Identification of new drug candidates against Borrelia burgdorferi using high-throughput screening

Venkata Raveendra Pothineni, Dhananjay Wagh, Mustafeez Mujtaba Babar, Mohammed Inayathullah, David Solow-Cordero, Kwang-Min Kim, Aneesh V Samineni, Mansi B Parekh, Lobat Tayebi, Jayakumar Rajadas, Venkata Raveendra Pothineni, Dhananjay Wagh, Mustafeez Mujtaba Babar, Mohammed Inayathullah, David Solow-Cordero, Kwang-Min Kim, Aneesh V Samineni, Mansi B Parekh, Lobat Tayebi, Jayakumar Rajadas

Abstract

Lyme disease is the most common zoonotic bacterial disease in North America. It is estimated that >300,000 cases per annum are reported in USA alone. A total of 10%-20% of patients who have been treated with antibiotic therapy report the recrudescence of symptoms, such as muscle and joint pain, psychosocial and cognitive difficulties, and generalized fatigue. This condition is referred to as posttreatment Lyme disease syndrome. While there is no evidence for the presence of viable infectious organisms in individuals with posttreatment Lyme disease syndrome, some researchers found surviving Borrelia burgdorferi population in rodents and primates even after antibiotic treatment. Although such observations need more ratification, there is unmet need for developing the therapeutic agents that focus on removing the persisting bacterial form of B. burgdorferi in rodent and nonhuman primates. For this purpose, high-throughput screening was done using BacTiter-Glo assay for four compound libraries to identify candidates that stop the growth of B. burgdorferi in vitro. The four chemical libraries containing 4,366 compounds (80% Food and Drug Administration [FDA] approved) that were screened are Library of Pharmacologically Active Compounds (LOPAC1280), the National Institutes of Health Clinical Collection, the Microsource Spectrum, and the Biomol FDA. We subsequently identified 150 unique compounds, which inhibited >90% of B. burgdorferi growth at a concentration of <25 µM. These 150 unique compounds comprise many safe antibiotics, chemical compounds, and also small molecules from plant sources. Of the 150 unique compounds, 101 compounds are FDA approved. We selected the top 20 FDA-approved molecules based on safety and potency and studied their minimum inhibitory concentration and minimum bactericidal concentration. The promising safe FDA-approved candidates that show low minimum inhibitory concentration and minimum bactericidal concentration values can be chosen as lead molecules for further advanced studies.

Keywords: BacTiter-Glo assay; Borrelia burgdorferi; Lyme disease; high-throughput screening; persisters.

Figures

Figure 1
Figure 1
Inhibition assay of drugs on CA8 strain. Notes: Effect of drugs on Borrelia cell viability was studied with drugs: (A) tetraethylthiuram disulfide, doxorubicin hydrochloride, and epirubicin hydrochloride and (B) azlocillin sodium and cephalothin sodium. The control has no drugs. The results represent mean ± SD. Abbreviation: SD, standard deviation.
Figure 2
Figure 2
Time kill curves for B. burgdorferi s.s. isolates CA8 with (A) azlocillin sodium and (B) cefotaxime acid. Notes: The Borrelia was grown in the drug concentrations of 0.625 µM, 1.25 µM, 2.5 µM, and 5 µM. Experiment was performed with triplicates by the investigation of growth using conventional cell counts, and data were reported as the mean of triplicate. The control has no drugs. Abbreviation:B. burgdorferi, Borrelia burgdorferi.

References

    1. Kuehn BM. CDC estimates 300,000 US cases of Lyme disease annually. JAMA. 2013;310:1110.
    1. Vollmer SA, Feil EJ, Chu CY, et al. Spatial spread and demographic expansion of Lyme borreliosis spirochaetes in Eurasia. Infect Genet Evol. 2013;14:147–155.
    1. Brownstein JS, Holford TR, Fish D. Effect of climate change on Lyme disease risk in North America. Ecohealth. 2005;2:38–46.
    1. Esposito S, Bosis S, Sabatini C, Tagliaferri L, Principi N. Borrelia burgdorferi infection and Lyme disease in children. Int J Infect Dis. 2013;17:e153–e158.
    1. Borchers AT, Keen CL, Huntley AC, Gershwin ME. Lyme disease: a rigorous review of diagnostic criteria and treatment. J Autoimmun. 2015;57:82–115.
    1. Bockenstedt LK, Radolf JD. Xenodiagnosis for posttreatment Lyme disease syndrome: resolving the conundrum or adding to it? Clin Infect Dis. 2014;58:946–948.
    1. Feng J, Wang T, Shi W, et al. Identification of novel activity against Borrelia burgdorferi persisters using an FDA approved drug library. Emerg Microbes Infect. 2014;3:e49.
    1. Bockenstedt LK, Gonzalez DG, Haberman AM, Belperron AA. Spirochete antigens persist near cartilage after murine Lyme borreliosis therapy. J Clin Invest. 2012;122:2652–2660.
    1. Hodzic E, Feng S, Holden K, Freet KJ, Barthold SW. Persistence of Borrelia burgdorferi following antibiotic treatment in mice. Antimicrob Agents Chemother. 2008;52:1728–1736.
    1. Diterich I, Rauter C, Kirschning CJ, Hartung T. Borrelia burgdorferi-induced tolerance as a model of persistence via immunosuppression. Infect Immun. 2003;71:3979–3987.
    1. Steere AC, Schoen RT, Taylor E. The clinical evolution of Lyme arthritis. Ann Intern Med. 1987;107:725–731.
    1. Barthold SW, de Souza MS, Janotka JL, Smith AL, Persing DH. Chronic Lyme borreliosis in the laboratory mouse. Am J Pathol. 1993;143:959–971.
    1. Babady NE, Hall L, Abbenyi AT, et al. Evaluation of Mycobacterium avium complex clarithromycin susceptibility testing using SLOMYCO Sensititre panels and JustOne strips. J Clin Microbiol. 2010;48:1749–1752.
    1. Embers ME, Barthold SW, Borda JT, et al. Persistence of Borrelia burgdorferi in rhesus macaques following antibiotic treatment of disseminated infection. PLoS One. 2012;7:e29914.
    1. Hodzic E, Imai D, Feng S, Barthold SW. Resurgence of persisting non-cultivable Borrelia burgdorferi following antibiotic treatment in mice. PLoS One. 2014;9:e86907.
    1. Straubinger RK, Summers BA, Chang YF, Appel MJ. Persistence of Borrelia burgdorferi in experimentally infected dogs after antibiotic treatment. J Clin Microbiol. 1997;35:111–116.
    1. Marques A, Telford SR, 3rd, Turk SP, et al. Xenodiagnosis to detect Borrelia burgdorferi infection: a first-in-human study. Clin Infect Dis. 2014;58:937–945.
    1. Brorson Ø, Brorson SH, Scythes J, MacAllister J, Wier A, Margulis L. Destruction of spirochete Borrelia burgdorferi round-body propagules (RBs) by the antibiotic tigecycline. Proc Natl Acad Sci U S A. 2009;106:18656–18661.
    1. Miklossy J, Kasas S, Zurn AD, McCall S, Yu S, McGeer PL. Persisting atypical and cystic forms of Borrelia burgdorferi and local inflammation in Lyme neuroborreliosis. J Neuroinflammation. 2008;5:1–18.
    1. Lantos PM, Auwaerter PG, Wormser GP. A systematic review of Borrelia burgdorferi morphologic variants does not support a role in chronic Lyme disease. Clin Infect Dis. 2014;58:663–671.
    1. Sharma B, Brown AV, Matluck NE, Hu LT, Lewis K. Borrelia burgdorferi, the causative agent of Lyme disease, forms drug-tolerant persister cells. Antimicrob Agents Chemother. 2015;59:4616–4624.
    1. Feng J, Auwaerter PG, Zhang Y. Drug combinations against Borrelia burgdorferi persisters in vitro: eradication achieved by using daptomycin, cefoperazone and doxycycline. PLoS One. 2015;10:e0117207.
    1. Sapi E, Kaur N, Anyanwu S, et al. Evaluation of in-vitro antibiotic susceptibility of different morphological forms of Borrelia burgdorferi. Infect Drug Resist. 2011;4:97–113.
    1. Pavia C, Inchiosa MA, Jr, Wormser GP. Efficacy of short-course ceftriaxone therapy for Borrelia burgdorferi infection in C3H mice. Antimicrob Agents Chemother. 2002;46:132–134.
    1. Wormser GP, Daniels TJ, Bittker S, Cooper D, Wang G, Pavia CS. Failure of topical antibiotics to prevent disseminated Borrelia burgdorferi infection following a tick bite in C3H/HeJ mice. J Infect Dis. 2012;205:991–994.
    1. Pavia CS, Wormser GP. Culture of the entire mouse to determine whether cultivable Borrelia burgdorferi persists in infected mice treated with a five-day course of Ceftriaxone. Antimicrob Agents Chemother. 2014;58:6701–6703.
    1. Lefas G, Chaconas G. High-throughput screening identifies three inhibitor classes of the telomere resolvase from the Lyme disease spirochete. Antimicrob Agents Chemother. 2009;53:4441–4449.
    1. Cornell KA, Primus S, Martinez JA, Parveen N. Assessment of methylthioadenosine/S-adenosylhomocysteine nucleosidases of Borrelia burgdorferi as targets for novel antimicrobials using a novel high-throughput method. J Antimicrob Chemother. 2009;63:1163–1172.
    1. Feng J, Shi W, Zhang S, Zhang Y. Identification of new compounds with high activity against stationary phase Borrelia burgdorferi from the NCI compound collection. Emerg Microbes Infect. 2015;4:e31.
    1. Feng J, Weitner M, Shi W, Zhang S, Sullivan D, Zhang Y. Identification of additional anti-persister activity against Borrelia burgdorferi from an FDA drug library. Antibiotics. 2015;4:397.
    1. Nygren P, Larsson R. Drug repositioning from bench to bedside: tumour remission by the antihelmintic drug mebendazole in refractory metastatic colon cancer. Acta Oncol. 2014;53:427–428.
    1. Jahchan NS, Dudley JT, Mazur PK, et al. A drug repositioning approach identifies tricyclic antidepressants as inhibitors of small cell lung cancer and other neuroendocrine tumors. Cancer Discov. 2013;3:1364–1377.
    1. Wagh D, Pothineni VR, Inayathullah M, Liu S, Kim KM, Rajadas J. Borreliacidal activity of Borrelia metal transporter A (BmtA) binding small molecules by manganese transport inhibition. Drug Des Devel Ther. 2015;9:805.
    1. Kraiczy P, Weigand J, Wichelhaus TA, et al. In vitro activities of fluoroquinolones against the spirochete Borrelia burgdorferi. Antimicrob Agents Chemother. 2001;45:2486–2494.
    1. Hunfeld KP, Rodel R, Wichelhaus TA. In vitro activity of eight oral cephalosporins against Borrelia burgdorferi. Int J Antimicrob Agents. 2003;21:313–318.
    1. Veinovic G, Cerar T, Strle F, et al. In vitro susceptibility of European human Borrelia burgdorferi sensu stricto strains to antimicrobial agents. Int J Antimicrob Agents. 2013;41:288–291.
    1. Feng J, Wang T, Zhang S, Shi W, Zhang Y. An optimized SYBR Green I/PI assay for rapid viability assessment and antibiotic susceptibility testing for Borrelia burgdorferi. PLoS One. 2014;9:e111809.
    1. Feng J, Wang T, Shi W, et al. Identification of novel activity against Borrelia burgdorferi persisters using an FDA approved drug library. Emerg Microbes Infect. 2014;3:e49.
    1. Marques A. Chronic Lyme disease: a review. Infect Dis Clin North Am. 2008;22:341–360. vii–viii.
    1. Terekhova D, Sartakova ML, Wormser GP, Schwartz I, Cabello FC. Erythromycin resistance in Borrelia burgdorferi. Antimicrob Agents Chemother. 2002;46:3637–3640.
    1. Kannan K, Kanabar P, Schryer D, et al. The general mode of translation inhibition by macrolide antibiotics. Proc Natl Acad Sci U S A. 2014;111:15958–15963.
    1. Steel HC, Theron AJ, Cockeran R, Anderson R, Feldman C. Pathogen-and host-directed anti-inflammatory activities of macrolide antibiotics. Mediators Inflamm. 2012;2012:584262.
    1. Shinkai M, Henke MO, Rubin BK. Macrolide antibiotics as immunomodulatory medications: proposed mechanisms of action. Pharmacol Ther. 2008;117:393–405.
    1. Leach KL, Brickner SJ, Noe MC, Miller PF. Linezolid, the first oxazolidinone antibacterial agent. Ann N Y Acad Sci. 2011;1222:49–54.
    1. Van Bambeke F. Macrolides and ketolides. In: Vinks A, Derendorf H, Mouton JW, editors. Fundamentals of Antimicrobial Pharmacokinetics and Pharmacodynamics. New York, NY: Springer; 2014. pp. 257–278.
    1. Yuan-shu Q, Qi-nan W, Yu-fu J. Pharmacokinetic study of leucomycin in healthy volunteers [J] Zhongguo Kang Sheng Su Za Zhi. 1990;2:008.
    1. Lin AH, Murray RW, Vidmar TJ, Marotti KR. The oxazolidinone eperezolid binds to the 50S ribosomal subunit and competes with binding of chloramphenicol and lincomycin. Antimicrob Agents Chemother. 1997;41:2127–2131.
    1. Dryden MS. Linezolid pharmacokinetics and pharmacodynamics in clinical treatment. J Antimicrob Chemother. 2011;66(Suppl 4):iv7–iv15.
    1. Keel RA, Schaeftlein A, Kloft C, et al. Pharmacokinetics of intravenous and oral linezolid in adults with cystic fibrosis. Antimicrob Agents Chemother. 2011;55:3393–3398.
    1. Viaggi B, Paolo AD, Danesi R, et al. Linezolid in the central nervous system: comparison between cerebrospinal fluid and plasma pharmacokinetics. Scand J Infect Dis. 2011;43:721–727.
    1. Kutscha-Lissberg F, Hebler U, Muhr G, Köller M. Linezolid penetration into bone and joint tissues infected with methicillin-resistant staphylococci. Antimicrob Agents Chemother. 2003;47:3964–3966.
    1. Pérez-Arnaiz C, Busto N, Leal JM, García B. New insights into the mechanism of the DNA/doxorubicin interaction. J Phys Chem B. 2014;118:1288–1295.
    1. Kizek R, Adam V, Hrabeta J, et al. Anthracyclines and ellipticines as DNA-damaging anticancer drugs: recent advances. Pharmacol Ther. 2012;133:26–39.
    1. Danesi R, Fogli S, Gennari A, Conte P, Del Tacca M. Pharmacokinetic-pharmacodynamic relationships of the anthracycline anticancer drugs. Clin Pharmacokinet. 2002;41:431–444.
    1. Pucci MJ, Wiles JA. Antimicrobials. New York, NY: Springer; 2014. Bacterial Topoisomerase Inhibitors: Quinolones and Beyond; pp. 307–326.
    1. Stass H, Kubitza D. Pharmacokinetics and elimination of moxifloxacin after oral and intravenous administration in man. J Antimicrob Chemother. 1999;43(Suppl B):83–90.
    1. Malik M, Drlica K. Moxifloxacin lethality against Mycobacterium tuberculosis in the presence and absence of chloramphenicol. Antimicrob Agents Chemother. 2006;50:2842–2844.
    1. Niki Y. Pharmacokinetics and safety assessment of tosufloxacin tosilate. J Infect Chemother. 2002;8:1–18.
    1. Fernandes R, Amador P, Prudêncio C. β-Lactams: chemical structure, mode of action and mechanisms of resistance. Rev Med Microbiol. 2013;24:7–17.
    1. Guay DR. Cefdinir: an expanded-spectrum oral cephalosporin. Ann Pharmacother. 2000;34:1469–1477.
    1. Berezin SK. Valinomycin as a classical anionophore: mechanism and ion selectivity. J Membr Biol. 2015;248:713–726.
    1. Zaldívar-Machorro VJ, López-Ortiz M, Demare P, Regla I, Muñoz-Clares RA. The disulfiram metabolites S-methyl-N,N- diethyldithiocarbamoyl sulfoxide and S-methyl-N,N-diethylthiocarbamoyl sulfone irreversibly inactivate betaine aldehyde dehydrogenase from Pseudomonas aeruginosa, both in vitro and in situ, and arrest bacterial growth. Biochimie. 2011;93:286–295.

Source: PubMed

3
Subskrybuj