1
|
Shen Y, Zhang H, Xue M, Zheng C, Chen Q. HSV-1 as a gene delivery platform for cancer gene therapy. Trends Pharmacol Sci 2025; 46:324-336. [PMID: 40069043 DOI: 10.1016/j.tips.2025.02.006] [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: 01/23/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 04/06/2025]
Abstract
Herpes simplex virus type 1 (HSV-1) is a DNA virus with strong replication capabilities, a large genomic payload (≥30 kb), and low toxicity, making it a prominent vector in cancer gene therapy. Clinically approved oncolytic HSV-1 (oHSV-1) variants, such as T-VEC and G47Δ, demonstrate safety and efficacy in localized tumors, but face challenges in treating metastatic disease. To address this issue, next-generation oHSV-1 designs focus on precision targeting and immune remodeling through the delivery of multiple exogenous genes. In this review, we provide an overview of the inherent characteristics of oHSV-1 as a gene delivery platform, focusing on its genetic modification strategies, safety challenges in clinical applications, and future directions to maximize its therapeutic potential.
Collapse
Affiliation(s)
- Yangkun Shen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Hucheng Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
2
|
Kulbay M, Tuli N, Mazza M, Jaffer A, Juntipwong S, Marcotte E, Tanya SM, Nguyen AXL, Burnier MN, Demirci H. Oncolytic Viruses and Immunotherapy for the Treatment of Uveal Melanoma and Retinoblastoma: The Current Landscape and Novel Advances. Biomedicines 2025; 13:108. [PMID: 39857692 PMCID: PMC11762644 DOI: 10.3390/biomedicines13010108] [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: 12/09/2024] [Revised: 12/25/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Intraocular malignant tumors are rare; however, they can cause serious life-threatening complications. Uveal melanoma (UM) and retinoblastoma (RB) are the most common intraocular tumors in adults and children, respectively, and come with a great disease burden. For many years, several different treatment modalities for UM and RB have been proposed, with chemotherapy for RB cases and plaque radiation therapy for localized UM as first-line treatment options. Extraocular extension, recurrence, and metastasis constitute the major challenges of conventional treatments. To overcome these obstacles, immunotherapy, which encompasses different treatment options such as oncolytic viruses, antibody-mediated immune modulations, and targeted immunotherapy, has shown great potential as a novel therapeutic tool for cancer therapy. These anti-cancer treatment options provide numerous advantages such as selective cancer cell death and the promotion of an anti-tumor immune response, and they prove useful in preventing vision impairment due to macular and/or optic disc involvement. Numerous factors such as the vector choice, route of administration, dosing, and patient characteristics must be considered when engineering an oncolytic virus or other forms of immunotherapy vectors. This manuscript provides an in-depth review of the molecular design of oncolytic viruses (e.g., virus capsid proteins and encapsulation technologies, vectors for delivery, cell targeting) and immunotherapy. The most recent advances in preclinical- and clinical-phase studies are further summarized. The recent developments in virus-like drug conjugates (i.e., AU011), oncolytic viruses for metastatic UM, and targeted immunotherapies have shown great results in clinical trials for the future clinical application of these novel technologies in the treatment algorithm of certain intraocular tumors.
Collapse
Affiliation(s)
- Merve Kulbay
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
| | - Nicolas Tuli
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - Massimo Mazza
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - Armaan Jaffer
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 2V5, Canada
- Research Excellence Cluster in Vision, University of British Columbia, Vancouver, BC V5Z 3N9, Canada
| | - Sarinee Juntipwong
- Kellogg Eye Center, Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI 48105, USA
| | - Emily Marcotte
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Stuti Misty Tanya
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
| | - Anne Xuan-Lan Nguyen
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Miguel N. Burnier
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Hakan Demirci
- Kellogg Eye Center, Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
3
|
Liu Y, Fang S, Wang P, Zhang J, Liu F. Olaparib Enhances the Efficacy of Third-Generation Oncolytic Adenoviruses Against Glioblastoma by Modulating DNA Damage Response and p66shc-Induced Apoptosis. CNS Neurosci Ther 2024; 30:e70124. [PMID: 39552450 PMCID: PMC11570871 DOI: 10.1111/cns.70124] [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: 07/22/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/19/2024] Open
Abstract
AIMS Patients with glioblastoma multiforme (GBM) do not benefit from current cancer treatments, and their prognosis is dismal. This study aimed to investigate the potential synergistic effects of TS-2021, a third-generation oncolytic adenovirus, combined with the PARP inhibitor olaparib in GBM. METHODS TS-2021's impact on p66shc-induced apoptosis, DNA damage response, and poly (ADP-ribose) polymerase (PARP) activation was evaluated in GBM cells. The synergistic effect of TS-2021 and olaparib was examined in GBM cell lines and an immunocompetent mouse model of GBM. Mechanistic studies focused on the role of p66shc phosphorylation in the observed effects. RESULTS TS-2021 triggered p66shc-induced apoptosis, DNA damage response, and PARP activation. The combination of TS-2021 and olaparib synergistically increased cell apoptosis and DNA damage and reduced PARP expression compared to monotherapy. Olaparib promoted TS-2021 replication and release in GBM cells. Mechanistically, olaparib combined with TS-2021 upregulated p66shc phosphorylation, enhancing tumor cell apoptosis. In vivo, the combination therapy inhibited tumor growth and prolonged survival, confirming the synergistic effect. CONCLUSION This study is the first to suggest that TS-2021 sensitizes GBM cells with wild-type BRCA1/2 to olaparib. The combination of TS-2021 and olaparib shows a synergistic therapeutic effect against GBM, providing a promising treatment strategy.
Collapse
Affiliation(s)
- Yida Liu
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| | - Peiwen Wang
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing Tiantan Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Laboratory of Biomedical MaterialsBeijingChina
| |
Collapse
|
4
|
Liu S, Li M, Sun F, Zhang J, Liu F. Enhancing the immune effect of oHSV-1 therapy through TLR3 signaling in uveal melanoma. J Cancer Res Clin Oncol 2023; 149:901-912. [PMID: 36030435 DOI: 10.1007/s00432-022-04272-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/07/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Uveal melanoma (UM) is the most common primary intraocular malignant tumor in adults, with patients having a low overall survival rate. Oncolytic viruses (OVs) have been shown effective as monotherapy or combined with immunotherapy in the treatment of UM. Oncolytic herpes simplex type I virus (oHSV-1) was found to alter gene expression and immune function in UMs. We investigated whether a combination treatment would be more effective in treating UM and reactive immune cells. METHODS RNA sequencing analysis were used to identify the effect of oHSV-1 infection in UM cells and protein changes were validated by western blot. Cell viability assays were performed through UM cell lines (MUM2B, 92.1, and MP41) and retinal pigment epithelial cell line (ARPE-19) to identify the efficacy and safety of the combination treatment. Western blot, qRT-PCR, cell viability assay and immunocytochemistry were performed to discover the reactivation of immune cells (U937 and HMC3). RESULTS Through RNA sequencing analysis and in vitro molecular biology assays, this study tested the ability of oHSV-1 combined with the TLR3 agonist poly(I:C) to re-activate the TLR3 meditated NF-ƙB signaling pathway and further increase the anti-tumor activity of UM cells and macrophages, including the stimulation of macrophage polarization and proliferation. CONCLUSIONS These findings indicate that the treatment of UM with a combination of oHSV-1 and poly(I:C) generates immune responses and enhances anti-tumoral activity, suggesting the need for further investigations and clinical trials of this combination.
Collapse
Affiliation(s)
- Sisi Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Dongjiao Minxiang 1, Dongcheng District, Beijing, 100730, China
| | - Mingxin Li
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, No. 119 Nansihuan West Road, Fengtai District, Beijing, 100070, China
| | - Fengqiao Sun
- Department of Neurosurgery, Peking University International Hospital, Peking University Health Science Center, Peking University, Shengming Kexueyuan 1, Changping District, Beijing, 102206, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, No. 119 Nansihuan West Road, Fengtai District, Beijing, 100070, China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, No. 119 Nansihuan West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
5
|
Hao P, Zhang C, Ma H, Wang R. Enhanced tumor inhibiting effect of 131I-BDI-1-based radioimmunotherapy and cytosine deaminase gene therapy modulated by a radio-sensitive promoter in nude mice bearing bladder cancer. JOURNAL OF RADIATION RESEARCH 2023; 64:85-90. [PMID: 36418230 PMCID: PMC9855308 DOI: 10.1093/jrr/rrac075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/05/2022] [Indexed: 06/16/2023]
Abstract
Radioimmunotherapy (RIT) has great potential in cancer therapy. However, its efficacy in numerous tumors is restricted due to myelotoxicity, thereby limiting the dose of radionuclide. To increase tumor radiosensitivity, we incorporated the recombinant lentivirus into the EJ cells (bladder cancer [BC] cells), and examined the combined anti-tumor effects of RIT with 131I-BDI-1(131I-monoclonal antibody against human BC-1) and gene therapy (GT). The recombinant lentivirus was constructed and packed. The animal xenograft model was built and when the tumor reached about 0.5 cm in diameter, the mice were randomly separated into four groups: (1) RIT + GT: the xenografts were continuously incorporated with the recombinant lentivirus for two days. And 7.4 MBq 131I-BDI-1 was IV-injected, and 10 mg prodrug 5-fluorocytosine (FC) was IV-injected for 7 days, (2) RIT: same dose of 131I-BDI-1 as the previous group mice, (3) GT: same as the first group, except no 131I-BDI-1, and (4) Untreated. Compute tumor volumes in all groups. After 28 days the mice were euthanized and the tumors were extracted and weighed, and the inhibition rate was computed. The RIT + GT mice, followed by the RIT mice, exhibited markedly slower tumor growth, compared to the control mice. The tumor size was comparable between the GT and control mice. The tumor inhibition rates after 28 days of incubation were 42.85 ± 0.23%, 27.92 ± 0.21% and 0.57 ± 0.11% for the four groups, respectively. In conclusion, RIT, combined with GT, suppressed tumor development more effectively than RIT or GT alone. This data highlights the potent additive effect of radioimmune and gene therapeutic interventions against cancer.
Collapse
Affiliation(s)
- Pan Hao
- Department of Nuclear Medicine, LuHe Hospital, Capital Medical University, Beijing 101149, China
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Chunli Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Huan Ma
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Rongfu Wang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
6
|
Pereira GDM, Bormio Nunes JH, Cruz ÁB, Pereira DH, Buglio KE, Ruiz ALT, de Carvalho JE, Frajácomo SCL, Lustri WR, Bergamini FR, Corbi PP. Synthesis, spectroscopic characterization, antibacterial activity and antiproliferative profile of a new silver(I) complex of 5-fluorocytosine. J Fluor Chem 2023. [DOI: 10.1016/j.jfluchem.2023.110096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
7
|
Microbial cytosine deaminase is a programmable anticancer prodrug mediating enzyme: antibody, and gene directed enzyme prodrug therapy. Heliyon 2022; 8:e10660. [PMID: 36164544 PMCID: PMC9508425 DOI: 10.1016/j.heliyon.2022.e10660] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/26/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Cytosine deaminase (CDA) is a non-mammalian enzyme with powerful activity in mediating the prodrug 5-fluorcytosine (5-FC) into toxic drug 5-fluorouracil (5-FU), as an alternative directed approach for the traditional chemotherapies and radiotherapies of cancer. This enzyme has been frequently reported and characterized from various microorganisms. The therapeutic strategy of 5-FC-CDA involves the administration of CDA followed by the prodrug 5-FC injection to generate cytotoxic 5-FU. The antiproliferative activity of CDA-5-FC elaborates from the higher activity of uracil pathway in tumor cells than normal ones. The main challenge of the therapeutic drug 5-FU are the short half-life, lack of selectivity and emergence of the drug resistance, consistently to the other chemotherapies. So, mediating the 5-FU to the tumor cells by CDA is one of the most feasible approaches to direct the drug to the tumor cells, reducing its toxic effects and improving their pharmacokinetic properties. Nevertheless, the catalytic efficiency, stability, antigenicity and targetability of CDA-5-FC, are the major challenges that limit the clinical application of this approach. Thus, exploring the biochemical properties of CDA from various microorganisms, as well as the approaches for localizing the system of CDA-5-FC to the tumor cells via the antibody directed enzyme prodrug therapy (ADEPT) and gene directed prodrug therapy (GDEPT) were the objectives of this review. Finally, the perspectives for increasing the therapeutic efficacy, and targetability of the CDA-5-FC system were described.
Collapse
|
8
|
EMT-Related Gene Signature Predicts the Prognosis in Uveal Melanoma Patients. JOURNAL OF ONCOLOGY 2022; 2022:5436988. [PMID: 35990996 PMCID: PMC9391141 DOI: 10.1155/2022/5436988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 11/18/2022]
Abstract
Background. Uveal melanoma (UVM) is the most common primary intraocular malignancy in adults. Epithelial-mesenchymal transition (EMT) is an essential regulator of the UVM’s immune microenvironment. However, the precise role of EMT in UVM remains to be explored and the development of a related treatment strategy is urgently needed. Methods. Multiomics data and clinical information for TCGA-UVM were used to identify the EMT subtypes and analyze their regulatory role in the immune microenvironment in UVM. A machine-learning method based on the identified subtypes was utilized to construct the EMT feature-based prognostic model. External validation cohorts GSE84976 and GSE22138 were employed to validate the model’s robustness. Immunotherapy cohort IMvigor210 was used to explore the model’s potential to predict immunotherapy responsiveness. Results. Two EMT subtypes were identified in UVM. The role of EMT in shaping the immune microenvironment and regulating cancer-immunity circle of UVM was analyzed. A robust prognostic model was presented and validated to predict patient prognosis. The model also predicted patient’s immune features and immunotherapy responsiveness. Conclusion. The EMT-mediated immune features in UVM were illustrated, providing a reliable model to facilitate precise UVM treatment. This research may assist in decision-making during clinical UVM therapy.
Collapse
|
9
|
Zhu G, Zhang Q, Zhang J, Liu F. Targeting Tumor-Associated Antigen: A Promising CAR-T Therapeutic Strategy for Glioblastoma Treatment. Front Pharmacol 2021; 12:661606. [PMID: 34248623 PMCID: PMC8264285 DOI: 10.3389/fphar.2021.661606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/09/2021] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor T cells (CAR-T) therapy is a prospective therapeutic strategy for blood cancers tumor, especially leukemia, but it is not effective for solid tumors. Glioblastoma (GBM) is a highly immunosuppressive and deadly malignant tumor with poor responses to immunotherapies. Although CAR-T therapeutic strategies were used for glioma in preclinical trials, the current proliferation activity of CAR-T is not sufficient, and malignant glioma usually recruit immunosuppressive cells to form a tumor microenvironment that hinders CAR-T infiltration, depletes CAR-T, and impairs their efficacy. Moreover, specific environments such as hypoxia and nutritional deficiency can hinder the killing effect of CAR-T, limiting their therapeutic effect. The normal brain lack lymphocytes, but CAR-T usually can recognize specific antigens and regulate the tumor immune microenvironment to increase and decrease pro- and anti-inflammatory factors, respectively. This increases the number of T cells and ultimately enhances anti-tumor effects. CAR-T therapy has become an indispensable modality for glioma due to the specific tumor-associated antigens (TAAs). This review describes the characteristics of CAR-T specific antigen recognition and changing tumor immune microenvironment, as well as ongoing research into CAR-T therapy targeting TAAs in GBM and their potential clinical application.
Collapse
Affiliation(s)
- Guidong Zhu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Laboratory of Biomedical Materials, Beijing, China.,Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan, China
| | - Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China.,Beijing Laboratory of Biomedical Materials, Beijing, China
| |
Collapse
|
10
|
D'Aguanno S, Mallone F, Marenco M, Del Bufalo D, Moramarco A. Hypoxia-dependent drivers of melanoma progression. J Exp Clin Cancer Res 2021; 40:159. [PMID: 33964953 PMCID: PMC8106186 DOI: 10.1186/s13046-021-01926-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia, a condition of low oxygen availability, is a hallmark of tumour microenvironment and promotes cancer progression and resistance to therapy. Many studies reported the essential role of hypoxia in regulating invasiveness, angiogenesis, vasculogenic mimicry and response to therapy in melanoma. Melanoma is an aggressive cancer originating from melanocytes located in the skin (cutaneous melanoma), in the uveal tract of the eye (uveal melanoma) or in mucosal membranes (mucosal melanoma). These three subtypes of melanoma represent distinct neoplasms in terms of biology, epidemiology, aetiology, molecular profile and clinical features.In this review, the latest progress in hypoxia-regulated pathways involved in the development and progression of all melanoma subtypes were discussed. We also summarized current knowledge on preclinical studies with drugs targeting Hypoxia-Inducible Factor-1, angiogenesis or vasculogenic mimicry. Finally, we described available evidence on clinical studies investigating the use of Hypoxia-Inducible Factor-1 inhibitors or antiangiogenic drugs, alone or in combination with other strategies, in metastatic and adjuvant settings of cutaneous, uveal and mucosal melanoma.Hypoxia-Inducible Factor-independent pathways have been also reported to regulate melanoma progression, but this issue is beyond the scope of this review.As evident from the numerous studies discussed in this review, the increasing knowledge of hypoxia-regulated pathways in melanoma progression and the promising results obtained from novel antiangiogenic therapies, could offer new perspectives in clinical practice in order to improve survival outcomes of melanoma patients.
Collapse
Affiliation(s)
- Simona D'Aguanno
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabiana Mallone
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Marco Marenco
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | | |
Collapse
|
11
|
Liu S, Liu F, Zhao M, Zhang J. Antitumor Efficacy of Oncolytic Herpes Virus Type 1 Armed with GM-CSF in Murine Uveal Melanoma Xenografts. Cancer Manag Res 2020; 12:11803-11812. [PMID: 33239914 PMCID: PMC7680789 DOI: 10.2147/cmar.s274605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background Uveal melanoma (UM) is the most common primary intraocular tumor in adults with a high incidence of metastasis. Standard care therapies for UM include enucleation and radiation, which are minimally effective in prolonging patient survival. Oncolytic virus treatment has become a new trend in cancer field. Of which, oncolytic herpes simplex virus type 1 (HSV-1) therapy is one of the most effective antitumor treatments. Here, we established an oncolytic HSV-1 encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), tested its efficacy in UM therapy, and investigated the innate immune response induced by this virus. Methods Oncolytic HSV-1 expressing GM-CSF (HSV-GM-CSF) was constructed, then verified using qPCR and Western blot assays. Cell viability assays and transmission electron microscopy were conducted on three UM cell lines, MUM2B, 92.1, and MP41, to assess the cell-killing ability and virus infection of this virus. For in vivo experiments, BALB/c-nude mice in situ UM xenografts were established to testify the efficacy of the oncolytic virus, oncolytic HSV-1, and HSV-GM-CSF groups, respectively. IVIS images, ocular volumes, mice weights, and survivals were tracked to see the efficacy of the virus. Hematoxylin and eosin staining, immunohistochemistry, and flow cytometry analyses were conducted to demonstrate the immune activity after virus treatment. Results All three tested UM cell lines were sensitive to infection by HSV-GM-CSF. In vivo xenograft experiments revealed that oncolytic virus HSV-1 reduced UM tumor volume and that oncolytic virus HSV-1 armed with GM-CSF enhanced the antitumor effect compared with unmodified HSV-1. The bodyweights of untreated control group mice were significantly lower than those of mice in either virus-treated group (HSV-1 or HSV-GM-CSF). Follow-up survivals were prolonged in the virus-treated groups compared with the control group and were prolonged to a greater extent in the HSV-GM-CSF group than in the HSV-1 group. Macrophage stimulation was observed following HSV-GM-CSF treatment. Conclusion Our results indicate that the recombinant oncolytic virus HSV-GM-CSF is a potential therapeutic treatment for UM.
Collapse
Affiliation(s)
- Sisi Liu
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, People's Republic of China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing 100070, People's Republic of China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, People's Republic of China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing 100070, People's Republic of China
| |
Collapse
|