Network


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

Hotspot


Dive into the research topics where Fangyi Zhu is active.

Publication


Featured researches published by Fangyi Zhu.


Nature | 2010

Chemical genetics of Plasmodium falciparum

W. Armand Guiguemde; Anang A. Shelat; David Bouck; Sandra Duffy; Gregory J. Crowther; Paul H. Davis; David C. Smithson; Michele C. Connelly; Julie Clark; Fangyi Zhu; María Belén Jiménez-Díaz; María Santos Martínez; Emily B. Wilson; Abhai K. Tripathi; Jiri Gut; Elizabeth R. Sharlow; Ian Bathurst; Farah El Mazouni; Joseph W. Fowble; Isaac P. Forquer; Paula L. McGinley; Steve Castro; Iñigo Angulo-Barturen; Santiago Ferrer; Philip J. Rosenthal; Joseph L. DeRisi; David J. Sullivan; John S. Lazo; David S. Roos; Michael K. Riscoe

Malaria caused by Plasmodium falciparum is a disease that is responsible for 880,000 deaths per year worldwide. Vaccine development has proved difficult and resistance has emerged for most antimalarial drugs. To discover new antimalarial chemotypes, we have used a phenotypic forward chemical genetic approach to assay 309,474 chemicals. Here we disclose structures and biological activity of the entire library—many of which showed potent in vitro activity against drug-resistant P. falciparum strains—and detailed profiling of 172 representative candidates. A reverse chemical genetic study identified 19 new inhibitors of 4 validated drug targets and 15 novel binders among 61 malarial proteins. Phylochemogenetic profiling in several organisms revealed similarities between Toxoplasma gondii and mammalian cell lines and dissimilarities between P. falciparum and related protozoans. One exemplar compound displayed efficacy in a murine model. Our findings provide the scientific community with new starting points for malaria drug discovery.


Journal of Biological Chemistry | 2010

Identification and characterization of the first small molecule inhibitor of MDMX.

Damon R. Reed; Ying Shen; Anang A. Shelat; Leggy A. Arnold; Antonio M. Ferreira; Fangyi Zhu; Nicholas Mills; David C. Smithson; Catherine Regni; Donald Bashford; Samantha A. Cicero; Brenda A. Schulman; Aart G. Jochemsen; R. Kiplin Guy; Michael A. Dyer

The p53 pathway is disrupted in virtually every human tumor. In ∼50% of human cancers, the p53 gene is mutated, and in the remaining cancers, the pathway is dysregulated by genetic lesions in other genes that modulate the p53 pathway. One common mechanism for inactivation of the p53 pathway in tumors that express wild-type p53 is increased expression of MDM2 or MDMX. MDM2 and MDMX bind p53 and inhibit its function by distinct nonredundant mechanisms. Small molecule inhibitors and small peptides have been developed that bind MDM2 in the p53-binding pocket and displace the p53 protein, leading to p53-mediated cell cycle exit and apoptosis. To date, peptide inhibitors of MDMX have been developed, but no small molecule inhibitors have been reported. We have developed biochemical and cell-based assays for high throughput screening of chemical libraries to identify MDMX inhibitors and identified the first MDMX inhibitor SJ-172550. This compound binds reversibly to MDMX and effectively kills retinoblastoma cells in which the expression of MDMX is amplified. The effect of SJ-172550 is additive when combined with an MDM2 inhibitor. Results from a series of biochemical and structural modeling studies suggest that SJ-172550 binds the p53-binding pocket of MDMX, thereby displacing p53. This lead compound is a useful chemical scaffold for further optimization of MDMX inhibitors that may eventually be used to treat pediatric cancers and various adult tumors that overexpress MDMX or have similar genetic lesions. When combined with selective MDM2 inhibitors, SJ-172550 may also be useful for treating tumors that express wild-type p53.


Proceedings of the National Academy of Sciences of the United States of America | 2014

(+)-SJ733, a clinical candidate for malaria that acts through ATP4 to induce rapid host-mediated clearance of Plasmodium

María Belén Jiménez-Díaz; Daniel H. Ebert; Yandira Salinas; Anupam Pradhan; Adele M. Lehane; Marie-Eve Myrand-Lapierre; Kathleen O’Loughlin; David M. Shackleford; Mariana Justino de Almeida; Angela K. Carrillo; Julie Clark; Adelaide S. M. Dennis; Jonathon Diep; Xiaoyan Deng; Sandra Duffy; Aaron N. Endsley; Greg Fedewa; W. Armand Guiguemde; María G. Gómez; Gloria Holbrook; Jeremy A. Horst; Charles C. Kim; Jian Liu; Marcus C. S. Lee; Amy Matheny; María Santos Martínez; Gregory Miller; Ane Rodríguez-Alejandre; Laura Sanz; Martina Sigal

Significance Useful antimalarial drugs must be rapidly acting, highly efficacious, and have low potential for developing resistance. (+)-SJ733 targets a Plasmodium cation-transporting ATPase, ATP4. (+)-SJ733 cleared parasites in vivo as quickly as artesunate by specifically inducing eryptosis/senescence in infected, treated erythrocytes. Although in vitro selection of pfatp4 mutants with (+)-SJ733 proceeded with moderate frequency, during in vivo selection of pbatp4 mutants, resistance emerged slowly and produced marginally resistant mutants with poor fitness. In addition, (+)-SJ733 met all other criteria for a clinical candidate, including high oral bioavailability, a high safety margin, and transmission blocking activity. These results demonstrate that targeting ATP4 has great potential to deliver useful drugs for malaria eradication. Drug discovery for malaria has been transformed in the last 5 years by the discovery of many new lead compounds identified by phenotypic screening. The process of developing these compounds as drug leads and studying the cellular responses they induce is revealing new targets that regulate key processes in the Plasmodium parasites that cause malaria. We disclose herein that the clinical candidate (+)-SJ733 acts upon one of these targets, ATP4. ATP4 is thought to be a cation-transporting ATPase responsible for maintaining low intracellular Na+ levels in the parasite. Treatment of parasitized erythrocytes with (+)-SJ733 in vitro caused a rapid perturbation of Na+ homeostasis in the parasite. This perturbation was followed by profound physical changes in the infected cells, including increased membrane rigidity and externalization of phosphatidylserine, consistent with eryptosis (erythrocyte suicide) or senescence. These changes are proposed to underpin the rapid (+)-SJ733-induced clearance of parasites seen in vivo. Plasmodium falciparum ATPase 4 (pfatp4) mutations that confer resistance to (+)-SJ733 carry a high fitness cost. The speed with which (+)-SJ733 kills parasites and the high fitness cost associated with resistance-conferring mutations appear to slow and suppress the selection of highly drug-resistant mutants in vivo. Together, our data suggest that inhibitors of PfATP4 have highly attractive features for fast-acting antimalarials to be used in the global eradication campaign.


Cancer Research | 2011

Targeting the p53 Pathway in Retinoblastoma with Subconjunctival Nutlin-3a

Rachel Brennan; Sara M. Federico; Cori Bradley; Jiakun Zhang; Jacqueline Flores-Otero; Matthew W. Wilson; Clinton F. Stewart; Fangyi Zhu; Kip Guy; Michael A. Dyer

Retinoblastoma is a rare childhood cancer of the retina that begins in utero and is diagnosed in the first years of life. The goals of retinoblastoma treatment are ocular salvage, vision preservation, and reduction of short- and long-term side effects without risking mortality because of tumor dissemination. To identify better chemotherapeutic combinations for the treatment of retinoblastoma, several groups have developed genetic mouse models and orthotopic xenograft models of human retinoblastoma for preclinical testing. Previous studies have implicated the MDMX protein in the suppression of the p53 pathway in retinoblastoma and shown that the MDM2/MDMX antagonist, Nutlin-3a, can efficiently induce p53-mediated cell death in retinoblastoma cell lines. However, Nutlin-3a cannot be administered systemically to treat retinoblastoma, because it has poor penetration across the blood-ocular barrier. Therefore, we developed an ocular formulation of Nutlin-3a, Nutlin-3a(OC), and tested the pharmacokinetics and efficacy of this new formulation in genetic and human retinoblastoma orthotopic xenograft models of retinoblastoma. Here, we show that Nutlin-3a(OC) specifically and efficiently targets the p53 pathway and that the combination of Nutlin-3a(OC) with systemic topotecan is a significantly better treatment for retinoblastoma than currently used chemotherapy in human orthotopic xenografts. Our studies provide a new standardized approach to evaluate and prioritize novel agents for incorporation into future clinical trials for retinoblastoma.


Journal of Medicinal Chemistry | 2012

Lead optimization of 3-carboxyl-4(1H)-quinolones to deliver orally bioavailable antimalarials.

Yiqun Zhang; Julie Clark; Michele C. Connelly; Fangyi Zhu; Jaeki Min; W. Armand Guiguemde; Anupam Pradhan; Lalitha V. Iyer; Anna Furimsky; Jason Gow; Toufan Parman; Farah El Mazouni; Margaret A. Phillips; Dennis E. Kyle; Jon C. Mirsalis; R. Kiplin Guy

Malaria is a protozoal parasitic disease that is widespread in tropical and subtropical regions of Africa, Asia, and the Americas and causes more than 800,000 deaths per year. The continuing emergence of multidrug-resistant Plasmodium falciparum drives the ongoing need for the development of new and effective antimalarial drugs. Our previous work has explored the preliminary structural optimization of 4(1H)-quinolone ester derivatives, a new series of antimalarials related to the endochins. Herein, we report the lead optimization of 4(1H)-quinolones with a focus on improving both antimalarial potency and bioavailability. These studies led to the development of orally efficacious antimalarials including quinolone analogue 20g, a promising candidate for further optimization.


Drug Metabolism and Disposition | 2011

Whole-Body Physiologically Based Pharmacokinetic Model for Nutlin-3a in Mice after Intravenous and Oral Administration

Fan Zhang; Michael Tagen; Stacy L. Throm; Jeremy P. Mallari; Laura Miller; R. Kiplin Guy; Michael A. Dyer; Richard T. Williams; Martine F. Roussel; Katie Nemeth; Fangyi Zhu; Jiakun Zhang; Min Lu; John C. Panetta; Nidal Boulos; Clinton F. Stewart

Nutlin-3a is an MDM2 inhibitor that is under investigation in preclinical models for a variety of pediatric malignancies, including retinoblastoma, rhabdomyosarcoma, neuroblastoma, and leukemia. We used physiologically based pharmacokinetic (PBPK) modeling to characterize the disposition of nutlin-3a in the mouse. Plasma protein binding and blood partitioning were assessed by in vitro studies. After intravenous (10 and 20 mg/kg) and oral (50, 100, and 200 mg/kg) dosing, tissue concentrations of nutlin-3a were determined in plasma, liver, spleen, intestine, muscle, lung, adipose, bone marrow, adrenal gland, brain, retina, and vitreous fluid. The PBPK model was simultaneously fit to all pharmacokinetic data using NONMEM. Nutlin-3a exhibited nonlinear binding to murine plasma proteins, with the unbound fraction ranging from 0.7 to 11.8%. Nutlin-3a disposition was characterized by rapid absorption with peak plasma concentrations at approximately 2 h and biphasic elimination consistent with a saturable clearance process. The final PBPK model successfully described the plasma and tissue disposition of nutlin-3a. Simulations suggested high bioavailability, rapid attainment of steady state, and little accumulation when administered once or twice daily at dosages up to 400 mg/kg. The final model was used to perform simulations of unbound tissue concentrations to determine which dosing regimens are appropriate for preclinical models of several pediatric malignancies.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery of trypanocidal thiosemicarbazone inhibitors of rhodesain and TbcatB

Jeremy P. Mallari; Anang A. Shelat; Aaron Kosinski; Conor R. Caffrey; Michele C. Connelly; Fangyi Zhu; James H. McKerrow; R. Kiplin Guy

Human African trypanosomiasis (HAT) is caused by the protozoan parasite Trypanosoma brucei. The cysteine proteases of T. brucei have been shown to be crucial for parasite replication and represent an attractive point for therapeutic intervention. Herein we describe the synthesis of a series of thiosemicarbazones and their activity against the trypanosomal cathepsins TbcatB and rhodesain, as well as human cathepsins L and B. The activity of these compounds was determined against cultured T. brucei, and specificity was assessed with a panel of four mammalian cell lines.


Journal of Medicinal Chemistry | 2009

Improvement of Pharmacological Properties of Irreversible Thyroid Receptor Coactivator Binding Inhibitors

Jong Yeon Hwang; Leggy A. Arnold; Fangyi Zhu; Aaron Kosinski; Thomas J. Mangano; Vincent Setola; Bryan L. Roth; R. Kiplin Guy

We have previously reported the discovery and preliminary structure activity relationships of a series of beta-aminoketones that disrupt the binding of coactivators to TR. However, the most active compounds had moderate inhibitory potency and relatively high cytotoxicity, resulting in narrow therapeutic index. Additionally, preliminary evaluation of in vivo toxicology revealed a significant dose related cardiotoxicity. Here we describe the improvement of pharmacological properties of thyroid hormone receptor coactivator binding inhibitors. A comprehensive survey of the effects of substitutents in key areas of the molecule was carried out based on mechanistic insight from the earlier report. This study revealed that both electron withdrawing and hydrophobic substituents on the aromatic ring led to higher potency. On the other hand, moving from an alkyl to a sulfonyl alkyl side chain led to reduced cytotoxicity. Finally, utilization of amine moieties having low pK(a)s resulted in lowered ion channel activity without any loss of pharmacological activity.


Bioorganic & Medicinal Chemistry | 2010

Synthesis and Structure-Activity Relationships of Antimalarial 4-oxo-3-carboxyl quinolones

Yiqun Zhang; W. Armand Guiguemde; Martina Sigal; Fangyi Zhu; Michele C. Connelly; Solomon Nwaka; R. Kiplin Guy

Malaria is endemic in tropical and subtropical regions of Africa, Asia, and the Americas. The increasing prevalence of multi-drug-resistant Plasmodium falciparum drives the ongoing need for the development of new antimalarial drugs. In this light, novel scaffolds to which the parasite has not been exposed are of particular interest. Recently, workers at the Swiss Tropical Institute discovered two novel 4-oxo-3-carboxyl quinolones active against the intra-erythrocytic stages of P. falciparum while carrying out rationally directed low-throughput screening of potential antimalarial agents as part of an effort directed by the World Health Organization. Here we report the design, synthesis, and preliminary pharmacologic characterization of a series of analogues of 4-oxo-3-carboxyl quinolones. These studies indicate that the series has good potential for preclinical development.


Journal of Medicinal Chemistry | 2011

Synthesis and evaluation of 7-substituted 4-aminoquinoline analogues for antimalarial activity.

Jong Yeon Hwang; Takashi Kawasuji; David J. Lowes; Julie Clark; Michele C. Connelly; Fangyi Zhu; W. Armand Guiguemde; Martina Sigal; Emily B. Wilson; Joseph L. DeRisi; R. Kiplin Guy

We previously reported that substituted 4-aminoquinolines with a phenyl ether substituent at the 7-position of the quinoline ring and the capability of intramolecular hydrogen bonding between the protonated amine on the side chain and a hydrogen bond acceptor on the amines alkyl substituents exhibited potent antimalarial activity against the multidrug resistant strain P. falciparum W2. We employed a parallel synthetic method to generate diaryl ether, biaryl, and alkylaryl 4-aminoquinoline analogues in the background of a limited number of side chain variations that had previously afforded potent 4-aminoquinolines. All subsets were evaluated for their antimalarial activity against the chloroquine-sensitive strain 3D7 and the chloroquine-resistant K1 strain as well as for cytotoxicity against mammalian cell lines. While all three arrays showed good antimalarial activity, only the biaryl-containing subset showed consistently good potency against the drug-resistant K1 strain and good selectivity with regard to mammalian cytotoxicity. Overall, our data indicate that the biaryl-containing series contains promising candidates for further study.

Collaboration


Dive into the Fangyi Zhu's collaboration.

Top Co-Authors

Avatar

R. Kiplin Guy

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Michele C. Connelly

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Julie Clark

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

W. Armand Guiguemde

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Martina Sigal

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

David C. Smithson

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Gloria Holbrook

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Lemoff

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Lei Yang

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge