Skip directly to site content Skip directly to page options Skip directly to A-Z link Skip directly to A-Z link Skip directly to A-Z link
Volume 21, Number 3—March 2015
Letter

Lack of Effect of Lamivudine on Ebola Virus Replication

Cite This Article

To the Editor: The unprecedented number of Ebola virus disease (EVD) cases in western Africa has compelled the world to consider experimental and off-label therapeutics to mitigate the current outbreak. For clinicians, approved drugs are an attractive solution because of known safety profiles and availability.

Oral lamivudine (GlaxoSmithKline, Brentford, UK), a US Food and Drug Administration–approved anti-HIV drug, has been suggested as a possible antiviral agent against Ebola virus (EBOV). In September 2014, a Liberian physician, Dr. Gorbee Logan, reported positive results while treating EVD with lamivudine (1). Thirteen of 15 patients treated with lamivudine survived presumed EVD and were declared virus free. Clinical confirmation of EVD in these cases remains to be verified.

Our laboratory had previously assessed this antiretroviral compound in drug screens against EBOV and observed no discernable antiviral activity. However, given the recent testimonials regarding lamivudine effectiveness in treating EBOV-infected patients in Africa, we conducted additional studies to determine whether our previous assertion that lamivudine lacked any direct antiviral activity was correct.

Lamivudine is a nucleoside analog reverse transcription inhibitor of HIV and hepatitis B virus that acts as a synthetic cytidine analog. Incorporation of the active triphosphate form into viral DNA results in chain termination. Studies have demonstrated that lamivudine is a weak inhibitor of mammalian α, β, and γ DNA polymerases (2). Lamivudine would not be expected to inhibit the replication of a negative-strand RNA virus.

The activity of lamivudine against EBOV infection was evaluated in a cell-based ELISA with 1995 isolate EBOV H. sapiens-tc/COD/1995/Kikwit (EBOV/Kik) (3). Three cell lines were tested: Vero E6 (African green monkey kidney, ATCC CRL-1586), Hep G2 (human hepatoma, ATCC HB-8065), and human monocyte-derived macrophages. Macrophages were generated by treating CD14+ cells for 7 days with macrophage colony–stimulating factor and conditioned medium. Cells were treated with compounds in 3-, 4-, or 8- point dose response curves with 2-fold dilutions starting at 80 µmol/L or 320 µmol/L oral lamivudine. Toremifene (T7204–5MG; Sigma-Aldrich, St. Louis, MO, USA) was used as a positive control for activity against EBOV and tested at 2-fold dilutions starting at 25 µmol/L. One hour after drug addition, the cells were infected at a multiplicity of infection of 0.5 or 1 with EBOV/Kik. Experiments were run on duplicate plates or the entire experiment was run on 2 separate days. At 48 hours after infection, cells were formalin-fixed and stained with a primary antibody against EBOV (antibodies against viral matrix protein or glycoprotein) and a secondary antibody (Alexa-488 or horseradish peroxidase).

No direct antiviral effect for lamivudine was observed at concentrations ≤320 µmol/L in Vero E6 cells (Table). Because optimal efficacy of the drug requires phosphorylation, lack of activity may be caused by poor phosphorylation in Vero E6 cells (6). Therefore, we also assessed HepG2 cells and primary human monocyte–derived macrophages sensitive to EBOV infection. Toremifene was included as a positive control. Toremifene is a US Food and Drug Administration–approved drug that was reported to have direct antiviral activity in cell culture and to protect mice infected with mouse-adapted EBOV (3). As expected, toremifene inhibited EBOV at low micromolar concentrations (Table).

Finally, we assessed the antiviral activity of the compounds against a recent isolate prototype from the current outbreak, EBOV H. sapiens-tc/GIN/2014/Guéckédou-C05 (EBOV/Gue) to test whether inhibition of EBOV/Gue by lamivudine was different from that of the reference Kikwit strain. In contrast to a known active compound (toremifene), lamivudine showed no direct antiviral activity.

The current data suggest that lamivudine does not directly inhibit EBOV RNA polymerase or replication of the virus. Systemic and off-target effects, while not previously described, might be possible. To address this possibility, we plan to assess lamivudine in the mouse model of EVD and will report these findings when available. However, on the basis of these in vitro tests, there is no foundation for recommending lamivudine for treatment of EVD in human patients.

Top

Acknowledgments

G.G.O. was named in patent #8,475,804 assigned to the US Army on approved drugs for use for filoviruses.

This study was supported by National Institute of Allergy and Infectious Diseases contract HHSN272200700016I.

Top

Lisa Hensley, Julie Dyall, Gene G. Olinger, and Peter B. JahrlingComments to Author 
Author affiliations: National Institutes of Health, Frederick, Maryland, USA

Top

References

  1. Cohen  E. Doctor treats Ebola with HIV drug in Liberia–seemingly successfully [videotape]. Atlanta: CNN Health; September 29, 2014. Running time: 2:08 minutes [cited 2014 Oct 28]. http://www.cnn.com/2014/09/27/health/ebola-hiv-drug/index.html?iref=allsearch.
  2. Hart  GJ, Orr  DC, Penn  CR, Figueiredo  HT, Gray  NM, Boehme  RE, Effects of (-)-2'-deoxy-3′-thiacytidine (3TC) 5′-triphosphate on human immunodeficiency virus reverse transcriptase and mammalian DNA polymerases alpha, beta, and gamma. Antimicrob Agents Chemother. 1992;36:168894 . DOIPubMedGoogle Scholar
  3. Johansen  LM, Brannan  JM, Delos  SE, Shoemaker  CJ, Stossel  A, Lear  C, FDA-approved selective estrogen receptor modulators inhibit Ebola virus replication Sci Transl Med. 2013;5:190ra79..DOIGoogle Scholar
  4. Schinazi  RF. Assessment of the relative potency of emtricitabine and lamivudine. J Acquir Immune Defic Syndr. 2003;34:2435, author reply 5–6. DOIPubMedGoogle Scholar
  5. Kruining  J, Heijtink  RA, Schalm  SW. Antiviral agents in hepatitis B virus transfected cell lines: inhibitory and cytotoxic effect related to time of treatment. J Hepatol. 1995;22:2637. DOIPubMedGoogle Scholar
  6. Furman  PA, Fyfe  JA, St Clair  MH, Weinhold  K, Rideout  JL, Freeman  GA, Phosphorylation of 3′-azido-3′-deoxythymidine and selective interaction of the 5′-triphosphate with human immunodeficiency virus reverse transcriptase. Proc Natl Acad Sci U S A. 1986;83:83337. DOIPubMedGoogle Scholar

Top

Table

Top

Cite This Article

DOI: 10.3201/eid2103.141862

Related Links

Top

Table of Contents – Volume 21, Number 3—March 2015

EID Search Options
presentation_01 Advanced Article Search – Search articles by author and/or keyword.
presentation_01 Articles by Country Search – Search articles by the topic country.
presentation_01 Article Type Search – Search articles by article type and issue.

Top

Comments

Please use the form below to submit correspondence to the authors or contact them at the following address:

Peter B. Jahrling, Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, B8200 Research Plaza Dr, Frederick, MD 21702, USA

Send To

10000 character(s) remaining.

Top

Page created: February 18, 2015
Page updated: February 18, 2015
Page reviewed: February 18, 2015
The conclusions, findings, and opinions expressed by authors contributing to this journal do not necessarily reflect the official position of the U.S. Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors' affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above.
file_external