to - dtic · 1202 slim, p.o. box 3333 *nev haven, connecticut 06510-8047 9.sosripigmonit oring...

30
UNCLASSIFIED AD NUMBER ADB181401 NEW LIMITATION CHANGE TO Approved for public release, distribution unlimited FROM Distribution authorized to U.S. Gov't. agencies only; Proprietary Information; 01 FEB 1992. Other requests shall be referred to Army Medical Research, Development, Acquisition, and Logistics Command, Fort Detrick, MD 21702-5012. AUTHORITY 31 May 1996, per memo MCMR-RMI-S [70-1y], DCS/Info Mgmt., Ft. Detrick, MD THIS PAGE IS UNCLASSIFIED

Upload: others

Post on 27-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

UNCLASSIFIED

AD NUMBER

ADB181401

NEW LIMITATION CHANGE

TOApproved for public release, distributionunlimited

FROMDistribution authorized to U.S. Gov't.agencies only; Proprietary Information; 01FEB 1992. Other requests shall be referredto Army Medical Research, Development,Acquisition, and Logistics Command, FortDetrick, MD 21702-5012.

AUTHORITY

31 May 1996, per memo MCMR-RMI-S [70-1y],DCS/Info Mgmt., Ft. Detrick, MD

THIS PAGE IS UNCLASSIFIED

Page 2: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

DEPARTMENT OF THE ARMYU.S. ARMY MEDICAL RESEARCH AND MATERIEL COMMAND

FORT DEIRICK, FREDERICK. MD 21702 5012

REPLV TOA1Y ENTICNC4

MO4R-RMI-S (70-1y) E 31 May 96

MEMORANDUM FOR Administrator, Defense Technical InformationCenter, ATTN: DTIC-OCP, Fort Belvoir,VA 22060-6218

SUBJECT: Request Change in Distribution Statement

1. The U.S. Army Medical Research and Materiel Command hasreexamined the need for the limitation assigned to technicalreports written for Contract Number DAMD17-86-C-6042. Requestthe limited distribution statement for Accession Document Numbers

1 AM I-W and, ' be changed to"Approved for public release; distribution unlimited.0 A copy ofthese reports should be released to the National TechnicalInformation Service. I2. Point of contact for this request is Mrs. Judy Pawlus atDSN 343-7322.

FOR THE COMMANDER:

I

O IS R. FAY IIILieutenant Colonel, MSDeputy Chief of Staff

for Information Management

S

S

( • •• • • •• •

4 l ii . . .

Page 3: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

REPRODUCTION QUALITY NOTICE

This document is the best quality available. The copy furnishedto DTIC contained pages that may have the following quality

problems:

" Pages smaller or larger than normal.

" Pages with background color or light colored printing.

" Pages with small type or poor printing; and or

" Pages with continuous tone material or colorphotographs.

Due to various output media available these conditions may ormay not cause poor legibility in the microfiche or hardcopy outputyou receive.

L. If this block is checked, the copy furnished to DTICcontained pages with color printing, that when reproduced inBlack and White, may change detail of the original copy.

0 0 0 0 0 4

Page 4: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

CONTRACT NO: DAMD17-86-C-6042

TITLE: DRUG DEVELOPMENT ACAINST VIRAL DISEASES(BIOLOGICAL TESTING)

PRINCIPAL INVESTIGATOR: Gregory R. Tignor

CONTRACTING ORGANIZATION: Yale University4School of Medicine

1202 SHM, P.O. Box 3333Now Haven, Connecticut 06510-8047

4REPORT DATE: February 1, 1992 &DTICELECTE !

TYPE OF REPORT: Annual/Final Report MAI 2 1J

PREPARED FOR: U.S. Army Medical Research, Development,Acquisition, and Logistics Command, (Provisional),Fort DetrickFrederick, Maryland 21702-5012

DISTRIBUTION STATEMENT: Distribution authorized to U.S.Government agencies only, Proprietary Information, Test andEvaluation, February 1, 1992. Other requests for this documentshall be referred to Commander, U.S. Army Medical Research,Development, Acquisition, and Logistics Command (Provisional),

4 Fort Detrick, Frederick, Maryland 21702-5012.

The views, opinions and/or findings contained in this report arethose of the author(s) and should not be construed as an officialDepartment of the Army position, policy or decision unless so

* designated by other documentation.

94-08446 7-0..4 .3 154zJ

4

Page 5: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

REPORT DOCUMENTATION PAGE O MB No 01C4-OW~

I.~~~~~ AGNYUS NY Ldfbf_7Z EOR AE3. REPORT TYPE AND DATES COVERED1 February 19921 nnual/Final (2/l/86 - 1/31/92)

4. TITLE AND SUBTITLE S. FUNIDING NUMBERS

Drug Development Against Viral Diseases ontract No.(Biological Testing) )AMD1 7-86-C- 6042

G. AUTHOR(S) 373Gregory H. Tignor 3M263763D807 .AD.059

UDA3093647. PERFORMING ORGANIZATION NAME(S) AND ADDRESS([$) B. PERFORMING ORGANIZATION

Yale UniversityREOTNMRSchool of Medicine1202 SliM, P.O. Box 3333*Nev Haven, Connecticut 06510-8047

9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING

U.S. Army Medical Research, Development AEC EOTNME

Acquisition, and ogistics AGECYREORTNUBE

FreerikMaryland 21702-5012WFredeickAR For Deric*Annual covers last year of research, February 1, 1991 - January 31,192

12a. O:S'IFJTiOPN AVAI.'A!aITY SI..TEMEWdT 12b. DISTRIBUTION CODE

Distribution authorized to U.S. Government 4agencies only; proprietary information, testand evaluation, June 6, 1969.

13i. 0BSTAAC! 'Maxsfrum 201 words)

Ribavirin and N-exides of adenosine were two groups of nucleoside analogues which were effective inreducing mortality in Congo-Crimean hemorrhaigic fever (CCHF) virus infected infant umice. A singlenucleotide change in dhe ribaviuin molecule could abrogate drug efficacy. During ribavirin treatment,thast I whp emsi inittsuedwsh-exac ul eotpic than the parent wh se i ivirus suggesting

tha hstcel odfid irs elcton ccrrd.Riavri teatren asoreduced lyphyticchoriomeningitis (LCM) virus mortality in adult mice although new drugs, including some N-oxidesof adenosine were identified which had a significantly greater effect. Both ribavirin and ana-Atreatroert reduced the number of tail lesions in vaccinia virus infected mice although an analogie ofadenosine-N-oxide with a 3.4-dimthyl group was even more effective. Four immunomodulatorssignificantly inhibited tail lesions in dhe vaccinia virus mouse model. These included quinolinaminie,aunpligen, recombinant ILI, and poly ICLC stabilized for injection. Ribavirin did not reducemortality of yellow fever virus infected prnms. An imznunomodulator, AM (Lederle), reducedviremia as did quinolinamine which reduced both viremia and morality. Ribavirin neatmen! did noteliminate skin lesions in vaccinia infected primates, but the titer of virus in vesicular scrapings wasreduced.

1?. S.JGJECT TERMS IS. NUMBER OF PAGES

Anti-Viral Drugs; Vaccinia Virus; Yellow Fever Virus;: 6 RIECOCrimean-Congo Hemorrhagic Fever Virus; PAD I 6 RIECO

1.SCiTY CLASSIFICATION itB SECURITY CLASSIFICATION .3. SECURITY CLASSIFICATION 20. LIMITATION OF ABSTRACTj1 F CREPORT OF THIS PAGE j OF ABSTRACT

I Unclassified IUnclassified IUnclassified Limited _!.S5J 75C40~.01;05500 Starsad Porm Z1)8 Cev 9

Page 6: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

S

FOREWORD

S

Opinions, interpretations, conclusions and recommendations arethose of the author and are not necessarily endorsed by the USArmy.

Where copyrighted material is quoted, permission has beenobtained to use such material. S

Where material from documents designated for limited-i-tribution is quoted, permission has been obtained to use the

material.

Citations of commercial organizations and trade names inEs report do not constitute an official Department of Armyendorsement or approval of the products or services of theseozganizations.

In ccnducting research using animals, the investigators)adhered to the wGuide for the Care and Use of LaboratoryAnimals,- prepared by the Comittee on Care and Use of LaboratoryAnimals of the Institute of Laboratory Resources, NationalResearch Council (NIH Publication No. 86-23, Revised 1985).

For the protection of human subjects, the investigctor(s)adhered to policies of applicable Federal Lay 45 CFR 46.

In conducting research utilizing recombinant DNA technoloqy, Sthe investigator(s) adhered to current guidelines promulgated bythe National Institutes of Health.

In the conduct of research utilizing recombinant DNA, the=hestigator(s) adhered to the NIH Guidelines for Research

Involving Recombinant DNA naleca.es.

In the conduct of research involving hazardous organisms,the investigatorts) adhered to the CDC-NIH Guide for Biosafety inNicrobiological and Biomedical Laboratories.

INTIS V1& I 5DTIC TAB PIrWj~atr DtUnamn, w--,ed 03

Dusti ,to

i-

0 0 0 S 00000

Page 7: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

1. TABLE OF CONTENTS1. TABLE OF CONTENTS ........ ...... .. . ..... . ............. 2d11. TESTING IN MUR&NE MODELS .......... . .......... . . ... .............................~.. . .... 3

A. THE CRIMEAN-CONGO HEMORRHAGIC FEVER VIRUS MODEL.....-..... ................. ..................................... 3I. SttmetOfPobe Udr3

3. Rakm sd n rentw~A Std.-.................... .................................- -... ...... ......... 4

L Nuu~ m ber Mada ofdustad............................... ...... .... ... ...................................4

b. Model of CCHF virus . ..... . . ... ... ........................... .... 4C. Dmioooafey .---------- -..-------........- ..-..

d. Anayisof 5a.---- . -~.

S o OfRaub --- -M CC-------- ..................------------ 6Tablet. RbudamoImwCnwAm.Caqs ion.nkfevw(CCIIF) vw-lemai hdmm=ca..-.7

b. Pedeeas o(CCUF vm i phmb.valo~e md ubevrilesmd imie7&. EllacotrilwkneemCCHlF worse wdsuuaybis the..ra narvai y ---------4 ies anloein doSCOWHdeL 9.........- -

G. D em A~ C ee .......m 103 M mLCH VOWS MODEL- - .-- .---- II

4. DrisiaOf L MVMV Rasui-------ts-_-- 12

6, DNEffmewlv110o* h1MV AnCOW Mhdes 1C THE VACCMA VIRUS, MODEL-..- 12

I. bia Rfhe ftbaae And (idappUe ud es.m wwln. iii mlm ... - - -. . 12. I3. R----------.....I

L V9W bWm (Xnil W i U& i inab.) 14mi i--------

Table 4. Veiai Mae; ad Ted VmTalLe.Nhdi~I

a. EAWemeas ad mian koctn o onlesn. .-- ------- 1

3.YLO eV AD chnBlMUOMatLsOU alei nd o,1

Table 4. amyleaja Efin111a Fre ( VSi1 Tod La Yehum__v

4 b.aaa n QujowimdYelwFyYi.Vena.... . . . . .2

C. YEVAYT VIRU AND ANIMALDUG

* D1. DISCUSSION AND ONCUNMCLO -. . . . . . . 22IV. LTRTUeeeswCLU67D........-.......... ----.. . . 23

V. CHOOO IAul oBIB is~vama with RAH Oid ALLvi .P. .CAT..I..NS............. ....................................... 25V. PSELPOR TDAN D G(EGREE3 GR uneiiANTED. ............. ............................................ 25

bVII.inrife n elo ee DVSiruTIs U ..... . . .. . . ......... ............................... 22

C.V C IM I USI RI A E ........ _ ........ ........ ....................................2..'

1.Efec f ib vnn on v rcnu eson n ri aes............... .. ...... ..................................................2

D . D I C U S I N N D. .. .. .. .. .... .. .. .. .. .. ... .. .. .. .. ... .. .. .. .. .. ... .. .. .. .. ... .. .. .. ..2rV LI E AT R CIE0.......... ................... .................. 0.. 0..... ... 0.......................... .

Page 8: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

.. : NG IN MURINE MODELS

A. THE CRIMEAN-CONGO HEMORRHAGIC FEVER VIRUS MODEL

1. Statement Of Problem Under StudyThe purpose of this work was to conduct primary testing of antiviral compounds in a mrine model

of Crimean-Congo hemorrhagic fever virus infection.

2. Background And Review Of Appropriate Literature.Infection of humans with Crimean-Congo hemorrhagic fever virus (CCHF) often results in a serious

illness followed by death. It has been estimated that about 33% of hospitalized patients with a CCHF virus 5diagnosis die(l). The etiologic agent of CCHF is a single-stranxded RNA virus of the Nairovirus genus Inthe Bunyaviridae family (2). It is distributed throughout much of Africa. Europe and Asia. Virus strainsfrom differing geopaphic regions have not been distinguished by those scrologic techniques which haveidentified geographically distinct virus mins from other virus families (3). The epidemiology of CCHFhas been reviewed in detail by ude (4,5).

There is no widely used or widely accepted vaccine for CCOF. CCHIF vaccine is not availablecotmmercially outside Bulgaria and the USSR. Administration of plasma from immune donors isconsidemd by some to be the stndard specific reatment for patients with CCHF i. However,immunotheapy in COHF is not without controversy. One view is that the variable uesults after hu ma

immunotherapy trials reflect the difficulty in getting donors with high antibody times 19i. Thr are otherproblems. Innmune plasma or globulin cannot be used prophylactically because the supply is limited andneutralizing antibody titers in convalescent plasmas ame low. In actual practice, only patients with a well-established CCHF diagnosis receive immune plasma. In one study, only transient improvement in clinical 0status was noted without a clear effec on viremia (6).

There has been limited experience with alternmte forms of CCHF therapy. During a 1984 outbreakof CCHF at Tygeberg Hospital in South Africa, two different antiviral agents, ribevirin and humanleukocyte interferon, were given. Ribavirin was used prophylactically in 6 of 9 CCHT case-contacts whohad sustained direct blood exposure by various means including needle-prick. Thuee individuals alsoreceived a shoi coms of interferon. Of the 6 treated contacts, one (who did not receive interferon)developed a mild case of CCHF. The 3 untreated contacts developed severe CCHF (7). South Africanpatients who wee treated with high doses of human leukocyte interferon either prophylactically ortherapeutically (together with plasma) developed severe side-effects; the practice was stopped [71. Theresults of this clinical trial did not provide an uncomplicated basis for continuing use of these two antiviralsduring future CCHF epidemics. Them were no laboratory data from the South African study showing drug

efficacy aglinst CCHF virus infections.Six years later, a brief rport was given of the result of ueating 12 CCHF paticnts widh ibavinn in

open-label trials in Soutn Africa (8, 9). When ribavirin therapy was started early (,4 days post-onset), 0 of7 patients died. However, of the seven survivors, only 2 had clinical laboratory markers associated with

3 S

Page 9: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

mortadly in historical (i.e.. untreated) controls. After late initiation of therapy (Z5 days post-onset). 3of 5 patients (60%) died.I

1. Rationale Used In Current Study.T1his present research was to find a scientific rationale for use of antivirals in CCHF virus infections.

The focus of the work was on ribavirin since it had been used in a human trial. Considerations invalved in .the design and execution of an antivirul drug study have been reviewed elsewhere (10).

According to prevailing criteria, CCHP virus infection is suitable for antiviral study. First, outbreaksof CCHF have been unpredictably sudden, frequent enough and of enough seriousnssm to warrant antiviral Ddrug study. Secondly. the potential for antigenic: or biologic variation among CCME virus strains increasesthe need for a broadly active antiviral drug. Tirdly, thi was a practical study because it could beincorporated into a broader studly desiged to fin antivirals against a wide spectium of virus diseases.

Prope models for COIF infection have been difficult to establish because frequently usedlaboratory animals show little, if any. sip of infection or disese (11. Also, laboratory work with CCHF hasbeeni limited because accidental infections which have had serious or even fata outcorm have toofrequently happened (11). Also little is known about the humans dises process. So, as has been done insimilar situations, both the candiat compound and the experimentail model itself had to be examined inthis study 110). The experimnental model used in these experiments was an infant mouse model based on 0

inw~ertenal(ip) inoculation of CCHP vkwu.

4. Experumattal Methodsa. Namnbgr and r~~n qldrs ered. Approximately 500 drugs fromn 4 classes were tested. 0

The four classes of drugs included said and optically protectd vuants. amine and derivatives, nucleosideanalogues with modified side chains and contuninant-free derivatives of natural products some of whichwer immimuomodulasors. Drop were cddso laboratory testng was done with only solubilityinformation. The chemical kkdetty and the relatonship betwee drugs were unknown by laboratoryworkm

b. Madel 01CCHF viru iWecdo#L Dmug wene tested for toxicity in 1-3 d oKd CharlesRiver strain CFl rndom-bred mice at 50 mu~kg. Eight pups were inoculated ip with each drug. Thosedrugs which were not toxic were tested aqint COIF, suai IbW 10200, passage I1I or 12 in infant mousebrain 131* This virus strain was isolated from a pool of ticks in Mbadan. Nigeria and was chosen for biologic 9safety reason becaus it had not been associated with either naturally occurrng or laborasory-associatedhuman disease.

Each drug was inoculatd ip into infant mice in a volume of 0.075 ad. Forty-five minutes later. 50LDSo's of virus were inoculated ip in a volume of 0.075 ml. Placebo-nrested mice were inoculated withtissue culture medium (DMEM). The virus was tirated simultaneously. In multiple drug dose tests, a totalof 4 more daily drug doses was given after injection of virus.

4

0S

Page 10: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

S.c e hou Ned and ob.srved daily in a biosafety level 3+ animal facility modified ICA

.. L.4 yLt tor nsork. Senuticl mice were tested serologically for intercurrent murine infections by the

Y.l University Division of Animal Cae. Ec. Biologic safety. Class 2 biological safety cabinets wen used in a biological safety level 3

laboratory for manipulations involving infectious or potentially infectious materials. Individual workerswere equipped with HEPA-filtered respirators and underwent serologic surveillance throughout the course Aof this project. Laboratory work was closely monitored by the University's Biological Safety AdvisoryCommittee and its representatives and by the Safety Officer of U.S. Army Medical Research Institute ofInfectious Diseases. Safety conditions on this project and in the University. at large, were reviewed in great 0depth by the Chairman of the Senate Subcommitte on Oversight Of Government Management of theCommittee on Governmental Affairs, United States Senate (12).

d. Analysis of daut. Geometric mean times to death were calculated for placebo (viruscontrol) mice (VC) and drug-treated mice. The geometric mean time to death was equal to the nth root(where n-the total number of animals) of the product of each day with moutality raised to the power of thenumber of animals dying on that day. In this calculation. survival was defined as 28 d.

METHOD FOR MEASURING ANlTWIRAL DRUG EFFECr

Vlt- GODMEJCbEAN SURVIVAL1ME(IRUG) 0

OBDM1EC AEAN SURVIVALTDIE (R.. MQ

Th vis rating of antiviral efficacy (VR) for each drug was equal to the ratio of the geometc mean time to

death for each drug divided by VC (13). When a single positive drug was identified, that drug wasinicorporated into each teat as a standard of teat sensitivity. T1he data were analyzed as described in an earlierpublication on CCHF virus from this laborory 13). 0

e. Pathogenesis of CCHF in Iram Mice. Pk cebo-neated and ribaviuin-ueawd mice were

each divided into two groups. one of which was held for observation. Virus was tivated daily from the othergroup by inoculating SW- 13 cells (supplied by Dr. D. Wan, U.S. Army Medical Research Institute ofInfectious Diseases) with blood seawn, liver, brain. spleen, and hear tissue homogenass. Tissues fromeight animals were pooled and examined in four replicate cultures at each interval. Mice were firtexsanguinated and then perfused with DMEM. Other tissues wee removed. weighed, homogenized,

clarified by light centfugation and the supematant fluid inoculated into freshly made cultures as describedin earlier pathogenesis experiments (14). Virus titers wee detrmined by fluorescence assay of infected

cells under semi-solid overlay and expressed as FFU's/inoculum as described earlier 131. The variation 0

between individual titers was less than 0.3 Iogl0.Infected tissues were also examined by immunofluorescence. Sections of liver tissue from 6 mice

were cut in 46 micron sections, mounted, fixed in acetone, stained with CCHF or control rabies virisantibody, washed in PBS. and stained with fluorescein-conjugated goat anti-mouse antibody (Antibodies 0

Incorporated, Davis. CA). Co-localization of virus and specific cell types was by peroxidase staining ofparaffin-embedded tissues using a commercial kit (ExtrAvidin Biotn Staining Kit, Sigma, St Louis, MO1iard virus-specific or cell-specific monoclonal or polyclonal antibodies. Mac- I antibody (monoclonal

5IS

4

Page 11: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

%I lj7O11L to a surface antigen found on mouse macrophage cells) was purvhjawd 0%Lorluflrctaily t~oehrinecr Staunheim Biochemicals. Indianapolis, IN) (15). This antibody has beenJm-~nbed as being weakly reactive with liver tissue (151. E

f SerLogy. Preparation and use of CCHF virus polyclonal antibody in IFA andneutrilization tests was as earlier descuibed[3l. Antibodies for immunofluorescence observation includedthose described ia ;.n earlier publication and others from the reference collection of the Yale ArbovinjsResearch Unit 121 Neutralization tests for monitoring the ptuity and identity of nbavirn-resstarlt viruswere done using, onstang antibody and varying virus dilutions. In this protocol, mice which escapedneutralization 0i C.. breakdmoqh mice) we.t examined by IFA to establish the identity of the virus in thetssues.

Hybridomas producing monoclonal neutralizing anti-COIF antibody were prepared afteriztmspleniinoculto~n~ of vuusj usig otherwis stndr mex (16. 17). The monoclonal antibodieswere isoyped using a commercial hit (Zymed Labauuzories. Inc., San Francisco, CA). Screening was doneby EFA and immFnprecpitationi using a method modified from Maso (18). PelleWe (100.000 1 for 2 h)lradiolbeled (35S) CCHF virus (rem C CU cel Ur supemnatant fluids was sulubilimod in RIPS buffer(10mM Tnis, pH7.5. 150 mM NaC! 5mM EDTrA. I % Na deooxycholaw. and I % Triton X- 100) containingp n ons inhibitors (PMSF 1mM, Apinin 10 lag/mI leupeptin 2 paajmI benzamidine 24igfml and ThCK 10&ag/m). Fifty Pal of solubiliind material wan incubated overnight at 40C with 1-2 id of clarifled monoclonalor polyclonal amic fluid. The immunopuncipiwed maserial was sedo rd with Protein A-Sepharosebeads (Siml) which had been wshd 3 domes previousy with RIPA buffer (RIPS buffer with 0. 1% SDS).The pellesad material was washeid 5X with RIPS buffer and the proteins were releaed from the beads byincubation in sample buffer for 5 win at IOOO The plrotins were sepaaed on a 10% msylamide gel 19

which was processed for nummadiopah sing Kodak film5. Sumoiny Of Rink.

&. Ribavuirfin di e CCHF itana ,noue imoulWhen given in a single dose early after infection. ribavirin was an effective unatment in doses as low as 50mglkg in the CCHF model. In 19 tst using an exact 50 LD50 virus dose, the mean GMTD was 15.2±0O.5days. an increas of 7.2 days (p=0.0001). Using a consmant 50 ua&kg dome of ribavirin. a din"c relationship(Comai codftdidu (r2.0.8) caimd between ribavirn GMTD and viru dome (25 to 200 LD50's). Thelowest increase in GM was 3.2 days (200±80 LD~o's; p.00) and the gratest. 12.0 (25±10 LD50's).(Table 1)

*Using 50 LDS's of virus the GMrTD aft multiple ribavirin doses (50 mg/kg. for 5Sd) was higher(20.4±0.3) than after a single dose (15.2±0.5). The GMMT after multiple injections of 100 mg/kg was21±0.4. However, a single 100 mg'kg dose was nearly as effective (20.2±0.4) as multiple dnses (l10)

Smgpig). (Table 1) Beginning treatment on day 5,at the earliest time of onset of clinical signs, had no effecton either mortality or GMTD.

6

0S .4 ... ..

Page 12: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

.... .:.ircd to the I0g p(Otectivc et'fCct P). The P1 was a direct measure ol red, Iced

::...ty ubuutble to nbavinn treatment. The correlation coefficient (r2) was 0.76. A increse in GM.TD

,: 3.2 da)s; (range 3.1-3.5) was equivalent to a 1.7 Pl.

These data were based on changes in GMTD. Experiments were then done to determine if increased

C.MTD, after ribavirin treatment was a result of decreased virus replicatien in a specific target organ. 0

T.ihle I. Ribavirin treatnent of Crimean-Congo hemon'haoc fever (CCHF) virus-infected infant mice.CCHF vrus strain 10200a Ribavrun/Placebo treamnent-- Oulwomec

Number Virus dose Drug dose Drug dose Percent Geometricof tests (LD5o) mg/kg schedule morality mean time

to death6 50±15 Placebo Single 100 8.0±0.6

19 50±15 50 Single 15 15.2±0.56 50±15 100 Single 2 20.2±0.4

2 25±t10 Placebo Single 100 8.6±0.4 02 25±10 50 Single 12 20.6±0.42 100:k24 Placebo Single 100 7.3±0.92 100±24 50 Single 37 14.4±0.52 125±33 Placebo Single 100 7.6±1.22 125±33 50 Single 37 13.3±0.62 175±50 Placebo Single 100 7.3±1.6

"175":00 so single so I11.0:±0.8

2 2000 Placebo Single 100 7.0±2.22 50 50 10.2±0.8

6 50W Placebo Muiple 100 7.4±026 50±2 50 Mulie 12 20.4±0.36 W*Z.2 100 , Multipe 0 21.0£-0.4 I

a Pamm 6 in MA mm brim ti . ip wAcdlmmb Mem (soot) w grea ip45 dw vm or du* wid 4 s d t ua. ommon otdve day& Th p" cebo ww

C 8- t0 amuamOakK Inh pWp

b. Pathogmens of CCHF Wlw in pacebo-wewed and rbaviin-read Wamt mice.

The liver was the wim y target organ in ip COIF virus inf-xcton of infant mice. Virus titers were

slightly higher in the liver than in the blood from day 3 to day 7 (death of the placebo-ouated mice).

Relative to ,us growth in liver tissue, virus appeared -cry late after infectio in other tissues including thehr un and heart. Virus was not isolated from the spleen after day 2. The differences in virus titer between

the liver and the brain, he and spleen we signifm (pe.. 0 5. paired -les). However, the differences in

tter between the liver und blood were not statistically significant.Ribavirin-treied mice (Ningle dne. 50 mg/kg) had significantly lower virus titers in liver tiscue

1'-) ()7. patred 2-wil t-test) ar.d a significantly reduced viremia (p=0.007, paired 2-tail t-test). Althoughvrus growth in liver tssue was suppressed by ribavirin tratment, a large viremia was detected beginning on

v S. The titer of the vuemia, in these experiments seemed higher than the virus titer in the liver, but the

7

p

0 0 0S 0 00 0

Page 13: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

%. .a.s rot tiIucally different (p--0.08, paired 2-tail test). Despite t ict viremia, none of theLLa':ed-rmce held for observation died. Mice were not examined after day 7. I

The target organ was examined by both IFA and immunoperoxidase techniques using both frozen

and paraffin-embedded tissue. Three d after inoculation of virus, CCHF virus antigen occurred in whatappeared morphologically to be Kupffer cells lining the liver sinusoids of placebo-treated mice. Anir= alstreated with a single dose of ribavirin had very little or no demonstrable CCHF antigen in these cells at thistime. (Figures illustrating these data were presented in the Annual Report, 1989)

Later in placebo-treated animals, CCHF virus antigen was present in many clusters of hepatocyteswidely distributed throughout liver tissue including occasional tissue macrophages. Infection of Kupffercells was not prominent. CCHF antigen was only occasionally seen in liver tissue of ribavirin-treatedanimals, sometimes in a putative Kupffer cell or sometimes in a hepatocyte.

Kupffer cells were identified by immunoperoxidase staining on serial sections of paraffin embeddedliver tissue sections using anti-Mac- I antibody; infected cells were identified using ant-CCHF antibody. In 0consecutive tissue slices, some Mac- I antibody-positive cells i.e.. Kupffer cells, were also positive withanti-CCHF antibody on day 3.

There was CCHF virus antigen in the liver tissue of animals treated with both single and multipledoses of ribavirin although the number of infected cells were few. Seven d after ip inoculation, infectiousvirus was isolated from the liver of mice treated with multiple doses of ribavirin (50 mg/kg for 5 days).

c. Effect of ribavinn on CCHF virus entry into the central nervous system.

In the experiments described above, evidence of CCHF virus infection of the liver was found in apparentlyhealthy ribavirin-treated mice. Despite a relatively high viremia, infection did not spread to the "rain orother organs. This result was different from that in placebo-treated animals where viremia was followed by 0

infection of other organs (brain and heart).

These results suggested that the protective effect of a single dose of ribavirin (50 mg/kg) diminishedwith time. Therefore, the experiment was repeated, focusing only on viremia in ribavirin- treated mice andusing a higher ribavirin dose (100 mg/kg) Additional doses of ribavirin were given on days 5 and 9. Virus Iisolations were not done before day 6. Viremia (log 10 FFU/ml) was found on days 6 (3.6±0.2), 7 (4.5±0.2),8 (2.6±0.2), 9 (2.4±0.3) and continued at that level through day 11. On day 12, the titer (log 10 FFU/rnl)decreased to 1.3±0.3; virus was not found on days 13 or 14. Despite the relatively prolonged viremia, viruswas isolated only from the liver (days 6-13), not from the brain, heart or spleen. Titers of liver virus werenot determined. Mice held for observation did not die.

Liver tissue from CCHF virus infected, ribavirin-treated mice (single dose, 100 -qg/kg) was titrateddaily by ip subinoculation into infant mice. The peak titer in the liver was found on day 8. Eight ribavirin-treated mice (single dose, 100 mg/kg) without clinical signs of illness were necropsied on the eighth dayaf:er ip virus injection (50 LD50). Virus was found in pooled serum and supernaant fluid from tissue 0homogenates of pooled liver but not other organs including brain, heart, and spleen. The infectious virustiter (loglo LD5s/ml) in the liver was 4.0 4pd 2.3 in the serum as determined by ip inoculation of infant

mice. Virus from liver tissue of ribavirin-.-cated mice grew very poorly in CER or SW-13 cells. For

8 •

Page 14: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

i ... i:du.d v.ells of Lab-Tek slide trays containing 5 x 104 CER cells, 2-10 positive c-1, CreC

c:..nr,,ed by IFA. Results were similar with SW-13 cells. Residual ribavirin in the tissue homogenates.,,u', have inhibited virus growth in vitro. Therefore, the virus was pelleted from the supernatant fluid by

ultracentrifugation (100,000g, 60 min), but virus infection of cultured CER cells was not increased.

Because of the variability between individual cultures in the number of infected cells, it was impossib'e todetermine in vitro ribavirin sensitivity reliably.

Attempts were made to determine ribavirin sensitivity in vivo. The original liver tissue virus fromribavirin-treated mice was used as source in an in vivo ribavirin drug test. Mice were infected with livervirus (50 ip LD50) and treated with a single dose of ribavirin at either 50 or 100 mg/kg. The GMTD in mice

treated with 50 mg/kg was unchanged from the placebo controls (mortality 8/8) and the GMTD in mice

treated with 100 mg/kg was increased by 1.6 days (mortality 8/8). Eight of eight (8/8) placebo-treated

animals died with an GMTD of 8.1 days.

Virus isolations were done by ip inoculation of infant mice using pooled tissues from two mice takenon days 7 and 8 from the two groups, one tiated with 100 mg/kg and one placeb,-treated. Virus wasisolated from the liver and serum in both treated and placebo-treated mice on days 7 and 8 but not fromother organs on either day. In treated mice (100 mg/kg), virus titers (log10 LDs0/ml) were as follows: Day7, liver, 4.2; serum 2.9; Day 8, liver 4.5, serum 2.7. Virus titers in placebo mice were as follows: Day 7,

liver 5.7; serum 5.3; Day 8 liver 5.2; serum 5.3. These data show that ribavirin treatment (100 mg/kg) 0

reduced virus titers in treated animals. In contrast to an earlier experiment with the parent virus, treatmentwith ribavirin at 50 mg/kg had no effect on virus titers (data not shown). Despite the relatively high viremiain placebo-treated mice (5.3 logl0 LD50/m), virus was not isolated from other tissues (brain, heart, spleen'.

This suggested a marked hepatotropism for the injected virus. 0 4Three blind passages of the hepatotropic virus through placebo-treated mice resulted in a loss of

strict hepatotropism. After three ip passages of liver tissue virus harvested on day 7, sick infected mice werenecropsied. Isolated virus had the following titers (logl0 LDs0/m): liver (6.1±0.2); serum (5.6±0.2); brain

(7.1±0.2); heart (6.6±0.2). Strict hepatic tropism was no longer in evidence.*d. Ribavirin analogues in the CCHF model. Three types of ribavirin analogues were tested:

(1) those with changes in the heterocyclic moiety; (2) those with changes in the glycosidic moiety; (3) C-nucleoside analogues. Three analogues closely related to the parent ribavirin (VR, 2.47±0.70) had similar

efficacy: (1) ribavirin triacetate (VR, 2.30±0.07) (2) ribamidine (2.63±0.45) and (3) AVS 4071, 1-beta-D-

ribosfuranosyl-1,2,3-triazole-3-methylamidate (VR, 2.3). Tiazofurin triacetate (VR. 1.2) and seleanzofurin 8(VR, 1.5±0.14) were less active in the CCHF model. Tiazofurin (VR 1.0) and the seleanzofurin 5'monophosphate (VR, 0.9) were inactive. Additional modification of tiazofurin triacetate, seleanzofurin, and

seleanzofurin 5' monophosphate resulted in either unchanged efficacy or toxicity problems.

9

0. . . . . . . . .. . . i -

i iii . . . . . . ... .. .. . . . . .. . ' " ' .. . ' i .. ..

Page 15: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

6. 1; 'u siion And Conclusions

This research was to test ribavirin and other compounds for antiviral activity against CCHF virus tosupport intervention with ribavirin during human CCHF virus outbreaks. We used an infant mouse modelbecause the virus does not obviously infect other commonly used laboratory animals. As a result, some

c'asses of drugs (i.e., some immunomodulators) could not be reliably tested in our model. In addition, thereis concern that metabolism of ritoavirin may be different in infant mice and in primates.

On the other hand, finding the early association of CCHF virus with Kupffer cells supplied

significant evidence suggesting the model bears relevance to htman disease. A series of published autopsy •

findings was summarized by Hoogstraal in 1979 [4]. After extensive pathologic examination of maay casesof fatal CCHF infection over a four year period, Karmysheva concluded that CCIF virus multiplied in cells(especially KupfYer cells) of the reticulo-endothelial system, in which specific antigens were detected byimmunofluorescence.

Published proof of CCHF virus growth in Kupffer cells is in agreement with our present research

findings. That is reassuring since some muine comaviruses and son arboviruses have a liver tropism ininfant mice similar to that shown in our experiments 20, 21, 22. Because of that, we have gone to elaboratelengths to eliminate intercurrent infection or contamination of reagents as an explanation for theexperimental results. Using a CC]HF virus neumalirng monoclonal antibody to confirm identity of resistat avirus was one step in establishing its purity and identity.

Our data reported in detail in previous Annual Reports suggest that ribavirin-resistant virus waspropagated in the liver of CCHF virus infected, ribavirin-treated mice. A dose regimen (single dose, 50mg/kg) that, with the parent virus, reduced virus titers and mortality was ineffective using vurus harvested 4from the liver of ribavirin-treated animals. Nevertheless, an increased dose of drug (single dose., 100 mg/kg)did. indeed, substantially reduce virus titers in both the liver and serum even though mortality was notreduced. Repeated passage of the liver virus in ribavirin-treated mice (multiple dose, 100 mg/kg) resultedin a loss of infectivity. These latter two observations suggest that, while partial resistance was apparent, itmay not be real. What is seen as partial resistance in this model may be related to a pharmacologic property Iof ribavirin unique to infant mouse tissue. For example, infant mouse liver cells could become resistant tothe effects of low doses of the drug thereby making sensitive virus appear resistant as postulated elsewhere

Evaluation of ribavirin analogues and other antiviral compounds in the CCHF model became moreimportant because alternative drugs or therapeutic procedures might be needed in clinical settings tominimize development of resistance. Our results can be put into the framework of ribavirin structure-

activity relationships which hav, been earlier evaluated [29]. We are in agreement with previousexperiments with other viruses in which addition of an amide group (C=O to C=N) to reduce toxicity did notchange ribavirin efficacy. However, addition of an amidine group (N-CH2-CH2--CH2) to reduce toxicity

destroyed drug efficacy. Adding an -OH to the amidine group to reduce toxicity and increase solubility did 5not restore efficacy. One possible explanation for loss of activity is ,iaL ;it amidine group inhibits ribavirin

phosphorylation.

10

0 0 0 0

Page 16: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

.::s wh ribavirin C-nucleoside analogues are different frnm those with olhcr Cis :

e' :.-..d In 23j. We did not find that ribavirin triacetate was significantly more effective than nbavirin. Inthe CCHF model, tiazofurin analogues (AVS I 1l, AVS 257) are inactive whereas selenazofurin analogues(AVS 253. AVS, 3705) ae active. We cannot address synergism in the CCHI-F model because combination

therapy was not used.

The other group of nucleoside analogues active in the CCHF model were N-oxides of adenosine. It

*as not altogether surprising that both ribavirin and adenosine analogues were active in the CCHF model.

Molecular models have confirmed the structural similarity between these two compounds although the

resemblance of ribavirin to adenosine is not readily apparent.

B. THE LCM VIRUS MODEL

1. Statement Of The Problem And Rationale Used.

Antivirals for arenavirus infections remain a high priority. The prootypefor this group of infections may beLassa fever. Lassa fever is an acute infection characterized by fever, headache, myalgia., pharyngitis,

vomitng, a relatively mild hemorrhagic diathesis, and a capillary leak syndrome with facial edema, adult

respiratory distress syndrome, shock and multiple organ failure. The disease in experimentally-infected

rhesus monkeys closely resembles that in humans. Ribavirin treatment initiated as late as 5 days after virusprevented lethal infection, reduced cell injury as reflected by serum trasaminase levels, and abrogated

virmeia, inspiring open-label trials, in which mortality in treated patients was compared with that in

historical controls. Despite shortcomings in design and anlaysis, a convincing case was made for efficacy of •

ribavirin. Antiviral treatment of experimental and human Lassa fever has not be associated with late

encephalitis or encephalopathy, as described in the other arenaviral hemorrhagic fevers (reviewed in [9]).A murine model of arenavirus infection infection was used to screen drugs for antiviral efficacy.

481 drugs wer tested in the LCMV model. The methods and calculations for determination of efficacy

were done by procedures described in deail above for CCHF virus. 1

2. Decription Of The LCMV ModelAdult mice ar inoculated with 50 mg/kg of dr.g i.p. in a volume of 0. 4 ms. Forty-five minutes later, mice

are inoculated with 50 LDS(Ys of LCM virus (LCMV) i.p. The virus strain was propagated by intracerebralpassage in inbred C3H mice. Random bred CF-I mice from Charles River were used for drug tests. The

identity of the virus strain was monitored by examination of infected mouse tissue by immunofluorescence.

3. Histopathology Of LCMV InfectionAdult random-bred mice inoculated with 50 LD50's of LCMV were examined on days 10 and I I post

inoculation. The details of our studies were presented in earlier Annual Reports. In summary, mice were

suffering from severe, multisystemic disease, with necrotizing inflammation of lymphcid tissues, parotid

salivary glands, pancreas, splenic red pulp, liver, intestine and mesentery. They also had mild focal

,, . 11

€ .... •• • • •• •

. . . .. .

.... . . . . .. . . ... . . . -. .. ... .... . . . ..,,,. . . . . .. .I

Page 17: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

... c .., ~. l iac jority of hcukocytes, regardless of type., in all tissues cxaminud were unldrgoir.i,,¢rois. No leb'ons were found in submaxillary or sublingual salivary gland, kidney, heart, eye, lacrimalgland, thyroid, trachea, or lung. Lesions were consistent with visceral LCM disease as described elsewhere(24.

4. Distribution Of LCMV Results pThe mean VR score and the mode of distribution of all drugs tested 1.0 and 1.077 respectivel). Therelationship between VR score and percentile ranking was presented in earlier Annual Reports. VR scoresof 1.5 or higher were in the 95% of all drugs tested.

S. Analysis Of Reprodtodbil.ty And Variation. bVariation in the test system was low. The standard deviation of ribavirin (AVS#l) was 0.4. In the CCHFsystem, the standard deviation of the VR+ AVS #1 was 0.6. Thus, there was less variation in the LCMmodel, but there was also less consistent positive effect on survival.

6. Drugs Effective In Both The LCMV And CCHF ModelsThirty-one drugs were identified as aeing of potential interest for additional tests of efficacy. Ribavirin waseffective in this model although the triacetate analogue (AVS 206) was more effective than the parent or anyother ribavirin analogue tested. The VR scores ranges from 1.2 to 1.6 depending upon virus dose used and

the number of drug doses given.Aside from ribavirin, the most promising, in terms of VR scores, was AVS# 4070 which had mean

VR's of 1.7 and 1.8 against test dose. of 100 and 63 LDS(Ys respectively. This drug had a higher VR scorein the LCMV model than we observed with any other drug including ribavirin (AVS# 1) or any of its tested

analogues.There were 6 drugs which were effective in both the LCMV and the CCHF models. In addition to *

ribavirin, (AVS#1, 206), there are 4 other drugs. They included AVS #4071, 4720,4796 and 4217. Data onthese individual drugs were presented in detail in previous Annual Reports (1989 & 1990).

SC. THE VACCINIA VIRUS MODEL

L Statement Of The Problem And Rationale Used.There is no evidence that the smallpox eradication program has been less than a total success. Nevertheless,the mili:ary continues to immunize its personnel with vaccinia virus. Because of the dearth of naturallyoccurring cases, the risk of vaccine-induced disease has begun to surpass the risk of naturally occurringdisease. This is particularly true since a large closed-population of military personnel is being activelyimmunized. Vaccine-induced side effects become both a significant disease burden and financial liability.

A high priority is development of an antiviral drug which could mitigate against the side effects frequentlyseen after vaccination. Toward this end, we have tested selected antiviral compounds and pimznunomodulators for their antiviral efficacy against vaccinia virus.

12

..

Page 18: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

-. pLbli: healih reasons for developing antivirals eticc:ive against vac'xira vlrus

. iease. The use of live recombinant vaccinia virus strains which express immunoreactive epitopes

farm pathogenic agents, holds great promise as a means of immunoprophylaxis against a variety of human

diseases 2 5 . However, the risk of disease from the vaccinia vector itself remains substantially untested and

therefore potentially significant2 6 . Development of antivirals could provide an additional safety margin for 0

that time when such recombinant vaccinia vaccines are in wide use both in military and civilian populations.

The testing system itself becomes a valuable adjunct in safety tests of vaccine constructs. We have studied

such a testing system.

134 O

J S

. . , . . . . . . _ . . . .. .. ,... ... _ . .. . .- , , n n n . .... . . I n n n In l U n m m l I I I I

Page 19: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

T ! )i <,'y and evolution cf tne " lesion model used in our studies is as follows. Boyle 2 7 first described

tail lesions in mice infected with vaccinia virus and the effect of some antivirals in reducing the number of

tail lesions. DeClercq 2 8 compared polyanions with interferon to determine their effect on vaccinia virus

lesion formation on the tails of mice. Smejkal et a12 9 determined antiviral and interferon-induing activities

of three benzo(c)fluorenone derivatives in mice. Morever, they introduced the staining of tail lesions with

1% fluorescein and 0.5% methylene blue. Jacoby et al.3C studied the pathogenesis of lesions produced by

vaccinia virus by virological, morpholgical and serological methods. They found the primary lesions were

typical pocks characterized by sequential development of epidermal necrosis, vesicle formation and

ulceration, and by dermal inflammation dominated by mononuclear cells. Seroconversion was shown on

day 8 after infection.

Our studies have focused on variables which have not been analyzed in previous studies of the tail

lesion model including the effect of virus strain, mouse strain, age and weight on induction of tail lesions. 0

In addition, we have tested several different categories of antivirals for efficacy in this model.

3. Results

a. Vaccinia virus strains and tail lesions.

The number of viral PFUs required to induce a tail lesion was determined for each virus strain after 0

inoculation of the highest possible virus dose (Table 2a) or after a standard amount of virus at the lowest

possible comparative dose (Table 2b). The four virus strains tested in CDI mice were of markedly different

virulence as measured by induction of tail lesions regardless of virus dose. The wild type and the Lister

strains were clearly the most virulent requiring fewer PFUs to produce a lesion than the other virus strains

regardless of the magnitude of the inoculum. The 3PP strain was the only one in which the magnitude of the

inoculum had an apparent effect on the number of tail le.ions observed. After a low dose inoculum, fewer

PFUs (284, 102-5) were required to produce one tail lesion whereas after a high dose inoculum, 62,797

(104.8) PFUs were required to produce one lesion. We have no explanation for this anomaly. In contrast,

regardless of virus dose inoculated, the plasmid strain was of reduced virulence perhaps because it lacked a S

functional TK gene. 17,942 (104.3) PFUs were required to produce a single lesion with this construct.

Lower virus doses did not produce lesions. The Copenhagen strain (alternately designated "Koppe") was of

intermediate virulence requiring between 733 (102-9) and 995 (103.0) PFUs to produce a lesion. Since this

strain was of intermediate virulence, it was used in subsequent experiments.

14 0

0.

Page 20: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

& ,...... . i,. ~d Tahd t.M~Ifn5 (

, ,Y LLSiONS AFTER INFECThM'N WITH HIGH DOSE VIRUS.-.. ,LS STRAIN VIRUS TITER PF'S LESIONS PFU/LESION

(PFU's,.1) INOCULATED ,MEAN)

WILDTYPE 398 398 46t11 9

USTER 3.981 3,981 616 60

COPENHAGEN 63.095 63,095 86±14 733

PLASMID 25.119 25.119 1.4+1.4 17.942

3PP 25.119 25.119 0.4±.4 62.797

B. DAY 7 LESIONS AFTER INFECTION WITH LOW DOSE VIRUS.VIRUS STRAIN vIRus TrER PFU'S LESIONS FUrILESION

MMFUSiV.1) INOCULATED (MEAN)

WILDTYPE 398 398 46±11 9

USTER 3981 396 6:1.6 66

3PP 25.119 398 1.411.4 284

COPENHAGEN 63.095 398 0.4±.4 995

PLASMID 25.119 398 0.0 ,

b. Relation between vaccinia (Koppe) virus dose, neurovindence, and tail lesions.There were two questions for which we sought initial answers. First, was there a relationship

between PFU titer and mouse irncerelra neurovirulence and secondly, was there a linear relationship

between virus PFU dose and the mean number of tail lesions produced?For the Koppe virus, a PFU titer of 103.9 corresponded to a suckling mouse inwacerebral LDSO

titer of 103.7 thereby establishing for this virus strain a close relationship between infant mouse

neurovirulence and PFU titer. Dilutions of stock virus ranging from undiluted stock to 1:100 were tested for

lesion production after tail vein inoculation of young adult CDI mice (19-20 grams). The mean number oflesions actually counted ranged from 70±15 (undiluted virus stock) to 5±10 (1:100 virus dilution). R-

squared, after linear regression of virus dilution on the number of lesions, was 0.942. Thus, over this virusdose range, there was a linear relationship between PFUs and lesions counted.

c. Effect of mouse age and strain on induction of tal lesions.The effect of mouse age on tail lesions was determined by repeating the experiment described above

using groups of ten CDI mice of 19.5 grams and 34.6 grams mean weight inoculated with 103.2 PFUs (1:5dilution of the stock virus). The meian number of lesions counted was 51.2±13.8 (Yi-d = 6.91 1+9) for the19.5 gram mice and was 28.6±3.9 (0I' = 5.3±.4) for 34.6 gram mice. At a lower dose of virus (1:100

dilution of the stock virus), the mean number of lesions was 2.8± 1.1 (" = 1.6t3) and 4.8±0.75 (V'" -

15

* *-

Page 21: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

'9.5 ,ind 34.6 grain mice respectively. Although there was no strong evidence of -in age

• t. ",l subsequent experiments were done in young adult mice between 19-20 grams.

Four different mouse stains were tested for development of tail lesions using the range of virus

concentrations described above. The dose responses were calculated for each mouse strain and compared by C!paired t-test for significance of differences. No significant differences were observed among the mouse 0

strains tested at any of the time points (days 6,7, 10) used for counting of tail lesions. CD 1mice were used

in subsequent experiments.

d. Vaccinia virus antigen (Koppe and plasmid strains) and tail lesions.

Vaccinia virus antigen (Koppe strain) was localized by staining of transverse annular sections of tail 0

tissue by indirect immunofluorescence. In the earlier stages after inoculation (up to day 3), antigen was

found only in isolated foci of cells surrounding individual hair follicles. These locations were extremely

focal and required examination of nearly the entire tail. Between days 3-5, antigen was found less

frequently i. this band of cells but appeared more diffusely distributed in the dermis. On days 6-8, antigen

could be detected in focal areas of the epidermis in massive concentrations. At this time and in the area of

antigen concentration, infiltrating inflammatory cells were observed. A similar early distribution of antigen

was detected after inoculation of the plasmid strain. However, antigen spread beyond the dennis was never

detected. Thus, development of tail lesions was associated with spread of virus antigen from an early focus

of infection in the dermis to a more pronounced. but still focal, infection of the epidermis. Virus antigen 0

was not prominently associated by immunofluorescence with sebaceous glands in the tail, muscle tissue, or

other organs examined including kidney, lung, liver and brain.

e. Active chemotherapeutic agents in the vaccinia tail lesion model.

Most drugs tested in thit model gave VR scores of 1.0. Drugs with VR scores of 1.4 were in the 0

highest 90% percentile of all drugs tested. The potency of the chemotherapeutic antivirals which were

active in inhibiting formation of mouse tail lesions following vaccinia virus infection (Koppe strain) is

summarized in Table 3. Active compounds include ribavirin, an A, adenosine N'-oxide and several of its

analogues. The relationship betwee in vitro activity and in vivo activity was excellent for adenosine N'-

oxide, but such cosrespondene was not absolute for other compounds. For example, ara A in cell culture

was not particularly poter. .VR of 0.8 to 1.4. (A VR of 1.0 means no drug effect.) Yet, in the mouse

model, depending upon -.e. - l and the virus dose, the VR score ranged from 0.9 to 3.7

suggesting that the drug , : :ful in animals than the tissue culture data suggested. Despite this

promise, the therapeutic index of arM A .. low (6-40) and alternative chemotherapeutic agents might be more

useful. The therapeutic index is a ratio of efficacy to toxicity and is a measure of safety. Compounds were

identified with a high therapeutic index, a high VR score in both cell culture and the tail lesion model.

Included, in particular, is analogue of adenosine-N'-oxide such as AVS 4224, a pro-drug with a substituent

3.4-dimethyl group. P

16

00

€ • •• • • •• ....e : I m l

. . . i ... .... ... ... ,i i H i ,i* 4J

Page 22: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

rabli 3. Summary of Acive Cbemotheraptulic Age~nts____________ ____________In the Vioccinia Virus Tail Lesion Model________

AVS U 1 Therapeutic J VR VR I Name or type of(thane culture) culture) We~on model lot______

I QSS 72-5650 3.4->7.2 1.0-2.3 adcnosine-N'-oxidePro-drop of adeomiae.N.ouide:

1986 >.487 3.4->7.2 1.0-1.3 substiattnL- 3-________ _________________ methyl

3607 992->1000 >2A.>2.7 1.2-1.5 substituint:________ ________ _________ aipla-medivi

3679 11167-10000 2.8-3.3 0.6-2.0 nsuitent 4-________I I_____ _____ _____ medioxy

42n4 >99, > 1762 >2.&.2.8 =2.-3.6 sibsziwusnt:_________ ________ _________ _________ 3.-dimethvi

Compounds other titan ademomae-NI-oide analogues:

1752 6-40 0.8-1.4 0.9-3.70 adenosinearabinoside

________ _______ __________________ (ars A)001 NtND 0.7-2.0 ribavirn7118 ND ND 1.4 rifampicin1214 50,119 ND 4.3 S'-deoxyadenosine

________ I_ I______I_ dialdehyde1tND me w r. dome.$Tl. usual drug schedule was one dome pno to vium inoculation follwe by 5 subsequent daily domes Mwi mewi number of tailesions was equal to tie aveag offXX wbere Xa is the number of lesions per moms. Tie viru raing (VR) was a maoo of die

average number of lesions in mice given DMEM as control divided by she average number of lesions in the drag-tteaed anials.

f. Immunomodu-- rs in the vaccinia virus tail lesion model.

An alternative antiviral therapy involves the use of imununomodulators to inhibit virus spread.Combined therapy could be useful in mitigating against selection of drug-resistant virus strains. Fiveimmunomodulators which have been widely studied elsewhere31, 32, 33, 34. were also studied in thepresent experiments. Of dhe five, four were found to be efficacious in significantly inhibiting tai lesions in Idie vaccinia virus mouse model. These include quinolinamnine (AVS 1018), ampligen (AVS 2149),recomrbinant 112 (AVS 5079), and poly ICLC stabilized for injection (AVS 1761). At the doses used.exogenous recombinant 112 had no effect on tail lesion formation. Tails from selected groups of micetreated with quinolinamine and anipligen were examined by indirect immnunofluorescence for detection ofvaccinia virus antigen. Although antigen was found in the dermis in the early days after infection, virusspread to the epidermis was not found in contrast to control mice. These results are in accord with diehypothesis that ,esimx formation is associated with spread of infection from the dermis to the epidermnis. In

.,trca:m.nt wkf. these two ammunomodulators appeared to have specifically inhibited virus growth

in epidermnal tissue.

17

Page 23: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

Tat le4. Immanomodulatom in the Vaccinla Vlruo Tal Lesion Model

VIRUS CONTROL DRUG Vhf PDRUG DOSE NMAN MEAN (t.TEST

Drug DOSEt Iogle PIFU'S LESIONS LESIONS CONTROL PAIRED(KOWE , ONE-TAIL)

UI m DRUG

qurAolmmmi 300 umwA 3.7 7.375t 0.333t 22.1 0.001

1.034 .167

quwin 100mgih 3.8 7AC4t 3A03t 2.34 0G 3

.708 -W

IL 2. 2.5 3a 104 3.8 7.60± 6473:t 1.17 0.526(mcombiust mits .708 .724

recoaI-m IO5 Wi 3.8 7.64± 2.514 2.9 00$M

.706 .915

ailioa 100 ams 3.8 7A (: itl 4A SAM63

.708 .62mliSelm so mgms 43 18A4 30 3.41 S

25 alwn 43 18A4 331 31 6.001

poly XM 20 Mm 3.8 7.604t I.603t 4.74 001

subized .706 .503fl u drg schele was r dons pew to vimoculAiion folowed by 5 t.asequ daily dosmhe mem umber of ad leo miso oal i do aon ofX woe Ka is Mle somber of leion per moo

VIle virtu o (Vt) mu a fho of theims i mr - DMF.M l d b e vanmber at leuionm dom hAesmd atls

4. Dbhvm Amd CmndusaueVaccinia virus sains mid constucts differ grady in die number of PFUs required to produce tail lesions inthe vaccinia vires mouse mode. The paldogenesis of lesion formaton appeared to involve virus spreadfrom an initdal focus in specific cells surrounding hair follicles to odier concentrated areas of the dermis andfri.ally, at the time of lesion development, to the epidermis. Antivirals which suppressed tail lesions, to a

greater or lesser degree, included ara-A, ribavirin, rifampicin, adenosine N'-oxide and selected analogues.

Inmunomodulators, including ampligen and recombinan: interferon suppressed lesions at very low doses.

Spread of virus infection from the dennis to the epidermis was inhibited as determined by

18 4

* 0I I I I --

Page 24: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

wS

....: . iL.Ne :tudies in the tul lesion model have suggctcd drugs which coulJ be t1tcd ,

.. . vir.i nuxiels of vaccinia virus inicction. In addition, these studies provide additional data on a

model which may be a useful adjunct in safety testing of recombinant vaccinia virus vaccines.

III. TESTING IN PRIMATE MODELS S

A. YELLOW FEVER AND ANTIVIRAL DRUGS

L Background and siteumut of the problemPreventive control of human disease by an antiviral subsune can be bent considered when it has a relativelybroad spectrum of activity. Otherwie, the time delay involved in identification of the causative agent mayrender application of an antviral impractical for prevention. Ribavirin is such an antiwial drug. In tissueculture and in rodents, it has broad antiviral activity. Howeve, efficacy against flavivinas infection has notbee convincinly demonstrated in labaromy infected prmate. One gol of the work in this section was to

4 test ribavirin for antiviral activity against yellow fever virus infection.

2. Summary of yellow ker-r vlrm tea.A. Dag reamwm at d. me o fand q*a virat ewsure.

Squirrel monkeys, Sa iicunew , were inoculated subcutaneously with 1000 LDSO of yellowfever virus, Daka 1270 strain, an the 86 moue brain isse piaage. Ribevirin (50 mgikg) was given 5subcutaneously at the time of virs inoculation ad daily theeaftr. Drug uaeomen had not effect onmorlity. One of two virus uifecwd annls survived whm I of3 drug oauWd animals survived. Thegeometric mean time to death for the onmol animals was 9.2 days compard to 9.4 days for drug-treatedanimals. rhe mean peak virnmna level (day 3 after virs inoculation) in €onrol animals was 6.8 lol PFU in * *contrast to S.I in treated animals. Surviving control uaimals had slightly higher HAI antibodies butneutralizing antdbodies were copnmable. Alkaline phospheaa levels were elevated in the conu animals,but only on day 3. This change was not conelaed with death. One conrol animal tueaed with drug alonedid not seauconvent.

b. Dn weamm bfore and qter vim cw p eIn a second experiment with squirrel monkeM drug aunet (50 mg/kg) was initia 3 days prior

to virus exposure and connudm for 8 days afier virus irecton. Prolonged survivaL but no statisticallysignificat reduction in mortality was seen in nbvimn-u d animls. Five of six (83%) animals infectedwith ellow fever virus alone died whereas 2/5 (40%) animals treated with ribavirin died. The geometric S

mean tme to death for control animals was 6.1 days and for armed animals, 15.6 days. Viremia levels wersignificantly reduced (84-999% reduction, p- 0.01) in those animals which were tet with ribavirin.

One very slriking difference between control and treated animals was observed byimmunotluorescent staining of tissues from moribund animlL Viral antigen was not detected in the central

nervous system in untreated animals. In contast, the central nervous system of drug-rwated animals washeavily infected with yellow fever virus as determined by immunofluorescence. Virus antigen was would inneuronal cell bodies and in processess projecting f'nm the cell body as these processes transversed brain

19

Jo 0 0 0 0 0 6l

Page 25: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

.iu~i:Nt ,lttatinmi iven nbavirin died of a syndrome different from that oi untrcated

. 1 type of altered pathogenesis, the late-stage entry into the CNS. has been seen in other vtrus-::ccted primates treated with ribavirin.

Alpha-beta interferon levels were low in both groups of animals with no differences observed

between the two groups. One untreated monkey (87) had detectable levels (1:150) beginning on the Rih day

after infection and lasting until the seventh day. Interestingly, this was a surviving monkey. However.

among nbavirin-taed animals, only one of three surviving animals developed detectable levels of

interferon (day 6-8). •

c. Concv im and hypohesisacarly, interfem development was Ime developing in fibaviuln-teamd animals suggesting that

interferon-stimulaIng dup or oherum m drup miglh be of beei in preventing death in

bavin-ated animals. This hypotes lead us into stu ies of izmunmodlam.

L YELLOW FEVER AND IMMUNOMODULATORS

L Dackgromd ad Unmue, of the probkmIn coasmt 0a conventional antiviral dha inhibits a pa cular enzyme nm universally Feete among

all vuses, i. mm luw cm provide prcon against a broad spactrm of vital infectons by Sevoking dthe p i ad nonpeciic antiviral deft en o( the hoL Thaerfom the ideny of the etioloicagetismarelev t rthem oftheemmem ieP C1246738 (AVS 1968).& aa linebydrcMoride (A), evokes cmplete ipmectidm in ,m - aains viral infections representing 3 out of 4

arbovir f Jies includinlg a flavivim. In addition, CAmpomid AVS 1018, a quinolbamine (QA) free

base. adnlavae in 2% lactic aid whvioa evoked exce protection ginst lethal vid infecionsrPePremmPdg bath Buy vAw md Flavivhw fmdlies. Therfore, these two drup were umd in d yellow

fever; pim -e model for eficacy in preveoft disease sympom.

2. RiIbm wih CL246736 (All, dedle) 0

The prophylactic posential of AH was evaluaed in squirrel monkeys that have natura resistance to

the memmopic mani of yellow fever virs infection Pe e the circulating viru titer was the mainindicasr of e amvind reactivit of t omoud. Orally anlsmed AH was given 24 h prior to

yellow fevr vm challenge. The ompomnd evoked a -d antiviral activity by entirely reducing 3 logsof circulating viru tim. The compoud was m ective we e m me was initiated 24 h

postinectio

& AN and rlm*ox

Srm from animals meaed with AH was assayed for alpha/beta interferon. The details of the

7,:e-rdure were presented in Annual Riports (1989. 1990) and ae summarized as follows. Test sera were

di!uted ten-fold in DMEM and rdjusted to pH 2 by addition of HO. held for 24 hours, and readjusted to pH

7.3. Half.log dilutions of each serum we.e incubated with Veto cells for I8 hours prior to infection of cells

with VSV-Indiana using a dose which was 100 PFUL

20

0S

0 000.

Page 26: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

. .... , ..... c u nn c s oI interfcron in monkeys until s.vcral dowcs of drug had

.,i. i.,., ,a Jd - .ud 1. Twenty-four hours after the first dose of drug, only one of four animals 4

...J ,i.cctjblc interferon. By day 2. all animals were positive. Interferon levels remained high through day

14. te end of the treatment schedule. These data suggest a reason for lack of drug efficacy when treatment

began after virus inoculation. At leas! two days of drug treatment arm required to induce detectable

int rferon.

b. Resid of Combined Treabment with AH and Ribavirin

Addition of AVS #1 to the experimental protocol resulted in an initial depression in viremia,

although later vinmia levels wen uniformly higher than those seen in animals treated with AH alone.

Drug treatment beginning after virus exposure produced no detectable differences from control

animals exposed to vis alone. Vinemia levels wo sifla in the two groups.

3. Raulta of Treatmme with Qutninamine (QA) 0

In cynomolgous monkey when QA was administemd (orally) beginning one day before infection

with the Asi stain of yellow fever virus and given every other day for 14 days, 8 logs of serum virus titer

were completely reduced. Although two of the three untreated infected monkeys died of yellow fever virus

ianfecion, all four Ied monkeys survived the yellow fever vins challenge.

Tae S. Prophylacti ffiacy e Free lase (AVS.101S) la Yellow Fever Viras.Infected CynmolgusMonkeys. ,Group I Tr"unen No. of Animals thaI Survivedioud -

I Vinus control 1/32 Viust + 4/43 Drug conol 2/2

8103 PFU yelow kvw vm ij Lm d a ty 0.620 m.hqg @m 1 oMy 1. 10 mgflk m ths O.4A.,10.12.14 ily m 2% KmmL

AL QiAa im (QA) and Inwfez A'.

It may be significa that imerferon in the serum of virus-infected, placebo-reated animals ncver

exceeded 100 unitsut any time 4uring the study. Those animals which died had low levels on day I and

never again during the study except for one monkey which had interferon at the time of death (day 5). All

virus-infected monkeys Oven drug in the study had detectable serum interferon on days S. 7, 8, 10, and 12.

It seems appawe from these resuld that the combination of virus-infection and drug treatment resulted in

-. r- e nt and prolong-d antcrf.-ron response than ",as observed in animals giver drug only.4 Aiteraubvely. the kinetics of the interferon response may have differed between those animals given both

virus arJ drug and those given drug only.

21

40

! "4

0 S0

Page 27: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

0 . .,.~,~~,. anine a Yd k'ellow Fever Vruas Viremia.;.c xt of d'ug-tzretment was most obvious on the suppression of viremia. As measured in Vero cells,

t.e drug effect was dramatic. Only the virus-infected animals had a viremia. Only one virus-infected, drug-

treated animal had a viremia, and it was transient, detected only on day 3 after infection. Viremia was not

detected in the serum of other virus-infected, drug-treated animals. As expected, animals which received

drug only did not have a viremia. A

C. VACCINIA VIRUS IN PRIMATES

Two cynomolgous monkeys which survived the yellow fever virus study were held for three weeks prior to

multi-site intradermal inoculation with wild-type vaccinia virus (5 sites, 0.1 ml, Koppe strain) describedpreviously repor These animals wen not producing detectable interferon and their blood chemistries and

CBCs were normal at the time of virus inoculation. Lesions, which were too numerous to count accuratelybecause of overlapping, developed on the chest at the site of subcutaneous inoculation seven days after i

inoculation resolved between 10 to 12 days after inoculation. Smears from vesicular scrapings done on dayseven were positively stained by immunofluorescence using vaccinia virus antibody (and CCHF viruscontrol antibody) prepared in mice. Multinucleate" jant cells containing intranuclear inclusions were alsoidentified in lesions on day 7. Infectious virus (between 103 and 104 TCID50 per ml, 6 samples, 3

replicate each) was isolated in Veto cells from scrapings taken on days 7 and 8 only.

I. Effect of ribavirin on vaccinia lesions in primatesSix weeks after conclusion of the yellow fever virus experiment, four surviving monkeys were inoculated

subcutaneously with vaccinia virus in the dosage described above. Two animals were treated orally withribavirin, 100 mg/kg daily beginning one day before inoculation of virus. On day 7, lesions appeared on the 0

chest of both the control and drug-treated animals. Smears from vesicular scrapings were antigen-positive

by immunofluorescence on day 7. Multinucleated giant cells were not seen in the ribavirin-treated animals.Infectious virus, and the virus titer from the scrapping was lower (6 samples, 3 replicates each) in the

ribavirin-ureated animals (mean titer, 101.7 /ml) as compared to the untreated controls (mean titer, 103 .3/ml).

D. DISCUSSION AND CONCLUSIONS

Ribavirin was the antiviral studied in the yellow fever virus primate model. The therapeutic index of

ribavirin in vitro for flaviviruses is lower than for bunyaviruses; no antiviral effect has been demonstrated

in rodent flavivirus models. In other experiments, prophylactic administration of ribavirin to monkeys failed S

to abrogate dengue viremia3 5 or to influence the course of yellow fever infection [Peters et al., Proc, Int.

Symp. on Yellow Fever and Dengue:, Rio de Janeiro 1988; Oswaldo Cruz lnst 19901. In our studies,cibavirin treatment markedly reduced viremia in monkeys although later invasion of the central nervoussystem was seen. This lead to a study in which we combined ribavirin treatment and immunomodulator

therapy. Combined treatment did not reduce viremia significantly whereas treatment with either ribavirin or

AH individually did reduce significantly vireruia. This failure may result from the complexities ofimnmunomodulator therapy (reviewed in [341).

22 0

S 00 0 0

Page 28: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

& nX~~iulaturs. All and QA, studied in this model are not double-stranded RNA s; 1

ai4'arently stimulate a variety of biological functions, induction of INF being -nost obvious. This isLxi, ed upon the fact that administration of arti-INIF serum abrogates their effect34. A two-armed pathway

appears to provide the antiviral mechanism of action. Via the upper arm, these imrnunomodulatorsstimulate INIF release by macrophages, T lymphocytes, and fibroblasts. The INF in turn activates enzymeswhich inhibit viral proliferation. Via the lower arm, INIF stimulates the specific and non-specific cellular or

humoral immune reactivity. In our squirrel monkey studies, ribavirin not only reduced viremia, but it also

delayed the onset of neutralizing antibody responses. Furthermore, only 1/5 animals treated with bothribavirin and AH (AVS 1968) had neutralizing antibody on day 7 whereas 415 placebo and 515 treated with

AH alone had antibody on day 7. T'hus, there may be antagonism between ribavirin and immunomodulatoreffects in primates. This is a question which should be further investigated.

The vaccine primate model deserves further exploration. Ribavirin treatment did not eliminatelesion formation. However, the virus titers in vesicular scrapings from the lesions were lower than inuntreated controls arid multinucleated giant cells were not seen in the scrapings. Whether giant cells are anormal aspect of vaccinia. infection is unknown from the available literature; such cells are often seen inherpes virus infection. The virus isolated was vaccinia virus as determined by neutralization tests; we hadno evidence of a concurrent herpes infection as determined by immnunofluorescence tests. In conclusion,while ribavirin trenert did not eliminate or reduce lesion formation, it did reduce dhe virus titer in the

vesicula scrapings.

IV. LITERATURE CITED

IGear MRS. Congo Fever4Crimean-Coogo Hemorrhagic Fever. In: Oea, J.H.S. ed. CRC Handbook of Viral and Ricketisial2 Hemonlagic Fevers. Florida: CRtC Press 196; 121-129.Casals 1; Tignor (1K. The Nairoviros genus: serological relationship. Ingervirol 1980; 14:1l44-147'.

3 Tignor OH; Smith Al, Cas J; Ezolkoli CD% Okoli J. Close relationship of Crimanen hemorrhagic fever-Congo (ClIP-C)stana by neinralizig sibody assays. Arm I Tr Med Hyg 19M; 29.676.685.

4 Hoogsraml ff. The epidemiology of tik-borne Qimean-Congo hemnagic fever in Asia Europe and Africa J Med Entomol4 197W; 15(4):307-417.

5 Watts DM; Ksiara TG; Linhicum KJ; Hoognraal H. Crimean-Congo hemwrtgic fever. In: Monath, 1? ed. The arbovinises:epidemiology and ecology. Vol 2. Plorift CRC Pnes 1981; pp. 177.222.

6 Van-Eeden P3; Vai-Eeden SF; Joubert JR; King JS; Va,-de-Wal BW; Michell WL A nosocomial outbreak of Crimean-Congohwnorrhagc fever at Tygerbeig Hospital. Part 11. Managementofatient. S Air Mad I 1985; 68(10): 718-721.

7 Van-de-Wal BW; Joubert JR; van-Eeftn Pl: King 13. A nosocomial outbreak of Crinean-Cango haernorrttagic fever atTygcrberg HospiWa. S Air Mod I11915; 63(10):729-732.

4 8 Swanepoel R; Leman P; Abbott JC: Sunt F); Orobbelew AA. Epientiology. diagnosis, clinical pathology anid treatment of

Criwnean-Congo Hamnorrhagic Feve (CCHF) in South Africa. In: Proc. Vilth Ind Cong Virol, Berlin. 1990; Abstr P70-003, N*9 Mosd TP. Approaches io the prophylaxis and therapy of the viral hemorrhagic fevers: ribavirin. in~terferon, and antibody.

Current Opinion in Infectious Diseases In Press.SGre RR. Search for anuvuia agents. Annu Rev Microbiol 1979; 33:335-53.

4 ~ Centers for Disease Control, National Institutes of Health. Arboviruses. In: U.S.Depannment of Aealth and Human Services.ed. 1984; Biosafety in Microbiological and Biomedical Laboratories. Washington: U.S. Government Printing Officc:77-78.

12 Tignor GH. Prepared statement. In: Committee On govemnmentai affairs. United States Senate, 100th Congress, 2nd Session.ad Hearings before the subcommit on oversight of government management. July 27a28 1988. Washington- USGovernment Printing Office 1988; pp 179-186.

23

0000 000

Page 29: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

1

,;%6.cd eP%; Huffman J H. Use of disposable micro tissue culture plates for antiviral and interferon induction slu.ies. ApplNI iriobiol 1971; 22:797-801. I

14 T..or GH; Shope RE; Bhau PN; Percy DH. Experimental infection of dogs and monkeys with two rabies scrogroup viruses.

Lagos bat and Mokola (TbAn27377). Clinical. serologic. virologic, and fluoemscent-anibody studies. J InfcA Dis 1973;12:471-478.

15 Ho M: Springer TA. Mac- I antigen: Qua',itative expression in macrophage populations and issu, and immunefluoresccnt

locali.auon in spleen. . Immunol 1982; 128:2281-2286.16. Spitz M; Spitz L. Thorpe R; Eugui E. lnzrasplaic primar immunization for production of monocional antibodies. J

Immunol 1984; 7&k39-43.17 Kohler G; Milstein C. Continuous cultres of fused cells secreting antibody of pre-defined specificity. Nature 1975; 256:495.

497.18 Mason PW. Maturation of Japeneee ecephalitis virm glycoprolein produced by infected mammalian and mosquito Cels.

Viral 1999; 169.354-364.19 Laemmh UK. Clavage of xtructrawl proens =dui the assembly of the head of bAwph T4. Nature (London) 1970;

227:680015.20 Ruebner. B.; Miyai IL The Kupffer cell reaction in murine aid human viral heptitis with pwficular reference so the oigin of

acidoplilic bodies. Am J Pashol 1962; 40: 425-435.21 Robins R. Syndiec antiviral agems. Chemica arid Engineering News 196; 64(4):135-146.22 Shope RE. Camme OR. Further studies on the srological relationship of group C ahIopod-borie viruses mian

application of ds relatioships to rapid identifcmion of types. Am J Trop Med Hyg 1962; 11(2): 283.290.23 Huggins J. W. Prospes for treatment of viral hemorrhaoc feven with rilavirin. a broad-spectrum antiviral drug. Reviews

of Infectios Diseases May-June. 1989.11, Supplement 4:S750-761.24 Lehmw-J)e, F. 1g62. The Mome in Biomedical Reamreh. Vol IL Diseases. Edited by H. L Foster. 3.D. Small amd 1.G.

Fox. New Yod. Academic Prea.25 Fisher-Hoch-S.P., J.P. McCormick. D. Auperin, B.G. Brown, M. Cator, G. Pem S. Ruo, A. Comty. L Brammer, & S.

Bane. 1969. Proection of dtess monkeys fom fatal Lssafever by vaccinsation with a ecombiimt vaccinia virusconsuming the Lma virus glycopsusei gen. Prc. NaL. Acad. ScL U.S.A. 86(1): 317-321.

26 Hruby,D.L 19. Prent and future applications of vaccmia vias ma vector. Vet-Pmiwd. 29(2-3): 281-292.27 Boyle, JJ., R.F. Haff. & R.C. Stwam 1967. Evaluation of amivira compounds by suppiusion of tail lesions in vaccinia-

infected mice. Antimicrobial. Agents aid ameods py-1966, 536-539.28 DaClarcq. E. & P. DeSoier. 1961. Effect of interfeo. polyacryiic acid, and polymediacrylic acid on tai lesions in mice

infected with varcina virus. Applied Microbiology 16:1314-1319. I29 Smejl. F.. D. Zele, J, . Krepelka, & I. Vancurova. 1964. Derivatives of bei(c)fluorem. VI. Antiviral and intmreron-

inducing activities of three bazo(C)fluonone derivatives in mace. Acta. Virol.29: 18.30 Jacoby. R.O.. P.N. Blats.ELA. JohnsonF.X. Paumro. 1963. Padiogenesis of vaccinia (IHD-) virus infection in

BALBA/cAnN mice. Lab. Animal. Sci. 33:435-441.31 Kende, M.. H. W. Lupton W. L. Rill. H. B. Levy. & P.O. Camonico. 19L. Enhanced therapeutic efracy of poly (ICLC)

and ribavirmn combirations against Rift Valley fever virus infection in mice. Anadmicibial Agents and Chemotherapy.(1987). 986-990

32 Levin, S. Interferon in acute vind infections. 1963. Eur. J. Pediar 140- 2-4.

33 Ho. M. 1962. Recent advatces in she study of interfron. PhmacoL Rev. 34:119-129.34 Kende, M.. H. W. Lupton. & P. G. Canonico. 1963. Treatment of experimental %'"al infections with immunomodu; -s.

Advces in Biosciences. 66. pp5l-63.35 Malinoski, FJ., Hasty, S.L. Ussery. M.A., and Dalrymple, I.M. Prophylactic rbavrin treatirmt of dengue type I infection inrhesus monkeys. Anitivral Res 1990. 13:139-150.

244i

4 0 0 • 0 0 .- 4

Page 30: TO - DTIC · 1202 SliM, P.O. Box 3333 *Nev Haven, Connecticut 06510-8047 9.SOSRIPIGMONiT ORING AGENCY NAMIE(S) ANO AOORESS(ES) 10. SPONSORING IMONITORING U.S. Army Medical Research,

..... L0h, CAL BIBLIOGRAPIHY OF ALL PUB3LICA I ,

A.L PVBZCA2IONS FFIOM 7ME ZABORATORY

Tlgnor. G. H. and Hanham. CA. Ribavirin efficacy in an in vivo model of Crimean-Congo hemorrhagic fever virus (CCHF) infection. Antivlral Research (1993) InPress

Tignor G. H.. Casals. J.. Shope. ILE. The yellow fever epidemic in Ethiopia. 1961-1962: retrospective serological evidence for concomitant Eboia or Ebola-like virusinfection. Transactions of the Royal Society of Tropical Medicine and Hygiene.87: 162. 1993. (supported by previous contract DAMD17-81-C- 1156 and 17-90-Z-0020)

Tignor. G. HI., Kende, M. and Hanham. C. A. Chemotherapeutic Prevention ofComplicatlns Caused by Vaccinia Virus-Vectored Immunogen. Annals of the NewYork Academy of Sciences. 1992. 334-343

Lee. Ch/a-Jean Antiviral Drugs In a Model of CCHF Virus Infection. 1989. MPH Essay.Department of Epidemiology and Public Health. Yale University School of Medicine.

A. G M 'VUBUMATOMS WWCR PRESWW DATA COWIAT7 N =S CONTRACT

Huggins: J. W. Prospects for treatment of viral hemorrhagic fevers with ribavirin. abroad-spectrum intivral drug. Reviews of Infectious Diseases May-June. 1989:U. Supplemert 4:5750-761.

Kende, M. Treatxr,-.t Srategies for Human Arbovirual Infections Applicable toVeterinar, Medicine. Annals of the New York Academy of Sciences. 1992. 297-313. * 6_

VT. PERSONNEL SUPPORTED AND DEGREES GRANTED

A. FACULTY:Dr. Gregcry H. TignorDr. Robert E. ShopeDr. Thomas Burrage

B. POST-DOCTORAL FELLOWS AND ASSOCIATESDr. Catherine A. HanhamDr. Feisha Zhao

C. STUDENTS AND DEGREE GRANTEDChia-Jean Lee, MPH

D. RESEARCH ASSOCIATES SRuben CedenoJames Washington

25

4 00 0 0 06