27 results on '"Lisa A. Santry"'
Search Results
2. Supplementary Data Figure 1 from Combining Vascular Normalization with an Oncolytic Virus Enhances Immunotherapy in a Preclinical Model of Advanced-Stage Ovarian Cancer
- Author
-
Jim Petrik, Byram W. Bridle, Sarah K. Wootton, Jack Lawler, Pierre P. Major, Amanda W.K. AuYeung, Jacob P. van Vloten, Lisa A. Santry, and Kathy Matuszewska
- Abstract
Supplementary Figure 1 Supplementary Data, Figure 1. Combination therapy results in decreased ascites fluid volume. At euthanasia, ascites fluid was aspirated from the abdomen of control and treated mice and the volume was quantified. Combined treatment with 3TSR and NDV(F3aa) resulted in a significant reduction in ascites volume compared to controls or other treatment groups. Means+/- SEM are shown. Data represents results from N=12 animals per experimental group. Data were analyzed by one-way analysis of variance with Tukey's multiple comparison test; *P
- Published
- 2023
3. Data from Combining Vascular Normalization with an Oncolytic Virus Enhances Immunotherapy in a Preclinical Model of Advanced-Stage Ovarian Cancer
- Author
-
Jim Petrik, Byram W. Bridle, Sarah K. Wootton, Jack Lawler, Pierre P. Major, Amanda W.K. AuYeung, Jacob P. van Vloten, Lisa A. Santry, and Kathy Matuszewska
- Abstract
Purpose:Intravenous delivery of oncolytic viruses often leads to tumor vascular shutdown, resulting in decreased tumor perfusion and elevated tumor hypoxia. We hypothesized that using 3TSR to normalize tumor vasculature prior to administration of an oncolytic Newcastle disease virus (NDV) would enhance virus delivery and trafficking of immunologic cell subsets to the tumor core, resulting in systemically enhanced immunotherapy and regression of advanced-stage epithelial ovarian cancer (EOC).Experimental Design:Using an orthotopic, syngeneic mouse model of advanced-stage EOC, we pretreated mice with 3TSR (4 mg/kg per day) alone or followed by combination with fusogenic NDV(F3aa) (1.0 × 108 plaque-forming units).Results:Treatment with 3TSR normalized tumor vasculature, enhanced blood perfusion of primary EOC tumors, and induced disease regression. Animals treated with combination therapy had the greatest reduction in primary tumor mass, ascites accumulation, and secondary lesions (50% of mice were completely devoid of peritoneal metastases). Combining 3TSR + NDV(F3aa) led to enhanced trafficking of immunologic cells into the primary tumor core.Conclusions:We have shown, for the first time, that NDV, like other oncolytic viruses, is a potent mediator of acute vascular shutdown and that preventing this through vascular normalization can promote regression in a preclinical model of advanced-stage ovarian cancer. This challenges the current focus on induction of intravascular thrombosis as a requisite for successful oncolytic virotherapy.See related commentary by Bykov and Zamarin, p. 1446
- Published
- 2023
4. Addition of an Fc-IgG induces receptor clustering and increases the in vitro efficacy and in vivo anti-tumor properties of the thrombospondin-1 type I repeats (3TSR) in a mouse model of advanced stage ovarian cancer
- Author
-
Sarah K. Wootton, Simone ten Kortenaar, Jack Lawler, Jim Petrik, Madison Pereira, Lisa A. Santry, Byram W. Bridle, Kathy Matuszewska, Duncan Petrik, and Kin-Ming Lo
- Subjects
CD36 ,Angiogenesis Inhibitors ,Carcinoma, Ovarian Epithelial ,Thrombospondin 1 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Receptor ,030304 developmental biology ,Ovarian Neoplasms ,0303 health sciences ,Neovascularization, Pathologic ,biology ,business.industry ,Obstetrics and Gynecology ,medicine.disease ,In vitro ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,Oncology ,Apoptosis ,Immunoglobulin G ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Receptor clustering ,business ,Ovarian cancer - Abstract
Objectives Tumor vasculature is structurally abnormal, with anatomical deformities, reduced pericyte coverage and low tissue perfusion. As a result of this vascular dysfunction, tumors are often hypoxic, which is associated with an aggressive tumor phenotype, and reduced delivery of therapeutic compounds to the tumor. We have previously shown that a peptide containing the thrombospondin-1 type I repeats (3TSR) specifically targets tumor vessels and induces vascular normalization in a mouse model of epithelial ovarian cancer (EOC). However, due to its small size, 3TSR is rapidly cleared from circulation. We now introduce a novel construct with the 3TSR peptide fused to the C-terminus of each of the two heavy chains of the Fc region of human IgG1 (Fc3TSR). We hypothesize that Fc3TSR will have greater anti-tumor activity in vitro and in vivo compared to the native compound. Methods Fc3TSR was evaluated in vitro using proliferation and apoptosis assays to investigate differences in efficacy compared to native 3TSR. In light of the multivalency of Fc3TSR, we also investigate whether it induces greater clustering of its functional receptor, CD36. We also compare the compounds in vivo using an orthotopic, syngeneic mouse model of advanced stage EOC. The impact of the two compounds on changes to tumor vasculature morphology was also investigated. Results Fc3TSR significantly decreased the viability and proliferative potential of EOC cells and endothelial cells in vitro compared to native 3TSR. High-resolution imaging followed by image correlation spectroscopy demonstrated enhanced clustering of the CD36 receptor in cells treated with Fc3TSR. This was associated with enhanced downstream signaling and greater in vitro and in vivo cellular responses. Fc3TSR induced greater vascular normalization and disease regression compared to native 3TSR in an orthotopic, syngeneic mouse model of advanced stage ovarian cancer. Conclusion The development of Fc3TSR which is greater in size, stable in circulation and enhances receptor activation compared to 3TSR, facilitates its translational potential as a therapy in the treatment of metastatic advanced stage ovarian cancer.
- Published
- 2022
5. Antibody-based protection against respiratory syncytial virus in mice and their offspring through vectored immunoprophylaxis
- Author
-
Amira D. Rghei, Jacob G. E. Yates, Jordyn A. Lopes, Xuiaoyan Zhan, Matthew M. Guilleman, Yanlong Pei, Laura P. van Lieshout, Lisa A. Santry, Byram W. Bridle, Khalil Karimi, Brad Thompson, Leonardo Susta, James E. Crowe, and Sarah K. Wootton
- Subjects
Genetics ,Molecular Medicine ,Molecular Biology - Published
- 2023
6. Production and purification of high-titer OrfV for preclinical studies in vaccinology and cancer therapy
- Author
-
Grant McFadden, Jessica A. Minott, Thomas M. McAusland, Byram W. Bridle, Jacob P. van Vloten, Sarah K. Wootton, Lisa A. Santry, Jim Petrik, and Joelle C. Ingrao
- Subjects
Cancer therapy ,OrfV ,QH426-470 ,Biology ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Protocol ,Genetics ,medicine ,High titer ,gradient ultracentrifugation ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,QH573-671 ,tangential flow filtration ,Cancer ,biology.organism_classification ,medicine.disease ,Virology ,3. Good health ,Parapoxvirus ovis ,poxvirus ,030220 oncology & carcinogenesis ,virus purification ,Parapoxvirus ,cancer therapy ,Molecular Medicine ,vaccinology ,Cytology - Abstract
Poxviruses have been used extensively as vaccine vectors for human and veterinary medicine and have recently entered the clinical realm as immunotherapies for cancer. We present a comprehensive method for producing high-quality lots of the poxvirus Parapoxvirus ovis (OrfV) for use in preclinical models of vaccinology and cancer therapy. OrfV is produced using a permissive sheep skin-derived cell line and is released from infected cells by repeated freeze-thaw combined with sonication. We present two methods for isolation and purification of bulk virus. Isolated virus is concentrated to high titer using polyethylene glycol to produce the final in vivo-grade product. We also describe methods for quantifying OrfV infectious virions and determining genomic copy number to evaluate virus stocks. The methods herein will provide researchers with the ability to produce high-quality, high-titer OrfV for use in preclinical studies, and support the translation of OrfV-derived technologies into the clinic., Graphical abstract, Methods for producing high-titer, high-purity OrfV, a poxvirus of the Parapoxvirus ovis genera and a promising oncolytic and vaccine vector, are described, as are methods for accurate titration of OrfV. These methods will provide researchers with the knowledge to produce in vivo-grade OrfV for preclinical studies.
- Published
- 2021
7. AAV-mediated delivery of actoxumab and bezlotoxumab results in serum and mucosal antibody concentrations that provide protection from C. difficile toxin challenge
- Author
-
Brad Thompson, Brenna A Y Stevens, Amira D Rghei, Laura P. van Lieshout, Sarah K. Wootton, Matthew M. Guilleman, Lisa A. Santry, and Yanlong Pei
- Subjects
0301 basic medicine ,biology ,medicine.drug_class ,Toxin ,Antibiotics ,Enterotoxin ,Clostridium difficile ,medicine.disease_cause ,Monoclonal antibody ,Virus ,3. Good health ,Microbiology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Bezlotoxumab ,030220 oncology & carcinogenesis ,Genetics ,biology.protein ,medicine ,Molecular Medicine ,Antibody ,Molecular Biology - Abstract
Clostridium difficile is the leading cause of antibiotic-associated nosocomial diarrhea in the developed world. When the host-associated colon microbiome is disrupted by the ingestion of antibiotics, C. difficile spores can germinate, resulting in infection. C. difficile secretes enterotoxin A (TcdA) and cytotoxin B (TcdB) that are responsible for disease pathology. Treatment options are limited as the bacterium demonstrates resistance to many antibiotics, and even with antibacterial therapies, recurrences of C. difficile are common. Actotoxumab and bezlotoxumab are human monoclonal antibodies that bind and neutralize TcdA and TcdB, respectively. In 2016, the US food and drug administration (FDA) approved bezlotoxumab for use in the prevention of C. difficile infection recurrence. To ensure the long-term expression of antibodies, gene therapy can be used. Here, adeno-associated virus (AAV)6.2FF, a novel triple mutant of AAV6, was engineered to express either actotoxumab or bezlotoxumab in mice and hamsters. Both antibodies expressed at greater than 90 μg/mL in the serum and were detected at mucosal surfaces in both models. Hundred percent of mice given AAV6.2FF-actoxumab survived a lethal dose of TcdA. This proof of concept study demonstrates that AAV-mediated expression of C. difficile toxin antibodies is a viable approach for the prevention of recurrent C. difficile infections.
- Published
- 2021
8. Tumour vasculature: Friend or foe of oncolytic viruses?
- Author
-
Jim Petrik, Ashley A. Stegelmeier, Kathy Matuszewska, Sarah K. Wootton, Robert C. Mould, Lisa A. Santry, Thomas M. McAusland, Byram W. Bridle, Jessica A. Minott, Jason P. Knapp, Pierre Major, and Jacob P. van Vloten
- Subjects
0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Immunology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,Vascular collapse ,medicine ,Humans ,Immunology and Allergy ,Oncolytic Virotherapy ,business.industry ,Effector ,3. Good health ,Oncolytic virus ,Oncolytic Viruses ,030104 developmental biology ,Cytokine ,030220 oncology & carcinogenesis ,Cancer research ,Immunotherapy ,business ,Shut down - Abstract
In the past two decades there have been substantial advances in understanding the anti-cancer mechanisms of oncolytic viruses (OVs). OVs can mediate their effects directly, by preferentially infecting and killing tumour cells. Additionally, OVs can indirectly generate anti-tumour immune responses. These differing mechanisms have led to a paradoxical divergence in strategies employed to further increase the potency of oncolytic virotherapies. On one hand, the tumour neovasculature is seen as a vital lifeline to the survival of the tumour, leading some to use OVs to target the tumour vasculature in hopes to starve cancers. Therapeutics causing vascular collapse can potentiate tumour hypoxia, nutrient restriction and pro-inflammatory cytokine release, which has shown promise in oncological studies. On the other hand, the same vasculature plays an important role for the dissemination of OVs, trafficking of effector cells and other therapeutics, which has prompted researchers to find ways of normalizing the vasculature to enhance infiltration of leukocytes and delivery of therapeutic agents. This article describes the recent developments of therapies aimed to shut down versus normalize tumour vasculature in order to inform researchers striving to optimize OV-based therapies.
- Published
- 2020
9. Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer
- Author
-
Jacob P van Vloten, Kathy Matuszewska, Mark A A Minow, Jessica A Minott, Lisa A Santry, Madison Pereira, Ashley A Stegelmeier, Thomas M McAusland, Elaine M Klafuric, Khalil Karimi, Joseph Colasanti, D Grant McFadden, James J Petrik, Byram W Bridle, and Sarah K Wootton
- Subjects
Pharmacology ,Oncolytic Virotherapy ,Ovarian Neoplasms ,Cancer Research ,Sheep ,Immunology ,Orf virus ,Carcinoma, Ovarian Epithelial ,Killer Cells, Natural ,Disease Models, Animal ,Mice ,Oncolytic Viruses ,Oncology ,Cell Line, Tumor ,Molecular Medicine ,Immunology and Allergy ,Animals ,Humans ,Female ,Licensure - Abstract
BackgroundNovel therapies are needed to improve outcomes for women diagnosed with ovarian cancer. Oncolytic viruses are multifunctional immunotherapeutic biologics that preferentially infect cancer cells and stimulate inflammation with the potential to generate antitumor immunity. Herein we describe Parapoxvirus ovis (Orf virus (OrfV)), an oncolytic poxvirus, as a viral immunotherapy for ovarian cancer.MethodsThe immunotherapeutic potential of OrfV was tested in the ID8 orthotopic mouse model of end-stage epithelial ovarian carcinoma. Immune cell profiling, impact on secondary lesion development and survival were evaluated in OrfV-treated mice as well as in Batf3 knockout, mice depleted of specific immune cell subsets and in mice where the primary tumor was removed. Finally, we interrogated gene expression datasets from primary human ovarian tumors from the International Cancer Genome Consortium database to determine whether the interplay we observed between natural killer (NK) cells, classical type 1 dendritic cells (cDC1s) and T cells exists and influences outcomes in human ovarian cancer.ResultsOrfV was an effective monotherapy in a murine model of advanced-stage epithelial ovarian cancer. OrfV intervention relied on NK cells, which when depleted abrogated antitumor CD8+ T-cell responses. OrfV therapy was shown to require cDC1s in experiments with BATF3 knockout mice, which do not have mature cDC1s. Furthermore, cDC1s governed antitumor NK and T-cell responses to mediate antitumor efficacy following OrfV. Primary tumor removal, a common treatment option in human patients, was effectively combined with OrfV for optimal therapeutic outcome. Analysis of human RNA sequencing datasets revealed that cDC1s correlate with NK cells in human ovarian cancer and that intratumoral NK cells correlate positively with survival.ConclusionsThe data herein support the translational potential of OrfV as an NK stimulating immunotherapeutic for the treatment of advanced-stage ovarian cancer.
- Published
- 2022
10. Quantifying Antigen-Specific T Cell Responses When Using Antigen-Agnostic Immunotherapies
- Author
-
Byram W. Bridle, Sarah K. Wootton, Jim Petrik, Lisa A. Santry, Jacob P. van Vloten, Grant McFadden, Thomas M. McAusland, and Khalil Karimi
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,T cell ,Major histocompatibility complex ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Genetics ,Medicine ,lcsh:QH573-671 ,Molecular Biology ,biology ,lcsh:Cytology ,business.industry ,Immunogenicity ,Cancer ,medicine.disease ,3. Good health ,lcsh:Genetics ,030104 developmental biology ,Epitope mapping ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Molecular Medicine ,business ,CD8 - Abstract
Immunotherapies are at the forefront of the fight against cancers, and researchers continue to develop and test novel immunotherapeutic modalities. Ideal cancer immunotherapies induce a patient’s immune system to kill their own cancer and develop long-lasting immunity. Research has demonstrated a critical requirement for CD8+ and CD4+ T cells in achieving durable responses. In the path to the clinic, researchers require robust tools to effectively evaluate the capacity for immunotherapies to generate adaptive anti-tumor responses. To study functional tumor-specific T cells, researchers have relied on targeting tumor-associated antigens (TAAs) or the inclusion of surrogate transgenes in pre-clinical models, which facilitate detection of T cells by using the targeted antigen(s) in peptide re-stimulation or tetramer-staining assays. Unfortunately, many pre-clinical models lack a defined TAA, and epitope mapping of TAAs is costly. Surrogate transgenes can alter tumor engraftment and influence the immunogenicity of tumors, making them less relevant to clinical tumors. Further, some researchers prefer to develop therapies that do not rely on pre-defined TAAs. Here, we describe a method to exploit major histocompatibility complex expression on murine cancer cell lines in a co-culture assay to detect T cells responding to bulk, undefined, tumor antigens. This is a tool to support the preclinical evaluation of novel, antigen-agnostic immunotherapies.
- Published
- 2019
11. Combining Vascular Normalization with an Oncolytic Virus Enhances Immunotherapy in a Preclinical Model of Advanced-Stage Ovarian Cancer
- Author
-
Byram W. Bridle, Jacob P. van Vloten, Jim Petrik, Amanda W. K. AuYeung, Pierre Major, Jack Lawler, Kathy Matuszewska, Sarah K. Wootton, and Lisa A. Santry
- Subjects
0301 basic medicine ,Oncolytic Newcastle Disease Virus ,Cancer Research ,Combination therapy ,Tumor hypoxia ,business.industry ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Primary tumor ,Virus ,Oncolytic virus ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Ovarian cancer ,business - Abstract
Purpose: Intravenous delivery of oncolytic viruses often leads to tumor vascular shutdown, resulting in decreased tumor perfusion and elevated tumor hypoxia. We hypothesized that using 3TSR to normalize tumor vasculature prior to administration of an oncolytic Newcastle disease virus (NDV) would enhance virus delivery and trafficking of immunologic cell subsets to the tumor core, resulting in systemically enhanced immunotherapy and regression of advanced-stage epithelial ovarian cancer (EOC). Experimental Design: Using an orthotopic, syngeneic mouse model of advanced-stage EOC, we pretreated mice with 3TSR (4 mg/kg per day) alone or followed by combination with fusogenic NDV(F3aa) (1.0 × 108 plaque-forming units). Results: Treatment with 3TSR normalized tumor vasculature, enhanced blood perfusion of primary EOC tumors, and induced disease regression. Animals treated with combination therapy had the greatest reduction in primary tumor mass, ascites accumulation, and secondary lesions (50% of mice were completely devoid of peritoneal metastases). Combining 3TSR + NDV(F3aa) led to enhanced trafficking of immunologic cells into the primary tumor core. Conclusions: We have shown, for the first time, that NDV, like other oncolytic viruses, is a potent mediator of acute vascular shutdown and that preventing this through vascular normalization can promote regression in a preclinical model of advanced-stage ovarian cancer. This challenges the current focus on induction of intravascular thrombosis as a requisite for successful oncolytic virotherapy. See related commentary by Bykov and Zamarin, p. 1446
- Published
- 2019
12. Intranasal vaccination with a Newcastle disease virus-vectored vaccine protects hamsters from SARS-CoV-2 infection and disease
- Author
-
Jonathan Audet, Sarah K. Wootton, Byram W. Bridle, Lily Chan, Lisa A. Santry, Logan Banadyga, Emelissa J. Valcourt, Darwyn Kobasa, Leonardo Susta, Kathy L. Frost, Marnie Willman, Anders Leung, Jessica A. Minott, Robert C. Mould, Nikesh Tailor, Alixandra Albietz, Bryce M. Warner, Jason P. Knapp, Pierre Major, Yanlong Pei, Yeganeh Mehrani, Robert Vendramelli, Phuc H. Pham, Kevin Tierney, Alexander Leacy, Heidi Wood, Jacob G. E. Yates, Mable Chan, David Safronetz, Stephanie A. Booth, Shihua He, and Bryan D. Griffin
- Subjects
COVID-19 Vaccines ,Science ,viruses ,Newcastle disease virus ,lyophilization ,Inflammation ,spike protein ,Newcastle disease ,Virus ,Article ,Disease Outbreaks ,Immune respons ,avian orthoavulavirus-1 ,Virology ,Pandemic ,Medicine ,Humans ,viral vectored vaccine ,skin and connective tissue diseases ,Multidisciplinary ,Lung ,lung pathology ,biology ,business.industry ,mucosal immunization ,SARS-CoV-2 ,Vaccination ,COVID-19 ,respiratory system ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,intranasal vaccination ,respiratory tract diseases ,Titer ,medicine.anatomical_structure ,Nasal administration ,medicine.symptom ,business - Abstract
The pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of Coronavirus Disease 2019 (COVID-19). Worldwide efforts are being made to develop vaccines to mitigate this pandemic. We engineered two recombinant Newcastle disease virus (NDV) vectors expressing either the full-length SARS-CoV-2 spike protein (NDV-FLS) or a version with a 19 amino acid deletion at the carboxy terminus (NDV-Δ19S). Hamsters receiving two doses (prime-boost) of NDV-FLS developed a robust SARS-CoV-2-neutralizing antibody response, with elimination of infectious virus in the lungs and minimal lung pathology at five days post-challenge. Single-dose vaccination with NDV-FLS significantly reduced SARS-CoV-2 replication in the lungs, but only mildly decreased lung inflammation. NDV-Δ19S-treated hamsters had a moderate decrease in SARS-CoV-2 titers in lungs and presented with severe microscopic lesions, suggesting that truncation of the spike protein was a less effective strategy. In summary, NDV-vectored vaccines represent a viable option for protection against COVID-19., Graphical Abstract
- Published
- 2021
13. AAV-mediated delivery of actoxumab and bezlotoxumab results in serum and mucosal antibody concentrations that provide protection from C. difficile toxin challenge
- Author
-
Matthew M, Guilleman, Brenna A Y, Stevens, Laura P, Van Lieshout, Amira D, Rghei, Yanlong, Pei, Lisa A, Santry, Brad, Thompson, and Sarah K, Wootton
- Abstract
Clostridium difficile is the leading cause of antibiotic-associated nosocomial diarrhea in the developed world. When the host-associated colon microbiome is disrupted by the ingestion of antibiotics, C. difficile spores can germinate, resulting in infection. C. difficile secretes enterotoxin A (TcdA) and cytotoxin B (TcdB) that are responsible for disease pathology. Treatment options are limited as the bacterium demonstrates resistance to many antibiotics, and even with antibacterial therapies, recurrences of C. difficile are common. Actotoxumab and bezlotoxumab are human monoclonal antibodies that bind and neutralize TcdA and TcdB, respectively. In 2016, the US food and drug administration (FDA) approved bezlotoxumab for use in the prevention of C. difficile infection recurrence. To ensure the long-term expression of antibodies, gene therapy can be used. Here, adeno-associated virus (AAV)6.2FF, a novel triple mutant of AAV6, was engineered to express either actotoxumab or bezlotoxumab in mice and hamsters. Both antibodies expressed at greater than 90 μg/mL in the serum and were detected at mucosal surfaces in both models. Hundred percent of mice given AAV6.2FF-actoxumab survived a lethal dose of TcdA. This proof of concept study demonstrates that AAV-mediated expression of C. difficile toxin antibodies is a viable approach for the prevention of recurrent C. difficile infections.
- Published
- 2020
14. Interference chromatography: a novel approach to optimizing chromatographic selectivity and separation performance for virus purification
- Author
-
Xiaojiao Shang, Renaud Jacquemart, James G. Stout, Jake M. Domm, Melissa Vandersluis, Pierre M. Major, Thomas M. McAusland, Sarah K. Wootton, Lisa A. Santry, and Mochao Zhao
- Subjects
Oncolytic Newcastle Disease Virus ,lcsh:Biotechnology ,Newcastle Disease ,Newcastle disease virus ,Biology ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Interference (communication) ,Virus purification ,lcsh:TP248.13-248.65 ,Animals ,Clinical efficacy ,030304 developmental biology ,Oncolytic Virotherapy ,0303 health sciences ,Chromatography ,Mice, Inbred BALB C ,Downstream processing ,Single ion ,Methodology Article ,Virion ,Single-use bioprocessing ,Fibroblasts ,3. Good health ,Oncolytic virus ,030220 oncology & carcinogenesis ,Viruses ,Interference chromatography ,Solid phases ,Female ,Adsorption ,Anion exchange membrane ,Biotechnology - Abstract
Background Oncolytic viruses are playing an increasingly important role in cancer immunotherapy applications. Given the preclinical and clinical efficacy of these virus-based therapeutics, there is a need for fast, simple, and inexpensive downstream processing methodologies to purify biologically active viral agents that meet the increasingly higher safety standards stipulated by regulatory authorities like the Food and Drug Administration and the European Agency for the Evaluation of Medicinal Products. However, the production of virus materials for clinical dosing of oncolytic virotherapies is currently limited—in quantity, quality, and timeliness—by current purification technologies. Adsorption of virus particles to solid phases provides a convenient and practical choice for large-scale fractionation and recovery of viruses from cell and media contaminants. Indeed, chromatography has been deemed the most promising technology for large-scale purification of viruses for biomedical applications. The implementation of new chromatography media has improved process performance, but low yields and long processing times required to reach the desired purity are still limiting. Results Here we report the development of an interference chromatography-based process for purifying high titer, clinical grade oncolytic Newcastle disease virus using NatriFlo® HD-Q membrane technology. This novel approach to optimizing chromatographic performance utilizes differences in molecular bonding interactions to achieve high purity in a single ion exchange step. Conclusions When used in conjunction with membrane chromatography, this high yield method based on interference chromatography has the potential to deliver efficient, scalable processes to enable viable production of oncolytic virotherapies.
- Published
- 2020
15. AAV-Vectored Expression of the Vascular Normalizing Agents 3TSR and Fc3TSR, and the Anti-Angiogenic Bevacizumab Extends Survival in a Murine Model of End-Stage Epithelial Ovarian Carcinoma
- Author
-
Ashley A. Stegelmeier, Lisa A. Santry, Matthew M. Guilleman, Kathy Matuszewska, Jessica A. Minott, Jacob G. E. Yates, Brenna A. Y. Stevens, Sylvia P. Thomas, Sierra Vanderkamp, Kiersten Hanada, Yanlong Pei, Amira D. Rghei, Jacob P. van Vloten, Madison Pereira, Brad Thompson, Pierre P. Major, James J. Petrik, Byram W. Bridle, and Sarah K. Wootton
- Subjects
ovarian cancer ,AOaV-1 ,gene therapy ,oncolytic virotherapy ,adeno-associated virus (AAV) ,vascular normalization ,viruses ,Medicine (miscellaneous) ,General Biochemistry, Genetics and Molecular Biology - Abstract
Epithelial ovarian cancer is the deadliest gynecological malignancy. The lack of effective treatments highlights the need for novel therapeutic interventions. The aim of this study was to investigate whether sustained adeno-associated virus (AAV) vector-mediated expression of vascular normalizing agents 3TSR and Fc3TSR and the antiangiogenic monoclonal antibody, Bevacizumab, with or without oncolytic virus treatment would improve survival in an orthotopic syngeneic mouse model of epithelial ovarian carcinoma. AAV vectors were administered 40 days post-tumor implantation and combined with oncolytic avian orthoavulavirus-1 (AOaV-1) 20 days later, at the peak of AAV-transgene expression, to ascertain whether survival could be extended. Flow cytometry conducted on blood samples, taken at an acute time point post-AOaV-1 administration (36 h), revealed a significant increase in activated NK cells in the blood of all mice that received AOaV-1. T cell analysis revealed a significant increase in CD8+ tumor specific T cells in the blood of AAV-Bevacizumab+AOaV-1 treated mice compared to control mice 10 days post AOaV-1 administration. Immunohistochemical staining of primary tumors harvested from a subset of mice euthanized 90 days post tumor implantation, when mice typically have large primary tumors, secondary peritoneal lesions, and extensive ascites fluid production, revealed that AAV-3TSR, AAV-Fc3TSR+AOaV-1, or AAV-Bevacizumab+AOaV-1 treated mice had significantly more tumor-infiltrating CD8+ T cells than PBS controls. Despite AAV-mediated transgene expression waning faster in tumor-bearing mice than in non-tumor bearing mice, all three of the AAV therapies significantly extended survival compared to control mice; with AAV-Bevacizumab performing the best in this model. However, combining AAV therapies with a single dose of AOaV-1 did not lead to significant extensions in survival compared to AAV therapies on their own, suggesting that additional doses of AOaV-1 may be required to improve efficacy in this model. These results suggest that vectorizing anti-angiogenic and vascular normalizing agents is a viable therapeutic option that warrants further investigation, including optimizing combination therapies.
- Published
- 2022
16. Use of Precision-Cut Lung Slices as an Ex Vivo Tool for Evaluating Viruses and Viral Vectors for Gene and Oncolytic Therapy
- Author
-
Robert C. Mould, Byram W. Bridle, Jondavid de Jong, John C. Bell, Sarah K. Wootton, Lisa A. Santry, Jacob P. van Vloten, María Carla Rosales Gerpe, and Adrian Pelin
- Subjects
0301 basic medicine ,Lung ,lcsh:QH426-470 ,lcsh:Cytology ,Vectors in gene therapy ,Biology ,3. Good health ,Viral vector ,Oncolytic virus ,Cell biology ,03 medical and health sciences ,lcsh:Genetics ,030104 developmental biology ,medicine.anatomical_structure ,Cancer cell ,Genetics ,medicine ,Tissue tropism ,Molecular Medicine ,lcsh:QH573-671 ,Molecular Biology ,Gene ,Ex vivo - Abstract
Organotypic slice cultures recapitulate many features of an intact organ, including cellular architecture, microenvironment, and polarity, making them an ideal tool for the ex vivo study of viruses and viral vectors. Here, we describe a procedure for generating precision-cut ovine and murine tissue slices from agarose-perfused normal and murine melanoma tumor-bearing lungs. Furthermore, we demonstrate that these precision-cut lung slices can be maintained up to 1 month and can be used for a range of applications, which include characterizing the tissue tropism of viruses that cannot be propagated in cell monolayers, evaluating the transducing properties of gene therapy vectors, and, finally, investigating the tumor specificity of oncolytic viruses. Our results suggest that ex vivo lung slices are an ideal platform for studying the tissue specificity and cancer cell selectivity of gene therapy vectors and oncolytic viruses prior to in vivo studies, providing justification for pre-clinical work.
- Published
- 2018
17. Production and Purification of High-Titer Newcastle Disease Virus for Use in Preclinical Mouse Models of Cancer
- Author
-
Jim Petrik, Leonardo Susta, Thomas M. McAusland, Pierre Major, Sarah K. Wootton, Byram W. Bridle, Lisa A. Santry, and Geoffrey A. Wood
- Subjects
0301 basic medicine ,animal structures ,Paramyxoviridae ,lcsh:QH426-470 ,viruses ,Newcastle disease virus ,Newcastle disease ,Virus ,Article ,03 medical and health sciences ,Genetics ,lcsh:QH573-671 ,Molecular Biology ,Pathogen ,oncolytic virus ,biology ,tangential flow filtration ,lcsh:Cytology ,allantoic fluid ,Embryonated ,RNA virus ,biology.organism_classification ,Virology ,3. Good health ,Oncolytic virus ,Titer ,lcsh:Genetics ,030104 developmental biology ,intravenous ,embryonic structures ,Molecular Medicine ,preclinical grade - Abstract
Newcastle disease virus (NDV) is a single-stranded, negative-sense RNA virus in the Paramyxoviridae family. Although primarily an avian pathogen, NDV is a potent oncolytic virus that has been shown to be safe and effective in a variety of preclinical cancer models and human clinical trials. To produce virus for oncolytic trials, NDV is commonly amplified in embryonated chicken eggs and purified from the allantoic fluid. Conventional methods for purifying virus from allantoic fluid often result in relatively low-titer preparations containing high levels of impurities, including immunogenic chicken host cell proteins from allantoic fluid. However, large quantities of virus need to be delivered intravenously to administer oncolytic NDV systemically to mice. This route of administration requires virus preparations that are both highly concentrated (to enable delivery of small volumes) and highly pure (to limit toxic effects from contaminants). Given the accumulation of promising preclinical and clinical data demonstrating the efficacy of NDV as an oncolytic agent, strategies for increasing the titer and purity of NDV preparations are sorely needed to allow for effective intravenous administration in mice. Here, we describe an optimized protocol for the rescue, production, and purification of high-titer in vivo-grade NDV for preclinical studies in mouse models. Keywords: Newcastle disease virus, oncolytic virus, tangential flow filtration, preclinical grade, intravenous, allantoic fluid
- Published
- 2018
18. Truncation of the enzootic nasal tumor virus envelope protein cytoplasmic tail increases Env-mediated fusion and infectivity
- Author
-
Sarah Wootton, Jacob P. van Vloten, Jondavid de Jong, María Carla Rosales Gerpe, Lisa A. Santry, and Scott R. Walsh
- Subjects
0301 basic medicine ,viruses ,Heterologous ,Biology ,Betaretrovirus ,Cell Line ,03 medical and health sciences ,Transduction (genetics) ,Viral Envelope Proteins ,Transduction, Genetic ,Viral entry ,Virology ,Animals ,Humans ,Tropism ,Sequence Deletion ,chemistry.chemical_classification ,Cell fusion ,Virus Internalization ,biology.organism_classification ,Jaagsiekte sheep retrovirus ,030104 developmental biology ,chemistry ,Glycoprotein - Abstract
Enzootic nasal tumor virus (ENTV) and Jaagsiekte sheep retrovirus (JSRV) are highly related ovine betaretroviruses that induce nasal and lung tumours in small ruminants, respectively. While the ENTV and JSRV envelope (Env) glycoproteins mediate virus entry using the same cellular receptor, the glycosylphosphatidylinositol-linked protein hyaluronoglucosaminidase, ENTV Env pseudovirions mediate entry into cells from a much more restricted range of species than do JSRV Env pseudovirions. Unlike JSRV Env, ENTV Env does not induce cell fusion at pH 5.0 or above, but rather requires a much lower pH (4.0-4.5) for fusion to occur. The cytoplasmic tail of retroviral envelope proteins is a key modulator of envelope-mediated fusion and pseudotype efficiency, especially in the context of virions composed of heterologous Gag proteins. Here we report that progressive truncation of the ENTV Env cytoplasmic tail improves transduction efficiency of pseudotyped retroviral vectors and that complete truncation of the ENTV Env cytoplasmic tail increases transduction efficiency to wild-type JSRV Env levels by increasing fusogenicity without affecting sensitivity to inhibition by lysosomotropic agents, subcellular localization or efficiency of inclusion into virions. Truncation of the cytoplasmic domain of ENTV Env resulted in a significant advantage in viral entry into all cell types tested, including foetal ovine lung and nasal cells. Taken together, we demonstrate that the cytoplasmic tail modulates the fusion activity of the ENTV Env protein and that truncation of this region enhances Eenv-mediated entry into target cells.
- Published
- 2017
19. Akt isoform specific effects in ovarian cancer progression
- Author
-
Manfred Jücker, Roger A. Moorehead, Jim Petrik, Nicolle M. Linnerth-Petrik, Sarah K. Wootton, and Lisa A. Santry
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cell Survival ,AKT1 ,Antineoplastic Agents ,Apoptosis ,AKT2 ,Akt inhibitors ,AKT3 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Akt isoforms ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,Protein Isoforms ,Neoplasm Metastasis ,RNA, Small Interfering ,Protein Kinase Inhibitors ,Protein kinase B ,Cell Proliferation ,Mice, Knockout ,Ovarian Neoplasms ,Tumor microenvironment ,business.industry ,Prognosis ,medicine.disease ,Primary tumor ,Tumor Burden ,tumor development ,Disease Models, Animal ,ovarian cancer ,030104 developmental biology ,Endocrinology ,Oncology ,Tumor progression ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Female ,Ovarian cancer ,business ,Proto-Oncogene Proteins c-akt ,Research Paper - Abstract
// Nicolle M. Linnerth-Petrik 1 , Lisa A. Santry 1 , Roger Moorehead 2 , Manfred Jucker 3 , Sarah K. Wootton 1, * , Jim Petrik 2, * 1 Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada 2 Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada 3 Center of Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany * These authors have contributed equally to this work Correspondence to: Jim Petrik, email: jpetrik@uoguelph.ca Keywords: ovarian cancer, Akt isoforms, Akt inhibitors, tumor development Received: March 21, 2016 Accepted: July 27, 2016 Published: August 11, 2016 ABSTRACT Ovarian cancer remains a significant therapeutic problem and novel, effective therapies are needed. Akt is a serine-threonine kinase that is overexpressed in numerous cancers, including ovarian. Mammalian cells express three Akt isoforms which are encoded by distinct genes. Although there are several Akt inhibitors in clinical trials, most indiscriminately target all isoforms. Current in vitro data and animal knockout experiments suggest that the Akt isoforms may have divergent roles. In this paper, we determined the isoform-specific functions of Akt in ovarian cancer cell proliferation in vitro and in ovarian cancer progression in vivo. For in vitro experiments, murine and human ovarian cancer cells were treated with Akt inhibitors and cell viability was assessed. We used two different in vivo approaches to identify the roles of Akt isoforms in ovarian cancer progression and their influence on the primary tumor and tumor microenvironment. In one experiment, wild-type C57Bl6 mice were orthotopically injected with ID8 cells with stable knockdown of Akt isoforms. In a separate experiment, mice null for Akt 1-3 were orthotopically injected with WT ID8 cells ( Figure 1 ). Our data show that inhibition of Akt1 significantly reduced ovarian cancer cell proliferation and inhibited tumor progression in vivo . Conversely, disruption of Akt2 increased tumor growth. Inhibition of Akt3 had an intermediate phenotype, but also increased growth of ovarian cancer cells. These data suggest that there is minimal redundancy between the Akt isoforms in ovarian cancer progression. These findings have important implications in the design of Akt inhibitors for the effective treatment of ovarian cancer.
- Published
- 2016
20. AAV Vectored Immunoprophylaxis for Filovirus Infections
- Author
-
Leonardo Susta, Amira D Rghei, Matthew M. Guilleman, Laura P. van Lieshout, Sarah K. Wootton, Lisa A. Santry, Byram W. Bridle, Khalil Karimi, and Sylvia P. Thomas
- Subjects
0301 basic medicine ,Zaire ebolavirus ,vector biology ,filoviruses ,lcsh:Medicine ,Review ,medicine.disease_cause ,Virus ,West africa ,Viral hemorrhagic fever ,adeno-associated virus vector ,03 medical and health sciences ,Human health ,0302 clinical medicine ,Immunity ,Medicine ,viral hemorrhagic fever ,Vector (molecular biology) ,vectored-immunoprophylaxis (VIP) ,General Immunology and Microbiology ,business.industry ,lcsh:R ,Public Health, Environmental and Occupational Health ,Outbreak ,medicine.disease ,Virology ,030104 developmental biology ,Infectious Diseases ,030220 oncology & carcinogenesis ,monoclonal antibodies ,zoonotic diseases ,business - Abstract
Filoviruses are among the deadliest infectious agents known to man, causing severe hemorrhagic fever, with up to 90% fatality rates. The 2014 Ebola outbreak in West Africa resulted in over 28,000 infections, demonstrating the large-scale human health and economic impact generated by filoviruses. Zaire ebolavirus is responsible for the greatest number of deaths to date and consequently there is now an approved vaccine, Ervebo, while other filovirus species have similar epidemic potential and remain without effective vaccines. Recent clinical success of REGN-EB3 and mAb-114 monoclonal antibody (mAb)-based therapies supports further investigation of this treatment approach for other filoviruses. While efficacious, protection from passive mAb therapies is short-lived, requiring repeat dosing to maintain therapeutic concentrations. An alternative strategy is vectored immunoprophylaxis (VIP), which utilizes an adeno-associated virus (AAV) vector to generate sustained expression of selected mAbs directly in vivo. This approach takes advantage of validated mAb development and enables vectorization of the top candidates to provide long-term immunity. In this review, we summarize the history of filovirus outbreaks, mAb-based therapeutics, and highlight promising AAV vectorized approaches to providing immunity against filoviruses where vaccines are not yet available.
- Published
- 2020
21. Use of Precision-Cut Lung Slices as an
- Author
-
María C, Rosales Gerpe, Jacob P, van Vloten, Lisa A, Santry, Jondavid, de Jong, Robert C, Mould, Adrian, Pelin, John C, Bell, Byram W, Bridle, and Sarah K, Wootton
- Subjects
Article - Abstract
Organotypic slice cultures recapitulate many features of an intact organ, including cellular architecture, microenvironment, and polarity, making them an ideal tool for the ex vivo study of viruses and viral vectors. Here, we describe a procedure for generating precision-cut ovine and murine tissue slices from agarose-perfused normal and murine melanoma tumor-bearing lungs. Furthermore, we demonstrate that these precision-cut lung slices can be maintained up to 1 month and can be used for a range of applications, which include characterizing the tissue tropism of viruses that cannot be propagated in cell monolayers, evaluating the transducing properties of gene therapy vectors, and, finally, investigating the tumor specificity of oncolytic viruses. Our results suggest that ex vivo lung slices are an ideal platform for studying the tissue specificity and cancer cell selectivity of gene therapy vectors and oncolytic viruses prior to in vivo studies, providing justification for pre-clinical work., Graphical Abstract
- Published
- 2018
22. AAV vector distribution in the mouse respiratory tract following four different methods of administration
- Author
-
Joelle C. Ingrao, Sarah K. Wootton, Lisa A. Santry, Geoffrey A. Wood, Laura P. van Lieshout, Darrick L. Yu, and Jondavid de Jong
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,lcsh:Biotechnology ,Genetic Vectors ,Respiratory System ,Gene Dosage ,Respiratory tract ,Biology ,Vectors in gene therapy ,medicine.disease_cause ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Adeno-associated virus ,Vector instillation ,lcsh:TP248.13-248.65 ,medicine ,Intubation, Intratracheal ,Intubation ,Animals ,Transgene expression ,Vector (molecular biology) ,Lung ,Administration, Intranasal ,Methodology Article ,Dependovirus ,3. Good health ,Mice, Inbred C57BL ,Intratracheal ,030104 developmental biology ,medicine.anatomical_structure ,Intranasal ,030220 oncology & carcinogenesis ,Immunology ,Respiratory epithelium ,Nasal administration ,Biotechnology - Abstract
Background Targeted delivery of gene therapy vectors to the mouse respiratory tract is often performed via intranasal or intratracheal administration; however, there can be a great deal of variability between these methods, which could potentially influence experimental results. Improving the accuracy and precision of lung delivery will not only reduce the number of animals required to detect statistically significant differences, but may reduce the variability of studies from different laboratories. Results Here we evaluated three different methods of adeno-associated virus (AAV) vector administration to the respiratory tract in mice (intranasal, intubation, and intratracheal injection) and discuss the advantages, challenges, and shortcomings of each. We also present a modified-intranasal delivery technique that is superior to passive administration of vector into the nares of anesthetized supine animals. Transgene expression was consistently visible in the nasal cavity, trachea, and proximal to middle aspect of all lung lobes for all four methods, whereas transgene expression was consistently observed in the most distal aspect of lung lobes only with the intubation and intratracheal injection techniques. AAV vector genome copy numbers in the lung were approximately four-fold lower in mice that received vector via intranasal administration in comparison to the other three methods of vector delivery. The modified intranasal, intubation and intratracheal injection methods of vector administration did not yield statistical differences in AAV vector genome copy numbers in the lung. With regard to reproducibility of vector distribution within and between animals, the modified-intranasal technique was superior. Conclusion Our results show that mode of AAV vector administration to the murine respiratory tract should be selected based on desired target site and skill of the researcher, and that appropriate technique selection may greatly influence experimental outcomes. Electronic supplementary material The online version of this article (doi:10.1186/s12896-017-0365-2) contains supplementary material, which is available to authorized users.
- Published
- 2017
23. Genetic characterization of small ruminant lentiviruses circulating in naturally infected sheep and goats in Ontario, Canada
- Author
-
Sarah K. Wootton, Lisa A. Santry, Paula I. Menzies, Alexander C. Gold, Jondavid de Jong, and Scott R. Walsh
- Subjects
Cancer Research ,Sequence analysis ,viruses ,Molecular Sequence Data ,Gene Products, gag ,Sheep Diseases ,Biology ,Virus ,Phylogenetics ,Virology ,Genetic variation ,Animals ,Cluster Analysis ,Caprine arthritis encephalitis virus ,Phylogeny ,Ontario ,Genetic diversity ,Goat Diseases ,Sheep ,Phylogenetic tree ,Immunodominant Epitopes ,Goats ,Lentivirus ,Genetic Variation ,Sequence Analysis, DNA ,Group-specific antigen ,biology.organism_classification ,Infectious Diseases ,Lentivirus Infections - Abstract
Maedi-visna virus (MVV) and caprine arthritis encephalitis virus (CAEV) are related members of a group of small ruminant lentiviruses (SRLVs) that infect sheep and goats. SRLVs are endemic in many countries, including Canada. However, very little is known about the genetic characteristics of Canadian SRLVs, particularly in the province of Ontario. Given the importance of surveillance and eradication programs for the control of SRLVs, it is imperative that the diagnostic tests used to identify infected animals are sensitive to local strains of SRLVs. The aim of this work was to characterize SRLV strains circulating in Ontario and to evaluate the variability of the immunodominant regions of the Gag protein. In this study, the nearly complete gag sequence of 164 SRLVs, from 130 naturally infected sheep and 32 naturally infected goats from Ontario, was sequenced. Animals belonged to distantly located single and mixed species (sheep and goats) farms. Ovine lentiviruses from the same farm tended to cluster more closely together than did caprine lentiviruses from the same farm. Sequence analysis revealed a higher degree of heterogeneity among the caprine lentivirus sequences with an average inter-farm pairwise DNA distance of 10% and only 5% in the ovine lentivirus group. Interestingly, amplification of SRLVs from ELISA positive sheep was successful in 81% of cases, whereas amplification of SRLV proviral DNA was only possible in 55% of the ELISA positive goat samples; suggesting that a significant portion of caprine lentiviruses circulating in Ontario possess heterogeneity at the primer binding sites used in this study. Sequences of sheep and goat SRLVs from Ontario were assembled into phylogenetic trees with other known SRLVs and were found to belong to sequence groups A2 and B1, respectively, as defined by Shah et al. (2004a). A novel caprine lentivirus with a pairwise genetic difference of 15.6-25.4% relative to other group B subtypes was identified. Thus we suggest the designation of a novel subtype, B4, within the caprine lentivirus-like cluster. Lastly, we demonstrate evidence of recombination between ovine lentiviruses. These results emphasize the broad genetic diversity of SRLV strains circulating in the province of Ontario and show that the gag region is suitable for phylogenetic studies and may be applied to monitor SRLV eradication programs.
- Published
- 2013
24. 422. Harnessing Immunogenic Cell Death to Potentiate the Oncolytic Effect of ORFV Infection in Murine Cancer Cells
- Author
-
Byram W. Bridle, Sarah K. Wootton, Lisa A. Santry, Jondavid de Jong, and Jacob P. vanVloten
- Subjects
0301 basic medicine ,Pharmacology ,03 medical and health sciences ,030104 developmental biology ,Drug Discovery ,Cancer cell ,Genetics ,Molecular Medicine ,Immunogenic cell death ,Biology ,Molecular Biology ,Virology ,Oncolytic virus - Published
- 2016
25. 66. Recombinant Newcastle Disease Virus as an Oncolytic Therapy for Ovarian and Prostate Cancers
- Author
-
Jacob P. vanVloten, Jim Petrik, Byram W. Bridle, Kathy Matuszewska, Sarah K. Wootton, and Lisa A. Santry
- Subjects
Pharmacology ,Melanoma ,Biology ,medicine.disease ,biology.organism_classification ,Virology ,Newcastle disease ,Virus ,Oncolytic virus ,Prostate cancer ,Immune system ,Drug Discovery ,Cancer cell ,Genetics ,medicine ,Molecular Medicine ,Ovarian cancer ,Molecular Biology - Abstract
Newcastle disease virus (NDV) is a member of the Avulavirus genus in the Paramyxoviridae family. NDV selectively replicates in tumor cells due to defects in antiviral and apoptotic signalling. It is the oncolytic virus with the longest history of use in clinical trials with a proven safety record as a monotherapy due to its strong induction of antiviral responses in non-transformed mammalian cells. In addition to its direct oncolytic effect, NDV also activates both innate and adaptive immune responses and therefore has strong immunotherapeutic potential. To enhance the fusogenicity of the virus, the fusion protein of recombinant LaSota NDV (rNDV) was engineered to express a multibasic cleavage and activation site (rNDV/F3aa). This virus is highly fusogenic leading to the formation of large syncytia that allows for efficient intratumoral spread and increased lytic potential. While NDV has demonstrated efficacy in a number of different cancer models, including melanoma and hepatocellular carcinoma, little information about its efficacy in ovarian and prostate cancer is available. To this end, we evaluated the oncolytic potential of rNDV/F3aa expressing enhanced green fluorescent protein (GFP) in a panel of prostate and ovarian cancer cell lines using a resazurin dye-based cell viability assay. Infection with rNDV/F3aa-GFP at a range of MOIs significantly reduced the viability of these cancer cells in vitro, but had little effect on normal untransformed cells. To evaluate the efficacy of NDV in vivo, we used a well-established orthotopic mouse model of epithelial ovarian cancer and a syngeneic murine RM9 prostate carcinoma model. Intravenous delivery of 1×10^8 PFU of rNDV/F3aa-GFP to ovarian tumor-bearing mice resulted in a significant reduction in the primary tumor size and number of metastatic abdominal tumours. Furthermore, 50% of the mice had a complete loss of ascites, which is an accumulation of fluid in the abdomen, as a result of advanced stage ovarian cancer. The apparent decrease in secondary disease after treatment with rNDV/F3aa-GFP is particularly promising as it is the metastatic lesions that ultimately cause high morbidity and mortality in patients with advanced ovarian cancers. A pilot study in the RM9 prostate model revealed that rNDV/F3aa-GFP was able to replicate and spread within the tumor and neovasculature after intratumoral or intravenous delivery of 1×10^8 PFU. Studies to confirm these results, as well as to evaluate the ability of rNDV/F3aa-GFP to induce tumor regression and increase survival are currently underway. Taken together, these data suggest that NDV/F3aa is a promising oncolytic for further development as an anti-cancer agent for the treatment of prostate and ovarian cancers.
- Published
- 2016
26. Adeno-associated virus vector mediated expression of an oncogenic retroviral envelope protein induces lung adenocarcinomas in immunocompetent mice
- Author
-
Darrick L. Yu, Sarah K. Wootton, Nicolle M. Linnerth-Petrik, and Lisa A. Santry
- Subjects
Lung Neoplasms ,lcsh:Medicine ,Signal transduction ,ERK signaling cascade ,medicine.disease_cause ,Mice ,0302 clinical medicine ,Molecular cell biology ,Signal Initiation ,Viral Envelope Proteins ,Akt signaling cascade ,lcsh:Science ,Adeno-associated virus ,0303 health sciences ,Multidisciplinary ,biology ,Mechanisms of Signal Transduction ,Signaling cascades ,Animal Models ,Dependovirus ,Jaagsiekte sheep retrovirus ,3. Good health ,Veterinary Diseases ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Viral Vectors ,Immunocompetence ,Research Article ,Genetically modified mouse ,MAPK signaling cascades ,MAP Kinase Signaling System ,Blotting, Western ,Genetic Vectors ,Microbiology ,Cell Growth ,Cell Line ,03 medical and health sciences ,Model Organisms ,Virology ,medicine ,PTEN ,Animals ,Humans ,Lung cancer ,Biology ,030304 developmental biology ,DNA Primers ,Base Sequence ,lcsh:R ,Veterinary Virology ,medicine.disease ,biology.organism_classification ,Mice, Inbred C57BL ,Animal Models of Infection ,Microscopy, Fluorescence ,Viruses and Cancer ,Immunology ,biology.protein ,Veterinary Science ,lcsh:Q ,Betaretrovirus ,Carcinogenesis ,Viral Transmission and Infection - Abstract
Lung cancer is the most common cause of cancer-related death worldwide. A poor overall survival rate of 16% necessitates the need for novel treatment strategies. Mouse models of lung cancer are important tools for analyzing the significance of somatic mutations in the initiation and progression of lung cancer. Of additional importance, however, are animal models of virally induced cancers. JSRV is a simple betaretrovirus that causes contagious lung cancer in sheep known as ovine pulmonary adenocarcinoma and closely resembles human lung adenocarcinoma. Previously we showed that expression of the JSRV envelope (Env) from an AAV vector induced lung tumors in immunodeficient mice, but not in immunocompetent mice. Because of the importance of studying lung cancer in the context of an intact immune system we sought to improve our mouse model. In this report, we employed the use of a strong JSRV enhancer-promoter combination to express Env at high levels and demonstrate for the first time, lung tumor induction in immunocompetent mice. This occurred despite a robust Env-specific antibody-mediated immune response. The PI3K/Akt and MAPK pathways were activated in both immunocompetent and immunodeficient mice, however, differential activation of PTEN, GSKα, p70S6K, p38MAPK, ATF2 and STAT5 was observed. A JSRV Env lung tumor-derived cell line was shown to have a similar signal transduction activation profile as Env-induced lung tumors in C57BL/6 mice. Given the similarities between our model and pulmonary adenocarcinomas in humans, and the ease with which tumors can be induced in any transgenic mouse, this system can be used to uncover novel mechanisms involved lung tumorigenesis.
- Published
- 2012
27. Opposing Functions of Akt Isoforms in Lung Tumor Initiation and Progression
- Author
-
Jim Petrik, Sarah K. Wootton, Lisa A. Santry, and Nicolle M. Linnerth-Petrik
- Subjects
Cell signaling ,Lung Neoplasms ,Carcinogenesis ,lcsh:Medicine ,Apoptosis ,Signal transduction ,medicine.disease_cause ,Bioinformatics ,Mice ,chemistry.chemical_compound ,Signal Initiation ,Molecular Cell Biology ,Protein Isoforms ,AKT signaling cascade ,Phosphorylation ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Mechanisms of Signal Transduction ,Signaling cascades ,Animal Models ,Phenotype ,Cell biology ,Knockout mouse ,Disease Progression ,Research Article ,Proto-Oncogene Proteins c-akt ,Mouse Models ,Phosphoinositide Signal Transduction ,Biology ,Research and Analysis Methods ,Microbiology ,Model Organisms ,Virology ,medicine ,Animals ,Phosphatidylinositol ,Protein kinase A ,Protein kinase B ,Cell Proliferation ,Biology and life sciences ,lcsh:R ,Disease Models, Animal ,chemistry ,Viruses and Cancer ,lcsh:Q - Abstract
BACKGROUND: The phosphatidylinositol 3-kinase-regulated protein kinase, Akt, plays an important role in the initiation and progression of human cancer. Mammalian cells express three Akt isoforms (Akt1-3), which are encoded by distinct genes. Despite sharing a high degree of amino acid identity, phenotypes observed in knockout mice suggest that Akt isoforms are not functionally redundant. The relative contributions of the different Akt isoforms to oncogenesis, and the effect of their deficiencies on tumor development, are not well understood. METHODS: Here we demonstrate that Akt isoforms have non-overlapping and sometimes opposing functions in tumor initiation and progression using a viral oncogene-induced mouse model of lung cancer and Akt isoform-specific knockout mice. RESULTS: Akt1 ablation significantly delays initiation of lung tumor growth, whereas Akt2 deficiency dramatically accelerates tumorigenesis in this mouse model. Ablation of Akt3 had a small, not statistically significant, stimulatory effect on tumor induction and growth by the viral oncogene. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and Ki67 immunostaining of lung tissue sections revealed that the delayed tumor induction in Akt1-/- mice was due to the inhibitory effects of Akt1 ablation on cell growth and survival. Conversely, the accelerated growth rate of lung tumors in Akt2-/- and Akt3-/- mice was due to increased cell proliferation and reduced tumor cell apoptosis. Investigation of Akt signaling in tumors from Akt knockout mice revealed that the lack of Akt1 interrupted the propagation of signaling in tumors to the critical downstream targets, GSK-3α/β and mTOR. CONCLUSIONS: These results demonstrate that the degree of functional redundancy between Akt isoforms in the context of lung tumor initiation is minimal. Given that this mouse model exhibits considerable similarities to human lung cancer, these findings have important implications for the design and use of Akt inhibitors for the treatment of lung cancer.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.