Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Aug 24.
Published in final edited form as: ACS Infect Dis. 2015 Mar 30;1(5):186–197. doi: 10.1021/id5000426

Pathophysiology of Ebola virus infection: Current challenges and future hopes

Andrea Rivera 1, Ilhem Messaoudi 1,#
PMCID: PMC7443712  NIHMSID: NIHMS1615488  PMID: 27622648

Abstract

The filoviruses, Ebola virus (EBOV) and Marburg virus (MARV), are amongst the deadliest viruses that cause disease in humans with reported case fatality rates of up to 90% in some outbreaks. The high virulence of EBOV and MARV is largely attributed to the ability of these viruses to interfere with host immune response. Currently, there are no approved vaccines or post-exposure therapeutics and treatment options for patients infected with EBOV are limited to supportive care. In this review, we discuss mechanisms of EBOV pathogenesis and its ability to subvert host immunity as well as several vaccines and therapeutics with respect to their evaluation in small animal models, nonhuman primates, and human clinical trials.

Keywords: Ebola virus, viral hemorrhagic fever, innate immunity, adaptive immunity, immunoevasion, antivirals, Ebola vaccines, Ebola therapeutics


Ebola virus (EBOV) is a filamentous enveloped virus containing a negative strand RNA genome 19 kb in length that encodes for a nucleoprotein (NP), glycoprotein (GP), RNA dependent RNA polymerase (L), and four structural proteins termed VP24, VP30, VP35 and VP40. Viral replication is carried out by NP, VP35 and L; the active polymerase complex is composed of VP35 (a polymerase cofactor) and L (polymerase) while NP drives RNA encapsidation. VP30 is a transcriptional activator and is also involved in nucleocapsid formation and assembly1. VP24 is a matrix protein that contributes to nucleocapsid formation while matrix protein VP40 facilitates budding of progeny virion from infected cells 1b, 2. Glycoprotein (GP) covers the surface of the virion and is the sole host attachment factor for EBOV 3. Moreover, EBOV expresses a two soluble forms of GP, sGP and ssGP through RNA editing 4.

Infection with EBOV results in severe viral hemorrhagic fever with fatality rates that can reach 90% in humans depending on the species. Infection with Zaire Ebola virus (EBOV), Sudan Ebola virus (SUDV) and Bundibugyo Ebola virus (BDBV) is associated with 70-90%, 50% and 40% mortality rate, respectively 5. In contrast, infection with either Tai Forest Ebola virus (TAFV) or Reston Ebola virus (RESTV) have not been associated with human fatalities 5. EBOV is transmitted by direct contact with infected bodily fluid. The incubation period ranges from 2 to 21 days with an average incubation period of 5-7 days. Early symptoms are general and nonspecific including fever, general malaise and myalgia. Following early symptoms, cases can show maculo-papulary rash, petichae, conjunctival hemorrhage, melena, hematemesis, shock and encephalopathy. During the terminal stage of disease, there is an increase in vascular permeability, massive tissue injury, dysregulation of the coagulation cascade and hemorrhage. Multiorgan failure and shock are usually the main causes of death 6.

Previous EBOV outbreaks have occurred mostly in remote regions of central Africa including the Democratic Republic of Congo, Sudan, Gabon and Uganda. However, in March 2014, the World Health Organization (WHO) reported an outbreak of Ebola virus disease originating in the Guinean capital, Conakry, marking the first large urban setting for EBOV transmission. This has led to the first known epidemic in West Africa and is the largest and longest outbreak since the virus was first discovered in 1976 7. Genome sequencing of the Guinea strain show 97% identity to EBOV 8. Sequencing studies of 99 EBOV genomes from 78 patients in Sierra Leone reveal an accumulation of inter- and intra-host genetic variation. Patterns of viral transmission suggest this variant diverged from central African lineages around 2004 9. As of March 8, 2015 the CDC reports a total 24,247 confirmed, probable and suspected cases of EBOV disease and 9,961 reported deaths in eight countries (Guinea, Liberia, Sierra Leone, Mali, Nigeria, Senegal, Spain and United States). Currently, there are no FDA-approved treatment strategies. Supportive care, consisting of oral fluid rehydration and nutritional supplementation, is the mainstay in treating Ebola hemorrhagic fever (EHF). The high mortality rate has caused concern to public health officials worldwide, prompting a sense of urgency to accelerate clinical trials to identify effective vaccines and post-exposure therapeutics.

Nonhuman primates, particularly cynomolgus macaques and rhesus macaques, are the gold standard animal models used in filovirus study because they are susceptible to infection by the same strains that cause disease in humans and exhibit strong similarity of viral hemorrhagic fever to humans 10. Small animal models, such as inbred mice and guinea pigs have also been used, however, these animal models require the use of adapted EBOV strains and do not exhibit the characteristic hemorrhagic manifestations seen with human and nonhuman primate infection 11. In a recent study using Collaborative Cross mice, animals exhibited a spectrum of disease outcomes following infection with mouse adapted EBOV (MA-EBOV), including hemorrhagic fever 12. However, MA-EBOV has diminished virulence in nonhuman primates. Of three rhesus macaques injected with a large challenge dose of 5000 pfu of MA-EBOV, two showed mild illness and survived infection 13. A single amino acid mutation in both NP and VP24 are the determinants of virulence of MA-EBOV in mice 14. Although MA-EBOV infection in mice correlated with evasion of IFN induced responses 14, the determinants for virulence of EBOV may differ between mouse and primate models. The purpose of this review is to summarize our current understanding of the pathogenesis of EBOV and discuss the various vaccine platforms and therapeutics currently being investigated.

Dysregulation of innate immunity by EBOV infection

Although EBOV targets a wide range of cell lineages, in vivo studies in nonhuman primates using immunohistochemistry and in-situ hybridization suggest that EBOV replicates preferentially in monocytes, macrophages, and dendritic cells 10b, 15. Infection of monocytes and macrophages triggers the robust expression of inflammatory mediators 16 (Figure 1). Indeed, several studies have shown that fatal infections were associated with uncontrolled secretion of proinflammatory cytokines, chemokines and growth factors such as IL-1β, IL-6, IL-8, IL-10, MCP-1, MIP-1α, MIP-1β and TNFα as well as nitric oxide and reactive oxygen species, which, at the time of death, can reach 5 to 1000 times the levels detected in survivors and healthy individuals 17. In contrast, survivors show a transient and moderate upregulation in levels of IL-1β, IL-6, TNFα, MIP1α and MIP1β early in the disease 17a. These studies suggest that protection from fatal EBOV infection may depend on an early yet regulated inflammatory response.

Figure 1. Dysregulation of Immune System by Ebola virus.

Figure 1.

Monocytes, macrophages, and dendritic cells are preferred sites of filovirus replication. Infection of monocytes and macrophages triggers robust expression of inflammatory mediators. Inflammatory mediators, reactive oxygen species and nitric oxide can induce apoptosis leading to lymphocyte death. Infection of dendritic cells impairs their maturation and suppresses type I (IFN) responses thereby preventing T cell activation. Production of EBOV soluble glycoprotein (sGP) usurps GP-specific antibodies.

Ebola virus infection is also characterized by a suppressed type I interferon (IFN) response, which is an integral part of the innate immune response against viral infections. This suppression is mediated by VP24, which prevents the nuclear transport of tyrosine phosphorylated STAT1 (pSTAT1), a key downstream outcome of IFN signaling, thereby preventing IFN-induced gene expression 18. VP24 binds the C-terminus of karyopherin α proteins (karyopherin α1, α5, α6), which normally transport pSTAT1 through the nuclear pore, therefore competing with pSTAT1 for binding the nuclear transporter 19. In addition to VP24, VP35 was also found to block activation of the transcription factor IFN regulatory factor 3 (IRF-3), thus decreasing IFN production 20. VP35 interacts with the SUMO E2 enzyme Ubc9 and the E3 ligase PIAS1, which in turn leads to increased SUMOylation and degradation of the transcription factor IRF-7 21. VP35 can also inhibit retinoic-acid inducible gene-I (RIG-I) helicase signaling by binding to dsRNA 22 or binding to PKR activating protein (PACT), which activates RIG-I 23. Moreover, VP35 can decrease IFN production by impairing IKKε and TBK-1 kinase function 24. Interference with RIG-I signaling can also inhibit the upregulation of a number of co-stimulatory molecules on dendritic cells (CD40, CD80, CD86, and MHC class II) needed to activate T cells 25. Additional, in vitro studies have shown that EBOV infection inhibits dendritic cells maturation into mobile, antigen presenting cells and impairs their ability to stimulate antigen specific T cell responses 26 (Figure 1).

Dysregulation of lymphocyte function by EBOV infection

One major consequence of EBOV infection is severe lymphopenia 10a. EBOV infection leads to loss of both human CD4+ and CD8+ T cells after 4 days of in vitro culture 27. Similarly, EBOV infection resulted in loss of peripheral CD4+ and CD8+ T cells in both mouse models at 2-3 dpi 28 and in nonhuman primates at 4 dpi 29. Significant loss of circulating natural killer (NK) cells was also described in mice 28 and in cynomolgus macaques 10b, 29. B cell loss is more controversial with some studies reporting the loss of B lymphocytes in mice 28 and macaques 15 and other studies reporting no changes in B cell counts in nonhuman primates 2930. Flow cytometric analysis of peripheral blood mononuclear cells (PBMCs) from humans and cynomolgus macaques infected with EBOV revealed an increase in the percentage of CD4+ and CD8+ T cells expressing the cell death receptor Fas (CD95) suggesting that apoptosis is the primary mechanism of lymphocyte death 17f, 29. Transmission electron microscopy and TUNEL staining confirmed lymphopenia in vivo and in vitro occurred by apoptosis 15. Increased plasma levels of apoptosis mediators such as soluble Fas (sFas) and 41/7 nuclear matrix protein, upregulation of Fas and FasL mRNA in PBMCs, and dramatic DNA fragmentation in leukocytes were detected during the terminal stage of human EBOV infection 31. Furthermore, PBMCs from survivors showed an upregulation of anti-apoptotic Bcl-2 mRNA whereas fatalities showed a significant decrease in Bcl-2 mRNA as well as a decrease in CD3, CD8, and TCR-Vβ mRNA 15, 31b.

The fact that EBOV does not replicate within lymphocytes during infection suggests that lymphocyte apoptosis is an indirect result of viral replication 15, 31a. Some of the proinflammatory mediators released by EBOV-infected monocytes such as TNFα, reactive oxygen species and nitric oxide have been shown to induce apoptosis 32 (Figure 1). Furthermore, immunohistochemistry, flow cytometry, and RNA analysis showed high expression of TNF related apoptosis inducing ligand (TRAIL) in 90% of EBOV infected adherent human monocytes/macrophages 17c. In addition to lymphopenia, EBOV infection interferes with T cell activation as evident by decreased expression of the activation marker CD44 26, 29. The absence of EBOV-specific T cell responses is consistent with the lack of T cell cytokines (IL-2, IL-4, IL-5, IL-12 and IFNγ) in the plasma of EBOV infected patients 17f, 33.

EBOV also subverts the host humoral immune response, as fatalities are associated with a lack of EBOV specific IgG antibodies 31b. The loss of CD4+ T cells, which are required for isotype class switching, may explain the lack of Ebola specific IgM and IgG antibodies observed in patients who succumb to infection. In contrast, asymptomatic EBOV infected patients developed IgM responses between 10-18 days and IgG responses between 17 −25 days specific to GP, NP and VP40 34. EBOV also subverts humoral responses through the production of sGP 30, which sequesters GP-specific antibodies needed to control viral replication 35 (Figure 1). The importance of antibodies in protection against EBOV infection is highlighted by passive immunization studies. During one of the EBOV outbreaks in the Democratic Republic of Congo, 7/8 patients treated with blood transfusions from 5 convalescent patients who generated EBOV-specific IgG antibodies, survived 36. Similarly, 100% of naïve mice were protected against infection when treated with serum from vaccinated mice that survived challenge with mouse-adapted EBOV 37.

Vaccines Against Ebola

Several vaccine platforms against EBOV have been developed. Replication deficient vaccines include DNA based vaccines, virus like particles (VLPs) and recombinant adenovirus vectors (rAd). Replication competent constructs include recombinant human parainfluenza virus 3 (rHPIV3), recombinant vesicular stomatitis virus (rVSV) and more recently recombinant rabies virus (RABV) and recombinant cytomegalovirus (CMV) 38. Two promising vaccine candidates, rAd and rVSV vectors expressing GP (Table 1), have entered clinical trials.

Table 1.

Vaccines against Ebola Virus

Vaccine rAd5/ChAd3 rVSV/ΔG/GP EBOV
Replication potential Replication deficient Replication competent
Mechanism of protection CD8+ T cell mediated Antibody mediated
Booster requirement Modified vaccinia Ankara
Durability 10 months with booster vaccination in nonhuman primates 12-18 months in rodents
14 months in nonhuman primates (MARV)
Clinical trial Phase II/III Phase II/III

Vaccination of cynomolgus macaques with the replication deficient rAd serotype 5 vector expressing EBOV-GP (rAd5-EBOVGP) resulted in the generation of EBOV-GP specific antibodies and GP-specific CD8+ T cell responses within 3 weeks of immunization and protected 100% of the animals against challenge 39. Passive transfer of EBOV-specific IgG purified from nonhuman primates vaccinated with DNA and rAd5 vectors expressing EBOV GP to naïve animals only protected 25% of naïve animals 6 or 16 hours before EBOV challenge, whereas, depletion of CD8+ T cells in vaccinated animals 4 days before challenge resulted in 4/5 nonhuman primates succumbing to infection, suggesting that CD8+ T cells play a more important role in protection compared to antibodies in this vaccine platform 40. Despite these promising results, humans have preexisting immunity against Ad5, which may interfere with the efficacy of the rAd5-EBOVGP vaccine 38. On the other hand, adenoviruses isolated from chimpanzees (ChAds) provide better vaccine vectors because of their low seroprevalence in humans. Immunization of cynomolgus macaques with a single inoculation of recombinant ChAd3 expressing EBOV GP (rChAd3-EBOVGP) resulted in complete protection when challenged with EBOV 5 weeks after vaccination but only conferred 50% protection when the animals were challenged 10 months after immunization. To improve long-term efficacy of this vaccine platform, a booster vaccination using recombinant modified vaccinia Ankara expressing EBOV-GP was added 8 weeks after the rChAd3-EBOVGP vaccination, which resulted in complete protection at 10 months 41. Phase I clinical trials of rChAd3 began in September 2014 by the National Institute of Allergy and Infectious Diseases (NIAID) to evaluate the vaccine’s safety and immunogenicity. Twenty adults were vaccinated with either 2 x 1010 or 2 x 1011 particles. Reactivity to rChAd3 was dose dependent as two participants developed fever 1 day after vaccination with the 2 x 1011 dose. Similarly, immunogenicity was also dose dependent with higher GP-specific antibodies and T-cell responses detected in the group that received the 2x1011 dose four weeks after vaccination 42. In parallel, the NIH partnered with the United Kingdom-based international consortium to test the safety and efficacy of rChAd3, at a dose of either 2 x 1010 or 2 x 1011, among 60 volunteers at the University of Oxford in England and 40 volunteers in Mali and have observed similar results in regards to safety and immune response 43. The vaccine will enter a Phase II/III trial called Partnership for Research on Ebola Vaccines in Liberia (PREVAIL). This study will enroll 27,000 healthy men and women to investigate rChAd3 and rVSV/ΔG/GP and is estimated to be completed in June 2016 43.

Vesicular stomatitis virus (VSV) is a nonsegmented, negative-stranded RNA virus and a member of the Rhabdoviridae family. Recombinant VSV, in which the VSV glycoprotein is replaced with EBOV GP (rVSV/ΔG/GP EBOV), confers 100% protection in nonhuman primates and mice challenged 28 days after immunization with EBOV and mouse-adapted EBOV respectively 45. Antibodies play a critical role in rVSV/ΔG/GP -mediated protection against EBOV. In a nonhuman primate study, only the animals depleted of CD4+ T cells during vaccination, which lacked GP-specific antibodies, succumbed to infection, suggesting that antibodies were required for protection 46. Because rVSV/ΔG/GP is a live-attenuated virus, several studies have investigated its safety. No toxicity was observed in over 80 nonhuman primates given rVSV/ΔG/GP MARV or rVSV/ΔG/GP EBOV vaccines 45a, 47. Moreover, this vaccine was safe in a mouse model of severe combined immunodeficiency (NOD-SCID) and in simian human immunodeficiency (SHIV) infected rhesus macaques 45b, 47b. Administration of a single dose of rVSV/ΔG/GP confers 100% protection against EBOV challenge for up to 6 months in macaques and 18 months in mice and guinea pigs and this protection correlates with GP-specific IgG titers 48,49. This vaccine platform has also provided protection in macaques against MARV for up to 14 months after vaccination, further demonstrating the durability of VSV based vaccines against filoviruses 50. Administration of a mixture vaccine containing rVSV/ΔG/GP from MARV, EBOV and SUDV to cynomolgus macaques resulted in their survival following challenge with MARV, EBOV, SUDV and TAFV, demonstrating that a single-injection of a multivalent vaccine is as efficacious as the administration of a single specificity vaccine 51. Finally, rVSV/ΔG/GP has also been shown to be effective post-exposure. Administration of rVSV/ΔG expressing EBOV or SUDV GP to rhesus macaques 20-30 minutes after challenge resulted in 50% and 100% protection, respectively 52. In March 2009 in Germany, a virologist working in a biosafety level 4 sustained a needle stick injury with a syringe that contained EBOV mixed with Freund’s adjuvant. A single dose of 5 x 107 pfu rVSV/ΔG/GP EBOV was administered to her 48 hours after the injury. Following post-exposure vaccination, the virologist developed a fever 12 hours later in addition to rVSV viremia detected via PCR for 2 days, but remained healthy during the 3-week observation period 53. During the 2014 outbreak, rVSV/ΔG/GP EBOV was used as an emergency post-exposure vaccination and administered 43 hours after a physician experienced a needle stick injury while working in an Ebola treatment unit in Sierra Leone 54. Although it is unknown if rVSV/ΔG/GP EBOV was effective as a post-exposure vaccination, the patient had a self-limited febrile syndrome and cytokine and Ebola glycoprotein specific adaptive immune response after vaccination54. The NIH and WHO have carried out Phase I clinical trials to test the safety and efficacy of rVSV/ΔG/GP EBOV, with a dose ranging from 3 x 106 pfu to 1 x 108 pfu, in various locations in the US, Europe and Africa. Although safety data from Phase I studies have not yet been published, there is sufficient safety and efficacy information to push the vaccine forward into the PREVAIL Phase II/III trial43. The CDC in collaboration with Sierra Leone has launched another Phase II/III trial called Sierra Leone Trial to Introduce a Vaccine Against Ebola (STRIVE). The study will test a dose of 2 x 107 pfu of rVSV/ΔG/GP on 6,000 participants55.

Therapeutics Against Ebola

Various therapeutic candidates that either directly inhibit viral replication, modulate clinical symptoms, or prolong survival time have been evaluated either as sole or adjunctive post-exposure therapies in rodent and nonhuman primates. A complete list of Ebola therapeutics is summarized in Table 2. Two anticoagulant recombinant proteins, recombinant nematode anticoagulant protein c2 (rNAPc2) and recombinant human activated protein C (rhAPC) have been evaluated in preventing increased coagulation during filovirus infection 59. However, both anticoagulants provided only partial protection in nonhuman primates challenged with EBOV 59. Administration of rNAPc2 to 6 rhesus macaques 10 minutes after EBOV challenge prolonged survival time from mean time-to-death of 8.3 days to 11.7 days with a 33% survival rate 59a. Treatment with rhAPC in 11 rhesus macaques starting 30-60 minutes after EBOV challenge and daily for 7 days increased mean time-to-death from 8.3 to 12.6 days with 2 of 11 rhesus macaques surviving 59b. Although rNAPc2 has gone through Phase II trials for use in prevention of venous thromboembolism after orthopedic surgery 60 and coronary revascularization 61, it has not been assessed in clinical trials for treatment of EBOV exposure. Additionally, rhAPC has not been evaluated in clinical trials for use against EBOV infection. Treatment with interferon beta (IFNβ) to 5 of EBOV infected rhesus macaques 18 hours and 1, 3, 5, 7 and 9 days post infection did not alter mortality but significantly increased mean time-to-death from 8.3 days to 13.8 days 62 and may be used as an adjunctive therapeutic.

Table 2.

Therapeutics for EBOV infection

Molecule Mechanism of Action Development Phase Reference
Favipiravir A nucleotide analog that inhibits RNA polymerase. Has been shown to protect mice against EBOV. Administered to one EBOV infected patient who survived.
Currently in Phase II trial.
63
PMOs AVI-6002 (AVI-7537 and AVI-7539)

AVI-6003 (AVI-7287 and AVI-7288)
AVI 7537 targets EBOV VP24; AVI-7539 targets EBOV VP35

AVI-7287 targets MARV VP24; AVI-7288 targets MARV NP
AVI-6002 has been shown to provide partial protection of rhesus macaques against EBOV. AVI-7537 confers sole protection observed. AVI-6003 has been shown to completely protect cynomolgus macaques from MARV infection.
Completed Phase I clinical trial.
71
ZMapp Monoclonal cocktail of antibodies against different epitopes of EBOV-GP. Has been shown to completely protect cynomologus macaques. Protected 5/7 patients infected with EBOV. Currently in Phase I clinical trial. 83,85
BCX4430 An adenosine analog that inhibits RNA transcription. Shown to protect cynomolgus macaques against Marburg virus.
In Phase I clinical trial.
66
Brincidofovir Inhibits DNA synthesis. Mechanism against RNA viruses is unknown. Shows efficacy against EBOV in vitro. Protected 1 of 2 US patients infected with EBOV. Withdrawn from Phase II clinical trial 69,68
TKM-Ebola Treatment with SNALP carrying siRNAs against EBOV L polymerase, VP24, and VP35. Protected macaques following EBOV challenge. Phase I trial were halted. FDA expanded access to TKM-Ebola on an emergency basis. 7475
rhAPC Anticoagulant Recombinant human activated protein C is an anticoagulant that prolongs survival time and provides partial protection in nonhuman primates. 59b
rNAPc2 Anticoagulant Recombinant nematode anticoagulant protein c2 prolongs survival time and provides partial protection in nonhuman primates. 59a
IFNβ Interferon Prolongs mean time-to-death in rhesus macaques infected with EBOV. May be possible adjunctive post-exposure therapy 62
Chloroquine The antiviral mechanism is unknown. FDA approved as an antimalarial agent. Through a screening study evaluating 1012 FDA-approved drugs, chloroquine was found to possess in vitro activity against EBOV. Found to protect mice against EBOV infection in vivo. 87
Selective estrogen receptor modulators: clomiphene and toremifene Inhibits EBOV entry. Clomiphene and toremifene inhibit EBOV infection in vitro and show partial protection in mice challenged with mouse adapted EBOV. 88
FGI compounds:
FGI-103,
FGI-104,
FGI-106
FGI compounds are broad-spectrum viral inhibitors.
Antiviral mechanism against EBOV is unknown.
Exhibited efficacy against EBOV in vitro and in mouse models. 89,90,91
SAH hydrolase inhibitor Deazaneplanocin A Targets SAH hydrolase, indirectly resulting in reduced methylation of 5’ cap of viral messenger RNA. May also increase IFNα production Early dose protected mice against EBOV challenge. 92
Glucosidase inhibitors: CM-10-18, IHVR11029, IHVR17028 and IHVR19029 Glucosidase inhibitors inhibit Endoplasmic reticulum α-glucosidases I and II and prevent maturation of viral glycosylated envelope and, inhibiting viral entry. CM-10-18, IHVR11029, IHVR17028 and IHVR19029 has been shown to suppress mortality of MARV and EBOV infection in mice. 93
Antioxidant NSC62914 Acts as a scavenger of reactive oxygen species and upregulates oxidative stress-induced genes. Exhibits anti-filovirus activity in vitro and in vivo in mice infected with EBOV or Marburg virus. 94
HSPA5 inhibitors Targets HSPA5 chaperone (EBOV-associated host factor). PMOs targeting HSPA5 reduced viral replication and protected mice against lethal EBOV infection. 95
SAH hydrolase inhibitor Neplanocin A, 3- Targets SAH hydrolase, indirectly resulting in reduced methylation of 5’ cap of viral messenger RNA Efficacy of Neplanocin A against EBOV has been shown in vitro. 96,97
Benzylpiperazine adamantine diamides Inhibits EBOV entry via cholesterol transporter Niemann-Pick C1 in vitro Shown to inhibit EBOV entry in vitro. 98,99
LJ-001 A rhodamine derivative, which prevents fusion of viral and cellular membranes and prevents viral entry. Shown to inhibit EBOV entry in vitro. 100
CMLDBU3402 Broad-spectrum antiviral Inhibits EBOV RNA transcription Shown to inhibit EBOV entry in vitro 101
Ion channel blockers: amiodarone, dronedarone, verapamil
Inhibit EBOV entry Shown to inhibit EBOV entry in vitro. 102
Heme oxygenase-1 (HO-1) Inhibit EBOV replication. Mechanism unknown HO-1 induction significantly suppressed EBOV replication in vitro. 103
Lamivudine Cytosine analog approved for treatment of HIV and hepatitis B virus. Unknown mechanism against EBOV Not yet evaluated in context of EBOV. Studies against EBOV in vitro are currently being conducted 104
duY11 A rigid amphipathic fusion inhibitor (RAFI) targets entry of enveloped viruses and may potentially inhibit entry of EBOV Not yet assessed in in vitro or in vivo studies against EBOV 105
Inhibitors of cathepsin L cleavage Prevention of cathepsin L cleavage of viral glycoproteins may inhibit EBOV entry. Not yet assessed in in vitro or in vivo studies 106
Statins Anti-inflammatory No in vitro or in vivo studies in efficacy against Ebola. However, may be used to treat viral sepsis during EBOV infection. FDA approved 107

Antivirals that have been evaluated as therapeutics against EBOV include nucleotide analogs (Favipiravir and BCX4430), Brincidofovir and antisense therapeutics (phosphorodiamidate morpholino oligomers (PMOs) and siRNAs) and are summarized in Table 2. Favipiravir (T-705) is an oral nucleotide analog that, when converted into its active metabolite ribofuranosyl triphosphate, inhibits the viral RNA-dependent RNA polymerase by directly competing with GTP. Currently licensed for influenza outbreaks, Favipiravir (T-705) was shown to suppress EBOV replication by 4 log10 units in Vero E6 cells 1 hour after infection 63. In a mouse model in which mice lack type I IFNα/β receptor and are susceptible to wild type EBOV, treatment with Favipiravir 6 hours after EBOV challenge resulted in complete survival of all five mice 63. To date, Favipiravir has been used to treat 1 French nurse infected with Ebola who recovered 64. Favipiravir entered Phase II evaluations in December 2014 in Guinea and is sponsored by Institut National de la Sante Et de la Recherche Medicale, France. Nucleoside analogue inhibitor BCX4430 is an adenosine analog with a C-nucleoside instead of the N-glycoside and a 1,4 imino group instead of the 1,4 oxygen. Once metabolized to the active triphosphate nucleotide form and after pyrophosphate cleavage, it is incorporated into the nascent viral RNA chain and terminates transcription. It is not incorporated into mammalian RNA or DNA. Administration of BCX4430 in cynomolgus macaque as late as 48 hours following infection with Marburg virus resulted in 100% protection 66. BioCryst Pharmaceuticals and NIAID have initiated a Phase I clinical trial of BCX4430 in the United Kingdom 67.

PMOs are synthetic antisense molecules that are able to target mRNA in a sequence-specific fashion and suppress translation through steric hindrance. AVI-6002 consists of PMO AVI-7537, which targets VP24, and AVI-7539, which targets VP35. AVI-6003 consists of PMO AVI-7287, which targets MARV VP24 and AVI-7288, which targets MARV NP. AVI-6002 protected 62.5% of rhesus monkeys against EBOV infection when given 30-60 minutes post infection and daily for 14 days. AVI-6003 protected 100% of cynomolgus macaques from MARV infection when given 30-60 minutes post infection and daily for 14 days 70. A follow up study discovered AVI-7537 alone protected 75% (6 out of 8) cynomolgus macaques against EBOV when administered 1 hour ± 30 minutes after challenge and daily for 14 days while treatment solely with AVI-7539 did not result in any survival past 10 days post infection 71 Results from a Phase I clinical trial evaluating the safety and pharmacokinetic profiles of AVI-6002 and AVI-6003 in two groups of 30 individuals revealed both AVI-6002 and AVI-6003 were safe and well tolerated with doses ranging from 0.005 to 4.5mg/kg per component72. Small-interfering RNA (siRNA) can inhibit translation of mRNA and has been shown to specifically downregulate MARV transcription of nucleocapsid protein in Vero E6 cells, significantly decreasing viral protein production and viral release 73. The design of stable nucleic acid-lipid particles (SNALP) to efficiently deliver siRNAs in vivo to target cells facilitated therapeutic evaluation. SNALP-encapsulated siRNAs targeting the EBOV polymerase L gene completely protected 5 guinea pigs from viremia and death when administered at a dose of 0.75mg/kg 1 hour after challenge and daily on days 1-6 post infection 74. Treatment with a SNALP carrying siRNAs targeting EBOV L polymerase, VP24, and VP35, called TKM-Ebola, resulted in the survival of 2/3 macaques that were given a 2mg/kg dose after 30 minutes and on days 1, 3, and 5 after EBOV challenge and 4/4 macaques that were a 2mg/kg dose after 30 minutes and on days 1, 2, 3, 4, 5, and 6 post challenge 75. However, Phase I trials of TKM-Ebola in early 2014 were halted after elevated cytokine levels were detected in healthy participants. In October 2014, the FDA expanded access to TKM-Ebola on an emergency basis 76. Brincidofovir (CMX001) is a lipid conjugate of cidofovir that can be converted intracellularly into cidofovir diphosphate, which has been shown to inhibit DNA polymerase. Although Brincidofovir is in clinical trials for diseases caused by DNA viruses, in vitro studies carried out at the CDC and NIH revealed Brincidofovir is active against Ebola virus 68. While the mechanism by which Brincidofovir acts against Ebola is unknown, Brincidofovir has been used to treat two patients infected with Ebola in the United States (one survived and one died). Although Chimerix initiated a Phase II clinical trial in October 2014 69, they withdrew the study in January 2015.

Given the large body of both experimental and clinical data supporting the role of antibodies in protection against EBOV infections, several monoclonal treatment modalities were tested. Human monoclonal antibody (mAb) KZ52, which targeted one epitope in EBOV-GP was able to protect guinea pigs 77, but failed to protect rhesus macaques against lethal challenge with EBOV strain Kikwit when administered 1 day before challenge and 4 days after challenge 78. On the other hand, the administration of a combination of two human-mouse chimeric neutralizing mAbs to three nonhuman primates 1 day prior to as well as 1 and 3 days after lethal EBOV challenge, resulted in protection of 1/3 nonhuman primates and prolonged time-to-death in a second animal 79. Passive transfer of polyclonal IgG purified from vaccinated NHPs that survived challenge with either EBOV or MARV as late as 48h after virus challenge protected naïve NHPs against both MARV and EBOV lethal challenge 80. Together with the previously failed trials of monoclonal antibodies, these observations strongly suggested the need to target multiple epitopes on GP in order to achieve protection against EBOV.

In order to develop a multi-specificity antibody cocktail, several EBOV GP-specific mAbs generated using the rVSV/ΔG/GP EBOV vaccine were evaluated in immunocompetent mice and guinea pigs individually or as pools of 3–4 mAbs. In contrast to individually administered mAbs, which were ineffective, pools of 3 mAbs were found to give complete protection in guinea pigs when administered 2 days post infection 81. These studies paved the way for the development of two cocktails of monoclonal antibodies (either chimeric (c) or human (h)) termed MB-003 (clones c13C6, h13F6 and c6D8) and ZMab (clones m1H3, m2G4 and m4G7) that mediated complete protection of nonhuman primates when administered 1–3 days post-EBOV challenge 82. Further refinement of these cocktails led to the generation of ZMapp™ containing the monoclonal antibodies with the highest efficacy (c13C6, c2G4 and c4G7). This combination is a highly effective post-exposure therapeutic that can protect nonhuman primates even when administered 5 days post-exposure to Ebola to symptomatic animals 83 (Table 2). ZMapp has been used to treat 7 patients infected with EBOV, resulting in 5 patients surviving 84. In February 2015, the NIAID initiated Phase I clinical trial evaluations of ZMapp in Liberia and the United States. The study will be conducted on 200 patients positive for Ebola virus infection who will be randomly assigned to one of two groups: the control group will receive the current standard of care while the second group will receive three infusions of ZMapp administered three days apart85.

Conclusion and Future Outlook

EBOV is one of the most feared pathogens due to its high lethality rates. Subversion of innate immune response by Ebola virus occurs at various levels from suppression of IFN response, generation of robust cytokine storm, and inhibition of dendritic cell maturation. Dysregulation of innate immune pathways together with the characteristic lymphopenia of Ebola prevent the development of cellular and humoral responses as evident by lack of EBOV-specific T and B cell responses, and sequestration of neutralizing antibodies. The current Ebola outbreak in West Africa has spurred a public health emergency and highlighted the need for licensed vaccines and post-exposure therapeutics. With the substantial amount of data pointing to the critical role for antibody-mediated protection against Ebola, it is imperative that research efforts focus on evaluating the safety and efficacy of monoclonal antibody cocktails. The highly promising antibody cocktail ZMapp is grown in tobacco plants, which is thought to take less time and money to produce than in rodents. However, clinical evaluations of ZMapp were delayed until early of 2015 due to the slow production of ZMapp. The production of one course treatment (14g) requires 78 tobacco plants to produce 86. Consequently, efforts must be allocated to developing novel methods to increase yields of the monoclonal antibody cocktail. Although using more traditional methods of generating monoclonal antibodies in rodents may be a slower route than tobacco plant production, this established technique might enable increased production in order to ensure enough of the treatment is available for further clinical evaluations and future outbreaks.

In addition to monoclonal cocktails, efforts to evaluate and license the rVSV/ΔG/GP EBOV vaccine should be increased. Compared to the rAd vaccine platform, advantages of the rVSV/ΔG/GP EBOV platform include limited pre-existing immunity, the ability to administer it mucosally, long duration of immunity after one dose, and its potential use as a post-exposure vaccination. With the Ebola outbreak slowing down, the opportunity to obtain efficacy data through human trials may decrease. In this case, the FDA animal rule should be used, in which laboratory animal data is used to show efficacy, in order to move vaccine candidates through the approval process and start stockpiling the most promising candidate. This will ensure that we have immediate immunization available to protect frontline healthcare workers and contain the spread of a future outbreak.

The 2014 outbreak highlights that Ebola virus disease in not limited to central Africa and is a global problem that requires global cooperation. The World Health Organization’s role in the event of an outbreak should be to effectively coordinate regional collaboration where data and expertise relevant to the outbreak in addition to critical information about disease surveillance is shared. Efforts to improve the infrastructure of healthcare in African nations should be undertaken, such as developing inexpensive systems to equip hospitals to be able to manage routine healthcare needs as well as outbreaks. Educating the public is also equally important, as many individuals in Ebola outbreak countries are fearful of modern health care, preventing those who have come into contact with the disease to seek health care and contributing the spread of the virus. Therefore, increased training of local health care force to build trust and effectively communicate with the public in times of crisis is urgently required. Community engagement is absolutely required especially when preventive measures are at odds with cultural beliefs and religious practices. Public health education regarding the dynamics of Ebola transmission is also urgently needed, especially with traditional burial practices, which involves washing and touching of the deceased. It is clear that our approaches in combatting Ebola virus disease will engage many areas ranging from vaccine and therapeutic discovery to improving healthcare infrastructure, training and community participation.

References

  • 1.(a) Muhlberger E; Lotfering B; Klenk HD; Becker S, Three of the four nucleocapsid proteins of Marburg virus, NP, VP35, and L, are sufficient to mediate replication and transcription of Marburg virus-specific monocistronic minigenomes. Journal of virology 1998, 72 (11), 8756–64 [DOI] [PMC free article] [PubMed] [Google Scholar]; (b) Dolnik O; Kolesnikova L; Becker S, Filoviruses: Interactions with the host cell. Cellular and molecular life sciences : CMLS 2008, 65 (5), 756–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.(a) Ruigrok RW; Schoehn G; Dessen A; Forest E; Volchkov V; Dolnik O; Klenk HD; Weissenhorn W, Structural characterization and membrane binding properties of the matrix protein VP40 of Ebola virus. Journal of molecular biology 2000, 300 (1), 103–12 [DOI] [PubMed] [Google Scholar]; (b) Huang Y; Xu L; Sun Y; Nabel GJ, The assembly of Ebola virus nucleocapsid requires virion-associated proteins 35 and 24 and posttranslational modification of nucleoprotein. Molecular cell 2002, 10 (2), 307–16 [DOI] [PubMed] [Google Scholar]; (c) Hartlieb B; Weissenhorn W, Filovirus assembly and budding. Virology 2006, 344 (1), 64–70. [DOI] [PubMed] [Google Scholar]
  • 3.Kawaoka Y, How Ebola virus infects cells. The New England journal of medicine 2005, 352 (25), 2645–6. [DOI] [PubMed] [Google Scholar]
  • 4.Volchkov VE; Volchkova VA; Slenczka W; Klenk HD; Feldmann H, Release of viral glycoproteins during Ebola virus infection. Virology 1998, 245 (1), 110–9. [DOI] [PubMed] [Google Scholar]
  • 5.(a) Kuhn JH; Bao Y; Bavari S; Becker S; Bradfute S; Brauburger K; Rodney Brister J; Bukreyev AA; Cai Y; Chandran K; Davey RA; Dolnik O; Dye JM; Enterlein S; Gonzalez JP; Formenty P; Freiberg AN; Hensley LE; Hoenen T; Honko AN; Ignatyev GM; Jahrling PB; Johnson KM; Klenk HD; Kobinger G; Lackemeyer MG; Leroy EM; Lever MS; Muhlberger E; Netesov SV; Olinger GG; Palacios G; Patterson JL; Paweska JT; Pitt L; Radoshitzky SR; Ryabchikova EI; Saphire EO; Shestopalov AM; Smither SJ; Sullivan NJ; Swanepoel R; Takada A; Towner JS; van der Groen G; Volchkov VE; Volchkova VA; Wahl-Jensen V; Warren TK; Warfield KL; Weidmann M; Nichol ST, Virus nomenclature below the species level: a standardized nomenclature for filovirus strains and variants rescued from cDNA. Archives of virology 2014, 159 (5), 1229–37 [DOI] [PMC free article] [PubMed] [Google Scholar]; (b) Lefebvre A; Fiet C; Belpois-Duchamp C; Tiv M; Astruc K; Aho Glele LS, Case fatality rates of Ebola virus diseases: a meta-analysis of World Health Organization data. Medecine et maladies infectieuses 2014, 44 (9), 412–6. [DOI] [PubMed] [Google Scholar]
  • 6.(a) Feldmann H; Geisbert TW, Ebola haemorrhagic fever. Lancet 2011, 377 (9768), 849–62 [DOI] [PMC free article] [PubMed] [Google Scholar]; (b) Bwaka MA; Bonnet MJ; Calain P; Colebunders R; De Roo A; Guimard Y; Katwiki KR; Kibadi K; Kipasa MA; Kuvula KJ; Mapanda BB; Massamba M; Mupapa KD; Muyembe-Tamfum JJ; Ndaberey E; Peters CJ; Rollin PE; Van den Enden E; Van den Enden E, Ebola hemorrhagic fever in Kikwit, Democratic Republic of the Congo: clinical observations in 103 patients. The Journal of infectious diseases 1999, 179 Suppl 1, S1–7. [DOI] [PubMed] [Google Scholar]
  • 7.(a)System Ebola Epidemiology Team, I. M., Update: ebola virus disease epidemic - west Africa, november 2014. MMWR. Morbidity and mortality weekly report 2014, 63 (46), 1064–6 [PMC free article] [PubMed] [Google Scholar]; (b) Team, W. H. O. E. R., Ebola virus disease in West Africa--the first 9 months of the epidemic and forward projections. N Engl J Med 2014, 371 (16), 1481–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Baize S; Pannetier D; Oestereich L; Rieger T; Koivogui L; Magassouba N; Soropogui B; Sow MS; Keita S; De Clerck H; Tiffany A; Dominguez G; Loua M; Traore A; Kolie M; Malano ER; Heleze E; Bocquin A; Mely S; Raoul H; Caro V; Cadar D; Gabriel M; Pahlmann M; Tappe D; Schmidt-Chanasit J; Impouma B; Diallo AK; Formenty P; Van Herp M; Gunther S, Emergence of Zaire Ebola virus disease in Guinea. The New England journal of medicine 2014, 371 (15), 1418–25. [DOI] [PubMed] [Google Scholar]
  • 9.Gire SK; Goba A; Andersen KG; Sealfon RS; Park DJ; Kanneh L; Jalloh S; Momoh M; Fullah M; Dudas G; Wohl S; Moses LM; Yozwiak NL; Winnicki S; Matranga CB; Malboeuf CM; Qu J; Gladden AD; Schaffner SF; Yang X; Jiang PP; Nekoui M; Colubri A; Coomber MR; Fonnie M; Moigboi A; Gbakie M; Kamara FK; Tucker V; Konuwa E; Saffa S; Sellu J; Jalloh AA; Kovoma A; Koninga J; Mustapha I; Kargbo K; Foday M; Yillah M; Kanneh F; Robert W; Massally JL; Chapman SB; Bochicchio J; Murphy C; Nusbaum C; Young S; Birren BW; Grant DS; Scheiffelin JS; Lander ES; Happi C; Gevao SM; Gnirke A; Rambaut A; Garry RF; Khan SH; Sabeti PC, Genomic surveillance elucidates Ebola virus origin and transmission during the 2014 outbreak. Science 2014, 345 (6202), 1369–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.(a) Jaax NK; Davis KJ; Geisbert TJ; Vogel P; Jaax GP; Topper M; Jahrling PB, Lethal experimental infection of rhesus monkeys with Ebola-Zaire (Mayinga) virus by the oral and conjunctival route of exposure. Arch Pathol Lab Med 1996, 120 (2), 140–55 [PubMed] [Google Scholar]; (b) Geisbert TW; Hensley LE; Larsen T; Young HA; Reed DS; Geisbert JB; Scott DP; Kagan E; Jahrling PB; Davis KJ, Pathogenesis of Ebola hemorrhagic fever in cynomolgus macaques: evidence that dendritic cells are early and sustained targets of infection. The American journal of pathology 2003, 163 (6), 2347–70 [DOI] [PMC free article] [PubMed] [Google Scholar]; (c) Geisbert TW; Young HA; Jahrling PB; Davis KJ; Larsen T; Kagan E; Hensley LE, Pathogenesis of Ebola hemorrhagic fever in primate models: evidence that hemorrhage is not a direct effect of virus-induced cytolysis of endothelial cells. The American journal of pathology 2003, 163 (6), 2371–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.(a) Bray M; Davis K; Geisbert T; Schmaljohn C; Huggins J, A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. The Journal of infectious diseases 1998, 178 (3), 651–61 [DOI] [PubMed] [Google Scholar]; (b) Connolly BM; Steele KE; Davis KJ; Geisbert TW; Kell WM; Jaax NK; Jahrling PB, Pathogenesis of experimental Ebola virus infection in guinea pigs. The Journal of infectious diseases 1999, 179 Suppl 1, S203–17. [DOI] [PubMed] [Google Scholar]
  • 12.Rasmussen AL; Okumura A; Ferris MT; Green R; Feldmann F; Kelly SM; Scott DP; Safronetz D; Haddock E; LaCasse R; Thomas MJ; Sova P; Carter VS; Weiss JM; Miller DR; Shaw GD; Korth MJ; Heise MT; Baric RS; de Villena FP; Feldmann H; Katze MG, Host genetic diversity enables Ebola hemorrhagic fever pathogenesis and resistance. Science 2014, 346 (6212), 987–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bray M; Hatfill S; Hensley L; Huggins JW, Haematological, biochemical and coagulation changes in mice, guinea-pigs and monkeys infected with a mouse-adapted variant of Ebola Zaire virus. Journal of comparative pathology 2001, 125 (4), 243–53. [DOI] [PubMed] [Google Scholar]
  • 14.Ebihara H; Takada A; Kobasa D; Jones S; Neumann G; Theriault S; Bray M; Feldmann H; Kawaoka Y, Molecular determinants of Ebola virus virulence in mice. PLoS pathogens 2006, 2 (7), e73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Geisbert TW; Hensley LE; Gibb TR; Steele KE; Jaax NK; Jahrling PB, Apoptosis induced in vitro and in vivo during infection by Ebola and Marburg viruses. Laboratory investigation; a journal of technical methods and pathology 2000, 80 (2), 171–86. [DOI] [PubMed] [Google Scholar]
  • 16.Stroher U; West E; Bugany H; Klenk HD; Schnittler HJ; Feldmann H, Infection and activation of monocytes by Marburg and Ebola viruses. Journal of virology 2001, 75 (22), 11025–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.(a) Baize S; Leroy EM; Georges AJ; Georges-Courbot MC; Capron M; Bedjabaga I; Lansoud-Soukate J; Mavoungou E, Inflammatory responses in Ebola virus-infected patients. Clinical & Experimental Immunology 2002, 128 (1), 163–168 [DOI] [PMC free article] [PubMed] [Google Scholar]; (b) Ebihara H; Rockx B; Marzi A; Feldmann F; Haddock E; Brining D; LaCasse RA; Gardner D; Feldmann H, Host response dynamics following lethal infection of rhesus macaques with Zaire ebolavirus. The Journal of infectious diseases 2011, 204 Suppl 3, S991–9 [DOI] [PMC free article] [PubMed] [Google Scholar]; (c) Hensley LE; Young HA; Jahrling PB; Geisbert TW, Proinflammatory response during Ebola virus infection of primate models: possible involvement of the tumor necrosis factor receptor superfamily. Immunol Lett 2002, 80 (3), 169–79 [DOI] [PubMed] [Google Scholar]; (d) Sanchez A; Lukwiya M; Bausch D; Mahanty S; Sanchez AJ; Wagoner KD; Rollin PE, Analysis of human peripheral blood samples from fatal and nonfatal cases of Ebola (Sudan) hemorrhagic fever: cellular responses, virus load, and nitric oxide levels. Journal of virology 2004, 78 (19), 10370–7 [DOI] [PMC free article] [PubMed] [Google Scholar]; (e) Villinger F; Rollin PE; Brar SS; Chikkala NF; Winter J; Sundstrom JB; Zaki SR; Swanepoel R; Ansari AA; Peters CJ, Markedly elevated levels of interferon (IFN)-gamma, IFN-alpha, interleukin (IL)-2, IL-10, and tumor necrosis factor-alpha associated with fatal Ebola virus infection. The Journal of infectious diseases 1999, 179 Suppl 1, S188–91 [DOI] [PubMed] [Google Scholar]; (f) Wauquier N; Becquart P; Padilla C; Baize S; Leroy EM, Human fatal zaire ebola virus infection is associated with an aberrant innate immunity and with massive lymphocyte apoptosis. PLoS neglected tropical diseases 2010, 4 (10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.(a) Reid SP; Valmas C; Martinez O; Sanchez FM; Basler CF, Ebola virus VP24 proteins inhibit the interaction of NPI-1 subfamily karyopherin alpha proteins with activated STAT1. Journal of virology 2007, 81 (24), 13469–77 [DOI] [PMC free article] [PubMed] [Google Scholar]; (b) Reid SP; Leung LW; Hartman AL; Martinez O; Shaw ML; Carbonnelle C; Volchkov VE; Nichol ST; Basler CF, Ebola virus VP24 binds karyopherin alpha1 and blocks STAT1 nuclear accumulation. Journal of virology 2006, 80 (11), 5156–67 [DOI] [PMC free article] [PubMed] [Google Scholar]; (c) Mateo M; Reid SP; Leung LW; Basler CF; Volchkov VE, Ebolavirus VP24 binding to karyopherins is required for inhibition of interferon signaling. Journal of virology 2010, 84 (2), 1169–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Xu W; Edwards MR; Borek DM; Feagins AR; Mittal A; Alinger JB; Berry KN; Yen B; Hamilton J; Brett TJ; Pappu RV; Leung DW; Basler CF; Amarasinghe GK, Ebola virus VP24 targets a unique NLS binding site on karyopherin alpha 5 to selectively compete with nuclear import of phosphorylated STAT1. Cell Host Microbe 2014, 16 (2), 187–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Basler CF; Mikulasova A; Martinez-Sobrido L; Paragas J; Muhlberger E; Bray M; Klenk HD; Palese P; Garcia-Sastre A, The Ebola virus VP35 protein inhibits activation of interferon regulatory factor 3. Journal of virology 2003, 77 (14), 7945–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chang TH; Kubota T; Matsuoka M; Jones S; Bradfute SB; Bray M; Ozato K, Ebola Zaire virus blocks type I interferon production by exploiting the host SUMO modification machinery. PLoS pathogens 2009, 5 (6), e1000493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Cardenas WB; Loo YM; Gale M Jr.; Hartman AL; Kimberlin CR; Martinez-Sobrido L; Saphire EO; Basler CF, Ebola virus VP35 protein binds double-stranded RNA and inhibits alpha/beta interferon production induced by RIG-I signaling. Journal of virology 2006, 80 (11), 5168–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Luthra P; Ramanan P; Mire CE; Weisend C; Tsuda Y; Yen B; Liu G; Leung DW; Geisbert TW; Ebihara H; Amarasinghe GK; Basler CF, Mutual antagonism between the Ebola virus VP35 protein and the RIG-I activator PACT determines infection outcome. Cell host & microbe 2013, 14 (1), 74–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Prins KC; Cardenas WB; Basler CF, Ebola virus protein VP35 impairs the function of interferon regulatory factor-activating kinases IKKepsilon and TBK-1. Journal of virology 2009, 83 (7), 3069–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lubaki NM; Ilinykh P; Pietzsch C; Tigabu B; Freiberg AN; Koup RA; Bukreyev A, The lack of maturation of Ebola virus-infected dendritic cells results from the cooperative effect of at least two viral domains. Journal of virology 2013, 87 (13), 7471–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mahanty S; Hutchinson K; Agarwal S; McRae M; Rollin PE; Pulendran B, Cutting edge: impairment of dendritic cells and adaptive immunity by Ebola and Lassa viruses. Journal of immunology 2003, 170 (6), 2797–801. [DOI] [PubMed] [Google Scholar]
  • 27.Gupta M; Spiropoulou C; Rollin PE, Ebola virus infection of human PBMCs causes massive death of macrophages, CD4 and CD8 T cell sub-populations in vitro. Virology 2007, 364 (1), 45–54. [DOI] [PubMed] [Google Scholar]
  • 28.Bradfute SB; Braun DR; Shamblin JD; Geisbert JB; Paragas J; Garrison A; Hensley LE; Geisbert TW, Lymphocyte death in a mouse model of Ebola virus infection. The Journal of infectious diseases 2007, 196 Suppl 2, S296–304. [DOI] [PubMed] [Google Scholar]
  • 29.Reed DS; Hensley LE; Geisbert JB; Jahrling PB; Geisbert TW, Depletion of peripheral blood T lymphocytes and NK cells during the course of ebola hemorrhagic Fever in cynomolgus macaques. Viral immunology 2004, 17 (3), 390–400. [DOI] [PubMed] [Google Scholar]
  • 30.Mehedi M; Falzarano D; Seebach J; Hu X; Carpenter MS; Schnittler HJ; Feldmann H, A new Ebola virus nonstructural glycoprotein expressed through RNA editing. Journal of virology 2011, 85 (11), 5406–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.(a) Baize S; Leroy EM; Mavoungou E; Fisher-Hoch SP, Apoptosis in fatal Ebola infection. Does the virus toll the bell for immune system? Apoptosis : an international journal on programmed cell death 2000, 5 (1), 5–7 [DOI] [PubMed] [Google Scholar]; (b) Baize S; Leroy EM; Georges-Courbot MC; Capron M; Lansoud-Soukate J; Debre P; Fisher-Hoch SP; McCormick JB; Georges AJ, Defective humoral responses and extensive intravascular apoptosis are associated with fatal outcome in Ebola virus-infected patients. Nature medicine 1999, 5 (4), 423–6. [DOI] [PubMed] [Google Scholar]
  • 32.(a) Rath PC; Aggarwal BB, TNF-induced signaling in apoptosis. Journal of clinical immunology 1999, 19 (6), 350–64 [DOI] [PubMed] [Google Scholar]; (b) Simon HU; Haj-Yehia A; Levi-Schaffer F, Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis : an international journal on programmed cell death 2000, 5 (5), 415–8 [DOI] [PubMed] [Google Scholar]; (c) Snyder CM; Shroff EH; Liu J; Chandel NS, Nitric oxide induces cell death by regulating anti-apoptotic BCL-2 family members. PloS one 2009, 4 (9), e7059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Leroy EM; Baize S; Debre P; Lansoud-Soukate J; Mavoungou E, Early immune responses accompanying human asymptomatic Ebola infections. Clinical & Experimental Immunology 2001, 124 (3), 453–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Leroy EM; Baize S; Volchkov VE; Fisher-Hoch SP; Georges-Courbot MC; Lansoud-Soukate J; Capron M; Debre P; McCormick JB; Georges AJ, Human asymptomatic Ebola infection and strong inflammatory response. Lancet 2000, 355 (9222), 2210–5. [DOI] [PubMed] [Google Scholar]
  • 35.Mohan GS; Li W; Ye L; Compans RW; Yang C, Antigenic subversion: a novel mechanism of host immune evasion by Ebola virus. PLoS pathogens 2012, 8 (12), e1003065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mupapa K; Massamba M; Kibadi K; Kuvula K; Bwaka A; Kipasa M; Colebunders R; Muyembe-Tamfum JJ, Treatment of Ebola hemorrhagic fever with blood transfusions from convalescent patients. International Scientific and Technical Committee. The Journal of infectious diseases 1999, 179 Suppl 1, S18–23. [DOI] [PubMed] [Google Scholar]
  • 37.Gupta M; Mahanty S; Bray M; Ahmed R; Rollin PE, Passive transfer of antibodies protects immunocompetent and imunodeficient mice against lethal Ebola virus infection without complete inhibition of viral replication. J Virol 2001, 75 (10), 4649–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Friedrich BM; Trefry JC; Biggins JE; Hensley LE; Honko AN; Smith DR; Olinger GG, Potential vaccines and post-exposure treatments for filovirus infections. Viruses 2012, 4 (9), 1619–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sullivan NJ; Geisbert TW; Geisbert JB; Shedlock DJ; Xu L; Lamoreaux L; Custers JH; Popernack PM; Yang ZY; Pau MG; Roederer M; Koup RA; Goudsmit J; Jahrling PB; Nabel GJ, Immune protection of nonhuman primates against Ebola virus with single low-dose adenovirus vectors encoding modified GPs. PLoS medicine 2006, 3 (6), e177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sullivan NJ; Hensley L; Asiedu C; Geisbert TW; Stanley D; Johnson J; Honko A; Olinger G; Bailey M; Geisbert JB; Reimann KA; Bao S; Rao S; Roederer M; Jahrling PB; Koup RA; Nabel GJ, CD8+ cellular immunity mediates rAd5 vaccine protection against Ebola virus infection of nonhuman primates. Nat Med 2011, 17 (9), 1128–1131. [DOI] [PubMed] [Google Scholar]
  • 41.Stanley DA; Honko AN; Asiedu C; Trefry JC; Lau-Kilby AW; Johnson JC; Hensley L; Ammendola V; Abbate A; Grazioli F; Foulds KE; Cheng C; Wang L; Donaldson MM; Colloca S; Folgori A; Roederer M; Nabel GJ; Mascola J; Nicosia A; Cortese R; Koup RA; Sullivan NJ, Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge. Nature medicine 2014, 20 (10), 1126–9. [DOI] [PubMed] [Google Scholar]
  • 42.Ledgerwood JE; DeZure AD; Stanley DA; Novik L; Enama ME; Berkowitz NM; Hu Z; Joshi G; Ploquin A; Sitar S; Gordon IJ; Plummer SA; Holman LA; Hendel CS; Yamshchikov G; Roman F; Nicosia A; Colloca S; Cortese R; Bailer RT; Schwartz RM; Roederer M; Mascola JR; Koup RA; Sullivan NJ; Graham BS; the VRCST, Chimpanzee Adenovirus Vector Ebola Vaccine - Preliminary Report. The New England journal of medicine 2014. [DOI] [PubMed] [Google Scholar]
  • 43.Diseases, N. I. o. A. a. I. PREVAIL Phase2/3 Clinical Trial of Investigational Ebola Vaccines. http://www.niaid.nih.gov/news/QA/Pages/EbolaVaxResultsQA.aspx (accessed March 7, 2015).
  • 44.Stover K NIH to Launch Human Safety Study of Ebola Vaccine Candidate. http://www.niaid.nih.gov/news/newsreleases/2014/Pages/EbolaVaxCandidate.aspx (accessed December 12, 2014).
  • 45.(a) Jones SM; Feldmann H; Stroher U; Geisbert JB; Fernando L; Grolla A; Klenk HD; Sullivan NJ; Volchkov VE; Fritz EA; Daddario KM; Hensley LE; Jahrling PB; Geisbert TW, Live attenuated recombinant vaccine protects nonhuman primates against Ebola and Marburg viruses. Nat Med 2005, 11 (7), 786–90 [DOI] [PubMed] [Google Scholar]; (b) Jones SM; Stroher U; Fernando L; Qiu X; Alimonti J; Melito P; Bray M; Klenk HD; Feldmann H, Assessment of a vesicular stomatitis virus-based vaccine by use of the mouse model of Ebola virus hemorrhagic fever. J Infect Dis 2007, 196 Suppl 2, S404–12. [DOI] [PubMed] [Google Scholar]
  • 46.Marzi A; Engelmann F; Feldmann F; Haberthur K; Shupert WL; Brining D; Scott DP; Geisbert TW; Kawaoka Y; Katze MG; Feldmann H; Messaoudi I, Antibodies are necessary for rVSV/ZEBOV-GP-mediated protection against lethal Ebola virus challenge in nonhuman primates. Proceedings of the National Academy of Sciences of the United States of America 2013, 110 (5), 1893–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.(a) Geisbert TW; Daddario-Dicaprio KM; Geisbert JB; Reed DS; Feldmann F; Grolla A; Stroher U; Fritz EA; Hensley LE; Jones SM; Feldmann H, Vesicular stomatitis virus-based vaccines protect nonhuman primates against aerosol challenge with Ebola and Marburg viruses. Vaccine 2008, 26 (52), 6894–900 [DOI] [PMC free article] [PubMed] [Google Scholar]; (b) Geisbert TW; Daddario-Dicaprio KM; Lewis MG; Geisbert JB; Grolla A; Leung A; Paragas J; Matthias L; Smith MA; Jones SM; Hensley LE; Feldmann H; Jahrling PB, Vesicular stomatitis virus-based ebola vaccine is well-tolerated and protects immunocompromised nonhuman primates. PLoS pathogens 2008, 4 (11), e1000225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Qiu X; Fernando L; Alimonti JB; Melito PL; Feldmann F; Dick D; Stroher U; Feldmann H; Jones SM, Mucosal immunization of cynomolgus macaques with the VSVDeltaG/ZEBOVGP vaccine stimulates strong ebola GP-specific immune responses. PloS one 2009, 4 (5), e5547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wong G; Audet J; Fernando L; Fausther-Bovendo H; Alimonti JB; Kobinger GP; Qiu X, Immunization with vesicular stomatitis virus vaccine expressing the Ebola glycoprotein provides sustained long-term protection in rodents. Vaccine 2014, 32 (43), 5722–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mire CE; Geisbert JB; Agans KN; Satterfield BA; Versteeg KM; Fritz EA; Feldmann H; Hensley LE; Geisbert TW, Durability of a vesicular stomatitis virus-based marburg virus vaccine in nonhuman primates. PloS one 2014, 9 (4), e94355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Geisbert TW; Geisbert JB; Leung A; Daddario-DiCaprio KM; Hensley LE; Grolla A; Feldmann H, Single-injection vaccine protects nonhuman primates against infection with marburg virus and three species of ebola virus. Journal of virology 2009, 83 (14), 7296–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.(a) Feldmann H; Jones SM; Daddario-DiCaprio KM; Geisbert JB; Stroher U; Grolla A; Bray M; Fritz EA; Fernando L; Feldmann F; Hensley LE; Geisbert TW, Effective post-exposure treatment of Ebola infection. PLoS pathogens 2007, 3 (1), e2. [DOI] [PMC free article] [PubMed] [Google Scholar]; (b) Geisbert TW; Daddario-DiCaprio KM; Williams KJ; Geisbert JB; Leung A; Feldmann F; Hensley LE; Feldmann H; Jones SM, Recombinant vesicular stomatitis virus vector mediates postexposure protection against Sudan Ebola hemorrhagic fever in nonhuman primates. J Virol 2008, 82 (11), 5664–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gunther S; Feldmann H; Geisbert TW; Hensley LE; Rollin PE; Nichol ST; Stroher U; Artsob H; Peters CJ; Ksiazek TG; Becker S; ter Meulen J; Olschlager S; Schmidt-Chanasit J; Sudeck H; Burchard GD; Schmiedel S, Management of accidental exposure to Ebola virus in the biosafety level 4 laboratory, Hamburg, Germany. The Journal of infectious diseases 2011, 204 Suppl 3, S785–90. [DOI] [PubMed] [Google Scholar]
  • 54.Lai L; Davey R; Beck A; Xu Y; Suffredini AF; Palmore T; Kabbani S; Rogers S; Kobinger G; Alimonti J; Link CJ Jr.; Rubinson L; Stroher U; Wolcott M; Dorman W; Uyeki TM; Feldmann H; Lane HC; Mulligan MJ, Emergency Postexposure Vaccination With Vesicular Stomatitis Virus-Vectored Ebola Vaccine After Needlestick. Jama 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.NIH STRIVE (Sierra Leone Trial to Introduce a Vaccine Against Ebola). https://clinicaltrials.gov/ct2/show/record/NCT02378753 (accessed March 7, 2015).
  • 56.Chaib F; Jasarevic T; Harris M WHO welcomes approval of a second Ebola vaccine trial in Switzerland. http://www.who.int/mediacentre/news/statements/2014/second-ebola-vaccine/en/ (accessed December 12, 2014).
  • 57.Fox M Joint Pain Interrupts Ebola Vaccine Trial. http://www.nbcnews.com/storyline/ebola-virus-outbreak/joint-pain-interrupts-ebola-vaccine-trial-n266516 (accessed Deceember 12, 2014).
  • 58.Stover K NIH Begins Early Human Clinical Trial of VSV Ebola Vaccine. http://www.niaid.nih.gov/news/newsreleases/2014/Pages/VSVEbolaVaccine.aspx (accessed December 12, 2014).
  • 59.(a) Geisbert TW; Hensley LE; Jahrling PB; Larsen T; Geisbert JB; Paragas J; Young HA; Fredeking TM; Rote WE; Vlasuk GP, Treatment of Ebola virus infection with a recombinant inhibitor of factor VIIa/tissue factor: a study in rhesus monkeys. Lancet 2003, 362 (9400), 1953–8 [DOI] [PubMed] [Google Scholar]; (b) Hensley LE; Stevens EL; Yan SB; Geisbert JB; Macias WL; Larsen T; Daddario-DiCaprio KM; Cassell GH; Jahrling PB; Geisbert TW, Recombinant human activated protein C for the postexposure treatment of Ebola hemorrhagic fever. The Journal of infectious diseases 2007, 196 Suppl 2, S390–9. [DOI] [PubMed] [Google Scholar]
  • 60.Lee A; Agnelli G; Buller H; Ginsberg J; Heit J; Rote W; Vlasuk G; Costantini L; Julian J; Comp P; van Der Meer J; Piovella F; Raskob G; Gent M, Dose-response study of recombinant factor VIIa/tissue factor inhibitor recombinant nematode anticoagulant protein c2 in prevention of postoperative venous thromboembolism in patients undergoing total knee replacement. Circulation 2001, 104 (1), 74–8. [DOI] [PubMed] [Google Scholar]
  • 61.Moons AH; Peters RJ; Bijsterveld NR; Piek JJ; Prins MH; Vlasuk GP; Rote WE; Buller HR, Recombinant nematode anticoagulant protein c2, an inhibitor of the tissue factor/factor VIIa complex, in patients undergoing elective coronary angioplasty. Journal of the American College of Cardiology 2003, 41 (12), 2147–53. [DOI] [PubMed] [Google Scholar]
  • 62.Smith LM; Hensley LE; Geisbert TW; Johnson J; Stossel A; Honko A; Yen JY; Geisbert J; Paragas J; Fritz E; Olinger G; Young HA; Rubins KH; Karp CL, Interferon-beta therapy prolongs survival in rhesus macaque models of Ebola and Marburg hemorrhagic fever. The Journal of infectious diseases 2013, 208 (2), 310–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Oestereich L; Ludtke A; Wurr S; Rieger T; Munoz-Fontela C; Gunther S, Successful treatment of advanced Ebola virus infection with T-705 (favipiravir) in a small animal model. Antiviral research 2014, 105, 17–21. [DOI] [PubMed] [Google Scholar]
  • 64.Trompiz G; Lawson H; Powell S First French Ebola patient leaves hospital. http://www.reuters.com/article/2014/10/04/us-health-ebola-france-idUSKCN0HT0D720141004. [Google Scholar]
  • 65.(a) WHO Ebola vaccines, therapies, and diagnostics. http://www.who.int/medicines/emp_ebola_q_as/en/; (b) NIH Efficacy of Favipiravir Against Ebola (JIKI). https://clinicaltrials.gov/ct2/show/record/NCT02329054?term=favipiravir+ebola&rank=1 (accessed March 7, 2015).
  • 66.Warren TK; Wells J; Panchal RG; Stuthman KS; Garza NL; Van Tongeren SA; Dong L; Retterer CJ; Eaton BP; Pegoraro G; Honnold S; Bantia S; Kotian P; Chen X; Taubenheim BR; Welch LS; Minning DM; Babu YS; Sheridan WP; Bavari S, Protection against filovirus diseases by a novel broad-spectrum nucleoside analogue BCX4430. Nature 2014, 508 (7496), 402–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.NIH A Phase 1 Study to Evaluate the Safety, Tolerability and Pharmacokinetics of BCX4430. https://clinicaltrials.gov/ct2/show/NCT02319772?term=BCX4430&rank=1 (accessed March 7, 2015).
  • 68.Schepers JT Chimerix Announces Emergency Investigational New Drug Applications for Brincidofovir Authorized by FDA for Patients with Ebola Virus Disease. http://ir.chimerix.com/releasedetail.cfm?releaseid=874647 (accessed December 12, 2014).
  • 69.Health, N. I. o. An Open-Label, Multicenter Study of the Safety and Anti Viral Activity of Brincidofovir (BCV, CMX001) for Ebola Virus Disease. https://clinicaltrials.gov/ct2/show/NCT02271347?term=chimerix&rank=4 (accessed December 13, 2014).
  • 70.Warren TK; Warfield KL; Wells J; Swenson DL; Donner KS; Van Tongeren SA; Garza NL; Dong L; Mourich DV; Crumley S; Nichols DK; Iversen PL; Bavari S, Advanced antisense therapies for postexposure protection against lethal filovirus infections. Nature medicine 2010, 16 (9), 991–4. [DOI] [PubMed] [Google Scholar]
  • 71.Warren TK; Whitehouse CA; Wells J; Welch L; Heald AE; Charleston JS; Sazani P; Reid SP; Iversen PL; Bavari S, A Single Phosphorodiamidate Morpholino Oligomer Targeting VP24 Protects Rhesus Monkeys against Lethal Ebola Virus Infection. mBio 2015, 6 (1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Heald AE; Iversen PL; Saoud JB; Sazani P; Charleston JS; Axtelle T; Wong M; Smith WB; Vutikullird A; Kaye E, Safety and pharmacokinetic profiles of phosphorodiamidate morpholino oligomers with activity against ebola virus and marburg virus: results of two single-ascending-dose studies. Antimicrobial agents and chemotherapy 2014, 58 (11), 6639–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Fowler T; Bamberg S; Moller P; Klenk HD; Meyer TF; Becker S; Rudel T, Inhibition of Marburg virus protein expression and viral release by RNA interference. The Journal of general virology 2005, 86 (Pt 4), 1181–8. [DOI] [PubMed] [Google Scholar]
  • 74.Geisbert TW; Hensley LE; Kagan E; Yu EZ; Geisbert JB; Daddario-DiCaprio K; Fritz EA; Jahrling PB; McClintock K; Phelps JR; Lee AC; Judge A; Jeffs LB; MacLachlan I, Postexposure protection of guinea pigs against a lethal ebola virus challenge is conferred by RNA interference. The Journal of infectious diseases 2006, 193 (12), 1650–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Geisbert TW; Lee AC; Robbins M; Geisbert JB; Honko AN; Sood V; Johnson JC; de Jong S; Tavakoli I; Judge A; Hensley LE; Maclachlan I, Postexposure protection of non-human primates against a lethal Ebola virus challenge with RNA interference: a proof-of-concept study. Lancet 2010, 375 (9729), 1896–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Young S FDA allows expanded use of experimental Ebola drug. http://www.cnn.com/2014/09/22/health/ebola-drug-tekmira/ (accessed December 12, 2014).
  • 77.Parren PW; Geisbert TW; Maruyama T; Jahrling PB; Burton DR, Pre- and postexposure prophylaxis of Ebola virus infection in an animal model by passive transfer of a neutralizing human antibody. Journal of virology 2002, 76 (12), 6408–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Oswald WB; Geisbert TW; Davis KJ; Geisbert JB; Sullivan NJ; Jahrling PB; Parren PW; Burton DR, Neutralizing antibody fails to impact the course of Ebola virus infection in monkeys. PLoS pathogens 2007, 3 (1), e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Marzi A; Yoshida R; Miyamoto H; Ishijima M; Suzuki Y; Higuchi M; Matsuyama Y; Igarashi M; Nakayama E; Kuroda M; Saijo M; Feldmann F; Brining D; Feldmann H; Takada A, Protective Efficacy of Neutralizing Monoclonal Antibodies in a Nonhuman Primate Model of Ebola Hemorrhagic Fever. PLoS ONE 2012, 7 (4), e36192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dye JM; Herbert AS; Kuehne AI; Barth JF; Muhammad MA; Zak SE; Ortiz RA; Prugar LI; Pratt WD, Postexposure antibody prophylaxis protects nonhuman primates from filovirus disease. Proc Natl Acad Sci U S A 2012, 109 (13), 5034–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Qiu X; Fernando L; Melito PL; Audet J; Feldmann H; Kobinger G; Alimonti JB; Jones SM, Ebola GP-specific monoclonal antibodies protect mice and guinea pigs from lethal Ebola virus infection. PLoS neglected tropical diseases 2012, 6 (3), e1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.(a) Qiu X; Audet J; Wong G; Pillet S; Bello A; Cabral T; Strong JE; Plummer F; Corbett CR; Alimonti JB; Kobinger GP, Successful treatment of ebola virus-infected cynomolgus macaques with monoclonal antibodies. Sci Transl Med 2012, 4 (138), 138ra81. [DOI] [PubMed] [Google Scholar]; (b) Olinger GG Jr.; Pettitt J; Kim D; Working C; Bohorov O; Bratcher B; Hiatt E; Hume SD; Johnson AK; Morton J; Pauly M; Whaley KJ; Lear CM; Biggins JE; Scully C; Hensley L; Zeitlin L, Delayed treatment of Ebola virus infection with plant-derived monoclonal antibodies provides protection in rhesus macaques. Proc Natl Acad Sci U S A 2012, 109 (44), 18030–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Qiu X; Wong G; Audet J; Bello A; Fernando L; Alimonti JB; Fausther-Bovendo H; Wei H; Aviles J; Hiatt E; Johnson A; Morton J; Swope K; Bohorov O; Bohorova N; Goodman C; Kim D; Pauly MH; Velasco J; Pettitt J; Olinger GG; Whaley K; Xu B; Strong JE; Zeitlin L; Kobinger GP, Reversion of advanced Ebola virus disease in nonhuman primates with ZMapp. Nature 2014, 514 (7520), 47–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.McCarthy M, US signs contract with ZMapp maker to accelerate development of the Ebola drug. Bmj 2014, 349, g5488. [DOI] [PubMed] [Google Scholar]
  • 85.Health, N. I. o., Libera-U.S. clinical research partnership opens trial to test Ebola treatments. 2015.
  • 86.Park A See How Ebola Drugs Grow in Tobacco Leaves. http://time.com/3457472/see-how-ebola-drugs-grow-in-tobacco-leaves/ (accessed March 7, 2015).
  • 87.Madrid PB; Chopra S; Manger ID; Gilfillan L; Keepers TR; Shurtleff AC; Green CE; Iyer LV; Dilks HH; Davey RA; Kolokoltsov AA; Carrion R Jr.; Patterson JL; Bavari S; Panchal RG; Warren TK; Wells JB; Moos WH; Burke RL; Tanga MJ, A systematic screen of FDA-approved drugs for inhibitors of biological threat agents. PloS one 2013, 8 (4), e60579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Johansen LM; Brannan JM; Delos SE; Shoemaker CJ; Stossel A; Lear C; Hoffstrom BG; Dewald LE; Schornberg KL; Scully C; Lehar J; Hensley LE; White JM; Olinger GG, FDA-approved selective estrogen receptor modulators inhibit Ebola virus infection. Science translational medicine 2013, 5 (190), 190ra79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Warren TK; Warfield KL; Wells J; Enterlein S; Smith M; Ruthel G; Yunus AS; Kinch MS; Goldblatt M; Aman MJ; Bavari S, Antiviral activity of a small-molecule inhibitor of filovirus infection. Antimicrobial agents and chemotherapy 2010, 54 (5), 2152–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kinch MS; Yunus AS; Lear C; Mao H; Chen H; Fesseha Z; Luo G; Nelson EA; Li L; Huang Z; Murray M; Ellis WY; Hensley L; Christopher-Hennings J; Olinger GG; Goldblatt M, FGI-104: a broad-spectrum small molecule inhibitor of viral infection. American journal of translational research 2009, 1 (1), 87–98. [PMC free article] [PubMed] [Google Scholar]
  • 91.Aman MJ; Kinch MS; Warfield K; Warren T; Yunus A; Enterlein S; Stavale E; Wang P; Chang S; Tang Q; Porter K; Goldblatt M; Bavari S, Development of a broad-spectrum antiviral with activity against Ebola virus. Antiviral research 2009, 83 (3), 245–51. [DOI] [PubMed] [Google Scholar]
  • 92.Bray M; Raymond JL; Geisbert T; Baker RO, 3-deazaneplanocin A induces massively increased interferon-alpha production in Ebola virus-infected mice. Antiviral research 2002, 55 (1), 151–9. [DOI] [PubMed] [Google Scholar]
  • 93.Chang J; Warren TK; Zhao X; Gill T; Guo F; Wang L; Comunale MA; Du Y; Alonzi DS; Yu W; Ye H; Liu F; Guo JT; Mehta A; Cuconati A; Butters TD; Bavari S; Xu X; Block TM, Small molecule inhibitors of ER alpha-glucosidases are active against multiple hemorrhagic fever viruses. Antiviral research 2013, 98 (3), 432–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Panchal RG; Reid SP; Tran JP; Bergeron AA; Wells J; Kota KP; Aman J; Bavari S, Identification of an antioxidant small-molecule with broad-spectrum antiviral activity. Antiviral research 2012, 93 (1), 23–9. [DOI] [PubMed] [Google Scholar]
  • 95.Reid SP; Shurtleff AC; Costantino JA; Tritsch SR; Retterer C; Spurgers KB; Bavari S, HSPA5 is an essential host factor for Ebola virus infection. Antiviral research 2014, 109, 171–4. [DOI] [PubMed] [Google Scholar]
  • 96.Zust R; Cervantes-Barragan L; Habjan M; Maier R; Neuman BW; Ziebuhr J; Szretter KJ; Baker SC; Barchet W; Diamond MS; Siddell SG; Ludewig B; Thiel V, Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5. Nature immunology 2011, 12 (2), 137–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ye W; Schneller SW, The enantiomers of the 1′,6′-isomer of neplanocin A: synthesis and antiviral properties. Bioorganic & medicinal chemistry 2014, 22 (19), 5315–9. [DOI] [PubMed] [Google Scholar]
  • 98.Cote M; Misasi J; Ren T; Bruchez A; Lee K; Filone CM; Hensley L; Li Q; Ory D; Chandran K; Cunningham J, Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection. Nature 2011, 477 (7364), 344–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Basu A; Li B; Mills DM; Panchal RG; Cardinale SC; Butler MM; Peet NP; Majgier-Baranowska H; Williams JD; Patel I; Moir DT; Bavari S; Ray R; Farzan MR; Rong L; Bowlin TL, Identification of a small-molecule entry inhibitor for filoviruses. Journal of virology 2011, 85 (7), 3106–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wolf MC; Freiberg AN; Zhang T; Akyol-Ataman Z; Grock A; Hong PW; Li J; Watson NF; Fang AQ; Aguilar HC; Porotto M; Honko AN; Damoiseaux R; Miller JP; Woodson SE; Chantasirivisal S; Fontanes V; Negrete OA; Krogstad P; Dasgupta A; Moscona A; Hensley LE; Whelan SP; Faull KF; Holbrook MR; Jung ME; Lee B, A broad-spectrum antiviral targeting entry of enveloped viruses. Proceedings of the National Academy of Sciences of the United States of America 2010, 107 (7), 3157–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Filone CM; Hodges EN; Honeyman B; Bushkin GG; Boyd K; Platt A; Ni F; Strom K; Hensley L; Snyder JK; Connor JH, Identification of a broad-spectrum inhibitor of viral RNA synthesis: validation of a prototype virus-based approach. Chemistry & biology 2013, 20 (3), 424–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Gehring G; Rohrmann K; Atenchong N; Mittler E; Becker S; Dahlmann F; Pohlmann S; Vondran FW; David S; Manns MP; Ciesek S; von Hahn T, The clinically approved drugs amiodarone, dronedarone and verapamil inhibit filovirus cell entry. The Journal of antimicrobial chemotherapy 2014, 69 (8), 2123–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hill-Batorski L; Halfmann P; Neumann G; Kawaoka Y, The cytoprotective enzyme heme oxygenase-1 suppresses Ebola virus replication. Journal of virology 2013, 87 (24), 13795–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Fauci Anthony S., F. S. C. NIH Ebola Update: Working Toward Treatments and Vaccines. http://directorsblog.nih.gov/2014/10/14/nih-ebola-update-working-toward-treatments-and-vaccines/.
  • 105.St Vincent MR; Colpitts CC; Ustinov AV; Muqadas M; Joyce MA; Barsby NL; Epand RF; Epand RM; Khramyshev SA; Valueva OA; Korshun VA; Tyrrell DL; Schang LM, Rigid amphipathic fusion inhibitors, small molecule antiviral compounds against enveloped viruses. Proceedings of the National Academy of Sciences of the United States of America 2010, 107 (40), 17339–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Elshabrawy HA; Fan J; Haddad CS; Ratia K; Broder CC; Caffrey M; Prabhakar BS, Identification of a broad-spectrum antiviral small molecule against severe acute respiratory syndrome coronavirus and Ebola, Hendra, and Nipah viruses by using a novel high-throughput screening assay. Journal of virology 2014, 88 (8), 4353–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Fedson DS, A practical treatment for patients with ebola virus disease. The Journal of infectious diseases 2015, 211 (4), 661–2. [DOI] [PubMed] [Google Scholar]

RESOURCES