1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Progress of oncolytic virotherapy for neuroblastoma

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          As a neuroendocrine tumor derived from the neural crest, neuroblastoma (NB) is the most common extracranial solid tumor in children. The prognosis in patients with low- and intermediate-risk NB is favorable while that in high-risk patients is often detrimental. However, the management of the considerably large proportion of high-risk patients remains challenging in clinical practice. Among various new approaches, oncolytic virus (OV) therapy offers great advantages in tumor treatment, especially for high-risk NB. Genetic modified OVs can target NB specifically without affecting normal tissue and avoid the widespread drug resistance issue in anticancer monotherapy. Meanwhile, its safety profile provides great potential in combination therapy with chemo-, radio-, and immunotherapy. The therapeutic efficacy of OV for NB is impressive from bench to bedside. The effectiveness and safety of OVs have been demonstrated and reported in studies on children with NB. Furthermore, clinical trials on some OVs (Celyvir, Pexa-Vec (JX-594) and Seneca Valley Virus (NTX-010)) have reported great results. This review summarizes the latest evidence in the therapeutic application of OVs in NB, including those generated in cell lines, animal models and clinical trials.

          Related collections

          Most cited references138

          • Record: found
          • Abstract: not found
          • Article: not found

          Recent advances in neuroblastoma.

          John Maris (2010)
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            ONCOLYTIC VIROTHERAPY

            Oncolytic virotherapy is an emerging treatment modality which uses replication competent viruses to destroy cancers. Advances in the past two years include preclinical proof of feasibility for a single-shot virotherapy cure, identification of drugs that accelerate intratumoral virus propagation, new strategies to maximize the immunotherapeutic potential of oncolytic virotherapy, and clinical confirmation of a critical viremic thereshold for vascular delivery and intratumoral virus replication. The primary clinical milestone was completion of accrual in a phase III trial of intratumoral herpes simplex virus therapy using talimogene laherparepvec for metastatic melanoma. Challenges for the field are to select ‘winners’ from a burgeoning number of oncolytic platforms and engineered derivatives, to transiently suppress but then unleash the power of the immune system to maximize both virus spread and anticancer immunity, to develop more meaningful preclinical virotherapy models and to manufacture viruses with orders of magnitude higher yields compared to established vaccine manufacturing processes.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Oncolytic viruses: a new class of immunotherapy drugs

              Key Points Oncolytic viruses mediate anti-tumour responses through a dual mechanism involving viral oncolysis of cancer cells and induction of host anti-tumour immunity. The molecular and cellular mechanisms of action are not fully elucidated but are likely to depend on viral replication within transformed cells, induction of primary cell death, interaction with tumour cell antiviral elements, release of danger signals and initiation of innate and adaptive anti-tumour immunity. A variety of native and genetically modified viruses have been utilized as oncolytic vectors in preclinical studies, which have demonstrated therapeutic activity against several types of cancer. Oncolytic viruses can be genetically modified to decrease pathogenicity, increase lytic potential and enhance immunogenicity, improving the risk–benefit ratio for clinical development. The approval of a modified adenovirus, H101, in China and the pending approval of a modified herpes simplex virus type 1 (HSV-1) encoding granulocyte–macrophage colony stimulating factor (GM-CSF), termed talimogene laherparepvec (T-VEC), by the US Food and Drug Administration (FDA) in the United States is likely to promote further drug development within this new class of cancer therapeutics. Oncolytic viruses face unique challenges in drug development, including the need for optimal clinical trial designs and response assessment that capture therapeutic responses, different regulatory and commercialization pathways, the need for live culture scale-up procedures, and novel biosafety concerns related to viral persistence in patients and transmission to household contacts and health-care providers. Supplementary information The online version of this article (doi:10.1038/nrd4663) contains supplementary material, which is available to authorized users.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Pediatr
                Front Pediatr
                Front. Pediatr.
                Frontiers in Pediatrics
                Frontiers Media S.A.
                2296-2360
                18 November 2022
                2022
                : 10
                : 1055729
                Affiliations
                Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects , Shanghai, China
                Author notes

                Edited by: Manuel Ramirez, Niño Jesús University Children's Hospital, Spain

                Reviewed by: Ramon Alemany, Catalan Institute of Oncology, Spain Javier Garcia-Castro, Carlos III Health Institute (ISCIII), Spain

                [* ] Correspondence: Rui Dong rdong@ 123456fudan.edu.cn
                [ † ]

                These authors have contributed equally to this work and share first authorship

                Specialty Section: This article was submitted to Pediatric Oncology, a section of the journal Frontiers in Pediatrics

                Abbreviations Ad, Adenovirus; CDX, Cell derived xenograft; GD2m, disialoganglioside mimotope; GEMMs, Genetically engineered mouse models; HCMV, human cytomegalovirus; HSV, Herpes simplex virus; hTERT, Human telomerase reverse transcriptase; MV, Measles virus; NB , Neuroblastoma ; oHSV, Oncolytic herpes simplex virus; OV , Oncolytic virus ; PDX, Patient-derived tumor xenograft; SINV, Sindbis virus; SVV, Seneca valley virus; TIMP3, tissue inhibitor of metalloproteinases 3; TME, Tumor microenvironment; VSV, Vesicular stomatitis viSrus; VV, Vaccinia virus.

                Article
                10.3389/fped.2022.1055729
                9716318
                36467495
                c907a028-bf54-4d24-b61e-55d84dbe0cfb
                © 2022 Chen, Dai, Zhan, Yang, Chen, Li, Zhou and Dong.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 28 September 2022
                : 03 November 2022
                Page count
                Figures: 1, Tables: 2, Equations: 0, References: 139, Pages: 0, Words: 0
                Funding
                Funded by: Cyrus Tang Foundation, doi 10.13039/100012515;
                Award ID: ZSBK0070
                Funded by: Shanghai Municipal Key Clinical Specialty
                Award ID: shslczdzk05703
                Funded by: National Natural Science Foundation of China, doi 10.13039/501100001809;
                Award ID: 82072782
                Categories
                Pediatrics
                Review

                oncolytic virus,neuroblastoma,mechanism,preclinical,clinical,combination therapy

                Comments

                Comment on this article