1
|
Soko GF, Kosgei BK, Meena SS, Ng YJ, Liang H, Zhang B, Liu Q, Xu T, Hou X, Han RPS. Extracellular matrix re-normalization to improve cold tumor penetration by oncolytic viruses. Front Immunol 2025; 15:1535647. [PMID: 39845957 PMCID: PMC11751056 DOI: 10.3389/fimmu.2024.1535647] [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: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Immunologically inert or cold tumors pose a substantial challenge to the effectiveness of immunotherapy. The use of oncolytic viruses (OVs) to induce immunogenic cell death (ICD) in tumor cells is a well-established strategy for initiating the cancer immunity cycle (CIC). This process promotes the trafficking and infiltration of CD8+ T cells into tumors, thereby eliciting a tumor-specific immune response. Despite the potential of OVs for handling cold tumors, clinical outcomes have fallen short of expectations. To better understand the obstacles faced by oncolytic virus immunotherapy (OVI), we would like to revisit the OV issue. Growing evidence indicates that limited intratumoral penetration and inadequate intratumoral distribution of OVs are critical factors contributing to the suboptimal response to OVI. Aberrant expressions of matrix proteins by cancer-associated fibroblasts (CAFs) alter the mechanical properties of the tumor extracellular matrix (ECM). This results in increased ECM desmoplasia and elevated intratumoral interstitial fluid pressure (IFP), creating physical barriers that impede the penetration and dissemination of OVs within tumors. This review explores the latest advancements in strategies designed to improve the intratumoral penetration of OVs to facilitate the penetration of tumor-infiltrating lymphocytes (TILs) into cold tumors. Additionally, we investigated current clinical trials and challenges associated with translating these strategies into clinical practice to improve patient outcomes.
Collapse
Affiliation(s)
- Geofrey F. Soko
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, China
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Benson K. Kosgei
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Stephene S. Meena
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, China
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Ying Jing Ng
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Huihui Liang
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Bing Zhang
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qingjun Liu
- Biosensor National Special Laboratory & Key Laboratory for Biomedical Engineering of Education Ministry, Dept. of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Tielong Xu
- Evidence-based Medicine Research Center, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xinju Hou
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, China
- Dept. of Rehabilitation, Nanchang Hongdu Hospital of Chinese Medicine, Nanchang, China
| | - Ray P. S. Han
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
2
|
Arias AG, Tovar-Martinez L, Asciutto EK, Mann A, Põšnograjeva K, Gracia LS, Royo M, Haugas M, Teesalu T, Smulski C, Ruoslahti E, Scodeller P. A Cyclic Peptide Targets Glioblastoma by Binding to Aberrantly Exposed SNAP25. Mol Pharm 2025; 22:363-376. [PMID: 39575977 DOI: 10.1021/acs.molpharmaceut.4c00958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Disease-specific changes in tumors and other diseased tissues are an important target of research because they provide clues about the pathophysiology of the disease as well as uncover potentially useful markers for diagnosis and treatment. Here, we report a new cyclic peptide, CESPLLSEC (CES), that specifically accumulated (homed) in intracranial U87MG and the WT-GBM model of glioblastoma (GBM) from intravenous (IV) injection, and associated with the vasculature. Affinity chromatography of U87MG tumor extracts on insolubilized CES peptide identified Synaptosomal Associated Protein 25 (SNAP25) as a candidate target molecule (receptor) for CES. Several results supported the identification of SNAP25 as the CES receptor. IV-injected FAM-CES colocalized with SNAP25 in the tumors, and direct binding studies showed specific binding of the CES peptide to recombinant human SNAP25. A CES peptide-drug conjugate designed for photodynamic therapy showed selective cytotoxicity to SNAP25+ glioblastoma cell lines. Specific accumulation of systemically injected anti-SNAP25 antibody in U87MG glioblastoma and labeling of intact U87MG cells with anti-SNAP in flow cytometry showed that SNAP25 is available from the circulation but not in normal tissues and that it is present at the cell surface. Using an array of ECM proteins and surface plasmon resonance revealed that SNAP25 binds moderately to collagen V and strongly to collagen VI. Modeling studies suggested that CES and collagen VI compete for the same binding site on SNAP25. Our results introduce CES as a valuable targeting peptide for drug delivery and its receptor SNAP25 as a possible molecular marker of interest for glioblastoma.
Collapse
Affiliation(s)
- Alberto G Arias
- Medical Physics Department, Gerencia de Área Aplicaciones Nucleares a la Salud (GAANS)Centro Atómico Bariloche, Avenida Bustillo 9500, San Carlos de Bariloche R8402AGP, Argentina
| | - Laura Tovar-Martinez
- Institute for Advanced Chemistry of Catalonia, IQAC-CSIC, Jordi Girona 18-26, Barcelona 08034, Spain
- Doctorate in Biotechnology, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain
| | - Eliana K Asciutto
- School of Science and Technology, National University of San Martin (UNSAM) and CONICET, Campus Migueletes, 25 de Mayo y Francia, San Martín 1650, Argentina
| | - Aman Mann
- Aivocode, Biolabs San Diego, 9276 Scranton Rd., San Diego, California 92121, United States
| | - Kristina Põšnograjeva
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia
| | - Lorena Simón Gracia
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia
| | - Miriam Royo
- Institute for Advanced Chemistry of Catalonia, IQAC-CSIC, Jordi Girona 18-26, Barcelona 08034, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Maarja Haugas
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia
| | - Cristian Smulski
- Medical Physics Department, Gerencia de Área Aplicaciones Nucleares a la Salud (GAANS)Centro Atómico Bariloche, Avenida Bustillo 9500, San Carlos de Bariloche R8402AGP, Argentina
| | - Erkki Ruoslahti
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd, La Jolla, California 92037, United States
| | - Pablo Scodeller
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia
- Institute for Advanced Chemistry of Catalonia, IQAC-CSIC, Jordi Girona 18-26, Barcelona 08034, Spain
| |
Collapse
|
3
|
Su J, Wu H, Yin C, Zhang F, Han F, Yu W. The hydrophobic cluster on the surface of protein is the key structural basis for the SDS-resistance of chondroitinase VhChlABC. MARINE LIFE SCIENCE & TECHNOLOGY 2024; 6:93-101. [PMID: 38433971 PMCID: PMC10902247 DOI: 10.1007/s42995-023-00201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 10/07/2023] [Indexed: 03/05/2024]
Abstract
The application of chondroitinase requires consideration of the complex microenvironment of the target. Our previous research reported a marine-derived sodium dodecyl sulfate (SDS)-resistant chondroitinase VhChlABC. This study further investigated the mechanism of VhChlABC resistance to SDS. Focusing on the hydrophobic cluster on its strong hydrophilic surface, it was found that the reduction of hydrophobicity of surface residues Ala181, Met182, Met183, Ala184, Val185, and Ile305 significantly reduced the SDS resistance and stability. Molecular dynamics (MD) simulation and molecular docking analysis showed that I305G had more conformational flexibility around residue 305 than wild type (WT), which was more conducive to SDS insertion and binding. The affinity of A181G, M182A, M183A, V185A and I305G to SDS was significantly higher than that of WT. In conclusion, the surface hydrophobic microenvironment composed of six residues was the structural basis for SDS resistance. This feature could prevent the binding of SDS and the destruction of hydrophobic packaging by increasing the rigid conformation of protein and reducing the binding force of SDS-protein. The study provides a new idea for the rational design of SDS-resistant proteins and may further promote chondroitinase research in the targeted therapy of lung diseases under the pressure of pulmonary surfactant. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-023-00201-1.
Collapse
Affiliation(s)
- Juanjuan Su
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237 China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, 266003 China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, 266003 China
| | - Hao Wu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237 China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, 266003 China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, 266003 China
| | - Chengying Yin
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237 China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, 266003 China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, 266003 China
| | - Fengchao Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237 China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, 266003 China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, 266003 China
| | - Feng Han
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237 China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, 266003 China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, 266003 China
| | - Wengong Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237 China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, 266003 China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, 266003 China
| |
Collapse
|
4
|
Kheirollahi A, Sadeghi S, Orandi S, Moayedi K, Khajeh K, Khoobi M, Golestani A. Chondroitinase as a therapeutic enzyme: Prospects and challenges. Enzyme Microb Technol 2024; 172:110348. [PMID: 37898093 DOI: 10.1016/j.enzmictec.2023.110348] [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: 05/22/2023] [Revised: 09/28/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
The chondroitinases (Chase) are bacterial lyases that specifically digest chondroitin sulfate and/or dermatan sulfate glycosaminoglycans via a β-elimination reaction and generate unsaturated disaccharides. In recent decades, these enzymes have attracted the attention of many researchers due to their potential applications in various aspects of medicine from the treatment of spinal cord injury to use as an analytical tool. In spite of this diverse spectrum, the application of Chase is faced with several limitations and challenges such as thermal instability and lack of a suitable delivery system. In the current review, we address potential therapeutic applications of Chase with emphasis on the challenges ahead. Then, we summarize the latest achievements to overcome the problems by considering the studies carried out in the field of enzyme engineering, drug delivery, and combination-based therapy.
Collapse
Affiliation(s)
- Asma Kheirollahi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Solmaz Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Orandi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiana Moayedi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Golestani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Hong SO, Kim J, Lee S, Shin J, Choi H, Lee E, Kang H, Lee H, Lee S, Yun N, An J, Choi H, Kim H, Kang W, Yoon Y, Kim S. Transgenic viral expression of PH-20, IL-12, and sPD1-Fc enhances immune cell infiltration and anti-tumor efficacy of an oncolytic virus. Mol Ther Oncolytics 2023; 30:301-315. [PMID: 37727704 PMCID: PMC10506102 DOI: 10.1016/j.omto.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
Oncolytic viruses are of significant clinical interest due to their ability to directly infect and kill tumors and enhance the anti-tumor immune response. Previously, we developed KLS-3010, a novel oncolytic virus derived from the International Health Department-White (IHD-W) strain vaccinia virus, which has robust tumoricidal effects. In the present study, we generated a recombinant oncolytic virus, KLS-3020, by inserting three transgenes (hyaluronidase [PH-20], interleukin-12 [IL-12], and soluble programmed cell death 1 fused to the Fc domain [sPD1-Fc]) into KLS-3010 and investigated its anti-tumor efficacy and ability to induce anti-tumor immune responses in CT26.WT and B16F10 mouse tumor models. A single injection of KLS-3020 significantly decreased tumor growth. The roles of the transgenes were investigated using viruses expressing each single transgene alone and KLS-3020. PH-20 promoted virus spread and tumor immune cell infiltration, IL-12 activated and reprogrammed T cells to inflammatory phenotypes, and sPD1-Fc increased intra-tumoral populations of activated T cells. The tumor-specific systemic immune response and the abscopal tumor control elicited by KLS-3020 were demonstrated in the CT26.WT tumor model. The insertion of transgenes into KLS-3020 increased its anti-tumor efficacy, supporting further clinical investigation of KLS-3020 as a novel oncolytic immunotherapy.
Collapse
Affiliation(s)
- Soon-Oh Hong
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Joonsung Kim
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Sungmin Lee
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Jaeil Shin
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Hwanjun Choi
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Eunjin Lee
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Hyesoo Kang
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Hyesun Lee
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Soondong Lee
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Naeun Yun
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Jiwon An
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Heonsik Choi
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| | - Hyeree Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Wonseok Kang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Yeup Yoon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sujeong Kim
- Institute of BioInnovation Research, Kolon Life Science, Seoul 07793, Republic of Korea
| |
Collapse
|
6
|
Lin YC, Chu YH, Liao WC, Chen CH, Hsiao WC, Ho YJ, Yang MY, Liu CH. CHST11-modified chondroitin 4-sulfate as a potential therapeutic target for glioblastoma. Am J Cancer Res 2023; 13:2998-3012. [PMID: 37559985 PMCID: PMC10408464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/23/2023] [Indexed: 08/11/2023] Open
Abstract
Aberrant chondroitin sulfate (CS) accumulation in glioblastoma (GBM) tissue has been documented, but the role of excessive CS in GBM progression and whether it can be a druggable target are largely unknown. The aim of this study is to clarify the biological functions of CHST11 in GBM cells, and evaluate therapeutic effects of blocking CHST11-derived chondroitin 4-sulfate (C4S). We investigated the expression of CHST11 in glioma tissue by immunohistochemistry, and analyzed CHST11 associated genes using public RNA sequencing datasets. The effects of CHST11 on aggressive cell behaviors have been studied in vitro and in vivo. We demonstrated that CHST11 is frequently overexpressed in GBM tissue, promoting GBM cell mobility and modulating C4S on GBM cells. We further discovered that CSPG4 is positively correlated with CHST11, and CSPG4 involved in CHST11-mediated cell invasiveness. In addition, GBM patients with high expression of CHST11 and CSPG4 have a significantly shorter survival time. We examined the effects of treating C4S-specific binding peptide (C4Sp) as a therapeutic agent in vitro and in vivo. C4Sp treatment attenuated GBM cell invasiveness and, notably, improved survival rate of orthotopic glioma cell transplant mice. Our results propose a possible mechanism of CHST11 in regulating GBM malignancy and highlight a novel strategy for targeting aberrant chondroitin sulfate in GBM cells.
Collapse
Affiliation(s)
- You-Cheng Lin
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Yin-Hung Chu
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Wen-Chieh Liao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Chia-Hua Chen
- Molecular Medicine Research Center, Chang Gung UniversityTaoyuan, Taiwan
| | - Wen-Chuan Hsiao
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical UniversityTaichung, Taiwan
| | - Meng-Yin Yang
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General HospitalTaichung, Taiwan
| | - Chiung-Hui Liu
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing UniversityTaichung, Taiwan
| |
Collapse
|
7
|
Scanlan H, Coffman Z, Bettencourt J, Shipley T, Bramblett DE. Herpes simplex virus 1 as an oncolytic viral therapy for refractory cancers. Front Oncol 2022; 12:940019. [PMID: 35965554 PMCID: PMC9364694 DOI: 10.3389/fonc.2022.940019] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
The need for efficacious and non-toxic cancer therapies is paramount. Oncolytic viruses (OVs) are showing great promise and are introducing new possibilities in cancer treatment with their ability to selectively infect tumor cells and trigger antitumor immune responses. Herpes Simplex Virus 1 (HSV-1) is a commonly selected OV candidate due to its large genome, relative safety profile, and ability to infect a variety of cell types. Talimogene laherparevec (T-VEC) is an HSV-1-derived OV variant and the first and only OV therapy currently approved for clinical use by the United States Food and Drug Administration (FDA). This review provides a concise description of HSV-1 as an OV candidate and the genomic organization of T-VEC. Furthermore, this review focuses on the advantages and limitations in the use of T-VEC compared to other HSV-1 OV variants currently in clinical trials. In addition, approaches for future directions of HSV-1 OVs as cancer therapy is discussed.
Collapse
Affiliation(s)
- Hayle Scanlan
- Rowan School of Medicine, RowanSOM-Jefferson Health-Virtua Our Lady of Lourdes Hospital, Stratford, NJ, United States
| | - Zachary Coffman
- Monroe Clinic Rural Family Medicine Program, The University of Illinois College of Medicine Rockford, Monroe, WI, United States
| | - Jeffrey Bettencourt
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Timothy Shipley
- Department of Biomedical Sciences, A.T. Still University School of Osteopathic Medicine in Arizona, Mesa, AZ, United States
| | - Debra E. Bramblett
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
- *Correspondence: Debra E. Bramblett,
| |
Collapse
|
8
|
Hong B, Sahu U, Mullarkey MP, Kaur B. Replication and Spread of Oncolytic Herpes Simplex Virus in Solid Tumors. Viruses 2022; 14:v14010118. [PMID: 35062322 PMCID: PMC8778098 DOI: 10.3390/v14010118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/11/2022] Open
Abstract
Oncolytic herpes simplex virus (oHSV) is a highly promising treatment for solid tumors. Intense research and development efforts have led to first-in-class approval for an oHSV for melanoma, but barriers to this promising therapy still exist that limit efficacy. The process of infection, replication and transmission of oHSV in solid tumors is key to obtaining a good lytic destruction of infected cancer cells to kill tumor cells and release tumor antigens that can prime anti-tumor efficacy. Intracellular tumor cell signaling and tumor stromal cells present multiple barriers that resist oHSV activity. Here, we provide a review focused on oncolytic HSV and the essential viral genes that allow for virus replication and spread in order to gain insight into how manipulation of these pathways can be exploited to potentiate oHSV infection and replication among tumor cells.
Collapse
|
9
|
Identification and Biochemical Characterization of a Surfactant-Tolerant Chondroitinase VhChlABC from Vibrio hyugaensis LWW-1. Mar Drugs 2021; 19:md19070399. [PMID: 34356824 PMCID: PMC8306027 DOI: 10.3390/md19070399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 11/16/2022] Open
Abstract
Chondroitinases, catalyzing the degradation of chondroitin sulfate (CS) into oligosaccharides, not only play a crucial role in understanding the structure and function of CS, but also have been reported as a potential candidate drug for the treatment of high CS-related diseases. Here, a marine bacterium Vibrio hyugaensis LWW-1 was isolated, and its genome was sequenced and annotated. A chondroitinase, VhChlABC, was found to belong to the second subfamily of polysaccharide lyase (PL) family 8. VhChlABC was recombinant expressed and characterized. It could specifically degrade CS-A, CS-B, and CS-C, and reached the maximum activity at pH 7.0 and 40 °C in the presence of 0.25 M NaCl. VhChlABC showed high stability within 8 h under 37 °C and within 2 h under 40 °C. VhChlABC was stable in a wide range of pH (5.0~10.6) at 4 °C. Unlike most chondroitinases, VhChlABC showed high surfactant tolerance, which might provide a good tool for removing extracellular CS proteoglycans (CSPGs) of lung cancer under the stress of pulmonary surfactant. VhChlABC completely degraded CS to disaccharide by the exolytic mode. This research expanded the research and application system of chondroitinases.
Collapse
|
10
|
The Effect of Herpes Simplex Virus-Type-1 (HSV-1) Oncolytic Immunotherapy on the Tumor Microenvironment. Viruses 2021; 13:v13071200. [PMID: 34206677 PMCID: PMC8310320 DOI: 10.3390/v13071200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022] Open
Abstract
The development of cancer causes disruption of anti-tumor immunity required for surveillance and elimination of tumor cells. Immunotherapeutic strategies aim for the restoration or establishment of these anti-tumor immune responses. Cancer immunotherapies include immune checkpoint inhibitors (ICIs), adoptive cellular therapy (ACT), cancer vaccines, and oncolytic virotherapy (OVT). The clinical success of some of these immunotherapeutic modalities, including herpes simplex virus type-1 derived OVT, resulted in Food and Drug Administration (FDA) approval for use in treatment of human cancers. However, a significant proportion of patients do not respond or benefit equally from these immunotherapies. The creation of an immunosuppressive tumor microenvironment (TME) represents an important barrier preventing success of many immunotherapeutic approaches. Mechanisms of immunosuppression in the TME are a major area of current research. In this review, we discuss how oncolytic HSV affects the tumor microenvironment to promote anti-tumor immune responses. Where possible we focus on oncolytic HSV strains for which clinical data is available, and discuss how these viruses alter the vasculature, extracellular matrix and immune responses in the tumor microenvironment.
Collapse
|
11
|
Zhou Z, Tian J, Zhang W, Xiang W, Ming Y, Chen L, Zhou J. Multiple strategies to improve the therapeutic efficacy of oncolytic herpes simplex virus in the treatment of glioblastoma. Oncol Lett 2021; 22:510. [PMID: 33986870 DOI: 10.3892/ol.2021.12771] [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: 10/22/2020] [Accepted: 03/29/2021] [Indexed: 11/06/2022] Open
Abstract
Oncolytic viruses have attracted widespread attention as biological anticancer agents that can selectively kill tumor cells without affecting normal cells. Although progress has been made in therapeutic strategies, the prognosis of patients with glioblastoma (GBM) remains poor and no ideal treatment approach has been developed. Recently, oncolytic herpes simplex virus (oHSV) has been considered a promising novel treatment approach for GBM. However, the therapeutic efficacy of oHSV in GBM, with its intricate pathophysiology, remains unsatisfactory due to several obstacles, such as limited replication and attenuated potency of oHSV owing to deletions or mutations in virulence genes, and ineffective delivery of the therapeutic virus. Multiple strategies have attempted to identify the optimal strategy for the successful clinical application of oHSV. Several preclinical trials have demonstrated that engineering novel oHSVs, developing combination therapies and improving methods for delivering oHSV to tumor cells seem to hold promise for improving the efficacy of this virotherapy.
Collapse
Affiliation(s)
- Zhengjun Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Junjie Tian
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Wenyan Zhang
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Wei Xiang
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Yang Ming
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China
| | - Ligang Chen
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan 646000, P.R. China.,Neurological Diseases and Brain Function Laboratory, Luzhou, Sichuan 646000, P.R. China
| | - Jie Zhou
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China.,Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan 646000, P.R. China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan 646000, P.R. China.,Neurological Diseases and Brain Function Laboratory, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
12
|
Nguyen HM, Saha D. The Current State of Oncolytic Herpes Simplex Virus for Glioblastoma Treatment. Oncolytic Virother 2021; 10:1-27. [PMID: 33659221 PMCID: PMC7917312 DOI: 10.2147/ov.s268426] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is a lethal primary malignant brain tumor with no current effective treatments. The recent emergence of immuno-virotherapy and FDA approval of T-VEC have generated a great expectation towards oncolytic herpes simplex viruses (oHSVs) as a promising treatment option for GBM. Since the generation and testing of the first genetically engineered oHSV in glioma in the early 1990s, oHSV-based therapies have shown a long way of great progress in terms of anti-GBM efficacy and safety, both preclinically and clinically. Here, we revisit the literature to understand the recent advancement of oHSV in the treatment of GBM. In addition, we discuss current obstacles to oHSV-based therapies and possible strategies to overcome these pitfalls.
Collapse
Affiliation(s)
- Hong-My Nguyen
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, School of Pharmacy, Abilene, TX, 79601, USA
| | - Dipongkor Saha
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, School of Pharmacy, Abilene, TX, 79601, USA
| |
Collapse
|
13
|
Zhang Q, Liu F. Advances and potential pitfalls of oncolytic viruses expressing immunomodulatory transgene therapy for malignant gliomas. Cell Death Dis 2020; 11:485. [PMID: 32587256 PMCID: PMC7316762 DOI: 10.1038/s41419-020-2696-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is an immunosuppressive, lethal brain tumor. Despite advances in molecular understanding and therapies, the clinical benefits have remained limited, and the life expectancy of patients with GBM has only been extended to ~15 months. Currently, genetically modified oncolytic viruses (OV) that express immunomodulatory transgenes constitute a research hot spot in the field of glioma treatment. An oncolytic virus is designed to selectively target, infect, and replicate in tumor cells while sparing normal tissues. Moreover, many studies have shown therapeutic advantages, and recent clinical trials have demonstrated the safety and efficacy of their usage. However, the therapeutic efficacy of oncolytic viruses alone is limited, while oncolytic viruses expressing immunomodulatory transgenes are more potent inducers of immunity and enhance immune cell-mediated antitumor immune responses in GBM. An increasing number of basic studies on oncolytic viruses encoding immunomodulatory transgene therapy for malignant gliomas have yielded beneficial outcomes. Oncolytic viruses that are armed with immunomodulatory transgenes remain promising as a therapy against malignant gliomas and will undoubtedly provide new insights into possible clinical uses or strategies. In this review, we summarize the research advances related to oncolytic viruses that express immunomodulatory transgenes, as well as potential treatment pitfalls in patients with malignant gliomas.
Collapse
Affiliation(s)
- Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
- Beijing Laboratory of Biomedical Materials, Beijing, 100070, China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
- Beijing Laboratory of Biomedical Materials, Beijing, 100070, China.
| |
Collapse
|
14
|
Development of oncolytic virotherapy: from genetic modification to combination therapy. Front Med 2020; 14:160-184. [PMID: 32146606 PMCID: PMC7101593 DOI: 10.1007/s11684-020-0750-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022]
Abstract
Oncolytic virotherapy (OVT) is a novel form of immunotherapy using natural or genetically modified viruses to selectively replicate in and kill malignant cells. Many genetically modified oncolytic viruses (OVs) with enhanced tumor targeting, antitumor efficacy, and safety have been generated, and some of which have been assessed in clinical trials. Combining OVT with other immunotherapies can remarkably enhance the antitumor efficacy. In this work, we review the use of wild-type viruses in OVT and the strategies for OV genetic modification. We also review and discuss the combinations of OVT with other immunotherapies.
Collapse
|
15
|
Chondroitin sulfate synthase 1 enhances proliferation of glioblastoma by modulating PDGFRA stability. Oncogenesis 2020; 9:9. [PMID: 32019907 PMCID: PMC7000683 DOI: 10.1038/s41389-020-0197-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/13/2019] [Accepted: 01/16/2020] [Indexed: 11/13/2022] Open
Abstract
Chondroitin sulfate synthases, a family of enzyme involved in chondroitin sulfate (CS) polymerization, are dysregulated in various human malignancies, but their roles in glioma remain unclear. We performed database analysis and immunohistochemistry on human glioma tissue, to demonstrate that the expression of CHSY1 was frequently upregulated in glioma, and that it was associated with adverse clinicopathologic features, including high tumor grade and poor survival. Using a chondroitin sulfate-specific antibody, we showed that the expression of CHSY1 was significantly associated with CS formation in glioma tissue and cells. In addition, overexpression of CHSY1 in glioma cells enhanced cell viability and orthotopic tumor growth, whereas CHSY1 silencing suppressed malignant growth. Mechanistic investigations revealed that CHSY1 selectively regulates PDGFRA activation and PDGF-induced signaling in glioma cells by stabilizing PDGFRA protein levels. Inhibiting PDGFR activity with crenolanib decreased CHSY1-induced malignant characteristics of GL261 cells and prolonged survival in an orthotopic mouse model of glioma, which underlines the critical role of PDGFRA in mediating the effects of CHSY1. Taken together, these results provide information on CHSY1 expression and its role in glioma progression, and highlight novel insights into the significance of CHSY1 in PDGFRA signaling. Thus, our findings point to new molecular targets for glioma treatment.
Collapse
|
16
|
Khan AR, Yang X, Du X, Yang H, Liu Y, Khan AQ, Zhai G. Chondroitin sulfate derived theranostic and therapeutic nanocarriers for tumor-targeted drug delivery. Carbohydr Polym 2020; 233:115837. [PMID: 32059890 DOI: 10.1016/j.carbpol.2020.115837] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/22/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022]
Abstract
The standard chemotherapy is facing the challenges of lack of cancer selectivity and development of drug resistance. Currently, with the application of nanotechnology, the rationally designed nanocarriers of chondroitin sulfate (CS) have been fabricated and their unique features of low toxicity, biocompatibility, and active and passive targeting made them drug delivery vehicles of the choice for cancer therapy. The hydrophilic and anionic CS could be incorporated as a building block into- or decorated on the surface of nanoformulations. Micellar nanoparticles (NPs) self-assembled from amphiphilic CS-drug conjugates and CS-polymer conjugates, polyelectrolyte complexes (PECs) and nanogels of CS have been widely implicated in cancer directed therapy. The surface modulation of organic, inorganic, lipid and metallic NPs with CS promotes the receptor-mediated internalization of NPs to the tumor cells. The potential contribution of CS and CS-proteoglycans (CSPGs) in the pathogenesis of various cancer types, and CS nanocarriers in immunotherapy, radiotherapy, sonodynamic therapy (SDT) and photodynamic therapy (PDT) of cancer are summarized in this review paper.
Collapse
Affiliation(s)
- Abdur Rauf Khan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, PR China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, PR China
| | - Xiyou Du
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, PR China
| | - Haotong Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, PR China
| | - Yuanxiu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, PR China
| | - Abdul Qayyum Khan
- Pakistan Council of Scientific and Industrial Research, Lahore, Pakistan
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
17
|
Sette P, Amankulor N, Li A, Marzulli M, Leronni D, Zhang M, Goins WF, Kaur B, Bolyard C, Cripe TP, Yu J, Chiocca EA, Glorioso JC, Grandi P. GBM-Targeted oHSV Armed with Matrix Metalloproteinase 9 Enhances Anti-tumor Activity and Animal Survival. Mol Ther Oncolytics 2019; 15:214-222. [PMID: 31890868 PMCID: PMC6926261 DOI: 10.1016/j.omto.2019.10.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/19/2019] [Indexed: 12/12/2022] Open
Abstract
The use of mutant strains of oncolytic herpes simplex virus (oHSV) in early-phase human clinical trials for the treatment of glioblastoma multiforme (GBM) has proven safe, but limited efficacy suggests that more potent vector designs are required for effective GBM therapy. Inadequate vector performance may derive from poor intratumoral vector replication and limited spread to uninfected cells. Vector replication may be impaired by mutagenesis strategies to achieve vector safety, and intratumoral virus spread may be hampered by vector entrapment in the tumor-specific extracellular matrix (ECM) that in GBM is composed primarily of type IV collagen. In this report, we armed our previously described epidermal growth factor receptor (EGFR)vIII-targeted, neuronal microRNA-sensitive oHSV with a matrix metalloproteinase (MMP9) to improve intratumoral vector distribution. We show that vector-expressed MMP9 enhanced therapeutic efficacy and long-term animal survival in a GBM xenograft model.
Collapse
Affiliation(s)
- Paola Sette
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Nduka Amankulor
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Aofei Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marco Marzulli
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Daniela Leronni
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Mingdi Zhang
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - William F. Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Balveen Kaur
- Department of Neurological Surgery, Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Chelsea Bolyard
- Department of Neurological Surgery, Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Timothy P. Cripe
- Division of Hematology/Oncology/Blood and Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH, USA
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Jianhua Yu
- Hematologic Malignancies & Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA, USA
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Division of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - E. Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s/Faulkner Hospital and Harvey Cushing Neuro-oncology Laboratories, Harvard Medicine School, Boston, MA, USA
- Center for Neuro-oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joseph C. Glorioso
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Paola Grandi
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
18
|
Zheng M, Huang J, Tong A, Yang H. Oncolytic Viruses for Cancer Therapy: Barriers and Recent Advances. MOLECULAR THERAPY-ONCOLYTICS 2019; 15:234-247. [PMID: 31872046 PMCID: PMC6911943 DOI: 10.1016/j.omto.2019.10.007] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oncolytic viruses (OVs) are powerful new therapeutic agents in cancer therapy. With the first OV (talimogene laherparepvec [T-vec]) obtaining US Food and Drug Administration approval, interest in OVs has been boosted greatly. Nevertheless, despite extensive research, oncolytic virotherapy has shown limited efficacy against solid tumors. Recent advances in viral retargeting, genetic editing, viral delivery platforms, tracking strategies, OV-based gene therapy, and combination strategies have the potential to broaden the applications of oncolytic virotherapy in oncology. In this review, we present several insights into the limitations and challenges of oncolytic virotherapy, describe the strategies mentioned above, provide a summary of recent preclinical and clinical trials in the field of oncolytic virotherapy, and highlight the need to optimize current strategies to improve clinical outcomes.
Collapse
Affiliation(s)
- Meijun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Jianhan Huang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Hui Yang
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, P.R. China
| |
Collapse
|
19
|
Recent advances in the therapeutic uses of chondroitinase ABC. Exp Neurol 2019; 321:113032. [PMID: 31398353 DOI: 10.1016/j.expneurol.2019.113032] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/19/2019] [Accepted: 08/03/2019] [Indexed: 12/18/2022]
Abstract
Many studies, using pre-clinical models of SCI, have demonstrated the efficacy of chondroitinase ABC as a treatment for spinal cord injury and this has been confirmed in laboratories worldwide and in several animal models. The aim of this review is report the current state of research in the field and to compare the relative efficacies of these new interventions to improve outcomes in both acute and chronic models of SCI. We also report new methods of chondroitinase delivery and the outcomes of two clinical trials using the enzyme to treat spinal cord injury in dogs and disc herniation in human patients. Recent studies have assessed the outcomes of combining chondroitinase with other strategies known to promote recovery following spinal cord injury and new approaches. Evidence is emerging that one of the most powerful combinations is that of chondroitinase with cell transplants. The particular benefits of each of the different cell types used for these transplant experiments are discussed. Combining chondroitinase with rehabilitation also improves outcomes. Gene therapy is an efficient method of enzyme delivery to the injured spinal cord and circumvents the issue of the enzyme's thermo-instability. Other methods of delivery, such as via nanoparticles or synthetic scaffolds, have shown promise; however, the outcomes from these experiments suggest that these methods of delivery require further optimization to achieve similar levels of efficacy to that obtained by a gene therapy approach. Pre-clinical models have also shown chondroitinase is efficacious in the treatment of other conditions, such as peripheral nerve injury, stroke, coronary reperfusion, Parkinson's disease and certain types of cancer. The wide range of conditions where the benefits of chondroitinase treatment have been demonstrated reflects the complex roles that chondroitin sulphate proteoglycans (its substrate) play in health and disease and warrants the enzyme's further development as a therapy.
Collapse
|
20
|
Zhang H, Wang R, Yu Y, Liu J, Luo T, Fan F. Glioblastoma Treatment Modalities besides Surgery. J Cancer 2019; 10:4793-4806. [PMID: 31598150 PMCID: PMC6775524 DOI: 10.7150/jca.32475] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 07/04/2019] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma multiforme (GBM) is commonly known as the most aggressive primary CNS tumor in adults. The mean survival of it is 14 to 15 months, following the standard therapy from surgery, chemotherapy, to radiotherapy. Efforts in recent decades have brought many novel therapies to light, however, with limitations. In this paper, authors reviewed current treatments for GBM besides surgery. In the past decades, only radiotherapy, temozolomide (TMZ), and tumor treating field (TTF) were approved by FDA. Though promising in preclinical experiments, therapeutic effects of other novel treatments including BNCT, anti-angiogenic therapy, immunotherapy, epigenetic therapy, oncolytic virus therapy, and gene therapy are still either uncertain or discouraging in clinical results. In this review, we went through current clinical trials, underlying causes, and future therapy designs to present neurosurgeons and researchers a sketch of this field.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Ruizhe Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuanqiang Yu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jinfang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Tianmeng Luo
- Department of Medical Affairs, Xiangya Hospital, Central South University, Chang Sha, Hunan Province, China
| | - Fan Fan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Center for Medical Genetics & Hunan Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Central South University Changsha, China
| |
Collapse
|
21
|
Totsch SK, Schlappi C, Kang KD, Ishizuka AS, Lynn GM, Fox B, Beierle EA, Whitley RJ, Markert JM, Gillespie GY, Bernstock JD, Friedman GK. Oncolytic herpes simplex virus immunotherapy for brain tumors: current pitfalls and emerging strategies to overcome therapeutic resistance. Oncogene 2019; 38:6159-6171. [PMID: 31289361 PMCID: PMC6771414 DOI: 10.1038/s41388-019-0870-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 12/25/2022]
Abstract
Malignant tumors of the central nervous system (CNS) continue to be a leading cause of cancer-related mortality in both
children and adults. Traditional therapies for malignant brain tumors consist of surgical resection and adjuvant chemoradiation;
such approaches are often associated with extreme morbidity. Accordingly, novel, targeted therapeutics for neoplasms of the CNS,
such as immunotherapy with oncolytic engineered herpes simplex virus (HSV) therapy, are urgently warranted. Herein, we discuss
treatment challenges related to HSV virotherapy delivery, entry, replication, and spread, and in so doing focus on host antiviral
immune responses and the immune microenvironment. Strategies to overcome such challenges including viral re-engineering,
modulation of the immunoregulatory microenvironment and combinatorial therapies with virotherapy, such as checkpoint inhibitors,
radiation, and vaccination are also examined in detail.
Collapse
Affiliation(s)
- Stacie K Totsch
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles Schlappi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kyung-Don Kang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Brandon Fox
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth A Beierle
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard J Whitley
- Division of Pediatric Infectious Disease, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - G Yancey Gillespie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joshua D Bernstock
- Avidea Technologies, Inc, Baltimore, MD, USA. .,Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Gregory K Friedman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
22
|
Prospect of Plasmacytoid Dendritic Cells in Enhancing Anti-Tumor Immunity of Oncolytic Herpes Viruses. Cancers (Basel) 2019; 11:cancers11050651. [PMID: 31083559 PMCID: PMC6562787 DOI: 10.3390/cancers11050651] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/30/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
The major type I interferon-producing plasmacytoid dendritic cells (pDC) surround and infiltrate certain tumors like malignant melanoma, head and neck cancer, and ovarian and breast cancer. The presence of pDC in these tumors is associated with an unfavorable prognosis for the patients as long as these cells are unstimulated. Upon activation by synthetic Toll-like receptor agonists or viruses, however, pDC develop cytotoxic activities. Viruses have the additional advantage to augment cytotoxic activities of pDC via lytic replication in malignant lesions. These effects turn cold tumors into hotspots, recruiting further immune cells to the site of inflammation. Activated pDC contribute to cross-presentation of tumor-associated antigens by classical dendritic cells, which induce cytotoxic T-cells in particular in the presence of checkpoint inhibitors. The modification of oncolytic herpes viruses via genetic engineering favorably affects this process through the enhanced production of pro-inflammatory cytokines, curbing of tumor blood supply, and removal of extracellular barriers for efficient viral spread. Importantly, viral vectors may contribute to stimulation of memory-type adaptive immune responses through presentation of tumor-related neo- and/or self-antigens. Eventually, both replication-competent and replication-deficient herpes simplex virus 1 (HSV-1) may serve as vaccine vectors, which contribute to tumor regression by the stimulation of pDC and other dendritic cells in adjuvant and neo-adjuvant situations.
Collapse
|
23
|
Alzahrani T, Eftimie R, Trucu D. Multiscale modelling of cancer response to oncolytic viral therapy. Math Biosci 2019; 310:76-95. [DOI: 10.1016/j.mbs.2018.12.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 12/29/2018] [Accepted: 12/29/2018] [Indexed: 12/29/2022]
|
24
|
Omidi-Ardali H, Aminian M, Golestani A, Shahaboddin ME, Maleki M. N∆89 and C∆274 Truncated Enzymes of Chondroitinase ABC I Regain More Imperturbable Microenvironments Around Structural Components in Comparison to their Wild Type. Protein J 2019; 38:151-159. [DOI: 10.1007/s10930-019-09821-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Kim Y, Lee J, Lee D, Othmer HG. Synergistic Effects of Bortezomib-OV Therapy and Anti-Invasive Strategies in Glioblastoma: A Mathematical Model. Cancers (Basel) 2019; 11:E215. [PMID: 30781871 PMCID: PMC6406513 DOI: 10.3390/cancers11020215] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/18/2022] Open
Abstract
It is well-known that the tumor microenvironment (TME) plays an important role in the regulation of tumor growth and the efficacy of anti-tumor therapies. Recent studies have demonstrated the potential of combination therapies, using oncolytic viruses (OVs) in conjunction with proteosome inhibitors for the treatment of glioblastoma, but the role of the TME in such therapies has not been studied. In this paper, we develop a mathematical model for combination therapies based on the proteosome inhibitor bortezomib and the oncolytic herpes simplex virus (oHSV), with the goal of understanding their roles in bortezomib-induced endoplasmic reticulum (ER) stress, and how the balance between apoptosis and necroptosis is affected by the treatment protocol. We show that the TME plays a significant role in anti-tumor efficacy in OV combination therapy, and illustrate the effect of different spatial patterns of OV injection. The results illustrate a possible phenotypic switch within tumor populations in a given microenvironment, and suggest new anti-invasion therapies.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Junho Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Donggu Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
26
|
Liao WC, Yen HR, Liao CK, Tseng TJ, Lan CT, Liu CH. DSE regulates the malignant characters of hepatocellular carcinoma cells by modulating CCL5/CCR1 axis. Am J Cancer Res 2019; 9:347-362. [PMID: 30906633 PMCID: PMC6405965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/25/2019] [Indexed: 06/09/2023] Open
Abstract
Abnormal expression of dermatan sulfate epimerase (DSE) has been found in many types of cancer, while its expression and biological functions in hepatocellular carcinoma (HCC) progression remains obscure. Here we report that DSE, the enzyme that catalyzes the conversion of chondroitin sulfate (CS) to dermatan sulfate (DS), is a critical mediator of malignant character in HCC, through regulation of CCL5 signaling. DSE mRNA and protein were downregulated frequently in HCC tumors, where these events were associated with advanced tumor stages, metastases, and poor survival. DSE-mediated tumor growth was evaluated in immune-deficient and immune-complement mice models. Restoring DSE expression in HCC cells suppressed tumor growth, as well as decreased IL-1β and CCL5 levels in transplanted tumor tissue. Mechanistic investigations revealed that the expression of DSE altered CCL5 signaling and cell surface binding in HCC cells. Accordingly, DSE suppressed CCL5-induced cell growth, migration, and invasion, whereas silencing of DSE enhanced CCL5-triggered malignant phenotypes. Inhibiting CCR1 activity with BX471 decreased CCL5-induced malignant characters caused by siRNA-mediated knockdown of DSE in HCC cells, establishing the critical role of the CCL5/CCR1 axis in mediating the effects of DSE expression. Taken together, our results suggest that DSE dysregulation contributes to the malignant behavior of HCC cells. This provides novel insight into the significance of DSE in CCL5 signaling and HCC pathogenesis.
Collapse
Affiliation(s)
- Wen-Chieh Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
- Department of Medical Education, Chung Shan Medical University HospitalTaichung, Taiwan
| | - Hung-Rong Yen
- School of Chinese Medicine, China Medical UniversityTaichung, Taiwan
- Department of Chinese Medicine, China Medical University HospitalTaichung, Taiwan
- Chinese Medicine Research Center, China Medical UniversityTaichung, Taiwan
| | - Chih-Kai Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
- Department of Medical Education, Chung Shan Medical University HospitalTaichung, Taiwan
| | - To-Jung Tseng
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
- Department of Medical Education, Chung Shan Medical University HospitalTaichung, Taiwan
| | - Chyn-Tair Lan
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
- Department of Medical Education, Chung Shan Medical University HospitalTaichung, Taiwan
| | - Chiung-Hui Liu
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
- Department of Medical Education, Chung Shan Medical University HospitalTaichung, Taiwan
| |
Collapse
|
27
|
Kahramanian A, Kuroda T, Wakimoto H. Construction of Oncolytic Herpes Simplex Virus with Therapeutic Genes of Interest. Methods Mol Biol 2019; 1937:177-188. [PMID: 30706396 DOI: 10.1007/978-1-4939-9065-8_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Herpes simplex virus (HSV) is one of the most extensively studied oncolytic virus platforms. The recent FDA approval of talimogene laherparepvec (T-VEC) has been accelerating translational research of oncolytic HSV (oHSV) as a promising therapeutic for refractory cancers such as glioblastoma, the deadliest primary malignancy in the brain. The large genome size of HSV readily allows arming of oHSV by incorporating therapeutic transgenes within the virus, as exemplified by T-VEC carrying GM-CSF, thereby enhancing the anticancer activity of oHSV. Here we describe a bacterial artificial chromosome-based method for construction of an oHSV expressing a transgene, which we routinely use in the laboratory to create a number of different recombinant oHSV bearing either therapeutic or reporter genes.
Collapse
Affiliation(s)
- Andranik Kahramanian
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Toshihiko Kuroda
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
PTEN expression by an oncolytic herpesvirus directs T-cell mediated tumor clearance. Nat Commun 2018; 9:5006. [PMID: 30479334 PMCID: PMC6258708 DOI: 10.1038/s41467-018-07344-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/22/2018] [Indexed: 12/19/2022] Open
Abstract
Engineered oncolytic viruses are used clinically to destroy cancer cells and have the ability to boost anticancer immunity. Phosphatase and tensin homolog deleted on chromosome 10 loss is common across a broad range of malignancies, and is implicated in immune escape. The N-terminally extended isoform, phosphatase and tensin homolog deleted on chromosome 10 alpha (PTENα), regulates cellular functions including protein kinase B signaling and mitochondrial adenosine triphosphate production. Here we constructed HSV-P10, a replicating, PTENα expressing oncolytic herpesvirus, and demonstrate that it inhibits PI3K/AKT signaling, increases cellular adenosine triphosphate secretion, and reduces programmed death-ligand 1 expression in infected tumor cells, thus priming an adaptive immune response and overcoming tumor immune escape. A single dose of HSV-P10 resulted in long term survivors in mice bearing intracranial tumors, priming anticancer T-cell immunity leading to tumor rejection. This implicates HSV-P10 as an oncolytic and immune stimulating therapeutic for anticancer therapy. Oncolytic viruses are a promising therapeutic approach for cancer treatment. The authors demonstrate the efficacy of an engineered HSV-1 expressing PTENα as an oncolytic and immune stimulating therapy against brain cancer metastases.
Collapse
|
29
|
Kim Y, Kang H, Powathil G, Kim H, Trucu D, Lee W, Lawler S, Chaplain M. Role of extracellular matrix and microenvironment in regulation of tumor growth and LAR-mediated invasion in glioblastoma. PLoS One 2018; 13:e0204865. [PMID: 30286133 PMCID: PMC6171904 DOI: 10.1371/journal.pone.0204865] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
The cellular dispersion and therapeutic control of glioblastoma, the most aggressive type of primary brain cancer, depends critically on the migration patterns after surgery and intracellular responses of the individual cancer cells in response to external biochemical cues in the microenvironment. Recent studies have shown that miR-451 regulates downstream molecules including AMPK/CAB39/MARK and mTOR to determine the balance between rapid proliferation and invasion in response to metabolic stress in the harsh tumor microenvironment. Surgical removal of the main tumor is inevitably followed by recurrence of the tumor due to inaccessibility of dispersed tumor cells in normal brain tissue. In order to address this complex process of cell proliferation and invasion and its response to conventional treatment, we propose a mathematical model that analyzes the intracellular dynamics of the miR-451-AMPK- mTOR-cell cycle signaling pathway within a cell. The model identifies a key mechanism underlying the molecular switches between proliferative phase and migratory phase in response to metabolic stress in response to fluctuating glucose levels. We show how up- or down-regulation of components in these pathways affects the key cellular decision to infiltrate or proliferate in a complex microenvironment in the absence and presence of time delays and stochastic noise. Glycosylated chondroitin sulfate proteoglycans (CSPGs), a major component of the extracellular matrix (ECM) in the brain, contribute to the physical structure of the local brain microenvironment but also induce or inhibit glioma invasion by regulating the dynamics of the CSPG receptor LAR as well as the spatiotemporal activation status of resident astrocytes and tumor-associated microglia. Using a multi-scale mathematical model, we investigate a CSPG-induced switch between invasive and non-invasive tumors through the coordination of ECM-cell adhesion and dynamic changes in stromal cells. We show that the CSPG-rich microenvironment is associated with non-invasive tumor lesions through LAR-CSGAG binding while the absence of glycosylated CSPGs induce the critical glioma invasion. We illustrate how high molecular weight CSPGs can regulate the exodus of local reactive astrocytes from the main tumor lesion, leading to encapsulation of non-invasive tumor and inhibition of tumor invasion. These different CSPG conditions also change the spatial profiles of ramified and activated microglia. The complex distribution of CSPGs in the tumor microenvironment can determine the nonlinear invasion behaviors of glioma cells, which suggests the need for careful therapeutic strategies.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
- Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
| | - Hyunji Kang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Gibin Powathil
- Department of Mathematics, Swansea University, Swansea, United Kingdom
| | - Hyeongi Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Dumitru Trucu
- Division of Mathematics, University of Dundee, Dundee, United Kingdom
| | - Wanho Lee
- National Institute for Mathematical Sciences, Daejeon, Republic of Korea
| | - Sean Lawler
- Department of neurosurgery, Brigham and Women’s Hospital & Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mark Chaplain
- School of Mathematics and Statistics, Mathematical Institute, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
30
|
Kloker LD, Yurttas C, Lauer UM. Three-dimensional tumor cell cultures employed in virotherapy research. Oncolytic Virother 2018; 7:79-93. [PMID: 30234074 PMCID: PMC6130269 DOI: 10.2147/ov.s165479] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Oncolytic virotherapy constitutes an upcoming alternative treatment option for a broad spectrum of cancer entities. However, despite great research efforts, there is still only a single US Food and Drug Administration/European Medicines Agency-approved oncolytic virus available for clinical use. One reason for that is the gap between promising preclinical data and limited clinical success. Since oncolytic viruses are biological agents, they might require more realistic in vitro tumor models than common monolayer tumor cell cultures to provide meaningful predictive preclinical evaluation results. For more realistic invitro tumor models, three-dimensional tumor cell-culture systems can be employed in preclinical virotherapy research. This review provides an overview of spheroid and hydrogel tumor cell cultures, organotypic tumor-tissue slices, organotypic raft cultures, and tumor organoids utilized in the context of oncolytic virotherapy. Furthermore, we also discuss advantages, disadvantages, techniques, and difficulties of these three-dimensional tumor cell-culture systems when applied specifically in virotherapy research.
Collapse
Affiliation(s)
- Linus D Kloker
- Department of Clinical Tumor Biology, University Hospital, University of Tübingen, Tübingen, Germany,
| | - Can Yurttas
- Department of General, Visceral and Transplant Surgery, University Hospital, University of Tübingen, Tübingen, Germany
| | - Ulrich M Lauer
- Department of Clinical Tumor Biology, University Hospital, University of Tübingen, Tübingen, Germany, .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Tübingen, Germany,
| |
Collapse
|
31
|
Liao WC, Liao CK, Tsai YH, Tseng TJ, Chuang LC, Lan CT, Chang HM, Liu CH. DSE promotes aggressive glioma cell phenotypes by enhancing HB-EGF/ErbB signaling. PLoS One 2018; 13:e0198364. [PMID: 29864158 PMCID: PMC5986151 DOI: 10.1371/journal.pone.0198364] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/17/2018] [Indexed: 01/23/2023] Open
Abstract
Remodeling of the extracellular matrix (ECM) in the tumor microenvironment promotes glioma progression. Chondroitin sulfate (CS) proteoglycans appear in the ECM and on the cell surface, and can be catalyzed by dermatan sulfate epimerase to form chondroitin sulfate/dermatan sulfate (CS/DS) hybrid chains. Dermatan sulfate epimerase 1 (DSE) is overexpressed in many types of cancer, and CS/DS chains mediate several growth factor signals. However, the role of DSE in gliomas has never been explored. In the present study, we determined the expression of DSE in gliomas by consulting a public database and conducting immunohistochemistry on a tissue array. Our investigation revealed that DSE was upregulated in gliomas compared with normal brain tissue. Furthermore, high DSE expression was associated with advanced tumor grade and poor survival. We found high DSE expression in several glioblastoma cell lines, and DSE expression directly mediated DS chain formation in glioblastoma cells. Knockdown of DSE suppressed the proliferation, migration, and invasion of glioblastoma cells. In contrast, overexpression of DSE in GL261 cells enhanced these malignant phenotypes and in vivo tumor growth. Interestingly, we found that DSE selectively regulated heparin-binding EGF-like growth factor (HB-EGF)-induced signaling in glioblastoma cells. Inhibiting epidermal growth factor receptor (EGFR) and ErbB2 with afatinib suppressed DSE-enhanced malignant phenotypes, establishing the critical role of the ErbB pathway in regulating the effects of DSE expression. This evidence indicates that upregulation of DSE in gliomas contributes to malignant behavior in cancer cells. We provide novel insight into the significance of DS chains in ErbB signaling and glioma pathogenesis.
Collapse
Affiliation(s)
- Wen-Chieh Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Kai Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - You-Huan Tsai
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - To-Jung Tseng
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Li-Ching Chuang
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chyn-Tair Lan
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hung-Ming Chang
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chiung-Hui Liu
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
32
|
Jaime-Ramirez AC, Dmitrieva N, Yoo JY, Banasavadi-Siddegowda Y, Zhang J, Relation T, Bolyard C, Wojton J, Kaur B. Humanized chondroitinase ABC sensitizes glioblastoma cells to temozolomide. J Gene Med 2018; 19. [PMID: 28087981 DOI: 10.1002/jgm.2942] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/06/2017] [Accepted: 01/08/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Malignant gliomas (glioblastomas; GBMs) are extremely aggressive and have a median survival of approximately 15 months. Current treatment modalities, which include surgical resection, radiation and chemotherapy, have done little to prolong the lives of GBM patients. Chondroitin sulfate proteoglycans (CSPG) are critical for cell-cell and cell-extracellular matrix (ECM) interactions and are implicated in glioma growth and invasion. Chondroitinase (Chase) ABC is a bacterial enzyme that cleaves chondroitin sulfate disaccharide chains from CSPGs in the tumor ECM. Wild-type Chase ABC has limited stability and/or activity in mammalian cells; therefore, we created a mutant humanized version (Chase M) with enhanced function in mammalian cells. METHODS We hypothesized that disruption of cell-cell and cell-ECM interactions by ChaseM and temozolomide (TMZ) will enhance the chemotherapeutic availability and sensitivity of glioma cells. RESULTS Utilizing primary patient-derived neurospheres, we found that ChaseM decreases glioma neurosphere aggregation in vitro. Furthermore, an oncolytic HSV-1 virus expressing secreted ChaseM (OV-ChaseM) enhanced viral spread and glioma cell killing compared to OV-Control, in vitro. OV-ChaseM plus TMZ combinatorial treatment resulted in a significant synergistic enhancement of glioma cell killing accompanied by an increase in apoptotic cell death. Intracellular flow cytometric analysis revealed a significant reduction in the phosphorylation of the pro-survival AKT protein following OV-ChaseM plus TMZ treatment. Lastly, in nude mice bearing intracranial GBM30 glioma xenografts, intratumoral OV-ChaseM plus TMZ (10 mg/kg by oral gavage) combination therapy resulted in a significant (p < 0.02) enhancement of survival compared to each individual treatment alone. CONCLUSIONS These data reveal that OV-ChaseM enhances glioma cell viral susceptibility and sensitivity to TMZ.
Collapse
Affiliation(s)
- Alena Cristina Jaime-Ramirez
- Department of Neurological Surgery, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| | - Nina Dmitrieva
- Department of Neurological Surgery, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| | - Ji Young Yoo
- Department of Neurological Surgery, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| | - Yeshavanth Banasavadi-Siddegowda
- Department of Neurological Surgery, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| | - Jianying Zhang
- Center for Biostatistics Biomedical Informatics Department, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| | - Theresa Relation
- Neuroscience Graduate Program and The Medical Scientist Training Program, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| | - Chelsea Bolyard
- Department of Neurological Surgery, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| | - Jeffrey Wojton
- Department of Neurological Surgery, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| | - Balveen Kaur
- Department of Neurological Surgery, The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital, and Solove Research Institute, Columbus, OH, USA
| |
Collapse
|
33
|
Liu CH, Lan CT, Chou JF, Tseng TJ, Liao WC. CHSY1 promotes aggressive phenotypes of hepatocellular carcinoma cells via activation of the hedgehog signaling pathway. Cancer Lett 2017; 403:280-288. [PMID: 28652022 DOI: 10.1016/j.canlet.2017.06.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/29/2022]
Abstract
Abnormal expression of chondroitin sulfate has been found in many types of cancer, while its biological functions in hepatocellular carcinoma (HCC) progression remain uninvestigated. Here, we report that chondroitin sulfate synthase 1 (CHSY1), the enzyme that mediates the polymerization step of chondroitin sulfate, is a critical mediator of malignant character in HCC that acts via modulating the activity of the hedgehog signaling. CHSY1 was up-regulated frequently in HCC where these events were associated with worse histologic grade and poor survival. Enforced expression of CHSY1 was sufficient to enhance cell growth, migration, invasion, and epithelial-mesenchymal transition, whereas silencing of CHSY1 suppressed these malignant phenotypes. Mechanistic investigations revealed that the increase of cell surface chondroitin sulfate by CHSY1 promoted sonic hedgehog binding and signaling. Inhibiting hedgehog pathway with vismodegib decreased CHSY1-induced migration, invasion, and lung metastasis of HCC cells, establishing the critical role of hedgehog signaling in mediating the effects of CHSY1 expression. Together, our results indicate that CHSY1 overexpression in HCC contributes to the malignant behaviors in cancer cells, we provide novel insights into the significance of chondroitin sulfate in hedgehog signaling and HCC pathogenesis.
Collapse
Affiliation(s)
- Chiung-Hui Liu
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan; Department of Medical Education, Chung Shan Medical University Hospital, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan
| | - Chyn-Tair Lan
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan; Department of Medical Education, Chung Shan Medical University Hospital, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan
| | - Jui-Feng Chou
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan
| | - To-Jung Tseng
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan; Department of Medical Education, Chung Shan Medical University Hospital, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan.
| | - Wen-Chieh Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan; Department of Medical Education, Chung Shan Medical University Hospital, No.110, Sec.1, Jianguo N. Rd, Taichung, Taiwan.
| |
Collapse
|
34
|
Bolyard C, Meisen WH, Banasavadi-Siddegowda Y, Hardcastle J, Yoo JY, Wohleb ES, Wojton J, Yu JG, Dubin S, Khosla M, Xu B, Smith J, Alvarez-Breckenridge C, Pow-Anpongkul P, Pichiorri F, Zhang J, Old M, Zhu D, Van Meir EG, Godbout JP, Caligiuri MA, Yu J, Kaur B. BAI1 Orchestrates Macrophage Inflammatory Response to HSV Infection-Implications for Oncolytic Viral Therapy. Clin Cancer Res 2016; 23:1809-1819. [PMID: 27852701 DOI: 10.1158/1078-0432.ccr-16-1818] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/04/2016] [Accepted: 10/27/2016] [Indexed: 01/10/2023]
Abstract
Purpose: Brain angiogenesis inhibitor (BAI1) facilitates phagocytosis and bacterial pathogen clearance by macrophages; however, its role in viral infections is unknown. Here, we examined the role of BAI1, and its N-terminal cleavage fragment (Vstat120) in antiviral macrophage responses to oncolytic herpes simplex virus (oHSV).Experimental Design: Changes in infiltration and activation of monocytic and microglial cells after treatment of glioma-bearing mice brains with a control (rHSVQ1) or Vstat120-expressing (RAMBO) oHSV was analyzed using flow cytometry. Co-culture of infected glioma cells with macrophages or microglia was used to examine antiviral signaling. Cytokine array gene expression and Ingenuity Pathway Analysis (IPA) helped evaluate changes in macrophage signaling in response to viral infection. TNFα-blocking antibodies and macrophages derived from Bai1-/- mice were used.Results: RAMBO treatment of mice reduced recruitment and activation of macrophages/microglia in mice with brain tumors, and showed increased virus replication compared with rHSVQ1. Cytokine gene expression array revealed that RAMBO significantly altered the macrophage inflammatory response to infected glioma cells via altered secretion of TNFα. Furthermore, we showed that BAI1 mediated macrophage TNFα induction in response to oHSV therapy. Intracranial inoculation of wild-type/RAMBO virus in Bai1-/- or wild-type non-tumor-bearing mice revealed the safety of this approach.Conclusions: We have uncovered a new role for BAI1 in facilitating macrophage anti-viral responses. We show that arming oHSV with antiangiogenic Vstat120 also shields them from inflammatory macrophage antiviral response, without reducing safety. Clin Cancer Res; 23(7); 1809-19. ©2016 AACR.
Collapse
Affiliation(s)
- Chelsea Bolyard
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - W Hans Meisen
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Yeshavanth Banasavadi-Siddegowda
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Jayson Hardcastle
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio
| | - Ji Young Yoo
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Eric S Wohleb
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Jeffrey Wojton
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio
| | - Jun-Ge Yu
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Otolaryngology, Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, Ohio
| | - Samuel Dubin
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Maninder Khosla
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Otolaryngology, Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, Ohio
| | - Bo Xu
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Jonathan Smith
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio
| | - Christopher Alvarez-Breckenridge
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Pete Pow-Anpongkul
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Flavia Pichiorri
- Department of Hematology, City of Hope Cancer Center, Duarte, California
| | - Jianying Zhang
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Matthew Old
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Otolaryngology, Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, Ohio
| | - Dan Zhu
- Departments of Neurosurgery and Hematology and Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Erwin G Van Meir
- Departments of Neurosurgery and Hematology and Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jonathan P Godbout
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Michael A Caligiuri
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Jianhua Yu
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Balveen Kaur
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio. .,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
35
|
Tedcastle A, Illingworth S, Brown A, Seymour LW, Fisher KD. Actin-resistant DNAse I Expression From Oncolytic Adenovirus Enadenotucirev Enhances Its Intratumoral Spread and Reduces Tumor Growth. Mol Ther 2016; 24:796-804. [PMID: 26708004 PMCID: PMC4886935 DOI: 10.1038/mt.2015.233] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 12/17/2015] [Indexed: 12/27/2022] Open
Abstract
Spread of oncolytic viruses through tumor tissue is essential to effective virotherapy. Interstitial matrix is thought to be a significant barrier to virus particle convection between "islands" of tumor cells. One way to address this is to encode matrix-degrading enzymes within oncolytic viruses, for secretion from infected cells. To test the hypothesis that extracellular DNA provides an important barrier, we assessed the ability of DNase to promote virus spread. Nonreplicating Ad5 vectors expressing actin-resistant DNase (aDNAse I), proteinase K (PK), hyaluronidase (rhPH20), and chondroitinase ABC (CABC) were injected into established DLD human colorectal adenocarcinoma xenografts, transcomplemented with a replicating Ad5 virus. Each enzyme improved oncolysis by the replicating adenovirus, with no evidence of tumor cells being shed into the bloodstream. aDNAse I and rhPH20 hyaluronidase were then cloned into conditionally-replicating group B adenovirus, Enadenotucirev (EnAd). EnAd encoding each enzyme showed significantly better antitumor efficacy than the parental virus, with the aDNAse I-expressing virus showing improved spread. Both DNase and hyaluronidase activity was still measurable 32 days postinfection. This is the first time that extracellular DNA has been implicated as a barrier for interstitial virus spread, and suggests that oncolytic viruses expressing aDNAse I may be promising candidates for clinical translation.
Collapse
Affiliation(s)
| | | | | | | | - Kerry D Fisher
- Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Hulou MM, Cho CF, Chiocca EA, Bjerkvig R. Experimental therapies: gene therapies and oncolytic viruses. HANDBOOK OF CLINICAL NEUROLOGY 2016; 134:183-197. [PMID: 26948355 DOI: 10.1016/b978-0-12-802997-8.00011-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Glioblastoma is the most common and aggressive primary brain tumor in adults. Over the past three decades, the overall survival time has only improved by a few months, therefore novel alternative treatment modalities are needed to improve clinical management strategies. Such strategies should ultimately extend patient survival. At present, the extensive insight into the molecular biology of gliomas, as well as into genetic engineering techniques, has led to better decision processes when it comes to modifying the genome to accommodate suicide genes, cytokine genes, and tumor suppressor genes that may kill cancer cells, and boost the host defensive immune system against neoantigenic cytoplasmic and nuclear targets. Both nonreplicative viral vectors and replicating oncolytic viruses have been developed for brain cancer treatment. Stem cells, microRNAs, nanoparticles, and viruses have also been designed. These have been armed with transgenes or peptides, and have been used both in laboratory-based experiments as well as in clinical trials, with the aim of improving selective killing of malignant glioma cells while sparing normal brain tissue. This chapter reviews the current status of gene therapies for malignant gliomas and highlights the most promising viral and cell-based strategies under development.
Collapse
Affiliation(s)
- M Maher Hulou
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Choi-Fong Cho
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - E Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rolf Bjerkvig
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg; Department of Biomedicine, University of Bergen, Norway
| |
Collapse
|
37
|
Scodeller P. Extracellular Matrix Degrading Enzymes for Nanocarrier-Based Anticancer Therapy. INTRACELLULAR DELIVERY III 2016. [DOI: 10.1007/978-3-319-43525-1_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
38
|
Kane JR, Miska J, Young JS, Kanojia D, Kim JW, Lesniak MS. Sui generis: gene therapy and delivery systems for the treatment of glioblastoma. Neuro Oncol 2015; 17 Suppl 2:ii24-ii36. [PMID: 25746089 DOI: 10.1093/neuonc/nou355] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Gene therapy offers a multidimensional set of approaches intended to treat and cure glioblastoma (GBM), in combination with the existing standard-of-care treatment (surgery and chemoradiotherapy), by capitalizing on the ability to deliver genes directly to the site of neoplasia to yield antitumoral effects. Four types of gene therapy are currently being investigated for their potential use in treating GBM: (i) suicide gene therapy, which induces the localized generation of cytotoxic compounds; (ii) immunomodulatory gene therapy, which induces or augments an enhanced antitumoral immune response; (iii) tumor-suppressor gene therapy, which induces apoptosis in cancer cells; and (iv) oncolytic virotherapy, which causes the lysis of tumor cells. The delivery of genes to the tumor site is made possible by means of viral and nonviral vectors for direct delivery of therapeutic gene(s), tumor-tropic cell carriers expressing therapeutic gene(s), and "intelligent" carriers designed to increase delivery, specificity, and tumoral toxicity against GBM. These vehicles are used to carry genetic material to the site of pathology, with the expectation that they can provide specific tropism to the desired site while limiting interaction with noncancerous tissue. Encouraging preclinical results using gene therapies for GBM have led to a series of human clinical trials. Although there is limited evidence of a therapeutic benefit to date, a number of clinical trials have convincingly established that different types of gene therapies delivered by various methods appear to be safe. Due to the flexibility of specialized carriers and genetic material, the technology for generating new and more effective therapies already exists.
Collapse
Affiliation(s)
- J Robert Kane
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Jason Miska
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Jacob S Young
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Deepak Kanojia
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Julius W Kim
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Maciej S Lesniak
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| |
Collapse
|
39
|
Hu L, Sun S, Wang T, Li Y, Jiang K, Lin G, Ma Y, Barr MP, Song F, Zhang G, Meng S. Oncolytic newcastle disease virus triggers cell death of lung cancer spheroids and is enhanced by pharmacological inhibition of autophagy. Am J Cancer Res 2015; 5:3612-3623. [PMID: 26885450 PMCID: PMC4731635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 11/07/2015] [Indexed: 06/05/2023] Open
Abstract
Lung cancer stem cells (CSCs) have recently been isolated from lung cancer patient samples and have been reported to be responsible for tumor initiation, treatment resistance and tumor recurrence. We have previously shown that oncolytic Newcastle disease virus (NDV), strain FMW (NDV/FMW) induces apoptosis in drug-resistant lung cancer cells. However, how NDV exerts its oncolytic effect on lung CSCs remains to be investigated. Here we show that NDV/FMW replicates in, and lyses CSC-enriched lung cancer spheroids and inhibits the 3D growth potential of lung cancer spheroid and agar colonies. We demonstrate that NDV/FMW triggers caspase-dependent apoptosis in lung cancer spheroids as shown by increased caspase-3 processing and Poly (ADP-ribose) polymerase (PARP) cleavage. Notably, NDV/FMW infection results in the degradation of microtubule-associated protein 1 light chain 3 (LC3) II and P62, two hallmarks of autophagy maturation, indicating that NDV/FMW promotes autophagy flux in lung cancer cell spheroids. This was further confirmed by the appearance of an increased number of double-membrane vesicles as detected by transmission electron microscopy. We also show that NDV/FMW promotes autophagy degradation in lung cancer spheroids via inhibition of the AKT/mTOR pathway. In addition, treatment of spheroids with the autophagy inhibitor, chloroquine increases NDV/FMW-induced cytotoxicity. Collectively, our data show that oncolytic NDV/FMW may be a potential strategy in targeting lung CSCs.
Collapse
Affiliation(s)
- Lulu Hu
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical UniversityDalian 116044, China
| | - Sulan Sun
- Biotherapy Research Center, Liaoning Cancer Hospital & Institute44 Xiaoheyan Road, Shenyang 110042, China
| | - Tianpeng Wang
- Department of Medical Image, Liaoning Cancer Hospital44 Xiaoheyan Road, Shenyang 110042, China
| | - Yingchun Li
- Biotherapy Research Center, Liaoning Cancer Hospital & Institute44 Xiaoheyan Road, Shenyang 110042, China
| | - Ke Jiang
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical UniversityDalian 116044, China
| | - Guibin Lin
- College of Bioscience and Biotechnology, Yangzhou University48 East, Wenhui Road, Yangzhou 225009, China
| | - Yan Ma
- School of Veterinary Medicine, Yangzhou University48 East, Wenhui Road, Yangzhou 225009, Jiangsu, China
| | - Martin P Barr
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences St. James’s Hospital & Trinity College DublinDublin, Ireland
| | - Fei Song
- Department of Neurosurgery, Second Affiliated Hospital, Dalian Medical UniversityDalian 116044, China
| | - Guirong Zhang
- Biotherapy Research Center, Liaoning Cancer Hospital & Institute44 Xiaoheyan Road, Shenyang 110042, China
| | - Songshu Meng
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical UniversityDalian 116044, China
| |
Collapse
|
40
|
Pediatric cancer gone viral. Part I: strategies for utilizing oncolytic herpes simplex virus-1 in children. MOLECULAR THERAPY-ONCOLYTICS 2015; 2:S2372-7705(16)30017-1. [PMID: 26436135 PMCID: PMC4589755 DOI: 10.1038/mto.2015.15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Progress for improving outcomes in pediatric patients with solid tumors remains slow. In addition, currently available therapies are fraught with numerous side effects, often causing significant life-long morbidity for long-term survivors. The use of viruses to kill tumor cells based on their increased vulnerability to infection is gaining traction, with several viruses moving through early and advanced phase clinical testing. The prospect of increased efficacy and decreased toxicity with these agents is thus attractive for pediatric cancer. In part I of this two-part review, we focus on strategies for utilizing oncolytic engineered herpes simplex virus (HSV) to target pediatric malignancies. We discuss mechanisms of action, routes of delivery, and the role of preexisting immunity on antitumor efficacy. Challenges to maximizing oncolytic HSV in children are examined, and we highlight how these may be overcome through various arming strategies. We review the preclinical and clinical evidence demonstrating safety of a variety of oncolytic HSVs. In Part II, we focus on the antitumor efficacy of oncolytic HSV in pediatric tumor types, pediatric clinical advances made to date, and future prospects for utilizing HSV in pediatric patients with solid tumors.
Collapse
|
41
|
Peters C, Rabkin SD. Designing Herpes Viruses as Oncolytics. MOLECULAR THERAPY-ONCOLYTICS 2015; 2:S2372-7705(16)30012-2. [PMID: 26462293 PMCID: PMC4599707 DOI: 10.1038/mto.2015.10] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oncolytic herpes simplex virus (oHSV) was one of the first genetically-engineered oncolytic viruses. Because herpes simplex virus (HSV) is a natural human pathogen that can cause serious disease, it is incumbent that it be genetically-engineered or significantly attenuated for safety. Here we present a detailed explanation of the functions of HSV-1 genes frequently mutated to endow oncolytic activity. These genes are non-essential for growth in tissue culture cells but are important for growth in post-mitotic cells, interfering with intrinsic antiviral and innate immune responses or causing pathology, functions dispensable for replication in cancer cells. Understanding the function of these genes leads to informed creation of new oHSVs with better therapeutic efficacy. Virus infection and replication can also be directed to cancer cells through tumor-selective receptor binding and transcriptional- or post-transcriptional miRNA-targeting, respectively. In addition to the direct effects of oHSV on infected cancer cells and tumors, oHSV can be 'armed' with transgenes that are: reporters, to track virus replication and spread; cytotoxic, to kill uninfected tumor cells; immune modulatory, to stimulate anti-tumor immunity; or tumor microenvironment altering, to enhance virus spread or to inhibit tumor growth. In addition to HSV-1, other alphaherpesviruses are also discussed for their oncolytic activity.
Collapse
Affiliation(s)
- Cole Peters
- Program in Virology, Harvard Medical School, Boston, MA, and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston MA
| | - Samuel D Rabkin
- Program in Virology, Harvard Medical School, Boston, MA, and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston MA
| |
Collapse
|
42
|
Kaufmann JK, Chiocca EA. Oncolytic virotherapy for gliomas: steps toward the future. CNS Oncol 2015; 2:389-92. [PMID: 25054659 DOI: 10.2217/cns.13.35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Johanna K Kaufmann
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham & Women's Hospital, Harvard Medical School, 4 Blackfan Circle, Harvard Institutes of Medicine, Room 914, Boston, MA 02115, USA
| | | |
Collapse
|
43
|
Kanai R, Rabkin SD. Combinatorial strategies for oncolytic herpes simplex virus therapy of brain tumors. CNS Oncol 2015; 2:129-42. [PMID: 23687568 DOI: 10.2217/cns.12.42] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Oncolytic viruses, such as the oncolytic herpes simplex virus (oHSV), are an exciting new therapeutic strategy for cancer as they are replication competent in tumor cells but not normal cells. In order to engender herpes simplex virus with oncolytic activity and make it safe for clinical application, mutations are engineered into the virus. Glioblastoma multiforme (GBM) is the most common and deadly primary brain tumor in adults. Despite many advances in therapy, overall survival has not been substantially improved over the last several decades. A number of different oHSVs have been tested as monotherapy in early-phase clinical trials for GBM and have demonstrated safety and anecdotal evidence of efficacy. However, strategies to improve efficacy are likely to be necessary to successfully treat GBM. Cancer treatment usually involves multimodal approaches, so the standard of care for GBM includes surgery, radiotherapy and chemotherapy. In preclinical GBM models, combinations of oHSV with other types of therapy have exhibited markedly improved activity over individual treatments alone. In this review, we will discuss the various combination strategies that have been employed with oHSV, including chemotherapy, small-molecule inhibitors, antiangiogenic agents, radiotherapy and expression of therapeutic transgenes. Effective combinations, especially synergistic ones, are clinically important not just for improved efficacy but also to permit lower and less-toxic doses and potentially overcome resistance.
Collapse
|
44
|
Jacobsen K, Russell L, Kaur B, Friedman A. Effects of CCN1 and Macrophage Content on Glioma Virotherapy: A Mathematical Model. Bull Math Biol 2015; 77:984-1012. [DOI: 10.1007/s11538-015-0074-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 03/02/2015] [Indexed: 12/14/2022]
|
45
|
Ding J. Oncolytic virus as a cancer stem cell killer: progress and challenges. Stem Cell Investig 2014; 1:22. [PMID: 27358868 DOI: 10.3978/j.issn.2306-9759.2014.12.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 12/21/2014] [Indexed: 01/15/2023]
Abstract
Oncolytic viruses (OVs), which were discovered more than one century ago, have been used in multiple clinical trials for cancer therapy. OVs specifically target cancer cells when sparing normal cells by exploiting biochemical differences between normal and tumor cells. Hence oncolytic virotherapy is more specific at targeting cancer cells compared with conventional anti-cancer therapy. Apart from the lack of specificity, conventional anti-cancer therapies also often witness relapse and incomplete cure of cancer. One hypothesis explaining this phenomenon is that a subpopulation of cancer cells, known as cancer stem cells (CSCs), are resistant to conventional therapies, possibly due to its self-renewal and differentiation abilities. With the discovery of CSCs, researchers have been trying to explain whether OVs are well suited to eliminate CSCs. Two explanations for postulating OVs as ideal candidates for cancer therapy have been proposed: first, OVs are not subject to the same mechanisms responsible for chemotherapy and radiation resistance; second, viruses could be harnessed to express therapeutic transgenes that specifically target the features unique to CSCs or the properties CSCs rely on for self-renewal and differentiation. Indeed, initial studies suggest that OVs could effectively target CSCs in multiple tumor types. The focus of this review is to highlight recent studies related to the application of OVs on targeting CSCs, based on which, the challenges and perspectives for further research in this field will also be discussed.
Collapse
Affiliation(s)
- Jingzhen Ding
- Department of Cellular and Molecular Medicine, Howard Hughes Medical Institute, University of California at San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
46
|
Rodríguez-García A, Giménez-Alejandre M, Rojas JJ, Moreno R, Bazan-Peregrino M, Cascalló M, Alemany R. Safety and efficacy of VCN-01, an oncolytic adenovirus combining fiber HSG-binding domain replacement with RGD and hyaluronidase expression. Clin Cancer Res 2014; 21:1406-18. [PMID: 25391696 DOI: 10.1158/1078-0432.ccr-14-2213] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor targeting upon intravenous administration and subsequent intratumoral virus dissemination are key features to improve oncolytic adenovirus therapy. VCN-01 is a novel oncolytic adenovirus that combines selective replication conditional to pRB pathway deregulation, replacement of the heparan sulfate glycosaminoglycan putative-binding site KKTK of the fiber shaft with an integrin-binding motif RGDK for tumor targeting, and expression of hyaluronidase to degrade the extracellular matrix. In this study, we evaluate the safety and efficacy profile of this novel oncolytic adenovirus. EXPERIMENTAL DESIGN VCN-01 replication and potency were assessed in a panel of tumor cell lines. VCN-01 tumor-selective replication was evaluated in human fibroblasts and pancreatic islets. Preclinical toxicity, biodistribution, and efficacy studies were conducted in mice and Syrian hamsters. RESULTS Toxicity and biodistribution preclinical studies support the selectivity and safety of VCN-01. Antitumor activity after intravenous or intratumoral administration of the virus was observed in all tumor models tested, including melanoma and pancreatic adenocarcinoma, both in immunodeficient mice and immunocompetent hamsters. CONCLUSIONS Oncolytic adenovirus VCN-01 characterized by the expression of hyaluronidase and the RGD shaft retargeting ligand shows an efficacy-toxicity prolife in mice and hamsters by intravenous and intratumoral administration that warrants clinical testing.
Collapse
Affiliation(s)
- Alba Rodríguez-García
- Translational Research Laboratory, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Juan J Rojas
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rafael Moreno
- Translational Research Laboratory, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Manel Cascalló
- VCN Biosciences, Sant Cugat del Vallès, Barcelona, Spain
| | - Ramon Alemany
- Translational Research Laboratory, IDIBELL-Institut Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
47
|
Abstract
Even after 20 years of granting orphan status for chondroitinase by US FDA, there is no visible outcome in terms of clinical use. The reasons are many. One of them could be lack of awareness regarding the biological application of the enzyme. The biological activity of chondroitinase is due to its ability to act on chondroitin sulfate proteoglycans (CSPGs). CSPGs are needed for normal functioning of the body. An increase or decrease in the level of CSPGs results in various pathological conditions. Chondroitinase is useful in conditions where there is an increase in the level of CSPGs, namely spinal cord injury, vitreous attachment and cancer. Over the last decade, various animal studies showed that chondroitinase could be a good drug candidate. Research focusing on developing a suitable carrier system for delivering chondroitinase needs to be carried out so that pharmacological activity observed in vitro and preclinical studies could be translated to clinical use. Further studies on distribution of chondroitinase as well need to be focused so that chondroitinase with desired attributes could be discovered. The present review article discusses about various biological applications of chondroitinase, drug delivery systems to deliver the enzyme and distribution of chondroitinase among microbes.
Collapse
Affiliation(s)
- Narayanan Kasinathan
- a Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal College of Pharmaceutical Sciences, Manipal University , Manipal , Karnataka , India
| | - Subrahmanyam M Volety
- a Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal College of Pharmaceutical Sciences, Manipal University , Manipal , Karnataka , India
| | - Venkata Rao Josyula
- a Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal College of Pharmaceutical Sciences, Manipal University , Manipal , Karnataka , India
| |
Collapse
|
48
|
Kim Y, Lee HG, Dmitrieva N, Kim J, Kaur B, Friedman A. Choindroitinase ABC I-mediated enhancement of oncolytic virus spread and anti tumor efficacy: a mathematical model. PLoS One 2014; 9:e102499. [PMID: 25047810 PMCID: PMC4105445 DOI: 10.1371/journal.pone.0102499] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 06/18/2014] [Indexed: 12/23/2022] Open
Abstract
Oncolytic viruses are genetically engineered viruses that are designed to kill cancer cells while doing minimal damage to normal healthy tissue. After being injected into a tumor, they infect cancer cells, multiply inside them, and when a cancer cell is killed they move on to spread and infect other cancer cells. Chondroitinase ABC (Chase-ABC) is a bacterial enzyme that can remove a major glioma ECM component, chondroitin sulfate glycosoamino glycans from proteoglycans without any deleterious effects in vivo. It has been shown that Chase-ABC treatment is able to promote the spread of the viruses, increasing the efficacy of the viral treatment. In this paper we develop a mathematical model to investigate the effect of the Chase-ABC on the treatment of glioma by oncolytic viruses (OV). We show that the model's predictions agree with experimental results for a spherical glioma. We then use the model to test various treatment options in the heterogeneous microenvironment of the brain. The model predicts that separate injections of OV, one into the center of the tumor and another outside the tumor will result in better outcome than if the total injection is outside the tumor. In particular, the injection of the ECM-degrading enzyme (Chase-ABC) on the periphery of the main tumor core need to be administered in an optimal strategy in order to infect and eradicate the infiltrating glioma cells outside the tumor core in addition to proliferative cells in the bulk of tumor core. The model also predicts that the size of tumor satellites and distance between the primary tumor and multifocal/satellite lesions may be an important factor for the efficacy of the viral therapy with Chase treatment.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Ohio State University, Columbus, Ohio, United States of America; Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| | - Hyun Geun Lee
- Department of Mathematics, Korea University, Seoul, Republic of Korea
| | - Nina Dmitrieva
- Department of Neurological Surgery, Ohio State University, Columbus, Ohio, United States of America
| | - Junseok Kim
- Department of Mathematics, Korea University, Seoul, Republic of Korea
| | - Balveen Kaur
- Department of Neurological Surgery, Ohio State University, Columbus, Ohio, United States of America
| | - Avner Friedman
- Department of Mathematics, Ohio State University, Columbus, Ohio, United States of America; Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
49
|
Abstract
Despite extensive research, current glioma therapies are still unsatisfactory, and novel approaches are pressingly needed. In recent years, both nonreplicative viral vectors and replicating oncolytic viruses have been developed for brain cancer treatment, and the mechanistic background of their cytotoxicity has been unveiled. A growing number of clinical trials have convincingly established viral therapies to be safe in glioma patients, and maximum tolerated doses have generally not been reached. However, evidence for therapeutic benefit has been limited: new generations of therapeutic vectors need to be developed in order to target not only tumor cells but also the complex surrounding microenvironment. Such therapies could also direct long-lasting immune responses toward the tumor while reducing early antiviral reactions. Furthermore, viral delivery methods are to be improved and viral spread within the tumor will have to be enhanced. Here, we will review the outcome of completed glioma virus therapy trials as well as highlight the ongoing clinical activities. On this basis, we will give an overview of the numerous strategies to enhance therapeutic efficacy of new-generation viruses and novel treatment regimens. Finally, we will conclude with approaches that may be crucial to the development of successful glioma therapies in the future.
Collapse
Affiliation(s)
| | - E. Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Calpain-dependent clearance of the autophagy protein p62/SQSTM1 is a contributor to ΔPK oncolytic activity in melanoma. Gene Ther 2014; 21:371-8. [PMID: 24553345 PMCID: PMC3975656 DOI: 10.1038/gt.2014.6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 12/17/2013] [Accepted: 01/06/2014] [Indexed: 02/07/2023]
Abstract
Oncolytic virotherapy is a promising strategy to reduce tumor burden through selective virus replication in rapidly proliferating cells. However, the lysis of slowly replicating cancer stem cells (CSC), which maintain neoplastic clonality, is relatively modest and the potential contribution of programmed cell death (PCD) pathways to oncolytic activity is still poorly understood. We show that the oncolytic virus ΔPK lyses CSC-enriched breast cancer and melanoma 3D spheroid cultures at low titers (0.1pfu/cell) and without resistance development and it inhibits the 3D growth potential (spheroids and agarose colonies) of melanoma and breast cancer cells. ΔPK induces calpain activation in both melanoma and breast cancer 3D cultures as determined by the loss of the p28 regulatory subunit, and 3D growth is restored by treatment with the calpain inhibitor PD150606. In melanoma, ΔPK infection also induces LC3-II accumulation and p62/SQSTM1 clearance, both markers of autophagy, and 3D growth is restored by treatment with the autophagy inhibitor chloroquine (CQ). However, expression of the autophagy-required protein Atg5 is not altered and CQ does not restore p62/SQSTM1 expression, suggesting that the CQ effect may be autophagy-independent. PD150606 restores expression of p62/SQSTM1 in ΔPK infected melanoma cultures, suggesting that calpain activation induces anti-tumor activity through p62/SQSTM1 clearance.
Collapse
|