85 results on '"Jay P. Powers"'
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. Discovery of Potent and Selective Methylenephosphonic Acid CD73 Inhibitors
- Author
-
Dillon H. Miles, Nigel Walker, Manmohan Reddy Leleti, Xiaoning Zhao, Eric T. Newcomb, Kenneth V. Lawson, Jaroslaw Kalisiak, Erick Allen Lindsey, Lixia Jin, Ada Chen, Laurent Debien, Guifen Xu, Ehesan U. Sharif, Norbert Sträter, Brandon Reid Rosen, Stephen W Young, Emma Scaletti, and Jay P. Powers
- Subjects
Adenosine ,Phosphorous Acids ,Drug Evaluation, Preclinical ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,GPI-Linked Proteins ,01 natural sciences ,Structure-Activity Relationship ,03 medical and health sciences ,Immune system ,ATP hydrolysis ,Drug Discovery ,medicine ,Extracellular ,Humans ,Ectonucleotidase ,5'-Nucleotidase ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Tumor microenvironment ,Binding Sites ,Adenosine receptor ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Enzyme ,chemistry ,Biochemistry ,Drug Design ,Molecular Medicine ,medicine.drug - Abstract
Solid tumors are often associated with high levels of extracellular ATP. Ectonucleotidases catalyze the sequential hydrolysis of ATP to adenosine, which potently suppresses T-cell and NK-cell functions via the adenosine receptors (A2a and A2b). The ectonucleotidase CD73 catalyzes the conversion of AMP to adenosine. Thus, increased CD73 enzymatic activity in the tumor microenvironment is a potential mechanism for tumor immune evasion and has been associated with poor prognosis in the clinic. CD73 inhibition is anticipated to restore immune function by skirting this major mechanism of adenosine generation. We have developed a series of potent and selective methylenephosphonic acid CD73 inhibitors via a structure-based design. Key binding interactions of the known inhibitor adenosine-5'-(α,β-methylene)diphosphate (AMPCP) with hCD73 provided the foundation for our early designs. The structure-activity relationship study guided by this structure-based design led to the discovery of 4a, which exhibits excellent potency against CD73, exquisite selectivity against related ectonucleotidases, and a favorable pharmacokinetic profile.
- Published
- 2021
7. Development of a Scalable Method for Manufacturing the Central Core of CD73 Inhibitor AB680
- Author
-
Manmohan Reddy Leleti, Jeremy Fournier, Steven D. Jacob, Eric F. Connor, Grange Rebecca Louise, Kenneth V. Lawson, Anh Tran, Jaroslaw Kalisiak, Jay P. Powers, and Xuelei Yan
- Subjects
Computer science ,Pancreatic cancer ,Organic Chemistry ,Core (graph theory) ,medicine ,Computational biology ,Physical and Theoretical Chemistry ,medicine.disease ,Small molecule - Abstract
AB680 is a highly potent CD73 small molecule inhibitor discovered and developed by Arcus Biosciences, currently in clinical trials for the treatment of pancreatic cancer. Here, we report the develo...
- Published
- 2020
8. Discovery of Potent and Selective PI3Kγ Inhibitors
- 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, Pei-Yu Chen, Jenna L. Jeffrey, Jesus Banuelos, Xuelei Yan, Samuel L Drew, Nigel Walker, Puja Dhanota, Lixia Jin, Jay P. Powers, Elaine Ginn, Guillaume Mata, Kent Wong, Jeremy Fournier, Joel W. Beatty, Rhiannon Thomas-Tran, Ulrike Schindler, Amber Pham, Matthew J. Walters, Jie Chen, Cesar Meleza, and Xiaoning Zhao
- Subjects
Gene isoform ,Stereochemistry ,Crystallography, X-Ray ,01 natural sciences ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,Animals ,Class Ib Phosphatidylinositol 3-Kinase ,Humans ,Binding site ,Phosphoinositide-3 Kinase Inhibitors ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Trifluoromethyl ,Bicyclic molecule ,Lipid signaling ,Rats ,0104 chemical sciences ,Molecular Docking Simulation ,010404 medicinal & biomolecular chemistry ,Enzyme ,chemistry ,Drug Design ,Molecular Medicine ,Selectivity ,Adenosine triphosphate - Abstract
The selective inhibition of the lipid signaling enzyme PI3Kγ constitutes an opportunity to mediate immunosuppression and inflammation within the tumor microenvironment but is difficult to achieve due to the high sequence homology across the class I PI3K isoforms. Here, we describe the design of a novel series of potent PI3Kγ inhibitors that attain high isoform selectivity through the divergent projection of substituents into both the "selectivity" and "alkyl-induced" pockets within the adenosine triphosphate (ATP) binding site of PI3Kγ. These efforts have culminated in the discovery of 5-[2-amino-3-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-5-yl]-2-[(1S)-1-cyclopropylethyl]-7-(trifluoromethyl)-2,3-dihydro-1H-isoindol-1-one (4, IC50 = 0.064 μM, THP-1 cells), which displays >600-fold selectivity for PI3Kγ over the other class I isoforms and is a promising step toward the identification of a clinical development candidate. The structure-activity relationships identified throughout this campaign demonstrate that greater γ-selectivity can be achieved by inhibitors that occupy an "alkyl-induced" pocket and possess bicyclic hinge-binding motifs capable of forming more than one hydrogen bond to the hinge region of PI3Kγ.
- Published
- 2020
9. Discovery of AB680: A Potent and Selective Inhibitor of CD73
- Author
-
Brandon Reid Rosen, Manmohan Reddy Leleti, Ada Chen, Sharon Zhao, Jenna L. Jeffrey, Norbert Sträter, Eric T. Newcomb, Jay P. Powers, Kenneth V. Lawson, Lijuan Fu, Dillon H. Miles, Uli Schindler, Wade Berry, Stephen W Young, Tim Park, Emma Scaletti, Lixia Jin, Joanne B.L. Tan, Ehesan U. Sharif, Laurent Debien, Erick Allen Lindsey, Jaroslaw Kalisiak, Susanne Moschütz, Matthew J. Walters, Guifen Xu, and Nigel Walker
- Subjects
Models, Molecular ,T cell ,CD8-Positive T-Lymphocytes ,Pharmacology ,GPI-Linked Proteins ,01 natural sciences ,Small Molecule Libraries ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,Immune system ,Pharmacokinetics ,Drug Discovery ,medicine ,Extracellular ,Animals ,Humans ,Structure–activity relationship ,5'-Nucleotidase ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Binding Sites ,Chemistry ,Catabolism ,Haplorhini ,Adenosine ,Rats ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,medicine.anatomical_structure ,Leukocytes, Mononuclear ,Molecular Medicine ,Protein Binding ,medicine.drug - Abstract
Extracellular adenosine (ADO), present in high concentrations in the tumor microenvironment (TME), suppresses immune function via inhibition of T cell and NK cell activation. Intratumoral generation of ADO depends on the sequential catabolism of ATP by two ecto-nucleotidases, CD39 (ATP → AMP) and CD73 (AMP → ADO). Inhibition of CD73 eliminates a major pathway of ADO production in the TME and can reverse ADO-mediated immune suppression. Extensive interrogation of structure-activity relationships (SARs), structure-based drug design, and optimization of pharmacokinetic properties culminated in the discovery of AB680, a highly potent (Ki = 5 pM), reversible, and selective inhibitor of CD73. AB680 is further characterized by very low clearance and long half-lives across preclinical species, resulting in a PK profile suitable for long-acting parenteral administration. AB680 is currently being evaluated in phase 1 clinical trials. Initial data show AB680 is well tolerated and exhibits a pharmacokinetic profile suitable for biweekly (Q2W) iv-administration in human.
- Published
- 2020
10. Development of a Scalable and Practical Synthesis of AB928, a Dual A2a/A2b Receptor Antagonist
- Author
-
Jenna L. Jeffrey, Laurent Debien, Dillon H. Miles, Manmohan Reddy Leleti, Brandon Reid Rosen, Jay P. Powers, and Ehesan U. Sharif
- Subjects
Chemistry ,medicine.drug_class ,Organic Chemistry ,Convergent synthesis ,medicine ,Antagonist ,Physical and Theoretical Chemistry ,DUAL (cognitive architecture) ,Pharmacology ,Receptor ,Receptor antagonist - Abstract
AB928 is a potent and selective dual antagonist of the A2a and A2b receptors, which is currently in clinical trials. Here, we report the development of two scalable and practical syntheses of AB928...
- Published
- 2020
11. Discovery of Potent and Selective Non-Nucleotide Small Molecule Inhibitors of CD73
- Author
-
Stephen W Young, Debashis Mandal, Sharon Zhao, Susanne Moschütz, Ada Chen, Elaine Ginn, Lixia Jin, Samuel L Drew, Nigel Walker, Erick Allen Lindsey, Jay P. Powers, Kenneth V. Lawson, Xuelei Yan, Anh Tran, Rhiannon Thomas-Tran, Amber Pham, Manmohan Reddy Leleti, Jenna L. Jeffrey, Steven D. Jacob, Norbert Sträter, Joel W. Beatty, Jeremy Fournier, and Laurent Debien
- Subjects
Membrane permeability ,Stereochemistry ,CHO Cells ,Crystallography, X-Ray ,GPI-Linked Proteins ,Binding, Competitive ,01 natural sciences ,03 medical and health sciences ,chemistry.chemical_compound ,Cricetulus ,Cricetinae ,Drug Discovery ,medicine ,Animals ,Humans ,Nucleotide ,5'-Nucleotidase ,Cells, Cultured ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Chemistry ,Triazoles ,Purinergic signalling ,Adenosine ,Small molecule ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Benzonitrile ,Enzyme ,Hepatocytes ,Molecular Medicine ,Nucleoside ,medicine.drug - Abstract
CD73 is an extracellular mediator of purinergic signaling. When upregulated in the tumor microenvironment, CD73 has been implicated in the inhibition of immune function through overproduction of adenosine. Traditional efforts to inhibit CD73 have involved antibody therapy or the development of small molecules, the most potent of which mimic the acidic and ionizable structure of the enzyme's natural substrate, adenosine 5'-monophosphate (AMP). Here, we report the systematic discovery of a novel class of non-nucleotide CD73 inhibitors that are more potent than all other nonphosphonate inhibitor classes reported to date. These efforts have culminated in the discovery of 4-({5-[4-fluoro-1-(2H-indazol-6-yl)-1H-1,2,3-benzotriazol-6-yl]-1H-pyrazol-1-yl}methyl)benzonitrile (73, IC50 = 12 nM) and 4-({5-[4-chloro-1-(2H-indazol-6-yl)-1H-1,2,3-benzotriazol-6-yl]-1H-pyrazol-1-yl}methyl)benzonitrile (74, IC50 = 19 nM). Cocrystallization of 74 with human CD73 demonstrates a competitive binding mode. These compounds show promise for the improvement of drug-like character via the attenuation of the acidity and low membrane permeability inherent to known nucleoside inhibitors of CD73.
- Published
- 2020
12. 583 Novel, potent, and selective inhibitors of hypoxia-inducible factor (HIF)-2α reverse pro-tumorigenic transcriptional programming in cancer, stromal, and immune cells
- Author
-
Jean Chan, Lixia Jin, Kelsey Sivick Gauthier, Stephen W Young, Matthew J. Walters, Balint Gal, Anh Tran, Kenneth V. Lawson, Elaine Ginn, Manmohan Reddy Leleti, Jennie Au, Soonweng Cho, Dana Piovesan, Xiaoning Zhao, Akshata Udyavar, Jay P. Powers, Ada Chen, Cesar Meleza, Brandon Reid Rosen, and Samuel L Drew
- Subjects
Tumor microenvironment ,Stromal cell ,Hypoxia-inducible factors ,Angiogenesis ,Chemistry ,Cancer cell ,Cancer research ,Gene silencing ,Cytokine secretion ,Gene signature ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 - Abstract
Background The microenvironment of solid tumors is hypoxic and requires induction of genes associated with metabolism, growth, proliferation, and angiogenesis for cancer cells to survive and metastasize. The master transcriptional regulators of hypoxia-induced genes are the HIF proteins, consisting of three distinct oxygen-regulated α monomers (HIF-1α, -2α, and -3α). In normoxia, hydroxylation of HIF-2α allows for recognition by the pVHL E3-ubiquitin ligase complex and proteasomal degradation. Exposure to hypoxia, or VHL mutation or silencing, leads to HIF-2α stabilization, dimerization with HIF-1β/ARNT, and transcription of pro-tumorigenic gene sets in a variety of cancer and non-cancer cell types in the tumor microenvironment. In patients, overexpression of HIF is associated with poor prognosis, and both preclinical and clinical evidence suggests that inhibiting HIF-2α is an effective strategy to mitigate tumor growth, particularly in clear cell renal cell carcinoma (ccRCC), warranting further development of HIF-2α inhibitors and investigation into the role of HIF-2α in various cellular and combinatorial settings. Methods Using a suite of assays to evaluate HIF-2α-specific effects, herein we describe pharmacological properties associated with novel, potent, and selective small-molecule inhibitors of HIF-2α. Results Optimized compounds inhibited HIF reporter transcription and VEGF secretion. Compounds that were biochemically confirmed to bind HIF-2α also inhibited HIF-2α-, but not HIF-1α-, mediated gene expression. Characterization of HIF-2α inhibition was expanded to human stromal and immune cell subsets. While compounds inhibited pro-angiogenic gene sets in endothelial cells, inhibiting HIF-2α in activated hypoxic T cells did not affect proliferation or cytokine secretion, suggesting that HIF-2α inhibitors would not impede T cell functionality in tumors. In contrast, in a M2-polarized macrophage model for suppressive tumor-associated macrophages, HIF-2α drove hypoxia-induced changes in the chemokine secretome that favored granulocytic rather than lymphocytic infiltration, an effect that was effectively reversed by HIF-2α inhibition. At the transcriptional level, mRNA-sequencing was used to define global gene sets impacted by HIF-2α inhibition in M2 macrophages. Additionally, in a set of liver, kidney, pancreatic, and colon cancer lines, CRISPR/Cas9-mediated gene editing was used to differentiate the transcriptomic profile driven by HIF-2α from that of HIF-1α or HIF-3α, allowing for the derivation of a HIF-2α-specific gene signature. Cancer cell and macrophage-derived signatures were applied to publicly available datasets to investigate cancer types, other than ccRCC, in which HIF-2α may play an important pathological role. Conclusions Collectively, these data support the development of our novel and selective HIF-2α inhibitors for the treatment of cancer and expand the indications that may benefit beyond ccRCC.
- Published
- 2020
13. Targeting Metabolism of Extracellular Nucleotides via Inhibition of Ectonucleotidases CD73 and CD39
- Author
-
Kenneth V. Lawson, Jenna L. Jeffrey, and Jay P. Powers
- Subjects
Drug ,media_common.quotation_subject ,Antineoplastic Agents ,GPI-Linked Proteins ,01 natural sciences ,Protein Structure, Secondary ,03 medical and health sciences ,Drug Delivery Systems ,Drug Discovery ,medicine ,Extracellular ,Humans ,Ectonucleotidase ,Nucleotide ,Enzyme Inhibitors ,5'-Nucleotidase ,030304 developmental biology ,media_common ,chemistry.chemical_classification ,0303 health sciences ,Tumor microenvironment ,Nucleotides ,Apyrase ,Metabolism ,Adenosine ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Enzyme ,chemistry ,Cancer research ,Molecular Medicine ,medicine.drug - Abstract
In the tumor microenvironment, unusually high concentrations of extracellular adenosine promote tumor proliferation through various immunosuppressive mechanisms. Blocking adenosine production by inhibiting nucleotide-metabolizing enzymes, such as ectonucleotidases CD73 and CD39, represents a promising therapeutic strategy that may synergize with other immuno-oncology mechanisms and chemotherapies. Emerging small-molecule ectonucleotidase inhibitors have recently entered clinical trials. This Perspective will outline challenges, strategies, and recent advancements in targeting this class with small-molecule inhibitors, including AB680, the first small-molecule CD73 inhibitor to enter clinical development. Specific case studies, including structure-based drug design and lead optimization, will be outlined. Preclinical data on these molecules and their ability to enhance antitumor immunity will be discussed.
- Published
- 2020
14. Abstract P253: Potent and selective AXL tyrosine kinase inhibition demonstrates significant anti-tumor efficacy in combination with standard of care therapeutics in preclinical models
- Author
-
Susan L. Paprcka, Subhasree Sridhar, Irene M. Luu, Salema Jafri, Dillon H. Miles, Suan Liu, Ruben Flores, Shiwei Qu, Manjunath Lamani, Sriivas Paladugu, Cesar Meleza, James Wu, Hema Singh, Yu Chen, Sean Cho, Akshata Udyavar, Angelo Kaplan, Enzo Stargnaro, Xiaoning Zhao, Lixia Jin, Manmohan R. Leleti, Stephen W. Young, Jay P. Powers, Matthew J. Walters, and Ester Fernandez-Salas
- Subjects
Cancer Research ,Oncology - Abstract
Background. AXL receptor tyrosine kinase (AXL) is a transmembrane protein that is overexpressed in a variety of tumors and correlates with poor prognosis in cancer patients. AXL is expressed in cancer and stromal cells and has been implicated in the development of resistance to chemotherapy, targeted therapies & immunotherapies. Activation of AXL by its ligand, growth arrest specific protein 6 (Gas6), or ligand-independent dimerization facilitates AXL phosphorylation, initiates signaling cascades that promote cancer cell proliferation, survival, and an immunosuppressive microenvironment. Here we present the discovery and characterization of a novel, highly potent and selective novel AXL inhibitor. Materials and Methods. The potency and specificity of the novel Arcus inhibitor against AXL and other kinases was determined using a panel of HTRF KinEASE-TK assays. Intracellular target engagement was evaluated by monitoring displacement of a competitive fluorescent tracer using an AXL NanoBRETTM intracellular kinase assay. To further assess the inhibitory effect of the novel Arcus inhibitor on AXL kinase activity, AXL autophosphorylation induced SH2 domain translocation was measured in cells using a PathHunterÒ U2OS AXL Functional assay system. Inhibition of AXL phosphorylation in cancer cells was evaluated by Western blot and levels of soluble and surface AXL were assessed by ELISA and flow cytometry, respectively. Pharmacokinetics (PK), pharmacodynamics (PD) and anti-tumor efficacy were evaluated in murine models. Results. A novel, potent, reversible, and highly selective AXL kinase inhibitor has been generated by Arcus. The novel inhibitor exhibits single-digit nanomolar potency in both biochemical and cell-based assays, retains significant activity in 100% human serum and does not show significant inhibition of the major CYP450 isoforms nor the hERG potassium channel. The novel Arcus molecule inhibits AXL phosphorylation mediated by both ligand-dependent Gas6 stimulation as well as ligand-independent autophosphorylation. AXL phosphorylation and subsequent signaling leads to receptor internalization, thereby decreasing both surface AXL expression and soluble AXL levels. AXL activity is inhibited in a concentration dependent manner significantly increasing both surface AXL expression and soluble AXL levels. More importantly, significant anti-tumor efficacy is observed in combination with targeted therapies in several in vivo models. Furthermore, AXL inhibition significantly reduces tumor growth after relapse to single-agent targeted therapy. Conclusions. A novel selective inhibitor of AXL tyrosine kinase activity has been developed that demonstrates single-digit nanomolar potency and inhibition of both ligand-dependent and ligand-independent AXL phosphorylation. Significant anti-tumor activity is observed in combination with targeted therapy and upon acquired resistance in xenograft models. Selective AXL inhibition is a promising therapeutic strategy to overcome resistance to chemotherapy, targeted therapy, and/or immunotherapy. Citation Format: Susan L. Paprcka, Subhasree Sridhar, Irene M. Luu, Salema Jafri, Dillon H. Miles, Suan Liu, Ruben Flores, Shiwei Qu, Manjunath Lamani, Sriivas Paladugu, Cesar Meleza, James Wu, Hema Singh, Yu Chen, Sean Cho, Akshata Udyavar, Angelo Kaplan, Enzo Stargnaro, Xiaoning Zhao, Lixia Jin, Manmohan R. Leleti, Stephen W. Young, Jay P. Powers, Matthew J. Walters, Ester Fernandez-Salas. Potent and selective AXL tyrosine kinase inhibition demonstrates significant anti-tumor efficacy in combination with standard of care therapeutics in preclinical models [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr P253.
- Published
- 2021
15. Abstract P206: AB521 potently and selectively inhibits pro-tumorigenic gene transcription by Hypoxia-Inducible Factor (HIF)-2α in vitro and in vivo
- Author
-
Kelsey E. Sivick Gauthier, Dana Piovesan, Soonweng Cho, Kenneth V. Lawson, Patrick G. Schweickert, Alejandra Lopez, Suan Liu, Timothy Park, Artur Mailyan, Jeremy T. A. Fournier, Joel W. Beatty, Samuel L. Drew, Jarek Kalisiak, Balint Gal, Guillaume Mata, Zhang Wang, Brandon R. Rosen, Clayton Hardman, Matthaw P. Epplin, Kai Yu, Karl T. Haelsig, Lixia Jin, Elaine Ginn, Jennie Au, Cesar A. Meleza, Joel Tencer, Amber Pham, Hyock J. Kwon, Stephen W. Young, Manmohan Leleti, Jay P. Powers, and Matthew J. Walters
- Subjects
Cancer Research ,Oncology - Abstract
Cells in the solid tumor microenvironment are frequently exposed to hypoxic conditions, necessitating molecular adaptations for survival. Of particular importance are transcriptional changes mediated by heterodimeric Hypoxia-Inducible Factor (HIF) proteins that consist of an oxygen-regulated α monomer (either HIF-1α, -2α, and -3α) coupled to a constitutively expressed β monomer (HIF-1β/ARNT). In normal oxygen conditions, HIF-2α is degraded following ubiquitination by the von Hippel-Lindau (pVHL) E3-ubiquitin ligase complex. Exposure to hypoxia, VHL mutation, or epigenetic silencing of pVHL leads to HIF-2α stabilization and transcription of pro-tumorigenic gene sets in both cancer and non-cancer cells. Inhibition of HIF-2α has been shown clinically to be an effective strategy to mitigate tumor growth, particularly in patients suffering from VHL disease or clear cell renal cell carcinoma (ccRCC), a cancer that has a particularly high prevalence of pVHL dysfunction. Applying a pharmacophore mapping and structure-based design approach, we identified a novel and potent small molecule HIF-2α inhibitor, AB521. AB521 avidly binds the HIF-2α PAS-B domain, preventing HIF-2α-mediated gene transcription. AB521 is characterized by a favorable preclinical pharmacokinetic profile and is projected to be suitable for once-daily dosing in humans. When delivered orally in mice, AB521 significantly regressed established 786-O xenograft tumors and decreased pharmacodynamic markers associated with HIF-2α in a dose-dependent manner. In vitro, AB521 potently inhibited HIF-2α-specific luciferase reporter transcription under high-serum conditions, VEGF protein secretion, colony formation in soft agar, and did not exhibit off-target cytotoxicity in 786-O cells. AB521 selectively inhibited HIF-2α-, but not HIF-1α-, mediated gene expression in hypoxic Hep3B hepatocellular carcinoma cells. AB521 also inhibited the transcriptional activity of endogenous HIF-2α in relevant human primary cell types, including endothelial cells and pro-tumorigenic M2-polarized macrophages. Importantly, inhibiting HIF-2α did not impact functionality of activated hypoxic human T cells, suggesting that AB521 would be favorable combination partner for I-O therapeutic agents. Indeed, expression of CD73, the primary enzyme responsible for synthesis of the immunosuppressive metabolite adenosine, was highly correlated with hypoxic signatures across several indications in publicly available bioinformatic datasets, suggesting combinations with adenosine pathway antagonists in ccRCC and beyond. In summary, AB521 is a novel and selective HIF-2α inhibitor with potent anti-tumor activity. Clinical evaluation of this molecule is expected to begin in the latter part of 2021. Citation Format: Kelsey E. Sivick Gauthier, Dana Piovesan, Soonweng Cho, Kenneth V. Lawson, Patrick G. Schweickert, Alejandra Lopez, Suan Liu, Timothy Park, Artur Mailyan, Jeremy T. A. Fournier, Joel W. Beatty, Samuel L. Drew, Jarek Kalisiak, Balint Gal, Guillaume Mata, Zhang Wang, Brandon R. Rosen, Clayton Hardman, Matthaw P. Epplin, Kai Yu, Karl T. Haelsig, Lixia Jin, Elaine Ginn, Jennie Au, Cesar A. Meleza, Joel Tencer, Amber Pham, Hyock J. Kwon, Stephen W. Young, Manmohan Leleti, Jay P. Powers, Matthew J. Walters. AB521 potently and selectively inhibits pro-tumorigenic gene transcription by Hypoxia-Inducible Factor (HIF)-2α in vitro and in vivo [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr P206.
- Published
- 2021
16. Abstract 1206: Discovery and characterization of AB521, a novel, potent, and selective hypoxia-inducible factor (HIF)-2α inhibitor
- Author
-
Stephen W. Young, Guillaume Mata, Joel W. Beatty, Matthew J. Walters, Kelsey Sivick Gauthier, Jeremy Fournier, Jennifer Au, Jay P. Powers, Dana Piovesan, Artur K. Mailyan, Kenneth V. Lawson, Manmohan Reddy Leleti, Elaine Ginn, Jarek Kalisiak, Xuelei Yan, Balint Gal, Zhang Wang, Lixia Jin, Samuel L Drew, and Cesar Meleza
- Subjects
Cancer Research ,Oncology ,Hypoxia-inducible factors ,Chemistry ,Cell biology - Abstract
A hypoxic environment is a common characteristic of solid tumors. Cancer cells adapt to hypoxia and become more aggressive by up-regulation of genes associated with metabolism, growth, proliferation, angiogenesis, and erythropoiesis. Hypoxia-inducible factors (HIFs) are the central driving force for the cellular response to hypoxia and regulate a vast array of these genes. HIFs are heterodimers composed of an oxygen-sensitive HIF-α subunit (HIF-1α, HIF-2α, and HIF-3α) and a constitutively expressed HIF-1β subunit. HIF-2α is constitutively synthesized and regulated in an oxygen-dependent manner. Proline residues present in the oxygen-dependent degradation domain of the HIF-2α subunit are hydroxylated and subject to ubiquitination via the von Hippel-Lindau (pVHL) E3 ligase complex for subsequent proteasomal degradation. Under hypoxia or pseudohypoxia, caused by loss of VHL function, this process is inhibited, allowing HIF-2α translocation to the nucleus where, in complex with HIF-1β/ARNT, it promotes transcription of various pro-tumorigenic gene sets. Inhibition of HIF-2α has been demonstrated to be an effective strategy to mitigate tumor growth in the clinic, particularly in clear cell renal cell carcinoma (ccRCC). Applying a pharmacophore mapping and structure-based design approach, we identified multiple novel series of small molecule HIF-2α inhibitors which avidly bind the HIF-2α PAS-B domain and disrupt dimerization with HIF-1β, preventing HIF-2α-mediated gene transcription. Interrogation of structure activity relationships and pharmacokinetic trends yielded highly optimized inhibitors, including AB521, which potently inhibits HIF-2α reporter transcription and VEGF secretion in a human ccRCC line. AB521 was confirmed to bind HIF-2α via multiple biochemical assays and inhibited HIF-2α-, but not HIF-1α-, mediated gene expression in a hepatocellular carcinoma cell line. Furthermore, AB521 is characterized by a favorable preclinical pharmacokinetic profile with high stability to human hepatocytes and no significant direct or time-dependent inhibition of the major CYP450 drug metabolizing enzymes. AB521 exhibits high bioavailability across preclinical species and is projected to possess a human pharmacokinetic profile suitable for once-daily dosing. In conclusion, we have discovered and extensively characterized a series of novel HIF-2α inhibitors, exemplified by AB521, which potently and selectively inhibits HIF-2α and possesses a favorable preclinical pharmacokinetic profile. The data described herein provides further support for the development of novel HIF-2α inhibitors for cancer therapy. Citation Format: Kenneth V. Lawson, Kelsey E. Sivick Gauthier, Artur K. Mailyan, Jeremy T. Fournier, Joel W. Beatty, Samuel L. Drew, Jarek Kalisiak, Balint Gal, Guillaume Mata, Zhang Wang, Xuelei Yan, Lixia Jin, Elaine Ginn, Dana Piovesan, Jennifer Au, Cesar A. Meleza, Stephen W. Young, Matthew J. Walters, Manmohan Leleti, Jay P. Powers. Discovery and characterization of AB521, a novel, potent, and selective hypoxia-inducible factor (HIF)-2α inhibitor [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1206.
- Published
- 2021
17. Discovery and characterization of potent and selective AXL receptor tyrosine kinase inhibitors for cancer therapy
- Author
-
D. Miles, C.N. Foley, Jay P. Powers, Matthew J. Walters, R. Grange, Manmohan Reddy Leleti, Y. Chen, X. Zhao, Lixia Jin, Steve Young, and S.L. Paprcka
- Subjects
Cancer Research ,Oncology ,AXL receptor tyrosine kinase ,Chemistry ,Cancer research ,Cancer therapy - Published
- 2020
18. Discovery and characterization of novel, potent, and selective hypoxiainducible factor (HIF)-2α inhibitors
- Author
-
A. Maliyan, C. Ada, Manmohan Reddy Leleti, Cesar Meleza, Kenneth V. Lawson, Jeremy Fournier, Brandon Reid Rosen, K. Sivick Gauthier, Matthew J. Walters, Akshata Udyavar, Steve Young, Dana Piovesan, Jay P. Powers, Lixia Jin, J. Au, Joel W. Beatty, Elaine Ginn, and S. Zhao
- Subjects
Cancer Research ,Oncology ,Chemistry ,Cancer research - Published
- 2020
19. Improved synthesis of sterically encumbered heteroaromatic biaryls from aromatic β-keto esters
- Author
-
Ehesan U. Sharif, Jay P. Powers, Nicholas S. Chan, Dillon H. Miles, Manmohan Reddy Leleti, and Brandon Reid Rosen
- Subjects
Steric effects ,010405 organic chemistry ,Aryl ,Organic Chemistry ,010402 general chemistry ,Condensation reaction ,01 natural sciences ,Biochemistry ,Combinatorial chemistry ,0104 chemical sciences ,chemistry.chemical_compound ,Column chromatography ,chemistry ,Drug Discovery ,Guanidine - Abstract
A protocol for the synthesis of hindered 4-aryl 2-aminopyrimidines from β–keto esters is described. The process employs trifluoroethanol as an essential additive to promote the guanidine condensation reaction, enabling the synthesis of 25 aryl- and heteroaryl substituted aminopyrimidines in good yields and high purities with no column chromatography. The conditions described herein are readily scalable and have been employed in the large-scale synthesis of the clinical A2a/A2bR antagonist AB928.
- Published
- 2020
20. 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
21. Discovery and Characterization of a Potent and Selective Inhibitor for Human Phosphoinositide‐3‐kinase γ
- Author
-
Guillaume Mata, Manmohan Reddy Leleti, Stephen W Young, Lixia Jin, Jenna L. Jeffrey, Jeremy Fournier, Ada Chen, Matthew J. Walters, Jay P. Powers, Xiaoning Zhao, Jesus Banuelos, Cesar Meleza, and Artur K. Mailyan
- Subjects
Phosphoinositide 3-kinase ,biology ,Biochemistry ,Chemistry ,Genetics ,biology.protein ,Molecular Biology ,Biotechnology - Published
- 2020
22. Kinetic and Mechanistic Characterization of a Potent and Selective Inhibitor for Human AXL Receptor Tyrosine Kinase
- Author
-
Matthew J. Walters, Yu Chen, Manmohan Reddy Leleti, Sean Cho, Jay P. Powers, Lixia Jin, Dillon H. Miles, Xiaoning Zhao, Annette Becker, Stephen W. Young, Corinne Nicole Foley, and Akshata Udyavar
- Subjects
Biochemistry ,AXL receptor tyrosine kinase ,Chemistry ,Genetics ,Molecular Biology ,Biotechnology - Published
- 2020
23. Safety, tolerability, and pharmacology of AB928, a novel dual adenosine receptor antagonist, in a randomized, phase 1 study in healthy volunteers
- Author
-
Gerhard Arold, Joanne B.L. Tan, Manmohan Reddy Leleti, Jenna L. Jeffrey, Devika Ashok, Renger G. Tiessen, Aimee Rieger, Lisa Seitz, Matthew J. Walters, Jay P. Powers, Joyson Joseph Karakunnel, and Lixia Jin
- Subjects
0301 basic medicine ,Adult ,Male ,Adolescent ,Administration, Oral ,Pharmacology ,CD8-Positive T-Lymphocytes ,Adenosine receptor antagonist ,03 medical and health sciences ,Food-Drug Interactions ,Young Adult ,0302 clinical medicine ,Pharmacokinetics ,Double-Blind Method ,Medicine ,Humans ,Pharmacology (medical) ,Receptor ,Cyclic AMP Response Element-Binding Protein ,business.industry ,Antagonist ,Middle Aged ,Adenosine receptor ,Adenosine ,Healthy Volunteers ,030104 developmental biology ,Oncology ,Tolerability ,Purinergic P1 Receptor Antagonists ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Female ,business ,medicine.drug - Abstract
Adenosine suppresses antitumor immune responses via A2a and A2b receptors expressed on intratumoral immune cells. This effect is mediated by increased cyclic adenosine 5′-monophosphate (AMP) levels and phosphorylation of cyclic AMP response element binding protein (CREB). We conducted a phase 1, placebo-controlled, single-ascending-dose (SAD) and multiple-ascending-dose (MAD) study to assess the safety, tolerability, pharmacokinetics (PK), including food effect (FE), and pharmacodynamics (PD) of oral AB928, a novel dual A2aR/A2bR antagonist, in healthy volunteers. AB928 doses between 10 and 200 mg once daily and 100 mg twice daily were evaluated. The study enrolled 85 subjects (randomized 3:1, AB928:placebo), 40 each in the SAD and MAD cohorts, and 5 in the FE cohort. AB928 was well tolerated up to the highest dose tested and did not affect any physiologic parameters potentially sensitive to adenosine inhibition. No safety concern was identified. The PK profile of AB928 was linear and dose-proportional, and a clear PK/PD correlation was demonstrated. Significant inhibition of adenosine receptor-mediated phosphorylated CREB was observed at peak plasma concentrations in all dose cohorts and at trough plasma concentrations in the higher-dose cohorts. AB928 plasma levels ≥1 μM were associated with ≥90% adenosine receptor inhibition. In the postprandial state, the rate of AB928 absorption decreased but the extent of absorption was unchanged. Together, these data support further clinical development of oral AB928 in cancer patients.
- Published
- 2018
24. Abstract C050: A novel, potent, and selective hypoxia-inducible factor (HIF)-2α antagonist
- Author
-
Xiaoning Zhao, Manmohan Reddy Leleti, Jay P. Powers, Steve Young, Tim Park, Samuel L Drew, Elaine Ginn, Anh Tran, Dana Piovesan, Kelsey Sivick Gauthier, Nikki Kimura, Cesar Meleza, Lixia Jin, Matthew J. Walters, Kenneth V. Lawson, and Ada Chen
- Subjects
Gene isoform ,Cancer Research ,Reporter gene ,Aryl hydrocarbon receptor nuclear translocator ,Chemistry ,Angiogenesis ,Cell ,medicine.anatomical_structure ,Oncology ,Hypoxia-inducible factors ,medicine ,Cancer research ,Transcriptional regulation ,Gene silencing - Abstract
The microenvironment of solid tumors is known to be hypoxic and requires induction of genes associated with metabolism, growth, proliferation, and angiogenesis for tumor cells to survive and metastasize. The master transcriptional regulator of hypoxia-induced genes is the Hypoxia-Inducible Factor (HIF), consisting of an oxygen-regulated alpha monomer, of which there are three isoforms (HIF-1α, HIF-2α, and HIF-3α), that can heterodimerize with a constitutively-expressed beta monomer (HIF-1β/ARNT) using Per-ARNT-SIM (PAS) protein-protein interaction domains. In normoxia, proline residues present in the oxygen-dependent degradation domain of the HIF-α subunits are hydroxylated, allowing for recognition by the von Hippel-Lindau (pVHL) E3-ubiquitin ligase complex and subsequent proteasomal degradation. Upon exposure to low oxygen conditions or in the case of VHL mutation or silencing, HIF-α subunits accumulate in the cell and mediate transcription of various pro-tumorigenic gene sets. In patients, overexpression of HIF is associated with poor prognosis, and both preclinical and clinical evidence is mounting that suggests inhibiting HIF-2α is a valid approach to destroy tumor cells, particularly in clear cell renal carcinoma, warranting development of next-generation inhibitors. Using a suite of in vitro and in vivo assays designed to evaluate HIF-2α-specific effects, herein we describe pharmacological properties associated with novel, potent, and selective small-molecule antagonists of HIF-2α. These compounds inhibited HIF-dependent reporter gene transcription as well as VEGF protein secretion in a human renal cell adenocarcinoma line. Compounds that were confirmed to bind the HIF-2α PAS-B domain by Microscale thermophoresis (MST) and Thermal shift assay (TSA) also significantly inhibited HIF-2α, but not HIF-1α, target gene expression in a hepatocellular carcinoma cell line (P Citation Format: Kelsey E Sivick Gauthier, Kenneth V Lawson, Dana Piovesan, Matthew J Walters, Ada Chen, Xiaoning Zhao, Cesar Meleza, Nikki Kimura, Tim Park, Steve Young, Anh Tran, Samuel L Drew, Lixia Jin, Manmohan Leleti, Elaine Ginn, Jay P Powers. A novel, potent, and selective hypoxia-inducible factor (HIF)-2α antagonist [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr C050. doi:10.1158/1535-7163.TARG-19-C050
- Published
- 2019
25. C5a Receptor (CD88) Blockade Protects against MPO-ANCA GN
- Author
-
Mark E. T. Penfold, Ronald J. Falk, Juan C. Jaen, Lin Gan, Daniel J. Dairaghi, Bao Lu, Thomas J. Schall, Lisa Seitz, Peiqi Hu, Craig Gerard, J. Charles Jennette, Norma P. Gerard, Hong Xiao, Yu Wang, Trageen Baumgart, Jay P. Powers, and Linda S. Ertl
- Subjects
biology ,Inflammation ,General Medicine ,medicine.disease ,C5a receptor ,Complement system ,Nephrology ,Proteinase 3 ,Myeloperoxidase ,Immunology ,Alternative complement pathway ,medicine ,biology.protein ,medicine.symptom ,Receptor ,Vasculitis - Abstract
Necrotizing and crescentic GN (NCGN) with a paucity of glomerular immunoglobulin deposits is associated with ANCA. The most common ANCA target antigens are myeloperoxidase (MPO) and proteinase 3. In a manner that requires activation of the alternative complement pathway, passive transfer of antibodies to mouse MPO (anti-MPO) induces a mouse model of ANCA NCGN that closely mimics human disease. Here, we confirm the importance of C5aR/CD88 in the mediation of anti-MPO–induced NCGN and report that C6 is not required. We further demonstrate that deficiency of C5a-like receptor (C5L2) has the reverse effect of C5aR/CD88 deficiency and results in more severe disease, indicating that C5aR/CD88 engagement enhances inflammation and C5L2 engagement suppresses inflammation. Oral administration of CCX168, a small molecule antagonist of human C5aR/CD88, ameliorated anti-MPO–induced NCGN in mice expressing human C5aR/CD88. These observations suggest that blockade of C5aR/CD88 might have therapeutic benefit in patients with ANCA-associated vasculitis and GN.
- Published
- 2014
26. AB928, a dual antagonist of the A 2a R and A 2b R adenosine receptors, leads to greater immune activation and reduced tumor growth when combined with chemotherapy
- Author
-
Matthew J. Walters, F. Yin, E. Sharif, Ferdie Soriano, Dana Piovesan, J. Tan, Ulrike Schindler, D. DiRenzo, Manmohan Reddy Leleti, L.C. Seitz, Jay P. Powers, D. Miles, T. Park, Joyson Joseph Karakunnel, Steve Young, Brandon Reid Rosen, A. Rieger, Lixia Jin, and D. Ashok
- Subjects
0301 basic medicine ,Cancer Research ,Chemotherapy ,Chemistry ,medicine.medical_treatment ,Antagonist ,Adenosine receptor ,03 medical and health sciences ,030104 developmental biology ,Oncology ,medicine ,Cancer research ,Tumor growth ,Immune activation - Published
- 2018
27. Abstract 3168: Methods for assessment of the 'adenosine fingerprint' in clinical trials of AB928
- Author
-
Daniel DiRenzo, Devika Ashok, Amy E. Anderson, Akshata Udyavar, Joanne B. Tan, Irene M. Luu, Kristen Zhang, Jenna L. Jeffrey, Lisa Seitz, Manmohan R. Leleti, Stephen W. Young, Jay P. Powers, and Matthew J. Walters
- Subjects
Cancer Research ,Oncology - Abstract
BACKGROUND: The tumor microenvironment (TME) contains high levels of immunosuppressive adenosine (ADO), which activates the A2aR and A2bR receptors on immune cells, leading to an ineffective anti-tumor response. Ecto-5’-nucleotidase (CD73) and tissue non-specific alkaline phosphatase (TNAP) are primarily responsible for the conversion of extracellular adenosine monophosphate (AMP) to ADO and exhibit both membrane-bound and secreted forms. We have previously shown that AB928, a dual A2aR/A2bR antagonist, rescues the immunosuppressive effects of ADO in experimental tumor models. Herein, we describe the development of assays to measure the expression and activity of adenosine-generating enzymes in human tumor samples and peripheral blood. These assays are being used to define an “adenosine fingerprint” that identifies tumor types and patients most sensitive to adenosine inhibition by AB928. METHODS: CD73 and TNAP immuno-histochemistry (IHC) and mRNA analysis were performed on sections of formalin fixed paraffin embedded (FFPE) tumor tissue. Circulating levels of CD73 were quantified with an in-house developed ELISA, and AMP-ase enzymatic activity in serum was determined using an AMP-GloTM(Promega) assay. Gene expression data were extracted from The Cancer Genome Atlas (TCGA) and expressed as a ratio of log2 counts per million per sample. RESULTS: TCGA data identified non-small cell lung (NSCLC), renal clear cell, triple-negative breast, ovarian, colorectal, and gastro-esophageal cancers as tumors that highly express the adenosine producing enzymes CD73 or TNAP. To confirm these gene expression patterns, IHC assays for both CD73 and TNAP were developed using normal and tumor human tissue. IHC for CD73 was strongest in NSCLC (54.3 +/- 11.2 µm2) and colorectal (22.5 +/- 8.1 µm2) adenocarcinomas, whereas prostate (1.0 +/- 0.3 µm2) cancer exhibited the weakest staining. In contrast, TNAP staining was strongest in ovarian cancer and NSCLC adenocarcinoma, whereas gastric and colorectal adenocarcinomas showed very little TNAP staining. Therefore, CD73 and TNAP IHC broadly recapitulate the gene expression patterns found in TCGA. A CD73-specific ELISA was developed using human cancer patient blood which established a range of circulating CD73 protein levels (2-8 ng/mL) and showed a strong correlation between plasma and serum levels (r2 = 0.94). To measure adenosine-generating enzyme activity in peripheral blood, the AMP-Glo biochemical assay was performed and showed strong concordance with the CD73 ELISA in cancer patient serum (r2 = 0.72). CONCLUSIONS: These assays provide a detailed picture of the adenosine-generating capacity in the local TME as well as the peripheral activity and levels of CD73/TNAP to better identify patients that may benefit from adenosine inhibition. Citation Format: Daniel DiRenzo, Devika Ashok, Amy E. Anderson, Akshata Udyavar, Joanne B. Tan, Irene M. Luu, Kristen Zhang, Jenna L. Jeffrey, Lisa Seitz, Manmohan R. Leleti, Stephen W. Young, Jay P. Powers, Matthew J. Walters. Methods for assessment of the “adenosine fingerprint” in clinical trials of AB928 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3168.
- Published
- 2019
28. Abstract A162: AB928, a dual antagonist of the A2aR and A2bR adenosine receptors, relieves adenosine-mediated immune suppression
- Author
-
Dana Piovesan, Ehesan U. Sharif, Bryan Handlos, Ulrike Schindler, Manmohan Reddy Leleti, Joanne Tan, Ferdie Soriano, Matthew J. Walters, Dillon H. Miles, Tim Park, Jenna L. Jeffrey, Jay P. Powers, Brandon Reid Rosen, and Daniel DiRenzo
- Subjects
0301 basic medicine ,Adenosine monophosphate ,Cancer Research ,Chemistry ,CD14 ,Immunology ,Dendritic cell ,Adenosine ,Adenosine receptor ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,EHNA ,medicine.drug - Abstract
Introduction: Adenosine, generated through the hydrolysis of extracellular adenosine monophosphate (AMP) by the ecto-nucleotidase CD73, is an important mechanism for immunosuppression in cancer development. Adenosine’s suppressive effects on immune cells are driven primarily through 2 of the 4 adenosine receptors, A2aR and A2bR. We have previously shown that adenosine-mediated suppression of T-cells can be blocked by the dual A2aR/A2bR antagonist, AB928. Herein, we show that AB928 is capable of relieving adenosine-mediated immune suppression using human in vitro cell cultures, advanced gene expression studies, and mouse syngeneic tumor models. Methods: The ability of AB928 to inhibit adenosine-mediated suppression of dendritic cell function in vitro was assessed using human monocyte-derived dendritic cells (moDC). Briefly, moDC were generated from freshly isolated CD14+ monocytes and differentiated with IL-4/GM-CSF for 7 days +/- adenosine/EHNA +/- AB928. Cells were then taken for NanoString analysis or placed in a mixed lymphocyte reaction (MLR) with CD4+ T-cells. Mouse syngeneic tumor studies were conducted using C57BL/6 mice inoculated with mouse mammary tumor AT3-OVA or melanoma B16-F10 cells. Tumors were subsequently treated with doxorubicin, oxaliplatin, or α-PD-1 +/- AB928. Results: Quantitative immunohistochemistry and analysis of public gene expression databases identified individual human tumor types that express high levels of adenosine processing enzymes. Most notably, non-small cell lung, renal, triple-negative breast, ovarian, colorectal, and gastroesophageal cancers were found to have the most favorable expression profiles for interventions targeting the adenosine pathway. Additionally, a high degree of correlation was found between transcript and protein measurements for CD73 (r2 = 0.87), illustrating the robust and reproducible nature of these techniques. In human in vitro cell cultures, moDC differentiated in the presence of adenosine showed a decreased ability to stimulate IFN-γ secretion from allogenic CD4+ T-cells in a MLR. This suppression was significantly reversed by addition of AB928. Next, multiplexed gene expression profiling using NanoString identified a cassette of 39 genes (>2.0 fold change, p Citation Format: Daniel DiRenzo, Dana Piovesan, Joanne Tan, Dillon H. Miles, Manmohan R. Leleti, Timothy Park, Ferdie Soriano, Bryan Handlos, Jenna L. Jeffrey, Ehesan U. Sharif, Brandon R. Rosen, Ulrike Schindler, Jay P. Powers, Matthew J. Walters. AB928, a dual antagonist of the A2aR and A2bR adenosine receptors, relieves adenosine-mediated immune suppression [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A162.
- Published
- 2019
29. Abstract A157: Preclinical pharmacokinetic and pharmacodynamic characterization of AB680, a small-molecule CD73 inhibitor for cancer immunotherapy
- Author
-
Matthew J. Walters, Tim Park, Kristen Zhang, Devika Ashok, Manmohan Reddy Leleti, Amber Pham, Kenneth V. Lawson, Ada Chen, Joanne B.L. Tan, Elaine Ginn, Xiaoning Zhao, Anderson Amy Elizabeth, Jarek Kalisiak, Jesus Banuelos, Jie Chen, Jay P. Powers, and Jenna Jeffreys
- Subjects
Cancer Research ,Tumor microenvironment ,Chemistry ,medicine.medical_treatment ,Immunology ,Cancer ,Pharmacology ,medicine.disease ,Immune system ,Pharmacokinetics ,Cancer immunotherapy ,medicine ,Potency ,IC50 ,CD8 - Abstract
Introduction: Extracellular adenosine (ADO), present at high concentrations in the tumor microenvironment (TME), suppresses immune function via inhibition of T-cell, natural killer (NK) cell, and dendritic cell (DC) activation. Intratumoral generation of ADO depends on the sequential catabolism of ATP by two ecto-nucleotidases CD39 (ATP→AMP) and CD73 (AMP→ADO). Inhibition of CD73 eliminates a major pathway of ADO production in the TME and can reverse ADO-mediated immune suppression. Here we present the preclinical characterization of AB680, a novel, highly potent, reversible and selective small-molecule inhibitor of CD73, currently in preclinical development as a potential antitumor agent. The link between CD73 levels present in different tissues, efficacy in mouse tumor models, plasma and tumor exposure, and projected human pharmacokinetic (PK) profile can be combined to provide an expected AB680 dosing strategy for the upcoming first-in-human clinical trial. Methods: The potency of AB680 against human CD73 was determined using CHO-CD73 cells, blood CD8+ T-cells, recombinant human CD73, and serum/plasma using either malachite green assay, AMP-Glo assay, or LCMS/MS. The selectivity of AB680 against related ecto-nucleotidases was also assessed using similar methods. Quantitation of soluble CD73 in mouse and human serum, and mouse tumor homogenates, was performed via in-house developed and validated ELISA or Western blot methods. Syngeneic mouse tumor models were established to assess the efficacy of AB680 at multiple doses. AB680 levels in plasma and tumor associated with each dosing regimen were determined via LCMS/MS. The potency of AB680 in an intratumoral setting was determined using various biochemical methods. The effects of AB680 on syngeneic tumor volumes were assessed in prophylactic and therapeutic settings. The PK properties of AB680 were evaluated in multiple preclinical species and a projected human dosing schedule for AB680 was determined via allometric scaling. Results: AB680 is a highly potent, reversible and selective inhibitor of CD73 activity (IC50 < 0.01 nM on human CD8+ T-cells), which retains its high potency in the presence of human serum. Inhibition of AMP hydrolysis by AB680 completely reversed ADO-mediated suppression of CD4+ and CD8+ T-cell effector function, as measured by cytokine secretion and proliferation. The in vivo efficacy of AB680, as measured by its ability to restrict tumor growth at doses as low as 10 mg/kg once daily, is reflective of the following parameters: 1) plasma half-life, 2) partitioning from blood to tumor compartment, 3) potency against soluble CD73 in plasma or serum, and 4) potency against membrane-bound CD73. The PK properties of AB680 in rodent and non-rodent species are characterized by very low clearance and long half-lives. The combination of in vitro potency against mouse and human soluble and membrane-bound CD73, in vivo tumor growth regulation observed in mouse models, quantification of mouse and human CD73 levels, and the projected human PK profile for AB680 have resulted in predicted human PK properties (projected half life: 4-14 days) suitable for intravenous dosing on a schedule consistent with typical monoclonal antibody dosing cycles. Conclusions: AB680 is a highly potent and selective small-molecule inhibitor of CD73 which can mitigate AMP and ADO-mediated tumor immunosuppression by potently blocking the production of ADO. AB680 exhibits a unique projected human PK profile suitable for parental administration and is expected to enter clinical development in 2018. Citation Format: Joanne B.L. Tan, Jie Chen, Elaine Ginn, Devika Ashok, Amy E Anderson, Jesus Banuelos, Kristen Zhang, Amber Pham, Timothy Park, Ada Chen, Xiaoning Zhao, Kenneth K.V. Lawson, Jenna Jeffreys, Jarek Kalisiak, Manmohan R. Leleti, Matthew J. Walters, Jay P. Powers. Preclinical pharmacokinetic and pharmacodynamic characterization of AB680, a small-molecule CD73 inhibitor for cancer immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A157.
- Published
- 2019
30. CCR9 inhibition does not interfere with the development of immune tolerance to oral antigens
- Author
-
Timothy J. Sullivan, Jay P. Powers, Juan C. Jaen, Karen Ebsworth, Matthew J. Walters, Penglie Zhang, and Thomas J. Schall
- Subjects
Mice, Knockout ,Chemokine ,CCR9 ,biology ,Ovalbumin ,business.industry ,Immunology ,Administration, Oral ,Oral tolerance ,Immune tolerance ,Mice ,Receptors, CCR ,Chemokine receptor ,Antigen ,Immune Tolerance ,biology.protein ,Animals ,Medicine ,Immunology and Allergy ,Antigens ,business - Abstract
Recent literature indicates that mice deficient in the chemokine receptor CCR9 (CCR9 −/− mice) are unable to generate oral tolerance. The present report describes how such inability can be overcome by increasing the dose of oral antigen. Pharmacological inhibition of CCR9 did not affect the generation of oral tolerance, regardless of antigen dose. These results highlight the inadequacy of genetic deletion of CCR9 when predicting the effects of pharmacological CCR9 inhibition on intestinal biology.
- Published
- 2013
- Full Text
- View/download PDF
31. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): part two
- Author
-
Casey Ager, Matthew Reilley, Courtney Nicholas, Todd Bartkowiak, Ashvin Jaiswal, Michael Curran, Tina C. Albershardt, Anshika Bajaj, Jacob F. Archer, Rebecca S. Reeves, Lisa Y. Ngo, Peter Berglund, Jan ter Meulen, Caroline Denis, Hormas Ghadially, Thomas Arnoux, Fabien Chanuc, Nicolas Fuseri, Robert W. Wilkinson, Nicolai Wagtmann, Yannis Morel, Pascale Andre, Michael B. Atkins, Matteo S. Carlino, Antoni Ribas, John A. Thompson, Toni K. Choueiri, F. Stephen Hodi, Wen-Jen Hwu, David F. McDermott, Victoria Atkinson, Jonathan S. Cebon, Bernie Fitzharris, Michael B. Jameson, Catriona McNeil, Andrew G. Hill, Eric Mangin, Malidi Ahamadi, Marianne van Vugt, Mariëlle van Zutphen, Nageatte Ibrahim, Georgina V. Long, Robyn Gartrell, Zoe Blake, Ines Simoes, Yichun Fu, Takuro Saito, Yingzhi Qian, Yan Lu, Yvonne M. Saenger, Sadna Budhu, Olivier De Henau, Roberta Zappasodi, Kyle Schlunegger, Bruce Freimark, Jeff Hutchins, Christopher A. Barker, Jedd D. Wolchok, Taha Merghoub, Elena Burova, Omaira Allbritton, Peter Hong, Jie Dai, Jerry Pei, Matt Liu, Joel Kantrowitz, Venus Lai, William Poueymirou, Douglas MacDonald, Ella Ioffe, Markus Mohrs, William Olson, Gavin Thurston, Cristian Capasso, Federica Frascaro, Sara Carpi, Siri Tähtinen, Sara Feola, Manlio Fusciello, Karita Peltonen, Beatriz Martins, Madeleine Sjöberg, Sari Pesonen, Tuuli Ranki, Lukasz Kyruk, Erkko Ylösmäki, Vincenzo Cerullo, Fabio Cerignoli, Biao Xi, Garret Guenther, Naichen Yu, Lincoln Muir, Leyna Zhao, Yama Abassi, Víctor Cervera-Carrascón, Mikko Siurala, João Santos, Riikka Havunen, Suvi Parviainen, Akseli Hemminki, Angus Dalgleish, Satvinder Mudan, Mark DeBenedette, Ana Plachco, Alicia Gamble, Elizabeth W. Grogan, John Krisko, Irina Tcherepanova, Charles Nicolette, Pooja Dhupkar, Ling Yu, Eugenie S. Kleinerman, Nancy Gordon, Italia Grenga, Lauren Lepone, Sofia Gameiro, Karin M. Knudson, Massimo Fantini, Kwong Tsang, James Hodge, Renee Donahue, Jeffrey Schlom, Elizabeth Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Cathie Foster, Alan Howell, Leslie Balch, Alyssa Knapp, John E. Leonard, Mark Paris, Terry Fisher, Siwen Hu-Lieskovan, Ernest Smith, Maurice Zauderer, William Fogler, Marilyn Franklin, Matt Thayer, Dan Saims, John L. Magnani, Jian Gong, Michael Gray, George Fromm, Suresh de Silva, Louise Giffin, Xin Xu, Jason Rose, Taylor H. Schreiber, Sofia R. Gameiro, Paul E. Clavijo, Clint T. Allen, James W. Hodge, Kwong Y. Tsang, Jane Grogan, Nicholas Manieri, Eugene Chiang, Patrick Caplazi, Mahesh Yadav, Patrick Hagner, Hsiling Chiu, Michelle Waldman, Anke Klippel, Anjan Thakurta, Michael Pourdehnad, Anita Gandhi, Ian Henrich, Laura Quick, Rob Young, Margaret Chou, Andrew Hotson, Stephen Willingham, Po Ho, Carmen Choy, Ginna Laport, Ian McCaffery, Richard Miller, Kimberly A. Tipton, Kenneth R. Wong, Victoria Singson, Chihunt Wong, Chanty Chan, Yuanhiu Huang, Shouchun Liu, Jennifer H. Richardson, W. Michael Kavanaugh, James West, Bryan A. Irving, Ritika Jaini, Matthew Loya, Charis Eng, Melissa L. Johnson, Alex A. Adjei, Mateusz Opyrchal, Suresh Ramalingam, Pasi A. Janne, George Dominguez, Dmitry Gabrilovich, Laura de Leon, Jeannette Hasapidis, Scott J. Diede, Peter Ordentlich, Scott Cruickshank, Michael L. Meyers, Matthew D. Hellmann, Pawel Kalinski, Amer Zureikat, Robert Edwards, Ravi Muthuswamy, Nataša Obermajer, Julie Urban, Lisa H. Butterfield, William Gooding, Herbert Zeh, David Bartlett, Olga Zubkova, Larissa Agapova, Marina Kapralova, Liudmila Krasovskaia, Armen Ovsepyan, Maxim Lykov, Artem Eremeev, Vladimir Bokovanov, Olga Grigoryeva, Andrey Karpov, Sergey Ruchko, Alexandr Shuster, Danny N. Khalil, Luis Felipe Campesato, Yanyun Li, Adam S. Lazorchak, Troy D. Patterson, Yueyun Ding, Pottayil Sasikumar, Naremaddepalli Sudarshan, Nagaraj Gowda, Raghuveer Ramachandra, Dodheri Samiulla, Sanjeev Giri, Rajesh Eswarappa, Murali Ramachandra, David Tuck, Timothy Wyant, Jasmin Leshem, Xiu-fen Liu, Tapan Bera, Masaki Terabe, Birgit Bossenmaier, Gerhard Niederfellner, Yoram Reiter, Ira Pastan, Leiming Xia, Yang Xia, Yangyang Hu, Yi Wang, Yangyi Bao, Fu Dai, Shiang Huang, Elaine Hurt, Robert E. Hollingsworth, Lawrence G. Lum, Alfred E. Chang, Max S. Wicha, Qiao Li, Thomas Mace, Neil Makhijani, Erin Talbert, Gregory Young, Denis Guttridge, Darwin Conwell, Gregory B. Lesinski, Rodney JM Macedo Gonzales, Austin P. Huffman, Ximi K. Wang, Ran Reshef, Andy MacKinnon, Jason Chen, Matt Gross, Gisele Marguier, Peter Shwonek, Natalija Sotirovska, Susanne Steggerda, Francesco Parlati, Amani Makkouk, Mark K. Bennett, Ethan Emberley, Tony Huang, Weiqun Li, Silinda Neou, Alison Pan, Jing Zhang, Winter Zhang, Netonia Marshall, Thomas U. Marron, Judith Agudo, Brian Brown, Joshua Brody, Christopher McQuinn, Matthew Farren, Hannah Komar, Reena Shakya, Thomas Ludwug, Y. Maurice Morillon, Scott A. Hammond, John W. Greiner, Pulak R. Nath, Anthony L. Schwartz, Dragan Maric, David D. Roberts, Aung Naing, Kyriakos P. Papadopoulos, Karen A. Autio, Deborah J. Wong, Manish Patel, Gerald Falchook, Shubham Pant, Patrick A. Ott, Melinda Whiteside, Amita Patnaik, John Mumm, Filip Janku, Ivan Chan, Todd Bauer, Rivka Colen, Peter VanVlasselaer, Gail L. Brown, Nizar M. Tannir, Martin Oft, Jeffrey Infante, Evan Lipson, Ajay Gopal, Sattva S. Neelapu, Philippe Armand, Stephen Spurgeon, John P. Leonard, Rachel E. Sanborn, Ignacio Melero, Thomas F. Gajewski, Matthew Maurer, Serena Perna, Andres A. Gutierrez, Raphael Clynes, Priyam Mitra, Satyendra Suryawanshi, Douglas Gladstone, Margaret K. Callahan, James Crooks, Sheila Brown, Audrey Gauthier, Marc Hillairet de Boisferon, Andrew MacDonald, Laura Rosa Brunet, William T. Rothwell, Peter Bell, James M. Wilson, Fumi Sato-Kaneko, Shiyin Yao, Shannon S. Zhang, Dennis A. Carson, Cristina Guiducci, Robert L. Coffman, Kazutaka Kitaura, Takaji Matsutani, Ryuji Suzuki, Tomoko Hayashi, Ezra E. W. Cohen, David Schaer, Yanxia Li, Julie Dobkin, Michael Amatulli, Gerald Hall, Thompson Doman, Jason Manro, Frank Charles Dorsey, Lillian Sams, Rikke Holmgaard, Krishnadatt Persaud, Dale Ludwig, David Surguladze, John S. Kauh, Ruslan Novosiadly, Michael Kalos, Kyla Driscoll, Hardev Pandha, Christy Ralph, Kevin Harrington, Brendan Curti, Wallace Akerley, Sumati Gupta, Alan Melcher, David Mansfield, David R. Kaufman, Emmett Schmidt, Mark Grose, Bronwyn Davies, Roberta Karpathy, Darren Shafren, Katerina Shamalov, Cyrille Cohen, Naveen Sharma, James Allison, Tala Shekarian, Sandrine Valsesia-Wittmann, Christophe Caux, Aurelien Marabelle, Brian M. Slomovitz, Kathleen M. Moore, Hagop Youssoufian, Marshall Posner, Poonam Tewary, Alan D. Brooks, Ya-Ming Xu, Kithsiri Wijeratne, Leslie A. A. Gunatilaka, Thomas J. Sayers, John P. Vasilakos, Tesha Alston, Simon Dovedi, James Elvecrog, Iwen Grigsby, Ronald Herbst, Karen Johnson, Craig Moeckly, Stefanie Mullins, Kristen Siebenaler, Julius SternJohn, Ashenafi Tilahun, Mark A. Tomai, Katharina Vogel, Eveline E. Vietsch, Anton Wellstein, Martin Wythes, Stefano Crosignani, Joseph Tumang, Shilpa Alekar, Patrick Bingham, Sandra Cauwenberghs, Jenny Chaplin, Deepak Dalvie, Sofie Denies, Coraline De Maeseneire, JunLi Feng, Kim Frederix, Samantha Greasley, Jie Guo, James Hardwick, Stephen Kaiser, Katti Jessen, Erick Kindt, Marie-Claire Letellier, Wenlin Li, Karen Maegley, Reece Marillier, Nichol Miller, Brion Murray, Romain Pirson, Julie Preillon, Virginie Rabolli, Chad Ray, Kevin Ryan, Stephanie Scales, Jay Srirangam, Jim Solowiej, Al Stewart, Nicole Streiner, Vince Torti, Konstantinos Tsaparikos, Xianxian Zheng, Gregory Driessens, Bruno Gomes, Manfred Kraus, Chunxiao Xu, Yanping Zhang, Giorgio Kradjian, Guozhong Qin, Jin Qi, Xiaomei Xu, Bo Marelli, Huakui Yu, Wilson Guzman, Rober Tighe, Rachel Salazar, Kin-Ming Lo, Jessie English, Laszlo Radvanyi, Yan Lan, Michael Postow, Yasin Senbabaoglu, Billel Gasmi, Hong Zhong, Cailian Liu, Daniel Hirschhorhn-Cymerman, Yuanyuan Zha, Gregory Malnassy, Noreen Fulton, Jae-Hyun Park, Wendy Stock, Yusuke Nakamura, Hongtao Liu, Xiaoming Ju, Rachelle Kosoff, Kimberly Ramos, Brandon Coder, Robert Petit, Michael Princiotta, Kyle Perry, Jun Zou, Ainhoa Arina, Christian Fernandez, Wenxin Zheng, Michael A. Beckett, Helena J. Mauceri, Yang-Xin Fu, Ralph R. Weichselbaum, Whitney Lewis, Yanyan Han, Yeting Wu, Chou Yang, Jing Huang, Dongyun Wu, Jin Li, Xiaoling Liang, Xiangjun Zhou, Jinlin Hou, Raffit Hassan, Thierry Jahan, Scott J. Antonia, Hedy L. Kindler, Evan W. Alley, Somayeh Honarmand, Weiqun Liu, Meredith L. Leong, Chan C. Whiting, Nitya Nair, Amanda Enstrom, Edward E. Lemmens, Takahiro Tsujikawa, Sushil Kumar, Lisa M. Coussens, Aimee L. Murphy, Dirk G. Brockstedt, Sven D. Koch, Martin Sebastian, Christian Weiss, Martin Früh, Miklos Pless, Richard Cathomas, Wolfgang Hilbe, Georg Pall, Thomas Wehler, Jürgen Alt, Helge Bischoff, Michael Geissler, Frank Griesinger, Jens Kollmeier, Alexandros Papachristofilou, Fatma Doener, Mariola Fotin-Mleczek, Madeleine Hipp, Henoch S. Hong, Karl-Josef Kallen, Ute Klinkhardt, Claudia Stosnach, Birgit Scheel, Andreas Schroeder, Tobias Seibel, Ulrike Gnad-Vogt, Alfred Zippelius, Ha-Ram Park, Yong-Oon Ahn, Tae Min Kim, Soyeon Kim, Seulki Kim, Yu Soo Lee, Bhumsuk Keam, Dong-Wan Kim, Dae Seog Heo, Shari Pilon-Thomas, Amy Weber, Jennifer Morse, Krithika Kodumudi, Hao Liu, John Mullinax, Amod A. Sarnaik, Luke Pike, Andrew Bang, Tracy Balboni, Allison Taylor, Alexander Spektor, Tyler Wilhite, Monica Krishnan, Daniel Cagney, Brian Alexander, Ayal Aizer, Elizabeth Buchbinder, Mark Awad, Leena Ghandi, Jonathan Schoenfeld, Elizabeth Lessey-Morillon, Lisa Ridnour, Neil H. Segal, Manish Sharma, Dung T. Le, Robert L. Ferris, Andrew D. Zelenetz, Ronald Levy, Izidore S. Lossos, Caron Jacobson, Radhakrishnan Ramchandren, John Godwin, A. Dimitrios Colevas, Roland Meier, Suba Krishnan, Xuemin Gu, Jaclyn Neely, John Timmerman, Claire I. Vanpouille-Box, Silvia C. Formenti, Sandra Demaria, Erik Wennerberg, Aranzazu Mediero, Bruce N. Cronstein, Michael P. Gustafson, AriCeli DiCostanzo, Courtney Wheatley, Chul-Ho Kim, Svetlana Bornschlegl, Dennis A. Gastineau, Bruce D. Johnson, Allan B. Dietz, Cameron MacDonald, Mark Bucsek, Guanxi Qiao, Bonnie Hylander, Elizabeth Repasky, William J. Turbitt, Yitong Xu, Andrea Mastro, Connie J. Rogers, Sita Withers, Ziming Wang, Lam T. Khuat, Cordelia Dunai, Bruce R. Blazar, Dan Longo, Robert Rebhun, Steven K. Grossenbacher, Arta Monjazeb, William J. Murphy, Scott Rowlinson, Giulia Agnello, Susan Alters, David Lowe, Nicole Scharping, Ashley V. Menk, Ryan Whetstone, Xue Zeng, Greg M. Delgoffe, Patricia M. Santos, Jian Shi, Greg Delgoffe, Misako Nagasaka, Ammar Sukari, Miranda Byrne-Steele, Wenjing Pan, Xiaohong Hou, Brittany Brown, Mary Eisenhower, Jian Han, Natalie Collins, Robert Manguso, Hans Pope, Yashaswi Shrestha, Jesse Boehm, W. Nicholas Haining, Kyle R. Cron, Ayelet Sivan, Keston Aquino-Michaels, Marco Orecchioni, Davide Bedognetti, Wouter Hendrickx, Claudia Fuoco, Filomena Spada, Francesco Sgarrella, Gianni Cesareni, Francesco Marincola, Kostas Kostarelos, Alberto Bianco, Lucia Delogu, Jessica Roelands, Sabri Boughorbel, Julie Decock, Scott Presnell, Ena Wang, Franco M. Marincola, Peter Kuppen, Michele Ceccarelli, Darawan Rinchai, Damien Chaussabel, Lance Miller, Andrew Nguyen, J. Zachary Sanborn, Charles Vaske, Shahrooz Rabizadeh, Kayvan Niazi, Steven Benz, Shashank Patel, Nicholas Restifo, James White, Sam Angiuoli, Mark Sausen, Sian Jones, Maria Sevdali, John Simmons, Victor Velculescu, Luis Diaz, Theresa Zhang, Jennifer S. Sims, Sunjay M. Barton, Angela Kadenhe-Chiweshe, Filemon Dela Cruz, Andrew T. Turk, Christopher F. Mazzeo, Andrew L. Kung, Jeffrey N. Bruce, Darrell J. Yamashiro, Eileen P. Connolly, Jason Baird, Marka Crittenden, David Friedman, Hong Xiao, Rom Leidner, Bryan Bell, Kristina Young, Michael Gough, Zhen Bian, Koby Kidder, Yuan Liu, Emily Curran, Xiufen Chen, Leticia P. Corrales, Justin Kline, Ethan G. Aguilar, Jennifer Guerriero, Alaba Sotayo, Holly Ponichtera, Alexandra Pourzia, Sara Schad, Ruben Carrasco, Suzan Lazo, Roderick Bronson, Anthony Letai, Richard S. Kornbluth, Sachin Gupta, James Termini, Elizabeth Guirado, Geoffrey W. Stone, Christina Meyer, Laura Helming, Nicholas Wilson, Robert Hofmeister, Natalie J. Neubert, Laure Tillé, David Barras, Charlotte Soneson, Petra Baumgaertner, Donata Rimoldi, David Gfeller, Mauro Delorenzi, Silvia A. Fuertes Marraco, Daniel E. Speiser, Tara S. Abraham, Bo Xiang, Michael S. Magee, Scott A. Waldman, Adam E. Snook, Wojciech Blogowski, Ewa Zuba-Surma, Marta Budkowska, Daria Salata, Barbara Dolegowska, Teresa Starzynska, Leo Chan, Srinivas Somanchi, Kelsey McCulley, Dean Lee, Nico Buettner, Feng Shi, Paisley T. Myers, Stuart Curbishley, Sarah A. Penny, Lora Steadman, David Millar, Ellen Speers, Nicola Ruth, Gabriel Wong, Robert Thimme, David Adams, Mark Cobbold, Remy Thomas, Mariam Al-Muftah, Michael KK Wong, Michael Morse, Joseph I. Clark, Howard L. Kaufman, Gregory A. Daniels, Hong Hua, Tharak Rao, Janice P. Dutcher, Kai Kang, Yogen Saunthararajah, Vamsidhar Velcheti, Vikas Kumar, Firoz Anwar, Amita Verma, Zinal Chheda, Gary Kohanbash, John Sidney, Kaori Okada, Shruti Shrivastav, Diego A. Carrera, Shuming Liu, Naznin Jahan, Sabine Mueller, Ian F. Pollack, Angel M. Carcaboso, Alessandro Sette, Yafei Hou, Hideho Okada, Jessica J. Field, Weiping Zeng, Vincent FS Shih, Che-Leung Law, Peter D. Senter, Shyra J. Gardai, Nicole M. Okeley, Jennifer G. Abelin, Abu Z. Saeed, Stacy A. Malaker, Jeffrey Shabanowitz, Stephen T. Ward, Donald F. Hunt, Pam Profusek, Laura Wood, Dale Shepard, Petros Grivas, Kerstin Kapp, Barbara Volz, Detlef Oswald, Burghardt Wittig, Manuel Schmidt, Julian P. Sefrin, Lars Hillringhaus, Valeria Lifke, Alexander Lifke, Anna Skaletskaya, Jose Ponte, Thomas Chittenden, Yulius Setiady, Eva Sivado, Vincent Thomas, Meddy El Alaoui, Sébastien Papot, Charles Dumontet, Mike Dyson, John McCafferty, Said El Alaoui, Praveen K. Bommareddy, Andrew Zloza, Frederick Kohlhapp, Ann W. Silk, Sachin Jhawar, Tomas Paneque, Jenna Newman, Pedro Beltran, Felicia Cao, Bang-Xing Hong, Tania Rodriguez-Cruz, Xiao-Tong Song, Stephen Gottschalk, Hugo Calderon, Sam Illingworth, Alice Brown, Kerry Fisher, Len Seymour, Brian Champion, Emma Eriksson, Jessica Wenthe, Ann-Charlotte Hellström, Gabriella Paul-Wetterberg, Angelica Loskog, Ioanna Milenova, Magnus Ståhle, Justyna Jarblad-Leja, Gustav Ullenhag, Anna Dimberg, Rafael Moreno, Ramon Alemany, Sharad Goyal, Ann Silk, Janice Mehnert, Nashat Gabrail, Jennifer Bryan, Daniel Medina, Leah Mitchell, Kader Yagiz, Fernando Lopez, Daniel Mendoza, Anthony Munday, Harry Gruber, Douglas Jolly, Steven Fuhrmann, Sasa Radoja, Wei Tan, Aldo Pourchet, Alan Frey, Ian Mohr, Matthew Mulvey, Robert H. I. Andtbacka, Merrick Ross, Sanjiv Agarwala, Kenneth Grossmann, Matthew Taylor, John Vetto, Rogerio Neves, Adil Daud, Hung Khong, Stephanie M. Meek, Richard Ungerleider, Scott Welden, Maki Tanaka, Matthew Williams, Sigrun Hallmeyer, Bernard Fox, Zipei Feng, Christopher Paustian, Carlo Bifulco, Sadia Zafar, Otto Hemminki, Simona Bramante, Lotta Vassilev, Hongjie Wang, Andre Lieber, Silvio Hemmi, Tanja de Gruijl, Anna Kanerva, Tameem Ansari, Srividya Sundararaman, Diana Roen, Paul Lehmann, Anja C. Bloom, Lewis H. Bender, Ian B. Walters, Jay A. Berzofsky, Fanny Chapelin, Eric T. Ahrens, Jeff DeFalco, Michael Harbell, Amy Manning-Bog, Alexander Scholz, Danhui Zhang, Gilson Baia, Yann Chong Tan, Jeremy Sokolove, Dongkyoon Kim, Kevin Williamson, Xiaomu Chen, Jillian Colrain, Gregg Espiritu Santo, Ngan Nguyen, Wayne Volkmuth, Norman Greenberg, William Robinson, Daniel Emerling, Charles G. Drake, Daniel P. Petrylak, Emmanuel S. Antonarakis, Adam S. Kibel, Nancy N. Chang, Tuyen Vu, Dwayne Campogan, Heather Haynes, James B. Trager, Nadeem A. Sheikh, David I. Quinn, Peter Kirk, Murali Addepalli, Thomas Chang, Ping Zhang, Marina Konakova, Katsunobu Hagihara, Steven Pai, Laurie VanderVeen, Palakshi Obalapur, Peiwen Kuo, Phi Quach, Lawrence Fong, Deborah H. Charych, Jonathan Zalevsky, John L. Langowski, Yolanda Kirksey, Ravi Nutakki, Shalini Kolarkar, Rhoneil Pena, Ute Hoch, Stephen K. Doberstein, John Cha, Zach Mallon, Myra Perez, Amanda McDaniel, Snjezana Anand, Darrin Uecker, Richard Nuccitelli, Eva Wieckowski, Ravikumar Muthuswamy, Roshni Ravindranathan, Ariana N. Renrick, Menaka Thounaojam, Portia Thomas, Samuel Pellom, Anil Shanker, Duafalia Dudimah, Alan Brooks, Yu-Lin Su, Tomasz Adamus, Qifang Zhang, Sergey Nechaev, Marcin Kortylewski, Spencer Wei, Clark Anderson, Chad Tang, Jonathan Schoenhals, Efrosini Tsouko, John Heymach, Patricia de Groot, Joe Chang, Kenneth R. Hess, Adi Diab, Padmanee Sharma, David Hong, James Welsh, Andrea J. Parsons, Jardin Leleux, Stephane Ascarateil, Marie Eve Koziol, Dina Bai, Peihong Dai, Weiyi Wang, Ning Yang, Stewart Shuman, Liang Deng, Patrick Dillon, Gina Petroni, David Brenin, Kim Bullock, Walter Olson, Mark E. Smolkin, Kelly Smith, Carmel Nail, Craig L. Slingluff, Meenu Sharma, Faisal Fa’ak, Louise Janssen, Hiep Khong, Zhilan Xiao, Yared Hailemichael, Manisha Singh, Christina Vianden, Willem W. Overwijk, Andrea Facciabene, Pierini Stefano, Fang Chongyung, Stavros Rafail, Michael Nielsen, Peter Vanderslice, Darren G. Woodside, Robert V. Market, Ronald J. Biediger, Upendra K. Marathi, Kevin Hollevoet, Nick Geukens, Paul Declerck, Nathalie Joly, Laura McIntosh, Eustache Paramithiotis, Magnus Rizell, Malin Sternby, Bengt Andersson, Alex Karlsson-Parra, Rui Kuai, Lukasz Ochyl, Anna Schwendeman, James Moon, Weiwen Deng, Thomas E. Hudson, Bill Hanson, Chris S. Rae, Joel Burrill, Justin Skoble, George Katibah, Michele deVries, Peter Lauer, Thomas W. Dubensky, Xin Chen, Li Zhou, Xiubao Ren, Charu Aggarwal, Drishty Mangrolia, Roger Cohen, Gregory Weinstein, Matthew Morrow, Joshua Bauml, Kim Kraynyak, Jean Boyer, Jian Yan, Jessica Lee, Laurent Humeau, Sandra Oyola, Susan Duff, David Weiner, Zane Yang, Mark Bagarazzi, Douglas G. McNeel, Jens Eickhoff, Robert Jeraj, Mary Jane Staab, Jane Straus, Brian Rekoske, Glenn Liu, Marit Melssen, William Grosh, Nikole Varhegyi, Nadejda Galeassi, Donna H. Deacon, Elizabeth Gaughan, Maurizio Ghisoli, Minal Barve, Robert Mennel, Gladice Wallraven, Luisa Manning, Neil Senzer, John Nemunaitis, Masahiro Ogasawara, Shuichi Ota, Kaitlin M. Peace, Diane F. Hale, Timothy J. Vreeland, Doreen O. Jackson, John S. Berry, Alfred F. Trappey, Garth S. Herbert, Guy T. Clifton, Mark O. Hardin, Anne Toms, Na Qiao, Jennifer Litton, George E. Peoples, Elizabeth A. Mittendorf, Lila Ghamsari, Emilio Flano, Judy Jacques, Biao Liu, Jonathan Havel, Vladimir Makarov, Timothy A. Chan, Jessica B. Flechtner, John Facciponte, Stefano Ugel, Francesco De Sanctis, George Coukos, Sébastien Paris, Agnes Pottier, Laurent Levy, Bo Lu, Federica Cappuccini, Emily Pollock, Richard Bryant, Freddie Hamdy, Adrian Hill, Irina Redchenko, Hussein Sultan, Takumi Kumai, Valentyna Fesenkova, Esteban Celis, Ingrid Fernando, Claudia Palena, Justin M. David, Elizabeth Gabitzsch, Frank Jones, James L. Gulley, Mireia Uribe Herranz, Hiroshi Wada, Atsushi Shimizu, Toshihiro Osada, Satoshi Fukaya, Eiji Sasaki, Milad Abolhalaj, David Askmyr, Kristina Lundberg, Ann-Sofie Albrekt, Lennart Greiff, Malin Lindstedt, Dallas B. Flies, Tomoe Higuchi, Wojciech Ornatowski, Jaryse Harris, Sarah F. Adams, Todd Aguilera, Marjan Rafat, Laura Castellini, Hussein Shehade, Mihalis Kariolis, Dadi Jang, Rie vonEbyen, Edward Graves, Lesley Ellies, Erinn Rankin, Albert Koong, Amato Giaccia, Reham Ajina, Shangzi Wang, Jill Smith, Mariaelena Pierobon, Sandra Jablonski, Emanuel Petricoin, Louis M. Weiner, Lorcan Sherry, John Waller, Mark Anderson, Alison Bigley, Chantale Bernatchez, Cara Haymaker, Harriet Kluger, Michael Tetzlaff, Natalie Jackson, Ivan Gergel, Mary Tagliaferri, Patrick Hwu, Mario Snzol, Michael Hurwitz, Theresa Barberi, Allison Martin, Rahul Suresh, David Barakat, Sarah Harris-Bookman, Charles Drake, Alan Friedman, Sara Berkey, Stephanie Downs-Canner, Robert P. Edwards, Tyler Curiel, Kunle Odunsi, Tullia C. Bruno, Brandon Moore, Olivia Squalls, Peggy Ebner, Katherine Waugh, John Mitchell, Wilbur Franklin, Daniel Merrick, Martin McCarter, Brent Palmer, Jeffrey Kern, Dario Vignali, Jill Slansky, Anissa S. H. Chan, Xiaohong Qiu, Kathryn Fraser, Adria Jonas, Nadine Ottoson, Keith Gordon, Takashi O. Kangas, Steven Leonardo, Kathleen Ertelt, Richard Walsh, Mark Uhlik, Jeremy Graff, Nandita Bose, Ravi Gupta, Nitin Mandloi, Kiran Paul, Ashwini Patil, Rekha Sathian, Aparna Mohan, Malini Manoharan, Amitabha Chaudhuri, Yu Chen, Jing Lin, Yun-bin Ye, Chun-wei Xu, Gang Chen, Zeng-qing Guo, Andrey Komarov, Alex Chenchik, Michael Makhanov, Costa Frangou, Yi Zheng, Carla Coltharp, Darryn Unfricht, Ryan Dilworth, Leticia Fridman, Linying Liu, Milind Rajopadhye, Peter Miller, Fernando Concha-Benavente, Julie Bauman, Sumita Trivedi, Raghvendra Srivastava, James Ohr, Dwight Heron, Uma Duvvuri, Seungwon Kim, Heather Torrey, Toshi Mera, Yoshiaki Okubo, Eva Vanamee, Rosemary Foster, Denise Faustman, Edward Stack, Daisuke Izaki, Kristen Beck, Dan Tong Jia, Paul Armenta, Ashley White-Stern, Douglas Marks, Bret Taback, Basil Horst, Laura Hix Glickman, David B. Kanne, Kelsey S. Gauthier, Anthony L. Desbien, Brian Francica, Justin L. Leong, Leonard Sung, Ken Metchette, Shailaja Kasibhatla, Anne Marie Pferdekamper, Lianxing Zheng, Charles Cho, Yan Feng, Jeffery M. McKenna, John Tallarico, Steven Bender, Chudi Ndubaku, Sarah M. McWhirter, Elena Gonzalez Gugel, Charles J. M. Bell, Adiel Munk, Luciana Muniz, Nina Bhardwaj, Fei Zhao, Kathy Evans, Christine Xiao, Alisha Holtzhausen, Brent A. Hanks, Nathalie Scholler, Catherine Yin, Pien Van der Meijs, Andrew M. Prantner, Cecile M. Krejsa, Leia Smith, Brian Johnson, Daniel Branstetter, Paul L. Stein, Juan C. Jaen, Joanne BL Tan, Ada Chen, Timothy Park, Jay P. Powers, Holly Sexton, Guifen Xu, Steve W. Young, Ulrike Schindler, Wentao Deng, David John Klinke, Hannah M. Komar, Gregory Serpa, Omar Elnaggar, Philip Hart, Carl Schmidt, Mary Dillhoff, Ming Jin, Michael C. Ostrowski, Madhuri Koti, Katrina Au, Nichole Peterson, Peter Truesdell, Gillian Reid-Schachter, Charles Graham, Andrew Craig, Julie-Ann Francis, Beatrix Kotlan, Timea Balatoni, Emil Farkas, Laszlo Toth, Mihaly Ujhelyi, Akos Savolt, Zoltan Doleschall, Szabolcs Horvath, Klara Eles, Judit Olasz, Orsolya Csuka, Miklos Kasler, Gabriella Liszkay, Eytan Barnea, Collin Blakely, Patrick Flynn, Reid Goodman, Raphael Bueno, David Sugarbaker, David Jablons, V. Courtney Broaddus, Brian West, Paul R. Kunk, Joseph M. Obeid, Kevin Winters, Patcharin Pramoonjago, Edward B. Stelow, Todd W. Bauer, Osama E. Rahma, Adam Lamble, Yoko Kosaka, Fei Huang, Kate A. Saser, Homer Adams, Christina E. Tognon, Ted Laderas, Shannon McWeeney, Marc Loriaux, Jeffery W. Tyner, Brian J. Druker, Evan F. Lind, Zhuqing Liu, Shanhong Lu, Lawrence P. Kane, Gulidanna Shayan, Julia Femel, Ryan Lane, Jamie Booth, Amanda W. Lund, Anthony Rodriguez, Victor H. Engelhard, Alessandra Metelli, Bill X. Wu, Caroline W. Fugle, Rachidi Saleh, Shaoli Sun, Jennifer Wu, Bei Liu, Zihai Li, Zachary S. Morris, Emily I. Guy, Clinton Heinze, Jasdeep Kler, Monica M. Gressett, Lauryn R. Werner, Stephen D. Gillies, Alan J. Korman, Hans Loibner, Jacquelyn A. Hank, Alexander L. Rakhmilevich, Paul M. Harari, Paul M. Sondel, Erica Huelsmann, Joseph Broucek, Dorothee Brech, Tobias Straub, Martin Irmler, Johannes Beckers, Florian Buettner, Elke Schaeffeler, Matthias Schwab, Elfriede Noessner, Alison Wolfreys, Andre Da Costa, John Silva, Andrea Crosby, Ludovicus Staelens, Graham Craggs, Annick Cauvin, Sean Mason, Alison M. Paterson, Andrew C. Lake, Caroline M. Armet, Rachel W. O’Connor, Jonathan A. Hill, Emmanuel Normant, Ammar Adam, Detlev M. Biniszkiewicz, Scott C. Chappel, Vito J. Palombella, Pamela M. Holland, Annette Becker, Manmohan R. Leleti, Eric Newcomb, Joanne B. L. Tan, Suthee Rapisuwon, Arash Radfar, Kellie Gardner, Geoffrey Gibney, Michael Atkins, Keith R. Rennier, Robert Crowder, Ping Wang, Russell K. Pachynski, Rosa M. Santana Carrero, Sarai Rivas, Figen Beceren-Braun, Scott Anthony, Kimberly S. Schluns, Deepali Sawant, Maria Chikina, Hiroshi Yano, Creg Workman, Elise Salerno, Ileana Mauldin, Donna Deacon, Sofia Shea, Joel Pinczewski, Thomas Gajewski, Stefani Spranger, Brendan Horton, Akiko Suzuki, Pamela Leland, Bharat H. Joshi, Raj K. Puri, Randy F. Sweis, Riyue Bao, Jason Luke, Marie-Nicole Theodoraki, Frances-Mary Mogundo, Haejung Won, Dayson Moreira, Chan Gao, Xingli Zhao, Priyanka Duttagupta, Jeremy Jones, Massimo D’Apuzzo, and Sumanta Pal
- Subjects
0301 basic medicine ,Pharmacology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Cancer ,Immunotherapy ,medicine.disease ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business - Abstract
O1 IL-15 primes an mTOR-regulated gene-expression program to prolong anti-tumor capacity of human natural killer cells #### Andreas Lundqvist1, Vincent van Hoef1, Xiaonan Zhang1, Erik Wennerberg2, Julie Lorent1, Kristina Witt1, Laia Masvidal Sanz1, Shuo Liang1, Shannon Murray3, Ola Larsson1
- Published
- 2016
32. Characterization of Pharmacologic and Pharmacokinetic Properties of CCX168, a Potent and Selective Orally Administered Complement 5a Receptor Inhibitor, Based on Preclinical Evaluation and Randomized Phase 1 Clinical Study
- Author
-
Daniel J. Dairaghi, Jay P. Powers, Juan C. Jaen, Trageen Baumgart, Yu Wang, Israel F. Charo, Ton Dang, Sarah Shugarts, Lisa Seitz, Daniel Johnson, Yibin Zeng, Lisa Lohr, Linda S. Ertl, Penglie Zhang, Pingchen Fan, Pirow Bekker, Manmohan Reddy Leleti, Thomas J. Schall, and Shichang Miao
- Subjects
0301 basic medicine ,Neutrophils ,Physiology ,Complement System ,Nipecotic Acids ,lcsh:Medicine ,Phases of clinical research ,Administration, Oral ,Drug research and development ,Monkeys ,Pharmacology ,Biochemistry ,C5a receptor ,White Blood Cells ,Mice ,Clinical trials ,0302 clinical medicine ,Animal Cells ,Immune Physiology ,Medicine and Health Sciences ,Medicine ,lcsh:Science ,Receptor ,Mammals ,Immune System Proteins ,Multidisciplinary ,Aniline Compounds ,Phase I clinical investigation ,Chemotaxis ,Hematology ,U937 Cells ,Healthy Volunteers ,Body Fluids ,Cell Motility ,Blood ,Tolerability ,Vertebrates ,Cellular Types ,Anatomy ,Research Article ,Primates ,Immune Cells ,Immunology ,Mice, Transgenic ,Blood Plasma ,03 medical and health sciences ,Pharmacokinetics ,In vivo ,Potency ,Animals ,Humans ,Receptor, Anaphylatoxin C5a ,Blood Cells ,business.industry ,lcsh:R ,Organisms ,Biology and Life Sciences ,Proteins ,Correction ,Cell Biology ,Research and analysis methods ,Macaca fascicularis ,030104 developmental biology ,Clinical medicine ,Immune System ,Amniotes ,lcsh:Q ,business ,030217 neurology & neurosurgery ,Ex vivo - Abstract
The complement 5a receptor has been an attractive therapeutic target for many autoimmune and inflammatory disorders. However, development of a selective and potent C5aR antagonist has been challenging. Here we describe the characterization of CCX168 (avacopan), an orally administered selective and potent C5aR inhibitor. CCX168 blocked the C5a binding, C5a-mediated migration, calcium mobilization, and CD11b upregulation in U937 cells as well as in freshly isolated human neutrophils. CCX168 retains high potency when present in human blood. A transgenic human C5aR knock-in mouse model allowed comparison of the in vitro and in vivo efficacy of the molecule. CCX168 effectively blocked migration in in vitro and ex vivo chemotaxis assays, and it blocked the C5a-mediated neutrophil vascular endothelial margination. CCX168 was effective in migration and neutrophil margination assays in cynomolgus monkeys. This thorough in vitro and preclinical characterization enabled progression of CCX168 into the clinic and testing of its safety, tolerability, pharmacokinetic, and pharmacodynamic profiles in a Phase 1 clinical trial in 48 healthy volunteers. CCX168 was shown to be well tolerated across a broad dose range (1 to 100 mg) and it showed dose-dependent pharmacokinetics. An oral dose of 30 mg CCX168 given twice daily blocked the C5a-induced upregulation of CD11b in circulating neutrophils by 94% or greater throughout the entire day, demonstrating essentially complete target coverage. This dose regimen is being tested in clinical trials in patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Trial Registration ISRCTN registry with trial ID ISRCTN13564773.
- Published
- 2016
33. Discovery and optimization of benzenesulfonanilide derivatives as a novel class of 11β-HSD1 inhibitors
- Author
-
Stefania Ursu, Daqing Sun, Yosup Rew, Juan C. Jaen, Jay P. Powers, Xiao He, Hua Tu, Dustin McMinn, and Michael DeGraffenreid
- Subjects
Stereochemistry ,Clinical Biochemistry ,Drug Evaluation, Preclinical ,Molecular Conformation ,Pharmaceutical Science ,Crystallography, X-Ray ,Transfection ,Biochemistry ,Piperazines ,11β hsd1 inhibitors ,Structure-Activity Relationship ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,Drug Discovery ,Humans ,Anilides ,Enzyme Inhibitors ,Piperazine ,Molecular Biology ,Biological evaluation ,chemistry.chemical_classification ,Sulfonamides ,Aniline Compounds ,Chemistry ,Organic Chemistry ,Combinatorial chemistry ,Sulfonamide ,Enzyme Activation ,HEK293 Cells ,Molecular Medicine ,hormones, hormone substitutes, and hormone antagonists - Abstract
A novel series of benzenesulfonanilide derivatives of 11β-HSD1 inhibitors were identified via modification of the sulfonamide core of the arylsulfonylpiperazine lead structures. The synthesis, in vitro biological evaluation, and structure–activity relationship of these compounds are presented. Optimization of this series rapidly resulted in the discovery of compounds (S)-10 and (S)-23 (11β-HSD1 SPA IC50 = 1.8 and 1.4 nM, respectively).
- Published
- 2012
34. Discovery of amide replacements that improve activity and metabolic stability of a bis-amide smoothened antagonist hit
- Author
-
Minqing Rong, Brian Lucas, Wendy Zhong, Guifen Xu, Gary Lee, Ben Jiang, Angela Chong, Qiuping Ye, Michael DeGraffenreid, Tom Huang, Dineli Wickramasinghe, Randall W. Hungate, Wade Aaron, Michael G. Johnson, Jessica Orf, Dustin McMinn, Matthew L. Brown, Richard J. Austin, Maria M. Toteva, Jacob Kaizerman, and Jay P. Powers
- Subjects
Molecular Structure ,Drug discovery ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Antagonist ,Pharmaceutical Science ,Stereoisomerism ,Amides ,Biochemistry ,Hedgehog signaling pathway ,In vitro ,High-Throughput Screening Assays ,Structure-Activity Relationship ,chemistry.chemical_compound ,In vivo ,Amide ,Drug Discovery ,Molecular Medicine ,Structure–activity relationship ,Enzyme Inhibitors ,Smoothened ,Molecular Biology ,Acyltransferases - Abstract
A bis-amide antagonist of Smoothened, a seven-transmembrane receptor in the Hedgehog signaling pathway, was discovered via high throughput screening. In vitro and in vivo experiments demonstrated that the bis-amide was susceptible to N-acyl transferase mediated amide scission. Several bioisosteric replacements of the labile amide that maintained in vitro potency were identified and shown to be metabolically stable in vitro and in vivo.
- Published
- 2011
35. Synthesis and optimization of novel 4,4-disubstituted cyclohexylbenzamide derivatives as potent 11β-HSD1 inhibitors
- Author
-
Hua Tu, Lisa Julian, Nigel Walker, Maren Gulsrud Willcockson, Yosup Rew, Michael DeGraffenreid, Zhulun Wang, Juan C. Jaen, Daqing Sun, Ron C. Kelly, Athena Sudom, Seb Caille, Jacob Kaizerman, Jay P. Powers, Xuelei Yan, Qiuping Ye, Stefania Ursu, Ben Jiang, Felix Gonzalez-Lopez de Turiso, Randall W. Hungate, and Dustin McMinn
- Subjects
medicine.drug_class ,Clinical Biochemistry ,Administration, Oral ,Pharmaceutical Science ,Carboxamide ,Crystallography, X-Ray ,Biochemistry ,Chemical synthesis ,11β hsd1 inhibitors ,Structure-Activity Relationship ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,Drug Discovery ,medicine ,Animals ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,biology ,Chemistry ,Organic Chemistry ,Highly selective ,Combinatorial chemistry ,Rats ,Macaca fascicularis ,Enzyme ,Enzyme inhibitor ,Benzamides ,Microsomes, Liver ,biology.protein ,Molecular Medicine ,Ex vivo - Abstract
The synthesis and SAR of a series of 4,4-disubstituted cyclohexylbenzamide inhibitors of 11β-HSD1 are described. Optimization rapidly led to potent, highly selective, and orally bioavailable inhibitors demonstrating efficacy in both rat and non-human primate ex vivo pharmacodynamic models.
- Published
- 2011
36. Abstract 5556: Combining adenosine receptor inhibition with AB928 and chemotherapy results in greater immune activation and tumor control
- Author
-
Matt J. Walters, Dillon H. Miles, Manmohan Reddy Leleti, Jay P. Powers, Eshan Sharif, Fang-Fang Yin, Tim Park, Dana Piovesan, Joanne Tan, Ulrike Schindler, Daniel DiRenzo, and Ferdie Soriano
- Subjects
0301 basic medicine ,Cancer Research ,Chemistry ,Adenosine receptor ,Adenosine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,medicine ,Cancer research ,Cytotoxic T cell ,Immunogenic cell death ,Doxorubicin ,CD8 ,medicine.drug - Abstract
INTRODUCTION: Extracellular adenosine triphosphate (ATP) is efficiently hydrolyzed to adenosine by ecto-nucleotidases CD39 and CD73, which converts adenosine-monophosphate (AMP) into adenosine (ADO). ADO suppresses immune responses including those of T cells, natural killer (NK) cells, and dendritic cells (DC) through activation of A2aR and A2bR receptors. Treatment of cancer cells with platinum-based and anthracycline chemotherapy has been shown to induce immunogenic cell death (ICD), characterized by increased extracellular ATP levels, and upregulation of CD39 and CD73, leading to the enhanced generation of adenosine. METHODS: The ability of AB928 to inhibit adenosine-mediated suppression of immune cell function was assessed using human CD4 and CD8 cells. BALB/c mice were inoculated with AT-3-OVA tumors, which express the model antigen ovalbumin, and subsequently treated with chemotherapy or AB928 alone, or a combination of both. The growth rate and immune composition of the tumors were assessed (flow cytometry). RESULTS: Consistent with the ability of adenosine to suppress immune function, AB928 inhibited the ability of adenosine to suppress CD4 or CD8 T cell activation. Tumor-bearing mice that have been treated with either doxorubicin (DOX, an anthracycline) or oxaliplatin (OX) exhibit increased expression of CD39 and CD73. Established AT-3-OVA tumors treated with AB928 alone had a small but significant decrease in their growth rate; similarly, tumors treated with chemotherapy exhibited a reduction in growth rate. Consistent with the ICD hypothesis, treatment of AT-3-OVA-bearing mice with DOX or OX resulted in an increase in OVA-specific CD8 T cells in the tumor. Concurrent treatment with AB928 and chemotherapy resulted in significantly reduced tumor growth rates, when compared to chemotherapy alone. Analysis of the tumor-infiltrating cell populations showed a significant increase in OVA-specific CD8 T cells, relative to those treated with chemotherapy alone. CONCLUSIONS: Treatment of tumor-bearing mice with a combination of AB928, a dual A2aR and A2bR antagonist, and ICD-inducing chemotherapy results in increased tumor antigen-specific CD8 T cell responses and significantly reduced tumor growth. AB928 is currently undergoing clinical evaluation. Citation Format: Matt J. Walters, Dana Piovesan, Joanne Tan, Daniel DiRenzo, Fangfang Yin, Dillon Miles, Manmohan R. Leleti, Tim Park, Ferdie Soriano, Eshan Sharif, Ulrike Schindler, Jay P. Powers. Combining adenosine receptor inhibition with AB928 and chemotherapy results in greater immune activation and tumor control [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 5556.
- Published
- 2018
37. Abstract 3769: Pharmacokinetic-pharmacodynamic relationship for AB928, a dual antagonist of the A2aR and A2bR adenosine receptors
- Author
-
Manmohan Reddy Leleti, Jay P. Powers, Matt J. Walters, Tim Park, Aimee Rieger, Dillon H. Miles, Joyson Joseph Karakunnel, Lisa Seitz, Brandon Reid Rosen, Adam Park, Ulrike Schindler, Lixia Jin, Devika Ashok, Steve Young, and Ferdie Soriano
- Subjects
Adenosine monophosphate ,Cancer Research ,medicine.medical_specialty ,Chemistry ,CD14 ,Adenosine ,Adenosine receptor ,chemistry.chemical_compound ,Endocrinology ,Oncology ,Internal medicine ,medicine ,Receptor ,Adenosine triphosphate ,CD8 ,medicine.drug ,Whole blood - Abstract
INTRODUCTION: CD73 converts extracellular adenosine monophosphate (AMP), derived from adenosine triphosphate, into adenosine (ADO). ADO suppresses immune responses, including those of T cells, natural killer (NK) cells and dendritic cells, via A2aR and A2bR receptors. Activation of these receptors results in increased intracellular levels of cyclic AMP (cAMP) and phosphorylation of the cAMP response element-binding protein (CREB). Monitoring the degree of pCREB induced by activation of the Aa2R and A2bR receptors in peripheral blood allows for a specific measurement of target engagement by AB928. In mice, the pCREB assay, coupled with changes in immune cell infiltrate and tumor growth rates, allows for a fuller understanding of the drug's PK/PD relationship. METHODS: Mouse and human whole blood was stimulated with the adenosine agonist NECA (5'-N-Ethylcarboxamidoadenosine) following in vitro spike in or in vivo dosing with AB928. Multi-color phospho-flow cytometry was used to assess levels of pCREB on immune cells. AB928 levels in plasma samples were determined using LC-MS-MS after protein precipitation. RESULTS: A2aR mRNA was the most prevalent adenosine receptor in CD4+ and CD8+ T cells. NK cells also express primarily A2aR, although A2bR was also detected. Dendritic cells and CD14+ monocytes expressed both A2aR and A2bR. NECA stimulation of mouse whole blood resulted in a significant increase in the levels of pCREB within CD8+ T cells with an EC50 of 100 nM, while in human whole blood the EC50 was higher at 700 nM. AB928 (100 nM) exhibited comparable shifts in the NECA dose response between human and murine whole blood, 15 and 16-fold, respectively. Due to the high level of adenosine in the tumor microenvironment, doses of AB928 associated with plasma levels that significantly inhibited 5 μM NECA in the whole-blood pCREB assay were selected for mouse efficacy studies. At these doses, significant reductions in tumor growth were noted. In human whole blood, approximately 90 nM AB928 was required to inhibit 50% of the 5 μM NECA-induced pCREB signal. Inhibition was observed on both CD4+ and CD8+ T cells. NECA-induced pCREB elevations were observed in CD14+ cells less frequently than T cells; however, when the signal was observed, AB928 inhibited it. The human pCREB assay is being utilized to monitor the PD responses in an ongoing clinical trial. Preliminary PK/PD results from this study will be presented. CONCLUSIONS: Systemic extent of receptor (A2aR and A2bR) occupancy by the novel dual antagonist AB928 can be assessed in humans and mice based on the extent of receptor-mediated CREB phosphorylation in blood lymphocytes. AB928 retains much of its inherent potency when competing against high concentrations of adenosine under physiologically relevant conditions (i.e., whole blood). Citation Format: Lisa Seitz, Devika Ashok, Manmohan R. Leleti, Jay P. Powers, Brandon Rosen, Dillon Miles, Lixia Jin, Adam Park, Tim Park, Steve Young, Ferdie Soriano, Aimee Rieger, Ulrike Schindler, Joyson Karakunnel, Matt J. Walters. Pharmacokinetic-pharmacodynamic relationship for AB928, a dual antagonist of the A2aR and A2bR adenosine receptors [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 3769.
- Published
- 2018
38. 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
39. AB680, a potent and selective CD73 small molecule inhibitor, reverses the AMP/adenosine-mediated impairment of immune effector cell activation by immune checkpoint inhibitors
- Author
-
A. Becker, Jay P. Powers, L. Jin, D. Swinarski, K. Zhang, S. Young, Matthew J. Walters, A. Chen, Jenna L. Jeffrey, F. Yin, J. Tan, Ulrike Schindler, Jaroslaw Kalisiak, and Kenneth V. Lawson
- Subjects
0301 basic medicine ,Cancer Research ,Chemistry ,Immune checkpoint inhibitors ,Adenosine ,Small molecule ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Immune effector cell ,medicine ,medicine.drug - Published
- 2018
40. Discovery of Novel, Potent Benzamide Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1) Exhibiting Oral Activity in an Enzyme Inhibition ex Vivo Model▽
- Author
-
Jinsong Liu, Daqing Sun, Tracy Bostick, Randall W. Hungate, Dustin McMinn, Xiao He, Michael DeGraffenreid, Lisa Julian, Juan C. Jaen, Mario Monshouwer, Qiuping Ye, Athena Sudom, Rebekah Choi, Yongmei Di, Yosup Rew, Zhulun Wang, Stefania Ursu, Xuelei Yan, Hua Tu, Seb Caille, Nigel Walker, and Jay P. Powers
- Subjects
Models, Molecular ,Administration, Oral ,Dehydrogenase ,Crystallography, X-Ray ,Cell Line ,Structure-Activity Relationship ,chemistry.chemical_compound ,Dogs ,In vivo ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,Benzamide ,chemistry.chemical_classification ,Molecular Structure ,biology ,In vitro ,Rats ,Macaca fascicularis ,Enzyme ,chemistry ,Biochemistry ,Enzyme inhibitor ,Benzamides ,Models, Animal ,biology.protein ,Molecular Medicine ,hormones, hormone substitutes, and hormone antagonists ,Ex vivo - Abstract
We report the discovery of potent benzamide inhibitors of 11beta-hydroxysteroid dehydrogenase (11beta-HSD1). The optimization and correlation of in vitro and in vivo metabolic stability will be described. Through modifications to our initial lead 2, we discovered pyridyl compound 13. This compound has a favorable pharmacokinetic profile across three species and showed a dose-dependent decrease in adipose 11beta-HSD1 activity in a monkey ex vivo pharmacodynamic model.
- Published
- 2008
41. Discovery and Initial SAR of Arylsulfonylpiperazine Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1)
- Author
-
Qiuping Ye, Daqing Sun, Jay P. Powers, Liang Tang, Craig Tomooka, Stefania Ursu, Hua Tu, Yongmei Di, Athena Sudom, Xuelei Yan, Juan C. Jaen, Shichang Miao, Nigel Walker, Ji Ma, Zhulun Wang, and Marc Labelle
- Subjects
Models, Molecular ,Stereochemistry ,Clinical Biochemistry ,Nitro compound ,Pharmaceutical Science ,Dehydrogenase ,Crystallography, X-Ray ,Biochemistry ,Isozyme ,Chemical synthesis ,Piperazines ,Small Molecule Libraries ,Mice ,Structure-Activity Relationship ,Oxidoreductase ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,Drug Discovery ,Animals ,Humans ,Sulfhydryl Compounds ,Enzyme Inhibitors ,Hydroxysteroid dehydrogenase ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,Molecular Structure ,biology ,Chemistry ,Organic Chemistry ,Stereoisomerism ,Molecular Weight ,Enzyme ,Enzyme inhibitor ,Drug Design ,biology.protein ,Molecular Medicine ,hormones, hormone substitutes, and hormone antagonists - Abstract
High-throughput screening of a small-molecule compound library resulted in the identification of a series of arylsulfonylpiperazines that are potent and selective inhibitors of human 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1). Optimization of the initial lead resulted in the discovery of compound (R)-45 (11β-HSD1 IC50 = 3 nM).
- Published
- 2008
42. An Efficient and Scalable One-Pot Double Michael Addition-Dieckmann Condensation for the Synthesis of 4,4-Disubstituted Cyclohexane β-Keto Esters
- Author
-
Felix Gonzalez-Lopez de Turiso, Daqing Sun, Yosup Rew, Sarah Bennett, Michael DeGraffenreid, Lisa Julian, Randall W. Hungate, Xuelei Yan, Sebastien Caille, Jay P. Powers, Jacob Kaizerman, and Dustin McMinn
- Subjects
Cyclohexane ,Nitrile ,Cyclohexanones ,Organic Chemistry ,Esters ,Condensation reaction ,Chemical synthesis ,Dieckmann condensation ,chemistry.chemical_compound ,chemistry ,Cascade reaction ,Michael reaction ,Organic chemistry ,Methyl acrylate - Abstract
A simple, scalable, and efficient one-pot methodology for the synthesis of 4,4-disubstituted cyclohexane beta-keto esters from benzylic nitriles or esters and methyl acrylate promoted by potassium tert-butoxide is described. The process relies on a tandem double Michael addition-Dieckmann condensation reaction, which results in the formation of three discrete carbon-carbon bonds in a single pot, including a quaternary center. The method allows for the convenient and rapid synthesis of a variety of 4-aryl-4-cyano-2-carbomethoxycyclohexanone and 4-aryl-2,4-biscarbomethoxycyclohexanone building blocks for use in natural products synthesis and medicinal chemistry.
- Published
- 2007
43. SAR studies on a novel series of human cytomegalovirus primase inhibitors
- Author
-
H. DiMaio, Xi Chen, Jay P. Powers, Lingming Liang, Matt Wright, C. Stein, M.G. Peterson, J. Adrian, V. Mayorga, F. Spector, D. Xu, Timothy D. Cushing, Juan C. Jaen, Shichang Miao, and Qiuping Ye
- Subjects
DNA Replication ,Ganciclovir ,Human cytomegalovirus ,Drug-Related Side Effects and Adverse Reactions ,viruses ,Clinical Biochemistry ,Administration, Oral ,Biological Availability ,Cytomegalovirus ,Pharmaceutical Science ,DNA Primase ,Antiviral Agents ,Biochemistry ,Virus ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Animals ,Humans ,Cytotoxicity ,Molecular Biology ,Organic Chemistry ,virus diseases ,Viral Load ,medicine.disease ,Virology ,In vitro ,Rats ,Pyrimidines ,chemistry ,Molecular Medicine ,Primase ,DNA ,medicine.drug ,Cidofovir - Abstract
A novel series of imidazolylpyrimidines were found to possess inhibitory activity against the human CMV UL70 primase. Extensive SAR studies on an HTS lead led to potent, orally bioavailable compounds with anti-CMV IC 50 values of 150 nM in both viral yield and viral DNA replication assays and with a much reduced cytotoxicity compared to marketed treatments ganciclovir and cidofovir.
- Published
- 2007
44. Discovery of potent, selective, and orally bioavailable inhibitors of interleukin-1 receptor-associate kinase-4
- Author
-
Zhaodan Cao, Zhulun Wang, Xiong Gao, Shichang Miao, Nigel Walker, Daqing Sun, Jingqian Liu, Xuelei Yan, Juan C. Jaen, Jay P. Powers, Sheree Johnstone, Matt Wright, Sarah E. Lively, Craig Tomooka, Athena Sudom, and Qiuping Ye
- Subjects
Benzimidazole ,Clinical Biochemistry ,Pharmaceutical Science ,Administration, Oral ,Plasma protein binding ,Interleukin-1 receptor ,Biochemistry ,Enzyme activator ,chemistry.chemical_compound ,Structure-Activity Relationship ,Interleukin-1 Receptor-Associated Kinases ,Drug Discovery ,Structure–activity relationship ,Animals ,Molecular Biology ,Protein Kinase Inhibitors ,Molecular Structure ,Kinase ,Drug discovery ,Organic Chemistry ,Rats ,Enzyme Activation ,chemistry ,Molecular Medicine ,Benzimidazoles ,Protein Binding - Abstract
In this Letter, we report the continued optimization of the N-acyl-2-aminobenzimidazole series, focusing in particular on the N-alkyl substituent and 5-position of the benzimidazole based on the binding mode and the early SAR. These efforts led to the discovery of 16, a highly potent, selective, and orally bioavailable inhibitor of IRAK-4.
- Published
- 2015
45. Crystal Structures of IRAK-4 Kinase in Complex with Inhibitors: A Serine/Threonine Kinase with Tyrosine as a Gatekeeper
- Author
-
Jinsong Liu, Holger Wesche, Shyun Li, Zhulun Wang, Merrill Ayres, Athena Sudom, Nigel Walker, and Jay P. Powers
- Subjects
Protein Conformation ,Molecular Sequence Data ,Glutamic Acid ,Biology ,Mitogen-activated protein kinase kinase ,Crystallography, X-Ray ,Protein Structure, Secondary ,Receptor tyrosine kinase ,Phosphates ,MAP2K7 ,Adenosine Triphosphate ,Structural Biology ,Humans ,Amino Acid Sequence ,c-Raf ,Enzyme Inhibitors ,MOLIMMUNO ,Molecular Biology ,Serine/threonine-specific protein kinase ,Binding Sites ,MAP kinase kinase kinase ,Protein-Tyrosine Kinases ,Interleukin-1 Receptor-Associated Kinases ,Biochemistry ,SIGNALING ,biology.protein ,Tyrosine ,Cyclin-dependent kinase 9 ,Tyrosine kinase ,Protein Binding - Abstract
Interleukin-1 (IL-1) receptor-associated kinase-4 (IRAK-4) is a serine/threonine kinase that plays an essential role in signal transduction by Toll/IL-1 receptors (TIRs). Here, we report the crystal structures of the phosphorylated human IRAK-4 kinase domain in complex with a potent inhibitor and with staurosporine to 2.0 and 2.2 A, respectively. The structures reveal that IRAK-4 has a unique tyrosine gatekeeper residue that interacts with the conserved glutamate from helix alphaC. Consequently, helix alphaC is "pulled in" to maintain the active orientation, and the usual pre-existing hydrophobic back pocket of the ATP-binding site is abolished. The peptide substrate-binding site is more open when compared with other protein kinases due to a marked movement of helix alphaG. The pattern of phosphate ligand interactions in the activation loop bears a close resemblance to that of a tyrosine kinase. Our results provide insights into IRAK-4 function and the design of selective inhibitors.
- Published
- 2006
- Full Text
- View/download PDF
46. Discovery and initial SAR of inhibitors of interleukin-1 receptor-associated kinase-4
- Author
-
Juan C. Jaen, Jay P. Powers, Nigel Walker, Zhulun Wang, Holger Wesche, Jinqian Liu, and Shyun Li
- Subjects
Alkylation ,High-throughput screening ,Clinical Biochemistry ,Pharmaceutical Science ,Protein Serine-Threonine Kinases ,Interleukin-1 receptor ,Biochemistry ,Structure-Activity Relationship ,Drug Discovery ,Structure–activity relationship ,Protein kinase A ,Protein Kinase Inhibitors ,Molecular Biology ,Molecular Structure ,biology ,Kinase ,Chemistry ,Organic Chemistry ,Intracellular Signaling Peptides and Proteins ,Amides ,Small molecule ,Cross-Linking Reagents ,Interleukin-1 Receptor-Associated Kinases ,Enzyme inhibitor ,biology.protein ,Cancer research ,Molecular Medicine ,Benzimidazoles ,Signal transduction - Abstract
High-throughput screening of a small-molecule compound library resulted in the identification of a novel series of N-acyl 2-aminobenzimidazoles that are potent inhibitors of interleukin-1 receptor-associated kinase-4.
- Published
- 2006
47. SAR and Mode of Action of Novel Non-Nucleoside Inhibitors of Hepatitis C NS5b RNA Polymerase
- Author
-
Zhulun Wang, Yang Li, M Greg Peterson, Derek E. Piper, Jinqian Liu, George Tonn, Jay P. Powers, Juan C. Jaen, Veronica Mayorga, Nigel Walker, Qiuping Ye, Gary Lee, John Anzola, and James M Chen
- Subjects
Models, Molecular ,Protein Conformation ,Stereochemistry ,viruses ,Hepatitis C virus ,Allosteric regulation ,Viral Nonstructural Proteins ,Crystallography, X-Ray ,medicine.disease_cause ,Structure-Activity Relationship ,chemistry.chemical_compound ,RNA polymerase ,Drug Discovery ,medicine ,Binding site ,Mode of action ,NS5B ,Polymerase ,chemistry.chemical_classification ,Avian Myeloblastosis Virus ,Binding Sites ,Diarrhea Viruses, Bovine Viral ,biology ,Thiones ,virus diseases ,DNA-Directed RNA Polymerases ,biochemical phenomena, metabolism, and nutrition ,digestive system diseases ,Thiazoles ,Enzyme ,chemistry ,Biochemistry ,biology.protein ,Reverse Transcriptase Inhibitors ,Molecular Medicine ,Allosteric Site - Abstract
Novel non-nucleoside inhibitors of the HCV RNA polymerase (NS5b) with sub-micromolar biochemical potency have been identified which are selective for the inhibition of HCV NS5b over other polymerases. The structures of the complexes formed between several of these inhibitors and HCV NS5b were determined by X-ray crystallography, and the inhibitors were found to bind in an allosteric binding site separate from the active site. Structure-activity relationships and structural studies have identified the mechanism of action for compounds in this series, several of which possess drug-like properties, as unique, reversible, covalent inhibitors of HCV NS5b.
- Published
- 2006
48. Abstract 2640: Small-molecule inhibitors of CD73, CD39 and A2aR: Three anti-cancer targets in the ATP/adenosine signaling pathway
- Author
-
Manmohan Reddy Leleti, Wan Hsin Lim, Fang-Fang Yin, Annette Becker, Ulrike Schindler, Juan C. Jaen, Yu Chen, Joanne B. Tan, Steve Young, Jay P. Powers, Matt J. Walters, and Ada Chen
- Subjects
0301 basic medicine ,Granzyme B production ,Cancer Research ,Chemistry ,T cell ,Purinergic signalling ,Molecular biology ,Adenosine ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,ATP hydrolysis ,medicine ,cAMP-dependent pathway ,IL-2 receptor ,Signal transduction ,medicine.drug - Abstract
INTRODUCTION: In the tumor micro-environment (TME) Adenosine (ADO) dampens the immune response towards cancer cells by inhibiting the cytotoxic activity of effector cells and promoting the proliferation of immunosuppressive cells. Extracellular ADO is generated by the two ecto-nucleotidases CD39 (ATP→AMP) and CD73 (AMP→ADO). In immune cells ADO signals primarily through the G-protein coupled receptor A2aR. Inhibition of ADO generation and signaling have been shown to be promising therapeutic approaches for the treatment of cancer. METHODS: Potent and highly selective small molecule inhibitors for A2aR, CD73 and CD39 have been designed and synthesized by the Medicinal Chemistry Department at Arcus Biosciences. Enzymatic assays: CD39 and CD73 activity was assayed by quantitating hydrolysis of ATP or AMP, respectively. Inorganic phosphate was detected using a colorimetric (malachite green) protocol. ADO receptor assays: The potency of A2aR antagonists and selectivity towards related receptors was determined by measuring cAMP elevation in stably-expressing CHO cells following NECA (ADO mimic) stimulation. CD8+ T cell assays: Activation, proliferation and effector function of CD8+ T cells were quantified following compound treatment in the presence and absence of ATP. Mixed Lymphocyte Reaction (MLR): Cytokine levels were determined in an MLR assay after compound treatment in the presence and absence of ATP. Tumor models: CT26 and B16F10 syngeneic tumor models were used to assess the therapeutic effect of the inhibitors. RESULTS: Potent and highly selective small molecules have been generated to block either ADO generation or ADO signaling. The therapeutic potential of these molecules was assessed in CD8+ T cell assays, MLR assays and in various tumor models. CD73 inhibition blocked the conversion of AMP to ADO. CD39 inhibition blocked the conversion of ATP to AMP and A2aR inhibition abolished the increases in intracellular cAMP following ADO stimulation. The compounds are highly selective relative to related enzymes/receptors, as well as a large panel of unrelated targets. Inhibition of any one of the three targets robustly reversed adenosine-mediated inhibition of proliferation, CD25 expression, and IFN-γ and granzyme B production by human CD8+ T-cells. Robust inhibition of tumor growth in combination with anti-PD1 antibody was observed for several of these compounds. CONCLUSIONS: Highly potent and selective inhibitors of each of the 3 molecular targets involved in the ATP/adenosine pathway have been identified. Their ability to interfere with the extracellular generation of ADO and the immune-suppressive effects of ADO, as well as their effects on experimental tumor biology, have been demonstrated in various in vitro and in vivo models. Citation Format: Ulrike Schindler, Joanne B. Tan, Matt Walters, Annette Becker, Fangfang Yin, Ada Chen, Yu Chen, Wan Hsin Lim, Steve Young, Manmohan Leleti, Jay Powers, Juan C. Jaen. Small-molecule inhibitors of CD73, CD39 and A2aR: Three anti-cancer targets in the ATP/adenosine signaling pathway [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2640. doi:10.1158/1538-7445.AM2017-2640
- Published
- 2017
49. Abstract 4572: Characterization of the potent and selective A2aR antagonist AB928 for the treatment of cancer
- Author
-
Manmohan Reddy Leleti, Annette Becker, Matt J. Walters, Juan C. Jaen, Fangfang Yi, Laurent Debien, Joanne B. Tan, Tim Park, Brandon Reid Rosen, Stefan Garrido-Shaqfeh, Jay P. Powers, Steve Young, Ehesan U. Sharif, and Wan Hsin Lim
- Subjects
0301 basic medicine ,Agonist ,Cancer Research ,Forskolin ,biology ,medicine.drug_class ,CD3 ,Antagonist ,CD28 ,Pharmacology ,Adenosine ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,IL-2 receptor ,Receptor ,medicine.drug - Abstract
Introduction: In the tumor micro-environment, extracellular ATP is sequentially hydrolyzed to adenosine by the ecto-nucleotidases CD39 (ATP→AMP) and CD73 (AMP→adenosine). Adenosine, through activation of the A2a receptor (A2aR), is a potent inhibitor of T-cell activation, resulting in an immunosuppressed phenotype. Thus, inhibition of A2aR has recently generated great interest in immuno-oncology. We present the characterization of a novel, selective, and highly potent small molecule antagonist of A2aR which is slated to enter the clinic in 2017. Methods: The cellular potency of A2aR antagonists was assessed as a function of decreased cAMP levels in CHO cells stably over-expressing hA2aR, a Gs coupled receptor, following stimulation with the agonist NECA. Experiments were conducted in the presence and absence of human serum. Selectivity against the Gi-coupled receptor A1R was assessed similarly as a function of cAMP elevation in CHO cells stably expressing hA1R, following pretreatment with forskolin and stimulation with NECA. The ability of AB928 to reverse adenosine-mediated immune suppression (25 μM) of human or mouse CD8+ T-cells was determined using standard CD3/CD28 activation conditions. CD25 expression and cytokine release were measured by flow cytometry and ELISA, respectively. The pharmacokinetic characteristics of AB928 were assessed in rodent and non-rodent species to facilitate calculation of a projected human dose. Results: AB928 represents a novel series of potent and selective compounds designed to inhibit adenosine-mediated A2aR activation. This molecule is different from most known A2aR antagonists in that it does not cross the blood brain barrier. AB928 inhibited NECA-mediated A2aR activation with a potency of Conclusions: AB928 is a potent, selective and peripherally restricted antagonist of the A2aR receptor which is slated to enter clinical development in 2017. Citation Format: Matt J. Walters, Joanne B. Tan, Annette Becker, Fangfang Yi, Tim Park, Manmohan R. Leleti, Brandon Rosen, Ehesan Sharif, Laurent Debien, Steve Young, Wan Hsin Lim, Stefan Garrido-Shaqfeh, Juan C. Jaen, Jay P. Powers. Characterization of the potent and selective A2aR antagonist AB928 for the treatment of cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4572. doi:10.1158/1538-7445.AM2017-4572
- Published
- 2017
50. Abstract B46: Small-molecule inhibitors of ecto-nucleotidase CD73 promote activation of human CD8+ T cells and have profound effects on tumor growth and immune parameters in experimental tumor models
- Author
-
Joanne Bl Tan, Steve Young, Erick Allen Lindsey, Juan C. Jaen, Jay P. Powers, Ada Chen, Manmohan Reddy Leleti, Annette Becker, Jaroslaw Kalisiak, and Ulrike Schindler
- Subjects
Adenosine monophosphate ,Granzyme B production ,Cancer Research ,T cell ,Immunology ,Biology ,Molecular biology ,Granzyme B ,chemistry.chemical_compound ,medicine.anatomical_structure ,Biochemistry ,chemistry ,medicine ,Interleukin 12 ,Cytotoxic T cell ,IL-2 receptor ,CD8 - Abstract
Introduction: The intra-tumoral generation of adenosine (ADO), a potent inhibitor of T-cell activation, depends on the coordinated and sequential cleavage of extracellular adenosine triphosphate (ATP) by the ecto-nucleotidases CD39 (which produces adenosine monophosphate, AMP) and CD73 (which hydrolyzes AMP to form ADO). For this reason, a number of anti-CD73 antibodies are being advanced into clinical trials; however, to date there have been few reports of potent, selective, small-molecule CD73 inhibitors, such as those described here. Methods: Ecto-nucleotidase activity was calculated using the Malachite green assay after 50-min incubation with 25µM AMP, in the presence of varying concentrations of test compound(s). The following systems were used. Endogenous expression: hCD73/SKOV-3 cells; hCD73/CD8 T cells. Stable over-expression: hCD73/CHO. Transient expression: mCD73/CHO; NTPDase2/CHO; NTPDase3/CHO; NTPDase8/CHO. Human CD8 T cells enriched from buffy coats or leukopaks were pre-treated with varying concentrations of CD73 inhibitors prior to addition of 50 μM AMP + 10 μM EHNA and activated with T cell Activation/Expansion kit (Miltenyi). In some experiments, exogenous recombinant human IL12p70 (1-10 ng/mL) was added to the culture. Activation (CD25) and effector functions (Granzyme B and IFNγ) were measured by flow cytometry. CT26 cells were implanted into the shaved right flank of 7-8 week old Balb/c mice and measured three times a week starting at 7 days. Mice were enrolled into 4 cohorts and dosed according to the following conditions when tumour volume reached ~100 mm3. Group 1: 1% HPMC (sc/QD) + 2A3 (10 mg/kg; IP/Q3D) Group 2: A000830 (30 mg/kg; sc/QD) + 2A3 (10 mg/kg; IP/Q3D) Group 3: 1% HPMC (sc/QD) + RMP1-14 (10 mg/kg; IP/Q3D) Group 4: A000830 (30 mg/kg; sc/QD) + RMP1-14 (10 mg/kg; IP/Q3D) For interim analysis, single cell suspension was generated from tissues, blocked (clone 2.4G2), and stained with antibodies. For intracellular FOXP3 staining, samples were fixed and stained using FOXP3/Transcription Factor Staining Buffer Set. Non-specific blocking was performed with 20% normal rat serum prior to addition of anti-mouse FOXP3 antibody. Results: We have designed a series of potent and specific small-molecule inhibitors of human and mouse CD73, represented by A000830 and A001190 with the following IC50 values in overexpression systems: A000830: Mouse IC50 (1 nM); Human IC50 (3 nM) A001190: Mouse IC50 (n.d.); Human IC50 (0.03 nM) A000830 and A001190 also blocked AMP hydrolysis by freshly isolated human CD8 T cells at potencies comparable to the over-expression systems. In in vitro models of AMP/ADO-driven inhibition of human CD8+ T-cell activation, A000830 and A001190 showed robust rescue of CD25 expression and granzyme B production. Complete rescue of IFNγ; production was achieved by adding exogenous IL12. In vivo, A000830 was well-tolerated in mice, resulting in sustained plasma concentrations above IC90. Therapeutic dosing of A000830 to these mice in combination with an α-PD1 antibody resulted in robust CT26 tumor growth inhibition, greater than either treatment alone. Conclusions: Cumulatively, these data provide the initial characterization of a novel class of potent and selective small-molecule CD73 inhibitors that effectively block the generation of ADO from extracellular ATP, reverse the ADO-driven inhibition of human T-cell activation, and display promising anti-tumor activity when dosed in combination with PD-1 blockade. Citation Format: Joanne BL Tan, Ada Chen, Manmohan Leleti, Annette Becker, Erick Lindsey, Jaroslaw Kalisiak, Jay P. Powers, Steve Young, Ulrike Schindler, Juan C. Jaen. Small-molecule inhibitors of ecto-nucleotidase CD73 promote activation of human CD8+ T cells and have profound effects on tumor growth and immune parameters in experimental tumor models. [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 B46.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.