Intratumoral injections of small doses of agonist anti-CD137 mAb to directly act on CD137+ TILs render systemic immunotherapeutic effects that are synergistic.

Slides:



Advertisements
Similar presentations
c-Myc expression in response to pre-TCR stimulation.
Advertisements

AF647-RIS is internalized by TAMs in vivo.
Selinexor combines with immune checkpoint blockade to slow B16F10 melanoma tumor growth. Selinexor combines with immune checkpoint blockade to slow B16F10.
Melanoma donor (MD) NK cells are functionally impaired/exhausted.
Successful Immunotherapy against a Transplantable Mouse Squamous Lung Carcinoma with Anti–PD-1 and Anti-CD137 Monoclonal Antibodies  Arantza Azpilikueta,
Human NK cell development in NOD/SCID mice receiving grafts of cord blood CD34+ cells by Christian P. Kalberer, Uwe Siegler, and Aleksandra Wodnar-Filipowicz.
William H. D. Hallett, Weiqing Jing, William R. Drobyski, Bryon D
Synergy among receptors on resting NK cells for the activation of natural cytotoxicity and cytokine secretion by Yenan T. Bryceson, Michael E. March, Hans-Gustaf.
by Bindu Varghese, Adam Widman, James Do, Behnaz Taidi, Debra K
Combined A2A receptor and PD-1 blockade is not effective in IFNγ−/− mice. Combined A2A receptor and PD-1 blockade is not effective in IFNγ−/− mice. AT-3ovadim.
by Anil Dangi, Lei Zhang, Xiaomin Zhang, and Xunrong Luo
H31m1-PDL1 cells form progressively growing tumors in WT mice.
Tumor development of poorly immunogenic LLC and B16F10 cells modified to produce GM-CSF was markedly inhibited. Tumor development of poorly immunogenic.
IL-6 is dispensable for the suppressive activity of MDSC on primary CD4+ T-cell activation. IL-6 is dispensable for the suppressive activity of MDSC on.
DQ661 improves survival in colon cancer model and potentiates activity of gemcitabine in KPC pancreatic cancer syngeneic model. DQ661 improves survival.
Gr-1+ MDSC in tumor-bearing mice produce IL-6.
PD-1 blockade enhances elotuzumab efficacy in mouse tumor models
HIF-1α is critically involved in hypoxia-induced CD137 upregulation.
Fig. 3 In situ vaccination with CpG and anti-OX40 is therapeutic in a spontaneous tumor model. In situ vaccination with CpG and anti-OX40 is therapeutic.
Volume 24, Issue 9, Pages (September 2016)
STAT3 regulates IgE class switching in a Th2-type response.
Volume 20, Issue 5, Pages (May 2012)
mpJX-594 effects on tumor burden and leukocyte influx.
Anti-CD96 combines with anti–CTLA-4 or anti–PD-1 to suppress experimental and spontaneous lung metastases. Anti-CD96 combines with anti–CTLA-4 or anti–PD-1.
PD-1–PD-L1 axis is highly expressed and is not IFNγ-dependent in a subcutaneous murine model of PDA. A, experimental design for establishment of subcutaneous.
CD8+ T cells were immunomodulated and required for the efficacy of anti–4-1BB/anti–PD-1 combination treatment. CD8+ T cells were immunomodulated and required.
T-bethi MP cells produce IFN-γ in response to IL-12.
Highly related T9 and T3 sarcoma cells show distinct tumor growth patterns but similar PD-L1 expression kinetics in vivo. Highly related T9 and T3 sarcoma.
Administration of necroptosis-targeting AAVs in conjunction within vivo promotes durable tumor clearance. Administration of necroptosis-targeting AAVs.
Fig. 2 In situ vaccination of CpG in combination with anti-OX40 antibody cures established local and distant tumors. In situ vaccination of CpG in combination.
CD8 T cells play a critical role in responses of TILs due to BRAF inhibition. CD8 T cells play a critical role in responses of TILs due to BRAF inhibition.
Reduced tumor growth in CCR5-deficient mice is associated with perturbed killing ability of Treg cells. Reduced tumor growth in CCR5-deficient mice is.
Conversion of CD11bhighCD27high NK cells into MDSCs leads to CD11bhighCD27high and CD11bhighCD27low NK cell reduction. Conversion of CD11bhighCD27high.
Possible mechanisms of recognition of MHCIlow tumor cells by NSCs
Stereotactic radiotherapy increases Tregs in tumors.
Anti-Flk-1 treatment inhibits growth of s. c. 4T1 and B16 tumors. s. c
Experimental metastasis inhibition by primary tumors is mediated by NK cells and IFNγ. Experimental metastasis inhibition by primary tumors is mediated.
PD-L1 expressed on edited T3 sarcoma cells prevents their immune elimination. PD-L1 expressed on edited T3 sarcoma cells prevents their immune elimination.
PVHA increased anti-PD-L1–mediated growth inhibition in 4T1/HAS3 tumors. PVHA increased anti-PD-L1–mediated growth inhibition in 4T1/HAS3 tumors. A, 4T1/HAS3.
ODM-203 administration results in immune activation in a syngenic model. ODM-203 administration results in immune activation in a syngenic model. Effect.
5FU-induced specific activation of CD8+ T cells.
CD4+ and CD8+ TILs express surface CD137.
CDN–treated MOC1 tumors demonstrate enhanced activation of innate and antigen-specific adaptive immunity. CDN–treated MOC1 tumors demonstrate enhanced.
Dual blockade of PD-1 and CTLA-4 directly activates CD4+Foxp3− cells in the absence of CD8+ or CD4+Foxp3+ cells. Dual blockade of PD-1 and CTLA-4 directly.
Combination CDN and PD-L1 mAb treatment of established MOC1 tumors produces consistent tumor rejection. Combination CDN and PD-L1 mAb treatment of established.
TILs upregulate CD137 in an HIF-1α–dependent fashion.
Expression of B7-H1, B7-DC, and PD-1 on B cells.
The CD8+ cytotoxic T-cell response in Ron TK−/− hosts in response to tumors is necessary and sufficient to block metastasis. The CD8+ cytotoxic T-cell.
Intratumoral changes in critical lymphocyte populations and numbers after NKTR-214 treatment. Intratumoral changes in critical lymphocyte populations and.
PD-1 and CD103 are coexpressed on CD8+ T cells but demonstrate distinct mechanisms of regulation. PD-1 and CD103 are coexpressed on CD8+ T cells but demonstrate.
Tumor-resident CD8+ T cells rapidly expand after IL-15/IL-15Rα complex treatment. Tumor-resident CD8+ T cells rapidly expand after IL-15/IL-15Rα complex.
Anti-SEMA4D antibody regulates immune-cell infiltration and inhibits growth of Tubo.A5 orthotopic mammary carcinoma. Anti-SEMA4D antibody regulates immune-cell.
Tumor protection induced by therapeutic PLG vaccination in combination with blockade antibodies. Tumor protection induced by therapeutic PLG vaccination.
Stk11/Lkb1 ablation directly promotes primary resistance to PD-L1/PD-1 blockade in immunocompetent murine models of Kras-mutant LUAC. Stk11/Lkb1-proficient/deficient.
TAMs directly contribute to tumor hypoxia.
TCR stimulation by agonistic mAb in vivo administration inhibits mouse T-ALL development. TCR stimulation by agonistic mAb in vivo administration inhibits.
Anti-CD40 activates TAMs and recruits inflammatory monocytes.
Comparative effect of anti-TIM3 against experimental tumors.
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.
Immune checkpoint molecule expression in primary and secondary tumors following radiotherapy. Immune checkpoint molecule expression in primary and secondary.
The effect of TGFβ on the post-RT increase of Tregs in tumors.
Immune response of CEA-transgenic mice transferred with OT-1 splenocytes and immunized with ovalbumin and adjuvant. Immune response of CEA-transgenic mice.
LSECtin, expressed by B16 cells, inhibits the tumor-specific immune responses both in vivo and in vitro. LSECtin, expressed by B16 cells, inhibits the.
Antibody-mediated blockade of the immune-inhibitory PD-1–PD-L1 signaling pathway prolongs survival in poly(I:C)-treated mice. Antibody-mediated blockade.
Hypoxia upregulates CD137 expression in T lymphocytes undergoing activation. Hypoxia upregulates CD137 expression in T lymphocytes undergoing activation.
Induction of PD-1 in B cells by TLR agonists and primary HCC-SN.
Ex vivo profiling of PD-1 blockade using MDOTS
Depletion of CD8+, CD4+, and Ly6G+ cells in subcutaneous TUBO tumor-bearing BALB/c mice treated with KM100 + MTX. Depletions were conducted by intraperitoneal.
CD38 regulates tumor growth and metastasis by adenosine-mediated CD8+ T-cell suppression. CD38 regulates tumor growth and metastasis by adenosine-mediated.
Bezafibrate increases the number of effector CTLs by enhancing their survival capacity and proliferation. Bezafibrate increases the number of effector.
Presentation transcript:

Intratumoral injections of small doses of agonist anti-CD137 mAb to directly act on CD137+ TILs render systemic immunotherapeutic effects that are synergistic with PD-L1 (B7-H1) systemic blockade. Intratumoral injections of small doses of agonist anti-CD137 mAb to directly act on CD137+ TILs render systemic immunotherapeutic effects that are synergistic with PD-L1 (B7-H1) systemic blockade. A, mice bearing subcutaneous CT26 palpable tumors for 7 days were treated intratumorally with three 5-μg doses of control rat IgG or anti-CD137 mAb on days 7, 9, and 11 or/and with 3 i.p. doses of 100 μg anti-B7-H1 blocking mAb. Individual follow-up of tumor sizes is presented for each experimental group, the fraction of tumor-free mice at the end of the experiment is given in each graph, and the survival curves are presented. i.t., intratumorally. B, top, FACS histograms show that CT26 cells do not express B7-H1 in culture but are inducible by culture for 48 hours in the presence of IFN-γ or when cells are freshly recovered from grafted tumors in syngenic mice. Bottom FACS histograms show bright expression of PD-1 on CD3+CD4+ and CD3+CD8+ TILs harvested from CT26 tumors. C, dot plots showing CD137 and PD-1 double immunostaining on gated CD8+ and CD4+ T cells as indicated. Background immunostainings with isotype-matched control mAb tagged with the same fluorochromes are presented. D, BALB/c mice were subcutaneously inoculated with CT26 cells on days 0 and 4 in opposite flanks (black and red as indicated in schematic representation of the experiments). On days 7, 9, and 11, the primary tumor received intratumoral injections of 5 μg of 1D8 mAb or control IgG. Graphs represent individual tumor sizes for the primary (black) and secondary (red) tumors. The fraction of mice completely rejecting tumors is provided with the corresponding black and red graphs. Asís Palazón et al. Cancer Discovery 2012;2:608-623 ©2012 by American Association for Cancer Research