Skip to main content

Repurposing of anti-malarial drugs for the treatment of tuberculosis: realistic strategy or fanciful dead end?

Abstract

Background

Drug repurposing offers a strategic alternative to the development of novel compounds, leveraging the known safety and pharmacokinetic profiles of medications, such as linezolid and levofloxacin for tuberculosis (TB). Anti-malarial drugs, including quinolones and artemisinins, are already applied to other diseases and infections and could be promising for TB treatment.

Methods

This review included studies on the activity of anti-malarial drugs, specifically quinolones and artemisinins, against Mycobacterium tuberculosis complex (MTC), summarizing results from in vitro, in vivo (animal models) studies, and clinical trials. Studies on drugs not primarily developed for TB (doxycycline, sulfonamides) and any novel developed compounds were excluded. Analysis focused on in vitro activity (minimal inhibitory concentrations), synergistic effects, pre-clinical activity, and clinical trials.

Results

Nineteen studies, including one ongoing Phase 1 clinical trial, were analysed: primarily investigating quinolones like mefloquine and chloroquine, and, to a lesser extent, artemisinins. In vitro findings revealed high MIC values for anti-malarials versus standard TB drugs, suggesting a limited activity. Synergistic effects with anti-TB drugs were modest, with some synergy observed in combinations with isoniazid or pyrazinamide. In vivo animal studies showed limited activity of anti-malarials against MTC, except for one study of the combination of chloroquine with isoniazid.

Conclusions

The repurposing of anti-malarials for TB treatment is limited by high MIC values, poor synergy, and minimal in vivo effects. Concerns about potential toxicity at effective dosages and the risk of antimicrobial resistance, especially where TB and malaria overlap, further question their repurposing. These findings suggest that focusing on novel compounds might be both more beneficial and rewarding.

Background

Drug repurposing, also known as drug repositioning, has emerged as an important strategy in contemporary medicine (Table 1). This approach, distinct from traditional drug discovery methods, leverages the known pharmacokinetics and safety profiles of existing drugs. By doing so, it side-steps the significant financial and time constraints inherent to the development and clinical testing of new compounds [1, 2]. Prime illustrations of this strategy include drugs like aspirin, initially intended for pain relief; sildenafil for hypertension and angina; and thalidomide for morning sickness; which have since been adapted for various other medical conditions [2]. Examples are relatively sparse for drugs from the field of infectious diseases. Notable instances include the repurposing of ketoconazole, an antifungal, for Cushing’s syndrome, and investigations into the use of levofloxacin and tetracyclines for Alzheimer’s disease [3]. Conversely, repurposing antimicrobials for different infectious diseases than their initial targets have proven beneficial, as seen with tetracycline for malaria [3] and linezolid for tuberculosis [4].

Table 1 Two distinct definitions of drug repurposing or repositioning

Recently, a tendency for a broader definition of repurposing has surfaced (Table 1), which includes the ‘repurposing’ of not only approved drugs but also investigational drugs which are being developed for other indications [1], often including novel computational models [5, 6]. While this approach is promising to identify novel compounds, most molecules may need modifications and must still pass through the demanding clinical evaluation process. Incorporation of investigational drugs requires a costly and lengthy clinical trial process, which contradicts the idea that drug repurposing is per se faster and less expensive.

Drug repurposing initiatives targeting new antimicrobial treatments typically begin with in vitro assays or, in more advanced stages, with pre-clinical rodent models. This approach has been a common thread in both bacterial and viral research [6,7,8,9]. However, the potential for an enormous disconnect between laboratory promise and clinical efficacy is highlighted using chloroquine and ivermectin in treating SARS-CoV-2. Despite initial laboratory indications suggesting potential, both compounds ultimately demonstrated no significant efficacy [10].

Tuberculosis (TB), caused by Mycobacterium tuberculosis complex (MTC), remains a leading cause of death globally, with approximately 1.6 million fatalities in 2022. Furthermore, the increasing prevalence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) TB poses significant treatment challenges [11, 12]. Consequently, there is an urgent need for novel anti-TB drugs. Trying to face this challenge, the past two decades have witnessed advancements such as the introduction of new anti-TB compounds (delamanid, bedaquiline) or novel treatment regimens intended to shorten treatment duration [13, 14].

Notably, the use of the existing antibiotic linezolid for TB treatment is often mentioned as a prime success story in drug repurposing [15]. Contrary to this, the use of the antibiotic levofloxacin for TB treatment is less commonly seen as 'true' repurposing; and is often simply considered an extension of the range of bacterial pathogens which are treatable with the drug. Despite these advances, the demand for new and repurposed anti-TB drugs continues unabated, addressing future issues of resistance, reducing toxic effects, identifying more effective drugs, and considering economic implications.

Compounds in the two major anti-malarial drug classes, quinolones and artemisinins, were developed and introduced exclusively for treating malaria. However, their potential for repurposing has been recognized, as several non-infectious disease indications have been well-established over time; or are currently being investigated (Table 2). Additionally, these drugs have been employed, or are under investigation, for treating various infections (Table 2).

Table 2 Examples of repurposing of anti-malarial drugs

Amidst the global scramble to find effective treatments against COVID-19, anti-malarials emerged prominently in research and clinical trials as repurposed drugs, albeit without proven efficacy. However, this spotlight on anti-malarial drugs reinvigorated interest in their repurposing potential, which could be pivotal in addressing other infectious diseases, notably tuberculosis (TB). Recognized for its efficiency and speed, drug repurposing is especially crucial in combatting difficult-to-treat pathogens for example those which develop resistance, including MDR and XDR-TB strains. Anti-malarials have also been the subject of research for their potential as repurposed drugs for TB [8, 24, 25]. Therefore, the objective of this review is to assess the impact of anti-malarials' repurposing for TB to determine whether such efforts have been effective, and should be pursued in the future; or rather reconsidered.

Methods

For this review, a PubMed search conducted in December 2023 focused on studies of anti-malarial drugs demonstrating in vitro and in vivo (using animal models) effects against Mycobacterium tuberculosis, or clinical effects on the treatment of tuberculosis. The search terms used were ‘antimalarial drugs tuberculosis’. Additionally, each individual antimalarial drug (Additional file 1: Table S1) of the two classes of quinolones and artemisinins was searched in combination with the term ‘tuberculosis’ (Additional file 1: Table S2). Furthermore, four clinical trial databases were screened for additional studies on this topic (EU Clinical Trial Register, ClinicalTrials.gov, BASEC, and ISRCTN registry). References in all relevant articles and their respective citations were also reviewed. All articles were screened based on their title and abstract using Rayyan Systems Inc (https://www.rayyan.ai/), and included studies that provide original data on the activity of established anti-malarial drug compounds (Additional file 1: Table S1) against MTC or their use in TB treatment.

Excluded were studies on anti-malarial agents initially developed as antibiotics for other infections, such as tetracyclines and clindamycin. Also omitted were drugs from classes primarily intended for non-malarial infections, such as those affecting folate synthesis (e.g., proguanil, pyrimethamine, sulfamethoxazole, or dapsone). Studies reporting in silico work, computer models, or investigations of novel compounds based on modifications of the molecular structures of existing anti-malarial drugs were also excluded. Consequently, compounds that have not yet been subjected to clinical investigation (see Table 1) were also excluded.

Study results were summarized according to their respective main methodologies applied, which were classified as: (i) In vitro activity of anti-malarial drugs against MTC in culture or in macrophage models, providing relevant quantitative measurements; (ii) in vitro activity of anti-malarial drugs against MTC in combination with other drugs (synergistic effects) in culture or in macrophage models; (iii) pre-clincial in vivo studies (animal models); and (iv) clinical trials in humans.

The publishing journal was assessed by verfiying the H-index reported by Scimago (https://www.scimagojr.com/) on 14/01/2024 which uses the Wikipedia defintion (https://en.wikipedia.org/wiki/H-index).

Results

A PubMed search yielded 1,187 hits, with an additional 86 identified through the screening of reference lists, ultimately culminating in 19 relevant papers (Additional file 1: Fig. S1). Upon closer examination, these papers encompass a range of study types: there are 19 instances of in vitro research reported within these papers, of which five investigate synergistic effects specifically. Additionally, there are three instances of in vivo animal research and a single ongoing Phase 1 clinical trial retrieved from ClinicalTrials.Gov (Table 3, Additional file 1: Tables S3–S6) [26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44]. These studies, predominantly published between 2006 and 2021, with a peak from 2016 to 2019, explored anti-malarial drug activity against MTC. Journal impact, assessed by Scimago H-indices, varied with most journals scoring over 100 (Additional file 1: Tables S3–S6). Notably, one 1990 study [27] on chloroquine had significant methodological limitations and high inhibitory concentrations compared to recent research (Additional file 1: Tables S3, S4). In several studies, the examination of the molecular structure of some anti-malarials yielded secondary outcomes related to anti-malarial drug activity against MTC [26, 36,37,38,39]. Other research compared anti-MTC activity of anti-malarial drugs to novel compounds [29, 30, 33, 40] or assessed activity against dormant MTC strains [28, 29, 31]. Eighteen studies focused on in-vitro anti-malarial activity against MTC (Additional file 1: Table S3), while five explored synergies with anti-TB drugs (Additional file 1: Table S4). Only three reported in-vivo effects in animal models (Additional file 1: Table S5).

Table 3 Summary of studies (n = 19) investigating the anti-malarial drug effects against M. tuberculosis complex

In vitro studies predominantly investigated quinolines, especially mefloquine, chloroquine, primaquine, tafenoquine, amodiaquine, and pyronaridine, while artemisinins and artesunate were less common (Table 3). These studies employed a variety of methodologies, notably viability detection assays like resazurin and Alamar blue. Some studies used liquid (BACTEC, MGIT) or solid media, which generally showed higher MIC values compared to the viability detection assays (Tables 3, Additional file 1: Tables S3, S4). Synergistic effects in vitro mainly involved isoniazid, pyrazinamide, and quinolones, with one study examining rifampicin and artemisinins [36] (Table 3, Additional file 1: Table S4).

The main findings are summarized in Table 4. All minimum inhibitory concentration (MIC) values were significantly higher than those for the breakpoints for anti-tuberculosis drugs (rifampicin: 1.2 µM = 1.0 µg/mL and isoniazid: 1.5 = 0.2 µg/mL) against MTC and half-maximal inhibitory concentration (IC50) values for anti-malarial drugs against Plasmodium falciparum. Notably, over 100 clinical isolates [32] showed MICs ranging from 11 – 42 µM for mefloquine. No inhibition by artemisinins was observed using MGIT or Ogawa solid media, in contrast to the resazurin-based method (Tables 3, Additional file 1: Table S3) [35]. Studies on mefloquine’s MICs in dormancy models showed similar effects to those in replicating MTC [29, 31, 40], with one study indicating equivalence to rifampicin in dormant MTC (MIC for mefloquine = rifampicin: 7 µM) [29]. Interestingly, a 1990 study using macrophage models reported chloroquine’s intracellular effect to be 50 times stronger than in culture [27], whereas other studies found more modest effects for mefloquine [31, 34] (Additional file 1: Table S3).

Table 4 Values for MIC, cytotoxicity and serum concentration of anti-malarial drugs compared to two major anti-tuberculosis drugs (all values in µM)*

Synergistic effects were negligible or modest for the anti-malarials chloroquine and tafenoquine [27, 28, 37] (Additional file 1: Table S3). However, the fractional inhibitory concentration index (FICI) of < 0.4 indicated synergy for mefloquine combined with pyrazinamide or isoniazid, and for artemisinin with isoniazid or amikacin [26, 36] (Additional file 1: Table S3). Other results for different drug combinatons showed a FICI of around 0.5, the threshold for synergism (Table 3, Additional file 1: Table S3).

In vivo studies demonstrated some effect of chloroquine, mefloquine, or artemisinins (Tables 3, S5). Most notably, in vivo studies in mice and guinea pigs found that combining isoniazid and chloroquine produced strong effects by various metrics [39] as specified in Tables 3 and Additional file 1: Table S5.

Regarding clinical trials, only one phase 1 trial was identified. This ongoing trial examines the tolerability of chloroquine combined with a standard quadruple TB drug regimen in healthy volunteers; results pending (Additional file 1: Table S5).

Discussion

The concept of repurposing clinically tested or approved drugs (Table 1), due to their established safety profiles, is a compelling approach for rapidly identifying new treatments. This strategy has also been applied to anti-malarial drugs for potential use in treating tuberculosis. Research often centres on the underlying structures of anti-malarials, using them as scaffolds for new compound development. This approach, illustrated by a review on quinolones [45], typically involves clinically non-tested compounds. Yet, this approach has been questioned for possibly negating the inherent benefits that were the original goal of reusing existing medications (Table 1). Chloroquine’s possible use as tuberculosis treatment is briefly discussed in a review by Rolain et al. [46], as is mefloquine [47]. A recent review expands on various anti-malarial drugs, although rather focusing on their possible mode of actions [48]. While all these reviews paint a rather optimistic picture of repurposing anti-malarial drugs, an in-depth review of the in vitro data on MIC values, synergistic effects, and in vivo data from animal models (Table 3, Additional file 1: Tables S3-S6) provides less support for this view.

Activity of anti-malarial drugs

Most minimum inhibitory concentration (MIC) data were obtained using colorimetric redox indicator assays (Additional file 1: Table S3), offering a rapid and simplified method for assessing drug resistance and screening new drugs [49]. However, MIC values determined through traditional liquid or solid media, which are standard in clinical microbiology, were significantly higher for mefloquine [32] and artemisinins [35]. Also, two studies which tested 113 and 228 clinical isolates, respectively [32, 41], reported higher MICs than those studies which used only a few laboratory-adapted strains (majority ≥ 21 μM mefloquine as compared to 4-12 μM, respectively) (see Tables 4, Additional file 1: S3 for details).

It is crucial to highlight that the MIC values for anti-malarial drugs are significantly higher compared to those reported for rifampicin and isoniazid, as demonstrated in Table 4. Additionally, the MIC values for these anti-malarial drugs against MTC are also markedly higher compared to their IC50 values against P. falciparum (Table 4). The serum levels of anti-malarials exceed the IC50 for P. falciparum which, interestingly, also includes rifampicin (Table 4). This explains why rifampicin has shown to have acceptable efficacy when combined with cotrimoxazole and isoniazid in clinical trials for the treatment of malaria, although parasitological cure rates were inferior [50]. However, the serum concentrations of anti-malarial drugs are 1–2 orders of magnitude lower than the MIC values for MTC. This disparity suggests that effective concentrations against MTC might only be achievable with significantly increased drug dosages.

It has been suggested that such MIC values could be achieved intracellularly [32]; however, the few studies that investigated this in macrophages report only modest effects [27, 31, 34, 42].

In-vitro studies show ambiguous results regarding the synergistic effects of anti-malarial and anti-TB drugs (Table 3, Additional file 1: Table S4). These effects are gauged using the fractional inhibitory concentration index (FICI) from a checkerboard assay, categorizing ≤ 0.5 as synergism, ≥ 4 as antagonism, and > 0.5—< 4 as indifferent or independent. Often, drugs considered synergistic barely meet the 0.5 FICI threshold (Table 4, Additional file 1: Table S4). Studies on dormant bacteria models (Additional file 1: Table S3) revealed similar MIC values for mefloquine and rifampicin at 7 µM [29], but this was primarily due to increased rifampicin MIC in dormant bacteria rather than its activity against active MTC. The clinical significance of these observations is unclear.

In assessing the activity of anti-malarial drugs in animal models for tuberculosis, the overall findings present a varied picture. Mefloquine and artemisinins, for example, demonstrated only moderate reductions in bacterial counts, ranging from 1.2 to 1.7 log CFU/mL and 2 to 3 log CFU/mL, respectively [33, 35]; while chloroquine use in mice and guinea pigs showed almost no impact on bacterial counts [39]. However, the addition of isoniazid to chloroquine led to considerable decreases in bacterial counts, sometimes resulting in sterilisation, along with reductions in histological lesion severity as well as relapse rates [39] (Tables 4, Additional file 1: Tabe S5).

Perhaps in line with this, the only identified clinical trial is an ongoing phase 1 trial which explores the safety of adding chloroquine to standard anti-TB medication in healthy volunteers (Additional file 1: Table S6).

Toxicity

The elevated MIC values of anti-malarial drugs against MTC, in contrast to achievable serum levels (Tables 3, 4), raise significant concerns about toxicity at higher dosages. The selectivity index (SI), calculated as the ratio of IC50% toxicity in cell assay to MIC, reveals low values for anti-malarials (single digits), suggesting a narrow therapeutic window, unlike the much higher values for rifampicin and isoniazid (based on values in Table 4). Consequently, serum concentrations approaching the MIC values for MTC could result in considerable toxicity, as evidenced by chloroquine toxicity studies in overdose, which conclude that death rates rise steeply at concentrations > 10 µmol/L [51].

It is important to note that dosages used in animal models for TB (Additional file 1: Table S5) are already at the higher end of what is recommended for malaria. Prolonged use of these drugs in TB treatment, which spans several months and involves patients with significant systemic infection, raises further safety concerns. Chloroquine and mefloquine have yielded an overall acceptable general safety profile in long-term malaria-chemoprophylactic use [52, 53], as well as chloroquine use in rheumatoid diseases [54]. However, specific long-term effects when added to other, potentially toxic TB treatment regimens must be carefully considered, as the case of prolonged use of linezolid illustrates [55].

Other implications and consequences

Repurposing antimicrobials for non-infectious diseases rises concerns about increasing antimicrobial resistance, though conclusive data are currently lacking [3]. The use of tetracyclines for malaria or linezolid and levofloxacin for TB, may have contributed to bacterial resistance; but a significant public health impact has not been reported so far. A key consideration in repurposing anti-malarials for TB treatment is the co-endemicity of malaria and TB, particularly in sub-Saharan Africa, where they affect overlapping populations. The prolonged use of anti-malarials for many months in TB treatment could potentially increase the risk of Plasmodium spp. parasites developing resistance. Certainly, this is of utmost importance for artemisinins. Contrary to this, chloroquine's ongoing current primary use against Plasmodium vivax (although there is shift towards applying ACT in the first place also against non-falciparum human-pathogenic Plasmodium) reduces its perceived risk in contributing to P. falciparum resistance. However, continued use could still foster resistance, also affecting the effectiveness of partner drugs in artemisinin-based combination therapy, such as mefloquine, lumefantrine, and amodiaquine, due to cross-resistance (markers: pfcrt and pfmdr1) [56]. Certainly, the long-term use of chloroquine in other situations, like in treating rheumatoid arthritis, affects only a relatively small and distinct overlapping population, and is unlikely to significantly influence Plasmodium resistance. Intriguingly, long-term chloroquine use seems to provide some limited protective effect against TB in rheumatoid patients undergoing immunosuppressive therapy [57, 58], although it was also associated with increased incidence of non-tuberculous mycobacterial infections [58].

Conclusions

The analysis has reviewed the possibility of repurposing anti-malarial drugs by leveraging their anti-mycobacterial properties for TB treatment. However, this potential appears to be limited by high MIC values, poor synergy, and minimal in vivo effects. Concerns about potential toxicity at effective dosages and the risk of antimicrobial resistance, especially where TB and malaria overlap, further question their repurposing. More importantly, it prompts a critical question: Is such a strategy both viable and beneficial? The co-endemicity of tuberculosis and malaria across many regions presents scenarios where co-infections are common, and using a drug to treat one condition may inadvertently influence the pathogen responsible for the other.

While the repurposing of medications like linezolid and fluoroquinolones has become integral to TB management, potentially impacting bacterial populations, the possible repercussions of such resistance appear to be manageable in public health terms, given the availability of alternative antibiotic classes and the possibly limited transmission of such bacteria. In contrast, the idea of adapting anti-malarials for TB treatment raises significant concerns due to the potential for inducing resistance in local parasite strains, thereby compromising the effectiveness of the limited arsenal of anti-malarial drugs.

Therefore, the clinical value of repurposing anti-malarials for TB is questionable, with scepticism regarding its ability to substantially alter TB treatment paradigms. In the light of these considerations, a cautious stance is advised, recommending against the systematic exploration of anti-malarials as TB treatments. The associated risks of resistance development, combined with limited chances of revolutionizing clinical practice, support this cautious approach. The pursuit of novel TB treatments should instead focus on strategies offering clear benefits and presenting a reduced risk of aggravating the global challenge of infectious diseases.

References

  1. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2018;18:41–58.

    Article  PubMed  Google Scholar 

  2. Jourdan JP, Bureau R, Rochais C, Dallemagne P. Drug repositioning: a brief overview. J Pharm Pharmacol. 2020;72:1145–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Talat A, Bashir Y, Khan AU. Repurposing of antibiotics: sense or non-sense. Front Pharmacol. 2022;13: 833005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Aguilar Diaz JM, Abulfathi AA, Te Brake LH, van Ingen J, Kuipers S, Magis-Escurra C, et al. New and repurposed drugs for the treatment of active tuberculosis: an update for clinicians. Respiration. 2023;102:83–100.

    Article  CAS  PubMed  Google Scholar 

  5. Urbina F, Puhl AC, Ekins S. Recent advances in drug repurposing using machine learning. Curr Opin Chem Biol. 2021;65:74–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Farha MA, Brown ED. Drug repurposing for antimicrobial discovery. Nat Microbiol. 2019;4:565–77.

    Article  CAS  PubMed  Google Scholar 

  7. Jampilek J. Drug repurposing to overcome microbial resistance. Drug Discov Today. 2022;27:2028–41.

    Article  CAS  PubMed  Google Scholar 

  8. Sharma K, Ahmed F, Sharma T, Grover A, Agarwal M, Grover S. Potential repurposed drug candidates for tuberculosis treatment: progress and update of drugs identified in over a decade. ACS Omega. 2023;8:17362–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Shahnawaz M, Safiya M, Zahoor A. Antituberculosis drug repurposing: a new hope for tackling multi-challenging TB in timely manner. In: Saxena SK (ed.). Drug Repurposing - Molecular Aspects and Therapeutic Applications. London IntechOpen; pp. 35–63. https://doi.org/10.5772/intechopen.95190.

  10. Grobusch MP, Del Portal R, Luyten C, Visser BJ, de Jong HK, Goorhuis A, et al. Overcoming publication and dissemination bias in infectious diseases clinical trials. Lancet Infect Dis. 2024;24:e189–95.

    Article  PubMed  Google Scholar 

  11. WHO. Global Tuberculosis Report 2022. Geneva, World Health Organization, 2022. https://www.who.int/teams/global-tuberculosis-programme/tb-reports/global-tuberculosis-report-2022 (accessed: 14/02/2024)

  12. Alsaad N, Wilffert B, van Altena R, de Lange WCM, van der Werf TS, Kosterink JGW, et al. Potential antimicrobial agents for the treatment of multidrug-resistant tuberculosis. Eur Respir J. 2014;43:884–97.

    Article  CAS  PubMed  Google Scholar 

  13. Lange C, Dheda K, Chesov D, Mandalakas AM, Udwadia Z, Horsburgh CR. Management of drug-resistant tuberculosis. Lancet. 2019;394:953–66.

    Article  CAS  PubMed  Google Scholar 

  14. Lange C, Barry CE, Horsburgh CR. Treatments of multidrug-resistant tuberculosis: light at the end of the tunnel. Am J Respir Crit Care Med. 2022;205:1142–4.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Thwaites G, Nguyen NV. Linezolid for drug-resistant tuberculosis. N Engl J Med. 2022;387:842–3.

    Article  CAS  PubMed  Google Scholar 

  16. Kamat S, Kumari M. Repurposing chloroquine against multiple diseases with special attention to SARS-CoV-2 and associated toxicity. Front Pharmacol. 2021;12: 576093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Al-Bari MA. Chloroquine analogues in drug discovery: new directions of uses, mechanisms of actions and toxic manifestations from malaria to multifarious diseases. J Antimicrob Chemother. 2015;70:1608–21.

    Article  PubMed  Google Scholar 

  18. Savarino A, Di Trani L, Donatelli I, Cauda R, Cassone A. New insights into the antiviral effects of chloroquine. Lancet Infect Dis. 2006;6:67–9.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Cohen HG, Reynolds TB. Comparison of metronidazole and chloroquine for the treatment of amoebic liver abscess. A controlled trial. Gastroenterology. 1975;69:35–41.

    Article  CAS  PubMed  Google Scholar 

  20. Pfarr KM, Krome AK, Al-Obaidi I, Batchelor H, Vaillant M, Hoerauf A, et al. The pipeline for drugs for control and elimination of neglected tropical diseases: 2. Oral anti-infective drugs and drug combinations for off- label use. Parasit Vectors. 2023;16:394.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Mandal AK, Abernathy T, Nelluri SN, Stitzel V. Is quinine effective and safe in leg cramps? J Clin Pharmacol. 1995;35:588–93.

    Article  CAS  PubMed  Google Scholar 

  22. Koga M, Suganuma A, Kikuchi T, Yoshimura Y, Shoji K, Kobayashi I, et al. Research Group on Chemotherapy of Tropical Diseases, Japan Primaquine plus clindamycin as a promising salvage therapy for Pneumocystis jirovecii pneumonia: a retrospective analysis in Japanese patients. J Infect Chemother. 2021;27:924–8.

    Article  CAS  PubMed  Google Scholar 

  23. Guan L, Wang H, Xu X, Fan H. Therapeutical utilization and repurposing of artemisinin and its derivatives: a narrative review. Adv Biol (Weinh). 2023;7: e2300086.

    Article  PubMed  Google Scholar 

  24. Cardoso NC, Oosthuizen CB, Peton N, Singh V. Drug Repurposing for Tuberculosis. In: Saxena SK (ed.). Drug Repurposing - Molecular Aspects and Therapeutic Applications. London IntechOpen; pp. 65–103. https://doi.org/10.5772/intechopen.95190.

  25. Huang Y, Yang Y, Liu G, Xu M, Hu D. New clinical application prospects of artemisinin and its derivatives: a scoping review. Infect Dis Poverty. 2023;12:115.

    Article  PubMed  PubMed Central  Google Scholar 

  26. dos Santos MC, Scaini JLR, Lopes MVC, Rodrigues BG, Silva NO, Borges CRL, et al. Mefloquine synergism with anti-tuberculosis drugs and correlation to membrane effects: biologic, spectroscopic and molecular dynamics simulations studies. Bioorg Chem. 2021;110: 104786. https://doi.org/10.1016/j.bioorg.2021.104786.

    Article  CAS  PubMed  Google Scholar 

  27. Crowle AJ, May MH. Inhibition of tubercle bacilli in cultured human macrophages by chloroquine used alone and in combination with streptomycin, isoniazid, pyrazinamide, and two metabolites of vitamin D3. Antimicrob Agents Chemother. 1990;34:2217–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sidrônio MGS, Castelo Branco APOT, Abbadi BL, Macchi F, Silveira MD, Lock A, et al. Effects of tafenoquine against active, dormant and resistant Mycobacterium tuberculosis. Tuberculosis. 2021;128:102089.

    Article  PubMed  Google Scholar 

  29. Jayaprakash S, Iso Y, Wan B, Franzblau SG, Kozikowski AP. Design, synthesis, and SAR studies of mefloquine-based ligands as potential antituberculosis agents. ChemMedChem. 2006;1:593–7.

    Article  CAS  PubMed  Google Scholar 

  30. Goncalves RSB, Kaiser CR, Loureno MCS, Bezerra FAFM, De Souza MVN, Wardell JL, et al. Mefloquine-oxazolidine derivatives, derived from mefloquine and arenecarbaldehydes: In vitro activity including against the multidrug-resistant tuberculosis strain T113. Bioorganic Med Chem. 2012;20:243–8.

    Article  CAS  Google Scholar 

  31. Bermudez LE, Meek L. Mefloquine and its enantiomers are active against Mycobacterium tuberculosis in vitro and in macrophages. Tuberc Res Treat. 2014;2014: 530815.

    PubMed  PubMed Central  Google Scholar 

  32. Krieger D, Vesenbeckh S, Schönfeld N, Bettermann G, Bauer TT, Rüssmann H, et al. Mefloquine as a potential drug against multidrug-resistant tuberculosis. Eur Respir J. 2015;46:1503–5.

    Article  PubMed  Google Scholar 

  33. Rodrigues-Junior VS, Villela AD, Gonçalves RSB, Abbadi BL, Trindade RV, López-Gavín A, et al. Mefloquine and its oxazolidine derivative compound are active against drug-resistant Mycobacterium tuberculosis strains and in a murine model of tuberculosis infection. Int J Antimicrob Agents. 2016;48:203–7.

    Article  CAS  PubMed  Google Scholar 

  34. Lougheed KEA, Taylor DL, Osborne SA, Bryans JS, Buxton RS. New anti-tuberculosis agents amongst known drugs. Tuberculosis (Edinb). 2009;89:364–70.

    Article  CAS  PubMed  Google Scholar 

  35. Choi WH. Novel pharmacological activity of artesunate and artemisinin: their potential as anti-tubercular agents. J Clin Med. 2017;10(6):30.

    Article  Google Scholar 

  36. Patel YS, Mistry N, Mehra S. Repurposing artemisinin as an anti-mycobacterial agent in synergy with rifampicin. Tuberculosis (Edinb). 2019;115:146–53.

    Article  CAS  PubMed  Google Scholar 

  37. Matt U, Selchow P, Dal Molin M, Strommer S, Sharif O, Schilcher K, et al. Chloroquine enhances the antimycobacterial activity of isoniazid and pyrazinamide by reversing inflammation-induced macrophage efflux. Int J Antimicrob Agents. 2017;50:55–62.

    Article  CAS  PubMed  Google Scholar 

  38. Danelishvili L, Shulzhenko N, Chinison JJJ, Babrak L, Hu J, Morgun A, et al. Mycobacterium tuberculosis proteome response to antituberculosis compounds reveals metabolic “escape” pathways that prolong bacterial survival. Antimicrob Agents Chemother. 2017;61:e00430-e517.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Mishra R, Kohli S, Malhotra N, Bandyopadhyay P, Mehta M, Munshi MH, et al. Targeting redox heterogeneity to counteract drug tolerance in replicating Mycobacterium tuberculosis. Sci Transl Med. 2019;11:6635.

    Article  Google Scholar 

  40. Mao J, Wang Y, Wan B, Kozikowski AP, Franzblau SG. Design, synthesis, and pharmacological evaluation of mefloquine-based ligands as novel antituberculosis agents. ChemMedChem. 2007;2:1624–30.

    Article  CAS  PubMed  Google Scholar 

  41. Cavanaugh JS, Jou R, Wu MH, Dalton T, Kurbatova E, Ershova J, et al. Susceptibilities of MDR Mycobacterium tuberculosis isolates to unconventional drugs compared with their reported pharmacokinetic/pharmacodynamic parameters. J Antimicrob Chemother. 2017;72:1678–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mori G, Orena BS, Franch C, Mitchenall LA, Godbole AA, Rodrigues L, et al. The EU approved antimalarial pyronaridine shows antitubercular activity and synergy with rifampicin, targeting RNA polymerase. Tuberculosis (Edinb). 2018;112:98–109.

    Article  CAS  PubMed  Google Scholar 

  43. Martini MC, Zhang T, Williams JT, Abramovitch RB, Weathers PJ, Shell SS. Artemisia annua and Artemisia afra extracts exhibit strong bactericidal activity against Mycobacterium tuberculosis. J Ethnopharmacol. 2020;262: 113191.

    Article  CAS  PubMed  Google Scholar 

  44. Zheng H, Colvin CJ, Johnson BK, Kirchhoff PD, Wilson M, Jorgensen-Muga K, et al. Inhibitors of Mycobacterium tuberculosis DosRST signaling and persistence. Nat Chem Biol. 2017;13:218–25.

    Article  CAS  PubMed  Google Scholar 

  45. Keri RS, Patil SA. Quinoline: a promising antitubercular target. Biomed Pharmacother. 2014;68:1161–75.

    Article  CAS  PubMed  Google Scholar 

  46. Rolain J-M, Colson P, Raoult D. Recycling of chloroquine and its hydroxyl analogue to face bacterial, fungal and viral infections in the 21st century. Int J Antimicrob Agents. 2007;30:297–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Palomino JC, Martin A. Is repositioning of drugs a viable alternative in the treatment of tuberculosis? J Antimicrob Chemother. 2013;68:275–83.

    Article  CAS  PubMed  Google Scholar 

  48. An Q, Li C, Chen Y, Deng Y, Yang T, Luo Y. Repurposed drug candidates for antituberculosis therapy. Eur J Med Chem. 2020;192: 112175.

    Article  CAS  PubMed  Google Scholar 

  49. Kontsevaya I, Werngren J, Holicka Y, Klaos K, Tran A, Nikolayevskyy V. Non-commercial phenotypic assays for the detection of Mycobacterium tuberculosis drug resistance: a systematic review. Eur J Clin Microbiol Infect Dis. 2020;39:415–26.

    Article  CAS  PubMed  Google Scholar 

  50. Genton B, Mueller I, Betuela I, Casey G, Ginny M, Alpers MP, et al. Rifampicin/Cotrimoxazole/Isoniazid versus mefloquine or quinine + sulfadoxine- pyrimethamine for malaria: a randomized trial. PLoS Clin Trials. 2006;1: e38.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Watson JA, Tarning J, Hoglund RM, Baud FJ, Megarbane B, Clemessy JL, et al. Concentration-dependent mortality of chloroquine in overdose. Elife. 2020;9: e58631.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. AlKadi HO. Antimalarial drug toxicity: a review. Chemotherapy. 2007;53:385–91.

    Article  CAS  PubMed  Google Scholar 

  53. Schlagenhauf P, Adamcova M, Regep L, Schaerer MT, Rhein HG. The position of mefloquine as a 21st century malaria chemoprophylaxis. Malar J. 2010;9:357.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Gisondi P, Piaserico S, Bordin C, Bellinato F, Tozzi F, Alaibac M, et al. The safety profile of hydroxychloroquine: major cutaneous and extracutaneous adverse events. Clin Exp Rheumatol. 2021;39:1099–107.

    Article  PubMed  Google Scholar 

  55. Veerman K, Goosen J, Spijkers K, Jager N, Heesterbeek P, Telgt D. Prolonged use of linezolid in bone and joint infections: a retrospective analysis of adverse effects. J Antimicrob Chemother. 2023;78:2660–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Haldar K, Bhattacharjee S, Safeukui I. Drug resistance in Plasmodium. Nat Rev Microbiol. 2018;16:156–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Lim CH, Chen HH, Chen YH, Chen DY, Huang WN, Tsai JJ, et al. The risk of tuberculosis disease in rheumatoid arthritis patients on biologics and targeted therapy: a 15-year real world experience in Taiwan. PLoS ONE. 2017;12: e0178035.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Brode SK, Jamieson FB, Ng R, Campitelli MA, Kwong JC, Paterson JM, et al. Increased risk of mycobacterial infections associated with anti-rheumatic medications. Thorax. 2015;70:677–82.

    Article  PubMed  Google Scholar 

Download references

Funding

No funding received.

Author information

Authors and Affiliations

Authors

Contributions

MPG and TH conceived the paper. CRdPL, TH, SMH and MPG contributed to data analysis and the writing of the first draft. All authors contributed to the final version of the manuscript and endorsed its submission.

Corresponding author

Correspondence to Martin P. Grobusch.

Ethics declarations

Competing interests

All authors declare no conflicts of interest related to this study.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1.

The additional file includes a detailed search strategy and comprehensive tables summarizing the results from all included studies.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hanscheid, T., del Portal Luyten, C.R., Hermans, S.M. et al. Repurposing of anti-malarial drugs for the treatment of tuberculosis: realistic strategy or fanciful dead end?. Malar J 23, 132 (2024). https://doi.org/10.1186/s12936-024-04967-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12936-024-04967-2

Keywords