
- Division of Immunology, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Jalan Boulevard Jenderal Sudirman 1688, Lippo Karawaci, Tangerang, Banten, 15811, Indonesia
| | - George Mathew
- Division of Immunology, Mochtar Riady Institute for Nanotechnology and Medical Science Group, University of Pelita Harapan, Jalan Boulevard Jenderal Sudirman 1688, Lippo Karawaci, Tangerang, Banten, 15811, Indonesia
| |
Collapse
|
14
|
Meng G, Fei Z, Fang M, Li B, Chen A, Xu C, Xia M, Yu D, Wei J. Fludarabine as an Adjuvant Improves Newcastle Disease Virus-Mediated Antitumor Immunity in Hepatocellular Carcinoma. Mol Ther Oncolytics 2019; 13:22-34. [PMID: 31011625 PMCID: PMC6461577 DOI: 10.1016/j.omto.2019.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 03/21/2019] [Indexed: 12/15/2022] Open
Abstract
In addition to direct oncolysis, oncolytic viruses (OVs) also induce antitumor immunity, also called viro-immunotherapy. Limited viral replication and immune-negative feedback are the major hurdles to effective viro-immunotherapy. In this study, we found that use of an adjuvant of fludarabine, a chemotherapeutic drug for chronic myeloid leukemia, increased the replication of Newcastle disease virus (NDV) by targeting signal transducer and activator of transcription 1 (STAT1), which led to enhanced oncolysis of hepatocellular carcinoma (HCC) cells. Moreover, fludarabine accelerated ubiquitin-proteasomal degradation by enhancing ubiquitylation rather than proteasomal activity. This resulted in accelerated degradation of phosphorylated STAT3 and indoleamine 2, 3-dioxygenase 1 (IDO1), whose expression was induced by NDV infection. In addition, fludarabine significantly increased the NDV-induced infiltration of NK cells and decreased the number of NDV-induced myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment. The aforementioned effects of fludarabine significantly improved NDV-mediated antitumor immunity and prolonged survival in mouse model of HCC. Our findings indicate the utility of fludarabine as an adjuvant for oncolytic anticancer viro-immunotherapy.
Collapse
Affiliation(s)
- Gang Meng
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Ziwei Fei
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - Mingyue Fang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - Binghua Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
- 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
| | - Chun Xu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Nanjing 210009, China
| | - Mao Xia
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, 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
|
15
|
Zhao J, Liu C, Zhang J, Huang X, Zhang G. Cytokine expression in chicken embryo fibroblasts in response to infection with virulent or lentogenic avian avulavirus 1 (AAvV-1). Microb Pathog 2019; 133:103556. [PMID: 31128172 DOI: 10.1016/j.micpath.2019.103556] [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: 02/15/2019] [Revised: 03/06/2019] [Accepted: 05/21/2019] [Indexed: 10/26/2022]
Abstract
To investigate cytokine expression in chicken embryo fibroblast (CEF) cells, a virulent avian avulavirus 1 (AAvV-1) strain called SG10 that rapidly causes 100% mortality in its host, and a vaccine strain (La Sota) were characterized. Real-time quantitative PCR was performed on RNA samples from CEF cells, which were collected at 0, 24, 48 and 72 h post-infection. The dynamic expression patterns of ten cytokines (TNF-α, IFN-α, IFN-β, IL-1β, IL-2, IL-6, IL-10, IL-13, IL-15 and IL-18) were investigated. The results showed that infection with lentogenic La Sota induced significantly higher levels of the antiviral cytokines IFN-α and IFN-β, proinflammatory cytokines IL-2, IL-15 and IL-18, and the anti-inflammatory cytokine IL-10, than did infection with virulent SG10. Furthermore, the SG10 strain induced dramatically higher levels of the inflammatory cytokine IL-6 than those observed in cells infected with La Sota. However, the expression patterns of the other cytokines that were tested did not show any obvious trends or statistically significant differences between cells infected with the virulent and avirulent strains. These data show that infection with lentogenic La Sota induced more effective immune responses and anti-viral effects than did infection with virulent SG10 in CEFs. Our data provide distinct expression patterns of IFNs and proinflammatory and anti-inflammatory cytokines to AAvV-1 by virulence in CEF cells.
Collapse
Affiliation(s)
- Jing Zhao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Changqing Liu
- Beijing Huadu Yukou Poultry Company Limited, Beijing, 101206, China
| | - Jiaojiao Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xiuying Huang
- Beijing Huadu Yukou Poultry Company Limited, Beijing, 101206, China
| | - Guozhong Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
16
|
Davola ME, Mossman KL. Oncolytic viruses: how "lytic" must they be for therapeutic efficacy? Oncoimmunology 2019; 8:e1581528. [PMID: 31069150 PMCID: PMC6492965 DOI: 10.1080/2162402x.2019.1596006] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022] Open
Abstract
Oncolytic viruses (OVs) preferentially target and kill cancer cells without affecting healthy cells through a multi-modal mechanism of action. While historically the direct killing activity of OVs was considered the primary mode of action, initiation or augmentation of a host antitumor immune response is now considered an essential aspect of oncolytic virotherapy. To improve oncolytic virotherapy, many studies focus on increasing virus replication and spread. In this article, we open for discussion the traditional dogma that correlates replication with the efficacy of OVs, pointing out several examples that oppose this principle.
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
| | - 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
|
17
|
Yurchenko KS, Jing Y, Shestopalov AM. Adaptation of the Newcastle Disease Virus to Cell Cultures for Enhancing Its Oncolytic Properties. Acta Naturae 2019; 11:66-73. [PMID: 31024750 PMCID: PMC6475870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Indexed: 11/23/2022] Open
Abstract
This study focuses on the adaptation of natural Newcastle disease virus (NDV) strains isolated from wild birds to human tumor cells. Many candidates for virotherapy are viruses pathogenic for human. During recombination of genetic material, there always exists a risk of getting a virus with an unstable genome. This problem can be solved by using natural apathogenic viruses as oncolytic agents. The Newcastle disease virus is the causative agent of contagious avian diseases. Its natural strains exhibit an antitumor effect and are considered safe for humans. As shown in earlier studies, the oncolytic properties of natural strains can be enhanced during adaptation to cell cultures, without interference in the virus genome. This study demonstrates that serial passaging increases the viral infectious titer in cancer cells. Moreover, the viability of tumor cells decreases post-infection when Newcastle disease virus strains are adapted to these cell cultures. The findings of this study complement the well-known data on the adaptation of the Newcastle disease virus to human cancer cells. Hence, it is possible to obtain a NDV strain with a more pronounced oncolytic potential during adaptation. This should be taken into account when choosing a strategy for designing anticancer drugs based on this virus.
Collapse
Affiliation(s)
- K. S. Yurchenko
- Federal State Budget Scientific Institution «Federal Research Center of Fundamental and Translational Medicine», Timakova Str. 2, 630117, Novosibirsk, Russia
| | - Yi. Jing
- Federal State Budgetary Educational Institution of higher professional education “Novosibirsk national research state University», Pirogova Str. 1, 630090, Novosibirsk, Russia
| | - A. M. Shestopalov
- Federal State Budget Scientific Institution «Federal Research Center of Fundamental and Translational Medicine», Timakova Str. 2, 630117, Novosibirsk, Russia
| |
Collapse
|
18
|
O’Bryan SM, Mathis JM. Oncolytic Virotherapy for Breast Cancer Treatment. Curr Gene Ther 2018; 18:192-205. [PMID: 30207220 PMCID: PMC7499349 DOI: 10.2174/1566523218666180910163805] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 06/20/2018] [Accepted: 09/06/2018] [Indexed: 12/24/2022]
Abstract
Breast cancer continues to be a leading cause of mortality among women. While at an early stage, localized breast cancer is easily treated; however, advanced stages of disease continue to carry a high mortality rate. The discrepancy in treatment success highlights that current treatments are insufficient to treat advanced-stage breast cancer. As new and improved treatments have been sought, one therapeutic approach has gained considerable attention. Oncolytic viruses are uniquely capable of targeting cancer cells through intrinsic or engineered means. They come in many forms, mainly from four major virus groups as defined by the Baltimore classification system. These vectors can target and kill cancer cells, and even stimulate immunotherapeutic effects in patients. This review discusses not only individual oncolytic viruses pursued in the context of breast cancer treatment but also the emergence of combination therapies with current or new therapies, which has become a particularly promising strategy for treatment of breast cancer. Overall, oncolytic virotherapy is a promising strategy for increased treatment efficacy for advanced breast cancer and consequently provides a unique platform for personalized treatments in patients.
Collapse
Affiliation(s)
- Samia M. O’Bryan
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - J. Michael Mathis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|