What are the new drugs for Malaria?

17 March 2025

Introduction to Malaria 
Malaria remains one of the most serious infectious diseases in the world, affecting vulnerable populations across tropical and subtropical regions. The disease is caused by parasites belonging to the Plasmodium species, with Plasmodium falciparum being responsible for the most severe and life‐threatening form, and Plasmodium vivax, Plasmodium malariae, Plasmodium ovale, and Plasmodium knowlesi also contributing to the overall burden. Annually, hundreds of millions are infected and nearly a million deaths occur, especially among children under five. The pathogenesis results from cycles of parasitic multiplication in red blood cells, which can lead to severe anemia, cerebral complications, and multi‐organ failure. The historical significance of malaria is also evident in its social and economic impact—impairing development and overwhelming health systems in endemic countries. 

Current Treatment Landscape 
For decades, the treatment of malaria has relied on a combination of monotherapies and combination therapies. Chloroquine once dominated the treatment landscape but lost its efficacy as resistant strains of P. falciparum emerged in the 1960s. Artemisinin-based combination therapies (ACTs) emerged as the gold standard following the widespread occurrence of chloroquine resistance; ACTs combine fast-acting artemisinin derivatives with longer lasting partner drugs such as lumefantrine, mefloquine, piperaquine, or amodiaquine. However, even ACTs are now facing emerging resistance, particularly in the Greater Mekong subregion and more recently in parts of Africa. The current therapeutic portfolio has also expanded to include prophylactic agents and anti-relapse treatments. Despite these advances, the persistence of drug-resistant parasites, suboptimal adherence to multi-day regimens, and other pharmacokinetic challenges underscore the urgent need for new drugs with novel mechanisms of action, improved efficacy, and simplified dosing regimens to ensure sustainable management and eventual eradication of the disease.

New Anti-Malarial Drugs 
Recently Approved Drugs 
Recent efforts to combat drug resistance and improve treatment outcomes have led to the development and regulatory approval of new anti-malarial agents. One of the most notable recent approvals is tafenoquine. Tafenoquine is a long-acting 8‑aminoquinoline that has been approved for the prophylaxis of malaria and, importantly, for preventing relapse in patients with Plasmodium vivax infections. Its extended half-life allows for a simplified dosing regimen compared to primaquine, reducing the challenges associated with adherence to a 14-day regimen. Tafenoquine offers the unique advantage of being effective against dormant liver-stage parasites (hypnozoites) that can cause relapses, thus filling an important therapeutic gap in malaria management. 

Another promising development in this arena is the advancement of ganaplacide as part of a combination therapy with lumefantrine. Unlike traditional ACTs, ganaplacide represents an entirely new class of antimalarials that does not contain artemisinin derivatives. This drug, which acts through a novel mechanism distinct from that of artemisinins, has advanced into pivotal Phase III studies in combination therapies. Early clinical studies have shown that the ganaplacide/lumefantrine combination demonstrates effective parasite clearance even against artemisinin-resistant strains, heralding a potential breakthrough in areas where first-line treatments are failing. 

It is also important to acknowledge that, while not yet approved, some drugs have recently received regulatory nods in some regions and are being rapidly adopted in national programs based on positive scientific opinion or emergency use provisions. These advances represent significant pivots in the therapeutic strategies against malaria, addressing shortcomings of older regimens and offering hope for a future where single-dose cures could become a reality.

Drugs in Clinical Trials 
In addition to already approved agents, a robust pipeline of candidate drugs is under evaluation in various stages of clinical trials. Several compounds have shown potential by targeting multiple stages of the malaria parasite’s lifecycle and by employing mechanisms of action that are distinct from those of current therapies. For example: 

• Artefenomel (OZ439): Artefenomel is a synthetic trioxolane designed to improve upon the short half-life of artemisinin by offering potent parasite clearance in a single dose. Clinical trials have indicated that artefenomel, when combined with partner drugs, can provide rapid parasite clearance even in regions beset by resistance, potentially supporting its use as part of a single-dose treatment regimen. 

• NITD609 (Cipargamin): Belonging to the spiroindolone class, NITD609 targets the parasite’s ATP4 ion pump, disrupting sodium homeostasis and leading to parasite death. This mode of action is substantially different from that of ACTs, and early clinical trials have revealed promising safety and efficacy profiles. NITD609 is progressing through clinical trial phases and is anticipated to provide an effective treatment option for drug-resistant malaria. 

• DSM265: This novel inhibitor targets the parasite’s dihydroorotate dehydrogenase (DHODH), a key enzyme in pyrimidine biosynthesis. DSM265 has shown rapid parasite clearance in early-phase clinical studies and might become a cornerstone of combination therapies aimed at reducing the parasite reservoir. 

• KAF156: Another compound in clinical trials, KAF156, displays broad-spectrum activity against blood-stage parasites and is being evaluated in both adult and pediatric populations. Its mechanism, though still under investigation, offers promise in terms of overcoming existing resistance patterns and providing a simplified dosing regimen. 

• P218: This candidate antimalarial targets dihydrofolate reductase (DHFR) in mutated strains of the parasite that resist traditional drugs such as pyrimethamine. P218’s design incorporates detailed structure-activity relationships to ensure efficacy against resistant strains, and early data from clinical studies are promising. 

• DDD107498: An emerging candidate, DDD107498 has demonstrated potent activity against multiple stages of the parasite in preclinical models, showing particular promise as a multi-stage active agent. Its progression into early clinical trials highlights its potential to contribute to combination therapies that could block transmission and treat symptomatic infections. 

Collectively, these candidate drugs reflect a strategic shift in drug discovery aimed at developing agents with novel, diverse mechanisms of action that target different metabolic pathways and cellular processes within the parasite. By combining these novel drugs with existing therapeutics, researchers hope not only to improve individual patient outcomes but also to reduce transmission at the community level through enhanced gametocytocidal activity and longer-lasting protection.

Drug Development and Efficacy 
Mechanism of Action 
Understanding the mechanism of action of new anti-malarial drugs is critical for their successful development, both to overcome existing drug resistance and to anticipate potential future hurdles. Recent compounds employ a variety of targets and pathways: 

• ATP4 Ion Pump Inhibition: NITD609, for instance, works by inhibiting PfATP4—a membrane ion pump that is essential for maintaining sodium homeostasis in the parasite. Disruption of this pump leads to an intracellular ionic imbalance, resulting in rapid parasite death. This mode of action is distinct from those of traditional therapies, offering a promising avenue in the fight against drug-resistant malaria. 

• DHODH Inhibition: DSM265’s inhibition of dihydroorotate dehydrogenase disrupts pyrimidine biosynthesis, which is essential for nucleic acid production and parasite replication. This target is attractive because it has limited overlap with human DHODH, potentially reducing adverse effects while ensuring selective toxicity against the parasite. 

• Inhibition of Folate Pathways: P218 is designed to overcome mutations in the DHFR gene, which confer resistance to antifolate drugs such as pyrimethamine. By optimizing interactions with the mutated enzyme, P218 is able to restore antifolate activity and effectively clear parasites. 

• Multi-Stage Activity and Gametocytocidal Effects: Artefenomel (OZ439) not only exhibits potent activity against asexual blood stages but has also been shown to act on gametocytes, thereby potentially reducing transmission. Likewise, ganaplacide and KAF156 are being studied for their ability to eliminate parasites at various lifecycle stages—from the intraerythrocytic stages to transmission-blocking effects. 

• Unique Molecular Scaffolds: Compounds such as DDD107498 represent a group of molecules with novel chemical architectures that interfere with essential parasite processes. Though the precise molecular target may still be under investigation, these compounds have demonstrated efficacy against both blood-stage parasites and latent liver stages in preclinical models. Such multi-target approaches are particularly welcome given the growing problem of resistance against single-target drugs. 

The diversity in mechanisms illustrates the current strategy of “target diversification” in malaria drug development. By addressing multiple facets of the parasite’s biology—from metabolic pathways and ion homeostasis to structural protein function—these new agents promise not only to broaden treatment options but also to delay the onset of resistance mechanisms that have historically undermined malaria control efforts.

Efficacy Studies and Results 
The efficacy of these new drugs has been evaluated in a range of preclinical and clinical settings, with emphasis on both parasite clearance and safety profiles: 

• Rapid Parasite Clearance: Studies have demonstrated that compounds targeting ATP4 (such as NITD609) can achieve rapid parasite clearance even in cases where ACTs are failing due to resistance. Parasite reduction ratios and clearance rates in early-phase clinical trials have shown significant improvements, sometimes reducing parasite loads within 48 hours. 

• Sustained Activity and Single-Dose Potential: Artefenomel (OZ439) has been highlighted for its potential to be administered as a single-dose treatment. Clinical trials have shown that when combined with other partner drugs, artefenomel achieves sufficient systemic exposure, rapid reduction of parasitemia, and maintains protective drug levels that may simplify dosing regimens, thus potentially improving patient compliance. 

• Combination Therapies: The ganaplacide/lumefantrine combination has undergone extensive testing in Phase II and III clinical studies, demonstrating high cure rates and good tolerability even in regions with documented artemisinin resistance. These studies have highlighted not only the rapid action of ganaplacide but also the sustained partner drug effect from lumefantrine, which together yield promising results in terms of efficacy and safety. 

• Homologous and Heterologous Parasite Clearance: DSM265 and KAF156 have both been effective in lowering parasite densities in a variety of patient populations, including adults and children. Efficacy trials measuring adequate clinical and parasitological response (ACPR) rates have shown promising outcomes, with some trials indicating cure rates surpassing 90% in areas where resistance has reduced the effectiveness of standard therapies. 

• Multi-Stage Targeting: The overall strategy behind many of these agents is their ability to inhibit parasite development at multiple stages. For instance, drugs with gametocytocidal properties not only treat clinical infections but also reduce the risk of transmission by killing or preventing the maturation of gametocytes. Early data from combination therapies incorporating such agents have indicated a substantial reduction in the infectious period post-treatment, an outcome that is vital for interrupting the cycle of transmission. 

The success of these efficacy studies reflects a shift in antimalarial drug development, where enhanced preclinical screening, advanced molecular modeling, and innovative clinical trial designs are used to capture a broad spectrum of data. These efforts have culminated in clinical results that are promising both in terms of rapid clearance kinetics and in achieving high cure rates even in regions beset by drug resistance.

Challenges and Future Directions 
Drug Resistance Issues 
Despite significant progress in the development of new drugs, one of the most pressing challenges in malaria treatment remains the emergence and spread of drug resistance. Resistance to artemisinins is now well documented, with mutations in the Kelch13 propeller region and other genetic markers contributing to reduced parasite susceptibility. As long as resistance develops, the window of opportunity for any drug is finite, and history has taught us that parasites adapt rapidly to selective pressures. Some of the new agents are designed with potential resistance-proofing in mind by targeting novel pathways not previously exploited by existing drugs. Nonetheless, the history of malaria treatment suggests that even these novel compounds may eventually encounter resistance if they are deployed as monotherapies. Therefore, many future strategies emphasize the use of combination therapies that target multiple pathways concurrently to minimize the likelihood of resistance development. 

Another challenge is the effective surveillance of drug efficacy in the field. In vivo and in vitro assays and molecular marker studies are essential to monitor emerging resistance. However, these methods vary in their sensitivity and standardization; therefore, the integration and harmonization of surveillance systems is urgently needed to capture real-time evidence of declining efficacy. Advances in genomic sequencing and mathematical modeling are playing critical roles in identifying resistance trends and informing policy adjustments, although substantial resource allocation and training remain significant hurdles. 

Future Research and Development 
Looking forward, the research and development agenda for anti-malarial drugs must continue to evolve to address both current challenges and anticipate future ones. There are several areas of focus: 

• Multi-targeted Approaches: Future drugs are expected not only to clear parasites rapidly but also to block transmission by exhibiting gametocytocidal activities. This multi-target strategy is essential for both clinical cure and reducing community-level transmission. Researchers are exploring compounds with activity against liver-stage parasites, blood-stage parasites, and sexual forms, thus tackling malaria at multiple fronts simultaneously. 

• Simplified Dosing Regimens: Simplifying treatment regimens remains a critical goal. The potential for single-dose cures, as indicated by artefenomel and the ganaplacide/lumefantrine combination, could dramatically improve patient adherence and reduce the risk of sub-therapeutic dosing that drives resistance. Future drug development efforts will likely focus on optimizing pharmacokinetic and pharmacodynamic properties to achieve sustained drug levels with simplified dosing schedules. 

• New Screening and Modeling Methods: The integration of high-throughput screening, in vitro assays, and in silico modeling has accelerated the identification of novel drug leads. Advanced techniques in molecular biology and genomics are now routinely employed to predict potential resistance mechanisms even before clinical deployment. These technologies, coupled with rigorous efficacy models, are expected to enhance the selection of effective candidates and streamline the development process. 

• Collaborative Networks and Funding: The fight against malaria is inherently a global and collaborative effort. Increased funding, better infrastructure for clinical trials in endemic regions, and international partnerships are all necessary to sustain the development pipeline. Organizations like the Medicines for Malaria Venture (MMV) have been instrumental in fostering public-private partnerships that have yielded several promising candidates. Continued collaboration among research institutions, industry players, and governments will be essential to ensure that novel drugs traverse the entire development pathway successfully. 

• Addressing Safety and Tolerability: While efficacy is paramount, long-term safety and tolerability remain critical, especially when drugs are intended for use in mass drug administration or in vulnerable populations such as children and pregnant women. Future research must balance efficacy with an acceptable safety profile by thoroughly investigating pharmacokinetic variability, potential drug-drug interactions, and long-term adverse event monitoring. 

• Integration of Transmission-Blocking Strategies: The ultimate goal of malaria eradication is not merely curing individual cases but also interrupting transmission within communities. Future research will increasingly focus on agents that have strong transmission-blocking properties, either as standalone compounds or as components of combination therapies. This involves not only targeting asexual blood stages but also the gametocyte forms that are responsible for infecting mosquitoes. 

• Personalized and Regionalized Therapies: Given the genetic diversity of Plasmodium strains across different geographic regions, a “one-size-fits-all” approach is unlikely to suffice. Tailoring therapies based on regional resistance patterns, local parasite genotypes, and patient demographics may enhance treatment outcomes. Research efforts in pharmacogenomics and personalized medicine will provide further insights into optimizing treatments for diverse populations. 

Detailed Conclusion 
In summary, the landscape of anti-malarial drug development is undergoing a profound transformation driven by both the urgent need to overcome drug resistance and the promise of innovative, multi-targeted therapies. The overview of malaria underscores a persistent global burden that has necessitated the evolution of treatment strategies from traditional monotherapies to complex combination regimens. Current first-line treatments, most notably ACTs, have served well but are increasingly compromised by emerging resistance. New anti-malarial drugs, such as tafenoquine—which has improved dosing schedules and is effective in preventing relapse in P. vivax infections—and novel agents like ganaplacide in combination with lumefantrine, artefenomel (OZ439), NITD609, DSM265, KAF156, P218, and DDD107498, are redefining our therapeutic arsenal. 

From a mechanistic standpoint, these new compounds employ a diverse range of targets—from the inhibition of vital ion pumps and crucial enzymes in nucleotide biosynthesis to targeting resistance-associated enzymes and exhibiting transmission-blocking effects. This target diversification is essential to outpace the adaptive resistance mechanisms of the parasite. Efficacy studies have demonstrated rapid parasite clearance, high cure rates, and potential for simplified, even single-dose regimens that could revolutionize patient adherence and reduce community-level transmission. These outcomes are supported by robust clinical trial data, in-depth pharmacokinetic and pharmacodynamic models, and innovative preclinical experiments that elucidate precise the molecular underpinnings of drug action. 

Despite these promising advances, considerable challenges remain. The specter of drug resistance continues to loom large, necessitating vigilant surveillance, streamlined drug development processes, and integrated treatment strategies that combine multiple mechanisms of action. Future research must address the sustainability of efficacious drug regimens by emphasizing multi-stage targeting, simplified dosing regimens, and rigorous safety evaluations. Moreover, expanding collaborative networks, increasing funding for research, and tailoring therapies to regional parasite profiles will be key to ensuring that the next generation of anti-malarial drugs can be effectively deployed in the field. 

In conclusion, the innovation in anti-malarial drug discovery represents a beacon of hope amid growing global challenges. While new drugs such as tafenoquine and ganaplacide/lumefantrine combinations are already redefining treatment standards, an active and dynamic pipeline—including artefenomel, NITD609, DSM265, KAF156, P218, and DDD107498—promises to further enhance our capacity to combat malaria. These advances, grounded in a deep understanding of molecular mechanisms and supported by extensive efficacy studies, pave the way for therapeutic regimens that are not only more effective but also better tailored to the needs of diverse and high-burden populations. The future of malaria treatment hinges on our ability to combine these new drugs with innovative strategies for surveillance, combination therapy, and regional customization—all of which are foundational to the ultimate goal of malaria elimination and eradication.

Curious to see how Eureka LS fits into your workflow? From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.