Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael B. Steele is active.

Publication


Featured researches published by Michael B. Steele.


Scientific Reports | 2013

Induction of antiviral genes by the tumor microenvironment confers resistance to virotherapy

Yu Ping Liu; Lukkana Suksanpaisan; Michael B. Steele; Stephen J. Russell; Kah Whye Peng

Oncolytic viruses obliterate tumor cells in tissue culture but not against the same tumors in vivo. We report that macrophages can induce a powerfully protective antiviral state in ovarian and breast tumors, rendering them resistant to oncolytic virotherapy. These tumors have activated JAK/STAT pathways and expression of interferon-stimulated genes (ISGs) is upregulated. Gene expression profiling (GEP) of human primary ovarian and breast tumors confirmed constitutive activation of ISGs. The tumors were heavily infiltrated with CD68+ macrophages. Exposure of OV-susceptible tumor cell lines to conditioned media from RAW264.7 or primary macrophages activated antiviral ISGs, JAK/STAT signaling and an antiviral state. Anti-IFN antibodies and shRNA knockdown studies show that this effect is mediated by an extremely low concentration of macrophage-derived IFNβ. JAK inhibitors reversed the macrophage-induced antiviral state. This study points to a new role for tumor-associated macrophages in the induction of a constitutive antiviral state that shields tumors from viral attack.


Human gene therapy. Clinical development | 2013

Safety Studies on Intravenous Administration of Oncolytic Recombinant Vesicular Stomatitis Virus in Purpose-Bred Beagle Dogs

Amy K. LeBlanc; Shruthi Naik; Gina Galyon; Nathan Jenks; Michael B. Steele; Kah Whye Peng; Mark J. Federspiel; Robert L. Donnell; Stephen J. Russell

VSV-IFNβ-NIS is a novel recombinant oncolytic vesicular stomatitis virus (VSV) with documented efficacy and safety in preclinical murine models of cancer. To facilitate clinical translation of this promising oncolytic therapy in patients with disseminated cancer, we are utilizing a comparative oncology approach to gather data describing the safety and efficacy of systemic VSV-IFNβ-NIS administration in dogs with naturally occurring cancer. In support of this, we executed a dose-escalation study in purpose-bred dogs to determine the maximum tolerated dose (MTD) of systemic VSV-hIFNβ-NIS, characterize the adverse event profile, and describe routes and duration of viral shedding in healthy, immune-competent dogs. The data indicate that an intravenous dose of 10(10) TCID50 is well tolerated in dogs. Expected adverse events were mild to moderate fever, self-limiting nausea and vomiting, lymphopenia, and oral mucosal lesions. Unexpected adverse events included prolongation of partial thromboplastin time, development of bacterial urinary tract infection, and scrotal dermatitis, and in one dog receiving 10(11) TCID50 (10 × the MTD), the development of severe hepatotoxicity and symptoms of shock leading to euthanasia. Viral shedding data indicate that detectable viral genome in blood diminishes rapidly with anti-VSV neutralizing antibodies detectable in blood as early as day 5 postintravenous virus administration. While low levels of viral genome copies were detectable in plasma, urine, and buccal swabs of dogs treated at the MTD, no infectious virus was detectable in plasma, urine, or buccal swabs at any of the doses tested. These studies confirm that VSV can be safely administered systemically in dogs, justifying the use of oncolytic VSV as a novel therapy for the treatment of canine cancer.


PLOS ONE | 2013

Mathematical Model for Radial Expansion and Conflation of Intratumoral Infectious Centers Predicts Curative Oncolytic Virotherapy Parameters

Kent R. Bailey; Amber C. Kirk; Shruthi Naik; Rebecca Nace; Michael B. Steele; Lukkana Suksanpaisan; Xing Li; Mark J. Federspiel; Kah Whye Peng; David Kirk; Stephen J. Russell

Simple, inductive mathematical models of oncolytic virotherapy are needed to guide protocol design and improve treatment outcomes. Analysis of plasmacytomas regressing after a single intravenous dose of oncolytic vesicular stomatitis virus in myeloma animal models revealed that intratumoral virus spread was spatially constrained, occurring almost exclusively through radial expansion of randomly distributed infectious centers. From these experimental observations we developed a simple model to calculate the probability of survival for any cell within a treated tumor. The model predicted that small changes to the density of initially infected cells or to the average maximum radius of infected centers would have a major impact on treatment outcome, and this was confirmed experimentally. The new model provides a useful and flexible tool for virotherapy protocol optimization.


Human gene therapy. Clinical development | 2016

Safety Studies in Tumor and Non-Tumor-Bearing Mice in Support of Clinical Trials Using Oncolytic VSV-IFNβ-NIS

Lianwen Zhang; Michael B. Steele; Nathan Jenks; Jacquelyn A. Grell; Lukkana Suksanpaisan; Shruthi Naik; Mark J. Federspiel; Martha Q. Lacy; Stephen J. Russell; Kah Whye Peng

Oncolytic VSV-IFNβ-NIS is selectively destructive to tumors. Here, we present the IND enabling preclinical rodent studies in support of clinical testing of vesicular stomatitis virus (VSV) as a systemic therapy. Efficacy studies showed dose-dependent tumor regression in C57BL/KaLwRij mice bearing syngeneic 5TGM1 plasmacytomas after systemic VSV administration. In contrast, the virus was effective at all doses tested against human KAS6/1 xenografts in SCID mice. Intravenous administration of VSV-mIFNβ-NIS is well tolerated in C57BL/6 mice up to 5 × 10(10) TCID50 (50% tissue culture infective dose)/kg with no neurovirulence, no cytokine storm, and no abnormalities in tissues. Dose-limiting toxicities included elevated transaminases, thrombocytopenia, and lymphopenia. Inactivated viral particles did not cause hepatic toxicity. Intravenously administered VSV was preferentially sequestered by macrophages in the spleen and liver. Quantitative RT-PCR analysis for total viral RNA on days 2, 7, 21, and 58 showed highest VSV RNA in day 2 samples; highest in spleen, liver, lung, lymph node, kidney, gonad, and bone marrow. No infectious virus was recovered from tissues at any time point. The no observable adverse event level and maximum tolerated dose of VSV-mIFNβ-NIS in C57BL/6 mice are 10(10) TCID50/kg and 5 × 10(10) TCID50/kg, respectively. Clinical translation of VSV-IFNβ-NIS is underway in companion dogs with cancer and in human patients with relapsed hematological malignancies and endometrial cancer.


Gynecologic Oncology | 2014

Oncolytic measles and vesicular stomatitis virotherapy for endometrial cancer

Yu Ping Liu; Michael B. Steele; Lukkana Suksanpaisan; Mark J. Federspiel; Stephen J. Russell; Kah Whye Peng; Jamie N. Bakkum-Gamez

OBJECTIVE Current adjuvant therapy for advanced-stage, recurrent, and high-risk endometrial cancer (EC) has not reduced mortality from this malignancy, and novel systemic therapies are imperative. Oncolytic viral therapy has been shown to be effective in the treatment of gynecologic cancers, and we investigated the in vitro and in vivo efficacy of the Edmonston strain of measles virus (MV) and vesicular stomatitis virus (VSV) on EC. METHODS Human EC cell lines (HEC-1-A, Ishikawa, KLE, RL95-2, AN3 CA, ARK-1, ARK-2, and SPEC-2) were infected with Edmonston strain MV expressing the thyroidal sodium iodide symporter, VSV expressing either human or murine IFN-β, or recombinant VSV with a methionine deletion at residue 51 of the matrix protein and expressing the sodium iodide symporter. Xenografts of HEC-1-A and AN3 CA generated in athymic mice were treated with intratumoral MV or VSV or intravenous VSV. RESULTS In vitro, all cell lines were susceptible to infection and cell killing by all 3 VSV strains except KLE. In addition, the majority of EC cell lines were defective in their ability to respond to type I IFN. Intratumoral VSV-treated tumors regressed more rapidly than MV-treated tumors, and intravenous VSV resulted in effective tumor control in 100% of mice. Survival was significantly longer for mice treated with any of the 3 VSV strains compared with saline. CONCLUSION VSV is clearly more potent in EC oncolysis than MV. A phase 1 clinical trial of VSV in EC is warranted.


Molecular Therapy | 2016

Perfusion Pressure Is a Critical Determinant of the Intratumoral Extravasation of Oncolytic Viruses

Amber Miller; Rebecca Nace; Camilo Ayala-Breton C; Michael B. Steele; Kent R. Bailey; Kah Whye Peng; Stephen J. Russell

Antitumor efficacy of oncolytic virotherapy is determined by the density and distribution of infectious centers within the tumor, which may be heavily influenced by the permeability and blood flow in tumor microvessels. Here, we investigated whether systemic perfusion pressure, a key driver of tumor blood flow, could influence the intratumoral extravasation of systemically administered oncolytic vesicular stomatitis virus (VSV) in myeloma tumor-bearing mice. Exercise was used to increase mean arterial pressure, and general anesthesia to decrease it. A recombinant VSV expressing the sodium iodide symporter (NIS), which concentrates radiotracers at sites of infection, was administered intravenously to exercising or anesthetized mice, and nuclear NIS reporter gene imaging was used to noninvasively track the density and spatial distribution of intratumoral infectious centers. Anesthesia resulted in decreased intratumoral infection density, while exercise increased the density and uniformity of infectious centers. Perfusion state also had a significant impact on the antitumor efficacy of the VSV therapy. In conclusion, quantitative dynamic radiohistologic imaging was used to noninvasively interrogate delivery of oncolytic virotherapy, highlighting the critical importance of perfusion pressure as a driver of intratumoral delivery and efficacy of oncolytic viruses.


Molecular Therapy | 2016

Robust Oncolytic Virotherapy Induces Tumor Lysis Syndrome and Associated Toxicities in the MPC-11 Plasmacytoma Model

Lianwen Zhang; Michael B. Steele; Nathan Jenks; Jacquelyn A. Grell; Marshall Behrens; Rebecca Nace; Shruthi Naik; Mark J. Federspiel; Stephen J. Russell; Kah Whye Peng

Tumor-selective oncolytic vesicular stomatitis viruses (VSVs) are being evaluated in clinical trials. Here, we report that the MPC-11 murine plasmacytoma model is so extraordinarily susceptible to oncolytic VSVs that a low dose of virus leads to extensive intratumoral viral replication, sustained viremia, intravascular coagulation, and a rapidly fatal tumor lysis syndrome (TLS). Rapid softening, shrinkage and hemorrhagic necrosis of flank tumors was noted within 1-2 days after virus administration, leading to hyperkalemia, hyperphosphatemia, hypocalcemia, hyperuricemia, increase in plasma cell free DNA, lymphopenia, consumptive coagulopathy, increase in fibrinogen degradation products, decreased liver function tests, dehydration, weight loss, and euthanasia or death after 5-8 days. Secondary viremia was observed but viral replication in normal host tissues was not detected. Toxicity could be mitigated by using VSVs with slowed replication kinetics, and was less marked in animals with smaller flank tumors. The MPC-11 tumor represents an interesting model to further study the complex interplay of robust intratumoral viral replication, tumor lysis, and associated toxicities in cases where tumors are highly responsive to oncolytic virotherapy.


Molecular Cancer Therapeutics | 2018

Comparative Oncology Evaluation of Intravenous Recombinant Oncolytic Vesicular Stomatitis Virus Therapy in Spontaneous Canine Cancer.

Shruthi Naik; Gina Galyon; Nathan Jenks; Michael B. Steele; Amber Miller; Sara D. Allstadt; Lukkana Suksanpaisan; Kah Whye Peng; Mark J. Federspiel; Stephen J. Russell; Amy K. LeBlanc

Clinical translation of intravenous therapies to treat disseminated or metastatic cancer is imperative. Comparative oncology, the evaluation of novel cancer therapies in animals with spontaneous cancer, can be utilized to inform and accelerate clinical translation. Preclinical murine studies demonstrate that single-shot systemic therapy with a vesicular stomatitis virus (VSV)-IFNβ-NIS, a novel recombinant oncolytic VSV, can induce curative remission in tumor-bearing mice. Clinical translation of VSV-IFNβ-NIS therapy is dependent on comprehensive assessment of clinical toxicities, virus shedding, pharmacokinetics, and efficacy in clinically relevant models. Dogs spontaneously develop cancer with comparable etiology, clinical progression, and response to therapy as human malignancies. A comparative oncology study was carried out to investigate feasibility and tolerability of intravenous oncolytic VSV-IFNβ-NIS therapy in pet dogs with spontaneous cancer. Nine dogs with various malignancies were treated with a single intravenous dose of VSV-IFNβ-NIS. Two dogs with high-grade peripheral T-cell lymphoma had rapid but transient remission of disseminated disease and transient hepatotoxicity that resolved spontaneously. There was no shedding of infectious virus. Correlative pharmacokinetic studies revealed elevated levels of VSV RNA in blood in dogs with measurable disease remission. This is the first evaluation of intravenous oncolytic virus therapy for spontaneous canine cancer, demonstrating that VSV-IFNβ-NIS is well-tolerated and safe in dogs with advanced or metastatic disease. This approach has informed clinical translation, including dose and target indication selection, leading to a clinical investigation of intravenous VSV-IFNβ-NIS therapy, and provided preliminary evidence of clinical efficacy and potential biomarkers that correlate with therapeutic response. Mol Cancer Ther; 17(1); 316–26. ©2017 AACR.


Molecular Therapy - Oncolytics | 2018

Preclinical Development of Oncolytic Immunovirotherapy for Treatment of HPVPOS Cancers

Lukkana Suksanpaisan; Rong Xu; Mulu Z. Tesfay; Carolyn Bomidi; Stefan Hamm; Rianna Vandergaast; Nathan Jenks; Michael B. Steele; Ayuko Ota-Setlik; Hinna Akhtar; Amara Luckay; Rebecca M. Nowak; Kah Whye Peng; John H. Eldridge; David K. Clarke; Stephen J. Russell; Rosa Maria Diaz

Immunotherapy for HPVPOS malignancies is attractive because well-defined, viral, non-self tumor antigens exist as targets. Several approaches to vaccinate therapeutically against HPV E6 and E7 antigens have been adopted, including viral platforms such as VSV. A major advantage of VSV expressing these antigens is that VSV also acts as an oncolytic virus, leading to direct tumor cell killing and induction of effective anti-E6 and anti-E7 T cell responses. We have also shown that addition of immune adjuvant genes, such as IFNβ, further enhances safety and/or efficacy of VSV-based oncolytic immunovirotherapies. However, multiple designs of the viral vector are possible—with respect to levels of immunogen expression and method of virus attenuation—and optimal designs have not previously been tested head-to-head. Here, we tested three different VSV engineered to express a non-oncogenic HPV16 E7/6 fusion protein for their immunotherapeutic and oncolytic properties. We assessed their profiles of efficacy and toxicity against HPVPOS and HPVNEG murine tumor models and determined the optimal route of administration. Our data show that VSV is an excellent platform for the oncolytic immunovirotherapy of tumors expressing HPV target antigens, combining a balance of efficacy and safety suitable for evaluation in a first-in-human clinical trial.


Molecular Therapy | 2015

74. The Mechanism of Hepatotoxicity from IV Administration of High Dose VSV

Lianwen Zhang; Michael B. Steele; Marshall B. Behrens; Mark J. Federspiel; Stephen J. Russell; Kah Whye Peng

Vesicular Stomatitis Virus (VSV) is a potent replication competent oncolytic virus (OV). Most individuals do not have pre-existing anti-VSV antibodies, making VSV an ideal OV for treatment of disseminated cancer. To tailor the virus for systemic use, VSV-IFNb-NIS was attenuated through genetic modification to encode interferon beta which restricts virus replication in nontransformed cells and a sodium iodide symporter (NIS) reporter gene to enable real time noninvasive imaging of the sites of virus infection in patients. Preclinical studies in mice show that, in contrast to expectations, dose limiting toxicity was hepatotoxicity, not neurotoxicity. To investigate the mechanism(s) underlying VSV induced hepatotoxicity, we first sought to determine if liver damage was due to interactions with viral particles or required virus infection.C57BL/6 mice were given saline, 109 TCID50 VSV-mIFNb-NIS or equivalent amounts of inactivated VSV-mIFNb-NIS intravenously. There was a significant increase in ALT and AST at 24h while no significant change was found in the animals receiving inactivated VSV. Studies with adenoviruses showed that Ad-induced hepatotoxicity was attenuated by depletion of macrophages in the liver and spleen using clodronate liposomes. Our studies in clodronate treated C57BL/6 mice showed even higher levels of ALT and AST enzymes in the blood with significantly increased cell damage in the liver and spleen by 24h if mice were given 109 VSV-IFNb-NIS. In contrast, ALT/AST levels did not change significantly in mice given heat inactivated VSV (104 infectious units but the same number of viral particles as the 109 stock). Similar data were also observed in Balb/c mice. In conclusion, our studies indicate that dose limiting toxicity from IV administration of high dose (4×1010 TCID50/kg) VSV-mIFNb-NIS in a mouse is not neurotoxicity, but hepatotoxicity. The mechanism of the hepatotoxicity is direct infection and killing of hepatocytes by live virus, and not due to the interaction of viral particles with macrophages and Kupffer cells. Hence, in contrast to adenovirus dosing (viral particles/kg), our data suggest that VSV dosing should be calibrated in units of infectious virus (TCID50/kg) and not numbers of viral particles per test subject. View Large Image | Download PowerPoint Slide View Large Image | Download PowerPoint Slide

Collaboration


Dive into the Michael B. Steele's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gina Galyon

University of Tennessee

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge