Oncolytic Herpes Simplex Virus

Backgrounds Featured Services Service Workflow Resources Related Sections

Oncolytic herpes simplex virus (oHSV) is one of the oncolytic viruses being studied in cancer therapeutic research and several oHSVs have been investigated in clinical trials. With years of experience in immunology and oncology, Creative Biolabs has established a comprehensive platform called OncoVirapy™ for providing oncolytic virus development service. By virtue of OncoVirapy™, we are able to design, engineer and generate the most efficacious oncolytic herpes viral vector and viral particles.

Backgrounds

HSV is a double-stranded DNA virus belonging to the Herpesviridae family. There're two existing variants of HSV, including HSV type 1 (HSV-1) and HSV type 2 (HSV-2). Among these two subtypes, HSV-1 has been widely investigated in cancer oncolytic viral therapy. HSV-1 contains a double-stranded DNA (dsDNA) genome of about 152 kb, which is arranged by two unique sequences: Unique Long (UL) and Unique Short (US), an icosadeltahedral capsid, an amorphous tegument, and an outer lipid bilayer envelope with glycoprotein spikes for viral entry. Some characteristics including large foreign genes capacity (estimated to be about 30 kb), neural sensitivity tropism and high titers of manufacture enable HSV-1 to be used as a viral-delivery candidate for cancer treatment. Although it is best known as a neurotropic virus due to its latency in neurons, HSV-1 actually can infect a wide variety of cell types and thus tumor types. Talimogene laherparepvec (T-Vec) was an oncolytic HSV that was first approved for the melanoma treatment by the Food and Drug Administration (FDA) in October 2015, and then successively approved in Europe, Australia, Switzerland, and Israel.

+- HSV-1 Gene Mutations in Oncolytic Virotherapy

+- Cases of oHSV Genetic Modifications

  • Safety

    HSV-1 can be genetically-engineered or significantly attenuated to confer safety by deleting or mutating some virulence genes.

  • Tumor targeting specificity

    HSV-1 genome has many genes that can be deleted, replaced or mutated to confer tumor-targeting specificity through tumor-selective receptor binding and transcriptional- or post-transcriptional miRNA-targeting, respectively.

  • "Armed" with transgenes

    HSV can be "armed" with transgenes that are: reporters, to track virus replication and spread; cytotoxic, to kill uninfected tumor cells; immune modulatory, to stimulate antitumor immunity; or tumor microenvironment altering, to enhance virus spread or to inhibit tumor growth.

    Oncolytic-Herpes-Simplex-Virus-1.jpg Fig.1 Map of HSV-1 genome and schematic representation of essential genes and non-essential genes. (Artusi, 2018)

Oncolytic virotherapy with mutants derived from HSV-1 exhibits significant antitumor effects in preclinical models. Several mutants have now been tested in clinical trials for a variety of cancer types, such as colorectal cancer, melanoma and osteosarcoma. Excitingly, some mutants have been found to be safe and effective. Table.1 listed several common genetic alterations of oHSVs.

Fig.1 Map of HSV-1 genome and schematic representation of essential genes and non-essential genes. (Artusi, 2018) Oncolytic-Herpes-Simplex-Virus-2.jpg

  • Thymidine kinase/UL23 (E)

    HSV-1 thymidine kinase (TK) is a multifunctional enzyme that catalyzes the creation of deoxythymidine 5'-phosphate (dTMP) from deoxythymidine, as well as phosphorylating deoxycytidine and nucleoside analogs, creating precursors for viral DNA synthesis. Loss of TK activity halts HSV-1 replication in nondividing cells, making TK mutants safer than wild type HSV-1. Dlsptk, an HSV-1 lacking TK, was the first genetically-engineered oncolytic virus devised to kill tumor cells while sparing normal cells.

  • ICP6/UL39 (E)

    ICP6 is the large subunit of the viral ribonucleotide reductase, which transforms ribonucleotides into deoxyribonucleotides (dNMP) by reducing the 2'-COH group. ICP6 is essential for HSV replication in noncycling cells, making ICP6-null oHSVs selective for dividing cells.

  • γ34.5 (ICP34.5)/RL1 (L)

    γ34.5, one of the most frequently mutated genes for creating oHSVs, has many functions. HSV-1 is diploid for the γ34.5 gene, and mature virions contain the protein in their tegument in order to deliver it upon entry. Attenuation of both copies of this gene leads to a reduction of neurovirulence associated with HSV-1 in vitro and in vivo. Despite the reduced replication in normal cells, HSV-1 virions deleted for ICP34.5 efficiently replicate in and are cytotoxic to a majority of glioma cell lines and primary tumor-derived cells.

  • ICP47

    ICP47 deletion enhances anti-tumor efficacy while retaining safety characteristics. G47Δ showed efficacy in all solid tumor models tested in vivo, such as hepatocellular carcinoma, schwannoma, prostate cancer, nasopharyngeal carcinoma, glioma, thyroid carcinoma, colorectal cancer, breast cancer and malignant peripheral nerve sheath tumor.

  • ICP0 (IE)

    ICP0 encodes a RING finger ubiquitin E3 ligase that targets cellular proteins. ICP0-null HSV-1 is extremely sensitive to IFN and PML-mediated disruption of the viral lifecycle. ICP0-null HSV-1 is extremely sensitive to IFN and PML-mediated disruption of the viral lifecycle.

  • UL56 (L)

    UL56 is another gene associated with pathogenicity and neuroinvasiveness. UL56 is a good candidate for mutation when creating a safer oHSV. In addition, it may contribute to tumor selectivity.

Featured Services

We provide a comprehensive range of HSV services based on our advanced OncoVirapy™ platform.

  • GOI cloning (e.g., antibody, bispecific antibody, cytokine, chemokine, shRNA or gRNA) and confirmation through restriction mapping and sequencing verification
  • Functional validation of transgenes
  • Expression cassette design with the most appropriate promoter to guarantee the high-level expression
  • Viral genes engineering (single gene or different genes combination)
  • Different construction strategies (homologous recombination, BAC system, etc.)
  • HSV amplification, purification, titering services
  • HSV whole genome sequencing (WGS) service

Oncolytic HSV has been widely used in pre-clinical and clinical cases; moreover, there's hope that its curative effect will be further enhanced through the combination of oHSV with both traditional and emerging therapeutics. With the development of modern genetic engineering techniques, more and more strategies have been discovered to optimize the construction of oncolytic HSV, to reduce toxicity, as well as increase the specificity and efficacy. Creative Biolabs offers a broad range of custom oncolytic HSV cloning and construction services at a reasonable cost and with quick turnaround time. Besides, we also provide in vitro and in vivo validation services for engineered oncolytic HSV to facilitate your whole project.

Service Workflow

With years of experience and continuous optimization, Creative Biolabs has developed a systematic workflow for oncolytic HSV construction as follows. In addition to the standard construction workflow, we also developed bacterial artificial chromosome (BAC) cloning technology, which significantly advances the capacity of herpesvirus researchers to manipulate virus genomes.

Oncolytic-Herpes-Simplex-Virus-3.jpg Fig.2 Workflow of oncolytic HSV construction at Creative Biolabs.

Resources

Use the resources in our library to help you understand your options and make critical decisions for your study.

VideosPodcastsSupport Knowledge
  • Oncolytic Virotherapy Basic Knowledge Review 00:13:23
    Oncolytic Virotherapy Basic Knowledge Review
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Introduction to Oncolytic Virotherapy 00:10:09
    Introduction to Oncolytic Virotherapy
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Ex Vivo Virotherapy with Myxoma Virus to Treat Cancer 00:08:29
    Ex Vivo Virotherapy with Myxoma Virus to Treat Cancer
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Immunohistochemistry for Tumor-Infiltrating Immune Cells After Oncolytic Virotherapy 00:09:40
    Immunohistochemistry for Tumor-Infiltrating Immune Cells After Oncolytic Virotherapy
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Detection of Tumor Antigen-Specific T-Cell Responses After Oncolytic Vaccination 00:11:47
    Detection of Tumor Antigen-Specific T-Cell Responses After Oncolytic Vaccination
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Evaluation of Oncolytic Virus-Induced Therapeutic Tumor Vaccination Effects in Murine Tumor Models 00:10:55
    Evaluation of Oncolytic Virus-Induced Therapeutic Tumor Vaccination Effects in Murine Tumor Models
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Delivery of Oncolytic Reovirus by Cell Carriers 00:16:09
    Delivery of Oncolytic Reovirus by Cell Carriers
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • In Vivo Bioimaging for Monitoring Intratumoral Virus Activity 00:09:15
    In Vivo Bioimaging for Monitoring Intratumoral Virus Activity
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Oncolytic Immunotherapy for Bladder Cancer Using Coxsackie A21 Virus 00:14:52
    Oncolytic Immunotherapy for Bladder Cancer Using Coxsackie A21 Virus
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Use of Liquid Patient Ascites Fluids as a Preclinical Model for Oncolytic Virus Activity 00:10:47
    Use of Liquid Patient Ascites Fluids as a Preclinical Model for Oncolytic Virus Activity
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Generating Primary Models of Human Cancer to Aid in the Development of Clinically Relevant Oncolytic Viruses 00:09:08
    Generating Primary Models of Human Cancer to Aid in the Development of Clinically Relevant Oncolytic Viruses
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Considerations for Clinical Translation of MG1 Maraba Virus-Ⅰ 00:11:28
    Considerations for Clinical Translation of MG1 Maraba Virus-Ⅰ
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Considerations for Clinical Translation of MG1 Maraba Virus-Ⅱ 00:13:32
    Considerations for Clinical Translation of MG1 Maraba Virus-Ⅱ
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Considerations for Clinical Translation of MG1 Maraba Virus-Ⅲ 00:09:14
    Considerations for Clinical Translation of MG1 Maraba Virus-Ⅲ
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Fluorescence In Situ Hybridization (FISH) Detection of Viral Nucleic Acids in Oncolytic H-1 Parvovirus-Treated Human Brain Tumors 00:10:34
    Fluorescence In Situ Hybridization (FISH) Detection of Viral Nucleic Acids in Oncolytic H-1 Parvovirus-Treated Human Brain Tumors
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
  • Mathematical Modeling of Oncolytic Virotherapy 00:09:22
    Mathematical Modeling of Oncolytic Virotherapy
    Subscribe-on-Apple-PodcastsSubscribe-on-Spotify
Introduction of Herpes Simplex Virus as Oncolytic Virus

Oncolytic viruses (OVs), specifically the herpes simplex virus (HSV), are emerging as a promising and effective treatment for cancer. Targeted OVs aim to kill tumor cells without harming normal cells, with HSV showing particular suitability. Genetic manipulation of HSV genes involved in evading host immune responses enhances its anti-tumor effects. Oncolytic HSV (oHSV) is undergoing extensive clinical trials, offering hope for improved curative effects when combined with conventional and emerging treatments. HSV-1, with its neurotropic nature, has been extensively studied for oncolytic viral therapy, displaying rapid replication, large genome modifiability, and potential for targeting tumor cells. Advances in clinical development, including retargeting strategies for systemic delivery, demonstrate promising outcomes in cancer treatment.

Applications of Oncolytic Viruses in Cancer Treatment

Oncolytic viruses (OVs), specifically the herpes simplex virus (HSV), are emerging as a promising and effective treatment for cancer. Targeted OVs aim to kill tumor cells without harming normal cells, with HSV showing particular suitability. Genetic manipulation of HSV genes involved in evading host immune responses enhances its anti-tumor effects. Oncolytic HSV (oHSV) is undergoing extensive clinical trials, offering hope for improved curative effects when combined with conventional and emerging treatments. HSV-1, with its neurotropic nature, has been extensively studied for oncolytic viral therapy, displaying rapid replication, large genome modifiability, and potential for targeting tumor cells. Advances in clinical development, including retargeting strategies for systemic delivery, demonstrate promising outcomes in cancer treatment.

Related Sections

Reference

  1. Artusi, S.; et al. Herpes simplex virus vectors for gene transfer to the central nervous system. Diseases. 2018, 6(3): 74.
! All products and services are for Research Use Only. Not For Clinical Use.
SERVICES

Online Inquiry