Network


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

Hotspot


Dive into the research topics where Margaret A. Phillips is active.

Publication


Featured researches published by Margaret A. Phillips.


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 Medicinal Chemistry | 2011

Structure-guided lead optimization of triazolopyrimidine-ring substituents identifies potent Plasmodium falciparum dihydroorotate dehydrogenase inhibitors with clinical candidate potential

José M. Coterón; Maria Marco; Jorge Esquivias; Xiaoyi Deng; Karen L. White; John White; Maria Koltun; Farah El Mazouni; Sreekanth Kokkonda; Kasiram Katneni; Ravi K. Bhamidipati; David M. Shackleford; Iñigo Angulo-Barturen; Santiago Ferrer; María Belén Jiménez-Díaz; Francisco Javier Gamo; Elizabeth J. Goldsmith; William N. Charman; Ian Bathurst; David M. Floyd; David Matthews; Jeremy N. Burrows; Pradipsinh K. Rathod; Susan A. Charman; Margaret A. Phillips

Drug therapy is the mainstay of antimalarial therapy, yet current drugs are threatened by the development of resistance. In an effort to identify new potential antimalarials, we have undertaken a lead optimization program around our previously identified triazolopyrimidine-based series of Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) inhibitors. The X-ray structure of PfDHODH was used to inform the medicinal chemistry program allowing the identification of a potent and selective inhibitor (DSM265) that acts through DHODH inhibition to kill both sensitive and drug resistant strains of the parasite. This compound has similar potency to chloroquine in the humanized SCID mouse P. falciparum model, can be synthesized by a simple route, and rodent pharmacokinetic studies demonstrated it has excellent oral bioavailability, a long half-life and low clearance. These studies have identified the first candidate in the triazolopyrimidine series to meet previously established progression criteria for efficacy and ADME properties, justifying further development of this compound toward clinical candidate status.


Journal of Medicinal Chemistry | 2008

Triazolopyrimidine-based dihydroorotate dehydrogenase inhibitors with potent and selective activity against the malaria parasite, Plasmodium falciparum

Margaret A. Phillips; Ramesh Gujjar; Nicholas A. Malmquist; John White; Farah El Mazouni; Jeffrey Baldwin; Pradipsinh K. Rathod

A Plasmodium falciparum dihydroorotate dehydrogenase ( PfDHODH) inhibitor that is potent ( KI = 15 nM) and species-selective (>5000-fold over the human enzyme) was identified by high-throughput screening. The substituted triazolopyrimidine and its structural analogues were produced by an inexpensive three-step synthesis, and the series showed good association between PfDHODH inhibition and parasite toxicity. This study has identified the first nanomolar PfDHODH inhibitor with potent antimalarial activity in whole cells (EC50 = 79 nM).


Journal of Medicinal Chemistry | 2009

Identification of a metabolically stable triazolopyrimidine-based dihydroorotate dehydrogenase inhibitor with antimalarial activity in mice.

Ramesh Gujjar; Alka Marwaha; Farah El Mazouni; John Kenneth White; Karen L. White; Sharon A. Creason; David M. Shackleford; Jeffrey Baldwin; William N. Charman; Frederick S. Buckner; Susan A. Charman; Pradip Rathod; Margaret A. Phillips

Plasmodium falciparum causes 1-2 million deaths annually. Yet current drug therapies are compromised by resistance. We previously described potent and selective triazolopyrimidine-based inhibitors of P. falciparum dihydroorotate dehydrogenase (PfDHODH) that inhibited parasite growth in vitro; however, they showed no activity in vivo. Here we show that lack of efficacy against P. berghei in mice resulted from a combination of poor plasma exposure and reduced potency against P. berghei DHODH. For compounds containing naphthyl (DSM1) or anthracenyl (DSM2), plasma exposure was reduced upon repeated dosing. Phenyl-substituted triazolopyrimidines were synthesized leading to identification of analogs with low predicted metabolism in human liver microsomes and which showed prolonged exposure in mice. Compound 21 (DSM74), containing p-trifluoromethylphenyl, suppressed growth of P. berghei in mice after oral administration. This study provides the first proof of concept that DHODH inhibitors can suppress Plasmodium growth in vivo, validating DHODH as a new target for antimalarial chemotherapy.


Science Translational Medicine | 2015

A long-duration dihydroorotate dehydrogenase inhibitor (DSM265) for prevention and treatment of malaria

Margaret A. Phillips; Julie Lotharius; Kennan Marsh; John White; Anthony Dayan; Karen L. White; Jacqueline W. Njoroge; Farah El Mazouni; Yanbin Lao; Sreekanth Kokkonda; Diana R. Tomchick; Xiaoyi Deng; Trevor Laird; Sangeeta N. Bhatia; Sandra March; Caroline L. Ng; David A. Fidock; Sergio Wittlin; Maria J. Lafuente-Monasterio; Francisco Javier Gamo–Benito; Laura Maria Sanz Alonso; María Santos Martínez; María Belén Jiménez-Díaz; Santiago Ferrer Bazaga; Iñigo Angulo-Barturen; John N. Haselden; James Louttit; Yi Cui; Arun Sridhar; Anna Marie Zeeman

The antimalarial drug DSM265 displays activity against blood and liver stages of Plasmodium falciparum and has a long predicted half-life in humans. Long-acting new treatment for drug-resistant malaria Malaria kills 0.6 million people annually, yet current malaria drugs are no longer fully effective because the parasite that causes malaria is becoming resistant to these agents. Phillips et al. have identified a new drug that kills both drug-sensitive and drug-resistant malaria parasites by targeting the ability of the parasite to synthesize the nucleotide precursors required for synthesis of DNA and RNA. This drug kills parasites in both the blood and liver and is sufficiently long-acting that it is expected to cure malaria after a single dose or to be effective if dosed weekly for chemoprevention. Malaria is one of the most significant causes of childhood mortality, but disease control efforts are threatened by resistance of the Plasmodium parasite to current therapies. Continued progress in combating malaria requires development of new, easy to administer drug combinations with broad-ranging activity against all manifestations of the disease. DSM265, a triazolopyrimidine-based inhibitor of the pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase (DHODH), is the first DHODH inhibitor to reach clinical development for treatment of malaria. We describe studies profiling the biological activity, pharmacological and pharmacokinetic properties, and safety of DSM265, which supported its advancement to human trials. DSM265 is highly selective toward DHODH of the malaria parasite Plasmodium, efficacious against both blood and liver stages of P. falciparum, and active against drug-resistant parasite isolates. Favorable pharmacokinetic properties of DSM265 are predicted to provide therapeutic concentrations for more than 8 days after a single oral dose in the range of 200 to 400 mg. DSM265 was well tolerated in repeat-dose and cardiovascular safety studies in mice and dogs, was not mutagenic, and was inactive against panels of human enzymes/receptors. The excellent safety profile, blood- and liver-stage activity, and predicted long half-life in humans position DSM265 as a new potential drug combination partner for either single-dose treatment or once-weekly chemoprevention. DSM265 has advantages over current treatment options that are dosed daily or are inactive against the parasite liver stage.


Journal of Biological Chemistry | 2009

An Alternative Polyamine Biosynthetic Pathway Is Widespread in Bacteria and Essential for Biofilm Formation in Vibrio cholerae

Jeongmi Lee; Vanessa Sperandio; Douglas E. Frantz; Jamie Longgood; Andrew Camilli; Margaret A. Phillips; Anthony J. Michael

Polyamines are small organic cations found in all cells, and the biosynthetic pathway is well described in eukaryotes and Escherichia coli. The characterized pathway uses decarboxylated S-adenosylmethionine as the aminopropyl group donor to form spermidine from putrescine by the key enzymes S-adenosylmethionine decarboxylase and spermidine synthase. We report here the in vivo characterization of an alternative polyamine biosynthetic pathway from Vibrio cholerae, the causative agent of human cholera. The pathway uses aspartate β-semialdehyde as the aminopropyl group donor and consists of a fused protein containing l-2,4-diaminobutyrate aminotransferase and l-2,4-diaminobutyrate decarboxylase, a carboxynorspermidine dehydrogenase (CANSDH), and a carboxynorspermidine decarboxylase (CANSDC). We show that in V. cholerae, this pathway is required for synthesis of both sym-norspermidine and spermidine. Heterologous expression of the V. cholerae pathway in E. coli results in accumulation of the nonnative polyamines diaminopropane and sym-norspermidine. Genetic deletion of the V. cholerae CANSDC led to accumulation of carboxynorspermidine, whereas deletion of either CANSDC or the putative CANSDH led to loss of sym-norspermidine and spermidine. These results allowed unambiguous identification of the gene encoding CANSDH. Furthermore, deletion of either CANSDH or CANSDC led to a 50–60% reduction in growth rate of planktonic cells and severely reduced biofilm formation, which could be rescued by exogenously supplied sym-norspermidine but not spermidine. The pathway was not required for infectivity in a mouse model of V. cholerae infection. Notably, the alternative polyamine biosynthetic pathway is widespread in bacteria and is likely to play a previously unrecognized role in the biology of these organisms.


Infectious disorders drug targets | 2010

Plasmodium dihydroorotate dehydrogenase: a promising target for novel anti-malarial chemotherapy

Margaret A. Phillips; Pradipsinh K. Rathod

Malaria remains a globally prevalent infectious disease that leads to significant morbidity and mortality. While there are a number of drugs approved for its treatment, drug resistance has compromised most of them, making the development of new drugs for the treatment and prevention of malaria essential. The completion of the Plasmodium falciparum genome and a growing understanding of parasite biology are fueling the search for novel drug targets. Despite this, few targets have been chemically validated in vivo. The pyrimidine biosynthetic pathway illustrates one of the best examples of successful identification of anti-malarial drug targets. This review focuses on recent studies to exploit the fourth enzyme in the de novo pyrimidine biosynthetic pathway of P. falciparum, dihydroorotate dehydrogenase (PfDHODH), as a new target for drug discovery. Several chemical scaffolds have been identified by high throughput screening as potent inhibitors of PfDHODH and these show strong selectivity for the malarial enzyme over that from the human host. Potent activity against parasites in whole cell models with good correlation between activity on the enzyme and the parasite have also been observed for a number of the identified series. Lead optimization of a triazolopyrimidine-based series has identified an analog with prolonged plasma exposure, that is orally bioavailable, and which shows good efficacy against the in vivo mouse model of the disease. These data provide strong evidence that PfDHODH is a validated target for the identification of new antimalarial chemotherapy. The challenge remains to identify compounds with the necessary combination of potency and metabolic stability to allow identification of a clinical candidate.


Current Topics in Medicinal Chemistry | 2011

State of the art in African trypanosome drug discovery.

Robert Jacobs; Bakela Nare; Margaret A. Phillips

African sleeping sickness is endemic in sub-Saharan Africa where the WHO estimates that 60 million people are at risk for the disease. Human African trypanosomiasis (HAT) is 100% fatal if untreated and the current drug therapies have significant limitations due to toxicity and difficult treatment regimes. No new chemical agents have been approved since eflornithine in 1990. The pentamidine analog DB289, which was in late stage clinical trials for the treatment of early stage HAT recently failed due to toxicity issues. A new protocol for the treatment of late-stage T. brucei gambiense that uses combination nifurtomox/eflornithine (NECT) was recently shown to have better safety and efficacy than eflornithine alone, while being easier to administer. This breakthrough represents the only new therapy for HAT since the approval of eflornithine. A number of research programs are on going to exploit the unusual biochemical pathways in the parasite to identify new targets for target based drug discovery programs. HTS efforts are also underway to discover new chemical entities through whole organism screening approaches. A number of inhibitors with anti-trypanosomal activity have been identified by both approaches, but none of the programs are yet at the stage of identifying a preclinical candidate. This dire situation underscores the need for continued effort to identify new chemical agents for the treatment of HAT.


Journal of Medicinal Chemistry | 2011

Lead-optimization of aryl and aralkyl amine based triazolopyrimidine inhibitors of Plasmodium falciparum dihydroorotate dehydrogenase with antimalarial activity in mice

Ramesh Gujjar; Farah El Mazouni; Karen L. White; John Kenneth White; Sharon A. Creason; David M. Shackleford; Xiaoyi Deng; William N. Charman; Ian Bathurst; Jeremy N. Burrows; David M. Floyd; David Matthews; Frederick S. Buckner; Susan A. Charman; Margaret A. Phillips; Pradipsinh K. Rathod

Malaria is one of the leading causes of severe infectious disease worldwide; yet, our ability to maintain effective therapy to combat the illness is continually challenged by the emergence of drug resistance. We previously reported identification of a new class of triazolopyrimidine-based Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) inhibitors with antimalarial activity, leading to the discovery of a new lead series and novel target for drug development. Active compounds from the series contained a triazolopyrimidine ring attached to an aromatic group through a bridging nitrogen atom. Herein, we describe systematic efforts to optimize the aromatic functionality with the goal of improving potency and in vivo properties of compounds from the series. These studies led to the identification of two new substituted aniline moieties (4-SF(5)-Ph and 3,5-Di-F-4-CF(3)-Ph), which, when coupled to the triazolopyrimidine ring, showed good plasma exposure and better efficacy in the Plasmodium berghei mouse model of the disease than previously reported compounds from the series.


Journal of Biological Chemistry | 2010

Novel Inhibitors of Plasmodium falciparum Dihydroorotate Dehydrogenase with Anti-malarial Activity in the Mouse Model

Michael Booker; Cecilia M. Bastos; Martin Kramer; Robert Barker; Renato Skerlj; Amar Bir Singh Sidhu; Xiaoyi Deng; Cassandra Celatka; Joseph F. Cortese; Jose E. Guerrero Bravo; Keila N. Crespo Llado; Adelfa E. Serrano; Iñigo Angulo-Barturen; María Belén Jiménez-Díaz; Sara Viera; Helen Garuti; Sergio Wittlin; Petros Papastogiannidis; Jing-wen Lin; Chris J. Janse; Shahid M. Khan; Manoj T. Duraisingh; Bradley I. Coleman; Elizabeth J. Goldsmith; Margaret A. Phillips; Benito Munoz; Dyann F. Wirth; Jeffrey D. Klinger; Roger Wiegand; Edmund Sybertz

Plasmodium falciparum, the causative agent of the most deadly form of human malaria, is unable to salvage pyrimidines and must rely on de novo biosynthesis for survival. Dihydroorotate dehydrogenase (DHODH) catalyzes the rate-limiting step in the pyrimidine biosynthetic pathway and represents a potential target for anti-malarial therapy. A high throughput screen and subsequent medicinal chemistry program identified a series of N-alkyl-5-(1H-benzimidazol-1-yl)thiophene-2-carboxamides with low nanomolar in vitro potency against DHODH from P. falciparum, P. vivax, and P. berghei. The compounds were selective for the parasite enzymes over human DHODH, and x-ray structural data on the analog Genz-667348, demonstrated that species selectivity could be attributed to amino acid differences in the inhibitor-binding site. Compounds from this series demonstrated in vitro potency against the 3D7 and Dd2 strains of P. falciparum, good tolerability and oral exposure in the mouse, and ED50 values in the 4-day murine P. berghei efficacy model of 13–21 mg/kg/day with oral twice-daily dosing. In particular, treatment with Genz-667348 at 100 mg/kg/day resulted in sterile cure. Two recent analogs of Genz-667348 are currently undergoing pilot toxicity testing to determine suitability as clinical development candidates.

Collaboration


Dive into the Margaret A. Phillips's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Farah El Mazouni

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Nick V. Grishin

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiaoyi Deng

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth J. Goldsmith

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lisa N. Kinch

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

John White

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Jeffrey Baldwin

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erin K. Willert

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge