53 results on '"Ada Chen"'
Search Results
2. 583 Novel, potent, and selective inhibitors of hypoxia-inducible factor (HIF)-2α reverse pro-tumorigenic transcriptional programming in cancer, stromal, and immune cells
- Author
-
Matthew Walters, Stephen Young, Ada Chen, Akshata Udyavar, Kelsey Sivick Gauthier, Dana Piovesan, Kenneth Lawson, Soonweng Cho, Jean Chan, Jennie Au, Cesar Meleza, Xiaoning Zhao, Anh Tran, Samuel Drew, Balint Gal, Brandon Rosen, Manmohan Leleti, Elaine Ginn, Lixia Jin, and Jay Powers
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
3. 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
- Full Text
- View/download PDF
4. Supplemental Figure 2 from The MDM2 Inhibitor AMG 232 Demonstrates Robust Antitumor Efficacy and Potentiates the Activity of p53-Inducing Cytotoxic Agents
- Author
-
Robert Radinsky, Angela Coxon, Jonathan D. Oliner, Richard Kendall, Stephen Kaufman, David Cordover, Jing Zhou, Ada Chen, Lixia Jin, Qiuping Ye, John Eksterowicz, Dongyin Yu, Rebecca Robertson, Anne Y. Saiki, Steven H. Olson, Tao Osgood, and Jude Canon
- Abstract
Supplemental Figure 2. AMG 232 treatment causes p53 stabilization, and increased p21 and MDM2 proteins in p53 wild-type cells.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplemental Figure 3 from The MDM2 Inhibitor AMG 232 Demonstrates Robust Antitumor Efficacy and Potentiates the Activity of p53-Inducing Cytotoxic Agents
- Author
-
Robert Radinsky, Angela Coxon, Jonathan D. Oliner, Richard Kendall, Stephen Kaufman, David Cordover, Jing Zhou, Ada Chen, Lixia Jin, Qiuping Ye, John Eksterowicz, Dongyin Yu, Rebecca Robertson, Anne Y. Saiki, Steven H. Olson, Tao Osgood, and Jude Canon
- Abstract
Supplemental Figure 3. Body weights of mice in xenograft efficacy studies
- Published
- 2023
- Full Text
- View/download PDF
6. Supplemental Figure 1 from The MDM2 Inhibitor AMG 232 Demonstrates Robust Antitumor Efficacy and Potentiates the Activity of p53-Inducing Cytotoxic Agents
- Author
-
Robert Radinsky, Angela Coxon, Jonathan D. Oliner, Richard Kendall, Stephen Kaufman, David Cordover, Jing Zhou, Ada Chen, Lixia Jin, Qiuping Ye, John Eksterowicz, Dongyin Yu, Rebecca Robertson, Anne Y. Saiki, Steven H. Olson, Tao Osgood, and Jude Canon
- Abstract
Supplemental Figure 1. AMG 232 chemical structure, and 3-D model of AMG 232 bound to MDM2 protein.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figures 1 - 2, Table 1 from Preclinical Evaluation of AMG 925, a FLT3/CDK4 Dual Kinase Inhibitor for Treating Acute Myeloid Leukemia
- Author
-
Kang Dai, Dineli Wickramasinghe, Alexander Kamb, Julio Medina, Lawrence R. McGee, Christophe Quéva, Timothy Carlson, Lily Liu, Ada Chen, Rachel Ngo, Sharon Zhao, Mei-Chu Lo, Jessica Orf, Grace Alba, Xianghong Wang, Justin Huard, Margaret Weidner, Lingming Liang, John Eksterowicz, David Hollenback, Suzanne Coberly, Mark Ragains, Ji Ma, Zhihong Li, Cong Li, and Kathleen Keegan
- Abstract
PDF file - 132KB, Figure S1. Plasma concentration-time profiles of AMG 925 in rat, dog, and cynomolgus monkey following oral administration. Figure S2. Inhibition of STAT5 and Rb phosphorylation by AMG 925 in MOLM13-Luc tumor cells in bone marrow. Table S1. Selectivity profile of AMG 925 (Kd for kinases with POC < 20 at 1 microM in KenomScan).
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary 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
Supplementary Data from Targeting CD73 with AB680 (Quemliclustat), a Novel and Potent Small-Molecule CD73 Inhibitor, Restores Immune Functionality and Facilitates Antitumor Immunity
- Published
- 2023
- Full Text
- View/download PDF
9. 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
- Full Text
- View/download PDF
10. 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
- Full Text
- View/download PDF
11. 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
- Full Text
- View/download PDF
12. 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
- Full Text
- View/download PDF
13. 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
- Full Text
- View/download PDF
14. 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
- Full Text
- View/download PDF
15. 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
- Full Text
- View/download PDF
16. Safeguard gaps and their managerial issues.
- Author
-
Rua-Huan Tsaih, Wanying Lin, and Ada Chen
- Published
- 2008
- Full Text
- View/download PDF
17. 258 AB308 is an anti-TIGIT antibody that enhances immune activation and anti-tumor immunity alone and in combination with other I-O therapeutic agents
- Author
-
Hema Singh, Kelsey Sivick Gauthier, Ferdie Soriano, Soonweng Cho, Xiaoning Zhao, Nigel Walker, Alejandra Lopez, Patrick Schweickert, Matthew Walters, Ada Chen, Dana Piovesan, and Stephen P. Young
- Subjects
Pharmacology ,Cancer Research ,biology ,Chemistry ,T cell ,CD226 ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Jurkat cells ,medicine.anatomical_structure ,Immune system ,Oncology ,TIGIT ,medicine ,biology.protein ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,CD155 ,Antibody ,CD8 ,RC254-282 - Abstract
BackgroundTIGIT (T-cell immunoreceptor with Ig and ITIM domains) is an inhibitory receptor expressed on natural killer (NK) cells, CD8< sup >+ T cells, CD4< sup >+ T cells and regulatory T cells (T < sub >regs). On the surface of these cells, TIGIT competes with another receptor, CD226, for shared receptor ligands (mainly CD155) that are expressed by cancer and antigen-presenting cells. Binding of CD155 to TIGIT results in immune suppression through multiple mechanisms. When TIGIT is blocked, binding of CD155 to CD226 promotes immune activation and anti-tumor immunity. We describe the preclinical characterization of AB308, a humanized wild-type IgG1 anti-TIGIT antibody that is currently undergoing clinical evaluation.MethodsBinding of AB308 to TIGIT and inhibition of the TIGIT/CD155 interaction were evaluated < i >in vitro. Functional assays were used to evaluate the immunomodulatory activity of AB308 alone or in combination with zimberelimab (anti-PD-1) or etrumadenant (a small molecule A< sub >2aA< sub >2b adenosine receptor antagonist). Surrogate Fc-silent and Fc-enabled antibodies that recognize mouse TIGIT or PD-1 were leveraged to interrogate TIGIT biology in syngeneic mouse tumor models.ResultsHuman tumor-infiltrating lymphocytes from a variety of cancer types expressed appreciable levels of TIGIT on relevant immune populations, including tumor reactive CD39< sup >+CD103< sup >+ CD8< sup >+ T cells and T< sub >regs. AB308 has a high binding affinity for human TIGIT, potently blocks the TIGIT-CD155 interaction, and induces Fcγ receptor (FcγR)-mediated signaling. In line with FcγRIII binding, AB308 also demonstrated the ability to induce NK cell-driven antibody-dependent cell-mediated cytotoxicity against TIGIT-expressing target cells. AB308 significantly increased IL-2 secretion by peripheral blood mononuclear cells activated with superantigen A, an activity that was further enhanced with zimberelimab. Blocking TIGIT with AB308 potently activated CD226 signaling in Jurkat T cells co-cultured with CD155-expressing cells, and combination of AB308 with etrumadenant in this system abrogated adenosine-mediated T cell suppression that occurred even in the presence of checkpoint inhibition. In mice, while combining Fc-silent or Fc-enabled anti-mouse TIGIT antibody with anti-PD-1 resulted in greater tumor growth inhibition than with anti-PD-1 alone, the activity of Fc-enabled anti-TIGIT was associated with intratumoral T< sub >regsdepletion.ConclusionsAB308 is a potent and highly effective anti-TIGIT antibody. Concurrent blockade of multiple immune checkpoints has the potential to confer effective and durable responses in the treatment of cancer. The data presented here support the clinical use of AB308 and provides a rationale for combination with zimberelimab and adenosine pathway blocking agents such as etrumadenant and CD73 small molecule inhibitor, AB680.Ethics ApprovalAnimal experiments were performed at Arcus Biosciences, Inc. in accordance with federal, state and Institutional guidelines and were approved by Arcus’ Institutional Animal Care and Use Committee.
- Published
- 2021
18. Evaluating the ICF Framework as a Model of Community Participation for People with Disabilities
- Author
-
Ada Chen
- Subjects
Rehabilitation ,Physical Therapy, Sports Therapy and Rehabilitation - Published
- 2022
- Full Text
- View/download PDF
19. Abstract 321: Inhibition of CD39 results in elevated ATP and activation of myeloid cells to promote anti-tumor immunity
- Author
-
Christine E. Bowman, Ada Chen, Damie Juat, Kaustubh Parashar, Julie Clor, Hema Singh, Ritu Kushwaha, Bryan Handlos, Suan Liu, Janine Kline, Xiaoning Zhao, Hyock Joo Kwon, David Green, Stephen W. Young, Ester Fernandez-Salas, Matthew J. Walters, and Nigel P. Walker
- Subjects
Cancer Research ,Oncology - Abstract
CD39 (ENTPD1) is an ecto-nucleoside triphosphate diphosphohydrolase expressed widely in the tumor microenvironment (TME) responsible for catalyzing the conversion of ATP to AMP. Inhibition of CD39 enzymatic activity can promote anti-tumor immune responses by increasing the immunostimulatory substrate ATP and decreasing the formation of the product AMP, a precursor to immunoinhibitory adenosine. CD39 inhibition has been shown to have an anti-tumor effect by activating dendritic cells, macrophages, and NK cells in the TME to promote antigen presentation and inflammatory cytokine release. AB598 is a novel antibody, highly specific for human and cynomolgus CD39. AB598 potently binds to CD39 expressed on human primary myeloid cells and tumor cells and potently inhibits both soluble and membrane-bound CD39 enzymatic activity. CD39 inhibition results in increased extracellular ATP (eATP) in the TME and subsequent activation of P2X and P2Y receptors. Myeloid cells highly express both CD39 and P2X7, the P2X receptor with the weakest ligand affinity for ATP (greater than 100 μM), and most likely to be specifically activated by very high levels of eATP resulting from CD39 inhibition. Thus, the myeloid compartment has emerged as a key target associated with the therapeutic effects of CD39 inhibition. AB598 retains the ability to bind and inhibit membrane bound CD39 in the presence of high ATP (400 μM). AB598 promotes maturation of human dendritic cells in the presence of ATP, as determined by increased expression of CD83 and CD86 and decreased expression of CD14. In human macrophages treated with ATP, AB598 activates the inflammasome, resulting in the secretion of pro-inflammatory IL-18 and IL-1β. The ATP-dependent effects of AB598 support a therapeutic rationale of combining it with immunogenic therapies that induce release of ATP, such as chemotherapy and radiation, which might represent a new paradigm in the treatment of advanced solid tumors. Citation Format: Christine E. Bowman, Ada Chen, Damie Juat, Kaustubh Parashar, Julie Clor, Hema Singh, Ritu Kushwaha, Bryan Handlos, Suan Liu, Janine Kline, Xiaoning Zhao, Hyock Joo Kwon, David Green, Stephen W. Young, Ester Fernandez-Salas, Matthew J. Walters, Nigel P. Walker. Inhibition of CD39 results in elevated ATP and activation of myeloid cells to promote anti-tumor immunity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 321.
- Published
- 2022
- Full Text
- View/download PDF
20. Anti-TIGIT antibodies promote immune activation relevant to targeting stem-like and tumor-specific T cells in combination with anti-PD-1
- Author
-
Kelsey E Sivick Gauthier, Dana Piovesan, Amber E de Groot, Gabrielle L Reiner, Patrick G Schweickert, Ferdie Soriano, Ada Chen, Hema Singh, Xiaoning Zhao, Lisa Seitz, Anita Reddy, Stephen W Young, Nigel Walker, and Matthew J Walters
- Subjects
Immunology ,Immunology and Allergy - Abstract
TIGIT is an inhibitory receptor expressed primarily on NK and T cell subsets, and binding to its cognate receptor ligand, CD155, results in multiple mechanisms of immunosuppression. Blocking the TIGIT-CD155 interaction in the context of cancer promotes anti-tumor immunity. We characterized cellular subsets that TIGIT blockade may impact and the pharmacology of two anti-TIGIT antibodies - representing functional (AB308) and non-functional (AB154) Fc domain classes - undergoing clinical evaluation. Surrogate antibodies were leveraged to interrogate TIGIT biology in mouse syngeneic tumor models. Human tumor-infiltrating lymphocytes from a variety of cancer types expressed appreciable levels of TIGIT on relevant immune populations, including Tregs and CD8+ T cells with tumor reactive or pre-dysfunctional stem-like phenotypes. Both antibodies potently bound TIGIT and blocked the TIGIT-CD155 interaction as well as displayed the predicted phenotypes in terms of Fcγ receptor (FcγR) engagement. In line with FcγRIII binding, AB308 demonstrated a capacity to induce ADCC against TIGIT-expressing target cells. Combination of anti-TIGIT antibodies with other therapeutic approaches that promote T cell activation resulted in enhanced immune responses. In mice, while combining Fc-silent or Fc-enabled anti-mouse TIGIT antibody with anti-PD-1 resulted in greater tumor growth inhibition than with anti-PD-1 alone, the activity of Fc-enabled anti-TIGIT was associated with intratumoral Treg depletion. These data provide a rationale for combination with immune-activating agents and support ongoing clinical evaluation of AB154 and AB308 with biomarker strategies focused on understanding the role of Fc functionality.
- Published
- 2022
- Full Text
- View/download PDF
21. 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
- Full Text
- View/download PDF
22. Measuring dispositional optimism in student Veterans: An item response theory analysis
- Author
-
Emre Umucu, Fong Chan, Jia Rung Wu, Beatrice Lee, Kanako Iwanaga, Jessica M. Brooks, and Ada Chen
- Subjects
050103 clinical psychology ,Psychometrics ,media_common.quotation_subject ,education ,05 social sciences ,Concurrent validity ,050109 social psychology ,Experimental and Cognitive Psychology ,humanities ,Exploratory factor analysis ,Classical test theory ,Test (assessment) ,Optimism ,Item response theory ,0501 psychology and cognitive sciences ,Psychology ,General Psychology ,Social Sciences (miscellaneous) ,Reliability (statistics) ,media_common ,Clinical psychology - Abstract
Background: The psychometric properties of the Life Orientation Test-Revised (LOT-R) have been established among college students, yet psychometric evidence is lacking for a sample of student Veterans in postsecondary education.Aims: The purpose of this study was to evaluate psychometric properties of the LOT-R for the assessment of dispositional optimism in student Veterans by using classical test theory (CTT) in conjunction with item response theory (IRT).Method: A sample of 205 student Veterans were recruited from universities across the United States. Exploratory factor analysis was conducted to test the unidimensionality of the LOT-R. A polychotomous IRT model using graded response model (GRM) was estimated. Reliability and concurrent validity of the LOT-R were tested.Results: CTT in conjunction with IRT validated that the LOT-R is a psychometrically sound unidimensional instrument for assessing the levels of dispositional optimism in student Veterans. The LOT-R was found to be associated wit...
- Published
- 2018
- Full Text
- View/download PDF
23. Design, Synthesis, and Structure-Activity Relationship Optimization of Pyrazolopyrimidine Amide Inhibitors of Phosphoinositide 3-Kinase γ (PI3Kγ).
- Author
-
Mata, Guillaume, Miles, Dillon H., Drew, Samuel L., Fournier, Jeremy, Lawson, Kenneth V., Mailyan, Artur K., Sharif, Ehesan U., Xuelei Yan, Beatty, Joel W., Banuelos, Jesus, Jie Chen, Ginn, Elaine, Ada Chen, Gerrick, Kimberline Y., Pham, Amber T., Wong, Kent, Soni, Divyank, Dhanota, Puja, Shaqfeh, Stefan G., and Meleza, Cesar
- Published
- 2022
- Full Text
- View/download PDF
24. 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
- Full Text
- View/download PDF
25. Discovery of AM-7209, a Potent and Selective 4-Amidobenzoic Acid Inhibitor of the MDM2–p53 Interaction
- Author
-
Jason Duquette, Xin Huang, Dongyin Yu, Yun Ling, Jing Zhou, Yu Chung Wang, Daqing Sun, Sarah Wortman, John Eksterowicz, Qiuping Ye, Min Jiang, Jonathan D. Oliner, Lixia Jin, Alexander M. Long, Ana Z. Gonzalez, Peter Yakowec, Yihong Li, Steven H. Olson, Anne Y. Saiki, Hilary Plake Beck, Yosup Rew, Lawrence R. McGee, Julio C. Medina, Jonathan B. Houze, Jiasheng Fu, Jude Canon, Mcintosh Joel, Ada Chen, Brian M. Fox, Mei-Chu Lo, Xuelei Yan, Paul L. Shaffer, Zhihong Li, Tao Osgood, and Xiaoning Zhao
- Subjects
Models, Molecular ,Carboxylic acid ,Mice, Nude ,Antineoplastic Agents ,Pharmacology ,Mice ,Structure-Activity Relationship ,Pharmacokinetics ,In vivo ,Drug Discovery ,Tumor Cells, Cultured ,Animals ,Humans ,Potency ,Structure–activity relationship ,IC50 ,Cell Proliferation ,chemistry.chemical_classification ,Molecular Structure ,Drug discovery ,Rational design ,Proto-Oncogene Proteins c-mdm2 ,chemistry ,Colonic Neoplasms ,Molecular Medicine ,Female ,Tumor Suppressor Protein p53 ,Protein Binding - Abstract
Structure-based rational design and extensive structure-activity relationship studies led to the discovery of AMG 232 (1), a potent piperidinone inhibitor of the MDM2-p53 association, which is currently being evaluated in human clinical trials for the treatment of cancer. Further modifications of 1, including replacing the carboxylic acid with a 4-amidobenzoic acid, afforded AM-7209 (25), featuring improved potency (KD from ITC competition was 38 pM, SJSA-1 EdU IC50 = 1.6 nM), remarkable pharmacokinetic properties, and in vivo antitumor activity in both the SJSA-1 osteosarcoma xenograft model (ED50 = 2.6 mg/kg QD) and the HCT-116 colorectal carcinoma xenograft model (ED50 = 10 mg/kg QD). In addition, 25 possesses distinct mechanisms of elimination compared to 1.
- Published
- 2014
- Full Text
- View/download PDF
26. Optimization beyond AMG 232: Discovery and SAR of sulfonamides on a piperidinone scaffold as potent inhibitors of the MDM2-p53 protein–protein interaction
- Author
-
Lawrence R. McGee, Jeffrey T. Mihalic, Yingcai Wang, Mei-Chu Lo, Jing Zhou, Steven H. Olson, Xiaoqi Chen, Frank Kayser, Jiang Zhu, Ada Chen, Jeffrey Deignan, Jonathan D. Oliner, Alexander M. Long, Daqing Sun, Ming Yu, Xin Huang, Qiuping Ye, Jiwen Jim Liu, Peter Yakowec, and Julio C. Medina
- Subjects
Models, Molecular ,Scaffold ,Clinical Biochemistry ,Molecular Conformation ,Pharmaceutical Science ,Acetates ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Protein–protein interaction ,Structure-Activity Relationship ,Drug Discovery ,medicine ,Animals ,Humans ,Mdm2 p53 ,Molecular Biology ,Piperidones ,Sulfonamides ,Dose-Response Relationship, Drug ,Chemistry ,Organic Chemistry ,Sulfonamide (medicine) ,Proto-Oncogene Proteins c-mdm2 ,Rats ,Molecular Medicine ,Drug Screening Assays, Antitumor ,Tumor Suppressor Protein p53 ,Protein Binding ,medicine.drug - Abstract
We recently reported on the discovery of AMG 232, a potent and selective piperidinone inhibitor of the MDM2-p53 interaction. AMG 232 is being evaluated in human clinical trials for cancer. Continued exploration of the N-alkyl substituent of this series, in an effort to optimize interactions with the MDM2 glycine-58 shelf region, led to the discovery of sulfonamides such as compounds 31 and 38 that have similar potency, hepatocyte stability and rat pharmacokinetic properties to AMG 232.
- Published
- 2014
- Full Text
- View/download PDF
27. Discovery of Potent and Simplified Piperidinone-Based Inhibitors of the MDM2–p53 Interaction
- Author
-
Jiang Zhu, Tao Osgood, Xin Huang, Anne Y. Saiki, John Eksterowicz, Jiwen Jim Liu, Yosup Rew, Xiaoning Zhao, Qiuping Ye, Steven H. Olson, Julio C. Medina, Michael W. Gribble, David Chow, Jiasheng Fu, Ming Yu, Zhihong Li, Daqing Sun, Jude Canon, Dustin McMinn, Paul L. Shaffer, Xuelei Yan, Yingcai Wang, Brian M. Fox, Mei-Chu Lo, Jonathan D. Oliner, Dongyin Yu, Ada Chen, Michael D. Bartberger, and Jing Zhou
- Subjects
Antitumor activity ,chemistry.chemical_compound ,Pharmacokinetics ,Chemistry ,Stereochemistry ,In vivo ,Organic Chemistry ,Drug Discovery ,Substituent ,Mdm2 p53 ,Biochemistry ,Combinatorial chemistry - Abstract
Continued optimization of the N-substituent in the piperidinone series provided potent piperidinone-pyridine inhibitors 6, 7, 14, and 15 with improved pharmacokinetic properties in rats. Reducing structure complexity of the N-alkyl substituent led to the discovery of 23, a potent and simplified inhibitor of MDM2. Compound 23 exhibits excellent pharmacokinetic properties and substantial in vivo antitumor activity in the SJSA-1 osteosarcoma xenograft mouse model.
- Published
- 2014
- Full Text
- View/download PDF
28. Selective and Potent Morpholinone Inhibitors of the MDM2–p53 Protein–Protein Interaction
- Author
-
Ada Chen, Steven H. Olson, Alexander M. Long, John Eksterowicz, Michael D. Bartberger, Jing Zhou, Yosup Rew, Lawrence R. McGee, Mei-Chu Lo, Anne Y. Saiki, Dongyin Yu, Jason Duquette, Xuelei Yan, Ana Z. Gonzalez, Hilary Plake Beck, Jonathan B. Houze, Yun Ling, Mcintosh Joel, Jonathan D. Oliner, Sarah Wortman, Jude Canon, Daqing Sun, Dustin McMinn, Brian M. Fox, Paul L. Shaffer, Xiaoning Zhao, Tao Osgood, Lixia Jin, Xin Huang, Zhihong Li, David Chow, Qiuping Ye, Yihong Li, Jiasheng Fu, Peter Yakowec, and Julio C. Medina
- Subjects
Models, Molecular ,Stereochemistry ,Morpholines ,Molecular Conformation ,Antineoplastic Agents ,Acetates ,Crystallography, X-Ray ,Protein–protein interaction ,Mice ,Structure-Activity Relationship ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Potency ,Structure–activity relationship ,Mdm2 p53 ,IC50 ,biology ,Chemistry ,Drug discovery ,Proto-Oncogene Proteins c-mdm2 ,Xenograft Model Antitumor Assays ,Rats ,Cell culture ,biology.protein ,Molecular Medicine ,Mdm2 ,Indicators and Reagents ,Tumor Suppressor Protein p53 - Abstract
We previously reported the discovery of AMG 232, a highly potent and selective piperidinone inhibitor of the MDM2-p53 interaction. Our continued search for potent and diverse analogues led to the discovery of novel morpholinone MDM2 inhibitors. This change to a morpholinone core has a significant impact on both potency and metabolic stability compared to the piperidinone series. Within this morpholinone series, AM-8735 emerged as an inhibitor with remarkable biochemical potency (HTRF IC50 = 0.4 nM) and cellular potency (SJSA-1 EdU IC50 = 25 nM), as well as pharmacokinetic properties. Compound 4 also shows excellent antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 41 mg/kg. Lead optimization toward the discovery of this inhibitor as well as key differences between the morpholinone and the piperidinone series will be described herein.
- Published
- 2014
- Full Text
- View/download PDF
29. 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
- Full Text
- View/download PDF
30. 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
- Full Text
- View/download PDF
31. Assessment of the Integrity of Compounds Stored in Assay-Ready Plates Using a Kinase Sentinel Assay
- Author
-
Shou-Hua Xiao, Cheng Su, Yichin Liu, John D. McCarter, Leeanne Zalameda, Thim Oung, Son Dang, Ada Chen, Xiaoning Zhao, Stephen W. Young, Xiaoyang Xia, Amanda Lembke, Heather Eastwood, and Laina Mercer
- Subjects
Chromatography ,Chemistry ,Drug Storage ,Organic Chemistry ,Analytical chemistry ,Compound management ,Fraction (chemistry) ,General Medicine ,Mass Spectrometry ,Lower temperature ,Computer Science Applications ,Solvent ,Drug Stability ,Drug Discovery ,Solvents ,Potency ,Dimethyl Sulfoxide ,A kinase ,Chromatography, Liquid - Abstract
Sentinel assays are a convenient adjunct to LC-MS purity assessment to monitor the integrity of compounds in pharmaceutical screening collections over time. To assess the stability of compounds stored both at room temperature and at -20°C in assay-ready plates that were either vacuum pack-sealed using a convenient industrial vacuum sealing method or individually sealed using conventional foil seals, a diverse collection of#126; 5,000 compounds was assayed using a robust biochemical kinase assay at intervals over a one year period. Assay results at each time point were compared to those of initial assay using a series of correlations of compound Percent of Control (POC) values as well as IC50 values of a subset of compounds in 200 nL or 500 nL plates. The fraction of hits in common between initial assays and assays at later time points ranged from 82% to 95% over one year and remained relatively constant over time with all storage temperatures or sealing methods tested. A majority of the hits that exhibited a consistent gradual trend to lower potency over one year storage were shifted to lower potency upon the rapid removal of DMSO solvent. Compound precipitation rather than compound decomposition is the likely reason for trends to lower potency for most such compounds over the storage period. Plates stored at room temperature featured a significantly higher fraction of hits that exhibited a trend to lower apparent potency than those stored at -20°C suggesting that this lower temperature is preferable for longer-term storage.
- Published
- 2013
- Full Text
- View/download PDF
32. 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
- Full Text
- View/download PDF
33. Abstract 1756: Discovery and characterization of AB680, a potent and selective small-molecule CD73 inhibitor for cancer immunotherapy
- Author
-
Matt J. Walters, Fang-Fang Yin, Debbie Swinarski, Jenna L. Jeffrey, Lixia Jin, Jarek Kalisiak, Ada Chen, Kenneth V. Lawson, Powers P. Jay, Steve Young, Ulrike Schindler, and Kristen Zhang
- Subjects
0301 basic medicine ,Cancer Research ,Chemistry ,medicine.medical_treatment ,T cell ,CD28 ,Pharmacology ,01 natural sciences ,Adenosine ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Immune system ,Oncology ,Cancer immunotherapy ,medicine ,IL-2 receptor ,Receptor ,IC50 ,medicine.drug - Abstract
Introduction. Extracellular adenosine (ADO) suppresses immune function via inhibition of T cell and NK cell activation and is present in high concentrations in the tumor micro-environment (TME). Intra-tumoral 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, non-redundant, pathway of ADO production in the TME and can reverse ADO-mediated immune suppression. Here we present the characterization of AB680, a novel, highly potent, reversible and selective small molecule inhibitor of CD73, currently in preclinical development as a potential anti-tumor agent. Methods. The potency of CD73 inhibitors was evaluated by measuring AMP hydrolysis by CHO-CD73 cells using a malachite green assay. Potency was also measured using human T-cells and soluble recombinant CD73. Selectivity against related ecto-nucleotidases was also assessed. In the presence of human serum, CD73 inhibition was measured by quantitation of AMP hydrolysis via luminescence. The ability of AB680 to reverse AMP-mediated immune suppression of human CD4+/CD8+ T cells was determined by adding exogenous AMP during T cell activation by anti-CD3/CD28/CD2 beads. The pharmacokinetic properties of AB680 were evaluated in multiple preclinical species. A projected human dosing schedule for AB680 was determined via allometric scaling. Results. AB680 is a highly potent, reversible and selective inhibitor of CD73 (IC50 < 0.01 nM on human CD8+ T-cells), which retains high potency in the presence of human serum. AB680 is > 10,000-fold selective against related ecto-nucleotidases and a large panel of unrelated enzymes, receptors, and ion channels. AB680 does not show significant inhibition of the major CYP450 isoforms or the hERG potassium channel. AB680 potently reverses AMP and ADO-mediated suppression of immune function in vitro. In the presence of high concentrations of AMP, AB680 robustly restored CD25 expression, IFN-γ production and proliferation of human CD4+ and CD8+ T-cells. The pharmacokinetics (PK) of AB680 were assessed in rodent and non-rodent species. The PK properties of AB680 are characterized by very low clearance and long half-lives in preclinical species, resulting in projected human PK properties suitable for intravenous (i.v.) dosing on a schedule consistent with typical mAb dosing cycles. High-dose infusions of AB680 in preclinical species were well tolerated. Conclusions. AB680 is a highly potent and selective small-molecule inhibitor of CD73 which can mitigate AMP and ADO-mediated tumor immunity by potently blocking the production of adenosine in the TME. AB680 exhibits a favorable projected human PK profile suitable for parental administration and is expected to enter clinical development in 2018. Citation Format: Kenneth V. Lawson, Lixia Jin, Jenna L. Jeffrey, Jarek Kalisiak, Fangfang Yin, Kristen Zhang, Ada Chen, Debbie Swinarski, Matt J. Walters, Steve Young, Ulrike Schindler, Powers P. Jay. Discovery and characterization of AB680, a potent and selective small-molecule CD73 inhibitor for cancer immunotherapy [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 1756.
- Published
- 2018
- Full Text
- View/download PDF
34. Safeguard gaps and their managerial issues
- Author
-
Wan-Ying Lin, Rua-Huan Tsaih, and Ada Chen
- Subjects
Value (ethics) ,Knowledge management ,Process (engineering) ,business.industry ,Business process ,Strategy and Management ,Data security ,Information security ,Industrial and Manufacturing Engineering ,Computer Science Applications ,Management Information Systems ,Safeguard ,Loan ,Industrial relations ,Business ,Marketing ,Information exchange - Abstract
PurposeTargeting the information security issue related to a bank loan process, this study aims to explore an approach that is user‐friendly to non‐information technology managers to be actively participative in the process of investigating the underlying information‐processing infrastructure.Design/methodology/approachA case study approach is used to analyze the safeguard gaps between the physical information assets (IAs) of a business process and their digital counterparts.FindingsThis study shows the existence of safeguard gaps between the physical IAs of a business process and their digital counterparts, explores a way of investigating such gaps, and provides (detection‐ and prevention‐oriented) managerial proposals to cope with the safeguard gap issue.Originality/valueThe existence of safeguard gaps releases (warning) signal to executives and executives should consult managers on how to manage these safeguard gaps. There are two types of managerial proposals to cope with the safeguard gap issue. The detection‐oriented proposals aim at ensuring early detection and interception of security breaches;, e.g. setting up a cross‐audit function to discover the security breaches. The prevention‐oriented proposals aim at alleviating the safeguard gaps;, e.g. re‐designing the processes of operation management and the associated risk management.
- Published
- 2008
- Full Text
- View/download PDF
35. Maternal immune activation alters placental histone-3 lysine-9 tri-methylation, offspring sensorimotor processing, and hypothalamic transposable element expression in a sex-specific manner
- Author
-
Holly DeRosa, Arianna Smith, Laurel Geist, Ada Cheng, Richard G. Hunter, and Amanda C. Kentner
- Subjects
Maternal immune activation ,Retrotransposons ,Transposable elements ,Epigenetics ,Placenta ,Prepulse inhibition ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 ,Neurophysiology and neuropsychology ,QP351-495 - Abstract
Animal models of maternal immune activation (MIA) are central to identifying the biological mechanisms that underly the association between prenatal infection and neuropsychiatric disorder susceptibility. Many studies, however, have limited their scope to protein coding genes and their role in mediating this inherent risk, while much less attention has been directed towards exploring the roles of the epigenome and transposable elements (TEs). In Experiment 1, we demonstrate the ability of MIA to alter the chromatin landscape of the placenta. We induced MIA by injecting 200 μg/kg (i.p.) of lipopolysaccharide (LPS) on gestational day 15 in Sprague-Dawley rats. We found a sex-specific rearrangement of heterochromatin 24-h after exposure to MIA, as evidenced by an increase in histone-3 lysine-9 trimethylation (H3K9me3). In Experiment 2, MIA was associated with long-term sensorimotor processing deficits as indicated by reduced prepulse inhibition (PPI) of the acoustic startle reflex in adult male and female offspring and an increased mechanical allodynia threshold in males. Analyses of gene expression within the hypothalamus-chosen for its involvement in the sex-specific pathogenesis of schizophrenia and the stress response-revealed significantly higher levels of the stress-sensitive genes Gr and Fkbp5. Deleterious TE expression is often a hallmark of neuropsychiatric disease and we found sex-specific increases in the expression of several TEs including IAP, B2 SINE, and LINE-1 ORF1. The data from this study warrant the future consideration of chromatin stability and TEs as part of the mechanism that drives MIA-associated changes in the brain and behavior.
- Published
- 2023
- Full Text
- View/download PDF
36. WET GROWTH: EFFECTS OF WATER POLICIES ON LAND USE IN THE AMERICAN WEST
- Author
-
Ada Chen and Ellen Hanak
- Subjects
Water conservation ,Land use ,Natural resource economics ,restrict ,Economics ,Population growth ,Environmental Science (miscellaneous) ,Development ,Water resource management ,American west ,Groundwater ,Externality ,Water trading - Abstract
Rapid population growth and increasing water development costs have prompted many western governments to condition residential development approval on the adequacy of water supplies. We examine the effects of these regulations on housing supply in Colorado and New Mexico using fixed-effects panel regressions. Our findings suggest that price-based tools to ensure water availability may be a preferred regulatory alternative to quantity restrictions. Attempts to restrict groundwater basin access have not unambiguously corrected negative externalities related to growth. Meanwhile, Colorado cities' aggressive use of impact fees has facilitated water resource development, without limiting growth.
- Published
- 2007
- Full Text
- View/download PDF
37. 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
- Full Text
- View/download PDF
38. 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
- Full Text
- View/download PDF
39. AMG 580: a novel small molecule phosphodiesterase 10A (PDE10A) positron emission tomography tracer
- Author
-
Hu Ye, Tim Kazules, Ji Ma, Dianna Lester-Zeiner, Essa Hu, Santosh Talreja, Carl Davis, Charlie Glaus, Oliver R. Thiel, Ning Chen, Geraldine Hill Della Puppa, James J. S. Treanor, Shannon Rumfelt, Hui Hannah Dou, Roxanne Kunz, David C. Immke, Jessica Able, Jennifer R. Allen, Silke Miller, Xiaoning Zhao, Hang Chen, Jianxia Shi, Jamie Wong, Toni Williamson, Christopher Biorn, Amy Porter, Klaus Michelsen, and Ada Chen
- Subjects
Male ,Fluorine Radioisotopes ,Phosphodiesterase Inhibitors ,Pharmacology ,Mass Spectrometry ,Rats, Sprague-Dawley ,Radioligand Assay ,Species Specificity ,medicine ,Distribution (pharmacology) ,Animals ,Humans ,Tissue Distribution ,medicine.diagnostic_test ,Molecular Structure ,Chemistry ,Drug discovery ,Phosphoric Diester Hydrolases ,Antagonist ,Phosphodiesterase ,Brain ,Stereoisomerism ,Surface Plasmon Resonance ,Small molecule ,Drug development ,Positron emission tomography ,Positron-Emission Tomography ,Pyrazines ,Molecular Medicine ,Benzimidazoles ,Female ,PDE10A ,Chromatography, Liquid ,Papio ,Protein Binding - Abstract
Phosphodiesterase 10A (PDE10A) inhibitors have therapeutic potential for the treatment of psychiatric and neurologic disorders, such as schizophrenia and Huntington's disease. One of the key requirements for successful central nervous system drug development is to demonstrate target coverage of therapeutic candidates in brain for lead optimization in the drug discovery phase and for assisting dose selection in clinical development. Therefore, we identified AMG 580 [1-(4-(3-(4-(1H-benzo[d]imidazole-2-carbonyl)phenoxy)pyrazin-2-yl)piperidin-1-yl)-2-fluoropropan-1-one], a novel, selective small-molecule antagonist with subnanomolar affinity for rat, primate, and human PDE10A. We showed that AMG 580 is suitable as a tracer for lead optimization to determine target coverage by novel PDE10A inhibitors using triple-stage quadrupole liquid chromatography-tandem mass spectrometry technology. [(3)H]AMG 580 bound with high affinity in a specific and saturable manner to both striatal homogenates and brain slices from rats, baboons, and human in vitro. Moreover, [(18)F]AMG 580 demonstrated prominent uptake by positron emission tomography in rats, suggesting that radiolabeled AMG 580 may be suitable for further development as a noninvasive radiotracer for target coverage measurements in clinical studies. These results indicate that AMG 580 is a potential imaging biomarker for mapping PDE10A distribution and ensuring target coverage by therapeutic PDE10A inhibitors in clinical studies.
- Published
- 2014
40. The MDM2 Inhibitor AMG 232 Demonstrates Robust Antitumor Efficacy and Potentiates the Activity of p53-Inducing Cytotoxic Agents
- Author
-
Tao Osgood, Steven H. Olson, Jonathan D. Oliner, Jude Canon, Robert Radinsky, John Eksterowicz, Richard Kendall, Qiuping Ye, Lixia Jin, Stephen Kaufman, Angela Coxon, Rebecca Robertson, David Cordover, Anne Y. Saiki, Ada Chen, Jing Zhou, and Dongyin Yu
- Subjects
Cancer Research ,DNA damage ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Pharmacology ,Acetates ,law.invention ,Mice ,law ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Cytotoxicity ,Piperidones ,Cell Proliferation ,biology ,Chemistry ,Cytotoxins ,Proto-Oncogene Proteins c-mdm2 ,Cell Cycle Checkpoints ,HCT116 Cells ,Xenograft Model Antitumor Assays ,Oncology ,Cell culture ,biology.protein ,MCF-7 Cells ,Mdm2 ,Suppressor ,Female ,Tumor Suppressor Protein p53 ,HT29 Cells ,MDM2 Inhibitor AMG-232 - Abstract
p53 is a critical tumor suppressor and is the most frequently inactivated gene in human cancer. Inhibition of the interaction of p53 with its negative regulator MDM2 represents a promising clinical strategy to treat p53 wild-type tumors. AMG 232 is a potential best-in-class inhibitor of the MDM2–p53 interaction and is currently in clinical trials. We characterized the activity of AMG 232 and its effect on p53 signaling in several preclinical tumor models. AMG 232 binds the MDM2 protein with picomolar affinity and robustly induces p53 activity, leading to cell-cycle arrest and inhibition of tumor cell proliferation. AMG 232 treatment inhibited the in vivo growth of several tumor xenografts and led to complete and durable regression of MDM2-amplified SJSA-1 tumors via growth arrest and induction of apoptosis. Therapeutic combination studies of AMG 232 with chemotherapies that induce DNA damage and p53 activity resulted in significantly superior antitumor efficacy and regression, and markedly increased activation of p53 signaling in tumors. These preclinical data support the further evaluation of AMG 232 in clinical trials as both a monotherapy and in combination with standard-of-care cytotoxics. Mol Cancer Ther; 14(3); 649–58. ©2015 AACR.
- Published
- 2014
41. Discovery of AMG 925, a FLT3 and CDK4 dual kinase inhibitor with preferential affinity for the activated state of FLT3
- Author
-
Mei-Chu Lo, Ji Ma, Julio C. Medina, Dineli Wickramasinghe, Mark L. Ragains, Derek E. Piper, Merrill Ayres, Zhihong Li, Alex J. Zhang, Xiaoqi Chen, Jeffrey Deignan, Zhulun Wang, Kanaka Pattabiraman, Kathleen S. Keegan, Xiaoning Zhao, Rachel Ngo, Nigel Walker, Lingming Liang, Margaret F. Weidner, Paul M. Wehn, Christophe Queva, Jason Duquette, Alexander Kamb, Ada Chen, Malgorzata Wanska, Jeffrey T. Mihalic, Kriti Modi, Pingchen Fan, Grace Q. Alba, Yunxiao Li, Michael W. Gribble, John Eksterowicz, Jacob Kaizerman, Dustin McMinn, Xianghong Wang, Michael DeGraffenreid, Steve Thibault, Xiaodong Wang, Kexue Li, Benjamin Fisher, Richard V. Connors, Wen Liu, Sarah E. Lively, Cong Li, Kang Dai, Jiasheng Fu, Justin N. Huard, Robert Cho, Lawrence R. McGee, Timothy J. Carlson, and Julia Suchomel
- Subjects
Programmed cell death ,Pyrimidine ,Cell ,chemistry.chemical_compound ,Structure-Activity Relationship ,Dogs ,Drug Discovery ,medicine ,Animals ,Cytochrome P-450 CYP3A ,Humans ,Naphthyridines ,Protein Kinase Inhibitors ,STAT5 ,biology ,Chemistry ,Kinase ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,U937 Cells ,Rats ,Macaca fascicularis ,medicine.anatomical_structure ,Biochemistry ,fms-Like Tyrosine Kinase 3 ,Cell culture ,biology.protein ,Molecular Medicine ,Cytochrome P-450 CYP3A Inhibitors ,Growth inhibition ,Lead compound ,Heterocyclic Compounds, 3-Ring - Abstract
We describe the structural optimization of a lead compound 1 that exhibits dual inhibitory activities against FLT3 and CDK4. A series of pyrido[4',3':4,5]pyrrolo[2,3-d]pyrimidine derivatives was synthesized, and SAR analysis, using cell-based assays, led to the discovery of 28 (AMG 925), a potent and orally bioavailable dual inhibitor of CDK4 and FLT3, including many FLT3 mutants reported to date. Compound 28 inhibits the proliferation of a panel of human tumor cell lines including Colo205 (Rb(+)) and U937 (FLT3(WT)) and induced cell death in MOLM13 (FLT3(ITD)) and even in MOLM13 (FLT3(ITD, D835Y)), which exhibits resistance to a number of FLT3 inhibitors currently under clinical development. At well-tolerated doses, compound 28 leads to significant growth inhibition of MOLM13 xenografts in nude mice, and the activity correlates with inhibition of STAT5 and Rb phosphorylation.
- Published
- 2014
42. Novel inhibitors of the MDM2-p53 interaction featuring hydrogen bond acceptors as carboxylic acid isosteres
- Author
-
Jing Zhou, Xiaoning Zhao, Yihong Li, Lixia Jin, David Chow, Lawrence R. McGee, Steven H. Olson, Paul L. Shaffer, Jonathan D. Oliner, Xin Huang, Ana Z. Gonzalez, Jonathan B. Houze, Yun Ling, Zhihong Li, Hilary Plake Beck, Ada Chen, Tao Osgood, Jason Duquette, Sarah Wortman, Dongyin Yu, Alexander M. Long, Xuelei Yan, Daqing Sun, Peter Yakowec, Yosup Rew, Brian M. Fox, Julio C. Medina, Qiuping Ye, Anne Y. Saiki, Jiasheng Fu, Mcintosh Joel, Jude Canon, Mei-Chu Lo, and John Eksterowicz
- Subjects
Models, Molecular ,Stereochemistry ,Carboxylic acid ,Myocytes, Smooth Muscle ,Carboxylic Acids ,Mice, Nude ,Stereoisomerism ,Antineoplastic Agents ,Bone Neoplasms ,Acetates ,Crystallography, X-Ray ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Drug Discovery ,Pyridine ,Tumor Cells, Cultured ,Structure–activity relationship ,Moiety ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Thiazole ,IC50 ,Cells, Cultured ,Piperidones ,Cell Proliferation ,chemistry.chemical_classification ,Osteosarcoma ,Molecular Structure ,Hydrogen bond ,Hydrogen Bonding ,Proto-Oncogene Proteins c-mdm2 ,Xenograft Model Antitumor Assays ,chemistry ,Drug Design ,Molecular Medicine ,Female ,Tumor Suppressor Protein p53 ,Protein Binding - Abstract
We previously reported the discovery of potent and selective morpholinone and piperidinone inhibitors of the MDM2-p53 interaction. These inhibitors have in common a carboxylic acid moiety that engages in an electrostatic interaction with MDM2-His96. Our continued search for potent and diverse inhibitors led to the discovery of novel replacements for these acids uncovering new interactions with the MDM2 protein. In particular, using pyridine or thiazole as isosteres of the carboxylic acid moiety resulted in very potent analogues. From these, AM-6761 (4) emerged as a potent inhibitor with remarkable biochemical (HTRF IC50 = 0.1 nM) and cellular potency (SJSA-1 EdU IC50 = 16 nM), as well as favorable pharmacokinetic properties. Compound 4 also shows excellent antitumor activity in the SJSA-1 osteosarcoma xenograft model with an ED50 of 11 mg/kg. Optimization efforts toward the discovery of these inhibitors as well as the new interactions observed with the MDM2 protein are described herein.
- Published
- 2014
43. Preclinical evaluation of AMG 925, a FLT3/CDK4 dual kinase inhibitor for treating acute myeloid leukemia
- Author
-
David Hollenback, Julio C. Medina, Lawrence R. McGee, Zhihong Li, Lingming Liang, Cong Li, Timothy J. Carlson, Sharon Zhao, Mei-Chu Lo, Suzanne Coberly, Grace Alba, Ada Chen, John Eksterowicz, Dineli Wickramasinghe, Justin N. Huard, Ji Ma, Margaret F. Weidner, Jessica Orf, Lily Liu, Alexander Kamb, Xianghong Wang, Rachel Ngo, Mark L. Ragains, Kathleen S. Keegan, Kang Dai, and Christophe Queva
- Subjects
Sorafenib ,Niacinamide ,Cancer Research ,Myeloid ,Pyridines ,Mice, Nude ,Apoptosis ,Pharmacology ,Receptor tyrosine kinase ,Piperazines ,Mice ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Naphthyridines ,Protein Kinase Inhibitors ,Cell Proliferation ,biology ,Dose-Response Relationship, Drug ,business.industry ,Cell growth ,Kinase ,Phenylurea Compounds ,Cell Cycle ,Myeloid leukemia ,Cancer ,Cyclin-Dependent Kinase 4 ,Neoplasms, Experimental ,U937 Cells ,medicine.disease ,Xenograft Model Antitumor Assays ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,fms-Like Tyrosine Kinase 3 ,biology.protein ,business ,Heterocyclic Compounds, 3-Ring ,medicine.drug ,Signal Transduction - Abstract
Acute myeloid leukemia (AML) remains a serious unmet medical need. Despite high remission rates with chemotherapy standard-of-care treatment, the disease eventually relapses in a major proportion of patients. Activating Fms-like tyrosine kinase 3 (FLT3) mutations are found in approximately 30% of patients with AML. Targeting FLT3 receptor tyrosine kinase has shown encouraging results in treating FLT3-mutated AML. Responses, however, are not sustained and acquired resistance has been a clinical challenge. Treatment options to overcome resistance are currently the focus of research. We report here the preclinical evaluation of AMG 925, a potent, selective, and bioavailable FLT3/cyclin-dependent kinase 4 (CDK4) dual kinase inhibitor. AMG 925 inhibited AML xenograft tumor growth by 96% to 99% without significant body weight loss. The antitumor activity of AMG 925 correlated with the inhibition of STAT5 and RB phosphorylation, the pharmacodynamic markers for inhibition of FLT3 and CDK4, respectively. In addition, AMG 925 was also found to inhibit FLT3 mutants (e.g., D835Y) that are resistant to the current FLT3 inhibitors (e.g., AC220 and sorafenib). CDK4 is a cyclin D–dependent kinase that plays an essential central role in regulating cell proliferation in response to external growth signals. A critical role of the CDK4–RB pathway in cancer development has been well established. CDK4-specific inhibitors are being developed for treating RB-positive cancer. AMG 925, which combines inhibition of two kinases essential for proliferation and survival of FLT3-mutated AML cells, may improve and prolong clinical responses. Mol Cancer Ther; 13(4); 880–9. ©2014 AACR.
- Published
- 2014
44. Methylation in hMLH1 promoter interferes with its binding to transcription factor CBF and inhibits gene expression
- Author
-
Ada Chen, Guoren Deng, Young S. Kim, and Erik Weiking Pong
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Transcription, Genetic ,Recombinant Fusion Proteins ,Response element ,CAAT box ,Adenocarcinoma ,Biology ,Decitabine ,Transfection ,Epigenetics of physical exercise ,Genes, Reporter ,Transcription (biology) ,Tumor Cells, Cultured ,Genetics ,Humans ,Gene Silencing ,Luciferases ,Promoter Regions, Genetic ,neoplasms ,Molecular Biology ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Binding Sites ,Nuclear Proteins ,nutritional and metabolic diseases ,Promoter ,DNA, Neoplasm ,Methylation ,DNA Methylation ,Molecular biology ,digestive system diseases ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,CCAAT-Binding Factor ,Gene Expression Regulation ,DNA methylation ,Azacitidine ,CpG Islands ,Electrophoresis, Polyacrylamide Gel ,Carrier Proteins ,Colorectal Neoplasms ,MutL Protein Homolog 1 ,Protein Binding - Abstract
Microsatellite instability (MSI) is caused by the dysfunction of mismatch repair genes, such as hMLH1, hMSH2. Loss of hMLH1 expression and methylation of CpG sites in hMLH1 promoter are frequently present in sporadic colorectal cancer with MSI. In this study, by transient transfection assay with constructs containing different lengths of hMLH1 promoter and a luciferase reporter gene, we located a proximal region of hMLH1 promoter, which plays a main role in regulating the gene. The fact that luciferase activities were high in all host cell lines regardless of their hMLH1 expression levels indicates that the transcription machinery is intact even in non-expressing cells. When hMLH1 promoter was in vitro methylated before transfection, the luciferase activities in the transfectants were significantly reduced. This observation indicates that methylation causes the inhibition of hMLH1 promoter activity. By electrophoretic mobility shift assay (EMSA), we identified a CCAAT box in this region, which specifically bound transcription factor CBF. Mutations in CCAAT box not only inhibited its binding to CBF factor, but also reduced its ability to drive the expression of luciferase gene. The role of CBF in activating transcription was further substantiated by inhibition of promoter activity with a plasmid expressing a dominant negative CBF-B mutant. Methylation at a CpG site two base pairs upstream of the CCAAT box inhibited the binding of CBF to CCAAT box. We conclude that methylation of an adjacent CpG site inhibits binding of the CBF transcription to the corresponding CCAAT box, and is one of the causes of hMLH1 gene silencing in colon cancer cells.
- Published
- 2001
- Full Text
- View/download PDF
45. The adoption of virtual banking: an empirical study
- Author
-
Huaiqing Wang, Yuan Pu Shao, S. Y. Liao, and Ada Chen
- Subjects
Strategic planning ,Hardware_MEMORYSTRUCTURES ,Computer Networks and Communications ,business.industry ,Theory of planned behavior ,Information technology ,Library and Information Sciences ,Virtual reality ,Empirical research ,Telephone banking ,Retail banking ,The Internet ,Business ,Marketing ,Information Systems - Abstract
Virtual banking is broadly defined in this paper as the provision of banking services via means other than traditional physical branches. Currently, virtual banking exists in the forms of ATM, phone banking, home banking and Internet banking. Understanding people's adoption intention of virtual banking can help financial institutions to formulate appropriate marketing strategies for new forms of banking. Theory of planned behavior (TPB) and innovation diffusion were used to study the adoption intention of virtual banking in a well-developed international financial city. The study finds that the relationships were found only partially explained by the TPB. Other results are interesting and useful for the strategic planning of IT in banking.
- Published
- 1999
- Full Text
- View/download PDF
46. Rational design and binding mode duality of MDM2-p53 inhibitors
- Author
-
Yosup Rew, Steve Schneider, Peter Yakowec, Lisa Julian, Jude Canon, Alexander M. Long, Xuelei Yan, Julio C. Medina, Xin Huang, Jonathan D. Oliner, Hilary Plake Beck, Felix Gonzalez-Lopez de Turiso, Dongyin Yu, Shou-Hua Xiao, Ada Chen, Mei-Chu Lo, Xiaoning Zhao, Anne Y. Saiki, David Chow, Michael D. Bartberger, Jing Zhou, Qiuping Ye, Tiffany L. Correll, Daqing Sun, Frank Kayser, Steven H. Olson, Jay P. Powers, Tao Osgood, Dustin McMinn, and Paul L. Shaffer
- Subjects
Models, Molecular ,Stereochemistry ,Morpholines ,Mice, Nude ,Antineoplastic Agents ,Crystallography, X-Ray ,Cocrystal ,Mice ,Structure-Activity Relationship ,Piperidines ,In vivo ,Drug Discovery ,Animals ,Humans ,IC50 ,Messenger RNA ,Crystallography ,biology ,Chemistry ,Circular Dichroism ,Rational design ,Proto-Oncogene Proteins c-mdm2 ,Stereoisomerism ,Small molecule ,Xenograft Model Antitumor Assays ,Pharmacodynamics ,Drug Design ,biology.protein ,Molecular Medicine ,Mdm2 ,Female ,Indicators and Reagents ,Tumor Suppressor Protein p53 - Abstract
Structural analysis of both the MDM2-p53 protein-protein interaction and several small molecules bound to MDM2 led to the design and synthesis of tetrasubstituted morpholinone 10, an MDM2 inhibitor with a biochemical IC50 of 1.0 μM. The cocrystal structure of 10 with MDM2 inspired two independent optimization strategies and resulted in the discovery of morpholinones 16 and 27 possessing distinct binding modes. Both analogues were potent MDM2 inhibitors in biochemical and cellular assays, and morpholinone 27 (IC50 = 0.10 μM) also displayed suitable PK profile for in vivo animal experiments. A pharmacodynamic (PD) experiment in mice implanted with human SJSA-1 tumors showed p21(WAF1) mRNA induction (2.7-fold over vehicle) upon oral dosing of 27 at 300 mg/kg.
- Published
- 2013
47. Abstract PR10: Small-molecule inhibitors of CD73 promote activation of human CD8+ T cells and have profound effects on tumor growth and immune parameters in experimental tumors
- Author
-
Manmohan Reddy Leleti, Juan C. Jaen, Ulrike Schindler, Jarek Kalisiak, Steve Young, Jay P. Powers, Shin Heng Chiou, Laurent Debien, Ada Chen, and Joanne Bl Tan
- Subjects
0301 basic medicine ,Granzyme B production ,Cancer Research ,T cell ,Immunology ,CD28 ,Biology ,Molecular biology ,Granzyme B ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,Cytotoxic T cell ,IL-2 receptor ,Cell activation ,CD8 - Abstract
Introduction: Intra-tumoral generation of adenosine (ADO), a potent inhibitor of T-cell activation, requires 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 ADO). For this reason, various anti-CD73 antibodies are being advanced into clinical trials; however, there are few reports of potent, selective, small-molecule CD73 inhibitors, such as those described here. Methods: Enzymatic assays: Ecto-nucleotidase activity was calculated based on the amount of inorganic phosphate (malachite green assay; absorbance at 620 nm) produced after 50-min incubation with 25μM AMP or ATP, in the presence of varying concentrations of test compound(s). The following systems were used. Endogenous expression: hCD73/SKOV-3 cells; mCD73/E771 cells. Stable over-expression: hCD73/CHO; hCD39/CHO. Transient expression: mCD73/CHO; NTPDase2/CHO; NTPDase3/CHO; NTPDase8/CHO. CD8+ T cell functional assays: Human (enriched from buffy coats or leukopaks) and mouse (isolated from spleen of C57BL/6 mice) CD8+ T cells were pretreated with CD73 inhibitor or buffer control. One hour later, cells were stimulated (anti-CD3/CD28; +/- 50 μM AMP) in the presence of ADO deaminase inhibitor (10 μM). 3 days after stimulation, cell activation (CD25), proliferation (CFSE) and effector function (IFN-γ and granzyme B) were quantified by flow cytometry. Tumor model: CT26 cells were subcutaneously implanted into Balb/c mice. When tumor volumes were ∼100 mm3, mice were enrolled into cohorts (1: vehicle control; 2: A000830; 3: PD-1 Ab; 4: A000830 + PD-1 Ab) of ≥13 mice each. Interim immune phenotyping was performed when Group 1 tumor volumes were 800-1,000 mm3. Single-cell suspensions were generated from tumors and spleens (gentleMACS). 106 cells were blocked using purified CD16/32 antibodies (clone 2.4G2) and stained with the appropriate antibody cocktails. Results: We have designed a series of potent and specific small-molecule inhibitors of human and mouse CD73, represented by A000830 (IC50 against human and mouse CD73 of 1.0 nM and 3 nM, respectively). A000830 blocked ADO generation from AMP by human ovarian cancer cells (SKOV-3) with IC50: 0.2 nM and >10,000-fold selectivity relative to other ecto-nucleotidases (CD39, NTPDase2, NTPDase3, NTPDase8) and a large panel of unrelated enzymes, receptors and ion channels. In in vitro models of ADO-driven inhibition of human and mouse CD8+ T-cell activation, A000830 showed robust rescue of proliferation, CD25 expression, and IFNγ and granzyme B production. A000830 was easily administered (and well tolerated) to mice, resulting in sustained plasma concentrations at least 1,000 times higher than its potency against mouse CD73. In the spleen of CT26 tumor-bearing Balb/c mice, CD39 expression was found mainly on natural killer (NK) cells and myeloid-lineage cells, while in the tumor CD39 was highly expressed on myeloid cells (myeloid-derived suppressor cells (MDSC) and dendritic cells (DC)) as well as CD4+ and CD8+ T cells. CD73 expression was limited (spleen and tumor) to NK cells and CD4+ and CD8+ T cells. Therapeutic dosing of A000830 to these mice (tumors ∼100 mm3 in size) in combination with anti-PD1 antibody produced robust tumor growth inhibition, greater than either treatment alone. This effect was associated with increased CD8:Treg ratios in tumors (but not spleens). Conclusions: We describe 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 and mouse T-cell activation, and display promising anti-tumor activity when dosed in combination with PD-1 blockade. Citation Format: Juan C. Jaen, Jay P. Powers, Ada Chen, Manmohan R. Leleti, Jarek Kalisiak, Ulrike Schindler, Joanne Bl Tan, Steve Young, Shin-Heng Chiou, Laurent Debien. Small-molecule inhibitors of CD73 promote activation of human CD8+ T cells and have profound effects on tumor growth and immune parameters in experimental tumors [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr PR10.
- Published
- 2016
- Full Text
- View/download PDF
48. Structure-based design of novel inhibitors of the MDM2-p53 interaction
- Author
-
Felix Gonzalez-Lopez de Turiso, Ada Chen, Steven H. Olson, Xin Huang, Jude Canon, Anne Y. Saiki, Michael D. Bartberger, Hilary Plake Beck, Jonathan D. Oliner, David J. Kopecky, Xuelei Yan, Jing Zhou, Maria M. Toteva, Min Jiang, Mei-Chu Lo, Alexander M. Long, Darin J. Gustin, Klaus Michelsen, Tao Osgood, Steve Schneider, Qiuping Ye, Jeffrey Deignan, Yosup Rew, Peter Yakowec, Daqing Sun, Xianyun Jiao, Lixia Jin, Dongyin Yu, David Chow, Julio C. Medina, Frank Kayser, Mark L. Ragains, Yihong Li, Xiaoning Zhao, and Brian M. Fox
- Subjects
Models, Molecular ,rho GTP-Binding Proteins ,Stereochemistry ,Transplantation, Heterologous ,Substituent ,Molecular Conformation ,Mice, Nude ,Stereoisomerism ,Antineoplastic Agents ,Plasma protein binding ,Acetates ,Ring (chemistry) ,Crystallography, X-Ray ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Animals ,Humans ,neoplasms ,Piperidones ,Cell Proliferation ,Rational design ,Proto-Oncogene Proteins c-mdm2 ,Rats ,Transplantation ,Macaca fascicularis ,chemistry ,Hepatocytes ,Molecular Medicine ,Drug Screening Assays, Antitumor ,Tumor Suppressor Protein p53 ,Neoplasm Transplantation ,Protein Binding - Abstract
Structure-based rational design led to the discovery of novel inhibitors of the MDM2–p53 protein–protein interaction. The affinity of these compounds for MDM2 was improved through conformational control of both the piperidinone ring and the appended N-alkyl substituent. Optimization afforded 29 (AM-8553), a potent and selective MDM2 inhibitor with excellent pharmacokinetic properties and in vivo efficacy.
- Published
- 2012
49. Abstract 3663: Discovery of sulfonamide-piperidinones as potent inhibitors of the MDM2-p53 protein-protein interaction
- Author
-
Zhihong Li, Ada Chen, Julio C. Medina, Jude Canon, Steve H. Olson, Jing Zhou, Tao Osgood, Daqing Sun, Xin Huang, Lawrence R. McGee, Mei-Chu Lo, Michael W. Gribble, Qiuping Ye, Jiasheng Fu, Xiaoning Zhao, Sarah Wortman, Paul L. Shaffer, Lixia Jin, Yosup Rew, Jonathan D. Oliner, Dongyin Yu, Anne Y. Saiki, and John Eksterowicz
- Subjects
Cancer Research ,Cell cycle checkpoint ,biology ,Cell growth ,Chemistry ,Sulfonamide (medicine) ,Cancer ,medicine.disease ,Protein–protein interaction ,Oncology ,Apoptosis ,Cancer cell ,medicine ,biology.protein ,Cancer research ,Mdm2 ,medicine.drug - Abstract
The p53 tumor suppressor is controlled by MDM2, which binds p53 and negatively regulates its transcriptional activity and stability. Many tumors overproduce MDM2 to impair p53 function. Therefore, restoration of p53 activity by inhibiting p53-MDM2 binding represents an attractive, novel approach to cancer therapy. We previously reported the discovery of AM-8553, a potent and selective piperidinone inhibitor of the MDM2-p53 interaction (Rew et al. J. Med. Chem. 2012, 55, 4936). We report here continued optimization of the N-alkyl substituent of this series, focused in particular on a previously underutilized interaction in a shallow cleft on the MDM2 surface that led to the discovery of a variety of extremely potent sulfonamides such as 14 with an IC50 of 5.3 nM in the cell proliferation assay. The compound 14 interacts specifically with the p53-binding pocket of MDM2 and releases the p53 protein from negative control. Treatment of cancer cells expressing wild-type p53 with sulfonamide 14 stabilizes p53 and activates the p53 pathway, leading to cell cycle arrest and apoptosis. The compound 14 showed excellent efficacy and caused tumor regression in the SJSA-1 tumor xenograft model. Citation Format: Zhihong Li, Jiasheng Fu, Yosup Rew, Michael W. Gribble, Jude Canon, Ada Chen, John Eksterowicz, Xin Huang, Lixia Jin, Mei-Chu Lo, Lawrence R. McGee, Tao Osgood, Anne Y. Saiki, Paul Shaffer, Daqing Sun, Sarah Wortman, Qiuping Ye, Dongyin Yu, Xiaoning Zhao, Jing Zhou, Jonathan D. Oliner, Steve H. Olson, Julio C. Medina. Discovery of sulfonamide-piperidinones as potent inhibitors of the MDM2-p53 protein-protein interaction. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3663. doi:10.1158/1538-7445.AM2015-3663
- Published
- 2015
- Full Text
- View/download PDF
50. Abstract A279: Preclinical evaluation of AMG 925, a FLT3/CDK4 dual kinase inhibitor
- Author
-
Ma Ji, Justin N. Huard, Lingming Liang, Ada Chen, Grace Alba, John Eksterowicz, Kang Dai, Lawrence R. McGee, Timothy J. Carlson, Mei-Chu Lo, Margret Weidner, Jessica Orf, Zhihong Li, Coberly Suzanne, David Hollenback, Alexander Kamb, Cong Li, Lily Liu, Rachel Ngo, Sharon Zhao, Julio C. Medina, Dineli Wickramasinghe, and Kathleen S. Keegan
- Subjects
Sorafenib ,Cancer Research ,Chemotherapy ,biology ,Cell growth ,Kinase ,business.industry ,medicine.medical_treatment ,Cancer ,Myeloid leukemia ,medicine.disease ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Oncology ,chemistry ,hemic and lymphatic diseases ,biology.protein ,medicine ,Cancer research ,business ,Quizartinib ,medicine.drug - Abstract
Acute myeloid leukemia (AML) remains a serious unmet medical need. Despite high remission rates with chemotherapy standard care treatment, the disease eventually relapses. Activating FLT3 mutations are found in approximately 30% of AML patients. Targeting FLT3 receptor tyrosine kinase has shown encouraging results in treating FLT3-mutated AML. Responses, however, are not sustained and acquired resistance has been a clinical challenge. Treatment options to overcome resistance are currently the focus of research. We report here preclinical evaluation of AMG 925, a potent, selective and bioavailable FLT3/CDK4 dual kinase inhibitor. The compound inhibited AML xenograft tumor growth by >99% without detectable body weight loss. AMG 925 was also found to inhibit FLT3 mutants (e.g, D835Y) that are resistant to the current FLT3 inhibitors (e.g., quizartinib/AC220, sorafenib). CDK4 is a cyclinD-dependent kinase that plays an essential central role in regulating cell proliferation in response to external growth signals. A critical role of the CDK4-Rb pathway in cancer development has been well established. CDK4 specific inhibitors are being developed for treating Rb positive cancer. AMG 925, which combines inhibition of two kinases essential for proliferation and survival of FLT3-mutated AML cells, may improve clinical response rates. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A279. Citation Format: Kang Dai, Kathleen Keegan, Zhihong Li, Ma Ji, Cong Li, John Eksterowicz, Coberly Suzanne, David Hollenback, Margret Weidner, Justin Huard, Lingming Liang, Grace Alba, Jessica Orf, Mei-Chu Lo, Sharon Zhao, Rachel Ngo, Ada Chen, Lily Liu, Timothy Carlson, Lawrence R. McGee, Julio Medina, Alexander Kamb, Dineli Wickramasinghe. Preclinical evaluation of AMG 925, a FLT3/CDK4 dual kinase inhibitor. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A279.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.