SUMMARY
The 2014-2015 outbreak of Ebola virus (EBOV), originating from Guinea, is now responsible for the infection of >20,000 people in 9 countries. Whereas past filovirus outbreaks in sub-Saharan Africa have been rapidly brought under control with comparably few cases, this outbreak has been particularly resistant to containment efforts. Both the general population and primary health care workers have been affected by this outbreak, with hundreds of doctors and nurses being infected in the line of duty. In the absence of approved therapeutics, several caregivers have turned to investigational new drugs as well as experimental therapies in an effort to save lives. This review aims to summarize the candidates currently under consideration for postexposure use in infected patients during the largest EBOV outbreak in history.
INTRODUCTION
The genus Ebolavirus (family Filoviridae) consists of five distinct virus species: Ebola virus (EBOV), Sudan virus, Bundibugyo virus, Taï Forest virus, and Reston virus. Along with the related viruses Marburg virus (MARV) and Ravn virus (RAVV) of the Marburgvirus genus, filoviruses are among the deadliest pathogens known to humans and nonhuman primates (NHPs), with case fatality rates in humans reaching as high as 90% in several past outbreaks. The EBOV outbreak in West Africa, identified by the World Health Organization (WHO) in mid-March 2014, is the largest documented filovirus outbreak by a significant margin. The numbers of cases and fatalities from this outbreak have outnumbered those of all past filovirus outbreaks combined. As of 11 March 2015, a total of 9,976 deaths from 24,282 cases have been reported (1), although authorities believe that both numbers are likely underreported because of the inability of overwhelmed responders to report the epidemiological data accurately. One report estimated the outbreak toll to be closer to 2.5 times the number of cases currently being reported by the WHO (2). At the height of the outbreak, the transmission of EBOV was intense and widespread in Guinea, Sierra Leone, and Liberia, with densely populated capitals in all three countries recording cases. Approximately 500 new infections were reported weekly as of early October 2014, with the number expected to double every 30 days if effective interventions are not implemented (3). The geographic spread and sustained presence of EBOV in these three countries has, for the first time, led to the importation of cases to other countries and resulted in clusters of new infections. The first instance, in which 8 of 20 total cases eventually succumbed to EBOV, was that of a Liberian citizen who had traveled to Nigeria by plane (4). The second instance was a Guinean traveling to Senegal by road, and fortunately, there were no secondary cases resulting from this individual (5). The third importation involved a Liberian patient with EBOV who traveled to the United States by plane; 2 nurses were subsequently infected while providing medical care to the patient (6). The fourth importation was from a U.S. physician who returned to New York City from Guinea, with no additional cases (7). The fifth and sixth international importations occurred in Mali; both cases had traveled by road from Guinea. While the former did not result in transmission, the latter resulted in 6 deaths among 8 reported cases. A nurse who fell ill upon returning to Scotland from West Africa in late December 2014 was also diagnosed with EBOV (8). With the outbreak not yet at an end, more infections may be possible in this evolving situation (9, 10).
A number of primary health care workers battling this outbreak have also been infected with EBOV in the line of duty. In addition to the two U.S. nurses mentioned above, a Spanish nurse was infected while caring for a patient who had been medically evacuated from Sierra Leone. This was the first instance of human-to-human EBOV transmission outside Africa (11). Also, hundreds of medical workers, local or from deployed international organizations, have also contracted the disease. The toll currently stands at 495 deaths out of 838 health care worker cases (1), devastating the already weak health care infrastructure. In the absence of licensed medical countermeasures and vaccines against EBOV, barrier techniques such as personal protective equipment, thorough decontamination, and vigilance still play the major role in keeping health workers safe, but these strategies have not been sufficiently effective, as evidenced by the number of infections.
Effective postexposure therapies are desperately needed to treat patients confirmed to be infected with EBOV. As response groups struggle to deal with mounting losses, several health workers and missionaries have already been repatriated or transferred to more resourceful countries in the hopes that receiving a higher standard of medical care will increase their chances of survival. An increasing number of investigational new drug (IND) applications are being approved by the Food and Drug Administration (FDA) for compassionate use in humans, with the intention to fast-track the most promising interventions toward clinical approval in the shortest time possible. In this review, we summarize the available preclinical and clinical results behind different strategies utilized during this outbreak and contemplate the requirements behind an effective, approved EBOV treatment.
As previously summarized in a review of postexposure therapies for filovirus infections (12), strategies for developing experimental postexposure therapeutics against EBOV revolve around preventing the development of filovirus-associated coagulopathies (recombinant nematode anticoagulant protein [rNAPc2] and recombinant human activated protein C [rhAPC]) (13, 14), inhibiting virus processes such as replication or translation (small interfering RNA [siRNA], BCX4430, or PMOplus) (15–17), boosting host immune responses by postexposure prophylaxis (vesicular stomatitis virus (VSV)-vectored virus expressing EBOV glycoprotein [VSVΔG/EBOVGP]) (18), or limiting viremia and virus spread (monoclonal antibody [MAb] cocktails ZMAb and MB-003) (19, 20). During the current outbreak, a particularly desirable strategy is the off-label use of approved or nearly approved nonspecific small-molecule drugs to treat EBOV infections in humans. The rationale behind the use of these drugs is that they have already been shown to have an acceptable safety profile in humans, therefore bypassing the need for phase I trials. These drugs are available in large quantities and are typically given orally, which increases their desirability because of the ease of administration in the early stages of the disease. If shown to be protective, these nonspecific drugs may be effective against not only EBOV but potentially also other filoviruses as well. TKM-Ebola (siRNA)- and ZMapp (antibody)-based compounds have been used in this outbreak. These compounds are specifically designed against EBOV and are relatively well characterized in terms of efficacy in the “gold-standard” NHP model; however, their safety profile is unknown. Blood transfusions from human survivors of EBOV, a controversial method because of the lack of data and safety concerns such as adverse immune-related reactions and the possibility of other blood-borne diseases being transmitted, have also been employed during this outbreak, in addition to intensive supportive care.
SMALL-MOLECULE DRUGS
Lamivudine
Lamivudine (developed by GlaxoSmithKline, United Kingdom), a nucleoside analog of cytidine, is a reverse transcriptase inhibitor. Studies have shown that lamivudine can inhibit the human immunodeficiency virus type 1 (HIV-1), HIV-2, and hepatitis B virus (HBV) reverse transcriptases in vitro (21, 22). Lamivudine has been approved by the FDA to be used in combination with other drugs to treat HIV infections in adults and children (23, 24) or alone to treat acute and chronic HBV infections (25, 26).
During this outbreak, a Liberian doctor used lamivudine to treat 15 infected patients, with 13 eventually surviving EBOV disease. The clinical and preclinical data regarding the effectiveness of this compound against EBOV were not made public. However, the Scientific and Technical Advisory Committee for Ebola Experimental Interventions (STAC-EE), an advisory body of the WHO, did not recommend further use of lamivudine to treat EBOV disease, as data presented to the STAC-EE did not appear to find a survival benefit of this drug (27). Furthermore, a recent in vitro study found that treatment with lamivudine at concentrations of up to 320 μmol/liter in EBOV-infected human monocyte-derived macrophages or Vero E6 (African green monkey kidney) or HepG2 (human hepatoma) cells did not have any direct antiviral effects (28).
Favipiravir
Favipiravir (developed by Toyama Chemical, Japan), a pyrazinecarboxamide derivative, is well characterized as a drug against influenza virus infections and acts by selectively inhibiting the activity of the viral RNA-dependent RNA polymerase (29, 30). Favipiravir has completed phase III clinical trials for the treatment of influenza (31), and several studies have supported it as a broad-spectrum antiviral against a panel of arenaviruses and bunyaviruses (32), West Nile virus (33), yellow fever virus (34), and others, which have been summarized in a review (35).
Recent studies have also shown that favipiravir is effective against aerosol Ebola virus E718 infection in immunodeficient A129 alpha/beta interferon (IFN-α/β) receptor knockout (IFNAR−/−) mice. When the drug was administered orally at a dose of 150 mg/kg of body weight, beginning at 1 h postchallenge and continuing twice daily for 14 days, all mice survived the challenge (36). Follow-up experiments investigated the ability of favipiravir to treat symptomatic EBOV disease in IFNAR−/− mice. At concentrations of 300 mg/kg/day, initiated 6 days after challenge and continuing for 1 week, all mice survived the lethal challenge. Importantly, this drug was able to reverse advanced Ebola virus infection, as evidenced by decreasing levels of the liver enzymes aspartate transaminase (AST) and alanine aminotransferase (ALT) as well as viremia after treatment (37). Favipiravir was one of the drugs used to treat an infected health worker repatriated to France during the outbreak (38), and clinical results are pending. A phase II trial for favipiravir against EBOV was also started on December 2014 in Guinea (39).
Brincidofovir
Brincidofovir (developed by Chimerix, USA) is a lipid-conjugated analog of cidofovir, which is converted into the active compound cidofovir diphosphate upon intracellular release of the drug. The drug acts by inhibiting DNA polymerase through incorporation into the cDNA strand during virus replication, thereby slowing chain extension or resulting in termination. The proofreading 3′-to-5′ exonuclease activity is also inhibited (40). Brincidofovir was shown to possess antiviral activity against cytomegalovirus (CMV) (41), adenovirus (Ad) (42), herpes simplex virus (43), and poxvirus (44) in animal models and is currently in phase III clinical trials against CMV and Ad infections (45, 46).
Brincidofovir was one of the drugs given to patients diagnosed with EBOV in the United States after the FDA approved its use on an emergency basis (47). A Liberian citizen in the United States who received brincidofovir later in the EBOV disease course died; however, several other patients were also given the same drug earlier in the disease course and survived. However, the potential mechanisms of actions of this drug are unknown because EBOV is not a DNA virus and does not undergo a double-stranded DNA (dsDNA) intermediate phase at any stage in its life cycle. Studies will be necessary to elucidate whether brincidofovir is also effective at inhibiting replication in RNA viruses. Clinical, animal, and in vitro data regarding the effectiveness of brincidofovir against EBOV are currently unavailable. A planned clinical trial for brincidofovir was cancelled by Chimerix as of January 2015 due to a lack of participants in Liberia, and it was reported that the trial would not be extended to neighboring Sierra Leone (48, 49).
SMALL INTERFERING RNA
TKM-Ebola
TKM-Ebola (developed by Tekmira Pharmaceuticals, Canada) consists of a cocktail of three siRNAs in the form of lipid nanoparticles, designed specifically to target regions in three EBOV genes: EBOV membrane-associated protein 24 (VP24), the EBOV polymerase complex protein VP35, and polymerase (L). The mechanism of TKM-Ebola is to interfere with the translation of the EBOV VP24, VP35, and L proteins from viral mRNA, which are required for evasion of host IFN responses and several viral processes, including virus assembly, transcription, and replication (50–53).
An intravenous (i.v.) bolus infusion of TKM-Ebola at 2 mg/kg, beginning at 30 min after EBOV challenge and continuing daily for 7 days, was found to be protective, as all NHPs survived the infection (15). A phase I clinical trial was initiated for this drug (54); however, following the observation of unintended cytokine release in participants that was induced by TKM-Ebola, the trial was initially put on clinical hold by the FDA (55). A partial lift was subsequently granted, in which the drug could be tested in EBOV-infected patients in response to the 2014-2015 outbreak (56), and several patients were administered TKM-Ebola, although clinical data are unavailable.
MONOCLONAL ANTIBODIES
ZMapp
ZMapp (developed jointly by the Public Health Agency of Canada and Mappbio Pharmaceuticals, USA) is an improved IgG MAb cocktail comprising MAbs from two precursor cocktails, ZMAb (providing MAbs c2G4 and c4G7) and MB-003 (providing MAb c13C6). The plant-derived antibodies are specific for the viral glycoprotein, which is the sole surface protein on the EBOV virion (57) and a main target for vaccine design because of its ability to elicit specific immune responses (58).
In preclinical studies, ZMapp was found to be efficacious in nonhuman primates when administered as an i.v. bolus at a concentration of 50 mg/kg in three separate doses spread evenly over 9 days. Complete survival was observed when ZMapp therapy was initiated up to 5 days after EBOV infection, and ZMapp was effective at reversing advanced EBOV disease symptoms, as evidenced by decreases in rash and viremia and elevated liver enzyme levels (59).
ZMapp was administered to 7 patients during the current outbreak under an emergency compassionate-use provision from the FDA, and clinical information is available from two U.S. health care providers who received ZMapp, combined with aggressive supportive therapy including hydration and electrolyte correction. Both patients had hypovolemia, hypokalemia, hypocalcemia, and hypoalbuminemia, and one patient also had substantial liver injury, which were factors associated with increased mortality (60) and indicative of advanced EBOV disease. The conditions of both patients improved with this combined treatment, and both patients survived EBOV infection. It was noted that there was a correlation between increasing antibody levels and decreasing viremia, as determined by reverse transcription-quantitative PCR (RT-qPCR), and subjective as well as objective improvements were observed shortly after the administration of the first ZMapp dose (61). However, this improvement occurred with other treatments as well, and the authors of this study could not definitively conclude that the administration of ZMapp had any survival benefit in these patients (61).
WHOLE-BLOOD OR PLASMA TRANSFUSIONS FROM CONVALESCENT SURVIVORS
Whole-blood transfusions are widely used in developing countries with limited resources committed toward health care. However, there are several concerns with this technique, as leukocytes have been associated with adverse effects, including febrile transfusion reactions, alloimmunization to leukocyte antigens, graft-versus-host disease, and the possibility of becoming infected with other blood-borne diseases, such as HIV, hepatitis viruses, CMV, and other viruses (62). Whole-blood transfusions from convalescent to infected patients were successful during the 1995 EBOV outbreak in Kikwit, Democratic Republic of Congo. Seven out of eight patients who received the transfusions survived, although several of the survivors already had detectable antibodies when they received the transfusion (63). However, the transfused patients also received comparatively better supportive care than did others, including infusions of glucose and electrolytes, treatment with antibiotics and antimalarial drugs, and food supplementation, limiting conclusions that can be drawn about the potential benefits of this treatment (63). Furthermore, a study in which sera were passively transferred from EBOV-immune survivor macaques was not successful in conferring protection to naive NHPs, but it should be noted that three of four recipient animals exhibited IgG titers of only 1:100 at 3 days after passive immunotherapy (64). Therefore, further investigation into passive immune therapy during a future EBOV outbreak is needed.
Patients seldom require all the components found in whole blood. As a safer alternative, immunotherapy with convalescent-phase serum, plasma, and polyclonal antibodies was used historically for many bacterial and viral diseases, including Haemophilus influenzae, scarlet fever, pertussis, measles, mumps, polio, the 1918 pandemic H1N1 influenza virus, and others (65). In the laboratory, a number of studies were performed to demonstrate the impact of antibodies on EBOV infection. Passive transfer of IgG from horses hyperimmunized to EBOV was evaluated for survival benefit to NHPs when given immediately after a lethal challenge. Although all treated NHPs succumbed to disease, there was a delay in the onset of viremia and clinical symptoms compared with control animals (66). An additional dose of equine IgG given to NHPs 5 days after challenge did not have any additional beneficial effects (67). However, NHPs were fully protected when given three doses of concentrated, polyclonal IgG from NHP survivors purified to 96% purity by fractionation in protein G columns, beginning 48 h after infection, with additional treatments at 4 and 8 days postinfection (68).
Whole-blood or plasma transfusions were deemed an ethically acceptable treatment modality by the WHO for use in the current outbreak (69), provided that risk assessments were carried out to minimize any known associated risks. For instance, the potential donor patient must be clinically asymptomatic and have twice tested negative for EBOV RNA in two independent blood samples taken at least 48 h apart. Donor blood must be blood group compatible and also tested for the possible presence of any blood-borne infections. The blood must be collected, prepared, stored, and transfused at facilities experienced in handling such processes. The appearance of neutralizing antibodies is late in general (several weeks or months after recovery), and levels are variable between patients. Donor blood is likely to vary in protective efficacy; therefore, the levels of total and neutralizing EBOV antibodies should be titrated if possible (69). During this outbreak, several infected patients received whole blood or plasma from convalescent survivors. Clinical trials are under way to assess the clinical benefits associated with blood or plasma transfusions (27).
SUPPORTIVE TREATMENT
Supportive care is based primarily on the management of and relief from disease symptoms, physical stress, as well as mental stress and therefore can be used regardless of the disease. The typical protocol for febrile illness was used in past filovirus outbreaks. Initial treatments include antimalarial drugs and antibiotics to eliminate the possibility of malaria as well as to prevent and treat secondary bacterial infections. Antiviral drugs used in past outbreaks include acyclovir and ribavirin. Other treatments include painkillers, sedatives, as well as anti-inflammatory, antidiarrheal, and antipsychotic drugs administered at the medical doctor's discretion (70). Intravenous rehydration was routinely used in later outbreaks (71); however, oral rehydration was encouraged whenever possible because of the risk of EBOV transmission through contaminated needles used for i.v. fluid administration. Coagulants such as fibrinogen and prothrombin were given to prevent hemorrhaging during a previous outbreak of MARV (70), although their use may be more restricted in resource-poor locations, as i.v. administrations are more difficult to perform properly for the reasons mentioned above. Anticoagulants such as heparin were also administered to some patients in response to certain disorders, such as disseminated intravascular coagulation (70). For patients in high-resource medical settings, such as those airlifted to the United States and Europe for intensive treatment, it is possible to monitor and rapidly respond to changes in the patient's renal, hepatic, and pulmonary/respiratory functions (61, 72). This is in part related to the high medical-personnel-to-patient ratio as well as the added ability to provide critical care such as noninvasive and invasive mechanical ventilation and continuous renal replacement therapy (73). As of 7 January 2015, among the 24 patients who received medical care for EBOV disease in the United States or Europe, 18 (75%) recovered, 5 died, and 1 is still undergoing intensive treatment (74). This represents a higher survival rate than that in West Africa. Supportive therapy has not been rigorously tested for efficacy as part of a randomized trial due to ethical issues and is expected in general to be associated with better outcomes. However, the survivor benefits associated with the details of this strategy under these conditions remain based on a general perception built upon years of experience that supportive therapy is not believed to be harmful to the patient.
OTHER POTENTIAL DRUGS FOR USE IN THE 2014-2015 OUTBREAK
A meeting convened by the WHO with a panel of experts to prioritize experimental drugs resulted in the consideration of two additional compounds not described above that may also have beneficial effects against EBOV infections in humans. Therapy with type I IFN is commonly used in the clinic to boost host antiviral responses for the treatment of chronic HBV (75), hepatitis C virus (HCV) (76), as well as human herpesvirus 8 (HHV-8) for Kaposi's sarcoma (77). In preclinical studies with EBOV, the administration of IFN-α2b to NHPs at a dose of 2 × 107 IU/kg/day beginning 18 h after challenge resulted in a delay of viremia development as well as a slight extension in the time to death of treated NHPs by 1 to 2 days (67). IFN-β treatment also increased the survival time for NHPs. Animals given 10.5 μg/kg of IFN-β beginning 18 h after challenge, with subsequent doses at 1, 3, 5, 7, and 9 days after infection, had significantly prolonged survival times, on average 5.5 days longer than those of untreated control animals (78).
Another candidate is the drug toremifene, an FDA-approved selective estrogen receptor modulator (SERM) identified by an in vitro screen of drugs approved by the FDA and drugs approved outside the United States for anti-EBOV activity (79). Toremifene is approved for the treatment of advanced breast cancer (80). SERMs act by binding to the estrogen receptor, causing conformational changes that lead to the initiation or suppression of target genes through interactions with coactivator or corepressor proteins, respectively (81). However, as estrogen receptor expression was not required for in vitro EBOV inhibition by toremifene, the drug likely works independently of the classical estrogen pathway. The activity of toremifene was also evaluated in vivo by using a mouse model of EBOV infection. Treatment was initiated beginning 1 h after challenge at a dose of 60 mg/kg by intraperitoneal injection, with subsequent doses at days 1, 3, 5, 7, and 9, which protected 50% of the infected animals (79).
POSTEXPOSURE PROPHYLAXIS WITH VSVΔG/EBOVGP
VSVΔG/EBOVGP is one of two vaccines that are currently undergoing phase I and II clinical trials in North America, Europe, and Africa as a potential candidate for the mass immunization of at-risk populations in response to the 2014-2015 outbreak (82, 83). With the recent exception of an adjuvanted adenovirus-vectored vaccine (84), VSVΔG/EBOVGP was at one time unique as the only vaccine that could also confer postexposure protection, with 50% efficacy in NHPs if given 30 min after EBOV challenge (18). The rapid protection induced by immunization with the VSV-vectored vaccine highlights its utility should instances of accidental exposure, such as a needlestick injury, occur in the laboratory or the field.
In 2009, the VSVΔG/EBOVGP vaccine was given under compassionate circumstances to a researcher who had a high-risk occupational EBOV exposure from a laboratory accident. The individual was given a single intramuscular (i.m.) dose of 5 × 107 PFU of VSVΔG/EBOVGP ∼48 h after the incident. Aside from a fever 12 h later and the detection of vesicular stomatitis virus (VSV) viremia by PCR for 2 days, the vaccine recipient remained healthy (85). In September 2014, a physician was potentially exposed to EBOV through a needlestick injury while working at an EBOV treatment center in Sierra Leone. The individual received an i.m. injection of 1 × 108 PFU of VSVΔG/EBOVGP ∼43 h after the accident. A fever as well as moderate to severe myalgia, chills, tiredness, and headache were noted 12 h after the injection but subsided over 3 to 4 days. VSV viremia was detectable by PCR for 4 days, and cytokine secretion as well as T-cell/plasmablast activation occurred early postvaccination (86). Both patients were not believed to have been infected with EBOV, as diagnostic tests for the EBOV nucleoprotein were consistently negative, and the VSVΔG/EBOVGP vaccine was not associated with any adverse effects in these two individuals.
DISCUSSION
The 2014-2015 EBOV outbreak has exposed the severe shortage of options in medical countermeasures at our disposal. The current process for the preclinical testing of candidate EBOV antivirals includes mouse, guinea pig, and NHP animal models, which fulfills the FDA's “two-animal rule.” The two-animal rule stipulates that in emergency scenarios, a drug can be made available on compassionate grounds provided that (i) it has shown efficacy in two different animal models for EBOV infections or efficacy in one well-characterized animal model that recapitulates the major hallmarks of EBOV disease and (ii) it does not cause adverse side effects in humans (87). Based on past experiences, some candidate treatments have been shown to be protective in lower-animal models but are less effective in the gold-standard NHP model. For instance, the EBOV-neutralizing MAb KZ52 demonstrated complete pre- and postexposure protection in guinea pigs with one dose (88) but was not effective when administered to NHPs (89). Along similar lines, a recombinant adenovirus expressing IFN-α (Ad-IFN-α) was fully protective when administered to mice (90) but was not protective in guinea pigs (91) and NHPs (92). The difference in protective efficacy is attributed to the observation that wild-type EBOV is more virulent in NHPs and more closely mimics the hallmarks of disease in humans than the adapted EBOV variants used for rodent studies (93). Therefore, the most promising compounds should be tested for efficacy and safety in NHPs before use in humans, even under compassionate-use provisions.
Owing to the severity of the EBOV outbreak in West Africa, regulatory approval bodies have modified their position toward the testing of potential clinical options in infected humans. A panel of WHO experts has unanimously concluded that it is ethical to use unapproved drugs in this outbreak, provided that there are preclinical safety and efficacy data supporting such use (94). When ZMapp was administered for the first time to two EBOV-infected health workers in Liberia during the outbreak, it meant that those responsible felt that the possibility of severe adverse effects from an untested cocktail was an acceptable risk, given the high mortality rates associated with EBOV disease. This was in stark contrast to events only several days earlier, when authorities declined to use ZMapp to treat a physician in Sierra Leone, despite the drug being made available at that time for compassionate use. These proceedings highlighted the ethical dilemma that faces governing aid organizations all the way down to the physician responsible for administering an untested drug. However, as a potential positive outcome of these events, the level of informed consent required from patients to necessitate the testing of experimental drugs under atypical circumstances is now established as a precedent. These events have also triggered a much-needed ethical discussion regarding the use of potentially efficacious but untested compounds or the off-label use of approved drugs in humans, along with a significant push to rapidly advance EBOV vaccines and therapeutics toward clinical licensure.
Clinical data on the effect of some experimental products administered to patients evacuated from West Africa are not readily available, and in many cases, adequate controls are not available for comparison. Therefore, it is difficult to compare the efficacies of the different treatment regimens at this time. Another confounding factor is the fact that many of the surviving patients were given several experimental therapies in order to maximize their chances of survival. Therefore, it will be very challenging to assess each intervention independently in relation to its survival benefit. Under these circumstances, the next best alternative would be to compare the clinical statuses of the patients shortly after the administration of a certain compound to determine which intervention had the most profound effects. Aside from objective clinical observations, one significant indicator would be a direct drop in EBOV viremia after treatment, as lower viremia has been correlated with survival from infection (95). Other possible survival indicators include blood chemistry parameters such as AST, blood urea nitrogen (BUN), creatinine, and albumin; the secretion of proinflammatory cytokines; or elevated thrombomodulin and ferritin levels, which were found to be associated with hemorrhaging (96).
This is the first instance in which a drug exists that could be used to treat patients postexposure during an EBOV outbreak (Table 1). ZMapp shows the most promising preclinical data in terms of efficacy in NHPs. While untested in a controlled clinical trial, in a small sample of seven people, ZMapp was well tolerated overall, and five patients who had received all three ZMapp doses survived the infection (97). Of note, eight additional patients were treated with ZMAb after supplies of ZMapp had been exhausted, and all patients survived the infection (our unpublished data). Available data support further investigation of antibody-based therapies, and controlled clinical trials should take place, preferably in the countries affected by the outbreak. Since ZMapp is expensive to synthesize in large quantities and the production process requires several months, other strategies should be combined with ZMapp to test whether the clinical benefits could be enhanced and lead to a more efficacious treatment with lower ZMapp doses. A previous study showed that the protective effects of ZMAb are enhanced in NHPs when supplemented with an Ad-IFN-α adjuvant (92). Effective postexposure treatment will likely consist of several components, including aggressive supportive care and a nonspecific antiviral to hinder EBOV pathogenesis and spread, before the control of viremia is achieved with an efficacious specific treatment such as ZMapp.
TABLE 1.
Compound | Developer(s) | Status before the 2014-2015 EBOV outbreak |
Current status | |
---|---|---|---|---|
Tested for efficacy against EBOV | Proof of safety in humans | |||
Lamivudine | GlaxoSmithKline | No | Yes, FDA approved against HIV | Not recommended by WHO for treatment of EBOV disease |
Favipiravir | Toyama Chemical | Yes, in mice | Yes, phase III against influenza virus | Phase II |
Brincidofovir | Chimerix | No | Yes, phase III against CMV and Ad infections | EBOV trial discontinued |
TKM-Ebola | Tekmira Pharmaceuticals | Yes, in NHPs | No | Phase I (partial suspension) |
ZMapp | Public Health Agency of Canada and MappBio Pharmaceuticals | Yes, in NHPs | No | Phase I/II |
Whole-blood or plasma transfusion | No | Yes, routine medical procedure in resource-poor countries | Still in use | |
Supportive treatment | No | Yes, routine medical procedure | Still in use | |
Type I IFN | Yes, delayed time to death in NHPs | Yes, used in clinic to treat HBV, HCV, and HHV-8 | Not yet used in patients during the 2014-2015 outbreak | |
Toremifene | Yes, in mice | Yes, FDA approved for use against breast cancer | Not yet used in patients during the 2014-2015 outbreak | |
VSVΔG/EBOVGP | Public Health Agency of Canada | Yes, in NHPs | No | Phase I/II (as a prophylactic) |
The 2014-2015 EBOV outbreak has provided the world with many tough lessons regarding the speed of research and development of protective medical countermeasures, the ethical considerations of the use of experimental therapies in humans under compassionate-use provisions, and the circumstances under which licensed drugs can be used off-label for the treatment of other diseases. This outbreak has also highlighted the various levels of preparedness among different countries to combat the rapid spread of infectious diseases with relative high case fatality rates. These findings must be incorporated into revised guidelines in order to ensure that any barriers regarding the emergency use of experimental treatments will not be repeated in the future, whether with EBOV or with other pathogens.
ACKNOWLEDGMENTS
We thank Pierre Rollin for the critical reading of the manuscript and his insightful thoughts for this review.
This work was supported by the Public Health Agency of Canada (PHAC) and was funded by a Canadian Safety and Security Program (CSSP) grant to G.P.K. G.W. is the recipient of a Banting postdoctoral fellowship from the Canadian Institutes of Health Research (CIHR).
Her Majesty the Queen in right of Canada holds a patent on MAbs 1H3, 2G4, and 4G7 (PCT/CA2009/000070), monoclonal antibodies for Ebola and Marburg viruses. G.W. and G.P.K. have read and agree with the contents in the manuscript. G.W. and G.P.K. have been extensively involved in the preclinical development and testing of the antibody cocktails ZMAb and ZMapp as well as experiments with the VSVΔG/EBOVGP vaccine mentioned in this review. G.W. and G.P.K. are not included on any related patents, other intellectual property, or contracts that may benefit them financially in the future. We declare no other competing interests.
Additionally, the findings and conclusions in this report are those of the authors and do not necessarily represent the views of the Centers for Disease Control and Prevention.
Biographies
Gary Wong received his Ph.D. in Medical Microbiology from the University of Manitoba in 2014 and carried out his research with the Special Pathogens Program at the National Microbiology Laboratory (NML), Public Health Agency of Canada (PHAC). He was recently awarded the prestigious Banting postdoctoral fellowship from the Canadian Institutes of Health Research (CIHR) to carry out his postdoctoral work in the Institute of Microbiology at the Chinese Academy of Sciences. Dr. Wong has had a long-standing interest in infectious diseases, dating back to when he watched the 1995 movie Outbreak, and currently has 7 years of laboratory and field experience working with biosafety level 4 (BSL-4) pathogens. Dr. Wong has published 25 peer-reviewed papers to date, and research highlights include identifying the immune parameters correlating with protection in Ad5- and VSV-vaccinated survivors, testing the monoclonal antibody cocktails ZMAb and ZMapp, and developing a guinea pig model for studying Ebola virus transmission.
Gary P. Kobinger received his Ph.D. from the University of Montréal in 1998 and completed his postdoctoral training at the University of Pennsylvania between 1999 and 2003. He is the chief of the Special Pathogens Program at the NML, PHAC. He also serves as an Associate and Adjunct Professor in the Departments of Medical Microbiology and Immunology, respectively, at the University of Manitoba and holds the position of Adjunct Professor at the Department of Pathology and Laboratory Medicine at the University of Pennsylvania. His research team is interested in the development and preclinical/clinical testing of novel vaccines/therapeutics against BSL-4 pathogens. As Canada's top expert on high-level-biocontainment pathogens, Dr. Kobinger has played an integral part over the past decade in the laboratory (with over 120 peer-reviewed publications) and in the field against infectious disease outbreaks, in collaboration with international aid groups, including the WHO and Médecins sans Frontières (MSF).
REFERENCES
- 1.WHO. 11 March 2015. Ebola situation report—11 March 2015. WHO, Geneva, Switzerland: http://apps.who.int/ebola/current-situation/ebola-situation-report-11-march-2015 Accessed 12 March 2015. [Google Scholar]
- 2.Meltzer MI, Atkins CY, Santibanez S, Knust B, Petersen BW, Ervin ED, Nichol ST, Damon IK, Washington ML. 2014. Estimating the future number of cases in the Ebola epidemic—Liberia and Sierra Leone, 2014-2015. MMWR Surveill Summ 63(Suppl 3):1–14. [PubMed] [Google Scholar]
- 3.Whitty CJ, Farrar J, Ferguson N, Edmunds WJ, Piot P, Leach M, Davies SC. 2014. Infectious disease: tough choices to reduce Ebola transmission. Nature 515:192–194. doi: 10.1038/515192a. [DOI] [PubMed] [Google Scholar]
- 4.WHO. 20 October 2014. Nigeria is now free of Ebola virus transmission. WHO, Geneva, Switzerland: http://www.who.int/mediacentre/news/ebola/20-october-2014/en/index2.html Accessed 15 January 2015. [Google Scholar]
- 5.WHO. 17 October 2014. The outbreak of Ebola virus disease in Senegal is over. WHO, Geneva, Switzerland: http://www.who.int/mediacentre/news/ebola/17-october-2014/en/ Accessed 15 January 2015. [Google Scholar]
- 6.ProMED-mail. 28 October 2014. Ebola virus disease—ex Africa (27): USA (Texas) second nurse better, test, quarantine. International Society for Infectious Diseases, Brookline, MA: http://www.promedmail.org/direct.php?id=2910297 Accessed 15 January 2015. [Google Scholar]
- 7.CDC. 23 October 2014. New York City reports positive test for Ebola in volunteer international aid worker. CDC, Atlanta, GA: http://www.cdc.gov/media/releases/2014/s1023-ebola-nyc.html Accessed 15 January 2015. [Google Scholar]
- 8.ProMED-mail. 3 January 2015. Ebola update (03): Africa, World, USA, UK, suspected, drugs, vaccines. International Society for Infectious Diseases, Brookline, MA: http://www.promedmail.org/direct.php?id=3069311 Accessed 15 January 2015. [Google Scholar]
- 9.WHO. 10 November 2014. Mali case, Ebola imported from Guinea. WHO, Geneva, Switzerland: http://www.who.int/mediacentre/news/ebola/10-november-2014-mali/en/ Accessed 15 January 2015. [Google Scholar]
- 10.WHO. 12 November 2014. Mali confirms its second fatal case of Ebola virus disease. WHO, Geneva, Switzerland: http://www.who.int/mediacentre/news/ebola/12-november-2014-mali/en/ Accessed 15 January 2015. [Google Scholar]
- 11.ProMED-mail. 8 November 2014. Ebola virus disease—ex Africa (32): Spanish nurse recovered, USA seeks patent. International Society for Infectious Diseases, Brookline, MA: http://www.promedmail.org/direct.php?id=2939861 Accessed 15 January 2015. [Google Scholar]
- 12.Wong G, Qiu X, Olinger GG, Kobinger GP. 2014. Post-exposure therapy of filovirus infections. Trends Microbiol 22:456–463. doi: 10.1016/j.tim.2014.04.002. [DOI] [PubMed] [Google Scholar]
- 13.Geisbert TW, Hensley LE, Jahrling PB, Larsen T, Geisbert JB, Paragas J, Young HA, Fredeking TM, Rote WE, Vlasuk GP. 2003. Treatment of Ebola virus infection with a recombinant inhibitor of factor VIIa/tissue factor: a study in rhesus monkeys. Lancet 362:1953–1958. doi: 10.1016/S0140-6736(03)15012-X. [DOI] [PubMed] [Google Scholar]
- 14.Hensley LE, Stevens EL, Yan SB, Geisbert JB, Macias WL, Larsen T, Daddario-DiCaprio KM, Cassell GH, Jahrling PB, Geisbert TW. 2007. Recombinant human activated protein C for the postexposure treatment of Ebola hemorrhagic fever. J Infect Dis 196(Suppl 2):S390–S399. doi: 10.1086/520598. [DOI] [PubMed] [Google Scholar]
- 15.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. 2010. Postexposure protection of non-human primates against a lethal Ebola virus challenge with RNA interference: a proof-of-concept study. Lancet 375:1896–1905. doi: 10.1016/S0140-6736(10)60357-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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. 2014. Protection against filovirus diseases by a novel broad-spectrum nucleoside analogue BCX4430. Nature 508:402–405. doi: 10.1038/nature13027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.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. 2010. Advanced antisense therapies for postexposure protection against lethal filovirus infections. Nat Med 16:991–994. doi: 10.1038/nm.2202. [DOI] [PubMed] [Google Scholar]
- 18.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. 2007. Effective post-exposure treatment of Ebola infection. PLoS Pathog 3:e2. doi: 10.1371/journal.ppat.0030002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Qiu X, Audet J, Wong G, Pillet S, Bello A, Cabral T, Strong JE, Plummer F, Corbett CR, Alimonti JB, Kobinger GP. 2012. Successful treatment of Ebola virus-infected cynomolgus macaques with monoclonal antibodies. Sci Transl Med 4:138ra81. doi: 10.1126/scitranslmed.3003876. [DOI] [PubMed] [Google Scholar]
- 20.Pettitt J, Zeitlin L, Kim DH, Working HC, Johnson JC, Bohorov O, Bratcher B, Hiatt E, Hume SD, Johnson AK, Morton J, Pauly MH, Whaley KJ, Ingram MF, Zovanyi A, Heinrich M, Piper A, Zelko J, Olinger GG. 2013. Therapeutic intervention of Ebola virus infection in rhesus macaques with the MB-003 monoclonal antibody cocktail. Sci Transl Med 5:199ra113. doi: 10.1126/scitranslmed.3006608. [DOI] [PubMed] [Google Scholar]
- 21.Coates JA, Cammack N, Jenkinson HJ, Jowett AJ, Jowett MI, Pearson BA, Penn CR, Rouse PL, Viner KC, Cameron JM. 1992. (-)-2′-Deoxy-3′-thiacytidine is a potent, highly selective inhibitor of human immunodeficiency virus type 1 and type 2 replication in vitro. Antimicrob Agents Chemother 36:733–739. doi: 10.1128/AAC.36.4.733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chang CN, Doong SL, Zhou JH, Beach JW, Jeong LS, Chu CK, Tsai CH, Cheng YC, Liotta D, Schinazi R. 1992. Deoxycytidine deaminase-resistant stereoisomer is the active form of (+/−)-2′,3′-dideoxy-3′-thiacytidine in the inhibition of hepatitis B virus replication. J Biol Chem 267:13938–13942. [PubMed] [Google Scholar]
- 23.Staszewski S, Keiser P, Montaner J, Raffi F, Gathe J, Brotas V, Hicks C, Hammer SM, Cooper D, Johnson M, Tortell S, Cutrell A, Thorborn D, Isaacs R, Hetherington S, Steel H, Spreen W. 2001. Abacavir-lamivudine-zidovudine vs indinavir-lamivudine-zidovudine in antiretroviral-naive HIV-infected adults: a randomized equivalence trial. JAMA 285:1155–1163. doi: 10.1001/jama.285.9.1155. [DOI] [PubMed] [Google Scholar]
- 24.Green H, Gibb DM, Walker AS, Pillay D, Butler K, Candeias F, Castelli-Gattinara G, Compagnucci A, Della Negra M, de Rossi A, Feiterna-Sperling C, Giaquinto C, Harper L, Levy J, Saidi Y, Wintergerst U. 2007. Lamivudine/abacavir maintains virological superiority over zidovudine/lamivudine and zidovudine/abacavir beyond 5 years in children. AIDS 21:947–955. doi: 10.1097/QAD.0b013e3280e087e7. [DOI] [PubMed] [Google Scholar]
- 25.Schmilovitz-Weiss H, Ben-Ari Z, Sikuler E, Zuckerman E, Sbeit W, Ackerman Z, Safadi R, Lurie Y, Rosner G, Tur-Kaspa R, Reshef R. 2004. Lamivudine treatment for acute severe hepatitis B: a pilot study. Liver Int 24:547–551. doi: 10.1111/j.1478-3231.2004.0983.x. [DOI] [PubMed] [Google Scholar]
- 26.Lau DT, Khokhar MF, Doo E, Ghany MG, Herion D, Park Y, Kleiner DE, Schmid P, Condreay LD, Gauthier J, Kuhns MC, Liang TJ, Hoofnagle JH. 2000. Long-term therapy of chronic hepatitis B with lamivudine. Hepatology 32:828–834. doi: 10.1053/jhep.2000.17912. [DOI] [PubMed] [Google Scholar]
- 27.WHO. 13 November 2014. WHO meeting of the Scientific and Technical Advisory Committee on Ebola Experimental Interventions—briefing note. WHO, Geneva, Switzerland: http://www.who.int/medicines/ebola-treatment/scientific_tech_meeting/en/# Accessed 15 January 2015. [Google Scholar]
- 28.Hensley LE, Dyall J, Olinger GG Jr, Jahrling PB. 2015. Lack of effect of lamivudine on Ebola virus replication. Emerg Infect Dis 21:550–552. doi: 10.3201/eid2103.141862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Furuta Y, Takahashi K, Kuno-Maekawa M, Sangawa H, Uehara S, Kozaki K, Nomura N, Egawa H, Shiraki K. 2005. Mechanism of action of T-705 against influenza virus. Antimicrob Agents Chemother 49:981–986. doi: 10.1128/AAC.49.3.981-986.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sangawa H, Komeno T, Nishikawa H, Yoshida A, Takahashi K, Nomura N, Furuta Y. 2013. Mechanism of action of T-705 ribosyl triphosphate against influenza virus RNA polymerase. Antimicrob Agents Chemother 57:5202–5208. doi: 10.1128/AAC.00649-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.ClinicalTrials.gov. 7 February 2013. T-705a multicenter study in adults subjects with uncomplicated influenza (FAVOR). Registration number NCT01728753 https://clinicaltrials.gov/ct2/show/NCT01728753 Accessed 15 January 2015. [Google Scholar]
- 32.Gowen BB, Wong MH, Jung KH, Sanders AB, Mendenhall M, Bailey KW, Furuta Y, Sidwell RW. 2007. In vitro and in vivo activities of T-705 against arenavirus and bunyavirus infections. Antimicrob Agents Chemother 51:3168–3176. doi: 10.1128/AAC.00356-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Morrey JD, Taro BS, Siddharthan V, Wang H, Smee DF, Christensen AJ, Furuta Y. 2008. Efficacy of orally administered T-705 pyrazine analog on lethal West Nile virus infection in rodents. Antiviral Res 80:377–379. doi: 10.1016/j.antiviral.2008.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Julander JG, Shafer K, Smee DF, Morrey JD, Furuta Y. 2009. Activity of T-705 in a hamster model of yellow fever virus infection in comparison with that of a chemically related compound, T-1106. Antimicrob Agents Chemother 53:202–209. doi: 10.1128/AAC.01074-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Furuta Y, Gowen BB, Takahashi K, Shiraki K, Smee DF, Barnard DL. 2013. Favipiravir (T-705), a novel viral RNA polymerase inhibitor. Antiviral Res 100:446–454. doi: 10.1016/j.antiviral.2013.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Smither SJ, Eastaugh LS, Steward JA, Nelson M, Lenk RP, Lever MS. 2014. Post-exposure efficacy of oral T-705 (favipiravir) against inhalational Ebola virus infection in a mouse model. Antiviral Res 104:153–155. doi: 10.1016/j.antiviral.2014.01.012. [DOI] [PubMed] [Google Scholar]
- 37.Oestereich L, Ludtke A, Wurr S, Rieger T, Munoz-Fontela C, Gunther S. 2014. Successful treatment of advanced Ebola virus infection with T-705 (favipiravir) in a small animal model. Antiviral Res 105:17–21. doi: 10.1016/j.antiviral.2014.02.014. [DOI] [PubMed] [Google Scholar]
- 38.ProMED-mail. 28 September 2014. Ebola virus disease—West Africa (180): Sierra Leone, Liberia, USA, drugs, Guinea. International Society for Infectious Diseases, Brookline, MA: http://www.promedmail.org/direct.php?id=2813292 Accessed 15 January 2015. [Google Scholar]
- 39.ClinicalTrials.gov. 5 March 2015. Efficacy of favipiravir against Ebola (JIKI). Registration number NCT02329054 https://clinicaltrials.gov/ct2/show/NCT02329054 Accessed 12 March 2015.
- 40.Hostetler KY. 2009. Alkoxyalkyl prodrugs of acyclic nucleoside phosphonates enhance oral antiviral activity and reduce toxicity: current state of the art. Antiviral Res 82:A84-98. doi: 10.1016/j.antiviral.2009.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bravo FJ, Bernstein DI, Beadle JR, Hostetler KY, Cardin RD. 2011. Oral hexadecyloxypropyl-cidofovir therapy in pregnant guinea pigs improves outcome in the congenital model of cytomegalovirus infection. Antimicrob Agents Chemother 55:35–41. doi: 10.1128/AAC.00971-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Toth K, Spencer JF, Dhar D, Sagartz JE, Buller RM, Painter GR, Wold WS. 2008. Hexadecyloxypropyl-cidofovir, CMX001, prevents adenovirus-induced mortality in a permissive, immunosuppressed animal model. Proc Natl Acad Sci U S A 105:7293–7297. doi: 10.1073/pnas.0800200105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Quenelle DC, Lampert B, Collins DJ, Rice TL, Painter GR, Kern ER. 2010. Efficacy of CMX001 against herpes simplex virus infections in mice and correlations with drug distribution studies. J Infect Dis 202:1492–1499. doi: 10.1086/656717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Parker S, Touchette E, Oberle C, Almond M, Robertson A, Trost LC, Lampert B, Painter G, Buller RM. 2008. Efficacy of therapeutic intervention with an oral ether-lipid analogue of cidofovir (CMX001) in a lethal mousepox model. Antiviral Res 77:39–49. doi: 10.1016/j.antiviral.2007.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.ClinicalTrials.gov. 16 May 2014. A study of the safety and efficacy of CMX001 for the prevention of cytomegalovirus (CMV) infection in CMV-seropositive (R+) hematopoietic stem cell transplant recipients. Registration number NCT01769170 https://clinicaltrials.gov/ct2/show/NCT01769170 Accessed 15 January 2015.
- 46.ClinicalTrials.gov. 9 October 2014. Phase III, open-labeled, multicenter study of the safety and efficacy of brincidofovir (CMX001) in the treatment of early versus late adenovirus infection (CMX001 Adv). Registration number NCT02087306 https://clinicaltrials.gov/ct2/show/NCT02087306 Accessed 15 January 2015.
- 47.ProMED-mail. 6 October 2014. Ebola virus disease—ex Africa (06): Spain case, USA case, US case medevaced. International Society for Infectious Diseases, Brookline, MA: http://www.promedmail.org/direct.php?id=2837374 Accessed 15 January 2015. [Google Scholar]
- 48.Kupferschmidt K, Cohen J. 2015. Infectious diseases. Ebola drug trials lurch ahead. Science 347:701–702. doi: 10.1126/science.347.6223.701. [DOI] [PubMed] [Google Scholar]
- 49.Médecins sans Frontières. 2015. Ebola drug trial in Liberia halted. Médecins sans Frontières, Geneva, Switzerland: http://www.msf.org/article/ebola-drug-trial-liberia-halted Accessed 19 March 2015. [Google Scholar]
- 50.Haasnoot J, de Vries W, Geutjes EJ, Prins M, de Haan P, Berkhout B. 2007. The Ebola virus VP35 protein is a suppressor of RNA silencing. PLoS Pathog 3:e86. doi: 10.1371/journal.ppat.0030086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hoenen T, Jung S, Herwig A, Groseth A, Becker S. 2010. Both matrix proteins of Ebola virus contribute to the regulation of viral genome replication and transcription. Virology 403:56–66. doi: 10.1016/j.virol.2010.04.002. [DOI] [PubMed] [Google Scholar]
- 52.Hoenen T, Groseth A, Kolesnikova L, Theriault S, Ebihara H, Hartlieb B, Bamberg S, Feldmann H, Stroher U, Becker S. 2006. Infection of naive target cells with virus-like particles: implications for the function of Ebola virus VP24. J Virol 80:7260–7264. doi: 10.1128/JVI.00051-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Volchkov VE, Volchkova VA, Chepurnov AA, Blinov VM, Dolnik O, Netesov SV, Feldmann H. 1999. Characterization of the L gene and 5′ trailer region of Ebola virus. J Gen Virol 80(Part 2):355–362. [DOI] [PubMed] [Google Scholar]
- 54.ClinicalTrials.gov. 31 July 2014. Safety, tolerability and pharmacokinetic first in human (FIH) study for intravenous (IV) TKM-100802. Registration number NCT02041715 http://clinicaltrials.gov/show/NCT02041715 Accessed 15 January 2015.
- 55.Gao J, Yin L. 2014. Drug development for controlling Ebola epidemic—a race against time. Drug Discov Ther 8:229–231. doi: 10.5582/ddt.2014.01040. [DOI] [PubMed] [Google Scholar]
- 56.ProMED-mail. 8 August 2014. Ebola virus disease—West Africa (120): MSF, Sierra Leone, drug, PAHO, aid. International Society for Infectious Diseases, Brookline, MA: http://www.promedmail.org/direct.php?id=2673098 Accessed 15 January 2015. [Google Scholar]
- 57.Feldmann H, Sanchez A, Geisbert TW. 2013. Filoviridae: Marburg and Ebola viruses, p 1410–1448. In Knipe DM, Howley PM, Cohen JI, Griffin DE, Lamb RA, Martin MA, Racaniello VR, Roizman B (ed), Fields virology, 6th ed Lippincott Williams & Wilkins, Philadelphia, PA. [Google Scholar]
- 58.Dowling W, Thompson E, Badger C, Mellquist JL, Garrison AR, Smith JM, Paragas J, Hogan RJ, Schmaljohn C. 2007. Influences of glycosylation on antigenicity, immunogenicity, and protective efficacy of Ebola virus GP DNA vaccines. J Virol 81:1821–1837. doi: 10.1128/JVI.02098-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.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. 2014. Reversion of advanced Ebola virus disease in nonhuman primates with ZMapp. Nature 514:47–53. doi: 10.1038/nature13777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rollin PE, Bausch DG, Sanchez A. 2007. Blood chemistry measurements and D-dimer levels associated with fatal and nonfatal outcomes in humans infected with Sudan Ebola virus. J Infect Dis 196(Suppl 2):S364–S371. doi: 10.1086/520613. [DOI] [PubMed] [Google Scholar]
- 61.Lyon GM, Mehta AK, Varkey JB, Brantly K, Plyler L, McElroy AK, Kraft CS, Towner JS, Spiropoulou C, Stroher U, Uyeki TM, Ribner BS. 2014. Clinical care of two patients with Ebola virus disease in the United States. N Engl J Med 371:2402–2409. doi: 10.1056/NEJMoa1409838. [DOI] [PubMed] [Google Scholar]
- 62.Erhabor O, Adias TC. 2011. From whole blood to component therapy: the economic, supply/demand need for implementation of component therapy in sub-Saharan Africa. Transfus Clin Biol 18:516–526. doi: 10.1016/j.tracli.2011.06.001. [DOI] [PubMed] [Google Scholar]
- 63.Mupapa K, Massamba M, Kibadi K, Kuvula K, Bwaka A, Kipasa M, Colebunders R, Muyembe-Tamfum JJ. 1999. Treatment of Ebola hemorrhagic fever with blood transfusions from convalescent patients. International Scientific and Technical Committee. J Infect Dis 179(Suppl 1):S18–S23. [DOI] [PubMed] [Google Scholar]
- 64.Jahrling PB, Geisbert JB, Swearengen JR, Larsen T, Geisbert TW. 2007. Ebola hemorrhagic fever: evaluation of passive immunotherapy in nonhuman primates. J Infect Dis 196(Suppl 2):S400–S403. doi: 10.1086/520587. [DOI] [PubMed] [Google Scholar]
- 65.Luke TC, Casadevall A, Watowich SJ, Hoffman SL, Beigel JH, Burgess TH. 2010. Hark back: passive immunotherapy for influenza and other serious infections. Crit Care Med 38:e66–e73. doi: 10.1097/CCM.0b013e3181d44c1e. [DOI] [PubMed] [Google Scholar]
- 66.Jahrling PB, Geisbert J, Swearengen JR, Jaax GP, Lewis T, Huggins JW, Schmidt JJ, LeDuc JW, Peters CJ. 1996. Passive immunization of Ebola virus-infected cynomolgus monkeys with immunoglobulin from hyperimmune horses. Arch Virol Suppl 11:135–140. [DOI] [PubMed] [Google Scholar]
- 67.Jahrling PB, Geisbert TW, Geisbert JB, Swearengen JR, Bray M, Jaax NK, Huggins JW, LeDuc JW, Peters CJ. 1999. Evaluation of immune globulin and recombinant interferon-alpha2b for treatment of experimental Ebola virus infections. J Infect Dis 179(Suppl 1):S224–S234. [DOI] [PubMed] [Google Scholar]
- 68.Dye JM, Herbert AS, Kuehne AI, Barth JF, Muhammad MA, Zak SE, Ortiz RA, Prugar LI, Pratt WD. 2012. Postexposure antibody prophylaxis protects nonhuman primates from filovirus disease. Proc Natl Acad Sci U S A 109:5034–5039. doi: 10.1073/pnas.1200409109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.WHO. 2014. Use of convalescent whole blood or plasma collected from patients recovered from Ebola virus disease. WHO, Geneva, Switzerland: http://apps.who.int/iris/bitstream/10665/135591/1/WHO_HIS_SDS_2014.8_eng.pdf?ua=1 Accessed 15 January 2015. [Google Scholar]
- 70.Clark DV, Jahrling PB, Lawler JV. 2012. Clinical management of filovirus-infected patients. Viruses 4:1668–1686. doi: 10.3390/v4091668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Guimard Y, Bwaka MA, Colebunders R, Calain P, Massamba M, De Roo A, Mupapa KD, Kibadi K, Kuvula KJ, Ndaberey DE, Katwiki KR, Mapanda BB, Nkuku OB, Fleerackers Y, Van den Enden E, Kipasa MA. 1999. Organization of patient care during the Ebola hemorrhagic fever epidemic in Kikwit, Democratic Republic of the Congo, 1995. J Infect Dis 179(Suppl 1):S268–S273. [DOI] [PubMed] [Google Scholar]
- 72.Wolf T, Kann G, Becker S, Stephan C, Brodt H-R, de Leuw P, Grünewald T, Vogl T, Kempf VAJ, Keppler OT, Zacharowski K. 2015. Severe Ebola virus disease with vascular leakage and multiorgan failure: treatment of a patient in intensive care. Lancet; 385:1428–1435. doi: 10.1016/S0140-6736(14)62384-9. [DOI] [PubMed] [Google Scholar]
- 73.Connor MJ Jr, Kraft C, Mehta AK, Varkey JB, Lyon GM, Crozier I, Stroher U, Ribner BS, Franch HA. 2015. Successful delivery of RRT in Ebola virus disease. J Am Soc Nephrol 26:31–37. doi: 10.1681/ASN.2014111057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.New York Times. 5 January 2015. How many Ebola patients have been treated outside of Africa? http://www.nytimes.com/interactive/2014/07/31/world/africa/ebola-virus-outbreak-qa.html?_r=0 Accessed 15 January 2015.
- 75.Marcellin P, Lau GK, Bonino F, Farci P, Hadziyannis S, Jin R, Lu ZM, Piratvisuth T, Germanidis G, Yurdaydin C, Diago M, Gurel S, Lai MY, Button P, Pluck N. 2004. Peginterferon alfa-2a alone, lamivudine alone, and the two in combination in patients with HBeAg-negative chronic hepatitis B. N Engl J Med 351:1206–1217. doi: 10.1056/NEJMoa040431. [DOI] [PubMed] [Google Scholar]
- 76.Hoofnagle JH, Seeff LB. 2006. Peginterferon and ribavirin for chronic hepatitis C. N Engl J Med 355:2444–2451. doi: 10.1056/NEJMct061675. [DOI] [PubMed] [Google Scholar]
- 77.Hauschild A, Petres-Dunsche C. 1992. Intralesional treatment of classical Kaposi sarcoma with interferon-alpha. Hautarzt 43:789–791. (In German.). [PubMed] [Google Scholar]
- 78.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. 2013. Interferon-beta therapy prolongs survival in rhesus macaque models of Ebola and Marburg hemorrhagic fever. J Infect Dis 208:310–318. doi: 10.1093/infdis/jis921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.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. 2013. FDA-approved selective estrogen receptor modulators inhibit Ebola virus infection. Sci Transl Med 5:190ra79. doi: 10.1126/scitranslmed.3005471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.National Cancer Institute. 16 October 2014. Toremifene. National Cancer Institute, Bethesda, MD: http://www.cancer.gov/cancertopics/druginfo/toremifene Accessed 15 January 2015. [Google Scholar]
- 81.Dutertre M, Smith CL. 2000. Molecular mechanisms of selective estrogen receptor modulator (SERM) action. J Pharmacol Exp Ther 295:431–437. [PubMed] [Google Scholar]
- 82.Cooper CL, Bavari S. 2015. A race for an Ebola vaccine: promises and obstacles. Trends Microbiol 23:65–66. doi: 10.1016/j.tim.2014.12.005. [DOI] [PubMed] [Google Scholar]
- 83.ClinicalTrials.gov. 11 November 2014. A study to find out if the new Ebola vaccine is safe and stimulates immunity that might protect adults in Kilifi, Kenya. Registration number NCT02296983 https://www.clinicaltrials.gov/ct2/show/study/NCT02296983 Accessed 28 February 2015.
- 84.Wong G, Richardson JS, Pillet S, Racine T, Patel A, Soule G, Ennis J, Turner J, Qiu X, Kobinger G. Adenovirus-vectored vaccine provides post-exposure protection to Ebola virus-infected nonhuman primates. J Infect Dis, in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.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. 2011. Management of accidental exposure to Ebola virus in the biosafety level 4 laboratory, Hamburg, Germany. J Infect Dis 204(Suppl 3):S785–S790. doi: 10.1093/infdis/jir298. [DOI] [PubMed] [Google Scholar]
- 86.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. 2015. Emergency postexposure vaccination with vesicular stomatitis virus-vectored Ebola vaccine after needlestick. JAMA 313:1249–1255. doi: 10.1001/jama.2015.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.FDA. 2014. Guidance for industry—product development under the animal rule. FDA, Washington, DC: http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm399217.pdf Accessed 15 January 2015. [Google Scholar]
- 88.Parren PW, Geisbert TW, Maruyama T, Jahrling PB, Burton DR. 2002. Pre- and postexposure prophylaxis of Ebola virus infection in an animal model by passive transfer of a neutralizing human antibody. J Virol 76:6408–6412. doi: 10.1128/JVI.76.12.6408-6412.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Oswald WB, Geisbert TW, Davis KJ, Geisbert JB, Sullivan NJ, Jahrling PB, Parren PW, Burton DR. 2007. Neutralizing antibody fails to impact the course of Ebola virus infection in monkeys. PLoS Pathog 3:e9. doi: 10.1371/journal.ppat.0030009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Richardson JS, Wong G, Pillet S, Schindle S, Ennis J, Turner J, Strong JE, Kobinger GP. 2011. Evaluation of different strategies for post-exposure treatment of Ebola virus infection in rodents. J Bioterror Biodef S1:1–7. doi: 10.4172/2157-2526.s1-007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Qiu X, Wong G, Fernando L, Ennis J, Turner JD, Alimonti JB, Yao X, Kobinger GP. 2013. Monoclonal antibodies combined with adenovirus-vectored interferon significantly extend the treatment window in Ebola virus-infected guinea pigs. J Virol 87:7754–7757. doi: 10.1128/JVI.00173-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Qiu X, Wong G, Fernando L, Audet J, Bello A, Strong J, Alimonti JB, Kobinger GP. 2013. mAbs and Ad-vectored IFN-alpha therapy rescue Ebola-infected nonhuman primates when administered after the detection of viremia and symptoms. Sci Transl Med 5:207ra143. doi: 10.1126/scitranslmed.3006605. [DOI] [PubMed] [Google Scholar]
- 93.Nakayama E, Saijo M. 2013. Animal models for Ebola and Marburg virus infections. Front Microbiol 4:267. doi: 10.3389/fmicb.2013.00267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Butler D. 2014. Ebola drug trials set to begin amid crisis. Nature 513:13–14. doi: 10.1038/513013a. [DOI] [PubMed] [Google Scholar]
- 95.Schieffelin JS, Shaffer JG, Goba A, Gbakie M, Gire SK, Colubri A, Sealfon RS, Kanneh L, Moigboi A, Momoh M, Fullah M, Moses LM, Brown BL, Andersen KG, Winnicki S, Schaffner SF, Park DJ, Yozwiak NL, Jiang PP, Kargbo D, Jalloh S, Fonnie M, Sinnah V, French I, Kovoma A, Kamara FK, Tucker V, Konuwa E, Sellu J, Mustapha I, Foday M, Yillah M, Kanneh F, Saffa S, Massally JL, Boisen ML, Branco LM, Vandi MA, Grant DS, Happi C, Gevao SM, Fletcher TE, Fowler RA, Bausch DG, Sabeti PC, Khan SH, Garry RF. 2014. Clinical illness and outcomes in patients with Ebola in Sierra Leone. N Engl J Med 371:2092–2100. doi: 10.1056/NEJMoa1411680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.McElroy AK, Erickson BR, Flietstra TD, Rollin PE, Nichol ST, Towner JS, Spiropoulou CF. 2014. Ebola hemorrhagic fever: novel biomarker correlates of clinical outcome. J Infect Dis 210:558–566. doi: 10.1093/infdis/jiu088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.McCarthy M. 2014. US signs contract with ZMapp maker to accelerate development of the Ebola drug. BMJ 349:g5488. doi: 10.1136/bmj.g5488. [DOI] [PubMed] [Google Scholar]