Abstract Methods and Results Results Introduction Conclusions

Slides:



Advertisements
Similar presentations
John A. Barrett Ph.D. Ziopharm Oncology, Boston MA 02129
Advertisements

Meredith Baker, MD PI: Bo Lu, MD – Radiation Oncology May 29th, 2014
Combination therapy with reovirus and PD-1 blockade effectively establishes tumor control via innate and adaptive immune responses Karishma Rajani1, Christopher.
Summary and Conclusion Hypothesis and Rationale
Up-Regulation of Activating Transcription Factor-5 Suppresses SAP Expression to Activate T Cells in Hemophagocytic Syndrome Associated with Epstein-Barr.
Volume 10, Issue 2, Pages (August 2004)
An anti-CD20–IL-2 immunocytokine is highly efficacious in a SCID mouse model of established human B lymphoma by Stephen D. Gillies, Yan Lan, Steven Williams,
Cyclin-Dependent Kinase 2 Promotes Tumor Proliferation and Induces Radio Resistance in Glioblastoma  Jia Wang, Tong Yang, Gaofeng Xu, Hao Liu, Chunying.
Molecular Therapy - Oncolytics
Antigen-Specific CD8 T Cells Can Eliminate Antigen-Bearing Keratinocytes with Clonogenic Potential via an IFN-γ-Dependent Mechanism  Rachel L. De Kluyver,
Volume 44, Issue 2, Pages (February 2016)
William H. D. Hallett, Weiqing Jing, William R. Drobyski, Bryon D
A humanized single-chain antibody against beta 3 integrin inhibits pulmonary metastasis by preferentially fragmenting activated platelets in the tumor.
Volume 15, Issue 2, Pages (February 2007)
CR3 (CD11b/CD18) and CR4 (CD11c/CD18) Are Involved in Complement-Independent Antibody-Mediated Phagocytosis of Cryptococcus neoformans  Carlos P Taborda,
by Norman Nausch, Ioanna E
Macrophages from C3-deficient mice have impaired potency to stimulate alloreactive T cells by Wuding Zhou, Hetal Patel, Ke Li, Qi Peng, Marie-Bernadette.
by Éric Aubin, Réal Lemieux, and Renée Bazin
by Jamie Honeychurch, Alison L. Tutt, Thomas Valerius, Ingmar A. F. M
Volume 13, Issue 4, Pages (October 2000)
by Bindu Varghese, Adam Widman, James Do, Behnaz Taidi, Debra K
Volume 42, Issue 2, Pages (February 2015)
Virally infected and matured human dendritic cells activate natural killer cells via cooperative activity of plasma membrane-bound TNF and IL-15 by Lazar.
Recombinant mumps virus as a cancer therapeutic agent
Molecular Therapy - Oncolytics
Volume 28, Issue 3, Pages (September 2015)
Maraba treatment sensitizes 4T1 tumors to immune checkpoint blockade
Volume 13, Issue 1, Pages (January 2006)
Volume 30, Issue 3, Pages (September 2016)
Molecular Therapy - Nucleic Acids
Volume 27, Issue 2, Pages (August 2007)
by Adrienne Sallets, Sophie Robinson, Adel Kardosh, and Ronald Levy
Volume 25, Issue 1, Pages (January 2014)
Volume 16, Issue 11, Pages (September 2016)
Volume 44, Issue 3, Pages (March 2016)
S100A15, an Antimicrobial Protein of the Skin: Regulation by E
by Kalpana Parvathaneni, and David W. Scott
Oncolytic VSV Primes Differential Responses to Immuno-oncology Therapy
Volume 27, Issue 4, Pages (October 2007)
Volume 13, Issue 2, Pages (October 2015)
Volume 22, Issue 1, Pages (January 2014)
Interleukin-18 and the Costimulatory Molecule B7-1 Have a Synergistic Anti-Tumor Effect on Murine Melanoma; Implication of Combined Immunotherapy for.
Volume 43, Issue 5, Pages (November 2015)
Reovirus FAST Protein Enhances Vesicular Stomatitis Virus Oncolytic Virotherapy in Primary and Metastatic Tumor Models  Fabrice Le Boeuf, Simon Gebremeskel,
Volume 12, Issue 5, Pages (November 2005)
Volume 23, Issue 7, Pages (May 2018)
Volume 22, Issue 1, Pages (January 2014)
Volume 12, Issue 2, Pages (February 2000)
Lukxmi Balathasan, Vera A
Volume 20, Issue 5, Pages (May 2012)
Volume 24, Issue 1, Pages (January 2016)
Molecular Therapy - Oncolytics
Volume 29, Issue 3, Pages (March 2016)
Sangeet Lal, Corey Raffel  Molecular Therapy - Oncolytics 
Volume 26, Issue 1, Pages (January 2018)
Volume 25, Issue 10, Pages (October 2017)
Molecular Therapy - Oncolytics
Volume 6, Issue 5, Pages (November 2002)
Sindbis Viral Vectors Transiently Deliver Tumor-associated Antigens to Lymph Nodes and Elicit Diversified Antitumor CD8+ T-cell Immunity  Tomer Granot,
Volume 26, Issue 4, Pages (April 2018)
Molecular Therapy - Nucleic Acids
Single-Shot, Multicycle Suicide Gene Therapy by Replication-Competent Retrovirus Vectors Achieves Long-Term Survival Benefit in Experimental Glioma  Chien-Kuo.
Dissecting the Multifactorial Causes of Immunodominance in Class I–Restricted T Cell Responses to Viruses  Weisan Chen, Luis C. Antón, Jack R. Bennink,
Molecular Therapy - Methods & Clinical Development
Adoptive transfer of gene-engineered CD4+ helper T cells induces potent primary and secondary tumor rejection by Maria Moeller, Nicole M. Haynes, Michael.
Volume 19, Issue 4, Pages (October 2003)
Efficacy of KM100 and/or MTX in BALB/c mice bearing subcutaneous TUBO tumors. Efficacy of KM100 and/or MTX in BALB/c mice bearing subcutaneous TUBO tumors.
PDL192 and inhibit the growth of xenograft tumors.
Volume 25, Issue 4, Pages (April 2017)
CD38 regulates tumor growth and metastasis by adenosine-mediated CD8+ T-cell suppression. CD38 regulates tumor growth and metastasis by adenosine-mediated.
Presentation transcript:

Abstract Methods and Results Results Introduction Conclusions Logo Placeholder PD-L1 Checkpoint Blockade Using a Single-Chain Variable Fragment Targeting PD-L1 Delivered by Retroviral Replicating Vector Toca 521 Enhances Anti-Tumor Effect in Cancer Models Amy H. Lin, Kader Yagiz, Andrew Hofacre, Anthony W. Munday, Fernando Lopez Espinoza, Daniel Mendoza, Cynthia Burrascano, Harry E. Gruber, Leah Mitchell, and Douglas J. Jolly Tocagen Inc., San Diego, CA 92109, USA Abstract Methods and Results Results scFv PD-L1 Inhibits Tumor Growth and Elicits Immune Memory Function in Tu-2449SC Mouse Model RRV-scFv-PDL1 Vectors Replicate Efficiency and Produce Viral Titers Immune checkpoint inhibitors are a breakthrough for immunotherapies in treating cancer patients. However, the response rate ranges from 20-30% across tumor types with a number of immune-related adverse events associated with treatment which can lead to a high rate of treatment discontinuation (~ 12-39% patients). An RRV expressing a single‑chain variable fragment targeting PD‑L1 (RRV‑scFv‑PDL1) has demonstrated that scFv PD-L1 binds specifically to both mouse and human PD-L1, and the binding specificity of scFv PD‑L1 was further confirmed by competitive ELISA showing that RRV‑generated scFv PD‑L1 was able to compete for target occupancy against a commercially available monoclonal antibody against PD‑L1. A bystander effect also has been observed with scFv PD‑L1 protein expression from RRV‑scFv‑PDL1 infected tumor cells in a dose-dependent manner showing saturated receptor binding to the cell surface PD‑L1 of bystander cells when co-cultured with as low as 10% scFv‑PD‑L1 expressing cells. In addition, in vivo mouse models showed that tumor cells infected with RRV-scFv-PDL1 conferred robust and durable immune-mediated anti-tumor activity superior to systemically administered anti-PD‑1 or anti‑PD‑L1 monoclonal antibodies. Local scFv PD-L1 production in the tumor and these results support that RRV‑scFv‑PDL1 checkpoint inhibition is potentially therapeutic and may provide an improved safety and efficacy profile compared to systemic monoclonal antibodies. The anti-tumor activity of RRV‑scFv‑PDL1 may be a consequence of the delivery approach, which provides a consistent high level of payload and bystander index which is localized within the tumor microenvironment. Furthermore, with selective local production, RRV‑scFv‑PDL1 may be therapeutically beneficial in combination with other entities as an immuno-oncology agent with less concern of combined autoimmunity adverse events. A B   TU/mL Std Dev RRV-scFv 2.09E+06 4.80E+05 RRV-scFv-HF 2.08E+06 6.73E+05 RRV-scFvFc 1.98E+06 4.38E+05 RRV-scFv Fc-HF 1.29E+06 1.87E+05 Mixture of 2 x 106 Tu-2449SC tumor cells (Mitchell et al., 2017) pre-transduced with RRV-scFv-PDL1 and RRV-GFP at indicated ratios were implanted subcutaneously in B6C3F1 mice (n = 10 per group). Tumor growth was monitored over time. Anti-PD-1 antibody (Clone J43) was included as a control and was intraperitoneally administered on day 0 (300µg/mouse), day 3, 6 and 9 (200µg/mouse). (B) All Mice that cleared their initial tumor implant from scFv treated groups (n = 15), were challenged with Tu-2449SC on the other side of the flank as were naïve control animals and tumor growth was monitored overtime. Naïve group (n=5) was included as a control. Error bars indicate the standard error of the dataset. scFv PD-L1 Competes with PD-1 for PD-L1 Binding Mouse PD-1/PD-L1 a-PD-L1 Ab scFv scFvFc Human PD-1/PD-L1 a-PD-L1 Ab scFv scFvFc scFv PD-L1 Inhibits Tumor Growth and Elicits Immune Memory Function in EMT6 Orthotopic Breast Cancer Model A B Wells in a 96-well plate were coated with 100 μL of recombinant mouse or human PD-L1 at 8 μg/mL. Various volume of supernatant from CT26 cells expressing scFv PD-L1 or scFvFc PD-L1 were co-incubated with His-tagged recombinant mouse or human PD-1 at 8 μg/mL followed by incubation with anti-His antibody for detection of PD-1 binding. Anti-PD-L1 antibodies were included as a positive control. Percentage of inhibition was calculated relative to supernatant from RRV-GFP infected cells that do not express scFv PD-L1 nor scFvFc PD-L1. Data are presented as means ± SD from one of three independent experiments. Introduction Background: RRV-scFv-PDL1 (Toca 521) RRV-scFv-PDL1 is a retroviral replicating vector (RRV) expressing a single‑chain variable fragment targeting PD‑L1 (scFv PD-L1) Buds off from tumor cells but does not lyse tumor cells as part of the virus life cycle Utilizes self-cleavage 2A peptide which allows proper separation of scFv PD-L1 from the viral envelope protein Selectively infects and produces scFv PD-L1 in tumor cells scFv PD-L1 produces from the infected tumor cells is secreted into the tumor microenvironment (A) Kaplan–Meier survival analysis of orthotopic breast cancer model. Mixture of 5 x 104 EMT6 tumor cells pre-transduced with RRV-scFv-PDL1 or RRV-GFP at indicated ratios were implanted in the mammary fat pad in BALB/C mice (n = 10 per group). Survival was monitored for 90 days. Anti-PD-1 antibody (Clone J43) was included as a control and was intraperitoneally administered on day 10 (300µg/mouse), day 13, 16, 19 (200µg/mouse). Animals with necrotic tumors were censored from analysis (indicated by tick marks). (B) Mice that survived initial tumor implant from scFv treated groups (n = 5) were challenged with EMT6 tumor cells on the flank and tumor growth was monitored overtime. Naïve animals were included as controls. Error bars indicate the standard error of the dataset. scFv PD-L1 Bystander Trans-Binding Activity to PD-L1 on the Cell Surface RRV-scFv-PDL1 infected cells RRV-GFP infected cells scFv PD-L1 PD-L1 Intracranial Injection of RRV-scFv-PDL1 (Toca 521) Improves Survival in Syngeneic Orthotopic Glioma Model Superinfection Resistant RRV-Infected Cells at Various Ratios (5-50%) RRV-GFP infected cells RRV-scFv-PDL1 infected cells Vector constructs of RRV-scFv-PDL1 (Toca 521): gag pol 4070A env 2A scFv PD-L1 U3-R-U5 PBS 5’ SS ψ 3’ SS PPT A B VH GS VL IgG1 Fc H F scFv scFvFc scFv-HF scFvFc-HF GFP HA 5/95 10/90 20/80 30/70 40/60 100% GFP 100% scFv-HF naïve 50/50 Proper protein translation and separation from the 2A peptide SU TM-2A Env 2A scFv-PDL1 scFv (A)n (A) Kaplan–Meier survival analysis of a syngeneic orthotopic glioma model. Mice in each group (n=10) were intracranially implanted with 1.4 x 104 of Tu-2449 cells. Survival analysis was monitored for 90 days. Mice in the experimental groups were injected with Toca 521 of 1 x 105 or 1 x 106 transduction unit (TU) at day 4 post tumor implant. Control groups are mice bearing 100% pre-transduced scFv PD-L1 expressing tumor cells and mice treated anti-PD-1 antibody (Clone J43) or isotype control. Anti-PD-L1 antibody was intraperitoneally administered at indicated time points (300µg/mouse induction dose followed by 200µg/mouse maintenance dose). Statistical significance of survival between mice treated with isotype and scFv PD-L1 or Toca 521 treated groups was determined by the Log-rank (Mantel-Cox) test and is indicated by the brackets; **** p = 0.0252, *** p = 0.0649, ** p = 0.0832, * p = 0.3960. (B) Mice which had survived from initial tumor implant from scFv treated or Toca 521 treated groups were challenged with Tu-2449SC tumor cells on the flank and tumor growth was monitored overtime. Mixtures of HA-tagged scFv PD-L1 (scFv-HF) expressing and GFP-tagged non-scFv PD-L1 expressing EMT6 cells at various ratios were stained with anti-HA antibody for detection of PD-L1 binding on the cell surface. The numbers on the second row indicate the ratios of scFv PD-L1 to non-scFv PD-L1 cells. Data presented are from one of three independent experiments. scFv PD-L1 Reverses PD-1/PD-L1 Mediated Immune Suppression Jurkat/PD-1 Raji SEE α-PD-L1 scFv PD-L1 Raji/PD-L1 Raji B/PD-L1 cells MHC B7 PD-L1 Jurkat/PD-1 T cells TCR CD28 PD-1 + - Co-incubation of Jurkat T cells with Raji B cells presenting the superantigen staphylococcal enterotoxin E (SEE), which binds to the TCR and to MHC class II molecules, leads to stimulation of Jurkat T cells and IL-2 production. Co-incubation of Jurkat T cells overexpressing PD-1 and Raji B cells overexpressing PD-L1 blocks the stimulation and IL-2 production. Anti-human PD-L1 antibody was included as a control. Data are presented as means ± SD from one of three independent experiments. hIL-2 [pg/mL] a-Flag a-GAPDH a-HA naive scFv scFv-HF scFvFc-HF scFvFc naive scFv scFv-HF scFvFc a-2A Env-2A TM-2A * scFvFc-HF Conclusions scFv PD-L1 can be expressed in the RRV-2A configuration and shows efficient separation from the viral envelope protein. scFv PD-L1 expressed and secreted from Toca 521 infected cells competes with PD-1 binding to PD-L1 in a dose- dependent manner. scFv PD-L1 can rescue PD-1/PD-L1 mediated immune suppression in vitro. scFv PD-L1 expressed from Toca 521 infected tumor cells shows anti-tumor response in several tumor models. With selective local production, Toca 521 may have the potential to improve safety and/or efficacy over current check-point inhibitors as a monotherapy or in combination. Proposed mechanism of action: Alleviate tumor suppression mediated via PD-1/PD-L1 and reinvigorate Teff function in the tumor microenvironment TCR CD28 PD-1 PD-L1 scFv PD-L1 Teff Cell Macrophage Tumor Cells © 2018 Tocagen Inc.