8 results on '"Nell Narasappa"'
Search Results
2. Supplementary Figure from Targeting CD73 with AB680 (Quemliclustat), a Novel and Potent Small-Molecule CD73 Inhibitor, Restores Immune Functionality and Facilitates Antitumor Immunity
- Author
-
Matthew J. Walters, Kelsey E. Sivick, Uli Schindler, Jay P. Powers, Stephen W. Young, Jaroslaw Kalisiak, Nikki Kimura, Timothy W. Park, Bhamini Purandare, Susan L. Paprcka, Fangfang Yin, Akshata R. Udyavar, Elaine Ginn, Ada Chen, Kristen Zhang, Daniel DiRenzo, Nell Narasappa, Jesus Banuelos, Annette Becker, Joanne B.L. Tan, and Dana Piovesan
- Abstract
Supplementary Figure from Targeting CD73 with AB680 (Quemliclustat), a Novel and Potent Small-Molecule CD73 Inhibitor, Restores Immune Functionality and Facilitates Antitumor Immunity
- Published
- 2023
3. Data from Targeting CD73 with AB680 (Quemliclustat), a Novel and Potent Small-Molecule CD73 Inhibitor, Restores Immune Functionality and Facilitates Antitumor Immunity
- Author
-
Matthew J. Walters, Kelsey E. Sivick, Uli Schindler, Jay P. Powers, Stephen W. Young, Jaroslaw Kalisiak, Nikki Kimura, Timothy W. Park, Bhamini Purandare, Susan L. Paprcka, Fangfang Yin, Akshata R. Udyavar, Elaine Ginn, Ada Chen, Kristen Zhang, Daniel DiRenzo, Nell Narasappa, Jesus Banuelos, Annette Becker, Joanne B.L. Tan, and Dana Piovesan
- Abstract
T cells play a critical role in the control of cancer. The development of immune checkpoint blockers (ICB) aimed at enhancing antitumor T-cell responses has revolutionized cancer treatment. However, durable clinical benefit is observed in only a subset of patients, prompting research efforts to focus on strategies that target multiple inhibitory signals within the tumor microenvironment (TME) to limit tumor evasion and improve patient outcomes. Adenosine has emerged as a potent immune suppressant within the TME, and CD73 is the major enzyme responsible for its extracellular production. CD73 can be co-opted within the TME to impair T-cell–mediated antitumor immunity and promote tumor growth. To target this pathway and block the formation of adenosine, we designed a novel, selective, and potent class of small-molecule inhibitors of CD73, including AB680 (quemliclustat), which is currently being tested in patients with cancer. AB680 effectively restored T-cell proliferation, cytokine secretion, and cytotoxicity that were dampened by the formation of immunosuppressive adenosine by CD73. Furthermore, in an allogeneic mixed lymphocyte reaction where CD73-derived adenosine had a dominant suppressive effect in the presence of PD-1 blockade, AB680 restored T-cell activation and function. Finally, in a preclinical mouse model of melanoma, AB680 inhibited CD73 in the TME and increased the antitumor activity of PD-1 blockade. Collectively, these data provide a rationale for the inhibition of CD73 with AB680 in combination with ICB, such as anti–PD-1, to improve cancer patient outcomes.
- Published
- 2023
4. Targeting CD73 with AB680 (Quemliclustat), a Novel and Potent Small-Molecule CD73 Inhibitor, Restores Immune Functionality and Facilitates Antitumor Immunity
- Author
-
Dana Piovesan, Joanne B.L. Tan, Annette Becker, Jesus Banuelos, Nell Narasappa, Daniel DiRenzo, Kristen Zhang, Ada Chen, Elaine Ginn, Akshata R. Udyavar, Fangfang Yin, Susan L. Paprcka, Bhamini Purandare, Timothy W. Park, Nikki Kimura, Jaroslaw Kalisiak, Stephen W. Young, Jay P. Powers, Uli Schindler, Kelsey E. Sivick, and Matthew J. Walters
- Subjects
Mice ,Cancer Research ,Adenosine ,Oncology ,Programmed Cell Death 1 Receptor ,Tumor Microenvironment ,Animals ,Humans ,Immune Checkpoint Inhibitors ,Melanoma - Abstract
T cells play a critical role in the control of cancer. The development of immune checkpoint blockers (ICB) aimed at enhancing antitumor T-cell responses has revolutionized cancer treatment. However, durable clinical benefit is observed in only a subset of patients, prompting research efforts to focus on strategies that target multiple inhibitory signals within the tumor microenvironment (TME) to limit tumor evasion and improve patient outcomes. Adenosine has emerged as a potent immune suppressant within the TME, and CD73 is the major enzyme responsible for its extracellular production. CD73 can be co-opted within the TME to impair T-cell–mediated antitumor immunity and promote tumor growth. To target this pathway and block the formation of adenosine, we designed a novel, selective, and potent class of small-molecule inhibitors of CD73, including AB680 (quemliclustat), which is currently being tested in patients with cancer. AB680 effectively restored T-cell proliferation, cytokine secretion, and cytotoxicity that were dampened by the formation of immunosuppressive adenosine by CD73. Furthermore, in an allogeneic mixed lymphocyte reaction where CD73-derived adenosine had a dominant suppressive effect in the presence of PD-1 blockade, AB680 restored T-cell activation and function. Finally, in a preclinical mouse model of melanoma, AB680 inhibited CD73 in the TME and increased the antitumor activity of PD-1 blockade. Collectively, these data provide a rationale for the inhibition of CD73 with AB680 in combination with ICB, such as anti–PD-1, to improve cancer patient outcomes.
- Published
- 2022
5. Design, Synthesis, and Structure–Activity Relationship Optimization of Pyrazolopyrimidine Amide Inhibitors of Phosphoinositide 3-Kinase γ (PI3Kγ)
- Author
-
Dillon H. Miles, Ada Chen, Manmohan Reddy Leleti, Divyank Soni, Stephen W Young, Artur K. Mailyan, Kenneth V. Lawson, Stefan G Shaqfeh, Ehesan U. Sharif, Jenna L. Jeffrey, Jesus Banuelos, Xuelei Yan, Samuel L Drew, Kimberline Gerrick, Nigel Walker, Puja Dhanota, Lixia Jin, Jay P. Powers, Elaine Ginn, Guillaume Mata, Kent Wong, Hema Singh, Jeremy Fournier, Joel W. Beatty, Ulrike Schindler, Amber Pham, Matthew J. Walters, Jie Chen, Cesar Meleza, Xiaoning Zhao, and Nell Narasappa
- Subjects
Male ,Pyrazolopyrimidine ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,chemistry.chemical_compound ,Immune system ,Amide ,Drug Discovery ,Animals ,Class Ib Phosphatidylinositol 3-Kinase ,Humans ,Structure–activity relationship ,Potency ,Phosphoinositide-3 Kinase Inhibitors ,Phosphoinositide 3-kinase ,biology ,Amides ,Phenotype ,In vitro ,Rats ,Molecular Docking Simulation ,Pyrimidines ,chemistry ,Biochemistry ,Drug Design ,biology.protein ,Molecular Medicine - Abstract
Phosphoinositide-3-kinase γ (PI3Kγ) is highly expressed in immune cells and promotes the production and migration of inflammatory mediators. The inhibition of PI3Kγ has been shown to repolarize the tumor immune microenvironment to a more inflammatory phenotype, thereby controlling immune suppression in cancer. Herein, we report the structure-based optimization of an early lead series of pyrazolopyrimidine isoindolinones, which culminated in the discovery of highly potent and isoform-selective PI3Kγ inhibitors with favorable drug-like properties. X-ray cocrystal structure analysis, molecular docking studies, and detailed structure-activity relationship investigations resulted in the identification of the optimal amide and isoindolinone substituents to achieve a desirable combination of potency, selectivity, and metabolic stability. Preliminary in vitro studies indicate that inhibition of PI3Kγ with compound 56 results in a significant immune response by increasing pro-inflammatory cytokine gene expression in M1 macrophages.
- Published
- 2021
6. A novel luciferase-based assay for the detection of Chimeric Antigen Receptors
- Author
-
Varun Sikri, Naman Sharma, Kenta Ito, Songjie Gong, Alberto Jeronimo, Venkatesh Natarajan, Sunju Choi, Magdalena Falat, Saurabh Deepak Chitnis, Nell Narasappa, Lalith Namburu, Queenie Qiu, Ankita Batra, Pooja Smruthi Keerthipati, Jason Braun, Hannalei Mae Zamora, Ramakrishnan Gopalakrishnan, Vishan Chaudhary, Hittu Matta, Michael Kahn, Rekha Prakash, Tomas Meza Stieben, Fatima Patel, Ruben Prins, Arta Zenunovic, Preet M. Chaudhary, Supriya Peshin, Allen Membreno, Katelyn Purvis, Dan Wang, Jyotirmayee Lenka, Andrei A. Kochegarov, and Jae Han Lee
- Subjects
0301 basic medicine ,Recombinant Fusion Proteins ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,lcsh:Medicine ,Immunotherapy, Adoptive ,Epitope ,Article ,Flow cytometry ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Protein purification ,medicine ,Humans ,Luciferase ,Lymphocytes ,lcsh:Science ,Luciferases ,Multidisciplinary ,Receptors, Chimeric Antigen ,medicine.diagnostic_test ,biology ,Chemistry ,lcsh:R ,Flow Cytometry ,Fusion protein ,Chimeric antigen receptor ,Receptors, Antigen ,030104 developmental biology ,Biochemistry ,biology.protein ,lcsh:Q ,Antibody ,030217 neurology & neurosurgery - Abstract
Chimeric Antigen Receptor-T (CAR-T) cell immunotherapy has produced dramatic responses in hematologic malignancies. One of the challenges in the field is the lack of a simple assay for the detection of CARs on the surface of immune effector cells. In this study, we describe a novel luciferase-based assay, termed Topanga Assay, for the detection of CAR expression. The assay utilizes a recombinant fusion protein, called Topanga reagent, generated by joining the extra-cellular domain of a CAR-target in frame with one of the marine luciferases or their engineered derivatives. The assay involves incubation of CAR expressing cells with the Topanga reagent, a few washes and measurement of luminescence. The assay can detect CARs comprising either immunoglobulin- or non-immunoglobulin-based antigen binding domains. We further demonstrate that addition of epitope tags to the Topanga reagent not only allows its convenient one step purification but also extends its use for detection of CAR cells using flow cytometry. However, crude supernatant containing the secreted Topanga reagent can be directly used in both luminescence and flow-cytometry based assays without prior protein purification. Our results demonstrate that the Topanga assay is a highly sensitive, specific, convenient, economical and versatile assay for the detection of CARs.
- Published
- 2019
7. Abstract A10: The dual A2aR/A2bR antagonist AB928 reverses adenosine-mediated immune suppression and inhibits tumor growth in vivo
- Author
-
Manmohan Reddy Leleti, Joanne B.L. Tan, Devika Ashok, Jay P. Powers, Steve Young, Daniel DiRenzo, Matthew J. Walters, Park Adam, Dana Piovesan, Nell Narasappa, Lisa Seitz, and Jenna L. Jeffrey
- Subjects
Cancer Research ,Tumor microenvironment ,Chemistry ,medicine.medical_treatment ,Immunology ,Antagonist ,Stimulation ,Immunotherapy ,Adenosine ,Adenosine receptor ,In vivo ,medicine ,Cancer research ,Phosphorylation ,medicine.drug - Abstract
Introduction: High levels of immunosuppressive adenosine are found in the tumor microenvironment, reaching 50-100 μM in experimental models. Adenosine exerts its effects on immune cells primarily through the adenosine receptors A2aR/A2bR, which increase intracellular levels of cyclic AMP, leading to CREB phosphorylation (pCREB). We have previously shown that the dual A2aR/A2bR antagonist AB928 is capable of inhibiting adenosine-induced pCREB in healthy human volunteer (HV) blood lymphocytes. AB928 has also been shown to relieve adenosine-mediated T-cell suppression in vitro and exhibit combinatorial effects with standard-of-care chemotherapeutics in mouse syngeneic tumor models. Herein, we show that AB928 is capable of inhibiting NECA-induced gene expression changes and CREB phosphorylation in non-small cell lung carcinoma (NSCLC) patient whole blood (WB). Additionally, observations from our in vitro human studies showing the combinatorial effect of AB928 and α-PD-1 were reproduced in B16F10 syngeneic tumors. Methods: Human WB was stimulated with 5 μM of NECA and flow cytometry was used to quantify AB928-mediated inhibition of pCREB and CD3ζ phosphorylation. B16F10 tumors were treated with α-PD-1 +/- AB928 and gene expression was determined from excised mouse tumors using the nCounter PanCancer panel. Results: To ensure AB928 can successfully inhibit the high levels of intratumoral adenosine, we found that 5 μM NECA provides maximal stimulation and is significantly more potent (>20 fold) than adenosine in the pCREB assay. Additional experiments demonstrated that AB928 has comparable potency in NECA-stimulated WB from both HV and NSCLC patients. In addition to blocking downstream signaling, NanoString analysis showed that AB928 could prevent NECA-stimulated gene expression changes in NSCLC WB. We also found that NECA stimulation, alone or in combination with PD-1 inhibition, significantly reduced proximal TCR signaling, leading to reduced levels of CD3ζ phosphorylation at TYR142 (pTYR142). These reduced pTYR142 levels, with and without α-PD-1, could be significantly rescued by AB928, suggesting that blocking adenosine immunosuppression may provide additional benefit to PD-1 inhibition in tumors. Consistent with these results, AB928 was capable of suppressing growth (volume in mm3) of B16-F10 tumors both as a single agent (vehicle: 462 +/- 58; AB928: 292 +/- 55; p Conclusions: Collectively, these results support a role for AB928 in relieving adenosine-mediated immunosuppression by blocking A2aR/A2bR-induced signaling events, gene expression changes, and suppressing tumor growth in vivo. Citation Format: Daniel M. DiRenzo, Nell Narasappa, Dana Piovesan, Devika Ashok, Park Adam, Jenna L. Jeffrey, Manmohan R. Leleti, Joanne B.L. Tan, Lisa Seitz, Steve W. Young, Jay P. Powers, Matthew J. Walters. The dual A2aR/A2bR antagonist AB928 reverses adenosine-mediated immune suppression and inhibits tumor growth in vivo [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 A10.
- Published
- 2020
8. Abstract 710: CD73 inhibitors (CD73i) reverse the AMP/adenosine-mediated impairment of immune effector cell activation by immune checkpoint inhibitors (ICI)
- Author
-
Tim Park, Jenna L. Jeffrey, Annette Becker, Jay P. Powers, Daniel DiRenzo, Fang-Fang Yin, Nell Narasappa, Jaroslaw Kalisiak, Joanne B. Tan, Ken Lawson, Ulrike Schindler, Kristen Zhang, and Matthew J. Walters
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Effector ,Chemistry ,Lymphocyte ,T cell ,CD28 ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Immune system ,Oncology ,TIGIT ,medicine ,Cancer research ,biology.protein ,Antibody - Abstract
INTRODUCTION: CD73 catalyzes the extracellular generation of adenosine (ADO) from adenosine monophosphate (AMP). ADO suppresses immune responses, including those of T cells, NK cells and dendritic cells through activation of A2aR and A2bR receptors. Exhausted T cells and NK cells express high levels of several immune checkpoint proteins, including PD-1 and TIGIT. We present here preclinical data on the ability of CD73i to reverse effector cell suppression from exposure to ADO even in the presence of ICI. METHODS: CD73i effects in a monotherapeutic setting were assessed by CD3/CD28/CD2 T cell stimulation and cytolytic assays. Combinatorial settings were assessed using mixed lymphocyte reactions (MLRs). In vivo effects of CD73i + ICI were determined using syngeneic tumor models. RESULTS: CD73 is expressed across a wide range of tumor types, including those with limited response to anti-PD-1 therapy. CD73i completely rescued AMP-mediated inhibition of T cell proliferation and effector function as well as NK cell cytolytic function. AMP abrogated the enhanced allogeneic CD4+ T cell activation and IFN-γ production mediated by blocking PD-1/PD-L1 and TIGIT, an effect that was reversed by CD73i. Mechanistically, addition of AMP in MLRs repressed expression of activation markers and immune checkpoint proteins. Thus, activation of the adenosinergic pathway may limit the efficacy of ICI. TCGA data from anti-PD-1-treated melanoma patients identified CD73 expression as a negative prognostic factor. Finally, co-administration of a CD73i with an anti-PD-1 mAb resulted in significant reduction of tumor volume associated with increases in immune cell infiltration. CONCLUSIONS: CD73 inhibition, alone or in combination with anti-PD-1 and anti-TIGIT antibodies, translates into potent enhancement of immune cell activation in a variety of studies. These data provide a rationale for CD73i + ICI combinations. Citation Format: Annette Becker, Nell Narasappa, Fangfang Yin, Kristen Zhang, Daniel DiRenzo, Timothy Park, Jaroslaw Kalisiak, Ken Lawson, Jenna Jeffrey, Jay P. Powers, Ulrike Schindler, Matthew J. Walters, Joanne B. Tan. CD73 inhibitors (CD73i) reverse the AMP/adenosine-mediated impairment of immune effector cell activation by immune checkpoint inhibitors (ICI) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 710.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.