Jennifer R. Hamilton, Gayathri Vijayakumar, Peter Palese  Cell Reports 

Slides:



Advertisements
Similar presentations
Continuous Delivery of Neutralizing Antibodies Elevate CCL2 Levels in Mice Bearing MCF10CA1d Breast Tumor Xenografts  Min Yao, Curtis Smart, Qingting.
Advertisements

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
Nrf2 Induces IL-17D to Mediate Tumor and Virus Surveillance
Molecular Therapy - Oncolytics
(A) Weight loss curves following intranasal infection of different mouse groups with COBRA, seasonal, or pandemic influenza viruses. (A) Weight loss curves.
Volume 20, Issue 7, Pages (August 2017)
DAI/ZBP1/DLM-1 Complexes with RIP3 to Mediate Virus-Induced Programmed Necrosis that Is Targeted by Murine Cytomegalovirus vIRA  Jason W. Upton, William J.
Volume 3, Issue 4, Pages (April 2013)
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.
Volume 22, Issue 3, Pages e5 (September 2017)
Brian Yordy, Norifumi Iijima, Anita Huttner, David Leib, Akiko Iwasaki 
Volume 18, Issue 11, Pages (November 2011)
Volume 10, Issue 6, Pages (December 2004)
Volume 4, Issue 3, Pages (September 2008)
Volume 20, Issue 12, Pages (December 2012)
Michael I. Ebright, MD, Jonathan S
Volume 18, Issue 11, Pages (November 2010)
Volume 26, Issue 9, Pages (September 2018)
Molecular Therapy - Oncolytics
Hemagglutinin-targeting Artificial MicroRNAs Expressed by Adenovirus Protect Mice From Different Clades of H5N1 Infection  Xinying Tang, Hongbo Zhang,
Volume 22, Issue 1, Pages e7 (July 2017)
Volume 21, Issue 6, Pages (November 2017)
Volume 4, Issue 4, Pages (October 2003)
Volume 12, Issue 1, Pages (July 2015)
Molecular Therapy - Oncolytics
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,
Volume 2, Issue 4, Pages (October 2000)
Volume 23, Issue 4, Pages (April 2015)
Volume 23, Issue 4, Pages (April 2015)
Volume 13, Issue 6, Pages (November 2015)
Volume 6, Issue 1, Pages (July 2009)
CD40, but Not CD40L, Is Required for the Optimal Priming of T Cells and Control of Aerosol M. tuberculosis Infection  Vanja Lazarevic, Amy J Myers, Charles.
Melissa B. Uccellini, Adolfo García-Sastre  Cell Reports 
Volume 22, Issue 1, Pages (January 2014)
Volume 21, Issue 6, Pages (November 2017)
Lukxmi Balathasan, Vera A
IL-12 affects Dermatophagoides farinae–induced IL-4 production by T cells from pediatric patients with mite-sensitive asthma  Takeshi Noma, MD, PhD, Izumi.
Volume 13, Issue 7, Pages (November 2015)
Volume 24, Issue 1, Pages (January 2016)
Nrf2 Induces IL-17D to Mediate Tumor and Virus Surveillance
Sangeet Lal, Corey Raffel  Molecular Therapy - Oncolytics 
Volume 18, Issue 5, Pages (May 2010)
Volume 3, Issue 1, Pages (January 2013)
Volume 6, Issue 3, Pages (September 2002)
Volume 18, Issue 12, Pages (December 2010)
Volume 46, Issue 4, Pages (April 2017)
Volume 24, Issue 7, Pages e5 (August 2018)
Molecular Therapy - Nucleic Acids
Newly Characterized Murine Undifferentiated Sarcoma Models Sensitive to Virotherapy with Oncolytic HSV-1 M002  Eric K. Ring, Rong Li, Blake P. Moore,
Volume 21, Issue 6, Pages (November 2017)
Molecular Therapy - Oncolytics
Volume 11, Issue 10, Pages (June 2015)
Hepatocyte Growth Factor Regulates the miR-206-HDAC4 Cascade to Control Neurogenic Muscle Atrophy following Surgical Denervation in Mice  Wooshik Choi,
Therapeutic Efficacy of G207, a Conditionally Replicating Herpes Simplex Virus Type 1 Mutant, for Gallbladder Carcinoma in Immunocompetent Hamsters  Kenji.
Volume 26, Issue 4, Pages (April 2018)
Volume 6, Issue 4, Pages (February 2014)
Single-Shot, Multicycle Suicide Gene Therapy by Replication-Competent Retrovirus Vectors Achieves Long-Term Survival Benefit in Experimental Glioma  Chien-Kuo.
Volume 20, Issue 4, Pages (April 2012)
Molecular Therapy - Oncolytics
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,
Molecular Therapy - Oncolytics
Fig. 2 NDR2-deficient mice are more vulnerable to RNA viral infection.
Volume 11, Issue 6, Pages (June 2012)
Presentation transcript:

A Recombinant Antibody-Expressing Influenza Virus Delays Tumor Growth in a Mouse Model  Jennifer R. Hamilton, Gayathri Vijayakumar, Peter Palese  Cell Reports  Volume 22, Issue 1, Pages 1-7 (January 2018) DOI: 10.1016/j.celrep.2017.12.025 Copyright © 2017 The Author(s) Terms and Conditions

Cell Reports 2018 22, 1-7DOI: (10.1016/j.celrep.2017.12.025) Copyright © 2017 The Author(s) Terms and Conditions

Figure 1 In Vitro and In Vivo Growth Characteristics of IAV-9H10 (A) Diagram of IAV engineered to express antibodies during infection. (B) Peak titer following viral growth in eggs at 33°C for 3 days. IAV-9H10 (harboring both PB1-9H10heavy chain and PA-9H10light chain segments) viruses containing single transgenic segments and WT IAV are shown. HC, heavy chain; LC, light chain. (C) Viral growth in eggs, 5 eggs per time point per virus. ∗Samples below the limit of detection (LOD). (D) Viral lung titers 2 days post intranasal infection with 5 × 105 PFUs of IAV-9H10 or WT IAV (n = 3 mice per group). (E and F) Following infection, (E) body weight loss and (F) survival were quantified (n = 4 mice per group). Experiments were performed in duplicate and all error bars represent SEM. Diagram is not to scale. Cell Reports 2018 22, 1-7DOI: (10.1016/j.celrep.2017.12.025) Copyright © 2017 The Author(s) Terms and Conditions

Figure 2 Antibody Is Produced during IAV-9H10 Infection In Ovo (A) Detection of transgenic segments in IAV-9H10. Viral RNA was isolated and RT-PCR was performed to amplify the terminal 300 bp of WT PB1 or PA. The expected PCR products for the IAV-9H10 PB1 and PA are 1,902 and 1,194 bp, respectively. (B) 9H10 quantity following inoculation with ∼10 PFUs, 3–4 eggs per time point, and assessed via ELISA. Eggs inoculated with WT IAV were assessed on day 3. ND, not detected. (C) Antibody expression is stable during passaging of IAV-9H10 in eggs. IAV-9H10 was diluted 10−6 and 100 μL was used to inoculate 3 eggs (passage 1 [P1]). Following viral growth at 33°C for 3 days, allantoic fluid was isolated, diluted 10−6, and injected back into 3 eggs. This was repeated for a total of 4 passages; 9H10 was quantified via ELISA. (B) and (C) were performed in duplicate and error bars represent SEM. No tx, no treatment. Cell Reports 2018 22, 1-7DOI: (10.1016/j.celrep.2017.12.025) Copyright © 2017 The Author(s) Terms and Conditions

Figure 3 Antibody Produced during IAV-9H10 Infection In Ovo Is Functional (A) ELISA assessing the binding of 9H10 to X-31 virus. Antibodies were diluted to 10 μg/mL, and 2-fold dilutions were performed in triplicate. OD, optical density. (B) Microneutralization assay comparing IAV-produced and hybridoma-produced 9H10 antibody. Both aliquots protect MDCK cells from infection with X-31. (C and D) Intranasal treatment with 10 μg 9H10 24 hr prior to infection protects mice from (C) morbidity and (D) mortality following X-31 challenge with 3× LD50 (n = 4 mice per group). Both 9H10hybridoma and 9H10IAV-9H10 protect mice from challenge. All experiments were performed in duplicate and error bars represent SEM. Cell Reports 2018 22, 1-7DOI: (10.1016/j.celrep.2017.12.025) Copyright © 2017 The Author(s) Terms and Conditions

Figure 4 IAV-CTLA4 Delays Growth of B16-F10 Melanoma Tumors In Vivo and Prolongs Overall Survival (A) B16-F10 cells undergo cell death in vitro following infection with WT IAV or IAV-9H10. Viability was assayed 48 hpi in quadruplicates and normalized to uninfected cells. (B) B16-F10 cells secrete antibody following IAV-9H10 infection at MOI = 5; 9H10 was quantified 48 hpi in triplicate via ELISA. ND, not detected. (C) B16-F10 cells do not support multicycle replication in the absence of TPCK-trypsin. Cells were infected at MOI = 0.01 with WT IAV, incubated ±TPCK-trypsin, and virus titrated in triplicate. (D) Experimental timeline for the assessment of IAV-CTLA4 as an oncolytic agent. B16-F10 cells were implanted in the right flank on day 0 and in the left flank on day 4. Right-flank tumors were intratumorally injected with WT IAV or IAV-CTLA4 (5.5 × 105 PFUs/injection) or PBS on days 5, 7, 9, and 11. From day 13 onward, tumor volumes were measured every 2 days. (E) Right-flank tumors treated with IAV-CTLA4 demonstrate delayed growth compared to PBS- and IAV WT-treated tumors. Mice with tumors exceeding 1,000 mm3 were sacrificed, and their last-measured tumor volumes were used for subsequent tumor measurements. Stars indicate when mice received right-flank intratumoral injections (n = 13, n = 8, and n = 9 mice per PBS, WT IAV, and IAV-CTLA4 groups, respectively). (F) An abscopal effect is induced by IAV-CTLA4 treatment. Left-flank, untreated tumors of mice administered IAV-CTLA4 demonstrate a delayed growth, compared to mice treated with WT IAV. NT, not tested; NS, not significant. Statistical analysis was performed using unpaired, two-tailed Student’s t tests (∗p ≤ 0.05, ∗∗p ≤ 0.01, and ∗∗∗p ≤ 0.001). Symbols indicate individual tumor volumes; note that on day 21 only one mouse remained in each of the PBS and WT IAV groups. For (E) and (F), data are shown until all PBS and WT IAV mice developed tumors >1,000 mm3. (G) IAV-CTLA4 treatment prolongs overall survival. LOD, limit of detection. Experiments were performed in duplicate and error bars represent SEM. Cell Reports 2018 22, 1-7DOI: (10.1016/j.celrep.2017.12.025) Copyright © 2017 The Author(s) Terms and Conditions