Network


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

Hotspot


Dive into the research topics where Aaron Nilsen is active.

Publication


Featured researches published by Aaron Nilsen.


Science Translational Medicine | 2013

Quinolone-3-Diarylethers: A New Class of Antimalarial Drug

Aaron Nilsen; Alexis N. LaCrue; Karen L. White; Isaac P. Forquer; R. Matthew Cross; Jutta Marfurt; Michael W. Mather; Michael J. Delves; David M. Shackleford; Fabián E. Sáenz; Joanne M. Morrisey; Jessica Steuten; Tina Mutka; Yuexin Li; Grennady Wirjanata; Eileen Ryan; Sandra Duffy; Jane Xu Kelly; Boni F. Sebayang; Anne-Marie Zeeman; Rintis Noviyanti; Robert E. Sinden; Clemens H. M. Kocken; Ric N. Price; Vicky M. Avery; Iñigo Angulo-Barturen; María Belén Jiménez-Díaz; Santiago Ferrer; Esperanza Herreros; Laura Sanz

ELQ-300, an investigational drug for treating and preventing malaria, shows potent transmission-blocking activity in rodent models of malaria. Taking the Bite Out of Malaria Malaria is spread from person to person by mosquitoes that inject 8 to 10 sporozoite forms of the parasite in a single bite. The sporozoites reproduce in the liver to produce 10,000 to 30,000 merozoites before the liver schizont ruptures and parasites flood into the bloodstream where the absolute parasite burden may increase to a thousand billion (1012) circulating parasites. Some of these parasites develop into gametocytes that may be ingested by another mosquito where they progress through ookinete, oocyst, and sporozoite stages to complete the cycle. Like quinine, most antimalarial drugs in use today target only the symptomatic blood stage. The efficacy of these drugs has been compromised by resistance, and so there is a pressing need for new drugs that target multiple stages of the parasite life cycle for use in malaria treatment and prevention. Clearly, it is advantageous to strike at the liver stage where parasite numbers are low, to diminish the likelihood of selecting for a resistant mutant and before the infection has a chance to weaken the defenses of the human host. In a new study, Nilsen and colleagues describe ELQ-300, a 4(1H)-quinolone-3-diarylether, which targets the liver and blood stages, including the forms that are crucial to disease transmission (gametocytes, zygotes, and ookinetes). In mouse models of malaria, a single oral dose of 0.03 mg/kg prevented sporozoite-induced infections, whereas four daily doses of 1 mg/kg achieved complete cures of patent infections. ELQ-300 is a preclinical candidate that may be coformulated with other antimalarials to prevent and treat malaria, with the potential to aid in eradication of the disease. The goal for developing new antimalarial drugs is to find a molecule that can target multiple stages of the parasite’s life cycle, thus impacting prevention, treatment, and transmission of the disease. The 4(1H)-quinolone-3-diarylethers are selective potent inhibitors of the parasite’s mitochondrial cytochrome bc1 complex. These compounds are highly active against the human malaria parasites Plasmodium falciparum and Plasmodium vivax. They target both the liver and blood stages of the parasite as well as the forms that are crucial for disease transmission, that is, the gametocytes, the zygote, the ookinete, and the oocyst. Selected as a preclinical candidate, ELQ-300 has good oral bioavailability at efficacious doses in mice, is metabolically stable, and is highly active in blocking transmission in rodent models of malaria. Given its predicted low dose in patients and its predicted long half-life, ELQ-300 has potential as a new drug for the treatment, prevention, and, ultimately, eradication of human malaria.


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

Endochin-like quinolones are highly efficacious against acute and latent experimental toxoplasmosis

J. Stone Doggett; Aaron Nilsen; Isaac P. Forquer; Keith W. Wegmann; Lorraine Jones-Brando; Robert H. Yolken; Claudia Bordón; Susan A. Charman; Kasiram Katneni; Tracey L. Schultz; Jeremy N. Burrows; David J. Hinrichs; Brigitte Meunier; Vern B. Carruthers; Michael K. Riscoe

Toxoplasma gondii is a widely distributed protozoan pathogen that causes devastating ocular and central nervous system disease. We show that the endochin-like quinolone (ELQ) class of compounds contains extremely potent inhibitors of T. gondii growth in vitro and is effective against acute and latent toxoplasmosis in mice. We screened 50 ELQs against T. gondii and selected two lead compounds, ELQ-271 and ELQ-316, for evaluation. ELQ-271 and ELQ-316, have in vitro IC50 values of 0.1 nM and 0.007 nM, respectively. ELQ-271 and ELQ-316 have ED50 values of 0.14 mg/kg and 0.08 mg/kg when administered orally to mice with acute toxoplasmosis. Moreover, ELQ-271 and ELQ-316 are highly active against the cyst form of T. gondii in mice at low doses, reducing cyst burden by 76–88% after 16 d of treatment. To investigate the ELQ mechanism of action against T. gondii, we demonstrate that endochin and ELQ-271 inhibit cytochrome c reduction by the T. gondii cytochrome bc1 complex at 8 nM and 31 nM, respectively. We also show that ELQ-271 inhibits the Saccharomyces cerevisiae cytochrome bc1 complex, and an M221Q amino acid substitution in the Qi site of the protein leads to >100-fold resistance. We conclude that ELQ-271 and ELQ-316 are orally bioavailable drugs that are effective against acute and latent toxoplasmosis, likely acting as inhibitors of the Qi site of the T. gondii cytochrome bc1 complex.


Journal of Medicinal Chemistry | 2014

Discovery, synthesis, and optimization of antimalarial 4(1H)-quinolone-3-diarylethers

Aaron Nilsen; Galen P. Miley; Isaac P. Forquer; Michael W. Mather; Kasiram Katneni; Yuexin Li; Sovitj Pou; April M. Pershing; Allison M. Stickles; Eileen Ryan; Jane X. Kelly; J. Stone Doggett; Karen L. White; David J. Hinrichs; Rolf Walter Winter; Susan A. Charman; Lev N. Zakharov; Ian Bathurst; Jeremy N. Burrows; Akhil B. Vaidya; Michael K. Riscoe

The historical antimalarial compound endochin served as a structural lead for optimization. Endochin-like quinolones (ELQ) were prepared by a novel chemical route and assessed for in vitro activity against multidrug resistant strains of Plasmodium falciparum and against malaria infections in mice. Here we describe the pathway to discovery of a potent class of orally active antimalarial 4(1H)-quinolone-3-diarylethers. The initial prototype, ELQ-233, exhibited low nanomolar IC50 values against all tested strains including clinical isolates harboring resistance to atovaquone. ELQ-271 represented the next critical step in the iterative optimization process, as it was stable to metabolism and highly effective in vivo. Continued analoging revealed that the substitution pattern on the benzenoid ring of the quinolone core significantly influenced reactivity with the host enzyme. This finding led to the rational design of highly selective ELQs with outstanding oral efficacy against murine malaria that is superior to established antimalarials chloroquine and atovaquone.


ChemBioChem | 2009

Iodothyronamines are Oxidatively Deaminated to Iodothyroacetic Acids in vivo

Warren J. L. Wood; Travis Geraci; Aaron Nilsen; Andrea E. DeBarber; Thomas S. Scanlan

3‐Iodothyronamine (T1AM) and 3,3′,5‐triiodothyroacetic acid (Triac) are bioactive metabolites of the hormone thyroxine (T4). In the present study, the ability of T1AM and 3,3′,5‐triiodothyronamine (T3AM) to be metabolized to 3‐iodothyroacetic acid (TA1) and Triac, respectively, was investigated. Both T1AM and T3AM were converted to their respective iodinated thyroacetic acid analogues in both cell and tissue extracts. This conversion could be significantly inhibited with the monamine oxidase (MAO) and semicarbazide‐sensitive amine oxidase (SSAO) inhibitor iproniazid. TA1 was found to be present in trace quantities in human serum and in substantial levels in serum from T1AM‐treated rats. These results demonstrate that iodothyronamines are substrates for amine oxidases and that this metabolism may be the source of the corresponding endogenous arylacetic acid products Triac and TA1.


PLOS Pathogens | 2015

Characterization of a Novel Human-Specific STING Agonist that Elicits Antiviral Activity Against Emerging Alphaviruses

Tina Sali; Kara Pryke; Jinu Abraham; Andrew Liu; Iris Archer; Rebecca Broeckel; Julia A. Staverosky; Jessica L. Smith; Ahmed Majeed Al-Shammari; Lisi Amsler; Kayla Sheridan; Aaron Nilsen; Daniel N. Streblow; Victor R. DeFilippis

Pharmacologic stimulation of innate immune processes represents an attractive strategy to achieve multiple therapeutic outcomes including inhibition of virus replication, boosting antitumor immunity, and enhancing vaccine immunogenicity. In light of this we sought to identify small molecules capable of activating the type I interferon (IFN) response by way of the transcription factor IFN regulatory factor 3 (IRF3). A high throughput in vitro screen yielded 4-(2-chloro-6-fluorobenzyl)-N-(furan-2-ylmethyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]thiazine-6-carboxamide (referred to herein as G10), which was found to trigger IRF3/IFN-associated transcription in human fibroblasts. Further examination of the cellular response to this molecule revealed expression of multiple IRF3-dependent antiviral effector genes as well as type I and III IFN subtypes. This led to the establishment of a cellular state that prevented replication of emerging Alphavirus species including Chikungunya virus, Venezuelan Equine Encephalitis virus, and Sindbis virus. To define cellular proteins essential to elicitation of the antiviral activity by the compound we employed a reverse genetics approach that utilized genome editing via CRISPR/Cas9 technology. This allowed the identification of IRF3, the IRF3-activating adaptor molecule STING, and the IFN-associated transcription factor STAT1 as required for observed gene induction and antiviral effects. Biochemical analysis indicates that G10 does not bind to STING directly, however. Thus the compound may represent the first synthetic small molecule characterized as an indirect activator of human STING-dependent phenotypes. In vivo stimulation of STING-dependent activity by an unrelated small molecule in a mouse model of Chikungunya virus infection blocked viremia demonstrating that pharmacologic activation of this signaling pathway may represent a feasible strategy for combating emerging Alphaviruses.


Antimicrobial Agents and Chemotherapy | 2015

ELQ-300 Prodrugs for Enhanced Delivery and Single-Dose Cure of Malaria

Galen P. Miley; Sovitj Pou; Rolf W. Winter; Aaron Nilsen; Yuexin Li; Jane Xu Kelly; Allison M. Stickles; Michael W. Mather; Isaac P. Forquer; April M. Pershing; Karen L. White; David M. Shackleford; Jessica Saunders; Gong Chen; Li Min Ting; Kami Kim; Lev N. Zakharov; Cristina Donini; Jeremy N. Burrows; Akhil B. Vaidya; Susan A. Charman; Michael K. Riscoe

ABSTRACT ELQ-300 is a preclinical candidate that targets the liver and blood stages of Plasmodium falciparum, as well as the forms that are crucial to transmission of disease: gametocytes, zygotes, and ookinetes. A significant obstacle to the clinical development of ELQ-300 is related to its physicochemical properties. Its relatively poor aqueous solubility and high crystallinity limit absorption to the degree that only low blood concentrations can be achieved following oral dosing. While these low blood concentrations are sufficient for therapy, the levels are too low to establish an acceptable safety margin required by regulatory agencies for clinical development. One way to address the challenging physicochemical properties of ELQ-300 is through the development of prodrugs. Here, we profile ELQ-337, a bioreversible O-linked carbonate ester prodrug of the parent molecule. At the molar equivalent dose of 3 mg/kg of body weight, the delivery of ELQ-300 from ELQ-337 is enhanced by 3- to 4-fold, reaching a maximum concentration of drug in serum (Cmax) of 5.9 μM by 6 h after oral administration, and unlike ELQ-300 at any dose, ELQ-337 provides single-dose cures of patent malaria infections in mice at low-single-digit milligram per kilogram doses. Our findings show that the prodrug strategy represents a viable approach to overcome the physicochemical limitations of ELQ-300 to deliver the active drug to the bloodstream at concentrations sufficient for safety and toxicology studies, as well as achieving single-dose cures.


Antimicrobial Agents and Chemotherapy | 2015

Subtle Changes in Endochin-Like Quinolone Structure Alter the Site of Inhibition within the Cytochrome bc1 Complex of Plasmodium falciparum

Allison M. Stickles; Mariana Justino de Almeida; Joanne M. Morrisey; Kayla Sheridan; Isaac P. Forquer; Aaron Nilsen; Rolf W. Winter; Jeremy N. Burrows; David A. Fidock; Akhil B. Vaidya; Michael K. Riscoe

ABSTRACT The cytochrome bc1 complex (cyt bc1) is the third component of the mitochondrial electron transport chain and is the target of several potent antimalarial compounds, including the naphthoquinone atovaquone (ATV) and the 4(1H)-quinolone ELQ-300. Mechanistically, cyt bc1 facilitates the transfer of electrons from ubiquinol to cytochrome c and contains both oxidative (Qo) and reductive (Qi) catalytic sites that are amenable to small-molecule inhibition. Although many antimalarial compounds, including ATV, effectively target the Qo site, it has been challenging to design selective Qi site inhibitors with the ability to circumvent clinical ATV resistance, and little is known about how chemical structure contributes to site selectivity within cyt bc1. Here, we used the proposed Qi site inhibitor ELQ-300 to generate a drug-resistant Plasmodium falciparum clone containing an I22L mutation at the Qi region of cyt b. Using this D1 clone and the Y268S Qo mutant strain, P. falciparum Tm90-C2B, we created a structure-activity map of Qi versus Qo site selectivity for a series of endochin-like 4(1H)-quinolones (ELQs). We found that Qi site inhibition was associated with compounds containing 6-position halogens or aryl 3-position side chains, while Qo site inhibition was favored by 5,7-dihalogen groups or 7-position substituents. In addition to identifying ELQ-300 as a preferential Qi site inhibitor, our data suggest that the 4(1H)-quinolone scaffold is compatible with binding to either site of cyt bc1 and that minor chemical changes can influence Qo or Qi site inhibition by the ELQs.


American Journal of Tropical Medicine and Hygiene | 2015

Inhibition of Cytochrome bc1 as a Strategy for Single-Dose, Multi-Stage Antimalarial Therapy

Allison M. Stickles; Li Min Ting; Joanne M. Morrisey; Yuexin Li; Michael W. Mather; Erin W. Meermeier; April M. Pershing; Isaac P. Forquer; Galen P. Miley; Sovitj Pou; Rolf W. Winter; David J. Hinrichs; Jane Xu Kelly; Kami Kim; Akhil B. Vaidya; Michael K. Riscoe; Aaron Nilsen

Single-dose therapies for malaria have been proposed as a way to reduce the cost and increase the effectiveness of antimalarial treatment. However, no compound to date has shown single-dose activity against both the blood-stage Plasmodium parasites that cause disease and the liver-stage parasites that initiate malaria infection. Here, we describe a subset of cytochrome bc1 (cyt bc1) inhibitors, including the novel 4(1H)-quinolone ELQ-400, with single-dose activity against liver, blood, and transmission-stage parasites in mouse models of malaria. Although cyt bc1 inhibitors are generally classified as slow-onset antimalarials, we found that a single dose of ELQ-400 rapidly induced stasis in blood-stage parasites, which was associated with a rapid reduction in parasitemia in vivo. ELQ-400 also exhibited a low propensity for drug resistance and was active against atovaquone-resistant P. falciparum strains with point mutations in cyt bc1. Ultimately, ELQ-400 shows that cyt bc1 inhibitors can function as single-dose, blood-stage antimalarials and is the first compound to provide combined treatment, prophylaxis, and transmission blocking activity for malaria after a single oral administration. This remarkable multi-stage efficacy suggests that metabolic therapies, including cyt bc1 inhibitors, may be valuable additions to the collection of single-dose antimalarials in current development.


Journal of Experimental Medicine | 2016

Radical cure of experimental babesiosis in immunodeficient mice using a combination of an endochin-like quinolone and atovaquone

Lauren Lawres; Aprajita Garg; Vidya P. Kumar; Igor Bruzual; Isaac P. Forquer; Isaline Renard; Azan Z. Virji; Pierre Boulard; Eduardo X. Rodriguez; Alexander J. Allen; Sovitj Pou; Keith W. Wegmann; Rolf W. Winter; Aaron Nilsen; Jialing Mao; Douglas A. Preston; Alexia A. Belperron; Linda K. Bockenstedt; David J. Hinrichs; Michael K. Riscoe; J. Stone Doggett; Choukri Ben Mamoun

Human babesiosis is a tick-borne multisystem disease, and current treatments have both adverse side effects and a significant rate of drug failure. Lawres et al. report that endochin-like quinolones, in combination with atovaquone, cure experimental babesiosis in immunodeficient mice.


Antimicrobial Agents and Chemotherapy | 2012

Sontochin as a Guide to the Development of Drugs against Chloroquine-Resistant Malaria

Sovitj Pou; Rolf W. Winter; Aaron Nilsen; Jane Xu Kelly; Yuexin Li; J. Stone Doggett; Erin W. Riscoe; Keith W. Wegmann; David J. Hinrichs; Michael K. Riscoe

ABSTRACT Sontochin was the original chloroquine replacement drug, arising from research by Hans Andersag 2 years after chloroquine (known as “resochin” at the time) had been shelved due to the mistaken perception that it was too toxic for human use. We were surprised to find that sontochin, i.e., 3-methyl-chloroquine, retains significant activity against chloroquine-resistant strains of Plasmodium falciparum in vitro. We prepared derivatives of sontochin, “pharmachins,” with alkyl or aryl substituents at the 3 position and with alterations to the 4-position side chain to enhance activity against drug-resistant strains. Modified with an aryl substituent in the 3 position of the 7-chloro-quinoline ring, Pharmachin 203 (PH-203) exhibits low-nanomolar 50% inhibitory concentrations (IC50s) against drug-sensitive and multidrug-resistant strains and in vivo efficacy against patent infections of Plasmodium yoelii in mice that is superior to chloroquine. Our findings suggest that novel 3-position aryl pharmachin derivatives have the potential for use in treating drug resistant malaria.

Collaboration


Dive into the Aaron Nilsen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sovitj Pou

Portland VA Medical Center

View shared research outputs
Top Co-Authors

Avatar

Isaac P. Forquer

Portland VA Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yuexin Li

Portland VA Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rolf W. Winter

Portland State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jane Xu Kelly

Portland State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge