1
|
Zhao JL, Lin BL, Luo C, Yi YL, Huang P, Chen Y, Zhao S, Huang ZJ, Ma XY, Huang L. Challenges and strategies toward oncolytic virotherapy for leptomeningeal metastasis. J Transl Med 2024; 22:1000. [PMID: 39501324 PMCID: PMC11539571 DOI: 10.1186/s12967-024-05794-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
Meningeal metastasis (LM) is commonly seen in the advanced stages of cancer patients, often leading to a rapid decline in survival time and quality of life. Currently, there is still a lack of standardized treatments. Oncolytic viruses (OVs) are a class of emerging cancer therapeutics with the advantages of selectively replicating in cancer cells, delivering various eukaryotic transgenes, inducing immunogenic cell death, and promoting anti-tumor immunity. Some studies applying OVs intrathoracically or intraperitoneally for the treatment of malignant pleural and peritoneal effusions have shown promising therapeutic effects. If OVs could be applied to treat LM, it would bring significant survival benefits to patients with LM. In this review, we analyzed past research on the use of viruses to treat meningeal metastasis, summarized the efficacy and safety demonstrated by the research results, and analyzed the feasibility of oncolytic virus therapy for meningeal metastasis. We also summarized the existing data to provide guidance for the development of OVs that can be injected into the cerebrospinal fluid (CSF).
Collapse
Affiliation(s)
- Jia-Li Zhao
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Bi-Lin Lin
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Chen Luo
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Yan-Ling Yi
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Peng Huang
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Yu Chen
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Sha Zhao
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Zhen-Jie Huang
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Xin-Yi Ma
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China
| | - Long Huang
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi, China.
| |
Collapse
|
2
|
Sakunchotpanit G, Patil MK, Venkatesh K, Rohan TZ, Cheng D, Nambudiri VE. Treatment of malignant melanoma with coxsackievirus A21 (V937): An emerging oncolytic virotherapy. Exp Dermatol 2024; 33:e15169. [PMID: 39207089 DOI: 10.1111/exd.15169] [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: 05/08/2024] [Revised: 08/06/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Despite rising melanoma incidence in recent decades, there is a trend towards overall decreased mortality, reflecting multiple factors including improved treatment options for metastatic disease. While local treatments are the mainstay for early-stage melanoma, metastatic disease necessitates systemic treatment, with oncolytic virotherapy emerging as a promising option. For this review, articles were retrieved from PubMed from 1964 through 2024. We conducted title, abstract and full-text screening in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to identify articles describing the use of coxsackievirus A21 (V937), either as monotherapy or as part of combination therapy for malignant melanoma. Fifteen articles met inclusion criteria, offering preclinical and clinical data on V937's efficacy in reducing tumour burden. In addition to reporting manageable safety profiles, clinical trial data examining intratumoral V937 combination therapy with pembrolizumab and ipilimumab also endorsed favourable objective response rates compared to immune checkpoint inhibitor monotherapy (47% vs. 38% and 21% vs. 10%, respectively). In contrast, intravenous V937 monotherapy failed to yield additional benefit in a cohort of patients with Stage IIIC/IV melanoma (n = 3) despite achieving detectable levels in tumour tissue (1 × 109 TCID50). Although small subsets of patients experienced severe adverse effects and study design limitations imposed constraints on collected data, evidence for the efficacy of V937 remains encouraging. With few clinical trials evaluating V937 in melanoma, additional data is required before routine usage in standard treatment for metastatic lesions.
Collapse
Affiliation(s)
- Goranit Sakunchotpanit
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Mihir K Patil
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Carle Illinois College of Medicine, Urbana, Illinois, USA
| | - Kaushik Venkatesh
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Z Rohan
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Debby Cheng
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Vinod E Nambudiri
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Swartz AR, Shieh Y, Gulasarian A, Curtis E, Hofmann CF, Baker JB, Templeton N, Olson JW. Glutathione affinity chromatography for the scalable purification of an oncolytic virus immunotherapy from microcarrier cell culture. Front Bioeng Biotechnol 2023; 11:1193454. [PMID: 37397964 PMCID: PMC10310922 DOI: 10.3389/fbioe.2023.1193454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Therapeutic viral vectors are an emerging technology with several clinical applications in gene therapy, vaccines, and immunotherapy. Increased demand has required the redevelopment of conventional, low-throughput cell culture and purification manufacturing methods such as static cell stacks and ultracentrifugation. In this work, scalable methods were investigated for the manufacture of an oncolytic virus immunotherapy application consisting of a prototype strain of coxsackievirus A21 (CVA21) produced in adherent MRC-5 cells. Cell culture was established in stirred-tank microcarrier bioreactors, and an efficient affinity chromatography method was developed for the purification of harvested CVA21 through binding of the viral capsids to an immobilized glutathione (GSH) ligand. Bioreactor temperature during infection was investigated to maximize titer, and a decrease in temperature from 37°C to 34°C yielded a two-three-fold increase in infectivity. After purification of the 34°C harvests, the GSH affinity chromatography elution not only maintained a >two-fold increase in infectivity and viral genomes but also increased the proportion of empty capsids compared to 37°C harvests. Using material generated from both infection temperature setpoints, chromatographic parameters and mobile phase compositions were studied at the laboratory scale to maximize infectious particle yields and cell culture impurity clearance. Empty capsids that co-eluted with full capsids from 34°C infection temperature harvests were poorly resolved across the conditions tested, but subsequent polishing anion exchange and cation exchange chromatography steps were developed to clear residual empty capsids and other impurities. Oncolytic CVA21 production was scaled-up 75-fold from the laboratory scale and demonstrated across seven batches in 250 L single-use microcarrier bioreactors and purified with customized, prepacked, single-use 1.5 L GSH affinity chromatography columns. The large-scale bioreactors controlled at 34°C during infection maintained a three-fold increase in productivity in the GSH elution, and excellent clearance of host cell and media impurities was observed across all batches. This study presents a robust method for the manufacture of an oncolytic virus immunotherapy application that may be implemented for the scalable production of other viruses and viral vectors which interact with glutathione.
Collapse
Affiliation(s)
- Andrew R. Swartz
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Yvonne Shieh
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Amanda Gulasarian
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Erik Curtis
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Carl F. Hofmann
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Jack B. Baker
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Neil Templeton
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Jessica W. Olson
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| |
Collapse
|
4
|
Das R, Langou S, Le TT, Prasad P, Lin F, Nguyen TD. Electrical Stimulation for Immune Modulation in Cancer Treatments. Front Bioeng Biotechnol 2022; 9:795300. [PMID: 35087799 PMCID: PMC8788921 DOI: 10.3389/fbioe.2021.795300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022] Open
Abstract
Immunotherapy is becoming a very common treatment for cancer, using approaches like checkpoint inhibition, T cell transfer therapy, monoclonal antibodies and cancer vaccination. However, these approaches involve high doses of immune therapeutics with problematic side effects. A promising approach to reducing the dose of immunotherapeutic agents given to a cancer patient is to combine it with electrical stimulation, which can act in two ways; it can either modulate the immune system to produce the immune cytokines and agents in the patient's body or it can increase the cellular uptake of these immune agents via electroporation. Electrical stimulation in form of direct current has been shown to reduce tumor sizes in immune-competent mice while having no effect on tumor sizes in immune-deficient mice. Several studies have used nano-pulsed electrical stimulations to activate the immune system and drive it against tumor cells. This approach has been utilized for different types of cancers, like fibrosarcoma, hepatocellular carcinoma, human papillomavirus etc. Another common approach is to combine electrochemotherapy with immune modulation, either by inducing immunogenic cell death or injecting immunostimulants that increase the effectiveness of the treatments. Several therapies utilize electroporation to deliver immunostimulants (like genes encoded with cytokine producing sequences, cancer specific antigens or fragments of anti-tumor toxins) more effectively. Lastly, electrical stimulation of the vagus nerve can trigger production and activation of anti-tumor immune cells and immune reactions. Hence, the use of electrical stimulation to modulate the immune system in different ways can be a promising approach to treat cancer.
Collapse
Affiliation(s)
- Ritopa Das
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Sofia Langou
- Department of Physiology and Neurobiology, University of Connecticut, Mansfield, CT, United States
| | - Thinh T. Le
- Department of Mechanical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Pooja Prasad
- Department of Cell and Molecular Biology, University of Connecticut, Mansfield, CT, United States
| | - Feng Lin
- Department of Mechanical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Thanh D. Nguyen
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
- Department of Mechanical Engineering, University of Connecticut, Mansfield, CT, United States
- Institute of Materials Science, University of Connecticut, Mansfield, CT, United States
| |
Collapse
|
5
|
Chis AA, Dobrea CM, Rus LL, Frum A, Morgovan C, Butuca A, Totan M, Juncan AM, Gligor FG, Arseniu AM. Dendrimers as Non-Viral Vectors in Gene-Directed Enzyme Prodrug Therapy. Molecules 2021; 26:5976. [PMID: 34641519 PMCID: PMC8512881 DOI: 10.3390/molecules26195976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 01/02/2023] Open
Abstract
Gene-directed enzyme prodrug therapy (GDEPT) has been intensively studied as a promising new strategy of prodrug delivery, with its main advantages being represented by an enhanced efficacy and a reduced off-target toxicity of the active drug. In recent years, numerous therapeutic systems based on GDEPT strategy have entered clinical trials. In order to deliver the desired gene at a specific site of action, this therapeutic approach uses vectors divided in two major categories, viral vectors and non-viral vectors, with the latter being represented by chemical delivery agents. There is considerable interest in the development of non-viral vectors due to their decreased immunogenicity, higher specificity, ease of synthesis and greater flexibility for subsequent modulations. Dendrimers used as delivery vehicles offer many advantages, such as: nanoscale size, precise molecular weight, increased solubility, high load capacity, high bioavailability and low immunogenicity. The aim of the present work was to provide a comprehensive overview of the recent advances regarding the use of dendrimers as non-viral carriers in the GDEPT therapy.
Collapse
Affiliation(s)
| | | | | | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (A.A.C.); (C.M.D.); (L.-L.R.); (A.B.); (M.T.); (A.M.J.); (F.G.G.); (A.M.A.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (A.A.C.); (C.M.D.); (L.-L.R.); (A.B.); (M.T.); (A.M.J.); (F.G.G.); (A.M.A.)
| | | | | | | | | | | |
Collapse
|
6
|
Perez C, Rico J, Guerrero C, Acosta O. Role of heat-shock proteins in infection of human adenocarcinoma cell line MCF-7 by tumor-adapted rotavirus isolates. COLOMBIA MEDICA (CALI, COLOMBIA) 2021; 52:e2024196. [PMID: 33911319 PMCID: PMC8054709 DOI: 10.25100/cm.v51i4.4196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background: Viruses are being used as alternative and complementary tools for treating cancers. Oncolytic viruses exhibit tumor tropism, ability to enhance anti-tumor immunity and ability to be used in combination with conventional chemotherapy and radiotherapy. We have recently selected some rotavirus isolates which are adapted to efficiently infect and kill tumor cell lines. Aim: We tested five tumor cell-adapted rotavirus isolates for their ability to infect the human adenocarcinoma cell line MCF-7. Methods: Cell surface membrane-associated proteins mediating virus particle attachment were characterized using ELISA, immunoprecipitation, FACS analysis, and antibody blocking. Results: It was found that heat shock proteins (HSPs) such as Hsp90, Hsp70, Hsp60, and Hsp40 are expressed on the cell surface forming complexes with protein disulfide isomerase (PDI), integrin β3, and heat shock cognate protein 70 (Hsc70) in lipid raft microdomains. Interaction of rotavirus isolates with these cellular proteins was further confirmed by a competition assay and an inhibition assay involving the HSPs tested. Conclusion: Our findings suggest that the tumor cell-adapted rotavirus isolates studied here offer a promising tool for killing tumor cells, thus encouraging further research into this topic, including animal models.
Collapse
Affiliation(s)
- Claudia Perez
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia. Universidad Nacional de Colombia Universidad Nacional de Colombia Faculty of Medicine Department of Physiological Sciences Bogota Colombia
| | - José Rico
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia. Universidad Nacional de Colombia Universidad Nacional de Colombia Faculty of Medicine Department of Physiological Sciences Bogota Colombia
| | - Carlos Guerrero
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia. Universidad Nacional de Colombia Universidad Nacional de Colombia Faculty of Medicine Department of Physiological Sciences Bogota Colombia
| | - Orlando Acosta
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia. Universidad Nacional de Colombia Universidad Nacional de Colombia Faculty of Medicine Department of Physiological Sciences Bogota Colombia
| |
Collapse
|
7
|
Franco-Luzón L, García-Mulero S, Sanz-Pamplona R, Melen G, Ruano D, Lassaletta Á, Madero L, González-Murillo Á, Ramírez M. Genetic and Immune Changes Associated with Disease Progression under the Pressure of Oncolytic Therapy in A Neuroblastoma Outlier Patient. Cancers (Basel) 2020; 12:cancers12051104. [PMID: 32354143 PMCID: PMC7281487 DOI: 10.3390/cancers12051104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/25/2020] [Accepted: 04/26/2020] [Indexed: 12/19/2022] Open
Abstract
Little is known about the effect of oncolytic adenovirotherapy on pediatric tumors. Here we present the clinical case of a refractory neuroblastoma that responded positively to Celyvir (ICOVIR-5 oncolytic adenovirus delivered by autologous mesenchymal stem cells) for several months. We analyzed samples during tumor evolution in order to identify molecular and mutational features that could explain the interactions between treatment and tumor and how the balance between both of them evolved. We identified a higher adaptive immune infiltration during stabilized disease compared to progression, and also a higher mutational rate and T-cell receptor (TCR) diversity during disease progression. Our results indicate an initial active role of the immune system controlling tumor growth during Celyvir therapy. The tumor eventually escaped from the control exerted by virotherapy through acquisition of resistance by the tumor microenvironment that exhausted the initial T cell response.
Collapse
Affiliation(s)
- Lidia Franco-Luzón
- Children Oncohematology Foundation, 28079 Madrid, Spain; (L.F.-L.); (L.M.)
| | - Sandra García-Mulero
- Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain;
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
| | - Rebeca Sanz-Pamplona
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
| | - Gustavo Melen
- Biomedical Research Foundation, Niño Jesús Children Hospital, 28009 Madrid, Spain; (G.M.); (Á.G.-M.)
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
| | - David Ruano
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
| | - Álvaro Lassaletta
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
| | - Luís Madero
- Children Oncohematology Foundation, 28079 Madrid, Spain; (L.F.-L.); (L.M.)
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
- Oncohematology Unit, Hospital Infantil Universitario Niño Jesús, 28009 Madrid, Spain
| | - África González-Murillo
- Biomedical Research Foundation, Niño Jesús Children Hospital, 28009 Madrid, Spain; (G.M.); (Á.G.-M.)
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
| | - Manuel Ramírez
- Biomedical Research Foundation, Niño Jesús Children Hospital, 28009 Madrid, Spain; (G.M.); (Á.G.-M.)
- La Princesa Institute of Health Research, 28006 Madrid, Spain; (D.R.); (Á.L.)
- Correspondence: ; Tel.: +34-9150-35938
| |
Collapse
|
8
|
Li L, Liu S, Han D, Tang B, Ma J. Delivery and Biosafety of Oncolytic Virotherapy. Front Oncol 2020; 10:475. [PMID: 32373515 PMCID: PMC7176816 DOI: 10.3389/fonc.2020.00475] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022] Open
Abstract
In recent years, oncolytic virotherapy has emerged as a promising anticancer therapy. Oncolytic viruses destroy cancer cells, without damaging normal tissues, through virus self-replication and antitumor immunity responses, showing great potential for cancer treatment. However, the clinical guidelines for administering oncolytic virotherapy remain unclear. Delivery routes for oncolytic virotherapy to patients vary in existing studies, depending on the tumor sites and the objective of studies. Moreover, the biosafety of oncolytic virotherapy, including mainly uncontrolled adverse events and long-term complications, remains a serious concern that needs to be accurately measured. This review provides a comprehensive and detailed overview of the delivery and biosafety of oncolytic virotherapy.
Collapse
Affiliation(s)
- Lizhi Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, China
| | - Shixin Liu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, China
| | - Duoduo Han
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, China
| | - Bin Tang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, China
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, National Health Commission Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, China
| |
Collapse
|
9
|
Hromic-Jahjefendic A, Lundstrom K. Viral Vector-Based Melanoma Gene Therapy. Biomedicines 2020; 8:E60. [PMID: 32187995 PMCID: PMC7148454 DOI: 10.3390/biomedicines8030060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
Gene therapy applications of oncolytic viruses represent an attractive alternative for cancer treatment. A broad range of oncolytic viruses, including adenoviruses, adeno-associated viruses, alphaviruses, herpes simplex viruses, retroviruses, lentiviruses, rhabdoviruses, reoviruses, measles virus, Newcastle disease virus, picornaviruses and poxviruses, have been used in diverse preclinical and clinical studies for the treatment of various diseases, including colon, head-and-neck, prostate and breast cancer as well as squamous cell carcinoma and glioma. The majority of studies have focused on immunotherapy and several drugs based on viral vectors have been approved. However, gene therapy for malignant melanoma based on viral vectors has not been utilized to its full potential yet. This review represents a summary of the achievements of preclinical and clinical studies using viral vectors, with the focus on malignant melanoma.
Collapse
Affiliation(s)
- Altijana Hromic-Jahjefendic
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | | |
Collapse
|
10
|
Abstract
Introduction: Immunotherapy has been introduced as a modern alternative for the treatment of various cancers, including the stimulation of the immune system by introduction of immunostimulatory molecules. Application of viral and non-viral vectors have provided a substantial contribution to improved delivery and expression of these immunostimulators.Areas covered: Alphavirus vectors, based on Semliki Forest virus, have allowed immunization with self-replicating RNA, recombinant virus particles, and layered DNA/RNA vectors. The attractive features of alphaviruses comprise their broad host range and extreme RNA replication in infected cells resulting in very high recombinant protein expression levels providing enhanced immune responses and an excellent basis for immunotherapy.Expert opinion: Immunization studies in animal tumor models have elicited strong humoral and cellular immune response, have provided prophylactic protection against tumor challenges, and have generated therapeutic efficacy in tumor-bearing animals. Clinical trials have indicated safe use of alphavirus vectors, making them attractive for cancer immunotherapy.
Collapse
|
11
|
Pérez C, Rico J, A. Guerrero C, Acosta O. Role of heat-shock proteins in infection of human adenocarcinoma cell line MCF-7 by tumor-adapted rotavirus isolates. Colomb Med (Cali) 2020. [DOI: 10.25100/cm.v52i1.4196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Background:
Viruses are being used as alternative and complementary tools for treating cancers. Oncolytic viruses exhibit tumor tropism, ability to enhance anti-tumor immunity and ability to be used in combination with conventional chemotherapy and radiotherapy. We have recently selected some rotavirus isolates which are adapted to efficiently infect and kill tumor cell lines.
Aim:
We tested five tumor cell-adapted rotavirus isolates for their ability to infect the human adenocarcinoma cell line MCF-7.
Methods:
Cell surface membrane-associated proteins mediating virus particle attachment were characterized using ELISA, immunoprecipitation, FACS analysis, and antibody blocking.
Results:
It was found that heat shock proteins (HSPs) such as Hsp90, Hsp70, Hsp60, and Hsp40 are expressed on the cell surface forming complexes with protein disulfide isomerase (PDI), integrin β3, and heat shock cognate protein 70 (Hsc70) in lipid raft microdomains. Interaction of rotavirus isolates with these cellular proteins was further confirmed by a competition assay and an inhibition assay involving the HSPs tested.
Conclusion:
Our findings suggest that the tumor cell-adapted rotavirus isolates studied here offer a promising tool for killing tumor cells, thus encouraging further research into this topic, including animal models.
Collapse
Affiliation(s)
- Claudia Pérez
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia
| | - José Rico
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia
| | - Carlos A. Guerrero
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia
| | - Orlando Acosta
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia
| |
Collapse
|
12
|
Taverner WK, Jacobus EJ, Christianson J, Champion B, Paton AW, Paton JC, Su W, Cawood R, Seymour LW, Lei-Rossmann J. Calcium Influx Caused by ER Stress Inducers Enhances Oncolytic Adenovirus Enadenotucirev Replication and Killing through PKCα Activation. Mol Ther Oncolytics 2019; 15:117-130. [PMID: 31890865 PMCID: PMC6931121 DOI: 10.1016/j.omto.2019.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/22/2019] [Indexed: 01/17/2023] Open
Abstract
Oncolytic viruses represent an emerging approach to cancer therapy. However, better understanding of their interaction with the host cancer cell and approaches to enhance their efficacy are needed. Here, we investigate the effect of chemically induced endoplasmic reticulum (ER) stress on the activity of the chimeric group B adenovirus Enadenotucirev, its closely related parental virus Ad11p, and the archetypal group C oncolytic adenovirus Ad5. We show that treatment of colorectal and ovarian cancer cell lines with thapsigargin or ionomycin caused an influx of Ca2+, leading to an upregulation in E1A transcript and protein levels. Increased E1A protein levels, in turn, increased levels of expression of the E2B viral DNA polymerase, genome replication, late viral protein expression, infectious virus particle production, and cell killing during Enadenotucirev and Ad11p, but not Ad5, infection. This effect was not due to the induction of ER stress, but rather the influx of extracellular Ca2+ and consequent increase in protein kinase C activity. These results underscore the importance of Ca2+ homeostasis during adenoviral infection, indicate a signaling pathway between protein kinase C and E1A, and raise the possibility of using Ca2+ flux-modulating agents in the manufacture and potentiation of oncolytic virotherapies.
Collapse
Affiliation(s)
- William K. Taverner
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Egon J. Jacobus
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - John Christianson
- NDORMS, Botnar Research Centre, University of Oxford, Headington, Oxford OX3 7LD, UK
| | - Brian Champion
- PsiOxus Therapeutics, Ltd., Milton Park, Abingdon OX14 3YS, UK
| | - Adrienne W. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide SA 5005, Australia
| | - James C. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide SA 5005, Australia
| | - Weiheng Su
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Ryan Cawood
- Oxford Genetics Ltd., Medawar Centre, Robert Robinson Avenue, Oxford OX4 4HG, UK
| | - Len W. Seymour
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Janet Lei-Rossmann
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, UK
| |
Collapse
|
13
|
Song H, Zhong LP, He J, Huang Y, Zhao YX. Application of Newcastle disease virus in the treatment of colorectal cancer. World J Clin Cases 2019; 7:2143-2154. [PMID: 31531310 PMCID: PMC6718777 DOI: 10.12998/wjcc.v7.i16.2143] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/21/2019] [Accepted: 07/20/2019] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is one of the main reasons of tumor-related deaths worldwide. At present, the main treatment is surgery, but the results are unsatisfactory, and the prognosis is poor. The majority of patients die due to liver or lung metastasis or recurrence. In recent years, great progress has been made in the field of tumor gene therapy, providing a new treatment for combating CRC. As oncolytic viruses selectively replicate almost exclusively in the cytoplasm of tumor cells and do not require integration into the host genome, they are safer, more effective and more attractive as oncolytic agents. Newcastle disease virus (NDV) is a natural RNA oncolytic virus. After NDV selectively infects tumor cells, the immune response induced by NDV’s envelope protein and intracellular factors can effectively kill the tumor without affecting normal cells. Reverse genetic techniques make NDV a vector for gene therapy. Arming the virus by inserting various exogenous genes or using NDV in combination with immunotherapy can also improve the anti-CRC capacity of NDV, and good results have been achieved in animal models and clinical treatment trials. This article reviews the molecular biological characteristics and oncolytic mechanism of NDV and discusses in vitro and in vivo experiments on NDV anti-CRC capacity and clinical treatment. In conclusion, NDV is an excellent candidate for cancer treatment, but more preclinical studies and clinical trials are needed to ensure its safety and efficacy.
Collapse
Affiliation(s)
- Hui Song
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Li-Ping Zhong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian He
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yong Huang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yong-Xiang Zhao
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
14
|
Wu T, Xiang Y, Liu T, Wang X, Ren X, Ye T, Li G. Oncolytic Vaccinia Virus Expressing Aphrocallistes vastus Lectin as a Cancer Therapeutic Agent. Mar Drugs 2019; 17:md17060363. [PMID: 31248066 PMCID: PMC6628141 DOI: 10.3390/md17060363] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022] Open
Abstract
Lectins display a variety of biological functions including insecticidal, antimicrobial, as well as antitumor activities. In this report, a gene encoding Aphrocallistes vastus lectin (AVL), a C-type lectin, was inserted into an oncolytic vaccinia virus vector (oncoVV) to form a recombinant virus oncoVV-AVL, which showed significant in vitro antiproliferative activity in a variety of cancer cell lines. Further investigations revealed that oncoVV-AVL replicated faster than oncoVV significantly in cancer cells. Intracellular signaling elements including NF-κB2, NIK, as well as ERK were determined to be altered by oncoVV-AVL. Virus replication upregulated by AVL was completely dependent on ERK activity. Furthermore, in vivo studies showed that oncoVV-AVL elicited significant antitumor effect in colorectal cancer and liver cancer mouse models. Our study might provide insights into a novel way of the utilization of marine lectin AVL in oncolytic viral therapies.
Collapse
Affiliation(s)
- Tao Wu
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Yulin Xiang
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Tingting Liu
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Xue Wang
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Xiaoyuan Ren
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Ting Ye
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Gongchu Li
- Zhejiang Sci-Tech University Hangzhou Gongchu Joint Institute of Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
15
|
Targeting Palbociclib-Resistant Estrogen Receptor-Positive Breast Cancer Cells via Oncolytic Virotherapy. Cancers (Basel) 2019; 11:cancers11050684. [PMID: 31100952 PMCID: PMC6563125 DOI: 10.3390/cancers11050684] [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: 04/26/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 12/26/2022] Open
Abstract
While clinical responses to palbociclib have been promising, metastatic breast cancer remains incurable due to the development of resistance. We generated estrogen receptor-positive (ER+) and ER-negative (ER−) cell line models and determined their permissiveness and cellular responses to an oncolytic adenovirus (OAd) known as Ad5/3-delta24. Analysis of ER+ and ER− palbociclib-resistant cells revealed two clearly distinguishable responses to the OAd. While ER+ palbociclib-resistant cells displayed a hypersensitive phenotype to the effects of the OAd, ER− palbociclib-resistant cells showed a resistant phenotype to the OAd. Hypersensitivity to the OAd in ER+ palbociclib-resistant cells correlated with a decrease in type I interferon (IFN) signaling, an increase in viral entry receptor expression, and an increase in cyclin E expression. OAd resistance in ER− palbociclib-resistant cells correlated with an increase in type I IFN signaling and a marked decrease in viral entry receptor. Using the OAd as monotherapy caused significant cytotoxicity to both ER+ and ER− palbociclib-sensitive cell lines. However, the addition of palbociclib increased the oncolytic activity of the OAd only in ER+ palbociclib-sensitive cells. Our studies provide a mechanistic base for a novel anti-cancer regimen composed of an OAd in combination with palbociclib for the treatment of ER+ breast cancer.
Collapse
|
16
|
Sivanandam V, LaRocca CJ, Chen NG, Fong Y, Warner SG. Oncolytic Viruses and Immune Checkpoint Inhibition: The Best of Both Worlds. MOLECULAR THERAPY-ONCOLYTICS 2019; 13:93-106. [PMID: 31080879 PMCID: PMC6503136 DOI: 10.1016/j.omto.2019.04.003] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cancer immunotherapy and the emergence of immune checkpoint inhibitors have markedly changed the treatment paradigm for many cancers. They function to disrupt cancer cell evasion of the immune response and activate sustained anti-tumor immunity. Oncolytic viruses have also emerged as an additional therapeutic agent for cancer treatment. These viruses are designed to target and kill tumor cells while leaving the normal cells unharmed. As part of this process, oncolytic virus infection stimulates anti-cancer immune responses that augment the efficacy of checkpoint inhibition. These viruses have the capability of transforming a “cold” tumor microenvironment with few immune effector cells into a “hot” environment with increased immune cell and cytokine infiltration. For this reason, there are multiple ongoing clinical trials that combine oncolytic virotherapy and immune checkpoint inhibitors. This review will detail the key oncolytic viruses in preclinical and clinical studies and highlight the results of their testing with checkpoint inhibitors.
Collapse
Affiliation(s)
- Venkatesh Sivanandam
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | - Nanhai G Chen
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Susanne G Warner
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
17
|
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
|
18
|
Lundstrom K. Self-Replicating RNA Viruses for RNA Therapeutics. Molecules 2018; 23:molecules23123310. [PMID: 30551668 PMCID: PMC6321401 DOI: 10.3390/molecules23123310] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022] Open
Abstract
Self-replicating single-stranded RNA viruses such as alphaviruses, flaviviruses, measles viruses, and rhabdoviruses provide efficient delivery and high-level expression of therapeutic genes due to their high capacity of RNA replication. This has contributed to novel approaches for therapeutic applications including vaccine development and gene therapy-based immunotherapy. Numerous studies in animal tumor models have demonstrated that self-replicating RNA viral vectors can generate antibody responses against infectious agents and tumor cells. Moreover, protection against challenges with pathogenic Ebola virus was obtained in primates immunized with alphaviruses and flaviviruses. Similarly, vaccinated animals have been demonstrated to withstand challenges with lethal doses of tumor cells. Furthermore, clinical trials have been conducted for several indications with self-amplifying RNA viruses. In this context, alphaviruses have been subjected to phase I clinical trials for a cytomegalovirus vaccine generating neutralizing antibodies in healthy volunteers, and for antigen delivery to dendritic cells providing clinically relevant antibody responses in cancer patients, respectively. Likewise, rhabdovirus particles have been subjected to phase I/II clinical trials showing good safety and immunogenicity against Ebola virus. Rhabdoviruses have generated promising results in phase III trials against Ebola virus. The purpose of this review is to summarize the achievements of using self-replicating RNA viruses for RNA therapy based on preclinical animal studies and clinical trials in humans.
Collapse
|
19
|
LaRocca CJ, Warner SG. Oncolytic viruses and checkpoint inhibitors: combination therapy in clinical trials. Clin Transl Med 2018; 7:35. [PMID: 30426287 PMCID: PMC6234197 DOI: 10.1186/s40169-018-0214-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/25/2018] [Indexed: 12/15/2022] Open
Abstract
Advances in the understanding of cancer immunotherapy and the development of multiple checkpoint inhibitors have dramatically changed the current landscape of cancer treatment. Recent large-scale phase III trials (e.g. PHOCUS, OPTiM) are establishing use of oncolytic viruses as another tool in the cancer therapeutics armamentarium. These viruses do not simply lyse cells to achieve their cancer-killing effects, but also cause dramatic changes in the tumor immune microenvironment. This review will highlight the major vector platforms that are currently in development (including adenoviruses, reoviruses, vaccinia viruses, herpesviruses, and coxsackieviruses) and how they are combined with checkpoint inhibitors. These vectors employ a variety of engineered capsid modifications to enhance infectivity, genome deletions or promoter elements to confer selective replication, and encode a variety of transgenes to enhance anti-tumor or immunogenic effects. Pre-clinical and clinical data have shown that oncolytic vectors can induce anti-tumor immunity and markedly increase immune cell infiltration (including cytotoxic CD8+ T cells) into the local tumor microenvironment. This "priming" by the viral infection can change a 'cold' tumor microenvironment into a 'hot' one with the influx of a multitude of immune cells and cytokines. This alteration sets the stage for subsequent checkpoint inhibitor delivery, as they are most effective in an environment with a large lymphocytic infiltrate. There are multiple ongoing clinical trials that are currently combining oncolytic viruses with checkpoint inhibitors (e.g. CAPTIVE, CAPRA, and Masterkey-265), and the initial results are encouraging. It is clear that oncolytic viruses and checkpoint inhibitors will continue to evolve together as a combination therapy for multiple types of cancers.
Collapse
Affiliation(s)
- Christopher J LaRocca
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA
| | - Susanne G Warner
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
20
|
NF-κB Signaling in Targeting Tumor Cells by Oncolytic Viruses-Therapeutic Perspectives. Cancers (Basel) 2018; 10:cancers10110426. [PMID: 30413032 PMCID: PMC6265863 DOI: 10.3390/cancers10110426] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/04/2018] [Accepted: 11/06/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, oncolytic virotherapy became a promising therapeutic approach, leading to the introduction of a novel generation of anticancer drugs. However, despite evoking an antitumor response, introducing an oncolytic virus (OV) to the patient is still inefficient to overcome both tumor protective mechanisms and the limitation of viral replication by the host. In cancer treatment, nuclear factor (NF)-κB has been extensively studied among important therapeutic targets. The pleiotropic nature of NF-κB transcription factor includes its involvement in immunity and tumorigenesis. Therefore, in many types of cancer, aberrant activation of NF-κB can be observed. At the same time, the activity of NF-κB can be modified by OVs, which trigger an immune response and modulate NF-κB signaling. Due to the limitation of a monotherapy exploiting OVs only, the antitumor effect can be enhanced by combining OV with NF-κB-modulating drugs. This review describes the influence of OVs on NF-κB activation in tumor cells showing NF-κB signaling as an important aspect, which should be taken into consideration when targeting tumor cells by OVs.
Collapse
|
21
|
Lundstrom K. Viral Vectors in Gene Therapy. Diseases 2018; 6:diseases6020042. [PMID: 29883422 PMCID: PMC6023384 DOI: 10.3390/diseases6020042] [Citation(s) in RCA: 310] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 01/02/2023] Open
Abstract
Applications of viral vectors have found an encouraging new beginning in gene therapy in recent years. Significant improvements in vector engineering, delivery, and safety have placed viral vector-based therapy at the forefront of modern medicine. Viral vectors have been employed for the treatment of various diseases such as metabolic, cardiovascular, muscular, hematologic, ophthalmologic, and infectious diseases and different types of cancer. Recent development in the area of immunotherapy has provided both preventive and therapeutic approaches. Furthermore, gene silencing generating a reversible effect has become an interesting alternative, and is well-suited for delivery by viral vectors. A number of preclinical studies have demonstrated therapeutic and prophylactic efficacy in animal models and furthermore in clinical trials. Several viral vector-based drugs have also been globally approved.
Collapse
|
22
|
Qu X, Tang Y, Hua S. Immunological Approaches Towards Cancer and Inflammation: A Cross Talk. Front Immunol 2018; 9:563. [PMID: 29662489 PMCID: PMC5890100 DOI: 10.3389/fimmu.2018.00563] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
The inflammation is the protective response of the body against various harmful stimuli; however, the aberrant and inappropriate activation tends to become harmful. The acute inflammatory response tends to resolved once the offending agent is subside but this acute response becomes chronic in nature when the body is unable to successfully neutralized the noxious stimuli. This chronic inflammatory microenvironment is associated with the release of various pro-inflammatory and oncogenic mediators such as nitric oxide (NO), cytokines [IL-1β, IL-2, interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α)], growth factor, and chemokines. These mediators make the inflammatory microenvironment more vulnerable toward tumorigenesis. The pro-inflammatory mediators released during the chronic inflammation tends to induce several molecular signaling cascades such as nuclear factor kappa B, MAPKinase, nuclear factor erythroid 2-related factor 2, phosphoinositide-3-kinase, Janus kinases/STAT, Wnt/B-catenin, and cyclic AMP response element binding protein. The immune system and its components have a pleiotropic effect on inflammation and cancer progression. Immune components such as T cells, natural killer cells, macrophages, and neutrophils either inhibit or enhance tumor initiation depending on the type of tumor and immune cells involved. Tumor-associated macrophages and tumor-associated neutrophils are pro-tumorigenic cells highly prevalent in inflammation-mediated tumors. Similarly, presence of T regulatory (Treg) cells in an inflammatory and tumor setting suppresses the immune system, thus paving the way for oncogenesis. However, Treg cells also inhibit autoimmune inflammation. By contrast, cytotoxic T cells and T helper cells confer antitumor immunity and are associated with better prognosis in patients with cancer. Cytotoxic T cells inflict a direct cytotoxic effect on cells expressing oncogenic markers. Currently, several anti-inflammatory and antitumor therapies are under trials in which these immune cells are exploited. Adoptive cell transfer composed of tumor-infiltrating lymphocytes has been tried for the treatment of tumors after their ex vivo expansion. Mediators released by cells in a tumorigenic and inflammatory microenvironment cross talk with nearby cells, either promoting or inhibiting inflammation and cancer. Recently, several cytokine-based therapies are either being developed or are under trial to treat such types of manifestations. Monoclonal antibodies directed against TNF-α, VEGF, and IL-6 has shown promising results to ameliorate inflammation and cancer, while direct administration of IL-2 has been shown to cause tumor regression.
Collapse
Affiliation(s)
- Xinglong Qu
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| | - Ying Tang
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| | - Shucheng Hua
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|