+ All Categories
Home > Documents > Therapeutics for filovirus infection: traditional approaches and progress towards in...

Therapeutics for filovirus infection: traditional approaches and progress towards in...

Date post: 03-Feb-2017
Category:
Upload: sina
View: 212 times
Download: 0 times
Share this document with a friend
20
1. Introduction 2. Filoviruses, hemorrhagic disease, and animal models 3. Drug discovery for filoviruses through library screening 4. The filovirus proteins and their functions 5. Compound library screening to identify viral entry inhibitors 6. Mechanistic protein functional studies reveal inhibitors with structural importance 7. Expert opinion Review Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design Amy C Shurtleff , Tam L Nguyen, David A Kingery & Sina Bavari U.S. Army Medical Research Institute of Infectious Diseases, Integrated Toxicology Division, Frederick, MD, USA Introduction: Ebolaviruses and marburgviruses cause severe and often lethal human hemorrhagic fevers. As no FDA-approved therapeutics are available for these infections, efforts to discover new therapeutics are important, especially because these pathogens are considered biothreats and emerging infectious diseases. All methods for discovering new therapeutics should be considered, including compound library screening in vitro against virus and in silico structure-based drug design, where possible, if sufficient biochemical and structural information is available. Areas covered: This review covers the structure and function of filovirus proteins, as they have been reported to date, as well as some of the current antiviral screening approaches. The authors discuss key studies mapping small-molecule modulators that were found through library and in silico screens to potential sites on viral proteins or host proteins involved in virus trafficking and pathogenesis. A description of ebolavirus and marburgvirus diseases and available animal models is also presented. Expert opinion: To discover novel therapeutics with potent efficacy using sophisticated computational methods, more high-resolution crystal structures of filovirus proteins and more details about the protein functions and host interaction will be required. Current compound screening efforts are finding active antiviral compounds, but an emphasis on discovery research to inves- tigate protein structures and functions enabling in silico drug design would provide another avenue for finding antiviral molecules. Additionally, targeting of protein-protein interactions may be a future avenue for drug discovery since disrupting catalytic sites may not be possible for all proteins. Keywords: ebolavirus, filovirus, glycoprotein, library screening, marburgvirus, protein interactions, rational drug design, small molecule, structure, structure-based drug design, viral entry mechanism, viral hemorrhagic fever Expert Opin. Drug Discov. (2012) 7(10):935-954 1. Introduction Structure-based drug design, or the rational design of drugs using structural insights gained from known three-dimensional structures of biological targets, is a sophisti- cated computationally enabled method which has been used to produce successful new treatments for diseases such as cancer, influenza and acquired immune defi- ciency syndrome (AIDS) [1-4]. The utility of this approach depends upon the deter- mination of the three-dimensional structures of proteins of interest, and for many highly virulent viruses, such protein structures have yet to be solved, making the immediate application of this method impossible. For example, the bcr-abl fusion protein, which is characteristic of chronic myelogenous leukemia and some other 10.1517/17460441.2012.714364 © 2012 Informa UK, Ltd. ISSN 1746-0441 935 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Gazi Univ. on 08/20/14 For personal use only.
Transcript

1. Introduction

2. Filoviruses, hemorrhagic

disease, and animal models

3. Drug discovery for filoviruses

through library screening

4. The filovirus proteins and

their functions

5. Compound library screening

to identify viral entry inhibitors

6. Mechanistic protein functional

studies reveal inhibitors with

structural importance

7. Expert opinion

Review

Therapeutics for filovirusinfection: traditional approachesand progress towards in silicodrug designAmy C Shurtleff†, Tam L Nguyen, David A Kingery & Sina Bavari†U.S. Army Medical Research Institute of Infectious Diseases, Integrated Toxicology Division,

Frederick, MD, USA

Introduction: Ebolaviruses and marburgviruses cause severe and often lethal

human hemorrhagic fevers. As no FDA-approved therapeutics are available

for these infections, efforts to discover new therapeutics are important,

especially because these pathogens are considered biothreats and emerging

infectious diseases. All methods for discovering new therapeutics should be

considered, including compound library screening in vitro against virus and

in silico structure-based drug design, where possible, if sufficient biochemical

and structural information is available.

Areas covered: This review covers the structure and function of filovirus

proteins, as they have been reported to date, as well as some of the current

antiviral screening approaches. The authors discuss key studies mapping

small-molecule modulators that were found through library and in silico

screens to potential sites on viral proteins or host proteins involved in virus

trafficking and pathogenesis. A description of ebolavirus and marburgvirus

diseases and available animal models is also presented.

Expert opinion: To discover novel therapeutics with potent efficacy using

sophisticated computational methods, more high-resolution crystal structures

of filovirus proteins and more details about the protein functions and host

interaction will be required. Current compound screening efforts are finding

active antiviral compounds, but an emphasis on discovery research to inves-

tigate protein structures and functions enabling in silico drug design would

provide another avenue for finding antiviral molecules. Additionally,

targeting of protein-protein interactions may be a future avenue for drug

discovery since disrupting catalytic sites may not be possible for all proteins.

Keywords: ebolavirus, filovirus, glycoprotein, library screening, marburgvirus, protein

interactions, rational drug design, small molecule, structure, structure-based drug design,

viral entry mechanism, viral hemorrhagic fever

Expert Opin. Drug Discov. (2012) 7(10):935-954

1. Introduction

Structure-based drug design, or the rational design of drugs using structural insightsgained from known three-dimensional structures of biological targets, is a sophisti-cated computationally enabled method which has been used to produce successfulnew treatments for diseases such as cancer, influenza and acquired immune defi-ciency syndrome (AIDS) [1-4]. The utility of this approach depends upon the deter-mination of the three-dimensional structures of proteins of interest, and for manyhighly virulent viruses, such protein structures have yet to be solved, making theimmediate application of this method impossible. For example, the bcr-abl fusionprotein, which is characteristic of chronic myelogenous leukemia and some other

10.1517/17460441.2012.714364 © 2012 Informa UK, Ltd. ISSN 1746-0441 935All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

types of malignancies was specifically targeted by imatinib, atyrosine kinase inhibitor designed through structure-baseddrug design and medicinal chemistry through a knowledgeof the structure of the protein target [5,6]. Zanamivir, a neur-aminidase inhibitor, which is approved for prophylaxis andtreatment of influenza Type A and B infections, was also dis-covered through structure-based drug design [7,8]. A paradigmfor structure-based drug design of anti-infective agents is thearray of approved drugs that have been developed to targetthe human immunodeficiency virus (HIV) protease [1]. HIVprotease cleaves the Phe(Tyr)-Pro bond for group-specificantigen and group-specific antigen polymerase precursor pro-teins. Structural studies of HIV protease function greatlyimproved the speed and efficiency of the rational design anddevelopment of the first protease inhibiting anti-HIV drugs:saquinavir, ritonavir, indinavir, and nelfinavir [1,9]. The deter-mination of the HIV protease structure and its active sitefacilitated the rational design and development of drugs thatblocked native substrate binding [10]. The anti-HIV drugsamprenavir and lopinavir, as well as those listed above, arepeptido-mimetics that were designed to bind to the activesite of the HIV protease dimer. A newer generation of anti-HIV drugs, characterized by atazanavir, fosamprenavir, anddarunavir, were designed to overcome mutations in HIVprotease that resulted in viral drug resistance.Such a wealth of rationally designed compounds is not avail-

able for many other highly pathogenic viruses. To date, thereexists only a handful of high-resolution crystal structures forintact filovirus proteins and consequently very few studies thatdescribe the interaction of a candidate therapeutic with a knownviral protein structure or functionally active site. Even fewer

studies describe an approach using classical in silico structure-based drug design [11,12]. Instead, filovirus researchers havehad to rely upon compound library screening to identify smallmolecules with drug-like properties, such as kinase inhibitorsgenistein and tyrphostin, as two examples [13-15]. A number ofcandidate therapeutics have been found through compoundlibrary screening or through design of antisense based products,and some of these candidates have been studied to delineatetheir mechanism of action [14,15]. For some of the available filo-virus candidate therapeutics, a description of how they mayinteract with a target protein is available, or can be predictedbased on published work [15-17]. In addition, an area with strongtherapeutic promise has been achieved through the design ofantisense-based products, thoroughly reviewed in [18].

2. Filoviruses, hemorrhagic disease,and animal models

The filoviruses are highly lethal zoonotic agents of viralhemorrhagic fevers that are of concern as emerging pathogensand potential bioweapons. These viruses emerge in unpredic-table sporadic outbreaks in certain areas of Africa and arepurported to have been weaponized in the former SovietUnion [19]. There are no available therapeutics which havebeen approved by the United States Food and Drug Admini-stration (FDA) for the treatment of filovirus infections, so thereis a great need for continued drug discovery efforts to protectagainst these lethal viruses. Due to their highly pathogenicnature, handling these viruses poses great risk for laboratorypersonnel. The lack of vaccines or therapeutics available fortreatment of laboratory personnel or the general public, drivestheir classification as Class 4 risk-group pathogens whichmust be handled at biosafety level 4 (BSL-4), the highest levelof biocontainment [20]. Research on BSL-4 agents is quitelimited because of constraints due to biosafety and biosecurity,and there is a short list of laboratories worldwide whereBSL-4 research can be performed [19,20].

Highly virulent filoviruses are endemic to parts of Africa,and some filoviruses with unknown human pathogenicityhave been found in the Philippines. Ebolaviruses were firstdiscovered during simultaneous outbreaks in Zaire (todaythe Democratic Republic of Congo) and Sudan in 1976.Today there are five known species in the Ebolavirus genus:Zaire ebolavirus (EBOV), Sudan ebolavirus (SUDV), Restonebolavirus (RESTV), Taı Forest ebolavirus (TAFV), andBundibugyo virus (BDBV) [19]. The Marburgvirus genusconsists of one only species, Marburg marburgvirus, whichhas two members: Marburg virus (MARV) and Ravn virus(RAVV) [21]. Marburgvirus was the first filovirus discovered,when in 1967 Yugoslavian laboratory workers manipulatingan African green monkey imported from Uganda becameinfected and initiated a 31-person outbreak of hemorrhagicfever in Marburg, Germany [22]. The case fatality rates ofEbola and Marburg hemorrhagic fevers range from 50 -- 90% and 23 -- 90 %, respectively [23,24].

Article highlights.

. The filoviruses are virulent, lethal human viruses forwhich FDA-approved therapeutics are unavailable.

. Rodent and nonhuman primate animal models offilovirus infection are in use for evaluation of efficacyof therapeutics.

. Drug discovery approaches for anti-filovirus compoundsinclude compound library screening against authenticvirus or pseudotyped viruses, and in silico structure-based drug design.

. General functions have long been known for most ofthe filovirus proteins, but many structural and functionaldetails are still unknown. The available structuralinformation may begin to inform in silico drug design,but more structural and functional information isneeded about viral proteins and any involvedhost proteins.

. Entry of filoviruses into host cells is an area of researchwhich has recently been addressed and some structure-based drug design techniques may be employed fordiscovery of new therapeutics directed against specifictarget processes such as entry.

This box summarizes key points contained in the article.

A. C. Shurtleff et al.

936 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

Outbreaks in human populations are rare and oftenself-limiting as person-to-person transmission is only docu-mented to occur upon close contact with blood or otherbodily fluids [25]. In experimental models of infection, aerosolpreparations of filoviruses are highly infectious, and have ledto concerns that these agents could be used in events ofmass dissemination [26,27]. In humans, the incubation periodsfor EBOV and MARV infection are generally 4 -- 7 days witha reported range of 2 -- 21 days and death often occurs within6 days of symptom onset [28]. The disease progresses quickly,with symptoms such as fever, chills, headache, malaise, andmyalgia typically appearing within 4 days after infection [28-31].Nausea, vomiting, abdominal pain, diarrhea, anorexia, a dry,non-productive cough, chest pain and shortness of breathhave all been reported as the predominant gastrointestinaland respiratory symptoms [24,32,33]. Vascular abnormalitiesinclude conjunctival injection, hypotension and edema [33].Hemorrhagic signs include development of petechiae on thetorso and arms, ecchymoses, bleeding from venipuncture sitesand mucous membranes, gingival bleeding, epistaxis, andvisceral hemorrhaging [28,33]. Currently, only supportive careor experimental therapeutics administered under specialprotocols can be offered to individuals with suspected orconfirmed cases of EBOV or MARV infection.

Animal models of filovirus infection are available to aid thestudy of disease pathology and evaluate potential therapeuticsand vaccines (Table 1) [18,34,35]. Mice and guinea pigs do notsuccumb to infection with wild-type ebolaviruses or marburgvi-ruses unless the virus has been passaged and adapted to viru-lence for the species [36-39]. For EBOV and MARV mousemodels, there is rapid viremia, high viral burden in the spleen,liver and multiple organ tissues, lymphopenia, thrombocytope-nia, and liver damage resulting in high AST and ALT levels inserum [38]. For both mouse models, there is an early inflamma-tory cytokine response, and lymphopenia and neutropenia areobserved, similar to the observations in guinea pig and nonhu-man primate infections. Mice do not demonstrate disseminatedintravascular coagulation (DIC), fibrin deposits, or skin rashessuch as petechiae, but some coagulation abnormalities arefound, such as induction of fibrin degradation products.Rodent models are useful for early stage discovery, as com-pounds can be evaluated in these models for antiviral effects,optimal delivery route and dosing frequency, dose level,prophylactic versus therapeutic efficacy studies and some earlyin vivo toxicity assessments [40,41]. Generally, due to small size,ease of handling, and possibility of working with larger groupsizes (where generally n is at least 10), mice are used as the initialmodels for testing, and success in this species generally warrantsprogression into the guinea pig efficacy model [42,43]. Althoughthese rodent infection models are valuable screening tools forevaluating virus-specific therapeutic agents, the nonhuman pri-mate (NHP) models are considered the most reliable predictorof therapeutic efficacy.

NHP are the most suitable model species to satisfy theFDA’s Animal Rule of Efficacy for licensure of therapeutics

and vaccines for agents where it is unethical or unfeasible toconduct human efficacy evaluations [34,44,45]. Several speciesof NHP are susceptible to infection with filoviruses [26,46-48].However, the majority of research is performed using rhesusmonkeys and cynomolgus macaques, as these animal modelsreliably reproduce human disease [49]. Fever in monkeys ismeasureable 3 -- 4 days after infection, and is coincidentwith onset of viremia [50]. A macropapular rash generallyappears by day 3 -- 4. Dehydration, anorexia and diarrheabegin approximately 4 days after infection [50-52]. Nearly allinfected animals appear ill by day 5 or 6, exhibiting moderateto severe depression in activity, dehydration, and hunchedposture; rashes with hemorrhage appear around day6 [46,48,50,53-55]. White blood cell counts and various bloodchemistry parameters are drastically altered during humanand NHP infection with EBOV or MARV. Lymphocytope-nia occurs around days 3 -- 4 postinfection [53-57]. Neutrophilcounts increase and comprise the majority of the circulatingleukocyte population; concomitantly monocytes, macro-phages and natural killer (NK) cell numbers decline. Thehemorrhagic manifestations include alterations in bloodcoagulation, fibrinolysis, thrombocytopenia, consumptionof coagulation factors, deficiency of anticoagulation, andincreased levels of fibrin degradation products [46,48,50,53-55].The average time to death after intramuscular (i.m.) challengewith 1000 plaque forming units (pfu) of EBOV is ~ 8 days forrhesus monkeys and ~ 6 days for cynomolgus macaques [28],which points to a longer therapeutic window for treatmentin models utilizing rhesus monkeys. In fact, research in thefilovirus field has frequently taken advantage of this longertimeframe for rhesus monkeys to manifest symptoms andsuccumb to disease, and have preferentially used this speciesover cynomolgus macaques for testing of small-moleculetherapeutics [43,58]. Even though most but not all filovirusresearchers have perpetuated the practice of evaluatingtherapeutics in rhesus monkeys, there has been no guidancefrom the FDA that this is the definitive model species fortherapeutics evaluation. While the human and NHP diseaseappear to be very similar, there is still a great amount ofresearch that can be done to characterize all of the animalmodels, to prove they are each suitably useful animal modelsfor therapeutics discovery. Table 1 provides a summary of use-ful animal models of filovirus infection for efficacy evaluationof small molecules.

3. Drug discovery for filovirusesthrough library screening

In the face of the challenges stated above, significant discoveriesand advances in therapeutics development have been made. Tocircumvent research limitations using authentic viral agents,detailed molecular biology studies have been performed withcloned genes, expressed viral proteins, and pseudotyped virusparticles. Pseudotyped viruses are useful tools for studyingvirus entry or egress in some systems [59]. These pseudotyped

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 937

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

Table

1.A

variety

ofanim

almodels

are

available

forthestudyoffilovirusinfection.

Species

Description

Advantagesofmodel

Disadvantagesofmodel

Ref.

Wild-typemice

Mouse-adaptedEBOVandMARV

cause

rapid

viremia

onset,high

viralburdenin

multiple

target

organtissues,

lymphopenia,

thrombocytopenia,andliver

damageresultingin

highserum

levelsofASTandALT

Smallsize

allowsforlargergroup

sizes,

inexpensive,easy

tohandle

inbiocontainment,requiresm

all

quantitiesoftest

compounddue

tosm

allbodysize,goodforinitial

testingoftherapeutics

invivo

toassess

potentialefficacy

Requiresuse

ofpassageadapted

virusesforlethalityandpathology.

Inherentstrongim

muneresponse

mayconfoundsomeresults.

PoormodelsforDIC,petechiae,

fibrinorcoagulationabnorm

alities.

FDA

doesnotpreferthismodel

system

butwillacceptdata

[38,118,177-179]

STAT1knockoutmice,Type-I

interferonresponse

knockoutmice

These

micesuccumbto

non

rodent-adaptedfilovirusinfection

deliveredbysubcutaneous

inoculation

Modelsmaybeusedfor

therapeutics

discovery

against

non-

passageadaptedvirusisolates

Modelsmaynotbeasusefulfor

immuneresponse

orvaccinestudies

[180,181]

Humanizedmice

ImmunodeficientIL2rg

nullmice

engraftedwithhumanim

mune

cells

willreplicate

wild-

typefiloviruses,

butdonot

succumbto

infection

Infectionofhumanim

munesystem

components

canbeevaluatedusing

anon-m

ouse

adaptedvirus

These

micedonotdemonstrate

lethalfilovirusdisease,provides

simply

aninfectionmodel

[182]

Guineapigs

Guineapig

passage-adaptedEBOV

andMARVdevelop:

febrile

illness

dehydration

viralreplicationin

multiple

organs

lack

rash/hemorrhageand

coagulationabnorm

alities

Disease

inguineapigsismore

similarto

humandisease

thanthat

ofmice.Resultsin

thismodelare

more

predictive

ofresultswhich

maybeobtainedin

NHPmedium

size

allowsreasonable

group

sizes(n

=5to

10)

Requiresuse

ofpassageadapted

virusesforlethalityandpathology.

FDA

doesnotpreferthismodel

system

butwillacceptdata.

Geneticknockoutstrainsand

immunologicalreagents

are

largely

unavailable

[37,39,47,48,183-186]

Nonhumanprimates(NHP)

Thismodelisthemost

similarto

humandisease

considering:

clinicalpresentation

hematology

serum

clinicalchemistry

anatomic

pathologyfindings

humoralandcellularim

mune

responses

meantimeto

death

is7--10days

Susceptible

towild-typeorclinical

virusisolates.

FDA

expectsdata

generatedin

thismodel.Complete

genomesequence

isavailable.

NHPspeciesare

alreadywidely

usedin

toxicologystudies

Expensive,available

inlim

ited

quantities,

leadingto

smallsample

size

pergroup.Usagepresents

ethicalchallenges.

Largebodysize

requireslargeamountoftest

compoundforevaluation

[28,34,48,53,187-190]

Thechallengedose

forNHPismost

frequently1000pfu,intramuscularinjection.

ALT:Alanineaminotransferase;AST:Aspartate

aminotransferase;DIC:Disseminatedintravascularcoagulation;FD

A:U.S.FoodandDrugAdministration;STAT1:signaltransducerandactivatoroftranscription1.

A. C. Shurtleff et al.

938 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

viruses take advantage of multiple parts of several differentviruses to study a specific process in the viral life cycle. Forexample, the core proteins of one virus packaging system suchas a lentivirus or vesicular stomatitis virus (VSV) are coexpressedin a producing cell line with the glycoprotein gene of the virusof interest plus a reporter gene like luciferase [59-61]. The endproduct is a single-cycle virion which is not replication compe-tent, but which can enter a target cell using the entry glycopro-tein of interest, such as filovirus GP1,2, thereby providing asystem in which to investigate virus-entry inhibitors. Thesepseudotyped viruses are useful tools for higher throughputscreening of libraries of small molecules for activity against viralentry and trafficking, and there are several hit compounds thathave been discovered using this approach [13,15,62]. There hasbeen a push to discover filovirus therapeutics using in vitrohigher throughput screens, since these methods could identifya possible lead candidate therapeutic, regardless of the

mechanism of action [63]. Once a hit candidate is found througheither library screens or structure-based drug design methods(Figure 1), it must be evaluated under rigorous in vitro condi-tions to determine its true specificity for and potency againstauthentic virus. Assays for cytotoxicity, ADMET and solubilitywill assess its chemical tractability as a molecule which could besuccessfully advanced as a safe and effective clinical candi-date [63,64]. Compounds with favorable activity and drug-likeproperties can proceed through in vivo efficacy studies in animalmodels. Mechanism of action studies are then performed tohelp define the antiviral activity of the compound and completethe data package to be evaluated by the FDA.

4. The filovirus proteins and their functions

Ebolaviruses and marburgviruses have 19kb genomes thatencode seven genes. For marburgviruses, these seven genes

Structure based approach

• Crystal structure of a protein/enzymatic site of importance is available • In-silico molecular dynamics and energy minimization algorithms identify candidate

small molecules with predicted binding • Synthetic chemistry is performed to produce

derivatives for screening

Small molecules found using these two approaches are screened in cell based assays againstpseudotyped viruses (BSL-2), GFP expressing or authentic viruses (BSL-4), in a high throughput format.

Hit candidates are identified andrescreened against authentic virus for

confirmation of antiviral activity

Mechanism of action studies (e.g.,):

• Viral entry/egress • Confocal microscopy • Effect of viral or host gene knockdown on viral growth, viral transcription/translation, etc.

• Mutational analysis

Continued screening: • In vitro assays for high antiviral potency • Specificity • Cytotoxicity • Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) • Favorable solubility properties • Agreement with Lipinski’s rule of 5 [59]. • Hits proceed through in vivo efficacy studies in animal models, and preclinical testing.

or pseudotyped virus

Authentic virus

Library based approach

• Libraries of thousands of small molecules are identified and purchased for execution of antiviral screening assays

OH

OH H3C

H3C

H3C

CH3

OO

O

O

N

NC2HF4

Figure 1. A general comparison of structure-based drug design and compound library screening approaches for the discovery

of new filovirus therapeutics.

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 939

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

Table

2.Filovirusprotein

functionsandavailabilityofcrystalstructures.

Protein

Generalfunctionsforeach

protein,andso

meknown

differencesbetw

eenEBOV

andMARV

(references)

Available

structures

Rationaldrugdesigned;potential

targetfortheprotein

NP

Nucleoprotein.Nucleocapsidandinclusionbody

form

ation;encapsulationofRNA

genomeand

antigenome,replicationandtranscriptionofviral

genome

[68].Part

oftheRNP.BindsVP35,andassociates

withVP30andL[69]

Notavailable

None;targetinteractionwithorbindingsitesonVP35,

VP30orL

VP30

Secondary

nucleoprotein,bindsNPin

inclusionbodies,

partofRNP.Oligomerizesforitsfunctionasa

transcriptionalactivator[191].ForEBOV,knownto

bindNP

andssRNA

[82,83]

Available

[78]

None;targetbasicpatchofVP30aroundtheLys180to

inhibittranscriptionalactivation

VP35

Associatesin

RNPcomplexes,bindsRNA.Part

of

replicase-transcriptase

holoenzymewithL,

initiates

transcription.EBOVVP35suppressesType-IIFN

production

[84,192]

Available

[69,92,93]

None;targetbasicpatchofVP35aroundtheArg312to

preventVP35bindingto

dsRNA

andpromote

IFN

production

Lprotein

RNAdependentRNA

polymerase;bindsVP35to

form

replicase-transcriptase

holoenzyme

[94]

Notavailable

None;poly-adenylation,proofreadingfunctionorinteraction

withZAP

VP40

Viralmatrix

protein.Accumulatesatcellularmembrane

proliferationsites,

responsible

forviralbudding

[104-107].

Associateswithmicrotubulesandoutercellmembranes.

MARVVP40(notEBOV)blocksIFN

andIL-6

[193].EBOV

VP40hastw

oLdomains:

aPTAPandaPPxY

L-domain,

butMARVhasonlyonePPxY

L-domain

[113]

Available

[98,101,102]

Compound5539-0062targetinginteractionofTsg101with

EBOVVP40atthePTAPmotif[12]

VP24

Secondary

matrix

protein,itcolocalizeswithVP40and

functionsin

nucleocapsidassembly

withNP

[19,68,116].

EBOV(notMARV)VP24inhibitscellularresponsesto

IFN

[120-122]

Available

[123]

None;targetVP24bindingto

STAT1,whichmayalter

interferonsignalingandkaryopherinsequestering

GP1,2

Viralspikesurface

andtransm

embraneprotein,servesto

bindcells

andpromote

entry[19,67].Somefunctional

domainsofGP1,2mayhave

immunosuppressive

functions[132,139,194,195]

Available

[151,196-198]

None;screeningfoundcompounds3.0

and3.47to

interact

withNPC-1

bindingto

cleavedGP1[17];compound7also

foundthroughscreeningactivities[15].Itinteractswitha

hydrophobicpocketattheinterface

ofGP1andGP2

sGP(EBOVonly)

Secretedglycoprotein

intheform

ofaparallel

homodim

er[67].Unknownfunction.sG

Pnotencodedin

MARVgenome

Notavailable

None;Functionandtargetunknown

ssGP(EBOVonly)

Secondary

secretedglycoprotein

withunknownfunction.

Secretedasamonomer.ssGPnotencodedin

MARV

genome

Notavailable

None;Functionandtargetunknown

A. C. Shurtleff et al.

940 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

are translated into seven unique proteins. However, alter-native mRNA translation results in a total of 9 ebolavirusproteins [65-67]. A significant body of literature is availablethat provides preliminary insights into how these proteinsare involved in the viral life cycle and pathogenesis (reviewedin [19]). Unfortunately, the crystal structures and functionaldomains for many of these proteins have not been describedin full detail. This lack of structural detail is a limitation tothe utility of structure-based drug design as an alternativemethod to rote library screening to find possible antiviralhits. Table 2 provides an overview of the filovirus proteinsand their functions.

4.1 NucleoproteinThe nucleoprotein (NP) is involved in nucleocapsid andinclusion body formation, encapsulation of RNA genomeand antigenome, and viral genome replication and transcrip-tion [68]. It is a central component of the ribonucleoproteincomplex (RNP) where it binds itself, another viral proteinVP35 at its interferon inhibitory domain, the secondarynucleoprotein known as VP30 and associates with L, the poly-merase [69]. The nucleoprotein gene is located upstream of allother genes in the filovirus RNA genome [70]. This gene sharesconserved sequence homology in central nucleotide sequenceregions with other members of the mononegavirales, para-myxoviruses and rhabdoviruses. There is variability in theC-terminal region, where there are multiple sites of phosphor-ylation that have been mapped for marburgvirus, variantMusoke (MARV-Mus) [19,65,71]. When phosphorylated byhost cell kinases, MARV-Mus NP can bind to itself, VP35,and VP30, and then incorporate into virions [19,72,73]. Studiesto map functional motifs and residues of importance toregions within the protein have been completed, and theregions responsible for NP--NP homodimerization, replica-tion and nucleocapsid-like structure formation have beenidentified [74,75]. Recent EBOV and MARV virus-like particle(VLP) and native virus studies using cryo-electron microscopyand tomography have described the nucleocapsid formationin detail, showing how NP, VP24, VP35, and VP40 arenecessary and sufficient to form a tight, structurally soundnucleocapsid [76,77]. At the time of this writing, neither thecrystal structure nor a description of any particular activesite of any filovirus NP was available. Continued investigationinto NP functions may reveal that there are specific sites thatmay be inhibited by specially designed small molecules.

4.2 VP30VP30 is thought to be a secondary nucleoprotein in filovi-ruses [78]. This protein colocalizes with NP in inclusion bodieswhen NP and VP30 are coexpressed in vitro [79]. VP30 andNP interact at VP30 C-terminal domain, through aminoacids (aa) 142-272, as shown through a crystal structure ofthe compact C-terminal domain [78]. VP30 oligomerizationis necessary for its function as a transcriptional activator butis not required for NP binding [80,81]. In vitro, the N-terminus

has been shown to bind single-stranded RNAs with a prefe-rence for filovirus RNAs [82,83]. The crystal structure of theC-terminal domain of VP30 demonstrates that this subunitfolds into a dimeric helical structure [78]. It is postulated thatVP30 dimers are the building blocks for the oligomerizationof VP30 into hexamers, and mutagenesis studies revealedseveral key interaction sites. The first site is Glu197, whichwas shown to be crucial for incorporation of VP30 into nucle-ocapsids. The second interaction site is a basic cluster ofthe amino acid residues Arg179, Lys180 and Lys183 inVP30 that are essential for nucleocapsid association and tran-scription activation [78]. Because this Lys180-centered, basiccluster is crucial for transcription activation, it may be feasiblethat a small molecule inhibitor that targets this site wouldinhibit transcription activation, and thus prove to be aneffective antiviral agent.

4.3 VP35Multifunctional VP35, a 35 kDa protein, is known to asso-ciate in RNP complexes and inclusion bodies, is likelyinvolved in replication, and functions as an innate immuneantagonist [69,84-86]. VP35 binds L protein to form the filo-viral replicase-transcriptase holoenzyme [85]. VP35 formshomodimers at aa 70 -- 120, and heterodimers withL protein at aa 70 -- 120 and 121 -- 219 [19,87].

One of the major functions of filovirus VP35 is to inhibitType-I interferon responses [84]. This protein is functionallyequivalent to HIV-1 Tat protein, which suppresses RNAi-based innate antiviral response of virus infected cells [88].Many reports describe that VP35 and VP35-NP complexesinhibit dsRNA and virus-mediated induction of interferonresponsive promoters leading to a general inhibition of thecellular immune response [19,84,89-91]. Crystal structures ofVP35 have been reported [69,92,93]. The structure of VP35 incomplex with an 8-base-pair dsRNA provides insights intothe functional role of VP35 in inhibition of Type-I interferonresponse in infection [69]. A small-molecule inhibitor couldpotentially be designed to block the binding of VP35to dsRNA targets, promoting downstream production ofinterferon. Mutagenesis studies showed that many aminoacid residues are crucial for the biological function ofVP35 [92]. The F239A, R312A, R322A, and K339A mutantswere shown to be unable to bind dsRNA and thus were alsounable to suppress IFN-b promoter activation relative towild-type VP35 [69]. Arg312, Arg322, and Lys339 form abasic patch on the VP35 protein surface. Presumably, asmall-molecule inhibitor that binds at this basic patch wouldprevent VP35 binding dsRNA and concomitantly allowexpression from the IFN-b promoter.

4.4 L proteinThe filoviral RNA-dependent RNA polymerase, the L protein,is the largest protein encoded in the 19kb genome. TheN-terminus of L protein binds to VP35 and together theymake up the replicase-transcriptase holoenzyme [94]. Bacterial

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 941

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

expression system production and gel electrophoresis puri-fication have been reported [82]. Detailed protein functionand interaction studies have not been reported in the publishedliterature. L protein may be responsible for polyadenylation. Lis a high-fidelity polymerase, with self-correcting features [95,96].L mRNAs are recognized by the host zinc finger antiviral pro-tein (ZAP). ZAP has been shown to decrease production ofinfectious EBOV and MARV in vitro [97]. The functionaldomains of L which are responsible for polyadenylation, self-correcting activities, and the interactions of ZAP with LmRNAs could all be further studied as candidate targets fortherapeutic intervention, and if structures become available,rational drug design methods could be applied. Designing ther-apeutics against L protein would require exacting protocols toensure that host polymerases are not also down-regulated.

4.5 VP40As the most abundant viral protein, VP40 is the viral matrixprotein and is encoded by the most conserved filovirusgene [12,65,96,98-100]. VP40 is the filovirus protein with themost functional data partly due to tractable expression andpurification from cellular systems [19]. The crystal structurehas been solved [98,101,102]. The crystal structure of aa44 -- 321 at 2 A resolution helped to describe the N-terminaldomain structure, and the C-terminal domain at aa201 -- 326 [98,101,102]. The hydrophobic interaction ofVP40 with cellular membranes is reminiscent of M (matrix)proteins from paramyxoviruses, measles, and respiratory syn-cytial virus (RSV) [103]. In fact, VP40 accumulates in areasof cellular membrane proliferation, where protein sorting forthe lysosome or the plasma membrane may occur [104].VP40 associates with cellular protrusions or filopodia in theabsence of any other viral proteins, and this membrane inter-action has been mapped to the C-terminus [100,105]. Theprotein alone will accumulate at membranes and causebudding of virus-like particles, which may be mediated bythe c-Abl1 tyrosine kinase [106,107]. Two compounds, nocoda-zole and paclitaxel, have been shown to reduce the release ofVP40-based VLPs into cell culture medium [106]. These twocompounds are microtubule depolymerization inhibitors.There is a region of sequence for VP40 from EBOV-Mayinga,which has homology to a tubulin binding motif found inmicrotubule-associated protein 2 (MAP2), and deletion ofthis region resulted in loss of binding of VP40 to the cellmembrane [108]. Thus, it appears that microtubule associationwith VP40 may occur and is important for egress of virionsfrom cells [106]. VP40 could not stabilize microtubulesin Niemann-Pick C1 (NPC-1)-deficient cells, which aredeficient in cholesterol transport properties, or cells treatedwith the cholesterol trafficking inhibitor U18666A [109].This suggests that cholesterol and/or its proper handlingand/or transport proteins may also be required for VP40function [109].Recently, a study provided insights into the structure and

function of filovirus VP40 [12]. This study demonstrated

that if the structure and function of filovirus proteins werebetter defined, then in silico drug design could provide insightinto directed therapeutics. The filovirus VP40 proteincontains one or more short amino acid sequences called theL-domain, or late budding domain, which serve to mediaterelease of virus particles. The EBOV L domains are PTAPand PPxY motifs, whereas the MARV VP40 L domain isa PPxY motif [110-113]. EBOV VP40 PTAP motif binds hostprotein Tsg101, and this has been visualized in live cellsas a mechanism for virus budding from host cells [12].A fluorescence-based technique called biomolecular comple-mentation was applied to VLP budding assays to prove thedirect interaction of Tsg101with EBOV VP40 at the PTAPmotif. This heterodimer may traffic to the plasma membranevia the microtubule network [12,108]. More importantly, theinvestigators screened in silico a 2.4 million compound libraryagainst the UEV domain of human Tsg101 which containsthe PTAP binding pocket, and identified potential inhibitorsof EBOV or MARV VP40 interaction with human Tsg101 atthe PTAP or PPxY binding pocket, for each virus, respec-tively [12]. Out of six inhibitors tested for ability to blockEBOV VP40 interactions with Tsg101, one small molecule,compound 5539-0062, reduced egress of EBOV VLPsin vitro by greater than 90% at 80 to 100 µM concentrations.In contrast, compound 5539-0062 only reduced MARV VLPegress by twofold, due to the lack of PTAP L-domain inMARV VP40 [12]. The compound was also found to directlyinhibit the fluorescence produced in the biomolecular com-plementation assay based on the interaction of Tsg101 andthe VP40 PTAP motif. Therefore, the small molecule wasshown to specifically inhibit VP40 at that site, as originallypredicted in silico. The molecule was tested against a livevesicular stomatitis virus, VSV-M40, modified to possessthe EBOV PTAP motif [114]. Compound 5539-0062 wasable to reduce growth of VSV-M40 by 3 -- 10 fold [12]. Thesestudies are promising, but results should be viewed with therecognition that compound 5539-0062 was not tested inassays with authentic viruses, and the most active concentra-tion was fairly high (above 80 µM). These studies demon-strate a proof of concept that if substantial structural dataexist, then in silico screening can be performed and effectiveantiviral hits can be obtained.

4.6 VP24Described as a secondary matrix protein, VP24 colocalizeswith VP40 in virions [65,73,115]. VP24 has not been shown tobind directly to VP40, the primary matrix protein [115].VP24 appears to be important for intracellular nucleocapsidassembly, and it may bind directly to NP [19,68,106,116]. TheVP24 from MARV-Mus may bind directly to GP2 at thecytoplasmic tail [19,117]. Changes to the NP and VP24 genesare responsible for most of the virulence associated withmouse adaptation of EBOV to cause lethal disease inmice [118]. VP24 may modulate the conversion of viralgenome from a form used in replication and transcription,

A. C. Shurtleff et al.

942 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

to a form ready to promote viral assembly and budding [119].VP24 appears to be involved in immunosuppression, throughinhibition of IFNa/b and IFNg signaling by sequesteringkaryopherin alpha proteins, and preventing movement ofphophorylated STAT1 to the nucleus [120-122]. A singleVP24 structure has recently been determined using x-ray crys-tallography and sheds light on this biological activity [123]. Itwas found that VP24 directly binds transcriptional activatorSTAT1, but how this binding exactly impacts function ofeither protein is unknown [123]. Structural differences inVP24 from RESTV and SUDV were observed [123]. Thesestructural differences led to postulation that perhaps RESTVVP24 may be less efficient at sequestering karyopherins andbinding STAT1, compared to the VP24 from the virulentSUDV species [123]. Now that these VP24 structures arereported, small molecules can be identified that bind to keyinteraction sites, and this may eventually lead to inhibitionof VP24 functions such as transcription, replication andimmunosuppression.

4.7 GlycoproteinThe glycoprotein genes of ebolavirus and marburgvirus differin their expression, but both function to produce the viral spikesurface glycoprotein that decorates the surface of virions, and isresponsible for virus entry into cells [67]. Only the ebolavirusglycoprotein gene encodes a frameshift reading site whichallows for a soluble glycoprotein (sGP) product to be madeand secreted from producing cells [67,124,125]. This sGP has notransmembrane domain or anchor. sGP released into thebloodstream may serve as an immune decoy during infec-tion [61]. The ebolavirus glycoprotein gene also encodes asecondary secreted glycoprotein of unknown function [126].The transmembrane spike GP1,2 is the full length transcript,and GPs differ by 37 -- 41 % nucleotide and 34 -- 43 % inamino acids across different ebolavirus species [67]. Ebolavirusand marburgvirus differ in glycoprotein sequence homologyby 55% at the nucleotide level and 67% at the amino acid level.The GP1,2 spike protein is a peplomer and a Type-Itransmembrane protein which is N- and O-glycosylated, phos-phorylated and post-translationally cleaved into two subunits,GP1 and GP2. The N-terminus of the glycoprotein precursorbecomes GP1 (about 160 -- 170 kDa) and theC-terminus becomes GP2 (about 38 -- 45 kDa). These twoproteins remain linked by a disulfide bond after host furincleavage [127]. N- and O-linked glycosylation accounts forabout one third of the molecular mass of the GP spike pro-teins [128]. GP1 is heavily glycosylated and responsible forattachment to host cells, but GP2 is responsible for fusion ofthe viral membrane with the host cell membrane, once thevirion has entered the lysosome [129-131].

GP1 and GP2 have functional domains which are reportedto have activities outside of binding and entry, and may beinvolved in pathogenesis. GP1 is reported to have about40 amino acid residues at the N-terminus which suppresslymphocyte blastogenesis in vitro, but the significance of this

activity in vivo has not yet been determined [132]. GP1 has amucin-like domain (MLD) that holds the glycans, and func-tions to prevent neutralizing antibodies from binding to theviral surface at GP1,2, and has been reported to have toxiceffects on cells [133-138]. GP2 is the transmembrane part ofthe spike glycoprotein and mainly functions as the fusion pep-tide, but an N-terminal immunosuppressive motif has alsobeen described [103,139,140]. This region may be cleaved andreleased to exert immunosuppressive activity [132,141].

A GP1,2 dependent model for filovirus entry has beendescribed through research involving studies of GP1,2 structureand through small molecule discovery [17,142-144]. External hostcell receptors have been evaluated and a variety of host cell fac-tors have been implicated in binding and entry: DC-SIGN/L-SIGN, hMGL, B-integrins, folate receptor A, and Tyrofamily receptors [145-149]. Once inside the endosomal/lysosomalcompartment, endosomal proteases cathepsins L and B arerequired for GP1 cleavage. This cleavage exposes the receptorbinding region and promotes fusion of viral membranes tocell membranes through GP2 fusion activities [142,143,147,150].Inside the lysosomal compartment, NPC-1 has been identifiedas the cellular factor required for entry of cells by GP2 throughthe cellular membrane [17,144]. Complete biological investi-gations such as these enable screening and mechanism ofentry studies like the ones presented below, and help directstructure-based drug discovery efforts.

5. Compound library screeningto identify viral entry inhibitors

A small molecule benzodiazepine derivative, named com-pound 7, was recently shown to inhibit filovirus entry [15].This molecule was discovered using a cell culture-basedhigh-throughput screen and a pseudotyped HIV virus express-ing EBOV-GP and containing a luciferase reporter gene(see Figure 1). Pseudotyped virus was screened against manydifferent compounds at a 25 µM concentration in a 96-wellplate format [59]. From a library of 52,000 compounds,18 compounds were found which had specific activity againstHIV/EBOV-GP, and no activity against HIV that had beenpseudotyped with VSV-G protein as a control for non-specific virus glycoprotein interaction [15]. These 18 hits hadgreater than 90% inhibitory activity against HIV/EBOV-GPand each compound was only cytotoxic to the indicatormonolayer at concentrations greater than 25 µM. Oncefound, these 18 compounds were tested against replication-competent EBOV expressing green fluorescent protein. Eightcompounds were identified with a 50% inhibitory concentra-tion (IC50) £ to 20 µM [15]. The cell cytotoxicity 50% level(CC50) was measured at > 50 µM for these compounds, there-fore the selectivity index (ratio of cytotoxicity to inhibitoryactivity) would be greater than 5. All eight molecules hadgood drug-like properties, but compound 7 demonstratedthe greatest potency, the most selective inhibition, andhad the most chemically “tractable” scaffold [64].

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 943

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

The analysis was taken even further by investigation ofcompound 7 with GP1 and GP2 using molecular modeling.The 3.4 A crystal structure of EBOV GP1 and GP2, whichwas solved in complex with a human neutralizing antibody(PDB code 3CSY), was selected as the template [151].Structural analysis of the GP1 and GP2 subunits revealed ahydrophobic pocket at their interface that could be thebinding site for compound 7. This site is demarked byVal66, Leu68, Asn69, Leu70, Leu184, and Leu186 fromGP1, and by Tyr517, Met548 and Leu558 from GP2(Figure 2). Subsequently, mutational studies of GP1 and GP2were performed in order to confirm the binding of compound7 to this hydrophobic site at the GP1-GP2 interface. Themutation work showed that even at concentrations as highas 50 µM, compound 7 exhibited little or no inhibition ofN69A and L70A mutants, and similarly, weakly inhibitedby £ 30% at a 25 µM concentration the L184A, I185A,L186A, K190A, and K191A mutants. Consistent with themolecular models, the results suggest that compound 7 bindsat or near amino acid residues Asn69, Leu70, Leu184, Ile185,Leu186, Lys190, and Lys191. Unfortunately, Basu et al. do

not present a binding model showing compound 7 in com-plex with GP1-GP2 in identified site. Instead, Basu et al.delineate the structure-activity relationship for compound 7.Twelve different analogues of compound 7 were assayed.The most potent of these analogues had an IC90 value of3.7 µM. Preliminary structure-activity relationship analysisshowed four sites (R1, R2, R3, and R4) that play a role inactivity (Figure 3). For R1 and R2, activity was increasedwhen the substituents were halogen atoms, and lost whenR1 and R2 were bulky benzo fusions. For R3, activity wasretained when the substituents were halogenated alkyl groupsand lost when the substituents were aromatic groups. For R4,small substituents at various positions can be tolerated. Forimproved structure-based drug design for anti-EBOV com-pounds, it is essential to determine the binding modes of theseactive benzodiazepine derivatives in the GP1-GP2 complex.This understanding would provide a structural basis for theiractivity and thus, become the platform for the design ofdrug candidates.

6. Mechanistic protein functional studiesreveal inhibitors with structural importance

An elegant body of work has recently shown that theNiemann-Pick C1 (NPC-1) protein is an essential host cellu-lar factor that viral GP1,2 binds for entry into cells at thelysosomal stage [17,144]. This discovery was made through ascreening effort investigating host cellular targets which couldimpede virus entry, as determined through the use of pseudo-typed VSV expressing the EBOV GP1,2 [17]. A benzylpipera-zine adamantine diamide derivative compound, named 3.0,inhibited growth of the VSV/EBOV-GP on vero cells over96 h postinfection. Adding a methoxycarbonyl benzyl groupat the ortho position increased the molecule’s potency by

Figure 2. Interface of the GP1 and GP2 subunits extracted from the 3CSY crystal structure [151] shows the binding site for

benzodiazepine derivatives identified by Basu et al. [15]. GP1 and GP2 are rendered in cyan and purple ribbon, respectively.

The surface of the hydrophobic binding site is shown in yellow.

N

HN

R2

R1

R3

R4

Figure 3. Preliminary structure-activity relationship of the

benzodiazepine-derivative chemotype determined by

Basuetal.showsfourpointsofdiversitythataffectactivity [15].

A. C. Shurtleff et al.

944 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

twofold (compound 3.47), resulting in decreased infectivityfor the VSV/EBOV-GP1,2. Endosomal protease cathepsin Bis required for EBOV GP1 cleavage, therefore the interactionof these new candidate molecules with cathepsin B was inves-tigated [142,143]. Through cellular activity studies and in vitrocathepsin activity assays, it was determined that compounds3.0 and 3.47 do not target cathepsin B protease [17]. NPC-1and cathepsin B do have distinct roles in EBOV entry witheach having a distinct timing for their activity [17,142,143].

These compounds induce formation of cytoplasmic vacuoleswhich are full of cholesterol, indicating that cholesterol uptakealong with its normal trafficking and regulation are hinderedby 3.0 and 3.47 [17]. Proteins known to regulate cholesterolaccumulation in late endosomes were evaluated for involve-ment and only NPC-1 was shown to be both essential toEBOV-GP infection and involved in cholesterol transport.Further, CHO cells lacking the NPC-1 protein could not beinfected with pseudotyped VSV/EBOV-GP, and experimentsexpressing recombinant mutants of NPC-1 on CHO cells indi-cated NPC-1 must be expressed, but need not be able to trans-port cholesterol for interaction with EBOV GP1,2 [17]. It wasalso shown that the GP1 component of EBOV-GP1,2 mustbe cleaved by the cathepsin B protease to bind to NPC-1,because engineered proteins that are made to the final cleavagephenotype would bind NPC-1 but those that were engineeredto be uncleavable did not bind the protein.

The NPC-1 protein is directly targeted by compounds3.0 and 3.47, as neither compound binds cleaved or wholeEBOV GP1,2. Instead, these compounds inhibit NPC-1 inter-action with the cleaved GP1 binding site. This was demon-strated through the use of a competitor compound designedto compete with 3.47 for NPC-1 binding. In addition,

NPC-1 over-expression conferred resistance to compounds3.0 and 3.47 [17]. Other small molecules, U18666A and imip-ramine, are known to cause a cellular phenotype similar tothat of NPC-1 deficiency, possibly because treatment withthese compounds inhibits late endosomal or lysosomal choles-terol transport [152,153]. U18666A and imipramine have beenshown to inhibit infection of vero cells and HAP1 cells byVSV/EBOV-GP1,2 when given at early stages of cellular infec-tion, but not when administered to cells late during infection.U18666A and imipramine may function as entry inhibitorsdue to interference with cholesterol transport, but they donot directly bind cleaved GP1 or NPC-1.

Given these data, NPC-1 is the host cellular factor to whichthe exposed N-terminal domain of GP1 binds, after theheavily glycosylated residues are cleaved off by endosomalcathepsin B protease. These studies, together with the struc-ture of the GP1,2 [151], have given us a clearer understandingof the activities of GP1 and GP2 in the lysosome and howNPC-1 may participate in enabling viral membrane fusionwith the cell membrane. Residues in the N-terminal domainof GP1 are interspersed with residues that make stabilizingcontacts with GP2. GP1 binding to NPC-1 may allow forGP2 to be freed up to change conformation and promotefusion of the viral membrane to the cellular membrane. Alter-natively, GP2 must also change conformation after GP1 bindsNPC-1, in order for the second step of GP2 to fuse. This exactmechanism is not yet solved.

NPC-1 is a large transmembrane protein that has beenproposed to be a proton-coupled transporter of sterols andother amphipathic molecules. The topology of NPC-1 isbuilt around 13 transmembrane helices and three majordomains [154,155]. The sterol sensing domain of NPC-1 exists

Figure 4. Crystal structure of the N-terminal domain of Niemann-Pick C1 (NPC1) in complex with cholesterol (PDB code

3GKI) [156]. NPC1 is shown in cyan ribbon and cholesterol in yellow CPK. The cholesterol binding site may also be the binding

site for compound 3.47 [17].

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 945

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

in the 5 transmembrane segment defined by helices 3-7. Thefirst domain is the N-terminal domain, which occurs onthe luminal side of the membrane and contains a Cys-rich leu-cine zipper motif as well as the sterol-binding site. The seconddomain is Loop C, which occurs on the luminal side and linkstransmembrane helices 2 and 3, and the third is Loop E whichlinks transmembrane 8 and 9, and is characterized by a Cys-rich ring finger motif. The concentration of Cys residues inthe N-terminal domain and Loop E makes NPC-1 susceptibleto suicide inhibitors. The structure of the N-terminal domainof NPC-1 in complex with cholesterol has been solved byx-ray crystallography (Figure 4) [156]. The 1.8 A structure ofNPC-1 (PDB code 3GKI) shows a deep, internal pocket forcholesterol binding. If the structure of NPC-1 is to be usedas a template for the structure-based design of more potentanalogues of compound 3.0 and 3.47, further biochemicalexperiments are necessary to determine the binding modesof these compounds in NPC-1.Furthermore, novel NPC-1 inhibitors may be gained from

the significant number of cholesterol-absorption inhibitors(CAI) that have been shown to target the closely relatedNiemann-Pick C1-Like 1 (NPC1L1) protein [157]. Whilehuman NPC-1 (uniprot code O15118) and human NPC1L1(uniprot code Q9UHC9) possess a sequence identity of36.9%, potential structural and functional similarity betweenNPC-1 and NPC1L1 may allow CAIs to also target NPC-1,providing a molecular basis for the development of anti-ebolavirus agents. The archetype among the CAI inhibitors isthe clinically approved drug ezetimibe (Zetia�) [158], which isan effective treatment for hypercholesterolemia. Biochemicaland proteomic studies delineated a mechanism of action forezetimibe, involving the binding of ezetimibe to the large extra-cellular loop C of NPC1L1, which prevents conformationalchanges in NPC1L1 that are essential for cholesterol transloca-tion across the membrane [159]. Potentially, as would ezetimibebind loop C in NPC1L1, compounds 3.0 and 3.47 bind tocorresponding loop C in NPC-1. The discovery of ezetimibewas further enriched by structure-activity relationship studiesof its azetidinone class of compound [160]. Moreover, thediscovery of ezetimibe opened the way for the developmentof other structural classes of NPC1L1 inhibitors, which arehighlighted by the recent disclosure of spiroimidazolidinoneNPC1L1 inhibitors [161,162].

7. Expert opinion

A key finding in this review is that there are many reportsdescribing solved filovirus protein structures (see Table 2). Inaddition, recent studies on host proteins such as NPC-1 andcAbl-1 highlight the crucial roles that host cellular factorsplay in the filovirus life cycle, making it possible that certainhost cellular factors are viable antiviral targets. Despite ourincreasing structural understanding of viral proteins VP30,VP35, VP40, VP24, and GP1,2, at present, a major weaknessin the field is that while some structures are available, many of

their functions and interactions remain to be fully elucidated.For example, it is known that NP, VP35, VP30, and Linteract to form the replication complex, but exactly howthese proteins interact and where might they physically bindone another is not yet known. If these interactions were betterunderstood, through a method such as bimolecular comple-mentation [16] compounds which could potentially block theassembly of the replication complex could be designed andtested in in vitro and in vivo models of filoviral replication.While there is a tremendous need for requisite biology studies,dissecting the biological process of viral proteins is slowed bythe rigors of a high biocontainment environment.

Continued screening of existing molecules is going toremain a critical activity in filovirus therapeutics discovery.To date, a variety of screening projects to look for new com-pounds with anti-filovirus activity, such as “compound 7” orto repurpose old ones (antidepressant imipramine or anti-malarial chloroquine) have been reported [15,144,163]. Screeningnot only provides lead compounds, but also may identifychemical probes that may be used to inform researchers aboutthe basic biology of filovirus infection [163]. As we continue toidentify ever increasing numbers of hits and lead compounds,and we are able to discern their potential mechanismsof action, we will be able to fully delineate the functions offiloviral proteins. These new functions may provide thebasis for rational drug design. As a classic example of this,Garcia M. et al. recently reported that VP40-mediated bud-ding of Ebola virus-like particles is mediated by phosphoryla-tion of Tyr13 on VP40 by the host cellular c-Abl1 tyrosinekinase [107]. In this work, viral release is decreased both byc-Abl1-specific small interfering RNA and by Abl-specifickinase inhibitors, imatinib (Gleevec) and nilotinib (Tasigna).Imatinib and nilotinib are approved drugs for the treatmentof chronic myeloid leukemia and are well known to specifi-cally target bcr-abl activity in leukemia. The ability of imati-nib and nilotinib to modulate the release of Ebola virus-likeparticles suggests that these two drugs may prove to be effica-cious short-term treatments against Ebola virus infection [107].Moreover, the anti-Ebola activity of two well-known anti-cancer drugs establishes a paradigm in which novel antiviraltherapeutics can be found in repurposed drugs and ininvestigational compounds.

In the coming years, research will continue to shed light onhow host proteins, such as cell-signaling molecules, stress-response proteins, or kinases and phosphatases, interact inthe virus life cycle. Small molecules with antioxidant proper-ties have shown anti-filovirus activity and may modulatemultiple host signaling pathways [164]. Other host proteins,such as Tsg101, Nedd4, actin, tubulin, some annnexins,heat-shock protein (HSP) A5, and ribosomal protein (RP)L18 have all been detected by proteomic analysis to be pack-aged inside EBOV and MARV virions [111,165-167]. siRNAstargeting HSPA5 and RP L18 in culture inhibited EBOVand MARV infection of 293T cells [167]. Continued drugscreening and viral biology studies will help to address

A. C. Shurtleff et al.

946 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

knowledge gaps about host protein interactions, hopefullyidentifying active sites of host or viral proteins through mech-anism of action studies. As these details become available,inhibition of host targets, or specific blocking of the site(s)of interaction of host targets with viral proteins may be thekey to unlocking good therapeutics for infection with highlyvirulent viruses. It will be important to specifically block thesesites of interaction, and not just inhibit the host protein acti-vity or its production, because such non-specific blocking orreduction of host proteins may lead to toxicity and unwantedside effects.

As an alternative to targeting catalytic sites, it is temptingto propose exploring the rational design of compoundsthat block essential protein-protein interactions (PPI), anapproach which has gotten some attention at recent biode-fense conferences as a method that could be employed whenlittle is known about target protein structure [168,169]. Consid-ering the limited number of known catalytic functions forfilovirus proteins, this may prove to be an avenue for potentialtherapeutics. The buried surfaces in PPIs are generally large(~ 1600 A) and it is thermodynamically improbable that arandom small molecule would disrupt the favorable energeticsenough to dissociate the interacting partners [170]. However,recent research has demonstrated that a small area of the inter-acting surface may actually account for the high-affinitybinding [171-173]. These compact regions of increased affinityhave been termed “hot-spots” and have been proposed to bea common motif for PPIs since these hot-spots are involvedin the binding of multiple partners for the same protein [171].Some recent work using bimolecular complementation tocharacterize the protein-protein interactions of VP40-VP40,NP-VP40, and NP-NP for EBOV and MARV has enhancedthe understanding of the role of these proteins in virus assem-bly and budding, and viral replication [12,16]. Using deletionmutant analysis, bimolecular complementation helpedpinpoint the protein sequences necessary for VP-40 selfinteractions and oligomerizations, and maybe soon somemolecules can be designed which mimic the structure andfunction of the interacting protein surfaces [16]. While there

are no specific examples of proteomimetics that have success-fully disrupted PPIs for filoviruses, examples of proteomimeticsas protease inhibitors do exist [169].

Comparatively speaking, data relating the structure andfunction of filovirus proteins is certainly less voluminousthan data available for protein constituents of HIV, cancersand other human pathogens of mainstream clinical impor-tance [3,6,7,9]. The substantial research funding that is allocatedfor cancer and AIDS has enabled scientists to generate thestructural and biochemical data needed to employ modernstructure-based drug design, and as a result, these fieldshave progressed much further than fields that are less wellfunded [174]. This limited funding combined with researchconstraints due to biosafety issues have restricted the availableapproaches for discovering lead candidates for therapeuticsand vaccines for biothreat agents [20,175]. Compounding theissue is the fact that it has been difficult to attract large phar-maceutical companies with state-of-the-art drug discoverytechniques to the field of biodefense due to the limited marketfor sale and distribution of their products [175]. The deve-lopment of therapeutics for biodefense or for use in lowersocio-economic countries with an unmet medical need offersa very limited return on investment compared to profitsfrom developing therapeutics for diseases of human clinicalimportance typically seen in the developed world [175,176].

The authors thank numerous colleagues and peers who arestriving to develop filovirus medical countermeasures andregret that space limitations may have prevented inclusion ofsome articles. Support for filovirus evaluations has been pro-vided by the Joint Science and Technologies Office/ChemBio Defense 00048_RD_T. The opinions, interpretations,conclusions and recommendations are those of the authorsand are not necessarily endorsed by the US Army.

Declaration of interest

This paper was sponsored by the Defense Threat ReductionAgency and Transformational Medical Technologies. Theauthors declare no other conflicts of interest.

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 947

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. De Clercq E. The design of drugs for

HIV and HCV. Nat Rev Drug Discov

2007;6:1001-18

2. Kubinyi H. Structure-based design of

enzyme inhibitors and receptor ligands.

Curr Opin Drug Discov Devel

1998;1:4-15

3. Kubinyi H. Chance favors the prepared

mind--from serendipity to rational drug

design. J Recept Signal Transduct Res

1999;19:15-39

4. Lu YH, Gao XQ, Wu M, et al.

Strategies on the development of small

molecule anticancer drugs for targeted

therapy. Mini Rev Med Chem

2011;11:611-24

5. Hochhaus A, Reiter A, Ernst T,

La Rosee P. Imatinib and

beyond--targeting activated tyrosine

kinases in myeloproliferative disorders.

Onkologie 2012;35(Suppl 1):34-41

6. Muller BA. Imatinib and its

successors--how modern chemistry has

changed drug development.

Curr Pharm Des 2009;15:120-33

7. von Itzstein M, Wu WY, Kok GB, et al.

Rational design of potent sialidase-based

inhibitors of influenza virus replication.

Nature 1993;363:418-23

8. von Itzstein M, Dyason JC, Oliver SW,

et al. A study of the active site of

influenza virus sialidase: an approach to

the rational design of novel anti-influenza

drugs. J Med Chem 1996;39:388-91

9. Wlodawer A, Vondrasek J. Inhibitors of

HIV-1 protease: a major success of

structure-assisted drug design. Annu Rev

Biophys Biomol Struct 1998;27:249-84

10. Louis JM, Ishima R, Torchia DA,

Weber IT. HIV-1 protease: structure,

dynamics, and inhibition. Adv Pharmacol

2007;55:261-98

11. Harty RN. No exit: targeting the

budding process to inhibit filovirus

replication. Antiviral Res 2009;81:189-97

12. Liu Y, Lee MS, Olson MA, Harty RN.

Bimolecular complementation to

visualize filovirus VP40-host

complexes in live mammalian cells:

toward the identification of budding

inhibitors. Adv Virol 2011;

2011:Article ID 341816. p. 10.. This paper describes one compound

identified using in silico drug design,

and is the first description of utilizing

this method for filoviruses.

13. Kolokoltsov AA, Saeed MF,

Freiberg AN, et al. Identification of

novel cellular targets for therapeutic

intervention against Ebola virus infection

by siRNA screening. Drug Dev Res

2009;70:255-65

14. Kolokoltsov AA, Adhikary S, Garver J,

et al. Inhibition of Lassa virus and Ebola

virus infection in host cells treated with

the kinase inhibitors genistein and

tyrphostin. Arch Virol 2012;157:121-7

15. Basu A, Li B, Mills DM, et al.

Identification of a small-molecule entry

inhibitor for filoviruses. J Virol

2011;85:3106-19.. A study exemplary of library screening

to find small molecule inhibitors of

EBOV. A molecular modeling analysis

predicted that the binding region for

this compound may be at GP1,2.

16. Liu Y, Stone S, Harty RN.

Characterization of filovirus

protein-protein interactions in

mammalian cells using bimolecular

complementation. J Infect Dis

2011;204(Suppl 3):S817-24

17. Cote M, Misasi J, Ren T, et al. Small

molecule inhibitors reveal Niemann-Pick

C1 is essential for Ebola virus infection.

Nature 2011;477:344-8.. This paper describes the NPC1 protein as

required for EBOV entry, and much of

the data were collected through detailed

mechanistic studies which elucidated

how the involved proteins function.

18. Warren TK, Shurtleff AC, Bavari S.

Advanced morpholino oligomers: A novel

approach to antiviral therapy.

Antiviral Res 2012;94(1):80-8

19. Kuhn JH. In: Calisher CH, editors.

Molecular Characteristics of Filoviruses:

a compendium of 40 years

of epidemiological, clinical, and

laboratory studies. SpringWien

New York; Vienna: 2008

20. CDC. Biosafety in Microbiological and

Biomedical Laboratories. 5th edition U.

S. Department of Health and Human

Services; Washington, D.C: 2009

21. Kuhn JH, Radoshitzky SR, Guth AC,

et al. Conserved receptor-binding

domains of Lake Victoria marburgvirus

and Zaire ebolavirus bind a common

receptor. J Biol Chem 2006;281:15951-8

22. Leroy EM, Gonzalez JP, Baize S. Ebola

and Marburg haemorrhagic fever viruses:

major scientific advances, but a relatively

minor public health threat for Africa.

Clin Microbiol Infect 2011;17:964-76

23. CDC. Outbreak Notice. Ebola

Outbreak in the District of

Bundibugyo; Uganda: 2008

24. CDC. Outbreak of Marburg virus

hemorrhagic fever--Angola, October 1,

2004-March 29, 2005. MMWR Morb

Mortal Wkly Rep 2005;54:308-9

25. Feldmann H, Jones SM, Schnittler HJ,

Geisbert T. Therapy and prophylaxis of

Ebola virus infections. Curr Opin

Investig Drugs 2005;6:823-30

26. Bazhutin NB, Belanov EF,

Spiridonov VA, et al. The effect of the

methods for producing an experimental

Marburg virus infection on the

characteristics of the course of the disease

in green monkeys. Vopr Virusol

1992;37:153-6

27. Leffel EK, Reed DS. Marburg and Ebola

viruses as aerosol threats.

Biosecur Bioterror 2004;2:186-91

28. Bente D, Gren J, Strong JE,

Feldmann H. Disease modeling for

Ebola and Marburg viruses.

Dis Model Mech 2009;2:12-17

29. Ebola haemorrhagic fever in Zaire,

1976. Bull World Health Organ

1978;56:271-93

30. Ebola haemorrhagic fever in Sudan,

1976. Report of a WHO/International

Study Team. Bull World Health Organ

1978;56:247-70

31. Bwaka MA, Bonnet MJ, Calain P, et al.

Ebola hemorrhagic fever in Kikwit,

Democratic Republic of the Congo:

clinical observations in 103 patients.

J Infect Dis 1999;179(Suppl 1):S1-7

32. Formenty P, Hatz C, Le Guenno B,

et al. Human infection due to Ebola

virus, subtype Cote d’Ivoire: clinical and

biologic presentation. J Infect Dis

1999;179(Suppl 1):S48-53

33. Outbreak of Ebola hemorrhagic fever

Uganda, August 2000-January 2001.

A. C. Shurtleff et al.

948 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

MMWR Morb Mortal Wkly Rep

2001;50:73-7

34. Shurtleff AC, Warren T, Bavari S.

Nonhuman primates as models for the

discovery and development of ebolavirus

therapeutics. Expert Opin Drug Discov

2011;6:1-18. A review thoroughly describing the

pathogenesis of ebolavirus infection of

NHP and the use of this model in the

FDA Animal Rule of Efficacy.

35. Warfield KL, Aman MJ. Advances in

virus-like particle vaccines for filoviruses.

J Infect Dis 2011;204(Suppl 3):S1053-9

36. Bray M, Davis K, Geisbert T, et al.

A mouse model for evaluation of

prophylaxis and therapy of Ebola

hemorrhagic fever. J Infect Dis

1999;179(Suppl 1):S248-58

37. Connolly BM, Steele KE, Davis KJ,

et al. Pathogenesis of experimental Ebola

virus infection in guinea pigs.

J Infect Dis 1999;179(Suppl 1):S203-17

38. Warfield KL, Bradfute SB, Wells J, et al.

Development and characterization of a

mouse model for Marburg hemorrhagic

fever. J Virol 2009;83:6404-15

39. Lofts LL, Ibrahim MS, Negley DL, et al.

Genomic differences between guinea

pig lethal and nonlethal Marburg

virus variants. J Infect Dis

2007;196(Suppl 2):S305-12

40. Enterlein S, Warfield KL, Swenson DL,

et al. VP35 knockdown inhibits Ebola

virus amplification and protects against

lethal infection in mice. Antimicrob

Agents Chemother 2006;50:984-93

41. Aman MJ, Kinch MS, Warfield K, et al.

Development of a broad-spectrum

antiviral with activity against Ebola

virus. Antiviral Res 2009;83:245-51

42. Warren TK, Warfield KL, Wells J, et al.

Advanced antisense therapies for

postexposure protection against

lethal filovirus infections.

Nat Med 2010;16:991-4

43. Geisbert TW, Lee AC, Robbins M, et al.

Postexposure protection of non-human

primates against a lethal Ebola virus

challenge with RNA interference:

a proof-of-concept study. Lancet

2010;375:1896-905

44. Snoy PJ. Establishing efficacy of human

products using animals: the US food

and drug administration’s “animal rule”.

Vet Pathol 2010;47:774-8

45. Draft Guidance for Industry. Essential

elements for address efficacy under the

animal rule. In: US Depart of Health

and Human Services FDA. Volumne 74

Fed Regist; 2009. p. 3610-11. This is the FDA draft guidance

document for studies to be executed

under the Animal Efficacy Rule.

46. Fisher-Hoch SP, Platt GS, Lloyd G,

et al. Haematological and biochemical

monitoring of Ebola infection in rhesus

monkeys: implications for patient

management. Lancet 1983;2:1055-8

47. Gonchar NI, Pshenichnov VA,

Pokhodiaev VA, et al. The sensitivity of

different experimental animals to the

Marburg virus. Vopr Virusol

1991;36:435-7

48. Ryabchikova EI, Kolesnikova LV,

Luchko SV. An analysis of features of

pathogenesis in two animal models of

Ebola virus infection. J Infect Dis

1999;179(Suppl 1):S199-202

49. Kortepeter MG, Bausch DG, Bray M.

Basic clinical and laboratory features of

filoviral hemorrhagic fever. University of

Chicago, Chicago, IL, USA. J Infect Dis

2011;204(Suppl 3):S810-16. A thorough compilation and review of

all available human clinical data from

filovirus infections.

50. Alves DA, Glynn AR, Steele KE, et al.

Aerosol exposure to the angola strain of

marburg virus causes lethal viral

hemorrhagic Fever in cynomolgus

macaques. Vet Pathol 2010;47:831-51

51. Baskerville A, Bowen ET, Platt GS, et al.

The pathology of experimental Ebola

virus infection in monkeys. J Pathol

1978;125:131-8

52. Bowen ET, Platt GS, Simpson DI, et al.

Ebola haemorrhagic fever: experimental

infection of monkeys. Trans R Soc Trop

Med Hyg 1978;72:188-91

53. Geisbert TW, Hensley LE, Larsen T,

et al. Pathogenesis of Ebola hemorrhagic

fever in cynomolgus macaques: evidence

that dendritic cells are early and

sustained targets of infection.

Am J Pathol 2003;163:2347-70

54. Geisbert TW, Daddario-DiCaprio KM,

Geisbert JB, et al. Marburg virus Angola

infection of rhesus macaques:

pathogenesis and treatment with

recombinant nematode anticoagulant

protein c2. J Infect Dis

2007;196(Suppl 2):S372-81

55. Hensley LE, Alves DA, Geisbert JB,

et al. Pathogenesis of marburg

hemorrhagic Fever in cynomolgus

macaques. J Infect Dis

2011;204(Suppl 3):S1021-31

56. Reed DS, Hensley LE, Geisbert JB, et al.

Depletion of peripheral blood T

lymphocytes and NK cells during

the course of ebola hemorrhagic

Fever in cynomolgus macaques.

Viral Immunol 2004;17:390-400

57. Johnson E, Jaax N, White J, Jahrling P.

Lethal experimental infections of rhesus

monkeys by aerosolized Ebola virus. Int J

Exp Pathol 1995;76:227-36

58. Warren TK, Warfield KL, Wells J, et al.

Antiviral activity of a small-molecule

inhibitor of filovirus infection.

Antimicrob Agents Chemother

2010;54:2152-9

59. Basu A, Mills DM, Bowlin TL.

High-throughput screening of viral

entry inhibitors using pseudotyped

virus. Curr Protoc Pharmacol

2011;Chapter 13:Unit 13B 3.. This work is a protocol chapter

describing use of pseudotyped viruses

in high throughput screens for viral

entry inhibitors.

60. Takada A, Robison C, Goto H, et al.

A system for functional analysis of Ebola

virus glycoprotein. Proc Natl Acad

Sci USA 1997;94:14764-9

61. Ito H, Watanabe S, Takada A,

Kawaoka Y. Ebola virus glycoprotein:

proteolytic processing, acylation, cell

tropism, and detection of neutralizing

antibodies. J Virol 2001;75:1576-80

62. Yermolina MV, Wang J, Caffrey M,

et al. discovery, synthesis, and biological

evaluation of a novel group of selective

inhibitors of filoviral entry. J Med Chem

2011;54:765-81

63. Panchal RG, Kota KP, Spurgers KB,

et al. Development of high-content

imaging assays for lethal viral pathogens.

J Biomol Screen 2010;15:755-65

64. Lipinski CA, Lombardo F, Dominy BW,

Feeney PJ. Experimental and

computational approaches to estimate

solubility and permeability in drug

discovery and development settings.

Adv Drug Deliv Rev 2001;46:3-26. This paper describes approaches to

estimate whether molecules may have

desirable chemical characteristics, such

as solubility and permeability, that

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 949

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

would would allow for successful drug

development.

65. Elliott LH, Kiley MP, McCormick JB.

Descriptive analysis of Ebola virus

proteins. Virology 1985;147:169-76. An early description of ebolavirus

proteins and their putative functions.

66. Elliott LH, Sanchez A, Holloway BP,

et al. Ebola protein analyses for the

determination of genetic organization.

Arch Virol 1993;133:423-36

67. Sanchez A, Trappier SG, Mahy BW,

et al. The virion glycoproteins of Ebola

viruses are encoded in two reading

frames and are expressed through

transcriptional editing. Proc Natl Acad

Sci USA 1996;93:3602-7

68. 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. Mol Cell 2002;10:307-16

69. Leung DW, Prins KC, Basler CF,

Amarasinghe GK. Ebolavirus VP35 is a

multifunctional virulence factor.

Virulence 2010;1:526-31.. A review describing much of the work

to date on VP35 crystal structure

analysis and the multiple functions

of VP35.

70. Kiley MP, Wilusz J, McCormick JB,

Keene JD. Conservation of the 3’

terminal nucleotide sequences of Ebola

and Marburg virus. Virology

1986;149:251-4

71. Barr J, Chambers P, Pringle CR,

Easton AJ. Sequence of the major

nucleocapsid protein gene of pneumonia

virus of mice: sequence comparisons

suggest structural homology between

nucleocapsid proteins of pneumoviruses,

paramyxoviruses, rhabdoviruses

and filoviruses. J Gen Virol

1991;72(Pt 3):677-85

72. Becker S, Huppertz S, Klenk HD,

Feldmann H. The nucleoprotein of

Marburg virus is phosphorylated.

J Gen Virol 1994;75(Pt 4):809-18

73. Becker S, Rinne C, Hofsass U, et al.

Interactions of Marburg virus

nucleocapsid proteins. Virology

1998;249:406-17

74. Watanabe S, Noda T, Kawaoka Y.

Functional mapping of the nucleoprotein

of Ebola virus. J Virol 2006;80:3743-51

75. Licata JM, Johnson RF, Han Z,

Harty RN. Contribution of ebola virus

glycoprotein, nucleoprotein, and VP24 to

budding of VP40 virus-like particles.

J Virol 2004;78:7344-51

76. Bharat TA, Noda T, Riches JD, et al.

Structural dissection of Ebola virus and

its assembly determinants using

cryo-electron tomography. Proc Natl

Acad Sci USA 2012;109(11):4275-80

77. Bharat TA, Riches JD, Kolesnikova L,

et al. Cryo-electron tomography of

Marburg virus particles and their

morphogenesis within infected cells.

PLoS Biol 2011;9:e1001196

78. Hartlieb B, Muziol T, Weissenhorn W,

Becker S. Crystal structure of the

C-terminal domain of Ebola virus

VP30 reveals a role in transcription and

nucleocapsid association. Proc Natl Acad

Sci USA 2007;104:624-9. This work describes the crystal

structure of VP30 and its roles in

transcriptional activation and

nucleocapsid assembly.

79. Modrof J, Muhlberger E, Klenk HD,

Becker S. Phosphorylation of

VP30 impairs ebola virus transcription.

J Biol Chem 2002;277:33099-104

80. Hartlieb B, Becker S. Characterization of

Ebola virus VP30 oligomerization.

Abstracts of the Annual Meeting of the

‘‘Gesellschaft feur Virologie [German

Society of Virology]’’- Joint Meeting

with the ‘‘Societa´ Italiana di Virologia

[Italian Society of Virology]’’; Eberhard-

Karls-Universiteat, Teubingen,Baden-Weurttemberg, Germany; 2004

81. Modrof J, Becker S, Muhlberger E.

Ebola virus transcription activator

VP30 is a zinc-binding protein. J Virol

2003;77:3334-8

82. John SP, Steffen S, Schmaljohn C,

Jonsson CB. In vitro expression,

purification and functional analysis

of ebola virus VP30, VP35 and

L proteins [abstract P25-1]. American

Society for Virology 23rd Annual

Meeting; McGill University, Montreal,

Quebec, Canada; 2004

83. John SP, Wang T, Steffen S, et al. Ebola

virus VP30 is an RNA binding protein.

J Virol 2007;81:8967-76

84. Basler CF, Wang X, Muhlberger E, et al.

The Ebola virus VP35 protein functions

as a type I IFN antagonist. Proc Natl

Acad Sci USA 2000;97:12289-94

85. 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. J Virol 1998;72:8756-64

86. Noda T, Aoyama K, Sagara H, et al.

Nucleocapsid-like structures of Ebola

virus reconstructed using electron

tomography. J Vet Med Sci

2005;67:325-8

87. Muhlberger E, Weik M, Volchkov VE,

et al. Comparison of the transcription

and replication strategies of marburg

virus and Ebola virus by using artificial

replication systems. J Virol

1999;73:2333-42

88. Haasnoot J, de Vries W, Geutjes EJ,

et al. The Ebola virus VP35 protein is a

suppressor of RNA silencing.

PLoS Pathog 2007;3:e86

89. Cardenas WB, Loo YM, Gale M Jr,

et al. Ebola virus VP35 protein

binds double-stranded RNA and

inhibits alpha/beta interferon production

induced by RIG-I signaling. J Virol

2006;80:5168-78

90. Feng Z, Cerveny M, Yan Z, He B. The

VP35 protein of Ebola virus inhibits the

antiviral effect mediated by

double-stranded RNA-dependent protein

kinase PKR. J Virol 2007;81:182-92

91. Hartman AL, Dover JE, Towner JS,

Nichol ST. Reverse genetic generation of

recombinant Zaire Ebola viruses

containing disrupted IRF-3 inhibitory

domains results in attenuated virus

growth in vitro and higher levels of

IRF-3 activation without inhibiting viral

transcription or replication. J Virol

2006;80:6430-40

92. Leung DW, Ginder ND, Fulton DB,

et al. Structure of the Ebola

VP35 interferon inhibitory domain.

Proc Natl Acad Sci USA

2009;106:411-16

93. Leung DW, Ginder ND, Nix JC, et al.

Expression, purification, crystallization

and preliminary X-ray studies of the

Ebola VP35 interferon inhibitory

domain. Acta Crystallogr Sect F Struct

Biol Cryst Commun 2009;65:163-5

94. Boehmann Y, Enterlein S, Randolf A,

Muhlberger E. A reconstituted

replication and transcription system for

Ebola virus Reston and comparison with

Ebola virus Zaire. Virology

2005;332:406-17

A. C. Shurtleff et al.

950 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

95. Cox NJ, McCormick JB, Johnson KM,

Kiley MP. Evidence for two subtypes of

Ebola virus based on oligonucleotide

mapping of RNA. J Infect Dis

1983;147:272-5

96. Towner JS, Khristova ML, Sealy TK,

et al. Marburgvirus genomics and

association with a large hemorrhagic fever

outbreak in Angola. J Virol

2006;80:6497-516

97. Muller S, Moller P, Bick MJ, et al.

Inhibition of filovirus replication by the

zinc finger antiviral protein. J Virol

2007;81:2391-400

98. Dessen A, Volchkov V, Dolnik O, et al.

Crystal structure of the matrix protein

VP40 from Ebola virus. EMBO J

2000;19:4228-36.. Together with references 99-101, this

set of papers describes the crystal

structure and membrane binding

properties of VP40.

99. Gomis-Ruth FX, Dessen A, Timmins J,

et al. The matrix protein VP40 from

Ebola virus octamerizes into pore-like

structures with specific RNA binding

properties. Structure 2003;11:423-33

100. Ruigrok RW, Schoehn G, Dessen A,

et al. Structural characterization and

membrane binding properties of the

matrix protein VP40 of Ebola virus.

J Mol Biol 2000;300:103-12

101. Dessen A, Forest E, Volchkov V, et al.

Crystallization and preliminary X-ray

analysis of the matrix protein from Ebola

virus. Acta Crystallogr D Biol Crystallogr

2000;56:758-60

102. Weissenhorn W. Structure of Viral

Proteins. In: Klenk H, Feldmann H,

editors. EBOLA and MARBURG

VIRUSES - Molecular and

Cellular Biology. Horizon Bioscience,

Wymondham, Norfolk, United

Kingdom; 2004;p. 27-57

103. Bukreyev AA, Volchkov VE, Blinov VM,

Netesov SV. The VP35 and

VP40 proteins of filoviruses. Homology

between Marburg and Ebola viruses.

FEBS Lett 1993;322:41-6

104. Kolesnikova L, Bugany H, Klenk HD,

Becker S. VP40, the matrix protein of

Marburg virus, is associated with

membranes of the late endosomal

compartment. J Virol 2002;76:1825-38

105. Jasenosky LD, Neumann G,

Lukashevich I, Kawaoka Y. Ebola virus

VP40-induced particle formation and

association with the lipid bilayer. J Virol

2001;75:5205-14

106. Noda T, Ebihara H, Muramoto Y, et al.

Assembly and budding of Ebolavirus.

PLoS Pathog 2006;2:e99

107. Garcia M, Cooper A, Shi W, et al.

Productive Replication of Ebola Virus Is

Regulated by the c-Abl1 Tyrosine Kinase.

Sci Transl Med 2012;4:123ra24

108. Ruthel G, Demmin GL, Kallstrom G,

et al. Association of ebola virus matrix

protein VP40 with microtubules. J Virol

2005;79:4709-19

109. Badie S, Kallstrom G, Lessick B, et al.

Abstract: Study of Ebola Virus and VLP

Release in NPC1 Cells Deficient in

Cholesterol Trafficking. ASM [American

Society for Microbiology] Biodefense

Research Meeting; Baltimore, Maryland,

USA; 2005

110. Harty RN, Brown ME, Wang G, et al.

A PPxY motif within the VP40 protein

of Ebola virus interacts physically and

functionally with a ubiquitin ligase:

implications for filovirus budding.

Proc Natl Acad Sci USA

2000;97:13871-6

111. Licata JM, Simpson-Holley M,

Wright NT, et al. Overlapping motifs

(PTAP and PPEY) within the Ebola virus

VP40 protein function independently as

late budding domains: involvement of

host proteins TSG101 and VPS-4.

J Virol 2003;77:1812-19

112. Urata S, Noda T, Kawaoka Y, et al.

Interaction of Tsg101 with Marburg

virus VP40 depends on the PPPY motif,

but not the PT/SAP motif as in the case

of Ebola virus, and Tsg101 plays a

critical role in the budding of Marburg

virus-like particles induced by VP40, NP,

and GP. J Virol 2007;81:4895-9

113. Dolnik O, Kolesnikova L, Stevermann L,

Becker S. Tsg101 is recruited by a late

domain of the nucleocapsid protein to

support budding of Marburg virus-like

particles. J Virol 2010;84:7847-56

114. Irie T, Licata JM, Harty RN. Functional

characterization of Ebola virus L-domains

using VSV recombinants. Virology

2005;336:291-8

115. Bamberg S, Kolesnikova L, Moller P,

et al. VP24 of Marburg virus influences

formation of infectious particles. J Virol

2005;79:13421-33

116. Noda T, Sagara H, Suzuki E, et al.

Ebola virus VP40 drives the formation of

virus-like filamentous particles along with

GP. J Virol 2002;76:4855-65

117. Bukreyev AA, Belanov EF, Blinov VM,

Netesov SV. Complete nucleotide

sequences of Marburg virus genes 5 and

6 encoding VP30 and VP24 proteins.

Biochem Mol Biol Int 1995;35:605-13

118. Ebihara H, Takada A, Kobasa D, et al.

Molecular determinants of Ebola virus

virulence in mice. PLoS Pathog

2006;2:e73

119. Han Z, Boshra H, Sunyer JO, et al.

Biochemical and functional

characterization of the Ebola virus

VP24 protein: implications for a role in

virus assembly and budding. J Virol

2003;77:1793-800

120. Mateo M, Reid SP, Leung LW, et al.

Ebolavirus VP24 binding to karyopherins

is required for inhibition of interferon

signaling. J Virol 2010;84:1169-75

121. Reid SP, Leung LW, Hartman AL, et al.

Ebola virus VP24 binds karyopherin

alpha1 and blocks STAT1 nuclear

accumulation. J Virol 2006;80:5156-67

122. Reid SP, Valmas C, Martinez O, et al.

Ebola virus VP24 proteins inhibit the

interaction of NPI-1 subfamily

karyopherin alpha proteins with activated

STAT1. J Virol 2007;81:13469-77

123. Zhang AP, Bornholdt ZA, Liu T, et al.

The Ebola Virus Interferon Antagonist

VP24 Directly Binds STAT1 and Has a

Novel, Pyramidal Fold. PLoS Pathog

2012;8:e1002550.. This paper compares VP24 structural

differences from pathogenic and

non-pathogenic ebolaviruses.

124. Volchkov VE, Becker S, Volchkova VA,

et al. GP mRNA of Ebola virus is edited

by the Ebola virus polymerase and by

T7 and vaccinia virus polymerases.

Virology 1995;214:421-30

125. Volchkova VA, Feldmann H, Klenk HD,

Volchkov VE. The nonstructural small

glycoprotein sGP of Ebola virus is

secreted as an antiparallel-orientated

homodimer. Virology 1998;250:408-14

126. Mehedi M, Falzarano D, Seebach J,

et al. A new Ebola virus nonstructural

glycoprotein expressed through

RNA editing. J Virol 2011;85:5406-14

127. Volchkov VE, Feldmann H,

Volchkova VA, Klenk HD. Processing of

the Ebola virus glycoprotein by the

proprotein convertase furin. Proc Natl

Acad Sci USA 1998;95:5762-7

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 951

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

128. Feldmann H, Nichol ST, Klenk HD,

et al. Characterization of filoviruses based

on differences in structure and

antigenicity of the virion glycoprotein.

Virology 1994;199:469-73

129. Saez-Cirion A, Gomara MJ, Agirre A,

Nieva JL. Pre-transmembrane sequence

of Ebola glycoprotein. Interfacial

hydrophobicity distribution and

interaction with membranes. FEBS Lett

2003;533:47-53

130. Adam B, Lins L, Stroobant V, et al.

Distribution of hydrophobic residues is

crucial for the fusogenic properties of the

Ebola virus GP2 fusion peptide. J Virol

2004;78:2131-6

131. Ruiz-Arguello MB, Goni FM,

Pereira FB, Nieva JL.

Phosphatidylinositol-dependent

membrane fusion induced by a putative

fusogenic sequence of Ebola virus.

J Virol 1998;72:1775-81.. This paper elucidated the structure of

EBOV GP when bound to a

neutralizing antibody from a

human survivor.

132. Chepurnov AA, Tuzova MN,

Ternovoy VA, Chernukhin IV.

Suppressive effect of Ebola virus on T

cell proliferation in vitro is provided by a

125-kDa GP viral protein. Immunol Lett

1999;68:257-61

133. Dowling W, Thompson E, Badger C,

et al. Influences of glycosylation on

antigenicity, immunogenicity, and

protective efficacy of ebola virus GP

DNA vaccines. J Virol 2007;81:1821-37

134. Yang ZY, Duckers HJ, Sullivan NJ, et al.

Identification of the Ebola virus

glycoprotein as the main viral

determinant of vascular cell cytotoxicity

and injury. Nat Med 2000;6:886-9

135. Chan SY, Ma MC, Goldsmith MA.

Differential induction of cellular

detachment by envelope glycoproteins of

Marburg and Ebola (Zaire) viruses.

J Gen Virol 2000;81:2155-9

136. Ray RB, Basu A, Steele R, et al. Ebola

virus glycoprotein-mediated anoikis of

primary human cardiac microvascular

endothelial cells. Virology

2004;321:181-8

137. Simmons G, Wool-Lewis RJ,

Baribaud F, et al. Ebola virus

glycoproteins induce global surface

protein down-modulation and loss of cell

adherence. J Virol 2002;76:2518-28

138. Volchkov VE, Volchkova VA,

Muhlberger E, et al. Recovery of

infectious Ebola virus from

complementary DNA: RNA editing of

the GP gene and viral cytotoxicity.

Science 2001;291:1965-9

139. Volchkov VE, Blinov VM, Netesov SV.

The envelope glycoprotein of Ebola virus

contains an immunosuppressive-like

domain similar to oncogenic retroviruses.

FEBS Lett 1992;305:181-4

140. Yaddanapudi K, Palacios G, Towner JS,

et al. Implication of a retrovirus-like

glycoprotein peptide in the

immunopathogenesis of Ebola and

Marburg viruses. FASEB J

2006;20:2519-30

141. Ignatyev GM, Volchkov VE, Blinov VM,

et al. Phenomenon of

immunosuppression caused by filoviruses.

Int J Immunorehabilitation

1994;1:Suppl Moscow

142. Schornberg K, Matsuyama S, Kabsch K,

et al. Role of endosomal cathepsins in

entry mediated by the Ebola virus

glycoprotein. J Virol 2006;80:4174-8

143. Chandran K, Sullivan NJ, Felbor U,

et al. Endosomal proteolysis of the Ebola

virus glycoprotein is necessary for

infection. Science 2005;308:1643-5

144. Carette JE, Raaben M, Wong AC, et al.

Ebola virus entry requires the cholesterol

transporter Niemann-Pick C1. Nature

2011;477:340-3

145. Shimojima M, Takada A, Ebihara H,

et al. Tyro3 family-mediated cell entry of

Ebola and Marburg viruses. J Virol

2006;80:10109-16

146. Chan SY, Speck RF, Ma MC,

Goldsmith MA. Distinct mechanisms of

entry by envelope glycoproteins of

Marburg and Ebola (Zaire) viruses.

J Virol 2000;74:4933-7

147. Kaletsky RL, Simmons G, Bates P.

Proteolysis of the Ebola virus

glycoproteins enhances virus binding and

infectivity. J Virol 2007;81:13378-84

148. Simmons G, Reeves JD, Grogan CC,

et al. DC-SIGN and DC-SIGNR bind

ebola glycoproteins and enhance

infection of macrophages and endothelial

cells. Virology 2003;305:115-23

149. Simmons G, Rennekamp AJ, Chai N,

et al. Folate receptor alpha and caveolae

are not required for Ebola virus

glycoprotein-mediated viral infection.

J Virol 2003;77:13433-8

150. Sanchez A. Analysis of filovirus entry

into vero e6 cells, using inhibitors of

endocytosis, endosomal acidification,

structural integrity, and cathepsin

(B and L) activity. J Infect Dis

2007;196(Suppl 2):S251-8

151. Lee JE, Fusco ML, Hessell AJ, et al.

Structure of the Ebola virus glycoprotein

bound to an antibody from a human

survivor. Nature 2008;454:177-82

152. Cenedella RJ. Cholesterol synthesis

inhibitor U18666A and the role of sterol

metabolism and trafficking in numerous

pathophysiological processes. Lipids

2009;44:477-87

153. Rodriguez-Lafrasse C, Rousson R,

Bonnet J, et al. Abnormal cholesterol

metabolism in imipramine-treated

fibroblast cultures. Similarities with

Niemann-Pick type C disease.

Biochim Biophys Acta

1990;1043:123-8

154. Davies JP, Ioannou YA. Topological

analysis of Niemann-Pick C1 protein

reveals that the membrane orientation

of the putative sterol-sensing domain

is identical to those of 3-hydroxy-3-

methylglutaryl-CoA reductase and sterol

regulatory element binding protein

cleavage-activating protein. J Biol Chem

2000;275:24367-74

155. King G, Sharom FJ. Proteins that bind

and move lipids: msbA and NPC1.

Crit Rev Biochem Mol Biol

2012;47:75-95

156. Kwon HJ, Abi-Mosleh L, Wang ML,

et al. Structure of N-terminal domain of

NPC1 reveals distinct subdomains for

binding and transfer of cholesterol. Cell

2009;137:1213-24

157. Chhabria MT, Mahajan BM. Update on

patented cholesterol absorption

inhibitors. Expert opin ther patents

2009;19:1083-107

158. Clader JW. The discovery of ezetimibe:

a view from outside the receptor.

J Med Chem 2004;47:1-9

159. Weinglass AB, Kohler M, Schulte U,

et al. Extracellular loop C of NPC1L1 is

important for binding to ezetimibe.

Proc Natl Acad Sci USA

2008;105:11140-5

160. Clader JW. Ezetimibe and other

azetidinone cholesterol absorption

inhibitors. Curr Top Med Chem

2005;5:243-56

A. C. Shurtleff et al.

952 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

161. Howell KL, DeVita RJ, Garcia-Calvo M,

et al. Spiroimidazolidinone

NPC1L1 inhibitors. Part 2:

structure-activity studies and in vivo

efficacy. Bioorg Med Chem Lett

2010;20:6929-32

162. McMasters DR, Garcia-Calvo M,

Maiorov V, et al. Spiroimidazolidinone

NPC1L1 inhibitors. 1: discovery by

3D-similarity-based virtual screening.

Bioorg Med Chem Lett 2009;19:2965-8

163. Wool-Lewis RJ, Bates P. Characterization

of Ebola virus entry by using

pseudotyped viruses: identification of

receptor-deficient cell lines. J Virol

1998;72:3155-60

164. Panchal RG, Reid SP, Tran JP, et al.

Identification of an antioxidant

small-molecule with broad-spectrum

antiviral activity. Antiviral Res

2011;93:23-9

165. Yasuda J, Nakao M, Kawaoka Y,

Shida H. Nedd4 regulates egress of Ebola

virus-like particles from host cells. J Virol

2003;77:9987-92

166. Han Z, Harty RN. Packaging of actin

into Ebola virus VLPs. Virol J 2005;2:92

167. Spurgers KB, Alefantis T, Peyser BD,

et al. Identification of essential

filovirion-associated host factors by serial

proteomic analysis and RNAi screen.

Mol Cell Proteomics 2010;9:2690-703

168. Fletcher S, Hamilton AD. Protein surface

recognition and proteomimetics: mimics

of protein surface structure and function.

Curr Opin Chem Biol 2005;9:632-8

169. Yin H, Hamilton AD. Strategies for

targeting protein-protein interactions

with synthetic agents. Angew Chem Int

Ed Engl 2005;44:4130-63

170. Lo Conte L, Chothia C, Janin J. The

atomic structure of protein-protein

recognition sites. J Mol Biol

1999;285:2177-98

171. Arkin MR, Wells JA. Small-molecule

inhibitors of protein-protein interactions:

progressing towards the dream. Nat Rev

Drug Discov 2004;3:301-17

172. Clackson T, Wells JA. A hot spot of

binding energy in a hormone-receptor

interface. Science 1995;267:383-6

173. DeLano WL. Unraveling hot spots in

binding interfaces: progress and

challenges. Curr Opin Struct Biol

2002;12:14-20

174. National Institutes of Health OoER.

NIH Research Portfolio Online

Reporting Tools (RePORT). Estimates of

Funding for Various Research, Condition

and Disease Categories: U.S. Department

of Health and Human Services, 2012

175. Greenstone H, Spinelli B, Tseng C, et al.

NIAID resources for developing new

therapies for severe viral infections.

Antiviral Res 2008;78:51-9

176. Bolken TC, Hruby DE. Discovery and

development of antiviral drugs for

biodefense: experience of a small

biotechnology company. Antiviral Res

2008;77:1-5

177. van der Groen G, Jacob W, Pattyn SR.

Ebola virus virulence for newborn mice.

J Med Virol 1979;4:239-40

178. Bray M, Davis K, Geisbert T, et al.

A mouse model for evaluation of

prophylaxis and therapy of Ebola

hemorrhagic fever. J Infect Dis

1998;178:651-61

179. Gibb TR, Bray M, Geisbert TW, et al.

Pathogenesis of experimental Ebola Zaire

virus infection in BALB/c mice.

J Comp Pathol 2001;125:233-42

180. Bray M. The role of the Type I

interferon response in the resistance of

mice to filovirus infection. J Gen Virol

2001;82:1365-73

181. Raymond J, Bradfute S, Bray M.

Filovirus infection of STAT-1 knockout

mice. J Infect Dis

2011;204(Suppl 3):S986-90

182. Shultz LD, Brehm MA, Bavari S,

Greiner DL. Humanized mice as a

preclinical tool for infectious disease and

biomedical research. Ann N Y Acad Sci

1245:50-4

183. Swenson DL, Warfield KL, Kuehl K,

et al. Generation of Marburg virus-like

particles by co-expression of glycoprotein

and matrix protein. FEMS Immunol

Med Microbiol 2004;40:27-31

184. Ryabchikova E, Strelets L,

Kolesnikova L, et al. Respiratory

Marburg virus infection in guinea pigs.

Arch Virol 1996;141:2177-90

185. Simpson DI, Zlotnik I, Rutter DA.

Vervet monkey disease. Experiment

infection of guinea pigs and monkeys

with the causative agent. Br J Exp Pathol

1968;49:458-64

186. Robin Y, Bres P, Camain R. Passage of

Marburg virus in guinea pigs. In:

Martini GAaS R, editors. Marburg virus.

Springer-Verlag; New York, NY:

1971. p. 117-22

187. Carrion R Jr, Ro Y, Hoosien K, et al.

A small nonhuman primate model for

filovirus-induced disease. Virology

2011;420:117-24

188. Alves DA, Glynn AR, Steele KE, et al.

Aerosol exposure to the angola

strain of marburg virus causes

lethal viral hemorrhagic Fever in

cynomolgus macaques. Vet Pathol

2010;47:831-51

189. Hensley LE, Alves DA, Geisbert JB,

et al. Pathogenesis of marburg

hemorrhagic Fever in cynomolgus

macaques. J Infect Dis

2011;204(Suppl 3):S1021-31

190. Fisher-Hoch SP, Platt GS, Neild GH,

et al. Pathophysiology of shock and

hemorrhage in a fulminating viral

infection (Ebola). J Infect Dis

1985;152:887-94

191. Hartlieb B, Modrof J, Muhlberger E,

et al. Oligomerization of Ebola virus

VP30 is essential for viral transcription

and can be inhibited by a synthetic

peptide. J Biol Chem 2003;278:41830-6

192. Hartman AL, Towner JS, Nichol ST.

A C-terminal basic amino acid motif of

Zaire ebolavirus VP35 is essential for

type I interferon antagonism and displays

high identity with the RNA-binding

domain of another interferon antagonist,

the NS1 protein of influenza A virus.

Virology 2004;328:177-84

193. Valmas C, Grosch MN, Schumann M,

et al. Marburg virus evades interferon

responses by a mechanism distinct from

ebola virus. PLoS Pathog

2010;6:e1000721

194. Kaletsky RL, Francica JR,

Agrawal-Gamse C, Bates P.

Tetherin-mediated restriction of filovirus

budding is antagonized by the Ebola

glycoprotein. Proc Natl Acad Sci USA

2009;106:2886-91

195. Bukreyev A, Volchkov VE, Blinov VM,

Netesov SV. The GP-protein of Marburg

virus contains the region similar to the

‘immunosuppressive domain’ of

oncogenic retrovirus P15E proteins.

FEBS Lett 1993;323:183-7

196. Lee JE, Fusco ML, Abelson DM, et al.

Techniques and tactics used in

determining the structure of the

trimeric ebolavirus glycoprotein.

Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design

Expert Opin. Drug Discov. (2012) 7(10) 953

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.

Acta Crystallogr D Biol Crystallogr

2009;65:1162-80

197. Lee JE, Saphire EO. Neutralizing

ebolavirus: structural insights into the

envelope glycoprotein and antibodies

targeted against it. Curr Opin Struct Biol

2009;19:408-17

198. Lee JE, Saphire EO. Ebolavirus

glycoprotein structure and mechanism of

entry. Future Virol 2009;4:621-35

AffiliationAmy C Shurtleff†1, Tam L Nguyen2,

David A Kingery1 & Sina Bavari1

†Author for correspondence1U.S. Army Medical Research Institute

of Infectious Diseases,

Integrated Toxicology Division,

Fort Detrick, 1425 Porter Street,

Frederick, MD 21702, USA

Tel: +1 301 619 8713;

Fax: +1 541 754 3545;

E-mail: [email protected] Cancer Institute Frederick,

Target Structure-based Drug Discovery Group,

SAIC-Frederick,

8490 Progress Dr, Suite 400,

Room 4093, Frederick,

MD 21702, USA

A. C. Shurtleff et al.

954 Expert Opin. Drug Discov. (2012) 7(10)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y G

azi U

niv.

on

08/2

0/14

For

pers

onal

use

onl

y.


Recommended