Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors

Tag: malaria

Expeditious recruitment of circulating memory CD8 T cells to the liver facilitates control of malaria

Circulating memory CD8 T cell trafficking and protective capacity during liver-stage malaria infection remains undefined. We find that effector memory CD8 T cells (Tem) infiltrate the liver within 6 hours after malarial or bacterial infections and mediate pathogen clearance. Tem recruitment coincides with rapid transcriptional upregulation of inflammatory genes in Plasmodium-infected livers. Recruitment requires CD8 T cell-intrinsic LFA-1 expression and the presence of liver phagocytes. Rapid Tem liver infiltration is distinct from recruitment to other non-lymphoid tissues in that it occurs both in the absence of liver tissue resident memory “sensing-and-alarm” function and ∼42 hours earlier than in lung infection by influenza virus. These data demonstrate relevance for Tem in protection against malaria and provide generalizable mechanistic insights germane to control of liver infections.

Mitchell N Lefebvre, Fionna A Surette, Scott M Anthony, Rahul Vijay, Isaac J Jensen, Lecia L Pewe, Lisa S Hancox, Natalija Van Braeckel-Budimir, Stephanie van de Wall, Stina L Urban, Madison R Mix, Samarcith P Kurup, Vladimir P Badovinac, Noah S Butler, John T Harty. Cell Rep. 2021 Nov 2;37(5):109956. doi: 10.1016/j.celrep.2021.109956.

Researchers implement new tool to screen drugs for treating malaria relapses

Steve Maher
Steve Maher, assistant research scientist in the Center for Tropical Emerging and Global Diseases, leads a team of researchers who have implemented a new screening tool to determine if a drug candidate kills hypnozoites, the cause of malaria relapses. (Photo by Donna Huber)

Globally, efforts to control malaria caused by Plasmodium vivax are lagging behind that of other species of Plasmodium due to its unique biology. A team of researchers at the University of Georgia, the Institute Pasteur of Cambodia, and Shoklo Malaria Research Unit in Thailand detail a new screening tool and report for the first time a method capable of discovering novel experimental drug compounds for use against vivax malaria. Their study was recently published in Scientific Reports.

The parasite species P. vivax is the most widespread cause of malaria. While not as deadly as malaria caused by P. falciparum, it can cause severe disease and has a significant impact on both national economies and personal finances, in part due to this species’ propensity to cause relapses.

Relapses are caused by hypnozoites, a form of the parasite residing in the liver, which can lie dormant for a period of time before causing another symptomatic blood infection. During this period of dormancy, hypnozoites are not susceptible to standard antimalarials, meaning a patient treated for a blood infection is not fully cured.

“With this assay, we can now tell earlier on in the drug discovery process if a compound is going to work against hypnozoites,” said Steven Maher, assistant research scientist at the Center for Tropical and Emerging Global Diseases and lead researcher on the study. “In this study, we were able to identify three new drugs that kill dormant hypnozoites.”

One of the drugs identified looks promising as a possible new treatment, though Maher said it will need more testing. The other two could be useful in studying hypnozoite biology and increase understanding of such things as the mechanisms of dormancy.

The team’s report also shows how two current antimalarial drugs, chloroquine and tafenoquine, synergistically work together to kill hypnozoites. However, these drugs cannot be administered to children and pregnant women (due to their known side effects), nor to people who lack the enzyme called G6PD. Up to 20% of the population in southeast Asia are G6PD deficient.

“The current drug therapies work well to treat the symptomatic blood stage of vivax malaria,” said Steven Maher. “However, in vivax malaria we need to eliminate hypnozoites to fully cure the patient, and for that we need new therapies.”

To compound the problem, typical mouse models used in malaria drug research can’t determine if the experimental compounds work against hypnozoites because the Plasmodium species that infects mice doesn’t produce them. Additionally, because the assays used as the first step in discovering potential new drug compounds focus on the blood stage of the parasite, researchers need a different kind of assay that will allow them to test these compounds on hypnozoites, which requires a stable culture of liver cells.

“It’s a challenge because you have to get samples from where the vivax malaria is endemic,” said Maher. “Liver cells don’t stay viable in culture for long, and these assays take eight days to show results. The assay itself is difficult to run, but we have a great team of researchers in Cambodia and Thailand that has really helped to make this possible.”

The team is continuing to build better tools to overcome the challenges drug discovery in P. vivax faces as they begin to test these drugs in animal models.

 

This story first appeared at UGA Research.

 

 

 

Probing the distinct chemosensitivity of Plasmodium vivax liver stage parasites and demonstration of 8-aminoquinoline radical cure activity in vitro

Improved control of Plasmodium vivax malaria can be achieved with the discovery of new antimalarials with radical cure efficacy, including prevention of relapse caused by hypnozoites residing in the liver of patients. We screened several compound libraries against P. vivax liver stages, including 1565 compounds against mature hypnozoites, resulting in one drug-like and several probe-like hits useful for investigating hypnozoite biology. Primaquine and tafenoquine, administered in combination with chloroquine, are currently the only FDA-approved antimalarials for radical cure, yet their activity against mature P. vivax hypnozoites has not yet been demonstrated in vitro. By developing an extended assay, we show both drugs are individually hypnozonticidal and made more potent when partnered with chloroquine, similar to clinically relevant combinations. Post-hoc analyses of screening data revealed excellent performance of ionophore controls and the high quality of single point assays, demonstrating a platform able to support screening of greater compound numbers. A comparison of P. vivax liver stage activity data with that of the P. cynomolgi blood, P. falciparum blood, and P. berghei liver stages reveals overlap in schizonticidal but not hypnozonticidal activity, indicating that the delivery of new radical curative agents killing P. vivax hypnozoites requires an independent and focused drug development test cascade.

Steven P. Maher, Amélie Vantaux, Victor Chaumeau, Adeline C. Y. Chua, Caitlin A. Cooper, Chiara Andolina, Julie Péneau, Mélanie Rouillier, Zaira Rizopoulos, Sivchheng Phal, Eakpor Piv, Chantrea Vong, Sreyvouch Phen, Chansophea Chhin, Baura Tat, Sivkeng Ouk, Bros Doeurk, Saorin Kim, Sangrawee Suriyakan, Praphan Kittiphanakun, Nana Akua Awuku, Amy J. Conway, Rays H. Y. Jiang, Bruce Russell, Pablo Bifani, Brice Campo, François Nosten, Benoît Witkowski & Dennis E. Kyle. Sci Rep 11, 19905 (2021). https://doi.org/10.1038/s41598-021-99152-9

UGA researchers developing new models for malaria drug development and testing

Chet Joyner and Dennis Kyle
CTEGD member Chet Joyner and CTEGD director Dennis Kyle receive a grant from the Bill & Melinda Gates Foundation for malaria drug development and testing

Two UGA researchers are working to make it easier to develop effective treatments for malaria, a disease that sickens millions worldwide and kills hundreds of thousands each year.

In tropical climates around the globe, malaria poses a grave risk to already vulnerable populations. In 2019, the World Health Organization estimated that there were 229 million clinical cases of malaria worldwide and 409,000 deaths, usually in children below the age of five.

Currently, developing and testing drugs for malaria requires scientists to work in areas where the disease is prevalent or to work with expensive, hard-to-source equipment.  Chester Joyner, an Assistant Professor in the Center for Vaccines and Immunology, and Dennis Kyle, Professor of Infectious Diseases and Cellular Biology, are working to reduce those barriers to malaria drug testing and development.

Joyner and Kyle aim to establish systems that rely on equipment most researchers can obtain: a petri dish. If successful, Joyner says this new culture system will reduce costs and be distributed more easily to advance drug and vaccine research. The University of Georgia College of Veterinary Medicine received a grant for malaria drug development and testing from the Bill & Melinda Gates Foundation.

Worldwide, there are many malaria-causing parasites that result in varying degrees of illness. Joyner and Kyle’s research focuses on defeating one of the most challenging: Plasmodium vivax. Unlike many other malaria parasites, P. vivax can lie dormant in the livers of its hosts—allowing the infected to travel abroad completely unaware that they’re carrying a potentially deadly passenger.

“Most infections with P. vivax are not due to new infections,” says Joyner. “These infections come from this parasite activating and potentially causing disease and sustaining transmission.”

Malaria disproportionately affects the poorest communities in the world, creating a cycle of disease and poverty that current treatments have improved but been unable to stop. However, treating the dormant forms of P. vivax has been particularly challenging because they can cause more harm than good in at-risk populations like pregnant women and people with certain blood conditions.

“We want researchers to have access to technologies to study P. vivax and develop new approaches to control and eliminate this parasite,” Joyner explains.

 

This article first appeared at https://give.uga.edu/uga-researchers-developing-new-models-for-malaria-drug-development-and-testing/

Researchers to test drug candidates to treat malaria

by Donna Huber

Belen Cassera
Belen Cassera is leading a research team that will test two new drugs for the treatment of malaria. The team’s work will be funded by a $3.7 million grant from the National Institutes of Health. (Photo credit: Amy Ware)

Though malaria was eliminated from the U.S. 70 years ago, the mosquito-borne disease caused by the Plasmodium parasite is still rampant in many parts of the world – nearly 40% of the world’s population is at risk of contracting it, and nearly 450,000 people die each year from it. With the rise of drug resistance, the current medical treatments aren’t enough to end this disease.

“Every drug treatment currently in use for malaria is showing resistance or reduced efficacy,” said Belen Cassera, a member of the University of Georgia’s Center for Tropical and Emerging Global Diseases. “Furthermore, there are very limited treatments for the most vulnerable – children and pregnant women. Over 60% of deaths are children under the age of 5.”

Cassera is co-leading the research team that recently received a $3.7 million grant from the National Institutes of Health to test two new drug candidates.

“These compounds are really promising as they are easy to synthesize, cheap, reliable, have a low toxicity profile, and kill the parasites fast,” said Cassera, associate professor in the Department of Biochemistry and Molecular Biology, part of the Franklin College of Arts and Sciences.

What’s unique about these compounds is that they can kill the parasite in three development stages in humans. Current treatments only target the blood stage, which is when clinical symptoms appear.

The life cycle of the Plasmodium parasite is complex. When an infected mosquito bites a person, just a small number of parasites – usually less than a hundred – are injected into the bite site and then travel to the liver, where they multiply in number to thousands. Once their numbers are sufficient enough, they invade the bloodstream and infect red blood cells.

When the number of parasites reaches 100 million, symptoms occur and some of the parasites develop into a sexual form, also known as the gametocyte stage. This is when symptoms occur. The sexual form is then transmitted back to the mosquito when the person is bitten again.

This complex life cycle makes it difficult to find a treatment that will eradicate the disease. Breaking the cycle of transmission between humans and mosquitos is key to accomplishing that goal. That’s why the team is excited about discovering compounds that can attack the parasite on multiple fronts.

“We are really a powerhouse team,” said Cassera. “We have a leading medicinal chemistry expert in Paul Carlier, the robust parasitology resources of UGA, and Max Totrov brings the machine-learning expertise to tie it all together.”

Cassera is a UGA Innovation Fellow, and she also credits the knowledge gained at UGA’s 2019 Innovation Bootcamp with helping her prepare a grant proposal that would be of particular interest to drug manufacturers.

Cassera has been working for several years to identify new drug candidates, along with Carlier, a professor in the Virginia Tech College of Science’s Department of Chemistry and director of the Virginia Tech Center for Drug Discovery, and Max Totrov, a computational chemist at Molsoft.

“We started working with the Malaria Box from Medicines for Malaria Venture, and the discoveries we made in basic malaria biochemistry and medicinal chemistry really springboarded us to a new level and led us in this new direction,” Cassera said.

Cassera is leading the testing of the new chemical variations of the antimalarial compounds prepared by Carlier for effectiveness in cellular and animal models.

“My lab will be looking at levels of toxicity, the potential for resistance, and how well they work both directly on the parasite and in infected mice,” she said. “We’ll be performing the studies for making the go/no-go decision for these compounds.”

A joint patent application for both drug candidates was recently filed, and the team is optimistic that their research will yield fast-acting candidates for advanced pre-clinical evaluation.

 

This story originally appeared at UGAResearch

Aminoalkoxycarbonyloxymethyl Ether Prodrugs with a pH-Triggered Release Mechanism: A Case Study Improving the Solubility, Bioavailability, and Efficacy of Antimalarial 4(1 H)-Quinolones with Single Dose Cures

Preclinical and clinical development of numerous small molecules is prevented by their poor aqueous solubility, limited absorption, and oral bioavailability. Herein, we disclose a general prodrug approach that converts promising lead compounds into aminoalkoxycarbonyloxymethyl (amino AOCOM) ether-substituted analogues that display significantly improved aqueous solubility and enhanced oral bioavailability, restoring key requirements typical for drug candidate profiles. The prodrug is completely independent of biotransformations and animal-independent because it becomes an active compound via a pH-triggered intramolecular cyclization-elimination reaction. As a proof-of-concept, the utility of this novel amino AOCOM ether prodrug approach was demonstrated on an antimalarial compound series representing a variety of antimalarial 4(1H)-quinolones, which entered and failed preclinical development over the last decade. With the amino AOCOM ether prodrug moiety, the 3-aryl-4(1H)-quinolone preclinical candidate was shown to provide single-dose cures in a rodent malaria model at an oral dose of 3 mg/kg, without the use of an advanced formulation technique.

Andrii Monastyrskyi, Fabian Brockmeyer, Alexis N LaCrue, Yingzhao Zhao, Steven P Maher, Jordany R Maignan, Vivian Padin-Irizarry, Yana I Sakhno, Prakash T Parvatkar, Ami H Asakawa, Lili Huang, Debora Casandra, Sherwin Mashkouri, Dennis E Kyle, Roman Manetsch. J Med Chem. 2021 May 12. doi: 10.1021/acs.jmedchem.0c01104.

Some conditions apply: Systems for studying Plasmodium falciparum protein function

Plasmodium falciparum life cycle
Fig 1. Conditional protein knockdown used throughout the Plasmodium falciparum life cycle.

Malaria, caused by infection with Plasmodium parasites, remains a significant global health concern. For decades, genetic intractability and limited tools hindered our ability to study essential proteins and pathways in Plasmodium falciparum, the parasite associated with the most severe malaria cases. However, recent years have seen major leaps forward in the ability to genetically manipulate P. falciparum parasites and conditionally control protein expression/function. The conditional knockdown systems used in P. falciparum target all 3 components of the central dogma, allowing researchers to conditionally control gene expression, translation, and protein function. Here, we review some of the common knockdown systems that have been adapted or developed for use in P. falciparum. Much of the work done using conditional knockdown approaches has been performed in asexual, blood-stage parasites, but we also highlight their uses in other parts of the life cycle and discuss new ways of applying these systems outside of the intraerythrocytic stages. With the use of these tools, the field’s understanding of parasite biology is ever increasing, and promising new pathways for antimalarial drug development are being discovered.

Heather M Kudyba, David W Cobb, Joel Vega-Rodríguez, Vasant Muralidharan. PLoS Pathog. 2021 Apr 22;17(4):e1009442. doi: 10.1371/journal.ppat.1009442. eCollection 2021 Apr.

Synthesis of Mono- and Bisperoxide-Bridged Artemisinin Dimers to Elucidate the Contribution of Dimerization to Antimalarial Activity

During the past decade, artemisinin as an antimalarial has been in the spotlight, in part due to the Nobel Prize in Physiology or Medicine awarded to Tu Youyou. While many studies have been completed detailing the significant increase in activity resulting from the dimerization of natural product artemisinin, activity increases unaccounted for by the peroxide bridge have yet to be researched. Here we outline the synthesis and testing for antimalarial activity of artemisinin dimers in which the peroxide bridge in one-half of the dimer is reduced, resulting in a dimer with one active and one deactivated artemisinin moiety.

Cynthia L Lichorowic, Yingzhao Zhao, Steven P Maher, Vivian Padín-Irizarry, Victoria C Mendiola, Sagan T de Castro, Jacob A Worden, Debora Casandra, Dennis E Kyle, Roman Manetsch. ACS Infect Dis. 2021 Apr 1. doi: 10.1021/acsinfecdis.1c00066

A redox-active crosslinker reveals an essential and inhibitable oxidative folding network in the endoplasmic reticulum of malaria parasites

Oxidative folding in the P. falciparum ER

Malaria remains a major global health problem, creating a constant need for research to identify druggable weaknesses in P. falciparum biology. As important components of cellular redox biology, members of the Thioredoxin (Trx) superfamily of proteins have received interest as potential drug targets in Apicomplexans. However, the function and essentiality of endoplasmic reticulum (ER)-localized Trx-domain proteins within P. falciparum has not been investigated. We generated conditional mutants of the protein PfJ2-an ER chaperone and member of the Trx superfamily-and show that it is essential for asexual parasite survival. Using a crosslinker specific for redox-active cysteines, we identified PfJ2 substrates as PfPDI8 and PfPDI11, both members of the Trx superfamily as well, which suggests a redox-regulatory role for PfJ2. Knockdown of these PDIs in PfJ2 conditional mutants show that PfPDI11 may not be essential. However, PfPDI8 is required for asexual growth and our data suggest it may work in a complex with PfJ2 and other ER chaperones. Finally, we show that the redox interactions between these Trx-domain proteins in the parasite ER and their substrates are sensitive to small molecule inhibition. Together these data build a model for how Trx-domain proteins in the P. falciparum ER work together to assist protein folding and demonstrate the suitability of ER-localized Trx-domain proteins for antimalarial drug development.

David W. Cobb, Heather M. Kudyba, Alejandra Villegas, Michael R. Hoopmann, Rodrigo P. Baptista, Baylee Bruton, Michelle Krakowiak, Robert L. Moritz, Vasant Muralidharan. PLoS Pathog. 2021 Feb 3;17(2):e1009293. doi: 10.1371/journal.ppat.1009293.

Resistance to some, but not other dimeric lindenane sesquiterpenoid esters is mediated by mutations in a Plasmodium falciparum esterase

Unique lindenane sesquiterpenoid dimers from Chloranthecae spp. were recently identified with promising in vitro antiplasmodial activity and potentially novel mechanisms of action. To gain mechanistic insights to this new class of natural products, in vitro selection of Plasmodium falciparum resistance to the most active antiplasmodial compound, chlorajaponilide C, was explored. In all selected resistant clones, the half-maximal effective concentration (EC50) of chlorajaponilide C increased >250-fold, and whole genome sequencing revealed mutations in the recently discovered P. falciparum prodrug activation and resistance esterase (PfPARE). Chlorajaponilide C was highly potent (mean EC50 = 1.6 nM, n=34) against fresh Ugandan P. falciparum isolates. Analysis of the structure-resistance relationships revealed that in vitro potency of a subset of lindenane sesquiterpenoid dimers was not mediated by PfPARE mutations. Thus, chlorajaponilide C, but not some related compounds, required parasite esterase activity for in vitro potency, and those compounds serve as the foundation for development of potent and selective antimalarials.

Joshua H Butler, Rodrigo P Baptista, Ana Lisa Valenciano, Bin Zhou, Jessica C Kissinger, Patrick K Tumwebaze, Philip J Rosenthal, Roland Cooper, Jian-Min Yue, Maria Belen Cassera. ACS Infect Dis. 2020 Sep 24. doi: 10.1021/acsinfecdis.0c00487