TY - JOUR A1 - Draganov, Dobrin D. A1 - Santidrian, Antonio F. A1 - Minev, Ivelina A1 - Duong, Nguyen A1 - Kilinc, Mehmet Okyay A1 - Petrov, Ivan A1 - Vyalkova, Anna A1 - Lander, Elliot A1 - Berman, Mark A1 - Minev, Boris A1 - Szalay, Aladar A. T1 - Delivery of oncolytic vaccinia virus by matched allogeneic stem cells overcomes critical innate and adaptive immune barriers JF - Journal of Translational Medicine N2 - Background Previous studies have identified IFNγ as an important early barrier to oncolytic viruses including vaccinia. The existing innate and adaptive immune barriers restricting oncolytic virotherapy, however, can be overcome using autologous or allogeneic mesenchymal stem cells as carrier cells with unique immunosuppressive properties. Methods To test the ability of mesenchymal stem cells to overcome innate and adaptive immune barriers and to successfully deliver oncolytic vaccinia virus to tumor cells, we performed flow cytometry and virus plaque assay analysis of ex vivo co-cultures of stem cells infected with vaccinia virus in the presence of peripheral blood mononuclear cells from healthy donors. Comparative analysis was performed to establish statistically significant correlations and to evaluate the effect of stem cells on the activity of key immune cell populations. Results Here, we demonstrate that adipose-derived stem cells (ADSCs) have the potential to eradicate resistant tumor cells through a combination of potent virus amplification and sensitization of the tumor cells to virus infection. Moreover, the ADSCs demonstrate ability to function as a virus-amplifying Trojan horse in the presence of both autologous and allogeneic human PBMCs, which can be linked to the intrinsic immunosuppressive properties of stem cells and their unique potential to overcome innate and adaptive immune barriers. The clinical application of ready-to-use ex vivo expanded allogeneic stem cell lines, however, appears significantly restricted by patient-specific allogeneic differences associated with the induction of potent anti-stem cell cytotoxic and IFNγ responses. These allogeneic responses originate from both innate (NK)- and adaptive (T)- immune cells and might compromise therapeutic efficacy through direct elimination of the stem cells or the induction of an anti-viral state, which can block the potential of the Trojan horse to amplify and deliver vaccinia virus to the tumor. Conclusions Overall, our findings and data indicate the feasibility to establish simple and informative assays that capture critically important patient-specific differences in the immune responses to the virus and stem cells, which allows for proper patient-stem cell matching and enables the effective use of off-the-shelf allogeneic cell-based delivery platforms, thus providing a more practical and commercially viable alternative to the autologous stem cell approach. KW - vaccinia KW - cancer KW - stem Cells KW - oncolysis KW - oncolytic virus KW - virotherapy KW - immunity KW - immunotherapy Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-226312 SN - 100 VL - 17 ER - TY - JOUR A1 - Minev, Boris R. A1 - Lander, Elliot A1 - Feller, John F. A1 - Berman, Mark A1 - Greenwood, Bernadette M. A1 - Minev, Ivelina A1 - Santidrian, Antonio F. A1 - Nguyen, Duong A1 - Draganov, Dobrin A1 - Killinc, Mehmet O. A1 - Vyalkova, Anna A1 - Kesari, Santosh A1 - McClay, Edward A1 - Carabulea, Gabriel A1 - Marincola, Francesco M. A1 - Butterfield, Lisa H. A1 - Szalay, Aladar A. T1 - First-in-human study of TK-positive oncolytic vaccinia virus delivered by adipose stromal vascular fraction cells JF - Journal of Translational Medicine N2 - Background ACAM2000, a thymidine kinase (TK)-positive strain of vaccinia virus, is the current smallpox vaccine in the US. Preclinical testing demonstrated potent oncolytic activity of ACAM2000 against several tumor types. This Phase I clinical trial of ACAM2000 delivered by autologous adipose stromal vascular fraction (SVF) cells was conducted to determine the safety and feasibility of such a treatment in patients with advanced solid tumors or acute myeloid leukemia (AML). Methods Twenty-four patients with solid tumors and two patients with AML participated in this open-label, non-randomized dose-escalation trial. All patients were treated with SVF derived from autologous fat and incubated for 15 min to 1 h with ACAM2000 before application. Six patients received systemic intravenous application only, one patient received intra-tumoral application only, 15 patients received combination intravenous with intra-tumoral deployment, 3 patients received intravenous and intra-peritoneal injection and 1 patient received intravenous, intra-tumoral and intra-peritoneal injections. Safety at each dose level of ACAM2000 (1.4 × 106 plaque-forming units (PFU) to 1.8 × 107 PFU) was evaluated. Blood samples for PK assessments, flow cytometry and cytokine analysis were collected at baseline and 1 min, 1 h, 1 day, 1 week, 1 month, 3 months and 6 months following treatment. Results No serious toxicities (> grade 2) were reported. Seven patients reported an adverse event (AE) in this study: self-limiting skin rashes, lasting 7 to 18 days—an expected adverse reaction to ACAM2000. No AEs leading to study discontinuation were reported. Viral DNA was detected in all patients’ blood samples immediately following treatment. Interestingly, in 8 patients viral DNA disappeared 1 day and re-appeared 1 week post treatment, suggesting active viral replication at tumor sites, and correlating with longer survival of these patients. No major increase in cytokine levels or correlation between cytokine levels and skin rashes was noted. We were able to assess some initial efficacy signals, especially when the ACAM2000/SVF treatment was combined with checkpoint inhibition. Conclusions Treatment with ACAM2000/SVF in patients with advanced solid tumors or AML is safe and well tolerated, and several patients had signals of an anticancer effect. These promising initial clinical results merit further investigation of therapeutic utility. Trial registration Retrospectively registered (ISRCTN#10201650) on October 22, 2018. KW - clinical trial KW - oncolytic vaccinia virus KW - stromal vascular fraction KW - immunotherapy of cancer Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-224105 VL - 17 ER - TY - JOUR A1 - Petrov, Ivan A1 - Gentschev, Ivaylo A1 - Vyalkova, Anna A1 - Elashry, Mohamed I. A1 - Klymiuk, Michele C. A1 - Arnhold, Stefan A1 - Szalay, Aladar A. T1 - Canine Adipose-Derived Mesenchymal Stem Cells (cAdMSCs) as a "Trojan Horse" in Vaccinia Virus Mediated Oncolytic Therapy against Canine Soft Tissue Sarcomas JF - Viruses N2 - Several oncolytic viruses (OVs) including various human and canine adenoviruses, canine distemper virus, herpes-simplex virus, reovirus, and members of the poxvirus family, such as vaccinia virus and myxoma virus, have been successfully tested for canine cancer therapy in preclinical and clinical settings. The success of the cancer virotherapy is dependent on the ability of oncolytic viruses to overcome the attacks of the host immune system, to preferentially infect and lyse cancer cells, and to initiate tumor-specific immunity. To date, several different strategies have been developed to overcome the antiviral host defense barriers. In our study, we used canine adipose-derived mesenchymal stem cells (cAdMSCs) as a “Trojan horse” for the delivery of oncolytic vaccinia virus Copenhagen strain to achieve maximum oncolysis against canine soft tissue sarcoma (CSTS) tumors. A single systemic administration of vaccinia virus-loaded cAdMSCs was found to be safe and led to the significant reduction and substantial inhibition of tumor growth in a CSTS xenograft mouse model. This is the first example that vaccinia virus-loaded cAdMSCs could serve as a therapeutic agent against CSTS tumors. KW - oncolytic virus KW - cancer KW - vaccinia virus KW - canine cancer cell lines KW - canine adipose-derived mesenchymal stem cells (cAdMSCs) KW - canine soft tissue sarcoma (CSTS) KW - canine cancer therapy Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-236007 VL - 12 IS - 7 ER - TY - THES A1 - Vyalkova, Anna T1 - Efficacy of approved Smallpox Vaccines in Human and Canine Cancer Therapy: Adipose - tissue derived Stem Cells (ADSC) take up VACV and serve as a protective vehicle for virus delivery to tumors T1 - Wirksamkeit zugelassener Pockenimpfstoffe in der Krebstherapie bei Menschen und Hunden: Aus Gewebe gewonnene Stammzellen (ADSC) nehmen VACV auf und dienen als schützendes Vehikel für den Viruseintrag in Tumore N2 - Cancer is one of the major causes of mortality in developed countries. In 2020, there were more than 19.3 million new cases of tumor malignancies worldwide, with more than 10 million deaths. The high rates of cancer cases and mortality necessitate extensive research and the development of novel cancer treatments and antitumor agents. In most cases, conventional treatment strategies for tumor therapy are based on chemotherapeutic treatment, which is supplemented with radiotherapy and/or surgical resection of solid tumors [1]. The use of chemotherapy for the treatment of cancer has significant side effects, the most dangerous of which is toxicity [2] [3]. Modern methods of treating tumors focus on specific drug delivery to the tumor site, actively targeting the tumor cells, as well as the reduction of side effects. One of the most promising current approaches is based on oncolytic viruses. Antitumor properties of viruses were documented at the beginning of the 20th century when some cancer patients recovered after acute viral infections, particularly influenza [4]. Vaccinia virus (VACV) is a member of the Poxviridae family, has natural antitumor properties, and provides a good basis for generating efficient recombinant oncolytic strains. Furthermore, VACV has never been shown to integrate into the host genome [5]. VACV is likely one of the safest and well-studied viruses due to extensive research being done in molecular biology and pathophysiology to investigate its potential as a vaccine for smallpox eradication programs. It has been administered to over 200 million people worldwide. VACV antitumor therapeutic effectiveness has been established in xenograft models with a variety of tumor types for human and canine cancers. Furthermore, recombinant oncolytic VACVs expressing genes encoding light-emitting proteins are a big improvement in a treatment strategy that combines tumor-specific therapies and diagnostics. Oncolytic virus treatments are effective in xenograft cancer models in mice, however, the significant improvements found in mice do not always translate to human cancer patients. These therapies should be tested in dogs with spontaneous cancer not only to offer well translatable information regarding the possible efficiency of viral therapy for human cancers but also to improve the health of our household pets as well. Spontaneous canine tumors are starting to be regarded as an essential model of human cancers that can reproduce the tumor microenvironment and immune response of cancer patients [6]. Just as data obtained in dog experiments can improve cancer therapy for human patients, these findings can also be used to improve treatment protocols in canine patients. Hundreds of studies and dozens of reviews have been published regarding the antitumor effects of various recombinants of VACV, but information on the anticancer features of initial, genetically-unmodified “naïve” VACV is still limited. In the first studies, we compared different wild-type, non-modified strains of VACV and tested their oncolytic properties on a panel of various cancer cells derived from different organs. In addition, we also tested a protection system based on the “Trojan horse” concept - using a combination of human Adipose tissue-derived Stem Cells (hADSC) and three different wild-type single plaque purified Vaccinia virus strains: W1, L1, and T1. We showed that all tested human cell lines (FaDu, MDA MB 231, HNT-13, HNT-35, and PC-3) are permissive to L0, W0, T0, L1, W1, and L1 infection. Furthermore, we tested the cytotoxicity of VACV in different cancer cell lines (A549, PC-3, MDA-MB 231, FaDu, HNT-13, HNT-25, and HNT-35). All strains lysed the cells, which was most visible at 96 hpi. We also showed that all tested strains could efficiently infect and multiply in hADSC at a high level. In our in vivo study, we tested the therapeutic efficacy of the wild-type Vaccinia viruses L1, W1, and T1 alone or in combination with hADSC. Wild-type VACV strains were tested for their oncolytic efficiency in human lung adenocarcinoma (A549) in a xenograft model. Treatment of A549 tumors with different doses of L1 and W1 as well as with a L1/ADSC or W1/ADSC combination led to significant tumor regression compared to the PBS control. Additionally, the treatment with L1 and W1 and the combination of L1/ADSC and W1/ADSC was well tolerated by the animals. In the case of the wild-type Tian Tan strain, results were not obtained due to the high cytotoxicity of this strain. Therefore, it should be attenuated for further studies. In the second part of the current study, we investigated the oncolytic effect of C1-opt1, W1 opt1, and L3-opt1 strains based on the wild-type Copenhagen, Wyeth, and Lister vaccines with additional expression of turboFP635. Replication and cytotoxicity assays demonstrated that all 3 viruses were able to infect, replicate in and kill canine tumor cell lines STSA-1 and CT1258 in a virus dose- and time- dependent fashion. Cytotoxicity and replication assays were also performed on cultured canine Adipose-derived Mesenchymal Stem Cells (cAdMSC). The results showed that the cells were lysed much slower than the tumor cells. It suggests that these cells can harbour the virus for a long-term period, allowing the virus to spread into the body and there is enough time to reach the primary tumor or metastases before the cell carrier is destroyed. The viral replication in cAdMSC in our study was lower than in canine cancer cells (STSA-1 and CT1258) at the same MOI. After being studied in cell culture, C1 opt1 and their combination with cAdMSC (C1-opt1/cAdMSC) were used in canine STSA 1 tumor bearing nude mice. We tested the oncolytic effect of the C1-opt1 virus alone and in combination with cAdMSC in the canine STSA-1 xenograft mouse model. Altogether, our findings have shown that both C1-opt1 and cAdMSC/C1-opt1 significantly reduced tumor size or eliminated the tumor. There was no significant difference between C1-opt1 alone and cAdMSC/C1-opt1. The virus particles were mostly found within the tumor after 24 dpi, some amount of virus particles were found in the lungs of mice injected with a combination of cAdMSC/C1-opt1 but not in the group injected with virus alone (cAdMSC might get stuck in the lungs and cause virus propagation there). Taken together, this study provided a proof-of-concept that hADSC/cAdMSC can be used as a carrier system for the “Trojan horse” concept. However, it should be confirmed in another experimental model system, such as canine patients. Moreover, these findings suggest that wild-type, non-modified strains of Vaccinia virus isolates can be considered promising candidates for oncolytic virotherapy, especially in combination with mesenchymal stem cells. N2 - Krebs wird zu einer der Hauptursachen für die Sterblichkeit in den Industrieländern. Im Jahr 2020 gab es weltweit mehr als 193 Millionen neue Fälle von tumormalen Erkrankungen mit mehr als 10 Millionen Todesfällen. Folglich erfordern die hohen Krebsfälle- und Mortalitätsraten umfangreiche Forschung und Entwicklung neuartiger Krebsbehandlungen und Antitumormittel. Konventionelle Behandlungsstrategien zur Tumortherapie basieren in den meisten Fällen auf einer chemotherapeutischen Behandlung, die durch Strahlentherapie und/oder chirurgische Resektion solider Tumoren ergänzt wird [1]. Die Verwendung von Chemotherapie zur Behandlung von Krebs hat erhebliche Nebenwirkungen, insbesondere die gefährlichste Intoxikation [2] [3]. ... KW - ADSC KW - AdMSC KW - Vaccinia virus (VACV) Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-253457 ER -