52 results on '"Hakan Gunaydin"'
Search Results
2. Abstract P5-16-10: RLY-2608: The first allosteric mutant- and isoform-selective inhibitor of PI3Kα, is efficacious as a single agent and drives regressions in combination with standard of care therapies in PIK3CA mutant breast cancer models
- Author
-
Ermira Pazolli, Randy Kipp, Alessandro Boezio, Hakan Gunaydin, Amanda Iskandar, Matthew Zubrowski, Bret Williams, Kelley Shortsleeves, Alexandre Larivee, Tom McLean, Klaus Michelsen, Hongtao Zeng, Jonathan LaRochelle, Joe Manna, Lucian DiPietro, Andre Lescarbeau, Mary Mader, Bindu Bennet, Jeremy Wilbur, Qi Wang, Levi Pierce, Iain Martin, James Watters, Pascal Fortin, and Donald Bergstrom
- Subjects
Cancer Research ,Oncology - Abstract
Inhibition of CDK4/6 combined with the estrogen receptor (ER) degrader fulvestrant significantly improves progression free survival and overall survival in advanced hormone receptor positive (HR+) breast cancer patients and is now the standard of care (SOC) in this disease. Up to 40% of HR+ breast cancers harbor PIK3CA mutations leading to activation of phosphoinositide 3-kinase alpha (PI3Kα), which has been associated with resistance to CDK4/6 inhibitors and fulvestrant. Therefore, PI3Kα inhibitor combinations with CDK4/6 inhibitors and/or fulvestrant are of high interest in HR+, PIK3CA mutant breast cancer. The therapeutic index of active site (orthosteric) inhibitors of PI3Kα has been limited by the dual issues of no clinically meaningful selectivity for mutant versus wild-type (WT) PI3Kα and off-isoform inhibitory activity. Alpelisib, the only approved orthosteric PI3Kα inhibitor, is emblematic of the class with toxicity related to inhibition of wild type PI3Kα and other PI3K isoforms resulting in sub-optimal inhibition of mutant PI3Kα, frequent discontinuation, and challenges in combining with CDK4/6 inhibitors. To overcome these limitations, we designed RLY-2608, the first allosteric, mutant, and isoform-selective inhibitor of PI3Kα. We solved the full-length cryo-EM structure of PI3Kα, performed long time-scale molecular dynamic simulations to elucidate conformational differences between WT and mutant PI3Kα, and leveraged these insights to enable the design of RLY-2608. RLY-2608 does not compete with orthosteric inhibitors for binding and associates 8x faster with mutant PI3Kα relative to WT PI3Kα. In biochemical assays, RLY-2608 inhibits kinase domain (H1047R) and helical domain (E542K, and E545K) mutant PI3Kα activity, demonstrating 1000-fold selective over the β, δ, and γ PI3K isoforms in biochemical assays and demonstrates exquisite selectivity across a panel of 322 kinases, with no other kinases showing >50% inhibition. We performed in vitro combinations in two HR+ PIK3CA mutant cell lines (MCF7: E545K; T47D: H1047R) and observed synergy between RLY-2608 and fulvestrant or CDK4/6 inhibitors. In vivo, we tested combinations of RLY-2608 with fulvestrant and/or the CDK4/6 inhibitor abemaciclib in the MCF7 xenograft model. Oral administration of RLY-2608 in combination with fulvestrant led to improved efficacy compared to either agent alone in a dose-dependent manner, with regressions observed in the combination arms at all doses. Furthermore, the triple combination of RLY-2608, fulvestrant, and abemaciclib resulted in superior efficacy compared to either the RLY-2608 + fulvestrant or RLY-2608 + abemaciclib doublets, with deep regressions observed in the triple combination arm. In addition, in vivo combination efficacy with fulvestrant and CDK4/6 inhibitors (palbociclib or abemaciclib) was assessed in patient-derived xenografts harboring the PIK3CA H1047R or E545K mutation along with a second site PIK3CA minor mutation. In these studies, combination benefit was observed with doses of RLY-2608 significantly lower than the dose required for maximum efficacy as a single agent. RLY-2608 synergizes in vitro with both anti-estrogen and CDK4/6 inhibitors in cell models of HR+/PIK3CA mutant breast cancer. RLY-2608 can be combined with fulvestrant and CDK4/6 inhibitors in vivo with tumor regressions observed in both cell- and patient-derived xenograft models. The pre-clinical profile of RLY-2608 supports the clinical development of RLY-2608 both in single agent and combination clinical trials in patients with PIK3CA mutant tumors, including HR+/PIK3CA mutant breast cancer. Citation Format: Ermira Pazolli, Randy Kipp, Alessandro Boezio, Hakan Gunaydin, Amanda Iskandar, Matthew Zubrowski, Bret Williams, Kelley Shortsleeves, Alexandre Larivee, Tom McLean, Klaus Michelsen, Hongtao Zeng, Jonathan LaRochelle, Joe Manna, Lucian DiPietro, Andre Lescarbeau, Mary Mader, Bindu Bennet, Jeremy Wilbur, Qi Wang, Levi Pierce, Iain Martin, James Watters, Pascal Fortin, Donald Bergstrom. RLY-2608: The first allosteric mutant- and isoform-selective inhibitor of PI3Kα, is efficacious as a single agent and drives regressions in combination with standard of care therapies in PIK3CA mutant breast cancer models [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P5-16-10.
- Published
- 2022
3. Discovery and Optimization of Potent, Selective, and Brain-Penetrant 1-Heteroaryl-1
- Author
-
David A, Candito, Vladimir, Simov, Anmol, Gulati, Solomon, Kattar, Ryan W, Chau, Blair T, Lapointe, Joey L, Methot, Duane E, DeMong, Thomas H, Graham, Ravi, Kurukulasuriya, Mitchell H, Keylor, Ling, Tong, Gregori J, Morriello, John J, Acton, Barbara, Pio, Weiguo, Liu, Jack D, Scott, Michael J, Ardolino, Theodore A, Martinot, Matthew L, Maddess, Xin, Yan, Hakan, Gunaydin, Rachel L, Palte, Spencer E, McMinn, Lisa, Nogle, Hongshi, Yu, Ellen C, Minnihan, Charles A, Lesburg, Ping, Liu, Jing, Su, Laxminarayan G, Hegde, Lily Y, Moy, Janice D, Woodhouse, Robert, Faltus, Tina, Xiong, Paul, Ciaccio, Jennifer A, Piesvaux, Karin M, Otte, Matthew E, Kennedy, David Jonathan, Bennett, Erin F, DiMauro, Matthew J, Fell, Santhosh, Neelamkavil, Harold B, Wood, Peter H, Fuller, and J Michael, Ellis
- Subjects
Indazoles ,Adenosine Triphosphate ,Leukocytes, Mononuclear ,Humans ,Animals ,Brain ,Parkinson Disease ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Protein Kinase Inhibitors ,Rats - Abstract
Inhibition of leucine-rich repeat kinase 2 (LRRK2) kinase activity represents a genetically supported, chemically tractable, and potentially disease-modifying mechanism to treat Parkinson's disease. Herein, we describe the optimization of a novel series of potent, selective, central nervous system (CNS)-penetrant 1-heteroaryl-1
- Published
- 2022
4. When Technocratic Appointments Signal Credibility
- Author
-
Hakan Gunaydin, William Spaniel, and Despina Alexiadou
- Subjects
021110 strategic, defence & security studies ,Sociology and Political Science ,05 social sciences ,Financial market ,0211 other engineering and technologies ,02 engineering and technology ,Technocracy ,Public administration ,Prime (order theory) ,0506 political science ,technocrats, experts, financial crises, credible policy signals, financial markets ,ddc:320 ,Credibility ,050602 political science & public administration ,Business - Abstract
How do prime ministers manage investors’ expectations during financial crises? We take a novel approach to this question by investigating ministerial appointments. When prime ministers appoint technocrats, defined as non-partisan experts, they forgo political benefits and can credibly signal their willingness to pay down their debt obligations. This reduces bond yields, but only at times when the market is sensitive to expected repayments—that is, during crises. To examine the theory, we develop an event study analysis that employs new data on the background of finance ministers in 21 Western and Eastern European democracies. We find that investors reward technocratic appointments by reducing a country’s borrowing costs. Consistent with the theory, technocratic appointments under crises predict lower bond yields. Our findings contribute to the literature on the interplay of financial markets and domestic politics.
- Published
- 2021
5. Optimization of brain-penetrant picolinamide derived leucine-rich repeat kinase 2 (LRRK2) inhibitors
- Author
-
Blair T. Lapointe, Jack D. Scott, Xin Cindy Yan, Haiqun Tang, Janice D Woodhouse, Kaleen Konrad Childers, Robert Faltus, Erin F. DiMauro, Solomon Kattar, Charles S. Yeung, Ravi Kurukulasuriya, Vladimir Simov, Hakan Gunaydin, Anmol Gulati, Joey L. Methot, Rachel L. Palte, Ellen C. Minnihan, Greg Morriello, J. Michael Ellis, Harold B. Wood, Santhosh Neelamkavil, Karin M. Otte, Michael J. Ardolino, Barbara Pio, Ping Liu, Laxminarayan G Hegde, Matthew J. Fell, Vanessa L. Rada, Peter Fuller, and Paul J Ciaccio
- Subjects
Pharmacology ,0303 health sciences ,Trifluoromethyl ,Chemistry ,Metabolite ,Organic Chemistry ,Pharmaceutical Science ,Pyrazole ,Leucine-rich repeat ,Biochemistry ,LRRK2 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Drug Discovery ,Molecular Medicine ,Kinome ,Penetrant (biochemical) ,Linker ,030304 developmental biology - Abstract
The discovery of potent, kinome selective, brain penetrant LRRK2 inhibitors is the focus of extensive research seeking new, disease-modifying treatments for Parkinson's disease (PD). Herein, we describe the discovery and evolution of a picolinamide-derived lead series. Our initial optimization efforts aimed at improving the potency and CLK2 off-target selectivity of compound 1 by modifying the heteroaryl C-H hinge and linker regions. This resulted in compound 12 which advanced deep into our research operating plan (ROP) before heteroaryl aniline metabolite 14 was characterized as Ames mutagenic, halting its progression. Strategic modifications to our ROP were made to enable early de-risking of putative aniline metabolites or hydrolysis products for mutagenicity in Ames. This led to the discovery of 3,5-diaminopyridine 15 and 4,6-diaminopyrimidine 16 as low risk for mutagenicity (defined by a 3-strain Ames negative result). Analysis of key matched molecular pairs 17 and 18 led to the prioritization of the 3,5-diaminopyridine sub-series for further optimization due to enhanced rodent brain penetration. These efforts culminated in the discovery of ethyl trifluoromethyl pyrazole 23 with excellent LRRK2 potency and expanded selectivity versus off-target CLK2.
- Published
- 2021
6. Commitment or expertise? Technocratic appointments as political responses to economic crises
- Author
-
Despina Alexiadou and Hakan Gunaydin
- Subjects
021110 strategic, defence & security studies ,Politics ,Sociology and Political Science ,Political economy ,Political science ,05 social sciences ,050602 political science & public administration ,0211 other engineering and technologies ,02 engineering and technology ,Technocracy ,0506 political science - Abstract
Why do prime ministers or presidents appoint non‐elected experts, also known as technocrats, during economic crises? Do they appoint them for their expertise or for their commitment to pro‐market reforms? Answering this question is crucial for understanding and predicting the longer‐term role of technocrats in democracies. With the aid of unique data on the political and personal background of finance ministers in 13 parliamentary and semi‐presidential European democracies this article shows that commitment, not expertise is the primary driver of technocratic appointments during major economic crises. Technocrats are preferred over experienced politicians when the latter lack commitment to policy reform. An important implication of the findings is that technocratic appointments to top economic portfolios in West European countries are unlikely to become the norm outside economic crises, assuming economic crises are short‐lived and not recurring.
- Published
- 2019
7. A Potent and Selective ULK1 Inhibitor Suppresses Autophagy and Sensitizes Cancer Cells to Nutrient Stress
- Author
-
Ronan C. O'Hagan, Abigail R. Solitro, Mark E. Scott, Jeffrey P. MacKeigan, Stuart D. Shumway, Hakan Gunaydin, Katie R. Martin, Peter Fuller, and Stephanie L. Celano
- Subjects
0301 basic medicine ,Multidisciplinary ,Functional Aspects of Cell Biology ,Kinase ,Chemistry ,Autophagy ,Mutant ,Regulator ,Cancer ,Therapeutics ,ULK1 ,medicine.disease ,Article ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,Tumor progression ,Cancer cell ,Cancer research ,medicine ,lcsh:Q ,lcsh:Science - Abstract
Summary In response to stress, cancer cells generate nutrients and energy through a cellular recycling process called autophagy, which can promote survival and tumor progression. Accordingly, autophagy inhibition has emerged as a potential cancer treatment strategy. Inhibitors targeting ULK1, an essential and early autophagy regulator, have provided proof of concept for targeting this kinase to inhibit autophagy; however, these are limited individually in their potency, selectivity, or cellular activity. In this study, we report two small molecule ULK1 inhibitors, ULK-100 and ULK-101, and establish superior potency and selectivity over a noteworthy published inhibitor. Moreover, we show that ULK-101 suppresses autophagy induction and autophagic flux in response to different stimuli. Finally, we use ULK-101 to demonstrate that ULK1 inhibition sensitizes KRAS mutant lung cancer cells to nutrient stress. ULK-101 represents a powerful molecular tool to study the role of autophagy in cancer cells and to evaluate the therapeutic potential of autophagy inhibition., Graphical Abstract, Highlights • ULK-101 has improved potency and selectivity when compared with SBI-0206965 • ULK-101 inhibits both the nucleation of autophagic vesicles and turnover • ULK-101 sensitizes KRAS-driven lung cancer cells to nutrient restriction • ULK-101 is a valuable molecular tool to study the function of ULK1 and autophagy, Therapeutics; Functional Aspects of Cell Biology; Cancer
- Published
- 2018
8. 1,2,4-Triazolsulfone: A novel isosteric replacement of acylsulfonamides in the context of Na V 1.7 inhibition
- Author
-
Emily A. Peterson, Stephanie D. Geuns-Meyer, Matthew Weiss, Hongbing Huang, Katrina W. Copeland, Violeta Yu, Erin F. DiMauro, Hua Gao, Alessandro Boezio, Russell Graceffa, Daniel S. La, Hakan Gunaydin, Robert T. Fremeau, Christiane Boezio, Margaret Chu-Moyer, Joseph Ligutti, Bryan D. Moyer, Kristin L. Andrews, and Robert S. Foti
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Gene isoform ,Aryl ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Context (language use) ,Biochemistry ,Combinatorial chemistry ,Sulfonamide ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Drug Discovery ,NAV1 ,Molecular Medicine ,Bioisostere ,Pharmacophore ,Selectivity ,Molecular Biology - Abstract
Recently, the identification of several classes of aryl sulfonamides and acyl sulfonamides that potently inhibit NaV1.7 and demonstrate high levels of selectivity over other NaV isoforms have been reported. The fully ionizable nature of these inhibitors has been shown to be an important part of the pharmacophore for the observed potency and isoform selectivity. The requirement of this functionality, however, has presented challenges associated with optimization toward inhibitors with drug-like properties and minimal off-target activity. In an effort to obviate these challenges, we set out to develop an orally bioavailable, selective NaV1.7 inhibitor, lacking these acidic functional groups. Herein, we report the discovery of a novel series of inhibitors wherein a triazolesulfone has been designed to serve as a bioisostere for the acyl sulfonamide. This work culminated in the delivery of a potent series of inhibitors which demonstrated good levels of selectivity over NaV1.5 and favorable pharmacokinetics in rodents.
- Published
- 2018
9. Strategy for Extending Half-life in Drug Design and Its Significance
- Author
-
Hakan Gunaydin, Scott A. Johnson, Brian R. Lahue, Blair T. Lapointe, J. Michael Ellis, Michael D. Altman, and Peter Fuller
- Subjects
0301 basic medicine ,Drug ,Chemistry ,media_common.quotation_subject ,Organic Chemistry ,Half-life ,Pharmacology ,030226 pharmacology & pharmacy ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Drug Discovery ,Lipophilicity ,Dose prediction ,Potency ,Constant (mathematics) ,media_common - Abstract
[Image: see text] Preclinical optimization of compounds toward viable drug candidates requires an integrated understanding of properties that impact predictions of the clinically efficacious dose. The importance of optimizing half-life, unbound clearance, and potency and how they impact dose predictions are discussed in this letter. Modest half-life improvements for short half-life compounds can dramatically lower the efficacious dose. The relationship between dose and half-life is nonlinear when unbound clearance is kept constant, whereas the relationship between dose and unbound clearance is linear when half-life is kept constant. Due to this difference, we show that dose is more sensitive to changes in half-life than changes in unbound clearance when half-lives are shorter than 2 h. Through matched molecular pair analyses, we also show that the strategic introduction of halogens is likely to increase half-life and lower projected human dose even though increased lipophilicity does not guarantee extended half-life.
- Published
- 2018
10. Who Can Reform the Labor Market? IMF Conditionality, Partisanship, and Labor Unions
- Author
-
Hakan Gunaydin
- Subjects
021110 strategic, defence & security studies ,Political economy ,Political science ,05 social sciences ,Political Science and International Relations ,050602 political science & public administration ,0211 other engineering and technologies ,Opposition (politics) ,02 engineering and technology ,Conditionality ,0506 political science - Abstract
Labor market reforms are critical for economic growth. Yet, they are politically contentious, and governments, more often than not, are faced with strong opposition from interest groups. Scholarly ...
- Published
- 2018
11. Abstract P251: Discovery and characterization of RLY-2608: The first allosteric, mutant, and isoform-selective inhibitor of PI3Kα
- Author
-
Ermira Pazolli, Randy Kipp, Alessandro Boezio, Hakan Gunaydin, Amanda Iskandar, Matthew Zubrowski, Bret Williams, Kelley Shortsleeves, Alexandre Larivee, Tom McLean, Klaus Michelsen, Hongtao Zeng, Jonathan LaRochelle, Joe Manna, Lucian DiPietro, Mary Mader, Bindu Bennet, Jeremy Wilbur, Qi Wang, Levi Pierce, Iain Martin, James Watters, Pascal Fortin, and Donald Bergstrom
- Subjects
Cancer Research ,Oncology - Abstract
Phosphoinositide 3-kinase alpha (PI3Kα) is the most frequently mutated kinase in solid tumors. Traditionally, the development of PI3Kα inhibitors has focused on the active, or orthosteric site. The therapeutic index of orthosteric inhibitors is limited by the lack of clinically meaningful selectivity for mutant versus wild-type (WT) PI3Kα and off-isoform activity. Alpelisib, the only approved orthosteric PI3Kα inhibitor, is emblematic of the class with toxicity related to inhibition of WT PI3Kα and other PI3K isoforms resulting in sub-optimal inhibition of mutant PI3Kα with reductions in dose intensity and frequent discontinuation. To overcome these limitations, we designed RLY-2608, the first allosteric, mutant, and isoform-selective PI3Kα inhibitor. We solved the full-length cryo-EM structure of PI3Kα, performed long time-scale molecular dynamic simulations to elucidate conformational differences between WT and mutant PI3Kα, and leveraged these insights to enable the design of RLY-2608. RLY-2608 does not compete with orthosteric inhibitors for binding and associates 8x faster with mutant PI3Kα relative to WT. In biochemical assays, RLY-2608 inhibits both kinase domain (H1047R) and helical domain (E542K, and E545K) mutant PI3Kα activity with 1000-fold selective over the β, δ, and γ PI3K isoforms in biochemical assays and demonstrates exquisite selectivity across a panel of 322 kinases, with no other kinases showing > 50% inhibition. In MCF10A cells engineered to express only mutant or WT PI3Kα, RLY-2608 inhibited phosphorylated AKT (pAKT) in a mutant-selective manner. Furthermore, pAKT and viability were significantly inhibited across a panel of cancer cell lines carrying hotspot PIK3CA mutations. RLY-2608 showed anti-tumor activity in both kinase and helical domain PIK3CA mutant in vivo xenograft models with marked regressions or stasis observed in all models. RLY-2608 was well tolerated, with pharmacodynamic modulation and efficacy observed in a dose dependent manner. Insulin levels measured as an indicator of glucose homeostasis were significantly lower when compared to orthosteric inhibitors, suggesting that RLY-2608 can achieve maximum efficacy by maintaining PI3Kα mutant target coverage throughout the dosing interval with significantly reduced impact on WT PI3Kα. In higher species, dosing of RLY-2608 for 28 days resulted in exposures exceeding mutant PI3Kα cellular PD IC90 throughout the dosing interval without elevated glucose levels or histopathological or ophthalmic findings associated with hyperglycemia. Compared to orthosteric inhibitors, RLY-2608 demonstrates preferential binding and inhibition of mutant PI3Kα, is highly selective across the kinome, and achieves in vivo efficacy without dysregulating glucose homeostasis. These results support clinical investigation of RLY-2608 as a differentiated mechanism for inhibition of oncogenic PI3Kα in patients with PIK3CA mutant tumors. Citation Format: Ermira Pazolli, Randy Kipp, Alessandro Boezio, Hakan Gunaydin, Amanda Iskandar, Matthew Zubrowski, Bret Williams, Kelley Shortsleeves, Alexandre Larivee, Tom McLean, Klaus Michelsen, Hongtao Zeng, Jonathan LaRochelle, Joe Manna, Lucian DiPietro, Mary Mader, Bindu Bennet, Jeremy Wilbur, Qi Wang, Levi Pierce, Iain Martin, James Watters, Pascal Fortin, Donald Bergstrom. Discovery and characterization of RLY-2608: The first allosteric, mutant, and isoform-selective inhibitor of PI3Kα [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr P251.
- Published
- 2021
12. Abstract 1455: RLY-4008, a novel precision therapy for FGFR2-driven cancers designed to potently and selectively inhibit FGFR2 and FGFR2 resistance mutations
- Author
-
Donald A. Bergstrom, Hudson Brandi M, Kamil Bruderek, Schoenherr Heike, Moustakas Demetri T, Jessica Casaletto, Dina Sharon, Lindsey Foster, Roberto Valverde, Pelin Ayaz, Songping Zhao, Dejan Maglic, Alexander B. Taylor, B. Barry Toure, Patrick O'Hearn, Nastaran Gerami-Moayed, James M. Watters, and Hakan Gunaydin
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Mutation ,Kinase ,business.industry ,Endometrial cancer ,Cancer ,medicine.disease ,medicine.disease_cause ,stomatognathic diseases ,Oncology ,Cell culture ,In vivo ,Fibroblast growth factor receptor ,embryonic structures ,medicine ,Cancer research ,Kinome ,business - Abstract
FGFR2 fusions, amplifications, and mutations are oncogenic drivers that occur across multiple tumor types. Clinical efficacy observed with pan-FGFR inhibitors has validated the driver status of FGFR2 in FGFR2 fusion-positive intrahepatic cholangiocarcinoma (ICC), however, FGFR1-mediated toxicities (hyperphosphatemia, tissue mineralization) and the emergence of on-target FGFR2 resistance mutations limit the efficacy of pan-FGFR inhibitors. To overcome these limitations, we designed RLY-4008, a potent and highly selective, FGFR2 inhibitor. Despite significant investment in traditional structure-based drug design, selective targeting of FGFR2 has not been achieved. We leveraged differences in conformational dynamics between FGFR2 and other FGFR isoforms observed through molecular dynamics simulations to enable the design of RLY-4008. RLY-4008 inhibits FGFR2 with low nanomolar potency and demonstrates > 200-fold selectivity over FGFR1, and > 80- and > 5000-fold selectivity over FGFR3 and FGFR4, respectively, in biochemical assays. Additionally, RLY-4008 demonstrates high kinome selectivity for FGFR2 against a panel of > 400 human kinases. RLY-4008 has strong activity against primary and acquired FGFR2 resistance mutations in cellular assays, and potent antiproliferative effects on FGFR2-altered human tumor cell lines. In vivo, RLY-4008 demonstrates dose-dependent FGFR2 inhibition and induces regression in multiple human xenograft tumor models, including FGFR2 fusion-positive ICC, gastric, and lung cancers, FGFR2-amplified gastric cancer, and FGFR2-mutant endometrial cancer. Strikingly, RLY-4008 induces regression in an FGFR2 fusion-positive ICC model harboring the FGFR2V564F gatekeeper mutation and an endometrial cancer model harboring the FGFR2N549K mutation, two mutations that drive clinical progression on current pan-FGFR inhibitors. In the FGFR2V564F model, pan-FGFR inhibitors are ineffective, even at maximally tolerated doses. Notably, treatment of these tumors with RLY-4008 induces rapid regression and restores body weight. In rat and dog toxicology studies, RLY-4008 is well tolerated and is not associated with hyperphosphatemia or tissue mineralization at exposures significantly above those required to induce regression in all models. In contrast to pan-FGFR inhibitors, RLY-4008 is highly selective for FGFR2 and demonstrates strong activity against FGFR2 resistance mutations, suggesting that RLY-4008 may have broader therapeutic potential via preventing and overcoming therapeutic resistance. Together, these data and the favorable pharmaceutical properties of RLY-4008 strongly support its clinical development in FGFR2-altered tumors. Citation Format: Jessica Casaletto, Dejan Maglic, B. Barry Toure, Alex Taylor, Heike Schoenherr, Brandi Hudson, Kamil Bruderek, Songping Zhao, Patrick O'Hearn, Nastaran Gerami-Moayed, Demetri Moustakas, Roberto Valverde, Lindsey Foster, Hakan Gunaydin, Pelin Ayaz, Dina Sharon, Donald Bergstrom, James Watters. RLY-4008, a novel precision therapy for FGFR2-driven cancers designed to potently and selectively inhibit FGFR2 and FGFR2 resistance mutations [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1455.
- Published
- 2021
13. Applications of parallel synthetic lead hopping and pharmacophore-based virtual screening in the discovery of efficient glycine receptor potentiators
- Author
-
Loren Berry, Hakan Gunaydin, Nagasree Chakka, Jeffrey R. Simard, Angel Guzman-Perez, Matthew H. Plant, Erin F. DiMauro, Kristin L. Andrews, Liyue Huang, Jacinthe Gingras, and Howard Bregman
- Subjects
0301 basic medicine ,Azetidine ,Drug Evaluation, Preclinical ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Receptors, Glycine ,Aminothiazole ,Drug Discovery ,Humans ,Glycine receptor ,ADME ,Pharmacology ,Sulfonamides ,Virtual screening ,Dose-Response Relationship, Drug ,Molecular Structure ,Drug discovery ,Chemistry ,Organic Chemistry ,General Medicine ,Potentiator ,Combinatorial chemistry ,Molecular Docking Simulation ,030104 developmental biology ,Pharmacophore - Abstract
Glycine receptors (GlyRs) are pentameric glycine-gated chloride ion channels that are enriched in the brainstem and spinal cord where they have been demonstrated to play a role in central nervous system (CNS) inhibition. Herein we describe two novel classes of glycine receptor potentiators that have been developed using similarity- and property-guided scaffold hopping enabled by parallel synthesis and pharmacophore-based virtual screening strategies. This effort resulted in the identification of novel, efficient and modular leads having favorable in vitro ADME profiles and high CNS multi-parameter optimization (MPO) scores, exemplified by azetidine sulfonamide 19 and aminothiazole sulfone (ent2)-20.
- Published
- 2017
14. The discovery of benzoxazine sulfonamide inhibitors of Na V 1.7: Tools that bridge efficacy and target engagement
- Author
-
Stefan I. McDonough, Min-Hwa Jasmine Lin, Erin F. DiMauro, Violeta Yu, Angel Guzman-Perez, Kristin Taborn, Christiane Bode, Thomas Kornecook, Thomas Dineen, Xin Huang, Robert T. Fremeau, Margaret Chu-Moyer, James R. Coats, Bingfan Du, Jeff S. McDermott, Hakan Gunaydin, Daniel S. La, Hua Gao, Bryan D. Moyer, Russell Graceffa, Alessandro Boezio, Charles Kreiman, Matthew Weiss, Hanh Nho Nguyen, David J. Matson, Joseph Ligutti, Christopher P Ilch, Isaac E. Marx, Emily A. Peterson, and Howard Bregman
- Subjects
0301 basic medicine ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Sulfonamide (medicine) ,Target engagement ,Pharmaceutical Science ,Genetic data ,Pharmacology ,Bioinformatics ,Biochemistry ,Sprague dawley ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Drug Discovery ,medicine ,Molecular Medicine ,Molecular Biology ,030217 neurology & neurosurgery ,medicine.drug - Abstract
The voltage-gated sodium channel NaV1.7 has received much attention from the scientific community due to compelling human genetic data linking gain- and loss-of-function mutations to pain phenotypes. Despite this genetic validation of NaV1.7 as a target for pain, high quality pharmacological tools facilitate further understanding of target biology, establishment of target coverage requirements and subsequent progression into the clinic. Within the sulfonamide class of inhibitors, reduced potency on rat NaV1.7 versus human NaV1.7 was observed, rendering in vivo rat pharmacology studies challenging. Herein, we report the discovery and optimization of novel benzoxazine sulfonamide inhibitors of human, rat and mouse NaV1.7 which enabled pharmacological assessment in traditional behavioral rodent models of pain and in turn, established a connection between formalin-induced pain and histamine-induced pruritus in mice. The latter represents a simple and efficient means of measuring target engagement.
- Published
- 2017
15. Informing the Selection of Screening Hit Series with in Silico Absorption, Distribution, Metabolism, Excretion, and Toxicity Profiles
- Author
-
James I. Fells, John M. Sanders, Jason E. Imbriglio, Suen Linda M, Andrew M. Haidle, Abbas Walji, Elizabeth Joshi, William D. Shipe, Nunzio Sciammetta, Hakan Gunaydin, Tjerk Bueters, Andreas Verras, Robert P. Sheridan, J. Christopher Culberson, Brian E. Mattioni, Marc A. Labroli, and Douglas C. Beshore
- Subjects
0301 basic medicine ,Prioritization ,Quantitative structure–activity relationship ,Drug discovery ,In silico ,Computational biology ,Biology ,Bioinformatics ,03 medical and health sciences ,030104 developmental biology ,Drug Discovery ,High-Throughput Screening Assays ,Molecular Medicine ,Selection (genetic algorithm) - Abstract
High-throughput screening (HTS) has enabled millions of compounds to be assessed for biological activity, but challenges remain in the prioritization of hit series. While biological, absorption, distribution, metabolism, excretion, and toxicity (ADMET), purity, and structural data are routinely used to select chemical matter for further follow-up, the scarcity of historical ADMET data for screening hits limits our understanding of early hit compounds. Herein, we describe a process that utilizes a battery of in-house quantitative structure–activity relationship (QSAR) models to generate in silico ADMET profiles for hit series to enable more complete characterizations of HTS chemical matter. These profiles allow teams to quickly assess hit series for desirable ADMET properties or suspected liabilities that may require significant optimization. Accordingly, these in silico data can direct ADMET experimentation and profoundly impact the progression of hit series. Several prospective examples are presented to ...
- Published
- 2017
16. Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency, Pharmacokinetics, and Metabolic Properties to Obtain Atropisomeric Quinolinone (AM-0466) that Affords Robust in Vivo Activity
- Author
-
Jessica Able, Benjamin C. Milgram, Loren Berry, Melanie Cooke, Liyue Huang, John Butler, Hongbing Huang, Violeta Yu, Kristin Taborn, John D. Roberts, Steven Altmann, Margaret Y. Chu-Moyer, John Yeoman, Jean Wang, Roman Shimanovich, Russell Graceffa, Matthew Weiss, Thomas Kornecook, Christopher P Ilch, Bryan D. Moyer, Christiane Boezio, Isaac E. Marx, Brian A. Sparling, Emily A. Peterson, Gwen Rescourio, Charles Kreiman, Elma Feric Bojic, Karina R. Vaida, Angel Guzman-Perez, Dawn Zhu, Hua Gao, Laurie B. Schenkel, Michael Jarosh, Hanh Nho Nguyen, Joseph Ligutti, Alessandro Boezio, Hakan Gunaydin, Daniel S. La, Thomas Dineen, Robert T. Fremeau, Robert S. Foti, Min-Hwa Jasmine Lin, Erin F. DiMauro, and John Stellwagen
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Bicyclic molecule ,CYP3A4 ,Stereochemistry ,Chemistry ,Sodium channel ,Sulfonamide ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,Drug Discovery ,Molecular Medicine ,Structure–activity relationship ,Selectivity ,CYP2C9 ,030217 neurology & neurosurgery - Abstract
Because of its strong genetic validation, NaV1.7 has attracted significant interest as a target for the treatment of pain. We have previously reported on a number of structurally distinct bicyclic heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. Herein, we report the discovery and optimization of a series of atropisomeric quinolinone sulfonamide inhibitors [Bicyclic sulfonamide compounds as sodium channel inhibitors and their preparation. WO 2014201206, 2014] of NaV1.7, which demonstrate nanomolar inhibition of NaV1.7 and exhibit high levels of selectivity over other sodium channel isoforms. After optimization of metabolic and pharmacokinetic properties, including PXR activation, CYP2C9 inhibition, and CYP3A4 TDI, several compounds were advanced into in vivo target engagement and efficacy models. When tested in mice, compound 39 (AM-0466) demonstrated robust pharmacodynamic activity in a NaV1.7-dependent model of histamine-induced pruritus (i...
- Published
- 2017
17. Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic Liabilities
- Author
-
Robert T. Fremeau, Isaac E. Marx, Emily A. Peterson, Charles Kreiman, Thomas Dineen, Hua Gao, Alessandro Boezio, Hakan Gunaydin, Min-Hwa Jasmine Lin, Steven Altmann, Elma Feric Bojic, Kristin Taborn, Robert S. Foti, Russell Graceffa, Daniel S. La, Liyue Huang, Matthew Weiss, Paul E. Rose, Angel Guzman-Perez, Beth D. Youngblood, Hongbing Huang, Violeta Yu, Dong Liu, Thomas Kornecook, Bryan D. Moyer, Howard Bregman, Hanh Nho Nguyen, Joseph Ligutti, Margaret Y. Chu-Moyer, Michael Jarosh, and Erin F. DiMauro
- Subjects
0301 basic medicine ,Pregnane X receptor ,CYP3A4 ,Chemistry ,Target engagement ,Pharmacology ,030226 pharmacology & pharmacy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Pharmacokinetics ,Pharmacodynamics ,Drug Discovery ,Lipophilicity ,NAV1 ,Molecular Medicine ,Potency - Abstract
Several reports have recently emerged regarding the identification of heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. The optimization of a series of internal NaV1.7 leads that address a number of metabolic liabilities including bioactivation, PXR activation, as well as CYP3A4 induction and inhibition led to the identification of potent and selective inhibitors that demonstrated favorable pharmacokinetic profiles and were devoid of the aforementioned liabilities. The key to achieving this within a series prone to transporter-mediated clearance was the identification of a small range of optimal cLogD values and the discovery of subtle PXR SAR that was not lipophilicity dependent. This enabled the identification of compound 20, which was advanced into a target engagement pharmacodynamic model where it exhibited robust reversal of histamine-induced scratching bouts in mice.
- Published
- 2017
18. Discovery of a Novel cGAMP Competitive Ligand of the Inactive Form of STING
- Author
-
Gottfried K. Schroeder, T. Ho, Altman, James P. Jewell, Matthew Lloyd Childers, Tony Siu, H. Hatch, Bo-Sheng Pan, J.M. Ellis, Brian M. Lacey, Hakan Gunaydin, Berengere Sauvagnat, G.A. Baltus, Shiyao Xu, and Charles A. Lesburg
- Subjects
010404 medicinal & biomolecular chemistry ,Sting ,010405 organic chemistry ,Chemistry ,Ligand ,Organic Chemistry ,Drug Discovery ,Druggability ,01 natural sciences ,Biochemistry ,Combinatorial chemistry ,0104 chemical sciences - Abstract
[Image: see text] Drugging large protein pockets is a challenge due to the need for higher molecular weight ligands, which generally possess undesirable physicochemical properties. In this communication, we highlight a strategy leveraging small molecule active site dimers to inhibit the large symmetric binding pocket in the STING protein. By taking advantage of the 2:1 binding stoichiometry, maximal buried interaction with STING protein can be achieved while maintaining the ligand physicochemical properties necessary for oral exposure. This mode of binding requires unique considerations for potency optimization including simultaneous optimization of protein–ligand as well as ligand–ligand interactions. Successful implementation of this strategy led to the identification of 18, which exhibits good oral exposure, slow binding kinetics, and functional inhibition of STING-mediated cytokine release.
- Published
- 2018
19. Structure-based design and development of (benz)imidazole pyridones as JAK1-selective kinase inhibitors
- Author
-
Yudith Garcia, Joon Jung, Craig R. Gibeau, Blair T. Lapointe, Rafael Fernandez, Sujal V. Deshmukh, Vladimir Simov, Jason D. Katz, Brian Kraybill, Sangita B. Patel, Jonathan R. Young, Hua Su, Fiona Elwood, Hakan Gunaydin, Tony Siu, and Christopher J. Dinsmore
- Subjects
Models, Molecular ,0301 basic medicine ,Pyridones ,Clinical Biochemistry ,Pharmaceutical Science ,Crystallography, X-Ray ,Biochemistry ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Ribose ,Humans ,Structure–activity relationship ,Kinome ,Protein Kinase Inhibitors ,Molecular Biology ,Janus kinase 1 ,Kinase ,Chemistry ,Organic Chemistry ,Janus Kinase 1 ,030104 developmental biology ,Tyrosine kinase 2 ,Drug Design ,030220 oncology & carcinogenesis ,Molecular Medicine ,Benzimidazoles ,Janus kinase ,Tyrosine kinase - Abstract
The mammalian Janus Kinases (JAK1, JAK2, JAK3 and TYK2) are intracellular, non-receptor tyrosine kinases whose activities have been associated in the literature and the clinic with a variety of hyperproliferative diseases and immunological disorders. At the onset of the program, it was hypothesized that a JAK1 selective compound over JAK2 could lead to an improved therapeutic index relative to marketed non-selective JAK inhibitors by avoiding the clinical AEs, such as anemia, presumably associated with JAK2 inhibition. During the course of the JAK1 program, a number of diverse chemical scaffolds were identified from both uHTS campaigns and de novo scaffold design. As part of this effort, a (benz)imidazole scaffold evolved via a scaffold-hopping exercise from a mature chemical series. Concurrent crystallography-driven exploration of the ribose pocket and the solvent front led to analogs with optimized kinome and JAK1 selectivities over the JAK2 isoform by targeting several residues unique to JAK1, such as Arg-879 and Glu-966.
- Published
- 2016
20. Probabilistic Approach to Generating MPOs and Its Application as a Scoring Function for CNS Drugs
- Author
-
Hakan Gunaydin
- Subjects
0301 basic medicine ,Computer science ,Organic Chemistry ,Probabilistic logic ,Bayesian inference ,computer.software_genre ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,Drug Discovery ,Probability distribution ,Data mining ,Function method ,computer ,Interpretability - Abstract
Multiparameter optimization (MPO) scoring functions are popular tools for providing guidance on how to design desired molecules in medicinal chemistry. The utility of a new probabilistic MPO (pMPO) scoring function method and its application as a scoring function for CNS drugs are described in this letter. In this new approach, a minimal number of statistically determined empirical boundaries is combined with the probability distribution of the desired molecules to define desirability functions. This approach attempts to minimize the number of parameters that define MPO scores while maintaining a high level of predictive power. Results obtained from a test-set of orally approved drugs show that the pMPO approach described here can be used to separate desired molecules from undesired ones with accuracy comparable to a Bayesian model with the advantage of better human interpretability. The application of this pMPO approach for blood-brain barrier penetrant drugs is also described.
- Published
- 2015
21. Identification of Kinases Responsible for p53-Dependent Autophagy
- Author
-
Mark E. Scott, Matthew G. Kortus, Edward Spooner, Ronan C. O'Hagan, Lisette P. Yco, Peter Fuller, Katie R. Martin, Stephanie L. Celano, Jeffrey P. MacKeigan, Stuart D. Shumway, Abigail R. Solitro, and Hakan Gunaydin
- Subjects
0301 basic medicine ,DNA damage ,02 engineering and technology ,CDC42 ,Biology ,Myotonic dystrophy ,Article ,03 medical and health sciences ,Downregulation and upregulation ,medicine ,lcsh:Science ,Multidisciplinary ,Functional Aspects of Cell Biology ,Effector ,Kinase ,Autophagy ,Cancer ,Cell Biology ,Biological Sciences ,021001 nanoscience & nanotechnology ,medicine.disease ,3. Good health ,Cell biology ,030104 developmental biology ,lcsh:Q ,0210 nano-technology - Abstract
Summary In cancer, autophagy is upregulated to promote cell survival and tumor growth during times of nutrient stress and can confer resistance to drug treatments. Several major signaling networks control autophagy induction, including the p53 tumor suppressor pathway. In response to DNA damage and other cellular stresses, p53 is stabilized and activated, while HDM2 binds to and ubiquitinates p53 for proteasome degradation. Thus blocking the HDM2-p53 interaction is a promising therapeutic strategy in cancer; however, the potential survival advantage conferred by autophagy induction may limit therapeutic efficacy. In this study, we leveraged an HDM2 inhibitor to identify kinases required for p53-dependent autophagy. Interestingly, we discovered that p53-dependent autophagy requires several kinases, including the myotonic dystrophy protein kinase-like alpha (MRCKα). MRCKα is a CDC42 effector reported to activate actin-myosin cytoskeletal reorganization. Overall, this study provides evidence linking MRCKα to autophagy and reveals additional insights into the role of kinases in p53-dependent autophagy., Graphical Abstract, Highlights • HDM2 inhibitors stabilize and activate p53 leading to robust autophagy induction • RNAi screen uncovers kinases involved in p53-dependent autophagy • ULK1 and the actin cytoskeleton kinase MRCKα mediate p53-induced autophagy, Biological Sciences; Cell Biology; Functional Aspects of Cell Biology
- Published
- 2018
22. 1,2,4-Triazolsulfone: A novel isosteric replacement of acylsulfonamides in the context of Na
- Author
-
Alessandro A, Boezio, Kristin, Andrews, Christiane, Boezio, Margaret, Chu-Moyer, Katrina W, Copeland, Erin F, DiMauro, Robert S, Foti, Robert T, Fremeau, Hua, Gao, Stephanie, Geuns-Meyer, Russell F, Graceffa, Hakan, Gunaydin, Hongbing, Huang, Daniel S, La, Joseph, Ligutti, Bryan D, Moyer, Emily A, Peterson, Violeta, Yu, and Matthew M, Weiss
- Subjects
Structure-Activity Relationship ,Sulfonamides ,Dose-Response Relationship, Drug ,Molecular Structure ,NAV1.7 Voltage-Gated Sodium Channel ,Microsomes, Liver ,Animals ,Humans ,Rats - Abstract
Recently, the identification of several classes of aryl sulfonamides and acyl sulfonamides that potently inhibit Na
- Published
- 2018
23. Informing the Selection of Screening Hit Series with in Silico Absorption, Distribution, Metabolism, Excretion, and Toxicity Profiles
- Author
-
John M, Sanders, Douglas C, Beshore, J Christopher, Culberson, James I, Fells, Jason E, Imbriglio, Hakan, Gunaydin, Andrew M, Haidle, Marc, Labroli, Brian E, Mattioni, Nunzio, Sciammetta, William D, Shipe, Robert P, Sheridan, Linda M, Suen, Andreas, Verras, Abbas, Walji, Elizabeth M, Joshi, and Tjerk, Bueters
- Subjects
Pharmacology ,Drug-Related Side Effects and Adverse Reactions ,Pharmaceutical Preparations ,Drug Discovery ,Animals ,Humans ,Quantitative Structure-Activity Relationship ,Computer Simulation ,Pharmacokinetics ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,High-Throughput Screening Assays - Abstract
High-throughput screening (HTS) has enabled millions of compounds to be assessed for biological activity, but challenges remain in the prioritization of hit series. While biological, absorption, distribution, metabolism, excretion, and toxicity (ADMET), purity, and structural data are routinely used to select chemical matter for further follow-up, the scarcity of historical ADMET data for screening hits limits our understanding of early hit compounds. Herein, we describe a process that utilizes a battery of in-house quantitative structure-activity relationship (QSAR) models to generate in silico ADMET profiles for hit series to enable more complete characterizations of HTS chemical matter. These profiles allow teams to quickly assess hit series for desirable ADMET properties or suspected liabilities that may require significant optimization. Accordingly, these in silico data can direct ADMET experimentation and profoundly impact the progression of hit series. Several prospective examples are presented to substantiate the value of this approach.
- Published
- 2017
24. The discovery of benzoxazine sulfonamide inhibitors of Na
- Author
-
Daniel S, La, Emily A, Peterson, Christiane, Bode, Alessandro A, Boezio, Howard, Bregman, Margaret Y, Chu-Moyer, James, Coats, Erin F, DiMauro, Thomas A, Dineen, Bingfan, Du, Hua, Gao, Russell, Graceffa, Hakan, Gunaydin, Angel, Guzman-Perez, Robert, Fremeau, Xin, Huang, Christopher, Ilch, Thomas J, Kornecook, Charles, Kreiman, Joseph, Ligutti, Min-Hwa, Jasmine Lin, Jeff S, McDermott, Isaac, Marx, David J, Matson, Stefan I, McDonough, Bryan D, Moyer, Hanh, Nho Nguyen, Kristin, Taborn, Violeta, Yu, and Matthew M, Weiss
- Subjects
Male ,Voltage-Gated Sodium Channel Blockers ,Analgesics ,Sulfonamides ,NAV1.7 Voltage-Gated Sodium Channel ,Pain ,Benzoxazines ,Rats ,Mice, Inbred C57BL ,Molecular Docking Simulation ,Rats, Sprague-Dawley ,Mice ,Animals ,Humans - Abstract
The voltage-gated sodium channel Na
- Published
- 2017
25. Sulfonamides as Selective Na
- Author
-
Russell F, Graceffa, Alessandro A, Boezio, Jessica, Able, Steven, Altmann, Loren M, Berry, Christiane, Boezio, John R, Butler, Margaret, Chu-Moyer, Melanie, Cooke, Erin F, DiMauro, Thomas A, Dineen, Elma, Feric Bojic, Robert S, Foti, Robert T, Fremeau, Angel, Guzman-Perez, Hua, Gao, Hakan, Gunaydin, Hongbing, Huang, Liyue, Huang, Christopher, Ilch, Michael, Jarosh, Thomas, Kornecook, Charles R, Kreiman, Daniel S, La, Joseph, Ligutti, Benjamin C, Milgram, Min-Hwa Jasmine, Lin, Isaac E, Marx, Hanh N, Nguyen, Emily A, Peterson, Gwen, Rescourio, John, Roberts, Laurie, Schenkel, Roman, Shimanovich, Brian A, Sparling, John, Stellwagen, Kristin, Taborn, Karina R, Vaida, Jean, Wang, John, Yeoman, Violeta, Yu, Dawn, Zhu, Bryan D, Moyer, and Matthew M, Weiss
- Subjects
Voltage-Gated Sodium Channel Blockers ,Analgesics ,Sulfonamides ,Pruritus ,NAV1.7 Voltage-Gated Sodium Channel ,Pain ,Quinolones ,Cell Line ,Rats ,Mice, Inbred C57BL ,Molecular Docking Simulation ,Structure-Activity Relationship ,Dogs ,Animals ,Protein Isoforms ,Capsaicin ,Histamine - Abstract
Because of its strong genetic validation, Na
- Published
- 2017
26. Sulfonamides as Selective Na
- Author
-
Matthew M, Weiss, Thomas A, Dineen, Isaac E, Marx, Steven, Altmann, Alessandro, Boezio, Howard, Bregman, Margaret, Chu-Moyer, Erin F, DiMauro, Elma, Feric Bojic, Robert S, Foti, Hua, Gao, Russell, Graceffa, Hakan, Gunaydin, Angel, Guzman-Perez, Hongbing, Huang, Liyue, Huang, Michael, Jarosh, Thomas, Kornecook, Charles R, Kreiman, Joseph, Ligutti, Daniel S, La, Min-Hwa Jasmine, Lin, Dong, Liu, Bryan D, Moyer, Hanh N, Nguyen, Emily A, Peterson, Paul E, Rose, Kristin, Taborn, Beth D, Youngblood, Violeta, Yu, and Robert T, Fremeau
- Subjects
Male ,Voltage-Gated Sodium Channel Blockers ,Receptors, Steroid ,Sulfonamides ,Pruritus ,NAV1.7 Voltage-Gated Sodium Channel ,Pregnane X Receptor ,Isoquinolines ,Cell Line ,Rats ,Mice, Inbred C57BL ,Structure-Activity Relationship ,Dogs ,Enzyme Induction ,Animals ,Cytochrome P-450 CYP3A ,Cytochrome P-450 CYP3A Inhibitors ,Humans ,Histamine - Abstract
Several reports have recently emerged regarding the identification of heteroarylsulfonamides as Na
- Published
- 2017
27. Correction to 'Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics to Enable in Vivo Target Engagement'
- Author
-
Robert T. Fremeau, Robert S. Foti, Hanh Nho Nguyen, Jeff S. McDermott, Jean Wang, Christiane Bode, Bingfan Du, Joseph Ligutti, Thomas Dineen, Hua Gao, Thomas Kornecook, Jonathan Roberts, Brian E. Hall, Charles Kreiman, Liyue Huang, Matthew Weiss, Jessica Able, Min-Hwa Jasmine Lin, Paul E. Rose, Isaac E. Marx, Emily A. Peterson, Violeta Yu, Erin F. DiMauro, Bryan D. Moyer, Daniel S. La, Beth D. Youngblood, Dong Liu, Margaret Y. Chu-Moyer, Hakan Gunaydin, and Howard Bregman
- Subjects
Pharmacokinetics ,business.industry ,In vivo ,Organic Chemistry ,Drug Discovery ,Target engagement ,Potency ,Medicine ,Pharmacology ,business ,Biochemistry - Abstract
Human genetic evidence has identified the voltage-gated sodium channel Na
- Published
- 2017
28. The Discovery and Hit-to-Lead Optimization of Tricyclic Sulfonamides as Potent and Efficacious Potentiators of Glycine Receptors
- Author
-
Shawn Ayube, Hao Chen, Yohannes Teffera, Jacinthe Gingras, Paul L. Shaffer, Paul Krolikowski, Angel Guzman-Perez, Kristin L. Andrews, Hakan Gunaydin, Richard Thomas Lewis, Klaus Michelsen, Jiali Hu, Liyue Huang, Sonya G. Lehto, Pamela Pegman, Erin F. DiMauro, Howard Bregman, Shuyan Yi, Xin Huang, Maosheng Zhang, Matthew H. Plant, and Jeffrey R. Simard
- Subjects
0301 basic medicine ,Male ,Allosteric regulation ,Pharmacology ,In Vitro Techniques ,Inhibitory postsynaptic potential ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Receptors, Glycine ,In vivo ,Drug Discovery ,Animals ,Humans ,Glycine receptor ,chemistry.chemical_classification ,Sulfonamides ,Chemistry ,Hit to lead ,Potentiator ,Mice, Inbred C57BL ,030104 developmental biology ,HEK293 Cells ,Molecular Medicine ,030217 neurology & neurosurgery ,Ex vivo ,Tricyclic - Abstract
Current pain therapeutics suffer from undesirable psychotropic and sedative side effects, as well as abuse potential. Glycine receptors (GlyRs) are inhibitory ligand-gated ion channels expressed in nerves of the spinal dorsal horn, where their activation is believed to reduce transmission of painful stimuli. Herein, we describe the identification and hit-to-lead optimization of a novel class of tricyclic sulfonamides as allosteric GlyR potentiators. Initial optimization of high-throughput screening (HTS) hit 1 led to the identification of 3, which demonstrated ex vivo potentiation of glycine-activated current in mouse dorsal horn neurons from spinal cord slices. Further improvement of potency and pharmacokinetics produced in vivo proof-of-concept tool molecule 20 (AM-1488), which reversed tactile allodynia in a mouse spared-nerve injury (SNI) model. Additional structural optimization provided highly potent potentiator 32 (AM-3607), which was cocrystallized with human GlyRα3cryst to afford the first descri...
- Published
- 2016
29. Development of Novel Dual Binders as Potent, Selective, and Orally Bioavailable Tankyrase Inhibitors
- Author
-
Zihao Hua, Bryan Egge, Erin L. Mullady, Hakan Gunaydin, John L. Buchanan, Steve Schneider, Xin Huang, Yohannes Teffera, Renee Emkey, Howard Bregman, Randy Serafino, Mary K. Stanton, Erin F. DiMauro, Jingzhou Liu, Virginia Berry, Douglas Saffran, Angel Guzman-Perez, Jennifer Dovey, Liyue Huang, Craig A. Strathdee, Susan M. Turci, Yan Gu, Paul S. Andrews, John Newcomb, Cindy Wilson, Ankita Mishra, Lisa Acquaviva, and Nagasree Chakka
- Subjects
Models, Molecular ,Tankyrases ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Chemistry ,Drug discovery ,Poly ADP ribose polymerase ,Wnt signaling pathway ,Administration, Oral ,Biological Availability ,Structure-Activity Relationship ,Biochemistry ,Drug Discovery ,biology.protein ,Humans ,Molecular Medicine ,Structure–activity relationship ,Enzyme Inhibitors ,Pharmacophore ,Axin Protein ,Polymerase - Abstract
Tankyrases (TNKS1 and TNKS2) are proteins in the poly ADP-ribose polymerase (PARP) family. They have been shown to directly bind to axin proteins, which negatively regulate the Wnt pathway by promoting β-catenin degradation. Inhibition of tankyrases may offer a novel approach to the treatment of APC-mutant colorectal cancer. Hit compound 8 was identified as an inhibitor of tankyrases through a combination of substructure searching of the Amgen compound collection based on a minimal binding pharmacophore hypothesis and high-throughput screening. Herein we report the structure- and property-based optimization of compound 8 leading to the identification of more potent and selective tankyrase inhibitors 22 and 49 with improved pharmacokinetic properties in rodents, which are well suited as tool compounds for further in vivo validation studies.
- Published
- 2013
30. Discovery of Novel, Induced-Pocket Binding Oxazolidinones as Potent, Selective, and Orally Bioavailable Tankyrase Inhibitors
- Author
-
Angel Guzman-Perez, Erin L. Mullady, Randy Serafino, Erin F. DiMauro, Hakan Gunaydin, Xin Huang, Liyue Huang, Jingzhou Liu, Virginia Berry, Bryan Egge, Nagasree Chakka, Cindy Wilson, Steve Schneider, John Newcomb, Yan Gu, Yohannes Teffera, Susan M. Turci, Ankita Mishra, Paul S. Andrews, Howard Bregman, and Craig A. Strathdee
- Subjects
Models, Molecular ,Molecular model ,Poly ADP ribose polymerase ,Administration, Oral ,Biological Availability ,In Vitro Techniques ,Pharmacology ,medicine.disease_cause ,Mice ,Structure-Activity Relationship ,Pharmacokinetics ,Transcription (biology) ,Drug Discovery ,medicine ,Animals ,Axin Protein ,Oxazolidinones ,Tankyrases ,Binding Sites ,Oncogene ,Chemistry ,Stereoisomerism ,Rats ,Bioavailability ,Biochemistry ,Microsomes, Liver ,Molecular Medicine ,Benzimidazoles ,Carcinogenesis - Abstract
Tankyrase (TNKS) is a poly-ADP-ribosylating protein (PARP) whose activity suppresses cellular axin protein levels and elevates β-catenin concentrations, resulting in increased oncogene expression. The inhibition of tankyrase (TNKS1 and 2) may reduce the levels of β-catenin-mediated transcription and inhibit tumorigenesis. Compound 1 is a previously described moderately potent tankyrase inhibitor that suffers from poor pharmacokinetic properties. Herein, we describe the utilization of structure-based design and molecular modeling toward novel, potent, and selective tankyrase inhibitors with improved pharmacokinetic properties (39, 40).
- Published
- 2013
31. Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics to Enable in Vivo Target Engagement
- Author
-
Min-Hwa Jasmine Lin, Paul E. Rose, Violeta Yu, Hakan Gunaydin, Robert T. Fremeau, Charles Kreiman, Daniel S. La, Joseph Ligutti, Matthew Weiss, Beth D. Youngblood, Thomas Dineen, Thomas Kornecook, Dong Liu, Bingfan Du, Brian E. Hall, Erin F. DiMauro, Jean Wang, Isaac E. Marx, Robert S. Foti, Emily A. Peterson, Jessica Able, Liyue Huang, Margaret Chu-Moyer, Jeff S. McDermott, Christiane Bode, Bryan D. Moyer, Howard Bregman, Jonathan Roberts, and Hua Gao
- Subjects
biology ,010405 organic chemistry ,Sodium channel ,Organic Chemistry ,Sulfonamide (medicine) ,Nav1.5 ,Pharmacology ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,chemistry.chemical_compound ,chemistry ,Pharmacokinetics ,In vivo ,Drug Discovery ,biology.protein ,Quinazoline ,medicine ,Potency ,Dosing ,medicine.drug - Abstract
Human genetic evidence has identified the voltage-gated sodium channel NaV1.7 as an attractive target for the treatment of pain. We initially identified naphthalene sulfonamide 3 as a potent and selective inhibitor of NaV1.7. Optimization to reduce biliary clearance by balancing hydrophilicity and hydrophobicity (Log D) while maintaining NaV1.7 potency led to the identification of quinazoline 16 (AM-2099). Compound 16 demonstrated a favorable pharmacokinetic profile in rat and dog and demonstrated dose-dependent reduction of histamine-induced scratching bouts in a mouse behavioral model following oral dosing.
- Published
- 2016
32. Application of a Parallel Synthetic Strategy in the Discovery of Biaryl Acyl Sulfonamides as Efficient and Selective NaV1.7 Inhibitors
- Author
-
Robert T. Fremeau, Joseph Ligutti, Paul E. Rose, Dong Liu, Elma Feric Bojic, Yan Wang, Hongbing Huang, Violeta Yu, Thomas Kornecook, Angel Guzman-Perez, Laurie B. Schenkel, Hakan Gunaydin, Stephen Altmann, Jean Wang, Kristin Taborn, Matthew Weiss, Margaret Y. Chu-Moyer, Michael Jarosh, Howard Bregman, Hua Gao, Robert S. Foti, Bryan D. Moyer, Brian E. Hall, Loren Berry, Nagasree Chakka, Josie Lee, Daniel Ortuno, and Erin F. DiMauro
- Subjects
0301 basic medicine ,Male ,Stereochemistry ,Cell Line ,03 medical and health sciences ,Radioligand Assay ,Structure-Activity Relationship ,0302 clinical medicine ,Drug Discovery ,Structure–activity relationship ,Animals ,Humans ,chemistry.chemical_classification ,Voltage-Gated Sodium Channel Blockers ,Sulfonamides ,Chemistry ,Pruritus ,NAV1.7 Voltage-Gated Sodium Channel ,Ligand (biochemistry) ,Sulfonamide ,Rats ,Mice, Inbred C57BL ,Molecular Docking Simulation ,030104 developmental biology ,Liver metabolism ,Benzamides ,Microsomes, Liver ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Female ,Selectivity ,030217 neurology & neurosurgery ,Histamine - Abstract
The majority of potent and selective hNaV1.7 inhibitors possess common pharmacophoric features that include a heteroaryl sulfonamide headgroup and a lipophilic aromatic tail group. Recently, reports of similar aromatic tail groups in combination with an acyl sulfonamide headgroup have emerged, with the acyl sulfonamide bestowing levels of selectivity over hNaV1.5 comparable to the heteroaryl sulfonamide. Beginning with commercially available carboxylic acids that met selected pharmacophoric requirements in the lipophilic tail, a parallel synthetic approach was applied to rapidly generate the derived acyl sulfonamides. A biaryl acyl sulfonamide hit from this library was elaborated, optimizing for potency and selectivity with attention to physicochemical properties. The resulting novel leads are potent, ligand and lipophilic efficient, and selective over hNaV1.5. Representative lead 36 demonstrates selectivity over other human NaV isoforms and good pharmacokinetics in rodents. The biaryl acyl sulfonamides reported herein may also offer ADME advantages over known heteroaryl sulfonamide inhibitors.
- Published
- 2016
33. Stacking with No Planarity?
- Author
-
Michael D. Bartberger and Hakan Gunaydin
- Subjects
0301 basic medicine ,Computer science ,Organic Chemistry ,Stacking ,computer.software_genre ,Biochemistry ,humanities ,Planarity testing ,03 medical and health sciences ,Crystallography ,030104 developmental biology ,Drug Discovery ,Side chain ,Molecule ,Data mining ,computer - Abstract
This viewpoint describes the results obtained from matched molecular pair analyses and quantum mechanics calculations that show unsaturated rings found in drug-like molecules may be replaced with their saturated counterparts without losing potency even if they are engaged in stacking interactions with the side chains of aromatic residues.
- Published
- 2016
34. Optimization of a Novel Quinazolinone-Based Series of Transient Receptor Potential A1 (TRPA1) Antagonists Demonstrating Potent in Vivo Activity
- Author
-
Beth D. Youngblood, Alessandro Boezio, Stephanie D. Geuns-Meyer, Maosheng Zhang, Angel Guzman-Perez, Laurie B. Schenkel, Yohannes Teffera, Weiya Wang, Philip R. Olivieri, Renee Emkey, Hakan Gunaydin, Josie H. Lee, Holly L. Deak, Violeta Yu, Russell Graceffa, Sonya G. Lehto, and Narender R. Gavva
- Subjects
0301 basic medicine ,Models, Molecular ,Biological Transport, Active ,Nerve Tissue Proteins ,CHO Cells ,Pharmacology ,In Vitro Techniques ,Madin Darby Canine Kidney Cells ,03 medical and health sciences ,Transient receptor potential channel ,chemistry.chemical_compound ,Structure-Activity Relationship ,0302 clinical medicine ,Cricetulus ,Dogs ,Transient Receptor Potential Channels ,Pharmacokinetics ,In vivo ,Drug Discovery ,Potency ,Animals ,Humans ,IC50 ,Quinazolinone ,TRPA1 Cation Channel ,Pain Measurement ,Oxadiazoles ,Dose-Response Relationship, Drug ,Antagonist ,food and beverages ,Small molecule ,High-Throughput Screening Assays ,Rats ,030104 developmental biology ,chemistry ,Purines ,Microsomes, Liver ,Quinazolines ,Molecular Medicine ,Calcium Channels ,psychological phenomena and processes ,030217 neurology & neurosurgery - Abstract
There has been significant interest in developing a transient receptor potential A1 (TRPA1) antagonist for the treatment of pain due to a wealth of data implicating its role in pain pathways. Despite this, identification of a potent small molecule tool possessing pharmacokinetic properties allowing for robust in vivo target coverage has been challenging. Here we describe the optimization of a potent, selective series of quinazolinone-based TRPA1 antagonists. High-throughput screening identified 4, which possessed promising potency and selectivity. A strategy focused on optimizing potency while increasing polarity in order to improve intrinsic clearance culminated with the discovery of purinone 27 (AM-0902), which is a potent, selective antagonist of TRPA1 with pharmacokinetic properties allowing for30-fold coverage of the rat TRPA1 IC50 in vivo. Compound 27 demonstrated dose-dependent inhibition of AITC-induced flinching in rats, validating its utility as a tool for interrogating the role of TRPA1 in in vivo pain models.
- Published
- 2016
35. De Novo Prediction of P-Glycoprotein-Mediated Efflux Liability for Druglike Compounds
- Author
-
Yaxiong Sun, Matthew Weiss, and Hakan Gunaydin
- Subjects
integumentary system ,In vivo ,In silico ,Organic Chemistry ,Drug Discovery ,biology.protein ,Efflux ,Computational biology ,Biology ,Bioinformatics ,Biochemistry ,P-glycoprotein - Abstract
P-glycoprotein (Pgp) is capable of recognizing and transporting a wide range of chemically diverse compounds in vivo. Overcoming Pgp-mediated efflux can represent a significant challenge when penetration into the central nervous system is required or within the context of developing anticancer therapies. While numerous in silico models have been developed to predict Pgp-mediated efflux, these models rely on training sets and are best suited to make interpolations. Therefore, it is desirable to develop ab initio models that can be used to predict efflux liabilities. Herein, we present a de novo method that can be used to predict Pgp-mediated efflux potential for druglike compounds. A model, which correlates the computed solvation free energy differences obtained in water and chloroform with Pgp-mediated efflux (in logarithmic scale), was successful in predicting Pgp efflux ratios for a wide range of chemically diverse compounds with a R(2) and root-mean-square error of 0.65 and 0.29, respectively.
- Published
- 2012
36. Structure-Based Design of Potent and Selective CK1γ Inhibitors
- Author
-
Xin Huang, Susan M. Turci, Zihao Hua, Randy Serafino, Liyue Huang, Jin Tang, Jason Brooks Human, Ryan White, Lisa Acquaviva, Jennifer Dovey, Barbara Grubinska, Oleg Epstein, Doug Saffran, Huilin Zhao, Steve Schneider, Howard Bregman, Hongbing Huang, Anne B. O’Connor, Violeta Yu, John Newcomb, Jonathan T. Goldstein, Nagasree Chakka, Matthew W. Martin, Vinod F. Patel, Craig A. Strathdee, Virginia Berry, Alexander M. Long, Cindy Wilson, Erin F. DiMauro, and Hakan Gunaydin
- Subjects
Gene isoform ,chemistry.chemical_classification ,business.industry ,Organic Chemistry ,Wnt signaling pathway ,LRP6 ,Pharmacology ,Biochemistry ,Enzyme ,chemistry ,Drug Discovery ,Cancer research ,Medicine ,Phosphorylation ,Casein kinase 1 ,Signal transduction ,business ,Cell potency - Abstract
Aberrant activation of the Wnt pathway is believed to drive the development and growth of some cancers. The central role of CK1γ in Wnt signal transduction makes it an attractive target for the treatment of Wnt-pathway dependent cancers. We describe a structure-based approach that led to the discovery of a series of pyridyl pyrrolopyridinones as potent and selective CK1γ inhibitors. These compounds exhibited good enzyme and cell potency, as well as selectivity against other CK1 isoforms. A single oral dose of compound 13 resulted in significant inhibition of LRP6 phosphorylation in a mouse tumor PD model.
- Published
- 2012
37. Computation of Accurate Activation Barriers for Methyl-Transfer Reactions of Sulfonium and Ammonium Salts in Aqueous Solution
- Author
-
Kendall N. Houk, William L. Jorgensen, Hakan Gunaydin, and Orlando Acevedo
- Subjects
Tetramethylammonium ,Aqueous solution ,Sulfonium ,Solvation ,Polarizable continuum model ,Computer Science Applications ,Reaction coordinate ,chemistry.chemical_compound ,Delocalized electron ,chemistry ,Physical chemistry ,Density functional theory ,Physics::Chemical Physics ,Physical and Theoretical Chemistry - Abstract
The energetics of methyl-transfer reactions from dimethylammonium, tetramethylammonium, and trimethylsulfonium to dimethylamine were computed with density functional theory, MP2, CBS-QB3, and quantum mechanics/molecular mechanics (QM/MM) Monte Carlo methods. At the CBS-QB3 level, the gas-phase activation enthalpies are computed to be 9.9, 15.3, and 7.9 kcal/mol, respectively. MP2/6-31+G(d,p) activation enthalpies are in best agreement with the CBS-QB3 results. The effects of aqueous solvation on these reactions were studied with polarizable continuum model, generalized Born/surface area (GB/SA), and QM/MM Monte Carlo simulations utilizing free-energy perturbation theory in which the PDDG/PM3 semiempirical Hamiltonian for the QM and explicit TIP4P water molecules in the MM region were used. In the aqueous phase, all of these reactions proceed more slowly when compared to the gas phase, since the charged reactants are stabilized more than the transition structure geometries with delocalized positive charges. In order to obtain the aqueous-phase activation free energies, the gas-phase activation free energies were corrected with the solvation free energies obtained from single-point conductor-like polarizable continuum model and GB/SA calculations for the stationary points along the reaction coordinate.
- Published
- 2015
38. Quantum Mechanical Design of Enzyme Active Sites
- Author
-
Fernando R. Clemente, Jason DeChancie, K. N. Houk, T. M. Handel,† and, Hakan Gunaydin, Adam J.T. Smith, Arnab B. Chowdry, and Xiyun Zhang
- Subjects
Models, Molecular ,Reaction mechanism ,Proline ,Stereochemistry ,Context (language use) ,Naphthols ,Catalysis ,Substrate Specificity ,Nitrophenols ,Reaction rate ,Cocaine ,Isomerism ,Aldol reaction ,Computational chemistry ,Molecule ,Acrolein ,Aldehydes ,Binding Sites ,Molecular Structure ,biology ,Chemistry ,Hydrolysis ,Organic Chemistry ,Water ,Active site ,Sarin ,Enzymes ,Enzyme Activation ,biology.protein ,Quantum Theory ,Peptides ,Isomerization - Abstract
The design of active sites has been carried out using quantum mechanical calculations to predict the rate-determining transition state of a desired reaction in presence of the optimal arrangement of catalytic functional groups (theozyme). Eleven versatile reaction targets were chosen, including hydrolysis, dehydration, isomerization, aldol, and Diels-Alder reactions. For each of the targets, the predicted mechanism and the rate-determining transition state (TS) of the uncatalyzed reaction in water is presented. For the rate-determining TS, a catalytic site was designed using naturalistic catalytic units followed by an estimation of the rate acceleration provided by a reoptimization of the catalytic site. Finally, the geometries of the sites were compared to the X-ray structures of related natural enzymes. Recent advances in computational algorithms and power, coupled with successes in computational protein design, have provided a powerful context for undertaking such an endeavor. We propose that theozymes are excellent candidates to serve as the active site models for design processes.
- Published
- 2008
39. How similar are enzyme active site geometries derived from quantum mechanical theozymes to crystal structures of enzyme-inhibitor complexes? Implications for enzyme design
- Author
-
Kendall N. Houk, Hakan Gunaydin, Xiyun Zhang, Yi-Lei Zhao, Adam J. T. Smith, Fernando R. Clemente, and Jason DeChancie
- Subjects
Bacillus ,Crystal structure ,Crystallography, X-Ray ,Biochemistry ,Catalysis ,Protein Structure, Secondary ,Article ,Substrate Specificity ,Protein structure ,Pseudomonas ,Escherichia coli ,Animals ,Humans ,Molecular Biology ,Binding Sites ,biology ,Chemistry ,Hydrogen bond ,Active site ,Hydrogen Bonding ,Models, Theoretical ,Enzymes ,Crystallography ,Models, Chemical ,Covalent bond ,Enzyme inhibitor ,biology.protein ,Quantum Theory ,Cattle ,Density functional theory ,Protein Binding - Abstract
Quantum mechanical optimizations of theoretical enzymes (theozymes), which are predicted catalytic arrays of biological functionalities stabilizing a transition state, have been carried out for a set of nine diverse enzyme active sites. For each enzyme, the theozyme for the rate-determining transition state plus the catalytic groups modeled by side-chain mimics was optimized using B3LYP/6–31G(d) or, in one case, HF/3–21G(d) quantum mechanical calculations. To determine if the theozyme can reproduce the natural evolutionary catalytic geometry, the positions of optimized catalytic atoms, i.e., covalent, partial covalent, or stabilizing interactions with transition state atoms, are compared to the positions of the atoms in the X-ray crystal structure with a bound inhibitor. These structure comparisons are contrasted to computed substrate–active site structures surrounded by the same theozyme residues. The theozyme/transition structure is shown to predict geometries of active sites with an average RMSD of 0.64 Å from the crystal structure, while the RMSD for the bound intermediate complexes are significantly higher at 1.42 Å. The implications for computational enzyme design are discussed.
- Published
- 2007
40. An Experimental and Computational Approach to Defining Structure/Reactivity Relationships for Intramolecular Addition Reactions to Bicyclic Epoxonium Ions
- Author
-
Paul E. Floreancig, Hakan Gunaydin, Kendall N. Houk, and Shuangyi Wan
- Subjects
Models, Molecular ,Addition reaction ,Molecular Structure ,Bicyclic molecule ,Chemistry ,Stereochemistry ,Oxocarbenium ,General Chemistry ,Biochemistry ,Article ,Catalysis ,Transition state ,Ion ,Colloid and Surface Chemistry ,Intramolecular force ,Epoxy Compounds ,Molecule ,Computer Simulation ,Selectivity - Abstract
In this manuscript we report that oxidative cleavage reactions can be used to form oxocarbenium ions that react with pendent epoxides to form bicyclic epoxonium ions as an entry to the formation of cyclic oligoether compounds. Bicyclic epoxonium ion structure was shown to have a dramatic impact on the ratio of exo- to endo-cyclization reactions, with bicyclo[4.1.0] intermediates showing a strong preference for endo-closures and bicyclo[3.1.0] intermediates showing a preference for exo-closures. Computational studies on the structures and energetics of the transition states using the B3LYP/6-31G(d) method provide substantial insight into the origins of this selectivity.
- Published
- 2007
41. Structure-based design of 2-aminopyridine oxazolidinones as potent and selective tankyrase inhibitors
- Author
-
Virginia Berry, Steve Schneider, Liyue Huang, Doug Saffran, Cindy Wilson, Randy Serafino, Angel Guzman-Perez, Lisa Acquaviva, Hongbing Huang, Erin F. DiMauro, Hakan Gunaydin, Howard Bregman, Jennifer Dovey, and Xin Huang
- Subjects
Gene isoform ,chemistry.chemical_classification ,Nicotinamide ,Poly ADP ribose polymerase ,Organic Chemistry ,Mutant ,Wnt signaling pathway ,Cancer ,Biology ,medicine.disease ,Biochemistry ,chemistry.chemical_compound ,Enzyme ,chemistry ,Drug Discovery ,medicine ,Cell potency - Abstract
Aberrant activation of the Wnt pathway has been implicated in the development and formation of many cancers. TNKS inhibition has been shown to antagonize Wnt signaling via Axin stabilization in APC mutant colon cancer cell lines. We employed structure-based design to identify a series of 2-aminopyridine oxazolidinones as potent and selective TNKS inhibitors. These compounds exhibited good enzyme and cell potency as well as selectivity over other PARP isoforms. Co-crystal structures of these 2-aminopyridine oxazolidinones complexed to TNKS reveal an induced-pocket binding mode that does not involve interactions with the nicotinamide binding pocket. Oral dosing of lead compounds 3 and 4 resulted in significant effects on several Wnt-pathway biomarkers in a three day DLD-1 mouse tumor PD model.
- Published
- 2013
42. Discovery of a class of novel tankyrase inhibitors that bind to both the nicotinamide pocket and the induced pocket
- Author
-
Yan Gu, Hakan Gunaydin, Cindy Wilson, Steve Schneider, Erin F. DiMauro, Xin Huang, and Howard Bregman
- Subjects
Models, Molecular ,Niacinamide ,Tankyrases ,Binding Sites ,Nicotinamide ,Chemistry ,Drug discovery ,Stereochemistry ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Biochemistry ,Drug Discovery ,Molecular Medicine ,Inhibitory concentration 50 ,Humans ,Binding site ,Enzyme Inhibitors - Abstract
Potent and selective inhibitors of tankyrases have recently been characterized to bind to an induced pocket. Here we report the identification of a novel potent and selective tankyrase inhibitor that binds to both the nicotinamide pocket and the induced pocket. The crystal structure of human TNKS1 in complex with this “dual-binder” provides a molecular basis for their strong and specific interactions and suggests clues for the further development of tankyrase inhibitors.
- Published
- 2013
43. 2-Phenylamino-6-cyano-1H-benzimidazole-based isoform selective casein kinase 1 gamma (CK1γ) inhibitors
- Author
-
Elizabeth M. Doherty, Stephanie J. Mercede, Vinod F. Patel, Susan M. Turci, Hakan Gunaydin, Cindy Wilson, Jie Yan, Hongbing Huang, Jonathan T. Goldstein, Zihao Hua, Nagasree Chakka, John Newcomb, Erin F. DiMauro, Howard Bregman, Matthew W. Martin, and Xin Huang
- Subjects
Models, Molecular ,Benzimidazole ,Clinical Biochemistry ,Pharmaceutical Science ,Mitogen-activated protein kinase kinase ,Biochemistry ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Casein Kinase I ,Drug Discovery ,Casein kinase 2, alpha 1 ,Structure–activity relationship ,Animals ,Humans ,Protein Isoforms ,Molecular Biology ,Protein Kinase Inhibitors ,beta Catenin ,Binding Sites ,Organic Chemistry ,Rats ,chemistry ,Molecular Medicine ,Cyclin-dependent kinase 9 ,Benzimidazoles ,Casein kinase 1 ,Casein kinase 2 - Abstract
Screening of the Amgen compound library led to the identification of 2-phenylamino-6-cyano-1H-benzimidazole 1a as a potent CK1 gamma inhibitor with excellent kinase selectivity and unprecedented CK1 isoform selectivity. Further structure-based optimization of this series resulted in the discovery of 1h which possessed good enzymatic and cellular potency, excellent CK1 isoform and kinase selectivity, and acceptable pharmacokinetic properties.
- Published
- 2012
44. Novel Binding Mode of a Potent and Selective Tankyrase Inhibitor
- Author
-
Hakan Gunaydin, Xin Huang, and Yan Gu
- Subjects
Models, Molecular ,Drugs and Devices ,Drug Research and Development ,Telomere Pathway ,Molecular Sequence Data ,lcsh:Medicine ,Biology ,Crystallography, X-Ray ,Biochemistry ,Tankyrases ,Molecular Cell Biology ,Chemical Biology ,Humans ,Amino Acid Sequence ,Binding site ,Enzyme Inhibitors ,lcsh:Science ,Wnt Signaling Pathway ,Multidisciplinary ,Binding Sites ,Molecular Structure ,Sequence Homology, Amino Acid ,lcsh:R ,Wnt signaling pathway ,Signaling Cascades ,Recombinant Proteins ,Cancer treatment ,Protein Structure, Tertiary ,Chemistry ,Medicine ,lcsh:Q ,Research Article ,Signal Transduction - Abstract
Tankyrases (TNKS1 and TNKS2) are key regulators of cellular processes such as telomere pathway and Wnt signaling. IWRs (inhibitors of Wnt response) have recently been identified as potent and selective inhibitors of tankyrases. However, it is not clear how these IWRs interact with tankyrases. Here we report the crystal structure of the catalytic domain of human TNKS1 in complex with IWR2, which reveals a novel binding site for tankyrase inhibitors. The TNKS1/IWR2 complex provides a molecular basis for their strong and specific interactions and suggests clues for further development of tankyrase inhibitors.
- Published
- 2012
45. Discovery of potent and highly selective thienopyridine Janus kinase 2 inhibitors
- Author
-
Josie Lee, Shen-Wu Wang, Alan C. Cheng, Elizabeth M. Doherty, Philip R. Olivieri, Qian Wan, Hakan Gunaydin, Robert D. Loberg, Joseph L. Kim, Stephanie D. Geuns-Meyer, Liqin Liu, Xin Huang, Laurie B. Schenkel, Jin Tang, Yan Gu, Renee Emkey, Mary C. Wells, Jeanne Pistillo, Holly L. Deak, Bin Wu, Violeta Yu, and Hui-Ling Wang
- Subjects
Models, Molecular ,Cell Membrane Permeability ,Thienopyridine ,Thienopyridines ,Swine ,Crystallography, X-Ray ,Structure-Activity Relationship ,hemic and lymphatic diseases ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Animals ,Humans ,Protein Kinase Inhibitors ,Janus kinase 2 ,biology ,Janus kinase 1 ,Kinase ,Drug discovery ,Chemistry ,Janus Kinase 1 ,Janus Kinase 2 ,Small molecule ,Protein Structure, Tertiary ,Protein kinase domain ,Biochemistry ,biology.protein ,Leukocytes, Mononuclear ,Molecular Medicine - Abstract
Developing Janus kinase 2 (Jak2) inhibitors has become a significant focus for small molecule drug discovery programs in recent years due to the identification of a Jak2 gain-of-function mutation in the majority of patients with myeloproliferative disorders (MPD). Here, we describe the discovery of a thienopyridine series of Jak2 inhibitors that culminates with compounds showing 100- to >500-fold selectivity over the related Jak family kinases in enzyme assays. Selectivity for Jak2 was also observed in TEL-Jak cellular assays, as well as in cytokine-stimulated peripheral blood mononuclear cell (PBMC) and whole blood assays. X-ray cocrystal structures of 8 and 19 bound to the Jak2 kinase domain aided structure–activity relationship efforts and, along with a previously reported small molecule X-ray cocrystal structure of the Jak1 kinase domain, provided structural rationale for the observed high levels of Jak2 selectivity.
- Published
- 2011
46. Mechanisms of Peroxynitrite Mediated Nitration of Tyrosine
- Author
-
Kendall N. Houk and Hakan Gunaydin
- Subjects
Nitrates ,Hydroxyl Radical ,Radical ,Nitrogen Dioxide ,Carbonates ,Protonation ,General Medicine ,Carbon Dioxide ,Toxicology ,Photochemistry ,Peroxide ,Decomposition ,Article ,chemistry.chemical_compound ,Peroxynitrous acid ,chemistry ,Nitration ,Peroxynitrous Acid ,Tyrosine ,Hydroxyl radical ,Peroxynitrite - Abstract
The mechanisms of tyrosine nitration by peroxynitrous acid or nitrosoperoxycarbonate were investigated with the CBS-QB3 method. Either the protonation of peroxynitrite, or a reaction with carbon dioxide gives a reactive peroxide intermediate. Peroxynitrous acid mediated nitration of phenol occurs via the unimolecular decomposition to give nitrogen dioxide and hydroxyl radicals. Nitrosoperoxycarbonate also undergoes unimolecular decomposition to give carbonate and nitrogen dioxide radicals. The reactions of tyrosine with the hydroxyl or carbonate radicals give a phenoxy radical intermediate. The reaction of the nitrogen dioxide with this radical intermediate followed by tautomerization gives nitrated tyrosine in both cases. According to CBS-QB3 calculations, the rate-limiting step for the nitration of phenol is the decomposition of peroxynitrous acid or of nitrosoperoxycarbonate.
- Published
- 2009
47. Molecular dynamics prediction of the mechanism of ester hydrolysis in water
- Author
-
Kendall N. Houk and Hakan Gunaydin
- Subjects
Models, Molecular ,Hydronium ,Molecular Structure ,Methyl formate ,Hydrolysis ,Inorganic chemistry ,Water ,Esters ,General Chemistry ,Biochemistry ,Catalysis ,Reversible reaction ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,chemistry ,Computational chemistry ,Amide ,Hydroxide ,Molecule ,Computer Simulation - Abstract
Hydrolysis reactions of the basic units of biological polymers with water, or the reverse reaction, the formation of ester, amide, ketal, or phosphate bonds, occur with very high activation barriers in the gas phase but occur much more rapidly in pure water. Car−Parrinello molecular dynamics simulations reported here show that the rate of hydrolysis of methyl formate in pure water is consistent with mechanisms involving cooperative catalysis by autoionization-generated hydroxide and hydronium, a process known to have an activation free energy of 23.8 kcal/mol. In this mechanism, autoionization is followed by rapid simultaneous acid−base catalysis.
- Published
- 2008
48. First-Principles Theory of Hydrogen Diffusion in Aluminum
- Author
-
Hakan Gunaydin, Kendall N. Houk, Vidvuds Ozoliņš, and Sergey V. Barabash
- Subjects
Materials science ,chemistry ,Hydrogen ,Aluminium ,Diffusion ,Enthalpy ,Lattice diffusion coefficient ,General Physics and Astronomy ,chemistry.chemical_element ,Effective diffusion coefficient ,Thermodynamics ,Density functional theory ,Thermal diffusivity - Abstract
Ab initio molecular dynamics simulations are used to obtain the activation enthalpy and preexponential factor for the lattice diffusion of hydrogen in aluminum between the temperatures 650 and 850 K: DeltaH double dagger=0.12+/-0.02 eV and D0=1.8 x 10(-7)m2/s. Vacancies are found to significantly decrease the apparent diffusivity due to their ability to bind one, two, or even six hydrogen atoms, causing a strong composition dependence and non-Arrhenius behavior of the effective diffusion coefficient.
- Published
- 2008
49. Vacancy-mediated dehydrogenation of sodium alanate
- Author
-
Kendall N. Houk, Hakan Gunaydin, and Vidvuds Ozoliņš
- Subjects
Multidisciplinary ,Hydrogen ,Hydride ,Nucleation ,chemistry.chemical_element ,Nanotechnology ,Activation energy ,Dissociation (chemistry) ,Hydrogen storage ,chemistry ,Vacancy defect ,Physical Sciences ,Physical chemistry ,Dehydrogenation - Abstract
Clarification of the mechanisms of hydrogen release and uptake in transition-metal-doped sodium alanate, NaAlH 4 , a prototypical high-density complex hydride, has fundamental importance for the development of improved hydrogen-storage materials. In this and most other modern hydrogen-storage materials, H 2 release and uptake are accompanied by long-range diffusion of metal species. Using first-principles density-functional theory calculations, we have determined that the activation energy for Al mass transport via AlH 3 vacancies is Q = 85 kJ/mol·H 2 , which is in excellent agreement with experimentally measured activation energies in Ti-catalyzed NaAlH 4 . The activation energy for an alternate decomposition mechanism via NaH vacancies is found to be significantly higher: Q = 112 kJ/mol·H 2 . Our results suggest that bulk diffusion of Al species is the rate-limiting step in the dehydrogenation of Ti-doped samples of NaAlH 4 and that the much higher activation energies measured for uncatalyzed samples are controlled by other processes, such as breaking up of AlH 4 − complexes, formation/dissociation of H 2 molecules, and/or nucleation of the product phases.
- Published
- 2008
50. Cyclopropanecarboxylic acid esters as potential prodrugs with enhanced hydrolytic stability
- Author
-
David M. Bender, Yu Takano, Jeffrey A Peterson, Hakan Gunaydin, K. N. Houk, and James R McCarthy
- Subjects
Alkane ,chemistry.chemical_classification ,Cyclopropanes ,Isodesmic reaction ,Molecular Structure ,Hydrolysis ,Organic Chemistry ,Acyclovir ,Esters ,Valine ,Prodrug ,Biochemistry ,Antiviral Agents ,Cyclopropane ,chemistry.chemical_compound ,chemistry ,Valacyclovir ,Organic chemistry ,Cyclopropanecarboxylic acid ,Molecule ,Prodrugs ,Physical and Theoretical Chemistry - Abstract
Esters of cyclopropanecarboxylic acid demonstrate a substantial increase in stability under both acid- and base-catalyzed hydrolytic conditions. Comparison of the stability of valacyclovir 13 with the cyclopropane analogue 14 shows that at 40 degrees C and pH 6 the half-life of 14 is >300 h while the value for 13 is 69.7 h. CBS-QB3 calculations on isodesmic reactions for transfer of groups from an alkane to an ester show that a cyclopropyl group provides hyperconjugative stabilization.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.