We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Overcoming drug resistance for macro parasites

    Mrigank Srivastava

    Division of Parasitology, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, UP, India

    Academy of Scientific & Innovative Research (AcSIR), New Delhi 110025, India

    &
    Shailja Misra-Bhattacharya

    *Author for correspondence:

    E-mail Address: shailja_bhattacharya@cdri.res.in

    Division of Parasitology, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226031, UP, India

    Academy of Scientific & Innovative Research (AcSIR), New Delhi 110025, India

    Published Online:https://doi.org/10.2217/fmb.15.73

    Helminth infections impose burden on human and livestock populations, and their control predominantly relies on periodic mass administration of anthelmintic drugs. However, recent emergence of drug resistance among parasites to currently available drugs raises serious problems for continuation of control strategies and achievement of elimination of parasitic diseases. This review discusses the problem of anthelmintic resistance in humans and livestock, and suggests steps that can be taken to overcome this problem. To achieve the goals of morbidity reduction or elimination of infection we need to develop novel tools, including more efficacious drugs, vaccines and/or antivectorial agents; new diagnostics for infection and assessment of drug efficacy; and markers for possible anthelmintic resistance. Harnessing the knowledge generated from sequencing of parasite genome sequences is the key to identify genes responsible for drug resistance, which can be used as a starting point for discovery of target-specific pharmacological or genetic modulation to test the functional importance of individual genes and pathways. Involvement of chemical genetic screens and Caenorhabditis elegans as a model system for drug discovery needs to be explored in greater detail. Collective effort from several quarters is needed to think of a world that is free of parasitic infections.

    References

    • 1 Hotez PJ, Brindley PJ, Bethony JM, King CH, Pearce EJ, Jacobson J. Helminth infections: the great neglected tropical diseases. J. Clin. Invest. 118(4), 1311–1321 (2008).
    • 2 Molyneux DH, Hotez PJ, Fenwick A. ‘Rapid-impact interventions’: how a policy of integrated control for Africa's neglected tropical diseases could benefit the poor. PLoS Med. 2(11), e336 (2005).
    • 3 Martin RJ. Modes of action of anthelmintic drugs. Vet. J. 154(1), 11–34 (1997).
    • 4 James CE, Hudson AL, Davey MW. Drug resistance mechanisms in helminths: is it survival of the fittest? Trends Parasitol. 25(7), 328–335 (2009).
    • 5 Kasinathan RS, Greenberg RM. Pharmacology and potential physiological significance of schistosome multidrug resistance transporters. Exp. Parasitol. 132(1), 2–6 (2012).
    • 6 Von Samson-Himmelstjerna G, Blackhall WJ, Mccarthy JS, Skuce PJ. Single nucleotide polymorphism (SNP) markers for benzimidazole resistance in veterinary nematodes. Parasitology 134(Pt 8), 1077–1086 (2007).
    • 7 Wolstenholme AJ, Kaplan RM. Resistance to macrocyclic lactones. Curr. Pharm. Biotechnol. 13(6), 873–887 (2012).
    • 8 Molento MB, Nielsen MK, Kaplan RM. Resistance to avermectin/milbemycin anthelmintics in equine cyathostomins – current situation. Vet. Parasitol. 185(1), 16–24 (2012).
    • 9 Martin RJ, Robertson AP, Buxton SK, Beech RN, Charvet CL, Neveu C. Levamisole receptors: a second awakening. Trends Parasitol. 28(7), 289–296 (2012).
    • 10 Kasinathan RS, Morgan WM, Greenberg RM. Genetic knockdown and pharmacological inhibition of parasite multidrug resistance transporters disrupts egg production in Schistosoma mansoni. PLoS Negl. Trop. Dis. 5(12), e1425 (2011).
    • 11 James CE, Davey MW. Increased expression of ABC transport proteins is associated with ivermectin resistance in the model nematode Caenorhabditis elegans. Int. J. Parasitol. 39(2), 213–220 (2009).
    • 12 Papadopoulos E, Gallidis E, Ptochos S. Anthelmintic resistance in sheep in Europe: a selected review. Vet. Parasitol. 189(1), 85–88 (2012).
    • 13 Choe KP, Leung CK, Miyamoto MM. Unique structure and regulation of the nematode detoxification gene regulator, SKN-1: implications to understanding and controlling drug resistance. Drug Metab. Rev. 44(3), 209–223 (2012).
    • 14 Keiser J, Utzinger J. Efficacy of current drugs against soil-transmitted helminth infections: systematic review and meta-analysis. JAMA 299(16), 1937–1948 (2008).
    • 15 Hooper PJ, Bradley MH, Biswas G, Ottesen EA. The Global Programme to Eliminate Lymphatic Filariasis: health impact during its first 8 years (2000–2007). Ann. Trop. Med. Parasitol. 103(Suppl. 1), S17–S21 (2009).
    • 16 Reynoldson JA, Behnke JM, Pallant LJ et al. Failure of pyrantel in treatment of human hookworm infections (Ancylostoma duodenale) in the Kimberley region of north west Australia. Acta Trop. 68(3), 301–312 (1997).
    • 17 Humphries D, Nguyen S, Boakye D, Wilson M, Cappello M. The promise and pitfalls of mass drug administration to control intestinal helminth infections. Curr. Opin. Infect. Dis. 25(5), 584–589 (2012).
    • 18 Prichard RK, Basanez MG, Boatin BA et al. A research agenda for helminth diseases of humans: intervention for control and elimination. PLoS Negl. Trop. Dis. 6(4), e1549 (2012).
    • 19 Ghedin E, Wang S, Spiro D et al. Draft genome of the filarial nematode parasite Brugia malayi. Science 317(5845), 1756–1760 (2007).
    • 20 Sommer RJ, Streit A. Comparative genetics and genomics of nematodes: genome structure, development, and lifestyle. Annu. Rev. Genet. 45, 1–20 (2011).
    • 21 Lespine A, Alvinerie M, Vercruysse J, Prichard RK, Geldhof P. ABC transporter modulation: a strategy to enhance the activity of macrocyclic lactone anthelmintics. Trends Parasitol. 24(7), 293–298 (2008).
    • 22 Taylor CM, Wang Q, Rosa BA et al. Discovery of anthelmintic drug targets and drugs using chokepoints in nematode metabolic pathways. PLoS Pathog. 9(8), e1003505 (2013).
    • 23 Liu K, Tian J, Xiang M, Liu X. How carnivorous fungi use three-celled constricting rings to trap nematodes. Protein Cell 3(5), 325–328 (2012).
    • 24 Harder A, Von Samson-Himmelstjerna G. Cyclooctadepsipeptides – a new class of anthelmintically active compounds. Parasitol. Res. 88(6), 481–488 (2002).
    • 25 Jones AK, Buckingham SD, Sattelle DB. Chemistry-to-gene screens in Caenorhabditis elegans. Nat. Rev. Drug Discov. 4(4), 321–330 (2005).