Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Wei-Sheng Huang is active.

Publication


Featured researches published by Wei-Sheng Huang.


Journal of Medicinal Chemistry | 2010

Discovery of 3-[2-(imidazo[1,2-b]pyridazin-3-yl)ethynyl]-4-methyl-N-{4-[(4-methylpiperazin-1-yl)methyl]-3-(trifluoromethyl)phenyl}benzamide (AP24534), a potent, orally active pan-inhibitor of breakpoint cluster region-abelson (BCR-ABL) kinase including the T315I gatekeeper mutant.

Wei-Sheng Huang; Chester A. Metcalf; Raji Sundaramoorthi; Yihan Wang; Dong Zou; R. Mathew Thomas; Xiaotian Zhu; Lisi Cai; David Wen; Shuangying Liu; Jan Antoinette C. Romero; Jiwei Qi; Ingrid Chen; Geetha Banda; Scott P. Lentini; Sasmita Das; Qihong Xu; Jeff Keats; Frank Wang; Scott Wardwell; Yaoyu Ning; Joseph Snodgrass; Marc I. Broudy; Karin Russian; Tianjun Zhou; Lois Commodore; Narayana I. Narasimhan; Qurish K. Mohemmad; John Iuliucci; Victor M. Rivera

In the treatment of chronic myeloid leukemia (CML) with BCR-ABL kinase inhibitors, the T315I gatekeeper mutant has emerged as resistant to all currently approved agents. This report describes the structure-guided design of a novel series of potent pan-inhibitors of BCR-ABL, including the T315I mutation. A key structural feature is the carbon-carbon triple bond linker which skirts the increased bulk of Ile315 side chain. Extensive SAR studies led to the discovery of development candidate 20g (AP24534), which inhibited the kinase activity of both native BCR-ABL and the T315I mutant with low nM IC(50)s, and potently inhibited proliferation of corresponding Ba/F3-derived cell lines. Daily oral administration of 20g significantly prolonged survival of mice injected intravenously with BCR-ABL(T315I) expressing Ba/F3 cells. These data, coupled with a favorable ADME profile, support the potential of 20g to be an effective treatment for CML, including patients refractory to all currently approved therapies.


Chemical Biology & Drug Design | 2011

Structural Mechanism of the Pan-BCR-ABL Inhibitor Ponatinib (AP24534): Lessons for Overcoming Kinase Inhibitor Resistance.

Tianjun Zhou; Lois Commodore; Wei-Sheng Huang; Yihan Wang; Mathew Thomas; Jeff Keats; Qihong Xu; Victor M. Rivera; William C. Shakespeare; Tim Clackson; David C. Dalgarno; Xiaotian Zhu

The BCR‐ABL inhibitor imatinib has revolutionized the treatment of chronic myeloid leukemia. However, drug resistance caused by kinase domain mutations has necessitated the development of new mutation‐resistant inhibitors, most recently against the T315I gatekeeper residue mutation. Ponatinib (AP24534) inhibits both native and mutant BCR‐ABL, including T315I, acting as a pan‐BCR‐ABL inhibitor. Here, we undertook a combined crystallographic and structure–activity relationship analysis on ponatinib to understand this unique profile. While the ethynyl linker is a key inhibitor functionality that interacts with the gatekeeper, virtually all other components of ponatinib play an essential role in its T315I inhibitory activity. The extensive network of optimized molecular contacts found in the DFG‐out binding mode leads to high potency and renders binding less susceptible to disruption by single point mutations. The inhibitory mechanism exemplified by ponatinib may have broad relevance to designing inhibitors against other kinases with mutated gatekeeper residues.


Journal of Medicinal Chemistry | 2016

Discovery of Brigatinib (AP26113), a Phosphine Oxide-Containing, Potent, Orally Active Inhibitor of Anaplastic Lymphoma Kinase

Wei-Sheng Huang; Shuangying Liu; Dong Zou; Mathew Thomas; Yihan Wang; Tianjun Zhou; Jan Antoinette C. Romero; Anna Kohlmann; Feng Li; Jiwei Qi; Lisi Cai; Timothy A. Dwight; Yongjin Xu; Rongsong Xu; Rory Dodd; Angela Toms; Lois Parillon; Xiaohui Lu; Rana Anjum; Sen Zhang; Frank Wang; Jeffrey Keats; Scott Wardwell; Yaoyu Ning; Qihong Xu; Lauren Moran; Qurish K. Mohemmad; Hyun Gyung Jang; Tim Clackson; Narayana I. Narasimhan

In the treatment of echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase positive (ALK+) non-small-cell lung cancer (NSCLC), secondary mutations within the ALK kinase domain have emerged as a major resistance mechanism to both first- and second-generation ALK inhibitors. This report describes the design and synthesis of a series of 2,4-diarylaminopyrimidine-based potent and selective ALK inhibitors culminating in identification of the investigational clinical candidate brigatinib. A unique structural feature of brigatinib is a phosphine oxide, an overlooked but novel hydrogen-bond acceptor that drives potency and selectivity in addition to favorable ADME properties. Brigatinib displayed low nanomolar IC50s against native ALK and all tested clinically relevant ALK mutants in both enzyme-based biochemical and cell-based viability assays and demonstrated efficacy in multiple ALK+ xenografts in mice, including Karpas-299 (anaplastic large-cell lymphomas [ALCL]) and H3122 (NSCLC). Brigatinib represents the most clinically advanced phosphine oxide-containing drug candidate to date and is currently being evaluated in a global phase 2 registration trial.


Clinical Cancer Research | 2016

The Potent ALK Inhibitor Brigatinib (AP26113) Overcomes Mechanisms of Resistance to First- and Second-Generation ALK Inhibitors in Preclinical Models

Sen Zhang; Rana Anjum; Rachel M. Squillace; Sara Nadworny; Tianjun Zhou; Jeff Keats; Yaoyu Ning; Scott Wardwell; David Miller; Youngchul Song; Lindsey Eichinger; Lauren Moran; Wei-Sheng Huang; Shuangying Liu; Dong Zou; Yihan Wang; Qurish K. Mohemmad; Emily Y. Ye; Narayana I. Narasimhan; Frank Wang; Juan J. Miret; Xiaotian Zhu; Tim Clackson; David C. Dalgarno; William C. Shakespeare; Victor M. Rivera

Purpose: Non–small cell lung cancers (NSCLCs) harboring ALK gene rearrangements (ALK+) typically become resistant to the first-generation anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor (TKI) crizotinib through development of secondary resistance mutations in ALK or disease progression in the brain. Mutations that confer resistance to second-generation ALK TKIs ceritinib and alectinib have also been identified. Here, we report the structure and first comprehensive preclinical evaluation of the next-generation ALK TKI brigatinib. Experimental Design: A kinase screen was performed to evaluate the selectivity profile of brigatinib. The cellular and in vivo activities of ALK TKIs were compared using engineered and cancer-derived cell lines. The brigatinib–ALK co-structure was determined. Results: Brigatinib potently inhibits ALK and ROS1, with a high degree of selectivity over more than 250 kinases. Across a panel of ALK+ cell lines, brigatinib inhibited native ALK (IC50, 10 nmol/L) with 12-fold greater potency than crizotinib. Superior efficacy of brigatinib was also observed in mice with ALK+ tumors implanted subcutaneously or intracranially. Brigatinib maintained substantial activity against all 17 secondary ALK mutants tested in cellular assays and exhibited a superior inhibitory profile compared with crizotinib, ceritinib, and alectinib at clinically achievable concentrations. Brigatinib was the only TKI to maintain substantial activity against the most recalcitrant ALK resistance mutation, G1202R. The unique, potent, and pan-ALK mutant activity of brigatinib could be rationalized by structural analyses. Conclusions: Brigatinib is a highly potent and selective ALK inhibitor. These findings provide the molecular basis for the promising activity being observed in ALK+, crizotinib-resistant patients with NSCLC being treated with brigatinib in clinical trials. Clin Cancer Res; 22(22); 5527–38. ©2016 AACR.


Journal of Medicinal Chemistry | 2013

Fragment growing and linking lead to novel nanomolar lactate dehydrogenase inhibitors.

Anna Kohlmann; Stephan G. Zech; Feng Li; Tianjun Zhou; Rachel M. Squillace; Lois Commodore; Matthew T. Greenfield; Xiaohui Lu; David P. Miller; Wei-Sheng Huang; Jiwei Qi; R. Mathew Thomas; Yihan Wang; Sen Zhang; Rory Dodd; Shuangying Liu; Rongsong Xu; Yongjin Xu; Juan J. Miret; Victor M. Rivera; Tim Clackson; William C. Shakespeare; Xiaotian Zhu; David C. Dalgarno

Lactate dehydrogenase A (LDH-A) catalyzes the interconversion of lactate and pyruvate in the glycolysis pathway. Cancer cells rely heavily on glycolysis instead of oxidative phosphorylation to generate ATP, a phenomenon known as the Warburg effect. The inhibition of LDH-A by small molecules is therefore of interest for potential cancer treatments. We describe the identification and optimization of LDH-A inhibitors by fragment-based drug discovery. We applied ligand based NMR screening to identify low affinity fragments binding to LDH-A. The dissociation constants (K(d)) and enzyme inhibition (IC(50)) of fragment hits were measured by surface plasmon resonance (SPR) and enzyme assays, respectively. The binding modes of selected fragments were investigated by X-ray crystallography. Fragment growing and linking, followed by chemical optimization, resulted in nanomolar LDH-A inhibitors that demonstrated stoichiometric binding to LDH-A. Selected molecules inhibited lactate production in cells, suggesting target-specific inhibition in cancer cell lines.


Journal of Medicinal Chemistry | 2009

9-(Arenethenyl)purines as Dual Src/Abl Kinase Inhibitors Targeting the Inactive Conformation: Design, Synthesis, and Biological Evaluation

Wei-Sheng Huang; Xiaotian Zhu; Yihan Wang; Mohammad Azam; David Wen; Raji Sundaramoorthi; R. Mathew Thomas; Shuangying Liu; Geetha Banda; Scott P. Lentini; Sasmita Das; Qihong Xu; Jeff Keats; Frank Wang; Scott Wardwell; Yaoyu Ning; Joseph Snodgrass; Marc I. Broudy; Karin Russian; George Q. Daley; John Iuliucci; David C. Dalgarno; Tim Clackson; Tomi K. Sawyer; William C. Shakespeare

A novel series of potent dual Src/Abl kinase inhibitors based on a 9-(arenethenyl)purine core has been identified. Unlike traditional dual Src/Abl inhibitors targeting the active enzyme conformation, these inhibitors bind to the inactive, DFG-out conformation of both kinases. Extensive SAR studies led to the discovery of potent and orally bioavailable inhibitors, some of which demonstrated in vivo efficacy. Once-daily oral administration of inhibitor 9i (AP24226) significantly prolonged the survival of mice injected intravenously with wild type Bcr-Abl expressing Ba/F3 cells at a dose of 10 mg/kg. In a separate model, oral administration of 9i to mice bearing subcutaneous xenografts of Src Y527F expressing NIH 3T3 cells elicited dose-dependent tumor shrinkage with complete tumor regression observed at the highest dose. Notably, several inhibitors (e.g., 14a, AP24163) exhibited modest cellular potency (IC50 = 300-400 nM) against the Bcr-Abl mutant T315I, a variant resistant to all currently marketed therapies for chronic myeloid leukemia.


Cancer Research | 2010

Abstract 3623: Efficacy and pharmacodynamic analysis of AP26113, a potent and selective orally active inhibitor of Anaplastic Lymphoma Kinase (ALK)

Victor M. Rivera; Rana Anjum; Frank Wang; Sen Zhang; Jeffrey Keats; Yaoyu Ning; Scott Wardwell; Lauren Moran; Emily Y. Ye; Dung Yu Chun; Qurish K. Mohemmad; Shuangying Liu; Wei-Sheng Huang; Yihan Wang; Mathew Thomas; Feng Li; Juan J. Miret; John Iuliucci; David C. Dalgarno; Narayana I. Narasimhan; Tim Clackson; William C. Shakespeare

Activating gene rearrangements of anaplastic lymphoma kinase (ALK) have been identified in anaplastic large cell lymphoma (ALCL; NPM-ALK) and non-small cell lung cancer (NSCLC; EML4-ALK). The dual Met/ALK inhibitor PF-02341066 (PF1066) has demonstrated promising clinical activity against tumors carrying activating ALK gene rearrangements (Kwak ASCO 2009: #3509) validating ALK as a therapeutic target. Previously, AP26113 was identified as a novel, potent, orally bioavailable ALK inhibitor with demonstrated selectivity over related receptor tyrosine kinase family members IGF-1R and InsR and no inhibition of Met. Here the efficacy and exposure/activity relationship of AP26113 was further characterized in preclinical models and compared to that of PF1066. In a panel of 7 EML4-ALK or NPM-ALK positive NSCLC and ALCL cell lines, the concentration of AP26113 that inhibited growth by 50% (GI50) ranged from 4.2 - 30.8 nM. In each cell line the GI50 for PF1066 was ∼10-fold greater (range 62 - 309 nM). In 4 cells lines tested, the IC50 for inhibition of ALK phosphorylation tracked with potency in cell proliferation assays and was 10-fold greater for PF1066 than AP26113. Across 3 ALK-negative NSCLC and ALCL cell lines the GI50s for AP26113 (503 - 2387 nM) and PF1066 (928 - 1773 nM) were similar. Overall, AP26113 exhibited ∼100-fold selectivity for ALK-positive lines compared with a ∼10-fold selectivity for PF1066. The in vivo activities of daily oral dosing of AP26113 (10, 25 and 50 mg/kg) and PF1066 (25, 50 and 100 mg/kg) were examined in Karpas-299 ALCL (2 week dosing) and H3122 NSCLC (3 week dosing) xenograft models. At the highest doses tested, strong regressions were achieved with AP26113, but not PF1066. Tumor growth inhibition by 25 mg/kg and 10 mg/kg doses of AP26113 in the ALCL and NSCLC models, respectively, was similar to that of 100 mg/kg PF1066. In a PK/PD study in the ALCL model, inhibition of ALK phosphorylation after administration of 100 mg/kg PF1066 was intermediate between that observed after administration of 10 or 25 mg/kg AP26113. Results from the analysis of plasma levels of each drug showed that AP26113 had equivalent efficacy to PF1066 at 4- to 10-fold lower levels of exposure (AUC and 24 h trough plasma levels). AP26113 demonstrated favorable properties including moderate in vitro plasma protein binding (≤77% in mouse, rat, monkey, and human plasma), negligible inhibition of major CYP isoforms (IC50 > 10 μM for 3A4, 2C9, 2D6), and good oral bioavailability (multiple animal species). In animal models, AP26113 was well-tolerated at and above predicted clinically effective plasma levels. In conclusion, these data demonstrate that AP26113 is a highly potent and selective inhibitor of ALK and support the clinical evaluation of AP26113 in patients with ALK-driven tumors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3623.


Bioorganic & Medicinal Chemistry Letters | 2008

Novel N9-arenethenyl purines as potent dual Src/Abl tyrosine kinase inhibitors.

Yihan Wang; William C. Shakespeare; Wei-Sheng Huang; Raji Sundaramoorthi; Scott P. Lentini; Sasmita Das; Shuangying Liu; Geeta Banda; David Wen; Xiaotian Zhu; Qihong Xu; Jeffrey Keats; Frank Wang; Scott Wardwell; Yaoyu Ning; Joseph Snodgrass; Mark I. Broudy; Karin Russian; David C. Dalgarno; Tim Clackson; Tomi K. Sawyer

Novel N(9)-arenethenyl purines, optimized potent dual Src/Abl tyrosine kinase inhibitors, are described. The key structural feature is a trans vinyl linkage at N(9) on the purine core which projects hydrophobic substituents into the selectivity pocket at the rear of the ATP site. Their synthesis was achieved through a Horner-Wadsworth-Emmons reaction of N(9)-phosphorylmethylpurines and substituted benzaldehydes or Heck reactions between 9-vinyl purines and aryl halides. Most compounds are potent inhibitors of both Src and Abl kinase, and several possess good oral bioavailability.


Chemical Biology & Drug Design | 2010

AP24163 Inhibits the Gatekeeper Mutant of BCR-ABL and Suppresses In vitro Resistance

Mohammad Azam; John T. Powers; William S. Einhorn; Wei-Sheng Huang; William C. Shakespeare; Xiaotian Zhu; David C. Dalgarno; Tim Clackson; Tomi K. Sawyer; George Q. Daley

Mutation in the ABL kinase domain is the principal mechanism of imatinib resistance in patients with chronic myelogenous leukaemia. The second generation BCR/ABL inhibitors nilotinib and dasatinib effectively inhibit most imatinib resistance variants, but are ineffective against the gatekeeper mutant, T315I. Gatekeeper mutation activates the kinase by stabilizing the hydrophobic spine. Here, we describe that the rationally designed compound AP24163 can inhibit native and gatekeeper mutants of the BCR/ABL kinase. Structural modelling suggests that AP24163 affects the flexibility of the P‐loop and destabilizes the active conformation by disrupting the hydrophobic spine. In vitro screening for drug resistance identified clones with compound mutations involving both the P‐loop and T315I. Our studies provide structural insights for the design of inhibitors against the gatekeeper mutant and suggest that up‐front combination therapy may be required to prevent the emergence of compound‐resistant mutations.


Chemical Biology & Drug Design | 2010

Structural Analysis of DFG‐in and DFG‐out Dual Src‐Abl Inhibitors Sharing a Common Vinyl Purine Template

Tianjun Zhou; Lois Commodore; Wei-Sheng Huang; Yihan Wang; Tomi K. Sawyer; William C. Shakespeare; Tim Clackson; Xiaotian Zhu; David C. Dalgarno

Bcr‐Abl is the oncogenic protein tyrosine kinase responsible for chronic myeloid leukemia (CML). Treatment of the disease with imatinib (Gleevec) often results in drug resistance via kinase mutations at the advanced phases of the disease, which has necessitated the development of new mutation‐resistant inhibitors, notably against the T315I gatekeeper mutation. As part of our efforts to discover such mutation resistant Abl inhibitors, we have focused on optimizing purine template kinase inhibitors, leading to the discovery of potent DFG‐in and DFG‐out series of Abl inhibitors that are also potent Src inhibitors. Here we present crystal structures of Abl bound by two such inhibitors, based on a common N9‐arenyl purine, and that represent both DFG‐in and ‐out binding modes. In each structure the purine template is bound deeply in the adenine pocket and the novel vinyl linker forms a non‐classical hydrogen bond to the gatekeeper residue, Thr315. Specific template substitutions promote either a DFG‐in or ‐out binding mode, with the kinase binding site adjusting to optimize molecular recognition. Bcr‐Abl T315I mutant kinase is resistant to all currently marketed Abl inhibitors, and is the focus of intense drug discovery efforts. Notably, our DFG‐out inhibitor, AP24163, exhibits modest activity against this mutant, illustrating that this kinase mutant can be inhibited by DFG‐out class inhibitors. Furthermore our DFG‐out inhibitor exhibits dual Src‐Abl activity, absent from the prototypical DFG‐out inhibitor, imatinib as well as its analog, nilotinib. The data presented here provides structural guidance for the further design of novel potent DFG‐out class inhibitors against Src, Abl and Abl T315I mutant kinases.

Collaboration


Dive into the Wei-Sheng Huang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiwei Qi

ARIAD Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge