Network


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

Hotspot


Dive into the research topics where Rolf W. Winter is active.

Publication


Featured researches published by Rolf W. Winter.


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.


Nature | 2009

Discovery of dual function acridones as a new antimalarial chemotype

Jane Xu Kelly; Martin J. Smilkstein; Reto Brun; Sergio Wittlin; Roland A. Cooper; Kristin D. Lane; Aaron Janowsky; Robert A. Johnson; Rozalia A. Dodean; Rolf W. Winter; David J. Hinrichs; Michael K. Riscoe

Preventing and delaying the emergence of drug resistance is an essential goal of antimalarial drug development. Monotherapy and highly mutable drug targets have each facilitated resistance, and both are undesirable in effective long-term strategies against multi-drug-resistant malaria. Haem remains an immutable and vulnerable target, because it is not parasite-encoded and its detoxification during haemoglobin degradation, critical to parasite survival, can be subverted by drug–haem interaction as in the case of quinolines and many other drugs. Here we describe a new antimalarial chemotype that combines the haem-targeting character of acridones, together with a chemosensitizing component that counteracts resistance to quinoline antimalarial drugs. Beyond the essential intrinsic characteristics common to deserving candidate antimalarials (high potency in vitro against pan-sensitive and multi-drug-resistant Plasmodium falciparum, efficacy and safety in vivo after oral administration, inexpensive synthesis and favourable physicochemical properties), our initial lead, T3.5 (3-chloro-6-(2-diethylamino-ethoxy)-10-(2-diethylamino-ethyl)-acridone), demonstrates unique synergistic properties. In addition to ‘verapamil-like’ chemosensitization to chloroquine and amodiaquine against quinoline-resistant parasites, T3.5 also results in an apparently mechanistically distinct synergism with quinine and with piperaquine. This synergy, evident in both quinoline-sensitive and quinoline-resistant parasites, has been demonstrated both in vitro and in vivo. In summary, this innovative acridone design merges intrinsic potency and resistance-counteracting functions in one molecule, and represents a new strategy to expand, enhance and sustain effective antimalarial drug combinations.


Current Medicinal Chemistry | 2005

Xanthones as Antimalarial Agents: Discovery, Mode of Action, and Optimization

Michael K. Riscoe; Jane Xu Kelly; Rolf W. Winter

It is believed that at no time in the history of the human race malaria has been absent. This disease, which is caused by protozoa of the genus Plasmodium, in all likelihood has been responsible for the death of about half of all people who ever lived. Even today, after attempts at intervention on a worldwide scale, malaria remains the most significant parasitic disease in the tropics and sub-tropics, where it causes at least 500 million clinical episodes and claims 1.5 million lives each year, mostly young children and pregnant women. Widespread resistance to the best and least expensive antimalarials, chloroquine and S/P (i.e., a combination of sulfadoxine and pyrimethamine), combined with an increasing tolerance to insecticides in the mosquito vector, threaten a global malaria tragedy unless new countermeasures are developed. For malaria therapy, the great panacea would be the development of a long-lasting vaccine, but until this becomes a reality, people living in and traveling to endemic regions must rely on a dwindling cache of more expensive drugs; many beyond the economic reach of impoverished people living in malarious regions of the world. Our course to recognition of xanthones as potential antimalarial agents took a rather circuitous route, involving both serendipity and empiricism, and is described together with mechanistic details of drug action. From a chance encounter with a sea urchin collected near the city of Cannon Beach on the Oregon coast to naturally occurring and functionalized xanthones, it is revealed how these compounds target the Plasmodium parasites most vulnerable feature--the digestive vacuole.


Journal of Neuroscience Research | 2004

α lipoic acid inhibits human T-cell migration: Implications for multiple sclerosis

Gail Marracci; Gabriel P. McKeon; Whitney E. Marquardt; Rolf W. Winter; Michael K. Riscoe; Dennis Bourdette

We have demonstrated previously the ability of the antioxidant α lipoic acid (ALA) to suppress and treat a model of multiple sclerosis (MS), relapsing experimental autoimmune encephalomyelitis (EAE). We describe the effects of ALA and its reduced form, dihydrolipoic acid (DHLA), on the transmigration of human Jurkat T cells across a fibronectin barrier in a transwell system. ALA and DHLA inhibited migration of Jurkat cells in a dose‐dependent fashion by 16–75%. ALA and DHLA reduced matrix metalloproteinase‐9 (MMP‐9) activity by 18–90% in Jurkat cell supernatants. GM6001, a synthetic inhibitor of MMP, reduced Jurkat cell migration, but not as effectively as ALA and DHLA did. Both ALA and DHLA downmodulated the surface expression of the α4β1 integrin (very late activation‐4 antigen; VLA‐4), which binds fibronectin and its endothelial cell ligand vascular cell adhesion molecule‐1 (VCAM‐1). Moreover, ALA, but not DHLA, reduced MMP‐9‐specific mRNA and extracellular MMP‐9 from Jurkat cells and their culture supernatants as detected by relative reverse transcriptase‐polymerase chain reaction (RT‐PCR) and enzyme‐linked immunosorbent assay (ELISA), respectively. ALA and DHLA inhibited Jurkat cell migration and have different mechanisms for inhibiting MMP‐9 activity. These data, coupled with its ability to treat relapsing EAE, suggest that ALA warrants investigation as a therapy for MS.


Antimicrobial Agents and Chemotherapy | 2002

Optimization of Xanthones for Antimalarial Activity: the 3,6-Bis-ω-Diethylaminoalkoxyxanthone Series

Jane Xu Kelly; Rolf W. Winter; David H. Peyton; David J. Hinrichs; Michael K. Riscoe

ABSTRACT Hydroxyxanthones have been identified as novel antimalarial agents. The compounds are believed to exert their activity by complexation to heme and inhibition of hemozoin formation. Modification of the xanthone structure was pursued to improve their antimalarial activity. Attachment of R-groups bearing protonatable nitrogen atoms was conducted to enhance heme affinity through ionic interactions with the propionate side chains of the metalloporphyrin and to facilitate drug accumulation in the parasite food vacuole. A series of 3,6-bis-ω-diethylaminoalkoxyxanthones with side chains ranging from 2 to 8 carbon atoms were prepared and evaluated. Measurement of heme affinity for each of the derivatives revealed a strong correlation (R2 = 0.97) between affinity and antimalarial potency. The two most active compounds in the series contained 5- and 6-carbon side chains and exhibited low nanomolar 50% inhibitory concentration (IC50) values against strains of chloroquine-susceptible and multidrug-resistant Plasmodium falciparum in vitro. Both of these xanthones exhibit stronger heme affinity (8.26 × 105 and 9.02 × 105 M−1, respectively) than either chloroquine or quinine under similar conditions and appear to complex heme in a unique manner.


Experimental Parasitology | 2011

Optimization of endochin-like quinolones for antimalarial activity

Rolf W. Winter; Jane Xu Kelly; Martin J. Smilkstein; David J. Hinrichs; Dennis R. Koop; Michael K. Riscoe

Our prior work on tricyclic acridones combined with a desire to minimize the tricyclic system led to an interest in antimalarial quinolones and a reexamination of endochin, an experimental antimalarial from the 1940s. In the present article, we show that endochin is unstable in the presence of murine, rat, and human microsomes which may explain its relatively poor antimalarial activity in mammalian systems. We also profile the structure-activity relationships of ≈ 30 endochin-like quinolone (ELQ) analogs and highlight features that are associated with enhanced metabolic stability, potent antiplasmodial activity against multidrug resistant strains of Plasmodium falciparum, and equal activity against an atovaquone-resistant clinical isolate. Our work also features an ELQ construct containing a polyethylene glycol carbonate pro-moiety that is highly efficacious by oral administration in a murine malaria model. These findings provide compelling evidence that development of ELQ therapeutics is feasible.


Antimicrobial Agents and Chemotherapy | 2007

Design, Synthesis, and Evaluation of 10-N-Substituted Acridones as Novel Chemosensitizers in Plasmodium falciparum

Jane Xu Kelly; Martin J. Smilkstein; Roland A. Cooper; Kristin D. Lane; Robert A. Johnson; Aaron Janowsky; Rozalia A. Dodean; David J. Hinrichs; Rolf W. Winter; Michael K. Riscoe

ABSTRACT A series of novel 10-N-substituted acridones, bearing alkyl side chains with tertiary amine groups at the terminal position, were designed, synthesized, and evaluated for the ability to enhance the potency of quinoline drugs against multidrug-resistant (MDR) Plasmodium falciparum malaria parasites. A number of acridone derivatives, with side chains bridged three or more carbon atoms apart between the ring nitrogen and terminal nitrogen, demonstrated chloroquine (CQ)-chemosensitizing activity against the MDR strain of P. falciparum (Dd2). Isobologram analysis revealed that selected candidates demonstrated significant synergy with CQ in the CQ-resistant (CQR) parasite Dd2 but only additive (or indifferent) interaction in the CQ-sensitive (CQS) D6. These acridone derivatives also enhanced the sensitivity of other quinoline antimalarials, such as desethylchloroquine (DCQ) and quinine (QN), in Dd2. The patterns of chemosensitizing effects of selected acridones on CQ and QN were similar to those of verapamil against various parasite lines with mutations encoding amino acid 76 of the P. falciparum CQ resistance transporter (PfCRT). Unlike other known chemosensitizers with recognized psychotropic effects (e.g., desipramine, imipramine, and chlorpheniramine), these novel acridone derivatives exhibited no demonstrable effect on the uptake or binding of important biogenic amine neurotransmitters. The combined results indicate that 10-N-substituted acridones present novel pharmacophores for the development of chemosensitizers against P. falciparum.


Journal of Inorganic Biochemistry | 2001

A spectroscopic investigation of the binding interactions between 4,5-dihydroxyxanthone and heme

Jane Xu Kelly; Rolf W. Winter; Michael K. Riscoe; David H. Peyton

In order to investigate one possible mechanism by which xanthones inhibit growth of malaria-causing Plasmodium parasites, optical and NMR spectroscopic studies were performed on a prototypic xanthone, 4,5-dihydroxyxanthone (45X2), upon its complexation to heme. The 45X2 x heme complex stoichiometry in aqueous solution was found to be 1:2; this interaction was non-cooperative, and exhibited a very similar heme complex dissociation constant (K(d)=5.1 x 10(-6)) as observed for the common antimalarial agents, chloroquine and quinine. The 45X2 x heme(2) complex formation was found to be both pH- and solvent-dependent, with clear evidence of the xanthone carbonyl moiety coordinating with the iron of heme. Hydrogen bonding between the hydroxyl groups of 45X2 and the propionate side chains of heme, as well as pi-pi stacking between both aromatic systems appeared to contribute to the overall stability of the 45X2 x heme(2) complex, as judged by 1H NMR. It was concluded that 45X2 forms a complex with a heme dimer in aqueous solution, and that this interaction can be generalized to account for its in vivo detrimental effect of parasite growth through an effective inhibition of hemozoin aggregate formation.


Chemistry: A European Journal | 2009

5-(1,2,3-Triazol-1-yl)tetrazole derivatives of an azidotetrazole via click chemistry.

Takashi Abe; Guo-Hong Tao; Young‐Hyuk Joo; Rolf W. Winter; Gary L. Gard; Jean'ne M. Shreeve

N-C bonded (non-bridged) 5-(1,2,3-triazol-1-yl)tetrazoles were synthesized by the Cu(I)-catalyzed 1,3-dipolar azide-alkyne cycloaddition click reaction using 5-azido-N-(propan-2-ylidene)-1H-tetrazole (1). For example, the click reaction of 1 in the presence of CuSO(4)5 H(2)O and Na ascorbate at 65-70 degrees C for 48 h in CH(3)CN/H(2)O co-solvent was found to be limited to only terminal alkynes that have electron-withdrawing groups, CF(3)C[triple chemical bond]CH (2 a) and SF(5)C[triple chemical bond]CH (2 b), giving rise to isopropylidene-[5-(4-trifluoromethyl-1,2,3-triazol-1-yl)tetrazol-1-yl]amine (3 a) and isopropylidene-[5-(4-pentafluorosulfanyl-1,2,3-triazol-1-yl)tetrazol-1-yl]amine (3 b) in 47 % and 66 % yields, respectively. When carried out under conditions using CuI and 2,6-lutidine as catalysts at 0 degrees C for 13 h in CHCl(3), the click reaction was versatile toward alkynes even those having electron-donating groups. Properties of new products were determined and compared with those of 1. Heats of formation, detonation pressures, detonation velocities and impact sensitivities are reported for these new 5-(1,2,3-triazol-1-yl)tetrazoles.


Molecular and Biochemical Parasitology | 2008

A drug-selected Plasmodium falciparum lacking the need for conventional electron transport

Martin J. Smilkstein; Isaac P. Forquer; Atsuko Kanazawa; Jane Xu Kelly; Rolf W. Winter; David J. Hinrichs; David M. Kramer; Michael K. Riscoe

Mitochondrial electron transport is essential for survival in Plasmodium falciparum, making the cytochrome (cyt) bc(1) complex an attractive target for antimalarial drug development. Here we report that P. falciparum cultivated in the presence of a novel cyt bc(1) inhibitor underwent a fundamental transformation in biochemistry to a phenotype lacking a requirement for electron transport through the cyt bc(1) complex. Growth of the drug-selected parasite clone (SB1-A6) is robust in the presence of diverse cyt bc(1) inhibitors, although electron transport is fully inhibited by these same agents. This transformation defies expected molecular-based concepts of drug resistance, has important implications for the study of cyt bc(1) as an antimalarial drug target, and may offer a glimpse into the evolutionary future of Plasmodium.

Collaboration


Dive into the Rolf W. Winter's collaboration.

Top Co-Authors

Avatar

Gary L. Gard

Portland State University

View shared research outputs
Top Co-Authors

Avatar

John A. Schlueter

Argonne National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Paul G. Nixon

Portland State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Urs Geiser

Argonne National Laboratory

View shared research outputs
Top Co-Authors

Avatar

J. Mohtasham

Portland State University

View shared research outputs
Top Co-Authors

Avatar

J. Wosnitza

Helmholtz-Zentrum Dresden-Rossendorf

View shared research outputs
Top Co-Authors

Avatar

Jane Xu Kelly

Portland State University

View shared research outputs
Top Co-Authors

Avatar

Aravinda M. Kini

Argonne National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge