Network


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

Hotspot


Dive into the research topics where Penchit Chitnumsub is active.

Publication


Featured researches published by Penchit Chitnumsub.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Malarial dihydrofolate reductase as a paradigm for drug development against a resistance-compromised target

Yongyuth Yuthavong; Bongkoch Tarnchompoo; Tirayut Vilaivan; Penchit Chitnumsub; Sumalee Kamchonwongpaisan; Susan A. Charman; Danielle N McLennan; Karen L. White; Livia Vivas; Emily Bongard; Chawanee Thongphanchang; Jarunee Vanichtanankul; Roonglawan Rattanajak; Uthai Arwon; Pascal Fantauzzi; Jirundon Yuvaniyama; William N. Charman; David Matthews

Malarial dihydrofolate reductase (DHFR) is the target of antifolate antimalarial drugs such as pyrimethamine and cycloguanil, the clinical efficacy of which have been compromised by resistance arising through mutations at various sites on the enzyme. Here, we describe the use of cocrystal structures with inhibitors and substrates, along with efficacy and pharmacokinetic profiling for the design, characterization, and preclinical development of a selective, highly efficacious, and orally available antimalarial drug candidate that potently inhibits both wild-type and clinically relevant mutated forms of Plasmodium falciparum (Pf) DHFR. Important structural characteristics of P218 include pyrimidine side-chain flexibility and a carboxylate group that makes charge-mediated hydrogen bonds with conserved Arg122 (PfDHFR-TS amino acid numbering). An analogous interaction of P218 with human DHFR is disfavored because of three species-dependent amino acid substitutions in the vicinity of the conserved Arg. Thus, P218 binds to the active site of PfDHFR in a substantially different fashion from the human enzyme, which is the basis for its high selectivity. Unlike pyrimethamine, P218 binds both wild-type and mutant PfDHFR in a slow-on/slow-off tight-binding mode, which prolongs the target residence time. P218, when bound to PfDHFR-TS, resides almost entirely within the envelope mapped out by the dihydrofolate substrate, which may make it less susceptible to resistance mutations. The high in vivo efficacy in a SCID mouse model of P. falciparum malaria, good oral bioavailability, favorable enzyme selectivity, and good safety characteristics of P218 make it a potential candidate for further development.


Parasitology | 2005

Malarial (Plasmodium falciparum) dihydrofolate reductase-thymidylate synthase: structural basis for antifolate resistance and development of effective inhibitors.

Yongyuth Yuthavong; Jirundon Yuvaniyama; Penchit Chitnumsub; Jarunee Vanichtanankul; Sudsanguan Chusacultanachai; Bongkoch Tarnchompoo; Tirayut Vilaivan; Sumalee Kamchonwongpaisan

Dihydrofolate reductase-thymidylate synthase (DHFR-TS) from Plasmodium falciparum, a validated target for antifolate antimalarials, is a dimeric enzyme with interdomain interactions significantly mediated by the junction region as well as the Plasmodium-specific additional sequences (inserts) in the DHFR domain. The X-ray structures of both the wild-type and mutant enzymes associated with drug resistance, in complex with either a drug which lost, or which still retains, effectiveness for the mutants, reveal features which explain the basis of drug resistance resulting from mutations around the active site. Binding of rigid inhibitors like pyrimethamine and cycloguanil to the enzyme active site is affected by steric conflict with the side-chains of mutated residues 108 and 16, as well as by changes in the main chain configuration. The role of important residues on binding of inhibitors and substrates was further elucidated by site-directed and random mutagenesis studies. Guided by the active site structure and modes of inhibitor binding, new inhibitors with high affinity against both wild-type and mutant enzymes have been designed and synthesized, some of which have very potent anti-malarial activities against drug-resistant P. falciparum bearing the mutant enzymes.


ACS Chemical Biology | 2009

Exploiting structural analysis, in silico screening, and serendipity to identify novel inhibitors of drug-resistant falciparum malaria.

Tina Dasgupta; Penchit Chitnumsub; Sumalee Kamchonwongpaisan; Cherdsak Maneeruttanarungroj; Sara E. Nichols; Theresa M. Lyons; Julian Tirado-Rives; William L. Jorgensen; Yongyuth Yuthavong; Karen S. Anderson

Plasmodium falciparum thymidylate synthase-dihydrofolate reductase (TS-DHFR) is an essential enzyme in folate biosynthesis and a major malarial drug target. This bifunctional enzyme thus presents different design approaches for developing novel inhibitors against drug-resistant mutants. We performed a high-throughput in silico screen of a database of diverse, drug-like molecules against a non-active-site pocket of TS-DHFR. The top compounds from this virtual screen were evaluated by in vitro enzymatic and cellular culture studies. Three compounds active to 20 microM IC(50)s in both wildtype and antifolate-resistant P. falciparum parasites were identified; moreover, no inhibition of human DHFR enzyme was observed, indicating that the inhibitory effects appeared to be parasite-specific. Notably, all three compounds had a biguanide scaffold. However, relative free energy of binding calculations suggested that the compounds might preferentially interact with the active site over the screened non-active-site region. To resolve the two possible modes of binding, co-crystallization studies of the compounds complexed with TS-DHFR enzyme were performed. Surprisingly, the structural analysis revealed that these novel, biguanide compounds do indeed bind at the active site of DHFR and additionally revealed the molecular basis by which they overcome drug resistance. To our knowledge, these are the first co-crystal structures of novel, biguanide, non-WR99210 compounds that are active against folate-resistant malaria parasites in cell culture.


Acta Crystallographica Section D-biological Crystallography | 2004

Characterization, crystallization and preliminary X-­ray analysis of bifunctional dihydrofolate reductase–thymidylate synthase from Plasmodium falciparum

Penchit Chitnumsub; Jirundon Yuvaniyama; Jarunee Vanichtanankul; Sumalee Kamchonwongpaisan; Walkinshaw; Yongyuth Yuthavong

The full-length pfdhfr-ts genes of the wild-type TM4/8.2 and the double mutant K1CB1 (C59R+S108N) from the genomic DNA of the corresponding Plasmodium falciparum parasite have been cloned into a modified pET(17b) plasmid and expressed in Escherichia coli BL21 (DE3) pLysS. Conditions for the expression and purification of the P. falciparum dihydrofolate reductase-thymidylate synthase (PfDHFR-TS) have been established that yield approximately 1 mg of the soluble active enzyme per litre of culture. The purified enzymes have been crystallized using a modified microbatch method with PEG 4000 as the primary precipitating agent. X-ray diffraction data were collected to 2.50 and 2.64 A resolution under cryogenic conditions from single crystals of the two PfDHFR-TS proteins in complex with NADPH, dUMP and either Pyr30 or Pyr39. Preliminary X-ray analysis indicated that the crystals belong to the orthorhombic space group P2(1)2(1)2(1), with two molecules per asymmetric unit and approximately 52% solvent content (VM approximately 2.6 A3 Da-1). The use of a particular type of baby oil in the microbatch setup appeared to be beneficial to PfDHFR-TS crystallization and a preliminary comparison with another commonly used oil is described.


Future Microbiology | 2006

Folate metabolism as a source of molecular targets for antimalarials.

Yongyuth Yuthavong; Sumalee Kamchonwongpaisan; Ubolsree Leartsakulpanich; Penchit Chitnumsub

Folate metabolism of the malaria parasites provides two targets for current antimalarials: dihydrofolate reductase and dihydropteroate synthase. Dihydrofolate reductase inhibitors have been used as antimalarials over the past few decades, often in combination with dihydropteroate synthase inhibitors. Resistance to these antifolate drugs developed through mutations in both target enzymes. However, limited mutation possibilities gave opportunities for the development of new drugs. Furthermore, other enzymes in the folate and related pathways are potential new targets that remain to be exploited. These include thymidylate synthase, an enzyme fused with dihydrofolate reductase in the same protein chain, serine hydroxymethyltransferase, methylene tetrahydrofolate dehydrogenase, methionine synthase and enzymes in the glycine cleavage pathway.


Journal of Medicinal Chemistry | 2015

Inhibitors of Plasmodial Serine Hydroxymethyltransferase (SHMT): Cocrystal Structures of Pyrazolopyrans with Potent Blood- and Liver-Stage Activities.

Matthias Witschel; Matthias Rottmann; Anatol Schwab; Ubolsree Leartsakulpanich; Penchit Chitnumsub; Michael Seet; Sandro Tonazzi; Geoffrey Schwertz; Frank Stelzer; Thomas Mietzner; Case W. McNamara; Frank Thater; Céline Freymond; Aritsara Jaruwat; Chatchadaporn Pinthong; Pinpunya Riangrungroj; Mouhssin Oufir; Matthias Hamburger; Pascal Mäser; Laura María Sanz-Alonso; Susan A. Charman; Sergio Wittlin; Yongyuth Yuthavong; Pimchai Chaiyen; François Diederich

Several of the enzymes related to the folate cycle are well-known for their role as clinically validated antimalarial targets. Nevertheless for serine hydroxymethyltransferase (SHMT), one of the key enzymes of this cycle, efficient inhibitors have not been described so far. On the basis of plant SHMT inhibitors from an herbicide optimization program, highly potent inhibitors of Plasmodium falciparum (Pf) and Plasmodium vivax (Pv) SHMT with a pyrazolopyran core structure were identified. Cocrystal structures of potent inhibitors with PvSHMT were solved at 2.6 Å resolution. These ligands showed activity (IC50/EC50 values) in the nanomolar range against purified PfSHMT, blood-stage Pf, and liver-stage P. berghei (Pb) cells and a high selectivity when assayed against mammalian cell lines. Pharmacokinetic limitations are the most plausible explanation for lack of significant activity of the inhibitors in the in vivo Pb mouse malaria model.


Antimicrobial Agents and Chemotherapy | 2012

Combined Spatial Limitation around Residues 16 and 108 of Plasmodium falciparum Dihydrofolate Reductase Explains Resistance to Cycloguanil.

Jarunee Vanichtanankul; Chayasith Uttamapinant; Penchit Chitnumsub; Tirayut Vilaivan; Yongyuth Yuthavong; Sumalee Kamchonwongpaisan

ABSTRACT Natural mutations of Plasmodium falciparum dihydrofolate reductase (PfDHFR) at A16V and S108T specifically confer resistance to cycloguanil (CYC) but not to pyrimethamine (PYR). In order to understand the nature of CYC resistance, the effects of various mutations at A16 on substrate and inhibitor binding were examined. Three series of mutations at A16 with or without the S108T/N mutation were generated. Only three mutants with small side chains at residue 16 (G, C, and S) were viable from bacterial complementation assay in the S108 series, whereas these three and an additional four mutants (T, V, M, and I) with slightly larger side chains were viable with simultaneous S108T mutation. Among these combinations, the A16V+S108T mutant was the most CYC resistant, and all of the S108T series ranged from being highly to moderately sensitive to PYR. In the S108N series, a strict requirement for alanine was observed at position 16. Crystal structure analyses reveal that in PfDHFR-TS variant T9/94 (A16V+S108T) complexed with CYC, the ligand has substantial steric conflicts with the side chains of both A16V and S108T, whereas in the complex with PYR, the ligand only showed mild conflict with S108T. CYC analogs designed to avoid such conflicts improved the binding affinity of the mutant enzymes. These results show that there is greater spatial limitation around the S108T/N residue when combined with the limitation imposed by A16V. The limitation of mutation of this series provides opportunities for drug design and development against antifolate-resistant malaria.


ACS Chemical Biology | 2011

Trypanosomal dihydrofolate reductase reveals natural antifolate resistance

Jarunee Vanichtanankul; Jirundon Yuvaniyama; Tirayut Vilaivan; Penchit Chitnumsub; Sumalee Kamchonwongpaisan; Yongyuth Yuthavong

Dihydrofolate reductase (DHFR) is a potential drug target for Trypanosoma brucei, a human parasite, which is the causative agent for African sleeping sickness. No drug is available against this target, since none of the classical antifolates such as pyrimethamine (PYR), cycloguanil, or trimethoprim are effective as selective inhibitors of T. brucei DHFR (TbDHFR). In order to design effective drugs that target TbDHFR, co-crystal structures with bound antifolates were studied. On comparison with malarial Plasmodium falciparum DHFR (PfDHFR), the co-crystal structures of wild-type TbDHFR reveal greater structural similarities to a mutant PfDHFR causing antifolate resistance than the wild-type enzyme. TbDHFR imposes steric hindrance for rigid inhibitors like PYR around Thr86, which is equivalent to Ser108Asn of the malarial enzymes. In addition, a missing residue on TbDHFR active-site loop together with the presence of Ile51 widens its active site even further than the structural effect of Asn51Ile, which is observed in PfDHFR structures. The structural similarities are paralleled by the similarly poor affinities of the trypanosomal enzyme for rigid inhibitors. Mutations of TbDHFR at Thr86 resulted in 10-fold enhancement or 7-fold reduction in the rigid inhibitors affinities for Thr86Ser or Thr86Asn, respectively. The co-crystal structure of TbDHFR with a flexible antifolate WR99210 suggests that its greater affinity result from its ability to avoid such Thr86 clash and occupy the widened binding space similarly to what is observed in the PfDHFR structures. Natural resistance to antifolates of TbDHFR can therefore be explained, and potential antifolate chemotherapy of trypanosomiasis should be possible taking this into account.


Antimicrobial Agents and Chemotherapy | 2010

Preclinical Evaluation of the Antifolate QN254, 5-Chloro- N′6′-(2,5-Dimethoxy-Benzyl)-Quinazoline-2,4,6-Triamine, as an Antimalarial Drug Candidate

Alexis Nzila; Matthias Rottmann; Penchit Chitnumsub; Stevens M. Kiara; Sumalee Kamchonwongpaisan; Cherdsak Maneeruttanarungroj; Bryan K. S. Yeung; Anne Goh; Suresh B. Lakshminarayana; Bin Zou; Josephine Wong; Ngai Ling Ma; Margaret Weaver; Thomas H. Keller; Véronique Dartois; Sergio Wittlin; Reto Brun; Yongyuth Yuthavong; Thierry T. Diagana

ABSTRACT Drug resistance against dihydrofolate reductase (DHFR) inhibitors—such as pyrimethamine (PM)—has now spread to almost all regions where malaria is endemic, rendering antifolate-based malaria treatments highly ineffective. We have previously shown that the di-amino quinazoline QN254 [5-chloro-N′6′-(2,5-dimethoxy-benzyl)-quinazoline-2,4,6-triamine] is active against the highly PM-resistant Plasmodium falciparum V1S strain, suggesting that QN254 could be used to treat malaria in regions with a high prevalence of antifolate resistance. Here, we further demonstrate that QN254 is highly active against Plasmodium falciparum clinical isolates, displaying various levels of antifolate drug resistance, and we provide biochemical and structural evidence that QN254 binds and inhibits the function of both the wild-type and the quadruple-mutant (V1S) forms of the DHFR enzyme. In addition, we have assessed QN254 oral bioavailability, efficacy, and safety in vivo. The compound displays favorable pharmacokinetic properties after oral administration in rodents. The drug was remarkably efficacious against Plasmodium berghei and could fully cure infected mice with three daily oral doses of 30 mg/kg. In the course of these efficacy studies, we have uncovered some dose limiting toxicity at higher doses that was confirmed in rats. Thus, despite its relative in vitro selectivity toward the Plasmodium DHFR enzyme, QN254 does not show the adequate therapeutic index to justify its further development as a single agent.


Acta Crystallographica Section D-biological Crystallography | 2014

The structure of Plasmodium falciparum serine hydroxymethyltransferase reveals a novel redox switch that regulates its activities

Penchit Chitnumsub; Wanwipa Ittarat; Aritsara Jaruwat; Krittikar Noytanom; Watcharee Amornwatcharapong; Wichai Pornthanakasem; Pimchai Chaiyen; Yongyuth Yuthavong; Ubolsree Leartsakulpanich

The crystal structure of P. falciparum SHMT revealed snapshots of an intriguing disulfide/sulfhydryl switch controlling the functional activity.

Collaboration


Dive into the Penchit Chitnumsub's collaboration.

Top Co-Authors

Avatar

Yongyuth Yuthavong

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar

Ubolsree Leartsakulpanich

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar

Sumalee Kamchonwongpaisan

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aritsara Jaruwat

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar

Jarunee Vanichtanankul

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wanwipa Ittarat

Thailand National Science and Technology Development Agency

View shared research outputs
Top Co-Authors

Avatar

Bongkoch Tarnchompoo

Thailand National Science and Technology Development Agency

View shared research outputs
Researchain Logo
Decentralizing Knowledge