Repurposing of the anti-malaria drug chloroquine for Zika Virus treatment and prophylaxis

Sergey A Shiryaev, Pinar Mesci, Antonella Pinto, Isabella Fernandes, Nicholas Sheets, Sujan Shresta, Chen Farhy, Chun-Teng Huang, Alex Y Strongin, Alysson R Muotri, Alexey V Terskikh, Sergey A Shiryaev, Pinar Mesci, Antonella Pinto, Isabella Fernandes, Nicholas Sheets, Sujan Shresta, Chen Farhy, Chun-Teng Huang, Alex Y Strongin, Alysson R Muotri, Alexey V Terskikh

Abstract

One of the major challenges of the current Zika virus (ZIKV) epidemic is to prevent congenital foetal abnormalities, including microcephaly, following ZIKV infection of pregnant women. Given the urgent need for ZIKV prophylaxis and treatment, repurposing of approved drugs appears to be a viable and immediate solution. We demonstrate that the common anti-malaria drug chloroquine (CQ) extends the lifespan of ZIKV-infected interferon signalling-deficient AG129 mice. However, the severity of ZIKV infection in these mice precludes the study of foetal (vertical) viral transmission. Here, we show that interferon signalling-competent SJL mice support chronic ZIKV infection. Infected dams and sires are both able to transmit ZIKV to the offspring, making this an ideal model for in vivo validation of compounds shown to suppress ZIKV in cell culture. Administration of CQ to ZIKV-infected pregnant SJL mice during mid-late gestation significantly attenuated vertical transmission, reducing the ZIKV load in the foetal brain more than 20-fold. Given the limited side effects of CQ, its lack of contraindications in pregnant women, and its worldwide availability and low cost, we suggest that CQ could be considered for the treatment and prophylaxis of ZIKV.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
CQ inhibits ZIKV infection and apoptosis in human neurospheres. (ad) Human iPSC-derived neurospheres were infected with ZIKVBR (multiplicity of infection = 1) and immediately treated with CQ at 5 µM, 20 µM, or 40 µM. MOCK = uninfected neurospheres. DMSO = DMSO treated neurospheres. Quantification of (a) neurosphere size at 96 h post-infection, (b) ZIKV-positive cells, (c) cleaved caspase 3 (CC3)-positive cells, and (d) CC3 and ZIKV double-positive cells. Data are the mean ± SEM of triplicates. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 compared with “ZIKV” one-way ANOVA with Dunnett’s multiple comparisons test.
Figure 2
Figure 2
CQ attenuates ZIKV infection in AG129 mice. (a) Survival curves for control and CQ- treated mice (n = 6 and 5, respectively). Note that 60% of the CQ-treated mice were euthanized on day 15. p = 0.0075 by log-rank Mantel–Cox test. (b) Weight loss following ZIKV infection in control and CQ-treated mice. Data are the mean ± SEM. *p < 0.05, **p < 0.01 compared with controls by unpaired t-test with Welch’s correction. (c,d) Visual appearance health scores in ZIKV-infected control (c) and CQ-treated (d) mice. Note that by day 13, all control mice had died but 60% of CQ-treated mice were alive.
Figure 3
Figure 3
Mouse model of chronic ZIKV infection for testing therapeutic interventions. (a) Female (n = 5) and (b) male (n = 8) SJL mice were infected with 108 PFU ZIKVBR retro-orbitally. Blood samples were taken every 3 days for 50 days and analysed for viral RNA by qRT-PCR. (c) qRT-PCR analysis of viral RNA in the testes of male mice 3 months after ZIKVBR infection. Mean ± SEM of 8 mice.
Figure 4
Figure 4
Horizontal and vertical sexual transmission of ZIKV in SJL mice. (a) Efficient sexual transmission of ZIKVBR was observed from infected males to uninfected females, but not from infected females to uninfected males. SJL male and female mice (n = 5) were infected with ZIKVBR (108 PFU retro-orbitally) and co-housed for 14 days with uninfected females and males, respectively. Viral RNA was measured by qRT-PCR of serum samples obtained from the sires and dams on day 14. Each point represents one animal. Data are the mean ± SD. n.d.: not detected (b) Vertical transmission of ZIKVBR to the offspring of infected sires and dams. RNA from the newborn heads was prepared on postnatal day 1 for qRT-PCR analysis of viral RNA. Each point represents one day 1 pup. Data are the mean ± SEM of the offspring of 4 infected dams/uninfected sires or 4 infected sires/uninfected dams as indicated.
Figure 5
Figure 5
CQ represses ZIKV infection in SJL mice and reduces vertical transmission. (a) Schematic of the experimental design. SJL dams were infected with ZIKV (2 × 105 PFU) on E12.5. On E13.5, they were randomly assigned to receive vehicle or CQ (30 mg/kg/day) in the drinking water. On E18.5, mice were euthanized for collection of blood and foetuses. (b) qRT-PCR of viral RNA. Each point represents one animal. Data are the mean ± SD of 3 (vehicle-treated) or 5 (CQ-treated) mice. *p < 0.05 by Student’s t-test. (c) qRT-PCR of viral RNA in foetal head extracts. Each point represents one foetus. Data are the mean ± SD of 8 foetuses pooled from 3 independent litters (control) or 5 foetuses pooled from 2 independent litters (CQ). *p < 0.05 by Student’s t-test. (d) Representative images of foetal brain sections from control, ZIKV-infected, and ZIKV-infected/CQ-treated mice on E18.5. Sections were stained with a primary antibody against Flavivirus Group Antigen (brown) and counterstained with Mayer’s hematoxylin (blue). Scale bar, 4 mm. (e) Quantification of ZIKV-infected cells in foetal brain sections from control, ZIKV-infected, and ZIKV-infected/CQ-treated mice. Data are the mean ± SEM of 6 sections per condition (3 embryos, 2 sections per embryo). *p < 0.05 compared with untreated ZIKV-infected mice by one-way ANOVA with Dunnett’s multiple comparisons test.

References

    1. Campos GS, Bandeira AC, Sardi SI. Zika Virus Outbreak, Bahia, Brazil. Emerg Infect Dis. 2015;21:1885–1886. doi: 10.3201/eid2110.150847.
    1. Castro LA, et al. Assessing real-time Zika risk in the United States. BMC Infect Dis. 2017;17:284. doi: 10.1186/s12879-017-2394-9.
    1. Azevedo RS, et al. Zika virus epidemic in Brazil. I. Fatal disease in adults: Clinical and laboratorial aspects. J Clin Virol. 2016;85:56–64. doi: 10.1016/j.jcv.2016.10.024.
    1. Cugola FR, et al. The Brazilian Zika virus strain causes birth defects in experimental models. Nature. 2016;534:267–271.
    1. Li, C. et al. Zika Virus Disrupts Neural Progenitor Development and Leads to Microcephaly in Mice. Cell Stem Cell, 10.1016/j.stem.2016.04.017 (2016).
    1. Mlakar J, et al. Zika Virus Associated with Microcephaly. N Engl J Med. 2016;374:951–958. doi: 10.1056/NEJMoa1600651.
    1. Melo AS, et al. Congenital Zika Virus Infection: Beyond Neonatal Microcephaly. JAMA Neurol. 2016;73:1407–1416. doi: 10.1001/jamaneurol.2016.3720.
    1. Cragan JD, et al. Baseline Prevalence of Birth Defects Associated with Congenital Zika Virus Infection - Massachusetts, North Carolina, and Atlanta, Georgia, 2013-2014. MMWR Morb Mortal Wkly Rep. 2017;66:219–222. doi: 10.15585/mmwr.mm6608a4.
    1. Beckham, J. D., Pastula, D. M., Massey, A. & Tyler, K. L. Zika Virus as an Emerging Global Pathogen: Neurological Complications of Zika Virus. JAMA Neurol, 10.1001/jamaneurol.2016.0800 (2016).
    1. Dirlikov E, et al. Guillain-Barré Syndrome During Ongoing Zika Virus Transmission - Puerto Rico, January 1-July 31, 2016. MMWR Morb Mortal Wkly Rep. 2016;65:910–914. doi: 10.15585/mmwr.mm6534e1.
    1. Wolfe MS, Cordero JF. Br Med J (Clin ResEd) 1985. Safety of chloroquine in chemosuppression of malaria during pregnancy; pp. 1466–1467.
    1. Delvecchio, R. et al. Chloroquine, an Endocytosis Blocking Agent, Inhibits Zika Virus Infection in Different Cell Models. Viruses8, 10.3390/v8120322 (2016).
    1. Cao, B., Parnell, L. A., Diamond, M. S. & Mysorekar, I. U. Inhibition of autophagy limits vertical transmission of Zika virus in pregnant mice. J Exp Med, 10.1084/jem.20170957 (2017).
    1. Shiryaev SA, et al. Characterization of the Zika virus two-component NS2B-NS3 protease and structure-assisted identification of allosteric small-molecule antagonists. Antiviral Res. 2017;143:218–229. doi: 10.1016/j.antiviral.2017.04.015.
    1. Tang H, et al. Zika Virus Infects Human Cortical Neural Progenitors and Attenuates Their Growth. Cell Stem Cell. 2016;18:587–590. doi: 10.1016/j.stem.2016.02.016.
    1. Morrison, T. E. & Diamond, M. S. Animal Models of Zika Virus Infection, Pathogenesis, and Immunity. J Virol91, 10.1128/JVI.00009-17 (2017).
    1. Rossi SL, et al. Characterization of a Novel Murine Model to Study Zika Virus. Am J Trop Med Hyg. 2016;94:1362–1369. doi: 10.4269/ajtmh.16-0111.
    1. Govero J, et al. Zika virus infection damages the testes in mice. Nature. 2016;540:438–442. doi: 10.1038/nature20556.
    1. Paz-Bailey, G. et al. Persistence of Zika Virus in Body Fluids - Preliminary Report. N Engl J Med, 10.1056/NEJMoa1613108 (2017).
    1. Frank, C. et al. Sexual transmission of Zika virus in Germany, April 2016. Euro surveillance: bulletin Européen sur les maladies transmissibles = European communicable disease bulletin21, 10.2807/1560-7917.ES.2016.21.23.30252 (2016).
    1. Moreira J, Peixoto TM, Siqueira AM, Lamas CC. Sexually acquired Zika virus: a systematic review. Clin Microbiol Infect. 2017;23:296–305. doi: 10.1016/j.cmi.2016.12.027.
    1. Cao, B., Diamond, M. S. & Mysorekar, I. U. Maternal-Fetal Transmission of Zika Virus: Routes and Signals for Infection. J Interferon Cytokine Res, 10.1089/jir.2017.0011 (2017).
    1. Fakeye TO, Fehintola FA, Ademowo OG, Walker O. Therapeutic monitoring of chloroquine in pregnant women with malaria. West Afr J Med. 2002;21:286–287.
    1. Lee SJ, et al. Chloroquine pharmacokinetics in pregnant and nonpregnant women with vivax malaria. Eur J Clin Pharmacol. 2008;64:987–992. doi: 10.1007/s00228-008-0500-z.
    1. Chandra RS, et al. Creative solutions to extraordinary challenges in clinical trials: methodology of a phase III trial of azithromycin and chloroquine fixed-dose combination in pregnant women in Africa. Malar J. 2013;12:122. doi: 10.1186/1475-2875-12-122.
    1. Radeva-Petrova, D., Kayentao, K., ter Kuile, F. O., Sinclair, D. & Garner, P. Drugs for preventing malaria in pregnant women in endemic areas: any drug regimen versus placebo or no treatment. Cochrane Database Syst Rev, CD000169, 10.1002/14651858.CD000169.pub3 (2014).
    1. Law I, et al. Transfer of chloroquine and desethylchloroquine across the placenta and into milk in Melanesian mothers. Br J Clin Pharmacol. 2008;65:674–679. doi: 10.1111/j.1365-2125.2008.03111.x.
    1. Levy M, Buskila D, Gladman DD, Urowitz MB, Koren G. Pregnancy outcome following first trimester exposure to chloroquine. Am J Perinatol. 1991;8:174–178. doi: 10.1055/s-2007-999371.
    1. Titus EO. Recent developments in the understanding of the pharmacokinetics and mechanism of action of chloroquine. Ther Drug Monit. 1989;11:369–379. doi: 10.1097/00007691-198907000-00001.
    1. Mackenzie AH. Dose refinements in long-term therapy of rheumatoid arthritis with antimalarials. Am J Med. 1983;75:40–45. doi: 10.1016/0002-9343(83)91269-X.
    1. Browning, D. J. In Hydroxychloroquine and Chloroquine Retinopathy 35–63 (Springer, 2014).
    1. Smit JM, Moesker B, Rodenhuis-Zybert I, Wilschut J. Flavivirus cell entry and membrane fusion. Viruses. 2011;3:160–171. doi: 10.3390/v3020160.
    1. Maramorosch, K., Shatkin, A. J., Murphy, F. A., Chambers, T. J. & Monath, T. P. The flaviviruses: structure, replication and evolution. Vol. 59 (Academic Press, 2003).
    1. Hsieh SC, et al. Highly conserved residues in the helical domain of dengue virus type 1 precursor membrane protein are involved in assembly, precursor membrane (prM) protein cleavage, and entry. J Biol Chem. 2014;289:33149–33160. doi: 10.1074/jbc.M114.610428.
    1. Barrows NJ, et al. A Screen of FDA-Approved Drugs for Inhibitors of Zika Virus Infection. Cell Host Microbe. 2016;20:259–270. doi: 10.1016/j.chom.2016.07.004.
    1. Bullard-Feibelman KM, et al. The FDA-approved drug sofosbuvir inhibits Zika virus infection. Antiviral Res. 2017;137:134–140. doi: 10.1016/j.antiviral.2016.11.023.
    1. Sacramento CQ, et al. The clinically approved antiviral drug sofosbuvir inhibits Zika virus replication. Sci Rep. 2017;7:40920. doi: 10.1038/srep40920.
    1. Li C, et al. 25-Hydroxycholesterol Protects Host against Zika Virus Infection and Its Associated Microcephaly in a Mouse Model. Immunity. 2017;46:446–456. doi: 10.1016/j.immuni.2017.02.012.
    1. Xu M, et al. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat Med. 2016;22:1101–1107. doi: 10.1038/nm.4184.
    1. Retallack H, et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci USA. 2016;113:14408–14413. doi: 10.1073/pnas.1618029113.
    1. Zmurko J, et al. The Viral Polymerase Inhibitor 7-Deaza-2′-C-Methyladenosine Is a Potent Inhibitor of In Vitro Zika Virus Replication and Delays Disease Progression in a Robust Mouse Infection Model. PLoS Negl Trop Dis. 2016;10:e0004695. doi: 10.1371/journal.pntd.0004695.
    1. Reznik SE, Ashby CR., Jr. Sofosbuvir: an antiviral drug with potential efficacy against Zika infection. Int J Infect Dis. 2017;55:29–30. doi: 10.1016/j.ijid.2016.12.011.
    1. Lanciotti RS, et al. Genetic and serologic properties of Zika virus associated with an epidemic, Yap State, Micronesia, 2007. Emerg Infect Dis. 2008;14:1232–1239. doi: 10.3201/eid1408.080287.

Source: PubMed

3
Abonnere