Haishan Wang
Singapore Science Park
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Haishan Wang.
Journal of Biomolecular Screening | 2006
Vasantha M. Nayagam; Xukun Wang; Yong Cheng Tan; Anders Poulsen; Kee Chuan Goh; Tony Ng; Haishan Wang; Hong Yan Song; BinHui Ni; Michael Entzeroth; Walter Stünkel
The nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase SIRT1 has been linked to fatty acid metabolism via suppression of peroxysome proliferator-activated receptor gamma (PPAR-γ) and to inflammatory processes by deacetylating the transcription factor NF-κB. First, modulation of SIRT1 activity affects lipid accumulation in adipocytes, which has an impact on the etiology of a variety of human metabolic diseases such as obesity and insulin-resistant diabetes. Second, activation of SIRT1 suppresses inflammation via regulation of cytokine expression. Using high-throughput screening, the authors identified compounds with SIRT1 activating and inhibiting potential. The biological activity of these SIRT1-modulating compounds was confirmed in cell-based assays using mouse adipocytes, as well as human THP-1 monocytes. SIRT1 activators were found to be potent lipolytic agents, reducing the overall lipid content of fully differentiated NIH L1 adipocytes. In addition, the same compounds have anti-inflammatory properties, as became evident by the reduction of the proinflammatory cytokine tumor necrosis factor-alpha (TNF-α). In contrast, a SIRT1 inhibitory compound showed a stimulatory activity on the differentiation of adipocytes, a feature often linked to insulin sensitization.
Molecular Cancer Therapeutics | 2010
Veronica Novotny-Diermayr; Kanda Sangthongpitag; Chang Yong Hu; Xiaofeng Wu; Nina Sausgruber; Pauline Yeo; Gediminas Greicius; Sven Pettersson; Ai Leng Liang; Yung Kiang Loh; Zahid Bonday; Kee Chuan Goh; Hannes Hentze; Stefan Hart; Haishan Wang; Kantharaj Ethirajulu; Jeanette Marjorie Wood
Although clinical responses in liquid tumors and certain lymphomas have been reported, the clinical efficacy of histone deacetylase inhibitors in solid tumors has been limited. This may be in part due to the poor pharmacokinetic of these drugs, resulting in inadequate tumor concentrations of the drug. SB939 is a new hydroxamic acid based histone deacetylase inhibitor with improved physicochemical, pharmaceutical, and pharmacokinetic properties. In vitro, SB939 inhibits class I, II, and IV HDACs, with no effects on other zinc binding enzymes, and shows significant antiproliferative activity against a wide variety of tumor cell lines. It has very favorable pharmacokinetic properties after oral dosing in mice, with >4-fold increased bioavailability and 3.3-fold increased half-life over suberoylanilide hydroxamic acid (SAHA). In contrast to SAHA, SB939 accumulates in tumor tissue and induces a sustained inhibition of histone acetylation in tumor tissue. These excellent pharmacokinetic properties translated into a dose-dependent antitumor efficacy in a xenograft model of human colorectal cancer (HCT-116), with a tumor growth inhibition of 94% versus 48% for SAHA (both at maximum tolerated dose), and was also effective when given in different intermittent schedules. Furthermore, in APCmin mice, a genetic mouse model of early-stage colon cancer, SB939 inhibited adenoma formation, hemocult scores, and increased hematocrit values more effectively than 5-fluorouracil. Emerging clinical data from phase I trials in cancer patients indicate that the pharmacokinetic and pharmacologic advantages of SB939 are translated to the clinic. The efficacy of SB939 reported here in two very different models of colorectal cancer warrants further investigation in patients. Mol Cancer Ther; 9(3); 642–52
Expert Opinion on Therapeutic Patents | 2009
Haishan Wang; Brian W. Dymock
Importance of the field: Following FDA approval of vorinostat in 2006, several novel HDAC inhibitors (HDACis) have entered clinical trials, and there are numerous published patent applications claiming novel HDACis which were optimized as potential drug candidates, designed for regional or systemic release, and created as dual or multifunctional inhibitors. Given the breadth and depth of recent reporting of novel HDACis, there has emerged a need to review the field from a chemists perspective in one compact article. Areas covered in this review: This review provides a summary of published patent applications claiming novel HDACis from 2007 until mid-2009, covering mainly classes I, II and IV anticancer HDACis including those that have recently advanced to the clinic. What the reader will gain: Readers will rapidly gain an overview of the majority of HDACi scaffolds with representative structure–activity relationships; they will learn how these new compounds were created, how their drug like properties were improved and which companies are the main players in the field. Take home message: Although competition in this field is intense, the future application of HDACis to treat human disease either as single agents or in combination with existing drugs holds real promise.
Journal of Medicinal Chemistry | 2011
Haishan Wang; Niefang Yu; Dizhong Chen; Ken Chi Lik Lee; Pek Ling Lye; Joyce Wei Wei Chang; Weiping Deng; Melvin Ng; Ting Lu; Mui Ling Khoo; Anders Poulsen; Kanda Sangthongpitag; Xiaofeng Wu; Changyong Hu; Kee Chuan Goh; Xukun Wang; Lijuan Fang; Kay Lin Goh; Hwee Hoon Khng; Siok Kun Goh; Pauline Yeo; Xin Liu; Zahid Bonday; Jeanette Marjorie Wood; Brian W. Dymock; Ethirajulu Kantharaj; Eric T. Sun
A series of 3-(1,2-disubstituted-1H-benzimidazol-5-yl)-N-hydroxyacrylamides (1) were designed and synthesized as HDAC inhibitors. Extensive SARs have been established for in vitro potency (HDAC1 enzyme and COLO 205 cellular IC(50)), liver microsomal stability (t(1/2)), cytochrome P450 inhibitory (3A4 IC(50)), and clogP, among others. These parameters were fine-tuned by carefully adjusting the substituents at positions 1 and 2 of the benzimidazole ring. After comprehensive in vitro and in vivo profiling of the selected compounds, SB939 (3) was identified as a preclinical development candidate. 3 is a potent pan-HDAC inhibitor with excellent druglike properties, is highly efficacious in in vivo tumor models (HCT-116, PC-3, A2780, MV4-11, Ramos), and has high and dose-proportional oral exposures and very good ADME, safety, and pharmaceutical properties. When orally dosed to tumor-bearing mice, 3 is enriched in tumor tissue which may contribute to its potent antitumor activity and prolonged duration of action. 3 is currently being tested in phase I and phase II clinical trials.
Journal of Medicinal Chemistry | 2012
Anthony D. William; Angeline C.-H. Lee; Kee Chuan Goh; Stéphanie Blanchard; Anders Poulsen; Ee Ling Teo; Harish Nagaraj; Chai Ping Lee; Haishan Wang; Meredith Williams; Eric T. Sun; Changyong Hu; Ramesh Jayaraman; Mohammed Khalid Pasha; Kantharaj Ethirajulu; Jeanette Marjorie Wood; Brian W. Dymock
Herein, we describe the design, synthesis, and SAR of a series of unique small molecule macrocycles that show spectrum selective kinase inhibition of CDKs, JAK2, and FLT3. The most promising leads were assessed in vitro for their inhibition of cancer cell proliferation, solubility, CYP450 inhibition, and microsomal stability. This screening cascade revealed 26 h as a preferred compound with target IC(50) of 13, 73, and 56 nM for CDK2, JAK2 and FLT3, respectively. Pharmacokinetic (PK) studies of 26 h in preclinical species showed good oral exposures. Oral efficacy was observed in colon (HCT-116) and lymphoma (Ramos) xenograft studies, in line with the observed PK/PD correlation. 26h (SB1317/TG02) was progressed into development in 2010 and is currently undergoing phase 1 clinical trials in advanced leukemias and multiple myeloma.
Journal of Computer-aided Molecular Design | 2012
Anders Poulsen; Anthony D. William; Stéphanie Blanchard; Angeline Lee; Harish Nagaraj; Haishan Wang; Eeling Teo; Evelyn Tan; Kee Chuan Goh; Brian W. Dymock
Macrocycles from our Aurora project were screened in a kinase panel and were found to be active on other kinase targets, mainly JAKs, FLT3 and CDKs. Subsequently these compounds became leads in our JAK2 project. Macrocycles with a basic nitrogen in the linker form a salt bridge with Asp86 in CDK2 and Asp698 in FLT3. This residue is conserved in most CDKs resulting in potent pan CDK inhibition. One of the main project objectives was to achieve JAK2 potency with 100-fold selectivity against CDKs. Macrocycles with an ether linker have potent JAK2 activity with the ether oxygen forming a hydrogen bond to Ser936. A hydrogen bond to the equivalent residues of JAK3 and most CDKs cannot be formed resulting in good selectivity for JAK2 over JAK3 and CDKs. Further optimization of the macrocyclic linker and side chain increased JAK2 and FLT3 activity as well as improving DMPK properties. The selective JAK2/FLT3 inhibitor 11 (Pacritinib, SB1518) has successfully finished phase 2 clinical trials for myelofibrosis and lymphoma. Another selective JAK2/FLT3 inhibitor, 33 (SB1578), has entered phase 1 clinical development for the non-oncology indication rheumatoid arthritis.
Journal of Medicinal Chemistry | 2012
Anthony D. William; Angeline C.-H. Lee; Anders Poulsen; Kee Chuan Goh; Babita Madan; Stefan Hart; Evelyn Tan; Haishan Wang; Harish Nagaraj; Dizhong Chen; Chai Ping Lee; Eric T. Sun; Ramesh Jayaraman; Mohammad Khalid Pasha; Kantharaj Ethirajulu; Jeanette Marjorie Wood; Brian W. Dymock
Herein, we describe the synthesis and SAR of a series of small molecule macrocycles that selectively inhibit JAK2 kinase within the JAK family and FLT3 kinase. Following a multiparameter optimization of a key aryl ring of the previously described SB1518 (pacritinib), the highly soluble 14l was selected as the optimal compound. Oral efficacy in the murine collagen-induced arthritis (CIA) model for rheumatoid arthritis (RA) supported 14l as a potential treatment for autoimmune diseases and inflammatory disorders such as psoriasis and RA. Compound 14l (SB1578) was progressed into development and is currently undergoing phase 1 clinical trials in healthy volunteers.
Journal of Molecular Modeling | 2013
Anders Poulsen; Anthony D. William; Stéphanie Blanchard; Harish Nagaraj; Meredith Williams; Haishan Wang; Angeline Lee; Eric T. Sun; Eeling Teo; Evelyn Tan; Kee Chuan Goh; Brian W. Dymock
AbstractA high-throughput screen against Aurora A kinase revealed several promising submicromolar pyrimidine-aniline leads. The bioactive conformation found by docking these leads into the Aurora A ATP-binding site had a semicircular shape. Macrocycle formation was proposed to achieve novelty and selectivity via ring-closing metathesis of a diene precursor. The nature of the optimal linker and its size was directed by docking. In a kinase panel screen, selected macrocycles were active on other kinase targets, mainly FLT3, JAK2, and CDKs. These compounds then became leads in a CDK/FLT3/JAK2 inhibitor project. Macrocycles with a basic nitrogen in the linker form a salt bridge with Asp86 in CDK2 and Asp698 in FLT3. Interaction with this residue explains the observed selectivity. The Asp86 residue is conserved in most CDKs, resulting in potent pan-CDK inhibition by these compounds. Optimized macrocycles generally have good DMPK properties, and are efficacious in mouse models of cancer. Compound 5 (SB1317/TG02), a pan-CDK/FLT3/JAK2 inhibitor, was selected for preclinical development, and is now in phase 1 clinical trials. FigureStructure of SB1317 (left). SB1317 docked into CDK2 (right)
Bioorganic & Medicinal Chemistry Letters | 2009
Haishan Wang; Niefang Yu; Hong Yan Song; Dizhong Chen; Yong Zou; Weiping Deng; Pek Ling Lye; Joyce Wei Wei Chang; Melvin Ng; Stéphanie Blanchard; Eric T. Sun; Kanda Sangthongpitag; Xukun Wang; Kee Chuan Goh; Xiaofeng Wu; Hwee Hoon Khng; Lijuan Fang; Siok Kun Goh; Wai Chung Ong; Zahid Bonday; Walter Stünkel; Anders Poulsen; Michael Entzeroth
A series of N-hydroxy-1,2-disubstituted-1H-benzimidazol-5-yl acrylamides were designed and synthesized as novel HDAC inhibitors. General SAR has been established for the substituents at positions 1 and 2, as well as the importance of the ethylene group and its attachment to position 5. Optimized compounds are much more potent than SAHA in both enzymatic and cellular assays. A representative compound, 23 (SB639), has demonstrated antitumor activity in a colon cancer xenograft model.
Drug Metabolism and Disposition | 2011
Ramesh Jayaraman; Venkatesh Pilla Reddy; Mohammed Khalid Pasha; Haishan Wang; Kanda Sangthongpitag; Pauline Yeo; Chang Yong Hu; Xiaofeng Wu; Liu Xin; Evelyn Goh; Lee Sun New; Kantharaj Ethirajulu
The preclinical absorption, distribution, metabolism, and excretion (ADME) properties of Pracinostat [(2E)-3-[2-butyl-1-[2-(diethylamino) ethyl]-1H-benzimidazol-5-yl]-N-hydroxyarylamide hydrochloride; SB939], an orally active histone deacetylase inhibitor, were characterized and its human pharmacokinetics (PK) was predicted using Simcyp and allometric scaling. SB939 showed high aqueous solubility with high Caco-2 permeability. Metabolic stability was relatively higher in dog and human liver microsomes than in mouse and rat. The major metabolites formed in human liver microsomes were also observed in preclinical species. Human cytochrome P450 (P450) phenotyping showed that SB939 was primarily metabolized by CYP3A4 and CYP1A2. SB939 did not significantly inhibit human CYP3A4, 1A2, 2D6, and 2C9 (>25 μM) but inhibited 2C19 (IC50 = 5.8 μM). No significant induction of human CYP3A4 and 1A2 was observed in hepatocytes. Plasma protein binding in mouse, rat, dog, and human ranged between ∼84 and 94%. The blood-to-plasma ratio was ∼1.0 in human blood. SB939 showed high systemic clearance (relative to liver blood flow) of 9.2, 4.5, and 1.5 l · h−1 · kg−1 and high volume of distribution at steady state (>0.6 l/kg) of 3.5, 1.7, and 4.2 l/kg in mouse, rat, and dog, respectively. The oral bioavailability was 34, 65, and ∼3% in mice, dogs, and rats, respectively. The predicted oral PK profile and parameters of SB939, using Simcyp and allometric scaling, were in good agreement with observed data in humans. Simcyp predictions showed lack of CYP3A4 and 2C19 drug-drug interaction potential for SB939. In summary, the preclinical ADME of SB939 supported its preclinical and clinical development as an oral drug candidate.