GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
  • 1
    Online-Ressource
    Online-Ressource
    American Society of Clinical Oncology (ASCO) ; 2020
    In:  Journal of Clinical Oncology Vol. 38, No. 5_suppl ( 2020-02-10), p. 21-21
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 38, No. 5_suppl ( 2020-02-10), p. 21-21
    Kurzfassung: 21 Background: Oncolytic virotherapy has been pursued by multiple companies and institutions with few candidates reaching the clinic and demonstrating limited efficacy. The therapeutic potential of oncolytic viruses can be severely restricted by innate and adaptive immune barriers. To overcome this obstacle, we load and protect tumor selective CAL1 oncolytic vaccinia virus into adipose-derived stem cells (AD-MSC) to generate a new therapeutic agent called SNV1(SuperNova1). Methods: SNV1s were generated by incubating AD-MSC with CAL1 virus. SNV1 was analyzed for its ability to kill cancer cell lines and protect virus in the presence of active neutralizing antibodies and complement. In animals, SNV1 was intratumorally injected in various xenograft and syngeneic models. Viral biodistribution was also evaluated by PCR. Immune infiltration were analyzed using flow cytometry. Results: Compared to the naked virus, SNV1 showed improved protection against the humoral barriers and efficient eradication of various human cancer cell lines in vitro. Intratumoral SNV1 treatment showed statistically significant and potentiated tumor growth inhibition compared to control or CAL1 naked virus treatment in all tested models (prostate, breast, melanoma, colon, and prostate cancers). Importantly, local administration of SNV1 induced systemic therapeutic effects. Five days after SNV1 administration, tumor infiltrating lymphocytes (TILs) from both treated and untreated tumors showed increased CD4 and CD8 T-cell populations. As well as decreased frequency of Tregs, and improved effector to Treg ratios, which was associated with inhibition of tumor growth at the treated tumor site and also at distant untreated sites. Ongoing and persistent virus infection could be detected in the treated tumor as late as 15 days after administration. Conclusions: This study demonstrates the ability of our cell-based platform to protect and potentiate oncolytic vaccinia virus by circumventing the innate and adaptive immune barriers, resulting in enhanced oncolytic virotherapy. These findings provide fundamental rationale for the development of cell-based platforms to maximize the therapeutic potential of various oncolytic viruses.
    Materialart: Online-Ressource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Clinical Oncology (ASCO)
    Publikationsdatum: 2020
    ZDB Id: 2005181-5
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Cancers, MDPI AG, Vol. 14, No. 24 ( 2022-12-13), p. 6136-
    Kurzfassung: We describe the repurposing and optimization of the TK-positive (thymidine kinase) vaccinia virus strain ACAM1000/ACAM2000™ as an oncolytic virus. This virus strain has been widely used as a smallpox vaccine and was also used safely in our recent clinical trial in patients with advanced solid tumors and Acute Myeloid Leukemia (AML). The vaccinia virus was amplified in CV1 cells and named CAL1. CAL1 induced remarkable oncolysis in various human and mouse cancer cells and preferentially amplified in cancer cells, supporting the use of this strain as an oncolytic virus. However, the therapeutic potential of CAL1, as demonstrated with other oncolytic viruses, is severely restricted by the patients’ immune system. Thus, to develop a clinically relevant oncolytic virotherapy agent, we generated a new off-the-shelf therapeutic called Supernova1 (SNV1) by loading CAL1 virus into allogeneic adipose-derived mesenchymal stem cells (AD-MSC). Culturing the CAL1-infected stem cells allows the expression of virally encoded proteins and viral amplification prior to cryopreservation. We found that the CAL1 virus loaded into AD-MSC was resistant to humoral inactivation. Importantly, the virus-loaded stem cells (SNV1) released larger number of infectious viral particles and virally encoded proteins, leading to augmented therapeutic efficacy in vitro and in animal tumor models.
    Materialart: Online-Ressource
    ISSN: 2072-6694
    Sprache: Englisch
    Verlag: MDPI AG
    Publikationsdatum: 2022
    ZDB Id: 2527080-1
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 6542-6542
    Kurzfassung: Background: Oncolytic virotherapy is a promising immuno-oncology approach that has not realized its potential due to rapid elimination by humoral immunity mediated by complement and neutralizing antibodies. We propose to use an adipose-derived mesenchymal stemcell-based platform,where the virus can be protected and amplified and potentiated inside the stem cells in order to minimize the clearance by anti-viral immunity. ACAM2000, the smallpox vaccine currently licensed in the U.S., is a clonal derivative of Dryvax® with reduced virulence and a well-documented safety profile in humans. This vaccinia virus strain can potentially be used as an oncolytic virus for cancer treatment. In this study, we evaluate the ability of ACAM2000 to (1) selectively kill cancer cells, (2) to be genetically modified without affecting its natural tumor selectivity, and to (3) determine if a stem cell-based platform can protect the virus from inactivation and potentiate its anti-tumor effects. Methods: ACAM2000 was amplified in CV1 cells and named CAL1. CAL1 was tested for its ability to replicate and selectively kill various human prostate cancer cell lines in vitro and in vivo. Additionally, CAL1 was loaded into adipose-derived mesenchymal stem cells to generate a new therapeutic agent called SuperNova1 (SNV1). Both CAL1 and SNV1 were tested for their ability to kill cancer cells in the presence of active complement and neutralizing antibodies in cell culture as well as in mice. Furthermore, CAL1 was used as the backbone to generate derivative CAL2 viruses using CRISPR/Cas9 technology to insert the gene encoding the fluorescent protein TurboFP into the intergenic locus between ORF-157 and ORF-158 of CAL1 without disrupting any existing CAL1 ORFs. Results: We showed that in vitro CAL1 preferentially infected, amplified in and lysed tumor cells and was also able to cause tumor regression in vivo without signs of toxicity. Furthermore, we demonstrated that the backbone of CAL1 can be used to engineer recombinant viruses, CAL2, that carry therapeutic genes without additionally attenuating the ability of the virus to amplify or kill tumor cells. SNV1 significantly enhanced protection of CAL1 virus from clearance by the immune system, leading to higher therapeutic efficacy. Furthermore, SNV1 provided instantly active viral particles for immediate infection and simultaneous release of therapeutic proteins in the injected tumors. Conclusions: CAL1 could be used as an oncolytic agent. We show here that a major advantage of using a cell-based platform to deliver and potentiate oncolytic vaccinia virus is the prevention of viral inactivation by the humoral immune system resulting in enhanced oncolytic viral therapy. Citation Format: Duong H. Nguyen, Thomas Herrmann, Ashley Alamillo, Forrest Neuharth, Alberto Gomez, Ivelina Minev, Barbara Härtl, Laura Schneider, Boris Minev, Dobrin Draganov, Antonio F. Santidrian. CAL1 vaccinia virus as oncolytic agent and potential use of cell-based platform to enhance its therapeutic effects [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6542.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...