Network


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

Hotspot


Dive into the research topics where Dachun Liu is active.

Publication


Featured researches published by Dachun Liu.


Journal of Medicinal Chemistry | 2014

Discovery of ABT-267, a pan-genotypic inhibitor of HCV NS5A.

David A. Degoey; John T. Randolph; Dachun Liu; John K. Pratt; Charles W. Hutchins; Pamela Donner; A. Chris Krueger; Mark A. Matulenko; Sachin V. Patel; Christopher E. Motter; Lissa T. Nelson; Ryan G. Keddy; Michael D. Tufano; Daniel D. Caspi; Preethi Krishnan; Neeta Mistry; Gennadiy Koev; Thomas Reisch; Rubina Mondal; Tami Pilot-Matias; Yi Gao; David W. A. Beno; Clarence J. Maring; Akhter Molla; Emily O. Dumas; Andrew Campbell; Laura Williams; Christine Collins; Rolf Wagner; Warren M. Kati

We describe here N-phenylpyrrolidine-based inhibitors of HCV NS5A with excellent potency, metabolic stability, and pharmacokinetics. Compounds with 2S,5S stereochemistry at the pyrrolidine ring provided improved genotype 1 (GT1) potency compared to the 2R,5R analogues. Furthermore, the attachment of substituents at the 4-position of the central N-phenyl group resulted in compounds with improved potency. Substitution with tert-butyl, as in compound 38 (ABT-267), provided compounds with low-picomolar EC50 values and superior pharmacokinetics. It was discovered that compound 38 was a pan-genotypic HCV inhibitor, with an EC50 range of 1.7-19.3 pM against GT1a, -1b, -2a, -2b, -3a, -4a, and -5a and 366 pM against GT6a. Compound 38 decreased HCV RNA up to 3.10 log10 IU/mL during 3-day monotherapy in treatment-naive HCV GT1-infected subjects and is currently in phase 3 clinical trials in combination with an NS3 protease inhibitor with ritonavir (r) (ABT-450/r) and an NS5B non-nucleoside polymerase inhibitor (ABT-333), with and without ribavirin.


Bioorganic & Medicinal Chemistry Letters | 2012

Novel Hepatitis C virus replicon inhibitors: Synthesis and structure–activity relationships of fused pyrimidine derivatives

A. Chris Krueger; Darold L. Madigan; David W. A. Beno; David A. Betebenner; Robert J. Carrick; Brian E. Green; Wenping He; Dachun Liu; Clarence J. Maring; Keith F. McDaniel; Hongmei Mo; Akhteruzzaman Molla; Christopher E. Motter; Tami Pilot-Matias; Michael D. Tufano; Dale J. Kempf

The synthesis of several pyrido[2,3-d]pyrimidine and pyrimido[4,5-d]pyrimidine analogs is described with one such analog possessing subnanomolar potency in both genotype 1a and 1b cell culture HCV replicon assays.


Journal of Medicinal Chemistry | 2017

Fragment-Based, Structure-Enabled Discovery of Novel Pyridones and Pyridone Macrocycles as Potent Bromodomain and Extra-Terminal Domain (BET) Family Bromodomain Inhibitors

Le Wang; John K. Pratt; Todd N. Soltwedel; George S. Sheppard; Steven D. Fidanze; Dachun Liu; Lisa A. Hasvold; Robert A. Mantei; James H. Holms; William J. McClellan; Michael D. Wendt; Carol K. Wada; Robin R. Frey; T. Matthew Hansen; Robert D. Hubbard; Chang H. Park; Leiming Li; Terrance J. Magoc; Daniel H. Albert; Xiaoyu Lin; Scott E. Warder; Peter Kovar; Xiaoli Huang; Denise Wilcox; Rongqi Wang; Ganesh Rajaraman; Andrew M. Petros; Charles W. Hutchins; Sanjay C. Panchal; Chaohong Sun

Members of the BET family of bromodomain containing proteins have been identified as potential targets for blocking proliferation in a variety of cancer cell lines. A two-dimensional NMR fragment screen for binders to the bromodomains of BRD4 identified a phenylpyridazinone fragment with a weak binding affinity (1, Ki = 160 μM). SAR investigation of fragment 1, aided by X-ray structure-based design, enabled the synthesis of potent pyridone and macrocyclic pyridone inhibitors exhibiting single digit nanomolar potency in both biochemical and cell based assays. Advanced analogs in these series exhibited high oral exposures in rodent PK studies and demonstrated significant tumor growth inhibition efficacy in mouse flank xenograft models.


Cancer Research | 2017

Preclinical Characterization of BET Family Bromodomain Inhibitor ABBV-075 Suggests Combination Therapeutic Strategies

Mai H. Bui; Xiaoyu Lin; Daniel H. Albert; Leiming Li; Lloyd T. Lam; Emily J. Faivre; Scott E. Warder; Xiaoli Huang; Denise Wilcox; Cherrie K. Donawho; George S. Sheppard; Le Wang; Steve D. Fidanze; John K. Pratt; Dachun Liu; Lisa A. Hasvold; Tamar Uziel; Xin Lu; Fred Kohlhapp; Guowei Fang; Steven W. Elmore; Saul H. Rosenberg; Keith F. McDaniel; Warren M. Kati; Yu Shen

ABBV-075 is a potent and selective BET family bromodomain inhibitor that recently entered phase I clinical trials. Comprehensive preclinical characterization of ABBV-075 demonstrated broad activity across cell lines and tumor models, representing a variety of hematologic malignancies and solid tumor indications. In most cancer cell lines derived from solid tumors, ABBV-075 triggers prominent G1 cell-cycle arrest without extensive apoptosis. In this study, we show that ABBV-075 efficiently triggers apoptosis in acute myeloid leukemia (AML), non-Hodgkin lymphoma, and multiple myeloma cells. Apoptosis induced by ABBV-075 was mediated in part by modulation of the intrinsic apoptotic pathway, exhibiting synergy with the BCL-2 inhibitor venetoclax in preclinical models of AML. In germinal center diffuse large B-cell lymphoma, BCL-2 levels or venetoclax sensitivity predicted the apoptotic response to ABBV-075 treatment. In vivo combination studies uncovered surprising benefits of low doses of ABBV-075 coupled with bortezomib and azacitidine treatment, despite the lack of in vitro synergy between ABBV-075 and these agents. The in vitro/in vivo activities of ABBV-075 described here may serve as a useful reference to guide the development of ABBV-075 and other BET family inhibitors for cancer therapy. Cancer Res; 77(11); 2976-89. ©2017 AACR.


Journal of Medicinal Chemistry | 2017

Discovery of N-(4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl)ethanesulfonamide (ABBV-075/mivebresib), a Potent and Orally Available Bromodomain and Extraterminal domain (BET) Family Bromodomain Inhibitor

Keith F. McDaniel; Le Wang; Todd N. Soltwedel; Steven D. Fidanze; Lisa A. Hasvold; Dachun Liu; Robert A. Mantei; John K. Pratt; George S. Sheppard; Mai H. Bui; Emily J. Faivre; Xiaoli Huang; Leiming Li; Xiaoyu Lin; Rongqi Wang; Scott E. Warder; Denise Wilcox; Daniel H. Albert; Terrance J. Magoc; Ganesh Rajaraman; Chang H. Park; Charles W. Hutchins; Jianwei J. Shen; Rohinton Edalji; Chaohong C. Sun; Ruth L. Martin; Wenqing Gao; Shekman Wong; Guowei Fang; Steven W. Elmore

The development of bromodomain and extraterminal domain (BET) bromodomain inhibitors and their examination in clinical studies, particularly in oncology settings, has garnered substantial recent interest. An effort to generate novel BET bromodomain inhibitors with excellent potency and drug metabolism and pharmacokinetics (DMPK) properties was initiated based upon elaboration of a simple pyridone core. Efforts to develop a bidentate interaction with a critical asparagine residue resulted in the incorporation of a pyrrolopyridone core, which improved potency by 9-19-fold. Additional structure-activity relationship (SAR) efforts aimed both at increasing potency and improving pharmacokinetic properties led to the discovery of the clinical candidate 63 (ABBV-075/mivebresib), which demonstrates excellent potency in biochemical and cellular assays, advantageous exposures and half-life both in animal models and in humans, and in vivo efficacy in mouse models of cancer progression and inflammation.


Bioorganic & Medicinal Chemistry Letters | 2017

Methylpyrrole inhibitors of BET bromodomains

Lisa A. Hasvold; George S. Sheppard; Le Wang; Steven D. Fidanze; Dachun Liu; John K. Pratt; Robert A. Mantei; Carol K. Wada; Robbert Hubbard; Yu Shen; Xiaoyu Lin; Xiaoli Huang; Scott E. Warder; Denise Wilcox; Leiming Li; F. Greg Buchanan; Lauren Smithee; Daniel H. Albert; Terrance J. Magoc; Chang H. Park; Andrew M. Petros; Sanjay C. Panchal; Chaohong Sun; Peter Kovar; Nirupama B. Soni; Steven W. Elmore; Warren M. Kati; Keith F. McDaniel

An NMR fragment screen for binders to the bromodomains of BRD4 identified 2-methyl-3-ketopyrroles 1 and 2. Elaboration of these fragments guided by structure-based design provided lead molecules with significant activity in a mouse tumor model. Further modifications to the methylpyrrole core provided compounds with improved properties and enhanced activity in a mouse model of multiple myeloma.


Cancer Research | 2016

Abstract 4718: ABBV-075, a novel BET family bromodomain inhibitor, represents a promising therapeutic agent for a broad spectrum of cancer indications

Aparna V. Sarthy; Leiming Li; Daniel H. Albert; Xiaoyu Lin; Warder Scott; Emily J. Faivre; Mai H. Bui; Xiaoli Huang; Denise Wilcox; Terry Magoc; Fritz G. Buchanan; Paul Tapang; George S. Sheppard; Le Wang; Steve D. Fidanze; John Pratt; Dachun Liu; Lisa A. Hasvold; Paul Hessler; Tamar Uziel; Lloyd T. Lam; Ganesh Rajaraman; Guowei Fang; Steven W. Elmore; Saul H. Rosenberg; Keith F. McDaniel; Warren M. Kati; Yu Shen

Small molecule inhibitors of the bromodomain and extraterminal domain (BET) proteins have emerged as a promising option for cancer therapy. ABBV-075 is a potent and selective BET family bromodomain inhibitor that recently entered Phase 1 clinical trials. It binds bromodomains of BRD2/4/T with similar affinities (Ki of 1-2.2 nM), but exhibits roughly 10-fold weaker potency towards BRD3 (Ki of 12.2 nM). ABBV-075 is highly selective for 18 bromodomain proteins tested (Kd > 1 μM; more than 600-fold selectivity vs. BRD4) and has moderate activity towards CREBBP (Kd = 87 μM; 54-fold selectivity vs. BRD4). ABBV-075 exhibited robust single agent activity in cell viability assays across cancer cell lines derived from solid tumors, leukemia and lymphomas. Further characterization of cancer cell responses to ABBV-075 indicated that ABBV-075 manifested diverse mechanisms of action in different cancer settings. These include 1): disruption of cell cycle control leading to G1 arrest followed by senescence, 2) inhibition of oncogenesis drivers leading to apoptosis and 3) potentially targeting tumor microenvironment to provide additional therapeutic benefit. Consistent with its broad spectrum of activities in vitro, ABBV-075 has comparable or superior efficacies to standard of care agents in flank xenograft mouse models of non-small-cell and small cell lung cancers, pancreatic, breast, prostate, head & neck cancers, multiple myeloma, diffuse large B cell lymphoma and leukemia. These results support the development of ABBV-075 in diverse hematological malignancies and solid tumor indications. Citation Format: Aparna Sarthy, Leiming Li, Daniel H. Albert, Xiaoyu Lin, Warder Scott, Emily Faivre, Mai H. Bui, Xiaoli Huang, Denise M. Wilcox, Terry Magoc, Fritz G. Buchanan, Paul Tapang, George S. Sheppard, Le Wang, Steve D. Fidanze, John Pratt, Dachun Liu, Lisa Hasvold, Paul Hessler, Tamar Uziel, Lloyd Lam, Ganesh Rajaraman, Guowei Fang, Steven W. Elmore, Saul H. Rosenberg, Keith McDaniel, Warren Kati, Yu Shen. ABBV-075, a novel BET family bromodomain inhibitor, represents a promising therapeutic agent for a broad spectrum of cancer indications. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4718.


Bioorganic & Medicinal Chemistry Letters | 2013

Aryl uracil inhibitors of hepatitis C virus NS5B polymerase: Synthesis and characterization of analogs with a fused 5,6-bicyclic ring motif

A. Chris Krueger; John T. Randolph; David A. Degoey; Pamela L. Donner; Charles A. Flentge; Douglas K. Hutchinson; Dachun Liu; Christopher E. Motter; Todd W. Rockway; Rolf Wagner; David W. A. Beno; Gennadiy Koev; Hock B. Lim; Jill M. Beyer; Rubina Mondal; Yaya Liu; Warren M. Kati; Kenton L. Longenecker; Akhteruzzaman Molla; Kent D. Stewart; Clarence J. Maring

The synthesis and structure-activity relationships of a novel aryl uracil series which contains a fused 5,6-bicyclic ring unit for HCV NS5B inhibition is described. Several analogs display replicon cell culture potencies in the low nanomolar range along with excellent rat pharmacokinetic values.


Journal of Medicinal Chemistry | 2018

Highlights of the Structure–Activity Relationships of Benzimidazole Linked Pyrrolidines Leading to the Discovery of the Hepatitis C Virus NS5A Inhibitor Pibrentasvir (ABT-530)

Rolf Wagner; John T. Randolph; Sachin V. Patel; Lissa T. Nelson; Mark A. Matulenko; Ryan G. Keddy; John K. Pratt; Dachun Liu; A. Chris Krueger; Pamela L. Donner; Douglas K. Hutchinson; Charles A. Flentge; David A. Betebenner; Todd W. Rockway; Clarence J. Maring; Teresa I. Ng; Preethi Krishnan; Tami Pilot-Matias; Christine A. Collins; Neeta S. Panchal; Thomas Reisch; Tatyana Dekhtyar; Rubina Mondal; DeAnne Stolarik; Yi Gao; Wenqing Gao; David W A Beno; Warren M. Kati

Curative interferon and ribavirin sparing treatments for hepatitis C virus (HCV)-infected patients require a combination of mechanistically orthogonal direct acting antivirals. A shared component of these treatments is usually an HCV NS5A inhibitor. First generation FDA approved treatments, including the component NS5A inhibitors, do not exhibit equivalent efficacy against HCV virus genotypes 1-6. In particular, these first generation NS5A inhibitors tend to select for viral drug resistance. Ombitasvir is a first generation HCV NS5A inhibitor included as a key component of Viekira Pak for the treatment of patients with HCV genotype 1 infection. Since the launch of next generation HCV treatments, functional cure for genotype 1-6 HCV infections has been achieved, as well as shortened treatment duration across a wider spectrum of genotypes. In this paper, we show how we have modified the anchor, linker, and end-cap architecture of our NS5A inhibitor design template to discover a next generation NS5A inhibitor pibrentasvir (ABT-530), which exhibits potent inhibition of the replication of wild-type genotype 1-6 HCV replicons, as well as improved activity against replicon variants demonstrating resistance against first generation NS5A inhibitors.


Journal of Medicinal Chemistry | 2018

Synthesis and Biological Characterization of Aryl Uracil Inhibitors of Hepatitis C Virus NS5B Polymerase: Discovery of ABT-072, a Trans-Stilbene Analog with Good Oral Bioavailability

John T. Randolph; A. Chris Krueger; Pamela L. Donner; John K. Pratt; Dachun Liu; Christopher E. Motter; Todd W. Rockway; Michael D. Tufano; Rolf Wagner; Hock B. Lim; Jill Beyer; Rubina Mondal; Neeta S. Panchal; Lynn Colletti; Yaya Liu; Gennadiy Koev; Warren M. Kati; Lisa E. Hernandez; David W. A. Beno; Kenton L. Longenecker; Kent D. Stewart; Emily O. Dumas; Akhteruzzaman Molla; Clarence J. Maring

ABT-072 is a non-nucleoside HCV NS5B polymerase inhibitor that was discovered as part of a program to identify new direct-acting antivirals (DAAs) for the treatment of HCV infection. This compound was identified during a medicinal chemistry effort to improve on an original lead, inhibitor 1, which we described in a previous publication. Replacement of the amide linkage in 1 with a trans-olefin resulted in improved compound permeability and solubility and provided much better pharmacokinetic properties in preclinical species. Replacement of the dihydrouracil in 1 with an N-linked uracil provided better potency in the genotype 1 replicon assay. Results from phase 1 clinical studies supported once-daily oral dosing with ABT-072 in HCV infected patients. A phase 2 clinical study that combined ABT-072 with the HCV protease inhibitor ABT-450 provided a sustained virologic response at 24 weeks after dosing (SVR24) in 10 of 11 patients who received treatment.

Collaboration


Dive into the Dachun Liu's collaboration.

Top Co-Authors

Avatar

Warren M. Kati

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John T. Randolph

TAP Pharmaceutical Products

View shared research outputs
Top Co-Authors

Avatar

Michael D. Tufano

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark A. Matulenko

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge