Molecular Therapy - Oncolytics

Slides:



Advertisements
Similar presentations
Manish R. Patel, DO, Blake A
Advertisements

Volume 14, Issue 3, Pages (September 2006)
Manish R. Patel, DO, Blake A
Molecular Therapy - Oncolytics
Molecular Therapy - Oncolytics
T Cell-Activating Mesenchymal Stem Cells as a Biotherapeutic for HCC
Volume 15, Issue 1, Pages (January 2007)
Jennifer R. Hamilton, Gayathri Vijayakumar, Peter Palese  Cell Reports 
Brian Hutzen, Chun-Yu Chen, Pin-Yi Wang, Les Sprague, Hayley M
Targeting an Oncolytic Influenza A Virus to Tumor Tissue by Elastase
Recombinant mumps virus as a cancer therapeutic agent
Molecular Therapy - Oncolytics
Molecular Therapy - Oncolytics
Novel Potent Chimeric Oncolytic Orthopoxvirus HOV-33 Kills Colorectal Cancer with Intratumoral and Intraperiteonal Delivery  Michael O'Leary, MD, Susanne.
Molecular Therapy - Oncolytics
Insertion of the Type-I IFN Decoy Receptor B18R in a miRNA-Tagged Semliki Forest Virus Improves Oncolytic Capacity but Results in Neurotoxicity  Tina.
Oncolytic Immunotherapy for Bladder Cancer Using Coxsackie A21 Virus
Volume 6, Issue 3, Pages (September 2002)
Volume 10, Issue 6, Pages (December 2004)
Volume 26, Issue 4, Pages (April 2018)
Michael I. Ebright, MD, Jonathan S
Genetically Engineered Multilineage-Differentiating Stress-Enduring Cells as Cellular Vehicles against Malignant Gliomas  Tomohiro Yamasaki, Shohei Wakao,
Volume 18, Issue 11, Pages (November 2010)
Volume 15, Issue 5, Pages (May 2007)
Volume 18, Issue 9, Pages (September 2010)
Suppression of Murine Colitis and its Associated Cancer by Carcinoembryonic Antigen- Specific Regulatory T Cells  Dan Blat, Ehud Zigmond, Zoya Alteber,
Volume 22, Issue 1, Pages (January 2014)
Incorporation of the B18R Gene of Vaccinia Virus Into an Oncolytic Herpes Simplex Virus Improves Antitumor Activity  Xinping Fu, Armando Rivera, Lihua.
Volume 9, Issue 6, Pages (June 2004)
Reovirus FAST Protein Enhances Vesicular Stomatitis Virus Oncolytic Virotherapy in Primary and Metastatic Tumor Models  Fabrice Le Boeuf, Simon Gebremeskel,
Efficacy and Safety of Doubly-Regulated Vaccinia Virus in a Mouse Xenograft Model of Multiple Myeloma  Muneyoshi Futami, Kota Sato, Kanji Miyazaki, Kenshi.
Volume 23, Issue 4, Pages (April 2015)
Volume 23, Issue 4, Pages (April 2015)
Hannah Chen, Padma Sampath, Weizhou Hou, Stephen H. Thorne 
Molecular Therapy - Methods & Clinical Development
Volume 15, Issue 3, Pages (March 2009)
Volume 22, Issue 6, Pages (June 2014)
Thomas S. Griffith, Elizabeth L. Broghammer  Molecular Therapy 
Volume 22, Issue 1, Pages (January 2014)
Volume 20, Issue 4, Pages (October 2016)
Lukxmi Balathasan, Vera A
EVA1A/TMEM166 Regulates Embryonic Neurogenesis by Autophagy
Volume 24, Issue 1, Pages (January 2016)
Molecular Therapy - Oncolytics
Sangeet Lal, Corey Raffel  Molecular Therapy - Oncolytics 
Molecular Therapy - Oncolytics
Chemovirotherapy of Pancreatic Adenocarcinoma by Combining Oncolytic Vaccinia Virus GLV-1h68 with nab-Paclitaxel Plus Gemcitabine  Eike Binz, Susanne.
Molecular Therapy - Oncolytics
Newly Characterized Murine Undifferentiated Sarcoma Models Sensitive to Virotherapy with Oncolytic HSV-1 M002  Eric K. Ring, Rong Li, Blake P. Moore,
Molecular Therapy - Nucleic Acids
Valerie Künzi, Patrick A
Volume 8, Issue 2, Pages (August 2003)
Molecular Therapy - Oncolytics
Volume 23, Issue 3, Pages (March 2015)
Volume 26, Issue 4, Pages (April 2018)
The Expression of MicroRNA-598 Inhibits Ovarian Cancer Cell Proliferation and Metastasis by Targeting URI  Feng Xing, Shuo Wang, Jianhong Zhou  Molecular.
Volume 20, Issue 4, Pages (April 2012)
Molecular Therapy - Oncolytics
Cowpox Virus: A New and Armed Oncolytic Poxvirus
Development of a Safe and Effective Vaccinia Virus Oncolytic Vector WR-Δ4 with a Set of Gene Deletions on Several Viral Pathways  Ernesto Mejías-Pérez,
The Enhanced Tumor Specificity of TG6002, an Armed Oncolytic Vaccinia Virus Deleted in Two Genes Involved in Nucleotide Metabolism  Johann Foloppe, Juliette.
Molecular Therapy - Methods & Clinical Development
Abscopal Effect in Non-injected Tumors Achieved with Cytokine-Armed Oncolytic Adenovirus  Riikka Havunen, João M. Santos, Suvi Sorsa, Tommi Rantapero,
Volume 20, Issue 6, Pages (June 2012)
Volume 20, Issue 4, Pages (April 2012)
Volume 3, Issue 5, Pages (May 2001)
Volume 18, Issue 2, Pages (February 2010)
Deletion of Apoptosis Inhibitor F1L in Vaccinia Virus Increases Safety and Oncolysis for Cancer Therapy  Adrian Pelin, Johann Foloppe, Julia Petryk, Ragunath.
Volume 18, Issue 10, Pages (October 2010)
Role of Cell-Penetrating Peptides in Intracellular Delivery of Peptide Nucleic Acids Targeting Hepadnaviral Replication  Bénédicte Ndeboko, Narayan Ramamurthy,
Presentation transcript:

Molecular Therapy - Oncolytics A Novel Oncolytic Chimeric Orthopoxvirus Encoding Luciferase Enables Real-Time View of Colorectal Cancer Cell Infection  Michael P. O’Leary, Susanne G. Warner, Sang-In Kim, Shyambabu Chaurasiya, Jianming Lu, Audrey H. Choi, Anthony K. Park, Yanghee Woo, Yuman Fong, Nanhai G. Chen  Molecular Therapy - Oncolytics  Volume 9, Pages 13-21 (June 2018) DOI: 10.1016/j.omto.2018.03.001 Copyright © 2018 The Author(s) Terms and Conditions

Figure 1 CF-33 Possesses Superior Replication versus Parental Strains and Is Robustly Cytotoxic against Colon Cancer Cells In Vitro in a Dose-Dependent Manner Parental virus strains and CF-33-infected HCT116 cells. (A) Secreted form of external enveloped virions (EEV) were measured from supernatant at 12 and 18 hr post-infection. (B) Lysates from infected HCT116 cells were measured at 24, 48, and 72 hr. Viral titers were measured via standard plaque assays. (C) CF-33 kills colon cancer cells HCT-116, SW620, and LoVo in a dose-dependent manner. Error bars indicate SD. Ordinary one-way ANOVA was used at each time point. *p < 0.05; **p < 0.01; ***p < 0.001. Fold change in PFU/cell is in comparison to titers of uninfected cells at 0 hr immediately prior to infection. Molecular Therapy - Oncolytics 2018 9, 13-21DOI: (10.1016/j.omto.2018.03.001) Copyright © 2018 The Author(s) Terms and Conditions

Figure 2 Virus-Encoded Luciferase Activity Is Dose Dependent In Vitro (A) HCT116 cells were infected with CF33-Fluc virus at indicated MOIs in a 6-well plate format. Cells were imaged 24 hr post-infection for bioluminescence using Lago-X imaging system. (B) Cells were infected as in (A), and cell lysates were collected 24 hr post-infection. Total luciferase activity was measured in relative lumominometer units (RLU) using luciferase assay kit. The differential luciferase expression, which seems stronger on the periphery, is attributed to confluence of plated cells, which was denser on inferior periphery of the wells. Moreover, the absence of central luciferase expression at lower MOIs is likely due to decreased levels of infectivity at the 24-hr time point, whereas absence of central luciferase expression at MOI of 3 is attributed to cell death. Error bars indicate SD. Molecular Therapy - Oncolytics 2018 9, 13-21DOI: (10.1016/j.omto.2018.03.001) Copyright © 2018 The Author(s) Terms and Conditions

Figure 3 Virally Induced Cell Death Occurs via Necroptotic Pathways (A) Virally infected fluorescent microscopy shows TUNEL assay controls demonstrating fluorescent green DNA damage indicative of apoptosis, whereas both uninfected cells and highly infected cells at a high-dose MOI of 5 demonstrate no evidence of DNA damage. (B) FACS analysis of annexin and phosphatidyl inostitol (PI) examining non-infected cells (left) and infected cells (right) 18 hr post-infection. This indicates that while many dead cells are present (indicated by PI), none are undergoing apoptosis as a result of viral infection (annexin). (C) Infected cells demonstrate significantly lower quantities of ATP than non-infected controls at 48 and 72 hr. Error bars indicate SD. Student’s t test was used. *** indicates statistical significance defined as p < 001 for 48-hr and 72-hr time points. (D) Calreticulin was analyzed via flow cytometry with and without viral infection, demonstrating increased caltreticulin from infected cells. (E) ATP secretion in supernatants was demonstrated to be at least 1.5-fold higher in infected cells (error bars indicate SD). (F) HMGB1 protein secretion in supernatants of infected cells was substantially more pronounced in infected versus non-infected cells. Molecular Therapy - Oncolytics 2018 9, 13-21DOI: (10.1016/j.omto.2018.03.001) Copyright © 2018 The Author(s) Terms and Conditions

Figure 4 CF33-Fluc Bioluminescence Corresponds with Tumor Viral Titers (A) Representative mouse received intratumoral (IT) injection of CF33-Fluc. Bioluminescence corresponds with viral titers and tumor regression. (B) Average intensity of bioluminescence in the region of interest (ROI) took longer to achieve in intravenous (IV) groups but had a sustained response that correlated with viral replication. (C) Similar IV versus IT tumor and organ titers 10 days post-infection. (D) At 50 days post-infection, prolonged tumor viral titer in IV group, but lower titer in IT group, corresponds with tumor regression and thereby decreased viral activity. Error bars indicate SD. Molecular Therapy - Oncolytics 2018 9, 13-21DOI: (10.1016/j.omto.2018.03.001) Copyright © 2018 The Author(s) Terms and Conditions

Figure 5 Virus Spread in the Tumor following Intratumor or Intravenous Injection and Examination of Vital Organs in Infected and Non-infected Mice (A) Whereas the IV group had a more infiltrative pattern of infection, the IT group had a more uniform leading edge of viral infectivity, likely indicating that efficacy was a direct function of oncolytic ability and effective viral delivery. (B) Representative slides of brain, kidney, liver, and spleen are shown demonstrating no differences in histopathological organ appearance based upon viral infection. Molecular Therapy - Oncolytics 2018 9, 13-21DOI: (10.1016/j.omto.2018.03.001) Copyright © 2018 The Author(s) Terms and Conditions

Figure 6 CF33-Fluc Shows Anti-tumor Activity in HCT116 Xenograft Model following Intratumoral and Intravenous Delivery Both IV (B) (eight mice/group with four sacrificed at day 10) and IT (A) (five mice/group with two sacrificed at day 10) delivery methods result in tumor growth abrogation compared to controls (four mice for IV PBS control, and two mice for IT control). Both IT and IV treatment groups had individual mice with complete pathologic tumor regression. *IT p = 0.037; *IV p = 0.034. Molecular Therapy - Oncolytics 2018 9, 13-21DOI: (10.1016/j.omto.2018.03.001) Copyright © 2018 The Author(s) Terms and Conditions