The objective of this proposal is to investigate the use of MYXV as potential oncolytic virotherapy agent against pancreatic cancer in preclinical animal models. The proposal focuses on MYXV, and is based on our previously published reports and supporting preliminary studies. MYXV is a rabbit-specific poxvirus with oncolytic activity against many types of human cancer models in vivo, including brain tumors, melanoma and rhabdoid tumors. In addition, we reported recently that MYXV is able to infect and kill pancreatic cancer cells in vitro. Our preliminary studies provided in the main body of the proposal show that MYXV exhibits potent oncolytic activity in both immunodeficient and immunocompetent animal models of pancreatic cancer. Based on these results, we seek to evaluate MYXV in combination with current chemotherapy regimens, especially gemcitabine, for pancreatic cancer. We hypothesize that MYXV will have potent oncolytic activity against pancreatic cancer in vivo and may be combined and/or engineered to enhance current chemotherapy treatments. Wildtype MYXV and recombinant MYXV """"""""armed"""""""" with chemosensitizing gene(s) will be evaluated as single agent therapies and in combination with current chemotherapy drugs approved for the treatment of pancreatic cancer. For the purposes of this grant, we propose to: 1) Evaluate the efficacy of wildtype MYXV oncolysis in murine models of pancreatic carcinoma. MYXV will be evaluated: a) as a single agent therapy compared to standard chemotherapies, b) in combination with gemcitabine and/or erlotinib, and as c) second line treatment therapy for chemotherapy resistant tumors. These experiments involve the use of the most common first line chemotherapies for pancreatic cancer and will therefore evaluate MYXV in the context of a clinically relevant scenario. Immunodeficient and immunocompetent murine models of pancreatic cancer will be established intraperitoneally (IP) and virus will be administered locally by the IP route. Tumor burden and survival curves will be compared between treatment groups in the presence or absence of gemcitabine and in gemcitabine refractory tumors to determine if MYXV virotherapy under the three regimes mentioned above results in an enhancement of therapeutic benefits as measured by tumor burden and survival. 2) Generate recombinant MYXV armed with chemosensitizing genes that will enhance gemcitabine-based chemotherapy. Recombinant """"""""armed"""""""" MYXVs that express deoxycytidine kinase (CDK) or the human equilibrative nucleoside transporter 1(hENT-1) will be engineered. These viruses will be characterized in vivo to determine if the expression of the transgenes enhances gemcitabine-based oncolysis of pancreatic cancer cells in the two models described above in Specific Aim 1. Tumor burden and survival curves will be compared between armed and wildtype MYXVs treated groups to determine if the use of an armed MYXV provides a therapeutic advantage over wildtype MYXV by enhancing gemcitabine-based chemotherapy in vivo. This proposal will be the first study to evaluate the oncolytic potential of MYXV in preclinical animal models of pancreatic cancer as single agent therapy and in combination with chemotherapy drugs, as well as the characterization of armed MYXV capable of sensitizing cells to gemcitabine chemotherapy. Thus, if successful, these pilot experiments will identify MYXV as an effective oncolytic virus that can be further developed as a novel, safe virotherapy for the treatment of pancreatic cancer. In particular, if the results are as positive as we anticipate, and given the excellent safety profile of MYXV, we will pursue the production of clinical grade stocks of MYXV and the filing for an investigational new drug application (IND) at the completion of the proposed study.

Public Health Relevance

Myxoma virus (MYXV), a rabbit-specific virus with no history of pathogenicity in humans, can specifically infect and kill a wide spectrum of human cancer cells. The purpose of this proposal is to investigate the potential use of MYXV as a novel virotherapy compatible with current chemotherapy regimens for pancreatic cancer. Currently, chemotherapy treatments for pancreatic cancer are limited and do not significantly improve survival. These experiments will determine if MYXV is a feasible oncolytic virus to use in combination with gemcitabine-based chemotherapies and if the addition of MYXV to these therapies results in an enhanced anti-tumor effect and survival benefit. If successful, these experiments will provide a new adjunct therapy for the treatment of pancreatic cancer involving wildtype or armed MYXV that will chemosensitize the infected pancreatic cancer cells to the toxic effects of gemcitabine resulting in an enhancement of the overall therapeutic effect. In particular, if the results are as positive as we anticipate, and given the safety of MYXV, we will pursue the production of clinical grade stocks of MYXV and the filing for an investigational new drug application (IND) to the Food and Drug Administration (FDA) at the completion of the proposed study.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Exploratory/Developmental Grants (R21)
Project #
1R21CA149869-01A1
Application #
8044924
Study Section
Developmental Therapeutics Study Section (DT)
Program Officer
Arya, Suresh
Project Start
2011-01-01
Project End
2012-12-31
Budget Start
2011-01-01
Budget End
2011-12-31
Support Year
1
Fiscal Year
2011
Total Cost
$191,183
Indirect Cost
Name
University of Florida
Department
Genetics
Type
Schools of Medicine
DUNS #
969663814
City
Gainesville
State
FL
Country
United States
Zip Code
32611
Chan, Winnie M; McFadden, Grant (2014) Oncolytic Poxviruses. Annu Rev Virol 1:119-141
Doty, Rosalinda A; Liu, Jia; McFadden, Grant et al. (2013) Histological evaluation of intratumoral myxoma virus treatment in an immunocompetent mouse model of melanoma. Oncolytic Virother 2:1-17
Sallam, Mohamed F; Al Ahmed, Azzam M; Abdel-Dayem, Mahmoud S et al. (2013) Ecological niche modeling and land cover risk areas for rift valley fever vector, culex tritaeniorhynchus giles in Jazan, Saudi Arabia. PLoS One 8:e65786
Zemp, Franz J; Lun, Xueqing; McKenzie, Brienne A et al. (2013) Treating brain tumor-initiating cells using a combination of myxoma virus and rapamycin. Neuro Oncol 15:904-20
Zemp, Franz J; McKenzie, Brienne A; Lun, Xueqing et al. (2013) Resistance to oncolytic myxoma virus therapy in nf1(-/-)/trp53(-/-) syngeneic mouse glioma models is independent of anti-viral type-I interferon. PLoS One 8:e65801
Chan, Winnie M; Rahman, Masmudur M; McFadden, Grant (2013) Oncolytic myxoma virus: the path to clinic. Vaccine 31:4252-8
Jenne, Craig N; Wong, Connie H Y; Zemp, Franz J et al. (2013) Neutrophils recruited to sites of infection protect from virus challenge by releasing neutrophil extracellular traps. Cell Host Microbe 13:169-80
Wennier, Sonia Tusell; Liu, Jia; Li, Shoudong et al. (2012) Myxoma virus sensitizes cancer cells to gemcitabine and is an effective oncolytic virotherapeutic in models of disseminated pancreatic cancer. Mol Ther 20:759-68
Li, Shoudong; Tong, Jessica; Rahman, Masmudur M et al. (2012) Oncolytic virotherapy for ovarian cancer. Oncolytic Virother 1:1-21
Madlambayan, Gerard J; Bartee, Eric; Kim, Manbok et al. (2012) Acute myeloid leukemia targeting by myxoma virus in vivo depends on cell binding but not permissiveness to infection in vitro. Leuk Res 36:619-24

Showing the most recent 10 out of 12 publications