GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (7)
  • Zhou, Shenghua  (7)
  • 1
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 3_Supplement ( 2020-03-01), p. B53-B53
    Abstract: Background: Agonists of the Stimulator of Interferon Genes (STING) pathway have great potential in cancer immunotherapy. Activation of STING in tumor cells and/or antige- presenting cells (APCs) can induce type I Interferon production, leading to the induction of innate and adaptive immune response. We recently reported the discovery of the cyclic dinucleotide SB 11285 as a potent, first-in-class, STING agonist. Herein, we report SB 11 and SB 12 as unique linkage analogs of SB 11285. Methods: (a) Synthesis: Several linkage analogs of SB 11285 were synthesized by standard solution-phase phosphoramidite chemistry and screened for STING agonistic activity using the previously described SZ14 reporter cell lines, and SB 11 and SB 12 were identified as lead compounds., (b) Binding affinity: Differential scanning fluorimetry was used to determine binding affinity of SB 11 and 12 to wtSTING, with 2´,3´-cGAMP being used as a positive control. (c) Induction of IRF3 and NF-KB: wtTHP-1 or R232-THP1 or RAW cells carrying the respective reporter constructs were treated with SB 11 and 12 or controls for 22hrs and induction of IRF3 and NF-KB was determined as % fold-change in luminescence compared to vehicle-treated cells. (d) Induction of cytokines: PBMCs and mBMDCs were treated with SB 11 and 12 at different doses, and cell supernatants were analyzed for IFN-β, TNF-α, and RANTES by ELISA or multiplexing Luminex assays. (e) In vivo studies: Both SB 11 and 12 were tested for antitumor efficacy in CT26 syngeneic mouse tumor model after intravenous (i.v.) administration at 3mg/kg on days 1,5,9,14. Results: (i) SB 11 and 12 demonstrated high binding affinity to human wild-type and mouse STING that is evident by 16° thermal shift. (ii) SB 11 and 12 showed potent induction of (a) STING-dependent IRF3 (EC50: 0.8 and 39.7 nM) and NF-κB (EC50: 9 and 5265 nM) signaling induction in wtTHP-1 cells; (b) IRF3 (EC50: 2.6 and 317 nM) and NF-κB (EC50: 83 and 3783 nM) induction in R232-THP-1 cells; (c) IRF3 (EC50: 67 and 46 nM) in RAW macrophages. (iii) Both SB 11 and SB 12 induced STING dependent secretion of IFN-β (~10 ng/ml) in mBMDCs. (iv) In the CT26 colon carcinoma syngeneic mouse models, both SB 11 and 12 showed potent antitumor activity when administered by i.v. route with 98 and 97% TGI and 91 and 79% TGD, respectively. Conclusion: SB 11 and SB 12 showed excellent safety and antitumor activity in syngeneic mouse model when administered by i.v. route. SB 11 and SB 12 were shown to cause STING-dependent activation of IRF3 and NF-κB signaling, as well as the induction of type I IFN signature and ISGs. Further preclinical studies of systemically administered analogs SB 11 and SB 12 are in progress. Citation Format: Shenghua Zhou, Sreerupa Challa, Diane Shmidt, Leena Suppiah, Vishal Nair, Anjaneyulu Sheri, Geeta Meher, Rayomand Gimi, Seetharamaiyer Padmanabhan, Dillon Cleary, Radhakrishnan Iyer. Development of SB 11 and SB 12, structurally unique linkage analogs of SB 11285 as STING agonists for Immuno-oncology [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr B53.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. B87-B87
    Abstract: Background: Cancer immunotherapy is a highly effective therapeutic option for cancer patients. However, the overall response rate with checkpoint inhibitors and other related modalities has been modest. Targeting innate immune signaling pathways that induce type I IFN production to reprogram tumor microenvironment and restore antitumor immunity represents a novel immunotherapeutic approach. We have previously reported that SB 11285 is a first-in-class synthetic cyclic dinucleotide STING agonist, which has demonstrated potent antitumor activity, when used alone or combined with other antitumor agents, in several syngeneic mouse and rat tumor models when administered by intratumoral, intravenous or intraperitoneal routes. Presented here are studies that provide additional insights into the mechanism of action of SB 11285 and analogs. Methods: (a) SB 11285 induces Type I IFN production and other cytokines in human PBMCs and PBMC-derived monocytes. PBMCs and monocytes, isolated from fresh PBMCs using pan monocyte isolation kit (Miltenyi Biotec), were stimulated with SB 11285. Type I IFNs and other cytokines were quantified using regular and multiplex ELISA assays. (b) SB 11285 directly binds STING. To address whether SB 11285 directly binds wild-type human STING, surface plasmon resonance assay was performed with a Biacore T200 device and a biotinylated SB 11285 analog (Biot-SB 11285). (c) Activity against STING polymorphs. Evaluation of SB 11285 and analogs was carried out using both SZ14 reporter cells (HEK293-derived) and HEK293T cells that stably or transiently express STING polymorphic variants. (d) Pharmacodynamic studies. Normal BALB/c mice were injected intravenously with SB 11285 or its analogs at 9 mg/kg. Serum, spleen, and liver samples were collected to quantify RANTES and TNF-α using ELISA. The expression of representative ISGs in spleen samples, including IRF7, IFIT2, and OAS1b were quantified using real-time PCR. (e) Determination of cellular uptake of SB 11285 by PBMCs. The STING agonist activity of Biot-SB 11285 in inducing type I IFN response was confirmed using SZ14 reporter cells and THP1-Dual-WT reporter cells. Human PBMCs were incubated with Biot-SB 11285 and the cellular uptake of compound by immune cells was evaluated using flow cytometry. Results and Conclusion: Our studies provide significant mechanistic insights into the STING agonistic activity of SB 11285 and analogs in that they: (a) induce Type I IFNs, other cytokines and chemokines in PBMCs and monocytes, monocyte-derived dendritic cells; (b) directly bind STING with nanomolar affinity; (c) activate multiple human STING polymorphic variants; (d) induce cytokines, chemokines, and ISGs in mice following i.v. injection; and (e) are effectively taken up by monocytes and other immune cells. SB 11285 is being advanced to human clinical trials. Citation Format: Shenghua Zhou, Sreerupa Challa, Vishal Nair, Geeta Meher, Anjaneyulu Sheri, Rayomand Gimi, Seetharamaiyer Padmanabhan, Dillon Cleary, Leena Suppiah, Diane Schmidt, Santosh Khedkar, Radhakrishnan Iyer. Mechanistic insights into the antitumor activity of SB 11285—a novel STING agonist [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr B87.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Immunology Research Vol. 8, No. 4_Supplement ( 2020-04-01), p. B88-B88
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. B88-B88
    Abstract: Background: Agonists of the Stimulator of Interferon Genes (STING) pathway have great potential in cancer immunotherapy. Activation of STING in tumor cells and/or antigen-presenting cells (APCs) can induce type I Interferon production leading to the induction of innate and adaptive immune response. We recently reported the discovery of the cyclic dinucleotide SB 11285 as a potent, first-in-class, STING agonist. Herein, we report that SB 11312, a metabolite of SB 11285, is also a potent antitumor agent in vitro and in vivo. Methods: (a) Synthesis: SB 11312 was synthesized by solution-phase standard phosphoramidite chemistry. (b) Binding affinity: Differential scanning fluorimetry was used to determine binding affinity of SB 11312 to all STING variants, with 2′3′-cGAMP being used as a positive control. (c) Induction of IRF and NF-KB: THP-1 cells carrying the respective reporter constructs were treated with SB 11312 or controls for 22hrs and induction of IRF and NF-KB was calculated as % fold-change in luminescence compared to vehicle-treated cells. (d) Induction of cytokines: PBMCs and mBMDCs were treated with SB 11312 at different doses, and cell supernatants were analyzed for cytokines by multiplexing assays. (e) In vivo studies: Dose-ranging studies of SB 11312 were performed using i.t. (10 to 100μg, days 1,2,4,6,8), and i.v. (3 to 6mg/kg, days 1,3,5,8) routes in the CT26 syngeneic mouse tumor model. (f) Pharmacodynamic studies: Following i.v. administration of SB 11312 at 3 mg/kg in the CT26 model, flow cytometry and multiplexing assays of blood, spleen and tumor tissues were carried out. Results: (i) SB 11312 demonstrated high binding affinity to human wild-type and STING polymorphic variants, as well as mouse STING. (ii) SB 11312 showed potent induction of (a) STING-dependent IRF and NF-KB signaling induction in wild-type and human STING variants as well as RAW macrophages, (b) secretion of cytokines associated with type I interferon signature in PBMCs and mBMDCs, and (c) interferon stimulated genes (ISGs) in PBMCs, mBMDCs and THP1 cells. In the A20 lymphoma and CT26 colon carcinoma syngeneic mouse models, SB 11312 showed potent antitumor activity when administered by intravenous (i.v.) and intratumoral (i.t.) routes. In PD study, cytokine analysis showed induction of type I interferon signature without significant systemic inflammatory response. Conclusion: SB 11312, an active metabolite of SB 11285 showed excellent safety and antitumor activity in syngeneic mouse models administered by multiple routes. The mechanism of action of SB 11312 was shown to be STING-dependent that is associated with activation of IRF and NF-KB signaling, induction of type I IFN signature and ISGs. SB 11312 is being advanced for additional studies for application in immuno-oncology. Citation Format: Sreerupa Challa, Leena Suppiah, Diane Schmidt, Dillon Cleary, Shenghua Zhou, Vishal Nair, Kris Iyer. SB 11312, an active metabolite of SB 11285, is a potent and systemically bioavailable STING agonist [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr B88.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Immunology Research Vol. 5, No. 3_Supplement ( 2017-03-01), p. B39-B39
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 5, No. 3_Supplement ( 2017-03-01), p. B39-B39
    Abstract: Background: Immunotherapy has recently emerged as a transformative approach for the treatment of cancer; nevertheless, many patients remain unresponsive to treatment. It is being recognized that induction of type I interferons (IFN) and interferon-stimulated genes (ISGs) in tumor cells and within the tumor microenvironment (TME) is essential for modulating the host-immune response and inducing apoptosis of tumor cells. Furthermore, the antigen-presenting cells within TME can cause induction of adaptive immune response, through priming of CD8+ T cells and tumor killing. Importantly, the DNA released from damaged cells and cancer cells can be sensed by cyclic GMP-AMP synthase (cGAS) leading to the synthesis of cyclic-GMP-AMP (2',3'-cGAMP), a second messenger that activates Stimulator of Interferon Genes (STING) pathway resulting in the production of type I IFN and ISGs. The cumulative effects of activation of innate and adaptive immune response can result in potent anti-cancer effects. Therefore, therapeutic agents that activate the cGAS-STING signaling pathway in tumor cells and TME are urgently needed. Herein, we describe the discovery of novel potent, first-in-class small molecules for application in immuno-oncology. Methods: Using structure-guided drug design, in conjunction with published crystal structures of cyclic dinucleotides bound to STING, a focused library of dinucleotide compounds was synthesized using phosphoramidite chemistry and evaluated for: (a) Induction of IFN signaling: The compounds were screened for the induction of Interferon regulatory factor (IRF), ISG54, and NF-κB using reporter assays. We used HEK293 cell line (SZ14) stably expressing ISG54 (ISRE)-promoter-driven firefly luciferase reporter gene for screening and the active compounds were further characterized in THP1 cells and human primary PBMCs. The IRF, ISG54, and NF-κB induction was calculated from % fold-change in luminescence compared to DMSO-treated cells and EC50s of the compounds were ascertained to identify active compounds, (b) Expression of IFN-β and IRF7 in THP1 cells: THP1 cells were treated with active compounds or controls for 22hrs. RNA was extracted and the expression of IFN-β, IRF7, was ascertained using semi-quantitative RT-PCR, (c) Induction of pathogen recognition receptors (PRRs) including RIG-I, MDA5, LGP2, and OAS-1 and ISG54: THP1 cells and PBMCs were treated with active compounds, 2',3'-cGAMP (control), or DMSO and the gene expression of different PRRs, ISGs, was determined by quantitative RT-PCR using ΔΔct method, (d) Induction of cGAS-STING signaling using reporter assays: HEK293 cells stably expressing ISG54 were transfected with plasmids encoding human cGAS (wild-type, or K384A, K400A, or K411A mutants) and treated with active compounds, poly (dA:dT) (positive control), or DMSO for 21 hrs. ISG54 induction was calculated as fold-change in luminescence compared to DMSO-treated controls. (e) Cytotoxicity assays: THP1 cells were treated with active compounds or DMSO control with Lipofectamine and cytotoxicity assessed using the CellTiter-Glo® Luminescent assays. Cytotoxicity was calculated from %-fold change in luminescence compared to DMSO-treated sample. Results: Through in vitro assays in conjunction with Structure Activity Relationship studies, we have identified potent compounds that activate cGAS-STING signaling pathway for induction of IRF, IFN, and NF-κB. These compounds also cause induction of expression of PRRs, including RIG-I, MDA5, LGP2, as well as, ISG54 and OAS-1. Conclusion: We have discovered potent, first-in-class agents that cause induction of IFN, NF-κB, ISGs, and PRRs. Further optimization and preclinical evaluation of the compounds for application in immuno-oncology is underway. Citation Format: Shenghua Zhou, Sreerupa Challa, Seetharamaiyer Padmanabhan, Anjaneyulu Sheri, Samantha Delaney, Geeta Meher, Dillon Cleary, Rayomand Gimi, Santosh Khedkar, Radhakrishnan Iyer. Novel dinucleotides that activate STING signaling for immuno-oncology. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr B39.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Immunology Research Vol. 8, No. 4_Supplement ( 2020-04-01), p. B96-B96
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. B96-B96
    Abstract: Background: The activation of innate and adaptive immunity via Stimulator of Interferon Genes (STING) signaling is a potentially transformative immunotherapeutic strategy in cancer. We have previously reported that SB 11285 is a novel and highly potent STING agonist that can be delivered by intravenous, intraperitoneal and intratumoral routes. We report here the antitumor and pharmacodynamic studies of SB 11285 in multiple orthotopic and subcutaneous syngeneic mouse tumor models. Methods: (1) Orthotopic NBT-II syngeneic rat bladder model: Rat bladder tumor cells were surgically implanted in the bladder wall of rats (n =10). After 6 days, SB 11285 was administered by i.v. @ 0.5, 1.5, and 3mg/kg on days 1, 5, 9,1 3, 17, 21, 25 and 31. Groups of rats treated with docetaxel at 7.5mg/kg i.v. and saline were used as positive and negative controls, respectively. At the end of dosing period, the bladders were harvested and evaluated by measurement of bladder weight and gross pathology. (2) Pharmacodynamic (PD) studies: Syngeneic mouse subcutaneous CT26 colon carcinoma and orthotopic 4T1 breast cancer models were used. In the CT26 model, when mean tumor volume (MTV) reached 125mm3, SB 11285 was administered at 3mg/kg i.v on days 1 and 5. In the orthotopic 4T1 model, SB 11285 was administered i.p. @ 10mg/kg on days 5, 7, 9, 11, 13, 17 and 19. Blood, lymph nodes, spleen and tumor were harvested and evaluated for (i) cytokine analysis by multiplexing assays and (ii) enumeration of CD4+, CD8+, Tregs, macrophages and MDSCs in blood, lymph nodes, spleen and tumor. Results: (1) In the orthotopic NBT-II rat bladder study, animals that received SB 11285 in all doses remained tumor-free based upon gross pathology and bladder weight measurements. SB 11285 was shown to be well tolerated and safe. (2) In both the 4T1 and CT26 PD studies, cytokine analysis of blood and tissues showed strong induction of type I interferon signature without significant systemic inflammatory response. Flow cytometric analysis of blood, lymph nodes, spleen and tumor revealed significant increase in macrophage & CD8+ T-cell infiltration and decrease in Tregs & CD4+ T-cell infiltration. Conclusion: SB 11285 showed potent antitumoral activity in multiple orthotopic and subcutaneous models that correlated with antitumoral immune response mediated by the induction of IFNs, and ISGs and other cytokines along with reduction of Tregs. SB 11285 is being advanced to human clinical trials. Citation Format: Sreerupa Challa, Balaji Ramachandran, Lalitha Vijayakrishnan, Douglas Weitzel, Shenghua Zhou, Kris Iyer. Pharmacodynamic studies of SB 11285, a systemically bioavailable STING agonist in orthotopic tumor models [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr B96.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Immunology Research Vol. 5, No. 3_Supplement ( 2017-03-01), p. B40-B40
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 5, No. 3_Supplement ( 2017-03-01), p. B40-B40
    Abstract: Immunotherapy has recently emerged as a transformative approach for the treatment of cancer; nevertheless, many patients remain unresponsive to treatment. Recent evidence suggests that the activation of Stimulator of Interferon Genes (STING) pathway in tumor cells and/or antigen presenting cells (APCs) within the tumor microenvironment (TME) can induce type I Interferon production leading to apoptosis of tumor cells, as well as, induction of adaptive immune response (through priming of CD8+ T cells to tumor-associated antigens) thereby providing a powerful anti-cancer strategy. Therefore, therapeutic agents that activate STING signaling pathway in tumor cells and APCs in the TME are urgently needed. Herein, we describe the discovery of highly potent and selective first-in-class STING agonists for application in immuno-oncology. Methods: Using structure-guided drug design, in conjunction with published crystal structures of different cyclic dinucleotides bound to STING, a focused library of nucleotide compounds was prepared using standard phosphoramidite chemistry. The compounds were screened for induction of Interferon regulatory factor (IRF), Interferon-stimulated gene 54 (ISG54), and NF-KB using reporter assays. We used HEK293 cell line stably expressing ISG54 (ISRE)-promoter-driven firefly luciferase reporter gene for initial hit discovery and the actives were further characterized in PBMCs and THP1 cells. The IRF and NF-kB induction was calculated from % fold-change in luminescence compared to DMSO-treated cells and EC50 of the compounds were ascertained using Xlfit. Lead STING agonists were further evaluated for: (a) Binding affinity: Binding assays were conducted by Differential Scanning Fluorimetry (DSF) and Tm was calculated using Thermal Shift software, (b) Induction of pathogen recognition receptors (PRRs), ISGs and Programmed Death Ligands 1 & 2 (PDL1, PDL2) genes: THP1 cells and PBMCs were treated with various concentrations of lead compounds or 2,'3'-cGAMP or DMSO and the gene expression of different PRRs, ISGs, PDL1, and PDL2 was determined by quantitative RT-PCR using ΔΔct method, (c) Apoptosis-inducing activity: PBMCs and THP1 cells were treated with various concentrations of lead compounds, 2',3'-cGAMP, or DMSO control and the apoptotic activity was evaluated using Caspase-Glo® 3/7 Assay (Promega), and (d) In vitro anti-tumor activity: STING-dependent anti-tumor activity of lead compounds in various tumor cell lines was assessed by either high-content imaging or through Cell titer Glo® Cytotoxicity Assay (Promega). Cell survival was calculated based upon % reduction of live cells compared to DMSO control. CC50 of the compounds were generated by curve fit in Xlfit. Results: Through in vitro assays in conjunction with Structure Activity Relationship (SAR) studies, we have identified several highly potent and selective first-in-class STING agonists. A promising lead nucleotide compound SB 11285 caused STING-dependent induction of: (a) IRF with an EC50 of 2 nM that is 1000-fold more potent than the natural STING agonist 2',3'-cGAMP, (b) NF-kB with an EC50 of 200 nM that is & gt;200-fold more potent than 2',3'-cGAMP, (c) selective apoptosis of human monocyte leukemic cell lines (CC50, 500 nM) as compared to normal PBMCs through induction of IFN, and NF-kB signaling, and (d) expression of various PRRs and ISGs including RIG-I, MDA-5, LGP2, ISG54 and OAS-1, as well as, PDL1 and PDL2. Finally, SB 11285 showed potent in vitro anti-tumor activity in multiple tumor cell lines. Conclusion: We have discovered highly potent first-in-class STING agonists that show excellent selectivity in induction of IFN, NF-KB, ISGs, and PRRs, and apoptosis of tumor-derived cell lines. The lead STING agonist SB 11285 has potent immune-modulating, as well as, anti-tumor activities and is being advanced for additional preclinical studies for application in immuno-oncology. Citation Format: Sreerupa Challa, Shenghua Zhou, Anjaneyulu Sheri, Seetharamaiyer Padmanabhan, Samantha Delaney, Geeta Meher, Dillon Cleary, Vishal Nair, Rayomand Gimi, Santosh Khedkar, Radhakrishnan Iyer. Nucleotide analogs as novel STING agonists for immuno-oncology. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr B40.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 4237-4237
    Abstract: Immune checkpoint inhibitor (ICI) anti-PD-1 antibody (mAb) monotherapy represented a significant advancement in cancer treatment, however most patients will ultimately progress, signaling the need for improvement. Combination approaches with other ICI mAbs may improve outcomes, however, systemic administration of immunostimulatory mAb combinations further increases risk for treatment-induced immune related adverse effects (irAEs). We have previously shown that intratumoral (IT) administration of PH-894, a self-delivering RNAi compound targeting BRD4 built on proprietary INTASYL™ technology, imparts tumor control in vitro and in vivo via effects on both T cells and tumor cells. Local priming of the immune response has shown promise toward potentiating the impact of systemic immunotherapy; therefore, we hypothesized that IT PH-894 might potentiate the efficacy of systemic anti-PD-1 mAb therapy. To assess this in vivo, an identical inoculum (5e05) of CT26 cells was implanted subcutaneously into the bilateral flanks of BALB/c mice. Vehicle (PBS; IT), anti-PD-1 mAb (0.1 mg/dose; intraperitoneal (IP)), PH-894 (0.5 mg/dose; IT) or combination anti-PD-1 mAb (0.1 mg/dose; IP) + PH-894 (0.5 mg/dose; IT) were administered, with IT doses limited to the tumor on the left flank only, on Days 1, 4, 7, 10 and 13. Tumors on the opposite flank were left untreated. Tumor volumes and body weights were recorded. Tumors, spleens, and lymph nodes were isolated for ex vivo mechanistic studies. The abscopal effects of IT PH-894 were confirmed in a bilateral Hepa1-6 model of murine hepatoma. In the CT26 model, as expected, growth of both tumors was inhibited with systemic anti-PD-1. Treatment with IT PH-894 provided a similar level of tumor control compared to treatment with IP anti-PD-1 mAb, both for the PH-894 directly-treated tumors, but in addition also towards the PH-894 untreated distal tumors, an abscopal impact. Additionally, IT PH-894 enhanced the antitumor efficacy of systemic anti-PD-1 mAb therapy, not only for the PH-894 locally treated tumors, but for the PH-894 untreated distal tumors. These data show that IT administered PH-894 potentiated the efficacy of systemic anti-PD-1 mAb, both locally and toward PH-894 untreated distal tumors. In addition, PH-894 silencing of BRD4 not only inhibited tumor growth at the local site of therapy, but also elicited an abscopal effect toward distal tumors, with efficacy similar to that provided by systemically administered anti-PD-1 mAb. These results indicate that IT PH-894 can potentiate the efficacy of systemic anti-PD-1 mAb, warranting further investigation of clinical applications in combination with anti-PD-1 mAbs toward improving patient outcomes. Citation Format: Benjamin Cuiffo, Melissa Maxwell, Dingxue Yan, Shenghua Zhou, Brianna Rivest, Andrew Boone, James Cardia, Simon P. Fricker. Local administration of BRD4-targeting self-delivering RNAi (PH-894) provides abscopal efficacy toward untreated distal tumors and potentiates the efficacy of systemic anti-PD-1 antibody therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 4237.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...