Network


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

Hotspot


Dive into the research topics where Fuqiang Ban is active.

Publication


Featured researches published by Fuqiang Ban.


Journal of Biological Chemistry | 2014

Selectively Targeting the DNA-binding Domain of the Androgen Receptor as a Prospective Therapy for Prostate Cancer

Kush Dalal; Mani Roshan-Moniri; Aishwariya Sharma; Huifang Li; Fuqiang Ban; Mohamed Hessein; Michael Hsing; Kriti Singh; Eric Leblanc; Scott M. Dehm; Emma S. Guns; Artem Cherkasov; Paul S. Rennie

Background: The androgen receptor (AR) is a transcription factor regulating progression of prostate cancer. Results: Developed compounds inhibit AR transcriptional activity in vitro and in vivo by selective targeting of the AR-DNA-binding domain (DBD). Conclusion: By targeting the DBD, the compounds differ from conventional anti-androgens. Significance: Anti-androgens with a novel mechanism of action have the potential to treat recurrent prostate cancer. The androgen receptor (AR) is a transcription factor that has a pivotal role in the occurrence and progression of prostate cancer. The AR is activated by androgens that bind to its ligand-binding domain (LBD), causing the transcription factor to enter the nucleus and interact with genes via its conserved DNA-binding domain (DBD). Treatment for prostate cancer involves reducing androgen production or using anti-androgen drugs to block the interaction of hormones with the AR-LBD. Eventually the disease changes into a castration-resistant form of PCa where LBD mutations render anti-androgens ineffective or where constitutively active AR splice variants, lacking the LBD, become overexpressed. Recently, we identified a surfaced exposed pocket on the AR-DBD as an alternative drug-target site for AR inhibition. Here, we demonstrate that small molecules designed to selectively bind the pocket effectively block transcriptional activity of full-length and splice variant AR forms at low to sub-micromolar concentrations. The inhibition is lost when residues involved in drug interactions are mutated. Furthermore, the compounds did not impede nuclear localization of the AR and blocked interactions with chromatin, indicating the interference of DNA binding with the nuclear form of the transcription factor. Finally, we demonstrate the inhibition of gene expression and tumor volume in mouse xenografts. Our results indicate that the AR-DBD has a surface site that can be targeted to inhibit all forms of the AR, including enzalutamide-resistant and constitutively active splice variants and thus may serve as a potential avenue for the treatment of recurrent and metastatic prostate cancer.


Journal of Medicinal Chemistry | 2013

Targeting the Binding Function 3 (BF3) Site of the Androgen Receptor Through Virtual Screening. 2. Development of 2-((2-phenoxyethyl) thio)-1H-benzimidazole Derivatives

Ravi Shashi Nayana Munuganti; Eric Leblanc; Peter Axerio-Cilies; Christophe Labrière; Kate Frewin; Kriti Singh; Mohamed D. Hassona; Nathan A. Lack; Huifang Li; Fuqiang Ban; Emma Tomlinson Guns; Robert J. Young; Paul S. Rennie; Artem Cherkasov

The human androgen receptor (AR) is a proven therapeutic target in prostate cancer. All current antiandrogens, such as Bicalutamide, Flutamide, Nilutamide, and Enzalutamide, target the buried hydrophobic androgen binding pocket of this protein. However, effective resistance mechanisms against these therapeutics exist such as mutations occurring at the target site. To overcome these limitations, the surface pocket of the AR called binding function 3 (BF3) was characterized as an alternative target for small molecule therapeutics. A number of AR inhibitors directly targeting the BF3 were previously identified by us ( J. Med. Chem. 2011 . 54 , 8563 ). In the current study, based on the prior results, we have developed structure-activity relationships that allowed designing a series of 2-((2-phenoxyethyl)thio)-1H-benzimidazole and 2-((2-phenoxyethyl)thio)-1H-indole as lead BF3 inhibitors. Some of the developed BF3 ligands demonstrated significant antiandrogen potency against LNCaP and Enzalutamide-resistant prostate cancer cell lines.


Journal of Medicinal Chemistry | 2014

Discovery of small-molecule inhibitors selectively targeting the DNA-binding domain of the human androgen receptor.

Huifang Li; Fuqiang Ban; Kush Dalal; Eric Leblanc; Kate Frewin; Dennis Ma; Hans Adomat; Paul S. Rennie; Artem Cherkasov

The human androgen receptor (AR) is considered as a master regulator in the development and progression of prostate cancer (PCa). As resistance to clinically used anti-AR drugs remains a major challenge for the treatment of advanced PCa, there is a pressing need for new anti-AR therapeutic avenues. In this study, we identified a binding site on the DNA binding domain (DBD) of the receptor and utilized virtual screening to discover a set of micromolar hits for the target. Through further exploration of the most potent hit (1), a structural analogue (6) was identified demonstrating 10-fold improved anti-AR potency. Further optimization resulted in a more potent synthetic analogue (25) with anti-AR potency comparable to a newly FDA-approved drug Enzalutamide. Site-directed mutagenesis demonstrated that the developed inhibitors do interact with the intended target site. Importantly, the AR DBD inhibitors could effectively inhibit the growth of Enzalutamide-resistant cells as well as block the transcriptional activity of constitutively active AR splice variants, such as V7.


Chemistry & Biology | 2014

Identification of a Potent Antiandrogen that Targets the BF3 Site of the Androgen Receptor and Inhibits Enzalutamide-Resistant Prostate Cancer

Ravi Shashi Nayana Munuganti; Mohamed D. Hassona; Eric Leblanc; Kate Frewin; Kriti Singh; Dennis Ma; Fuqiang Ban; Michael Hsing; Hans Adomat; Nada Lallous; Christophe Andre; Jon Paul Selvam Jonadass; Amina Zoubeidi; Robert N. Young; Emma Tomlinson Guns; Paul S. Rennie; Artem Cherkasov

There has been a resurgence of interest in the development of androgen receptor (AR) inhibitors with alternative modes of action to overcome the development of resistance to current therapies. We demonstrated previously that one promising strategy for combatting mutation-driven drug resistance is to target the Binding Function 3 (BF3) pocket of the receptor. Here we report the development of a potent BF3 inhibitor, 3-(2,3-dihydro-1H-indol-2-yl)-1H-indole, which demonstrates excellent antiandrogen potency and anti-PSA activity and abrogates the androgen-induced proliferation of androgen-sensitive (LNCaP) and enzalutamide-resistant (MR49F) PCa cell lines. Moreover, this compound effectively reduces the expression of AR-dependent genes in PCa cells and effectively inhibits tumor growth in vivo in both LNCaP and MR49F xenograft models. These findings provide evidence that targeting the AR BF3 pocket represents a viable therapeutic approach to treat patients with advanced and/or resistant prostate cancer.


Journal of Medicinal Chemistry | 2014

Discovery of 1H-indole-2-carboxamides as novel inhibitors of the androgen receptor binding function 3 (BF3).

Fuqiang Ban; Eric Leblanc; Huifang Li; Ravi Shashi Nayana Munuganti; Kate Frewin; Paul S. Rennie; Artem Cherkasov

To overcome resistance to conventional anti-androgens of human androgen receptor (AR), the allosteric site of the AR binding function 3 (BF3) was investigated as an alternative target for small molecule therapeutics. A library of 1H-indole-2-carboxamides were discovered as BF3 inhibitors and exhibited strong antiproliferative activity against LNCaP and enzalutamide-resistant prostate cancer cell lines. Several of the lead compounds may prove of particular benefit as a novel alternative treatment for castration-resistant prostate cancers.


Proteins | 2007

Indel-based targeting of essential proteins in human pathogens that have close host orthologue(s): Discovery of selective inhibitors for Leishmania donovani elongation factor-1α

Devki Nandan; Martin Lopez; Fuqiang Ban; Meilan Huang; Yvonne Y. Li; Neil E. Reiner; Artem Cherkasov

We propose a novel strategy for selective targeting of essential pathogen proteins that contain sizable indels (insertions/deletions) in their sequences compared with their host orthologues. This approach has been tested on elongation factor‐1α (EF‐1α) from the protozoan pathogen Leishmania donovani. Leishmania EF‐1α is 82% identical to the corresponding human orthologue, but possesses a 12 aminoacid sequence deletion compared with human EF‐1α. We used this indel‐differentiated region to design small molecules that selectively bind to leishmania EF‐1α and not to the human protein. Three unrelated molecules were identified with the capacity to inhibit protein synthesis in leishmania by up to 75% while exhibiting no effect on human protein translation. These candidates may serve as prototypes for future development of antiprotozoan therapeutics. More generally, these findings provide a basis for a novel drug design platform. This platform targets essential pathogen proteins that are highly conserved across species, and consequently would not typically be considered to be conventional drug targets. We anticipate that such indel‐directed targeting of essential proteins in microbial pathogens may help address the growing problem of antibiotic resistance. Proteins 2007.


Toxicology and Applied Pharmacology | 2009

In silico identification of anthropogenic chemicals as ligands of zebrafish sex hormone binding globulin

Nels Thorsteinson; Fuqiang Ban; Osvaldo Santos-Filho; Seyed M.H. Tabaei; Solange Miguel-Queralt; Caroline Underhill; Artem Cherkasov; Geoffrey L. Hammond

Anthropogenic compounds with the capacity to interact with the steroid-binding site of sex hormone binding globulin (SHBG) pose health risks to humans and other vertebrates including fish. Building on studies of human SHBG, we have applied in silico drug discovery methods to identify potential binders for SHBG in zebrafish (Danio rerio) as a model aquatic organism. Computational methods, including; homology modeling, molecular dynamics simulations, virtual screening, and 3D QSAR analysis, successfully identified 6 non-steroidal substances from the ZINC chemical database that bind to zebrafish SHBG (zfSHBG) with low-micromolar to nanomolar affinities, as determined by a competitive ligand-binding assay. We also screened 80,000 commercial substances listed by the European Chemicals Bureau and Environment Canada, and 6 non-steroidal hits from this in silico screen were tested experimentally for zfSHBG binding. All 6 of these compounds displaced the [(3)H]5alpha-dihydrotestosterone used as labeled ligand in the zfSHBG screening assay when tested at a 33 microM concentration, and 3 of them (hexestrol, 4-tert-octylcatechol, and dihydrobenzo(a)pyren-7(8H)-one) bind to zfSHBG in the micromolar range. The study demonstrates the feasibility of large-scale in silico screening of anthropogenic compounds that may disrupt or highjack functionally important protein:ligand interactions. Such studies could increase the awareness of hazards posed by existing commercial chemicals at relatively low cost.


Molecular Cancer Therapeutics | 2017

Bypassing drug-resistance mechanisms of prostate cancer with small-molecules that target androgen receptor chromatin interactions

Kush Dalal; Meixia Che; Nanette L. S. Que; Aishwariya Sharma; Rendong Yang; Nada Lallous; Hendrik Borgmann; Deniz Ozistanbullu; Ronnie Tse; Fuqiang Ban; Huifang Li; Kevin Tam; Mani Roshan-Moniri; Eric Leblanc; Martin Gleave; Daniel T. Gewirth; Scott M. Dehm; Artem Cherkasov; Paul S. Rennie

Human androgen receptor (AR) is a hormone-activated transcription factor that is an important drug target in the treatment of prostate cancer. Current small-molecule AR antagonists, such as enzalutamide, compete with androgens that bind to the steroid-binding pocket of the AR ligand–binding domain (LBD). In castration-resistant prostate cancer (CRPC), drug resistance can manifest through AR-LBD mutations that convert AR antagonists into agonists, or by expression of AR variants lacking the LBD. Such treatment resistance underscores the importance of novel ways of targeting the AR. Previously, we reported the development of a series of small molecules that were rationally designed to selectively target the AR DNA-binding domain (DBD) and, hence, to directly interfere with AR–DNA interactions. In the current work, we have confirmed that the lead AR DBD inhibitor indeed directly interacts with the AR-DBD and tested that substance across multiple clinically relevant CRPC cell lines. We have also performed a series of experiments that revealed that genome-wide chromatin binding of AR was dramatically impacted by the lead compound (although with lesser effect on AR variants). Collectively, these observations confirm the novel mechanism of antiandrogen action of the developed AR-DBD inhibitors, establishing proof of principle for targeting DBDs of nuclear receptors in endocrine cancers. Mol Cancer Ther; 16(10); 2281–91. ©2017 AACR.


Journal of Chemical Information and Modeling | 2017

Best Practices of Computer-Aided Drug Discovery: Lessons Learned from the Development of a Preclinical Candidate for Prostate Cancer with a New Mechanism of Action

Fuqiang Ban; Kush Dalal; Huifang Li; Eric Leblanc; Paul S. Rennie; Artem Cherkasov

Small-molecule drug design is a complex and iterative decision-making process relying on pre-existing knowledge and driven by experimental data. Low-molecular-weight chemicals represent an attractive therapeutic option, as they are readily accessible to organic synthesis and can easily be characterized.1 Their potency as well as pharmacokinetic and pharmacodynamic properties can be systematically and rationally investigated and ultimately optimized via expert science behind medicinal chemistry and methods of computer-aided drug design (CADD). In recent years, significant advances in molecular modeling techniques have afforded a variety of tools to effectively identify potential binding pockets on prospective targets, to map key interactions between ligands and their binding sites, to construct and assess energetics of the resulting complexes, to predict ADMET properties of candidate compounds, and to systematically analyze experimental and computational data to derive meaningful structure-activity relationships leading to the creation of a drug candidate. This Perspective describes a real case of a drug discovery campaign accomplished in a relatively short time with limited resources. The study integrated an arsenal of available molecular modeling techniques with an array of experimental tools to successfully develop a novel class of potent and selective androgen receptor inhibitors with a novel mode of action. It resulted in the largest academic licensing deal in Canadian history, totaling


Journal of Cheminformatics | 2017

SimBoost : a read-across approach for predicting drug–target binding affinities using gradient boosting machines

Tong He; Marten Heidemeyer; Fuqiang Ban; Artem Cherkasov; Martin Ester

142M. This project exemplifies the importance of team science, an integrative approach to drug discovery, and the use of best practices in CADD. We posit that the lessons learned and best practices for executing an effective CADD project can be applied, with similar success, to many drug discovery projects in both academia and industry.

Collaboration


Dive into the Fuqiang Ban's collaboration.

Top Co-Authors

Avatar

Artem Cherkasov

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Eric Leblanc

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Paul S. Rennie

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Huifang Li

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Kush Dalal

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Kate Frewin

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Michael Hsing

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Kriti Singh

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Mani Roshan-Moniri

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Mohamed D. Hassona

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge