antiviral therapy: current concepts and practicesantiviral therapy 147 oh idoxuridine...

37
CLINICAL MICROBIOLOGY REVIEWS, Apr. 1992, p. 146-182 Vol. 5, No. 2 0893-8512/92/020146-37$02.00/0 Copyright © 1992, American Society for Microbiology Antiviral Therapy: Current Concepts and Practices BONNIE BEAN Departments of Pathology and Medicine, Humana Hospital-Michael Reese, Chicago, Illinois 60616 INTRODUCTION .................................... 146 CELLULAR AND VIRAL REPLICATION .................................... 146 ANTIVIRAL AGENTS .................................... 149 Amantadine and Rimantadine .................................... 149 Ribavirin .................................... 153 Vidarabine .................................... 154 Acyclovir .................................... 154 Ganciclovir .................................... 156 Foscarnet .................................... 157 Zidovudine .................................... 158 Didanosine .................................... 159 Investigational Antiretroviral Agents .................................... 159 IMMUNOGLOBULINS .................................... 160 IMMUNOMODULATORS .................................... 162 PROBLEMS OF ANTWIRAL THERAPY .................................... 164 Resistance .................................... 164 Latency .................................... 165 Immunosuppression by Antiviral Agents .................................... 165 PROSPECTS FOR THE FUTURE .................................... 165 Liposomes .................................... 165 Combination Therapy .................................... 166 Computer-Aided Drug Design .................................... 166 Role of the Clinical Microbiology Laboratory .................................... 166 CONCLUSIONS .................................... 167 ACKNOWLEDGMENTS .................................... 167 REFERENCES .................................... 167 INTRODUCTION Interest in antiviral chemotherapy began in the 1950s, when the search for antitumor agents generated a great deal of interest in DNA synthesis inhibitors and produced a number of compounds capable of inhibiting viral DNA synthesis. Antiviral agents were first successfully adminis- tered to patients in the 1960s, when Bauer prevented disease by giving thiosemicarbazone (methisazone) to patients ex- posed to smallpox (20) and Kaufman greatly improved the healing of herpes keratitis by treating patients with topical idoxuridine (233). Progress was slow, however, because of the difficulty in finding compounds capable of inhibiting viruses while at the same time leaving host cell functions intact. With the late 1970s and early 1980s came develop- ment and marketing of acyclovir, the first antiviral agent nontoxic enough to be of value in treating a wide range of herpesvirus infections in ambulatory as well as seriously ill patients. The late 1980s and early 1990s are seeing an explosion in antiviral agents and in approaches to antiviral therapy that is fueled, in part, by the AIDS epidemic. This article reviews the basis of antiviral therapy, the agents themselves, the problems to be solved, and prospects for the future. CELLULAR AND VIRAL REPLICATION Like antibacterial agents, useful antiviral agents must have certain properties. They must reach their target organs, be active intracellularly as well as extracellularly, and be metabolically stable. Most important, they must inhibit virus replication without disturbing host cell function. Because viruses reproduce intracellularly and use host cell metabolic machinery in doing so, it was thought for many years that specific interference with viral replication was impossible. Experience with early antiviral compounds corroborated this view; the drugs were either too toxic or insufficiently potent to be of use (460). Two drugs that illustrate this well are idoxuridine (5-iodo-2'-deoxyuridine) and trifluorothymi- dine (5-trifluoromethyl-2'-deoxyuridine) (Fig. 1). Idoxuri- dine was first synthesized by Prusoff in 1959 (363) and was subsequently shown to inhibit a number of DNA viruses (for a review, see reference 364). When administered systemi- cally to patients, however, it did not decrease the risk of death from herpes encephalitis and it caused myelosuppres- sion in almost all patients who received it (40). On the other hand, the drug was effective and much less toxic when administered topically to patients with herpes keratitis (233). It is still used for this purpose. Recently, there has been a resurgence of interest in topical use of idoxuridine coupled with dimethyl sulfoxide for treatment of herpes labialis (450). Trifluorothymidine, first developed as an antitumor agent, was found to inhibit herpes simplex virus (HSV) in vitro, but its selectivity index (ratio of drug concentration causing cellular toxicity to that needed for viral inhibition) was very low compared with those of other agents (99) and it was not developed for systemic use. Like idoxuridine, it is used for topical treatment of herpes keratitis. Despite their shortcomings, these drugs were important in 146 on May 24, 2020 by guest http://cmr.asm.org/ Downloaded from

Upload: others

Post on 23-May-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

CLINICAL MICROBIOLOGY REVIEWS, Apr. 1992, p. 146-182 Vol. 5, No. 20893-8512/92/020146-37$02.00/0Copyright © 1992, American Society for Microbiology

Antiviral Therapy: Current Concepts and PracticesBONNIE BEAN

Departments of Pathology and Medicine, Humana Hospital-Michael Reese, Chicago, Illinois 60616

INTRODUCTION .................................... 146CELLULAR AND VIRAL REPLICATION .................................... 146ANTIVIRAL AGENTS.................................... 149Amantadine and Rimantadine.................................... 149Ribavirin.................................... 153Vidarabine.................................... 154Acyclovir.................................... 154Ganciclovir.................................... 156Foscarnet .................................... 157Zidovudine .................................... 158Didanosine.................................... 159Investigational Antiretroviral Agents .................................... 159

IMMUNOGLOBULINS.................................... 160IMMUNOMODULATORS.................................... 162PROBLEMS OF ANTWIRAL THERAPY .................................... 164

Resistance.................................... 164Latency.................................... 165Immunosuppression by Antiviral Agents .................................... 165

PROSPECTS FOR THE FUTURE .................................... 165Liposomes .................................... 165Combination Therapy .................................... 166Computer-Aided Drug Design.................................... 166Role of the Clinical Microbiology Laboratory.................................... 166

CONCLUSIONS.................................... 167ACKNOWLEDGMENTS.................................... 167REFERENCES.................................... 167

INTRODUCTION

Interest in antiviral chemotherapy began in the 1950s,when the search for antitumor agents generated a great dealof interest in DNA synthesis inhibitors and produced anumber of compounds capable of inhibiting viral DNAsynthesis. Antiviral agents were first successfully adminis-tered to patients in the 1960s, when Bauer prevented diseaseby giving thiosemicarbazone (methisazone) to patients ex-posed to smallpox (20) and Kaufman greatly improved thehealing of herpes keratitis by treating patients with topicalidoxuridine (233). Progress was slow, however, because ofthe difficulty in finding compounds capable of inhibitingviruses while at the same time leaving host cell functionsintact. With the late 1970s and early 1980s came develop-ment and marketing of acyclovir, the first antiviral agentnontoxic enough to be of value in treating a wide range ofherpesvirus infections in ambulatory as well as seriously illpatients. The late 1980s and early 1990s are seeing anexplosion in antiviral agents and in approaches to antiviraltherapy that is fueled, in part, by the AIDS epidemic. Thisarticle reviews the basis of antiviral therapy, the agentsthemselves, the problems to be solved, and prospects for thefuture.

CELLULAR AND VIRAL REPLICATION

Like antibacterial agents, useful antiviral agents musthave certain properties. They must reach their target organs,be active intracellularly as well as extracellularly, and be

metabolically stable. Most important, they must inhibit virusreplication without disturbing host cell function. Becauseviruses reproduce intracellularly and use host cell metabolicmachinery in doing so, it was thought for many years thatspecific interference with viral replication was impossible.Experience with early antiviral compounds corroboratedthis view; the drugs were either too toxic or insufficientlypotent to be of use (460). Two drugs that illustrate this wellare idoxuridine (5-iodo-2'-deoxyuridine) and trifluorothymi-dine (5-trifluoromethyl-2'-deoxyuridine) (Fig. 1). Idoxuri-dine was first synthesized by Prusoff in 1959 (363) and wassubsequently shown to inhibit a number of DNA viruses (fora review, see reference 364). When administered systemi-cally to patients, however, it did not decrease the risk ofdeath from herpes encephalitis and it caused myelosuppres-sion in almost all patients who received it (40). On the otherhand, the drug was effective and much less toxic whenadministered topically to patients with herpes keratitis (233).It is still used for this purpose. Recently, there has been aresurgence of interest in topical use of idoxuridine coupledwith dimethyl sulfoxide for treatment of herpes labialis (450).Trifluorothymidine, first developed as an antitumor agent,was found to inhibit herpes simplex virus (HSV) in vitro, butits selectivity index (ratio of drug concentration causingcellular toxicity to that needed for viral inhibition) was verylow compared with those of other agents (99) and it was notdeveloped for systemic use. Like idoxuridine, it is used fortopical treatment of herpes keratitis.

Despite their shortcomings, these drugs were important in

146

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 147

OH

IDOXURIDINE TRIFLUOROTHYMIDINE VIlFIG. 1. Some nucleoside analogs first used as antiviral agents.

that they demonstrated the feasibility of antiviral therapy inpatients. They also sustained interest in a continuing searchfor more selective agents. By the late 1970s, it was knownthat most human pathogenic viruses possess enzymes codedby the viruses themselves and not present in uninfected cells(327). Most of these enzymes are involved in viral nucleicacid synthesis, as discussed below. Their discovery repre-sented a major advance in antiviral therapy by making itpossible to direct efforts at finding specific inhibitors of theseenzymes rather than nonspecific inhibitors of viral growth incell culture.

Replication of viruses can be divided into several steps: (i)attachment to the cell, (ii) penetration, (iii) uncoating ofnucleic acid, (iv) transcription and translation of early (reg-ulatory) proteins, (v) nucleic acid synthesis, (vi) synthesis oflate (structural) proteins, (vii) assembly of mature virions,and (viii) release from the cell (for an example, see Fig. 2).All of these steps are potential targets for interference,although viral attachment, penetration, uncoating, assem-bly, and release closely resemble normal cellular processesand are thought to be carried out, in many instances, bycellular enzymes (210). It is at the point of nucleic acidsynthesis that viral processes diverge most from their cellu-lar counterparts and are most likely to require virus-specifiedenzymes. This is because eukaryotic cells contain double-stranded DNA and, when they replicate, make new DNAfrom the parental DNA template in the cell nucleus (Fig. 3).In addition, they transcribe mRNA from DNA and transportit into the cytoplasm, where proteins are translated. Viruses,on the other hand, may contain DNA or RNA as theirgenomic material, and the nucleic acid may be double orsingle stranded, circular or linear, and segmented or in onecontinuous piece. RNA may be either positive or negativestranded (having the same sense or direction as mRNA, andthus directly translatable, or having the opposite sense andnot directly translatable). Viruses may replicate in the nu-cleus, in the cytoplasm, or in both. Despite these differencesfrom their cellular hosts, viruses must be able to fit into hostcell synthetic pathways if they are to replicate successfully.They must present to the cell either a form of DNA that canbe transcribed directly into mRNA or a form of mRNA thatthe cell can recognize and translate into proteins (390). Manydifferent pathways have evolved by which viruses accom-plish this; some examples are shown in Fig. 3. Retroviruses,for example, carry single-stranded duplex RNA as theirgenomic material and, when infecting cells, must supply avirus-encoded reverse transcriptase to transcribe this RNA

OH

DARABINE

into cDNA which is then inserted into the host cell DNA(Fig. 2). Picornaviruses such as rhinoviruses and enterovi-ruses carry a positive single-stranded RNA that can bedirectly translated by host cell enzymes, but they mustencode an RNA replicase which allows use of the genomicRNA as a template for new negative-stranded RNA. Ortho-myxoviruses (influenza viruses), on the other hand, carry anegative-stranded RNA, and thus must supply an RNA-dependent RNA polymerase from which a plus-strandedmRNA can be made. Such unique enzymes, critical to viralreplication but unnecessary for cellular function, offer verygood targets for selective inhibition by antiviral agents. Theyare not the only enzyme targets, however.

Other enzymes such as nucleoside kinases and DNApolymerases are encoded by both cells and viruses (Fig. 4).The properties of such enzymes can differ greatly withrespect to substrate specificity, binding affinity for sub-strates, and susceptibility to inhibition by various com-pounds. These differences can be exploited in developingantiviral compounds. For example, thymidine kinases cata-lyze the first step in the preparation of pyrimidine nucleo-sides for incorporation into DNA, phosphorylation of the 5'carbon atom of the pentose ring (Fig. 4). Thymidine kinasespecified by HSV binds acyclovir much better than cellularthymidine kinase does and phosphorylates it 3 million timesfaster (235). DNA polymerases are responsible for incorpo-rating nucleotide triphosphates into growing DNA chains,and again, herpesvirus-specified DNA polymerases differfrom their cellular counterparts: they are 30 times moresusceptible to inhibition by acyclovir triphosphate than arethe alpha DNA polymerases of human origin (122). Differ-ences like these make it possible to identify compounds thatselectively inhibit viral functions. What advantage is there toviruses in encoding and carrying enzymes also made bycells? The answer is survival. Viruses carrying the extraenzymes have a wider host range and can infect cells that donot encode a critical enzyme or that express it only at acertain point in the cell cycle. HSV mutants that do notencode thymidine kinase, for example, are less capable ofestablishing reactivatable latent infections in neurons (whichare nondividing cells) than are thymidine kinase-encodingstrains (121, 142, 471).Many antiviral agents are nucleoside analogs, structures

that closely resemble the natural nucleosides used as build-ing blocks for DNA synthesis (Fig. 1 and 5). As such, thesedrugs are phosphorylated by cellular nucleoside kinases andincorporated into growing DNA chains by DNA polymer-

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

o ~~~~o

X I I <~~~~~C

I

IL

._

CZ

CZu

E

0

C

0 .

ILO 0

*a 0_ z

O Ck,

So

0.

~%>r%oV %,

nu(a

oc

Ji

Ani

z

o

z

IC

z

iCXz1-

z

o

z

z

At

148

la _*Xa~~~~

a

v ba0

pq a *00 CL

000OM44, In

021, z

4di bo

aCL

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 149

RETROVIRUSES

as du RNA ORTHOMYXOVIRUSESas RNA (-)

* REOVIRUSES

cDNA ds RNA

; % // * PICORNAVIRUSESa / t ss RNA(+) *J

crDsIA mRNA 4 proteins"

DNAsupercolled

HEPATITIS B VIRUSFIG. 3. Protein and nucleic acid synthetic pathways for eukaryotic cells (bold) and some representative human viruses (dashed lines). *,

need for a virus-specified enzyme; -, negative stranded; +, positive stranded. ss, single stranded; du, duplex; ds, double stranded.

ases, competing with the natural nucleosides as substratesfor both enzymes (Fig. 4). Many of the early antiviral agents,such as idoxuridine and trifluorothymidine, were equallygood substrates for both viral and cellular kinases. Eventhough they were slightly better inhibitors of viral than ofcellular polymerases (194, 365), they were activated andcapable of decreasing DNA synthesis in both infected anduninfected cells. Many of the newer nucleosides are lesstoxic, in part because there is at least one point in theirfunctional pathways which is specific for virus-infected cells.For example, acyclovir is activated only in virus-infectedcells, and zidovudine inhibits an enzyme, human immuno-deficiency virus (HIV) reverse transcriptase, not found innormal cells.Although the greatest success has been achieved by using

inhibitors of nucleic acid synthesis as antiviral agents, addi-tional enzymes are necessary for other steps in the viralreplication cycle and should be amenable to inhibition byputative antiviral compounds. Special interest in such com-pounds has been generated by the AIDS epidemic and theincreasingly obvious need to treat HIV infection with com-binations of drugs active at different sites of viral replication.Such combination therapy, it is hoped, will decrease drugtoxicity and reduce the chances of antiviral resistance de-veloping during treatment. Unfortunately, steps such as viralattachment and penetration and assembly and release arecatalyzed by cellular enzymes (210), and it has been difficultto find compounds that interfere specifically with theseevents. Nevertheless, some success has been achieved withamantadine (an inhibitor of uncoating) for management ofinfluenza, and recombinant soluble CD4 may yet proveeffective at inhibiting attachment of HIV to host lympho-cytes. Interferons, which inhibit viral mRNA transcriptionand protein synthesis, are also useful as antiviral agents. Inaddition, HIV proteases, which cleave precursor polypep-tides to form functional reverse transcriptases, are verypromising targets for selective inhibition.

ANTIVIRAL AGENTS

Amantadine and Rimantadine

The adamantanes, amantadine (1-aminoadamantane hy-drochloride) and its alpha-methyl derivative, rimantadine,are used for management of influenza A virus infections.They are cyclic amines with bulky, cagelike structuresunlike those of other known antiviral agents (Fig. 6). Theirmechanisms of action and spectra of antiviral activity areidentical, but rimantadine is metabolized differently fromamantadine, and it causes fewer central nervous system sideeffects such as insomnia and difficulty concentrating (110,514). It is also a more potent in vitro inhibitor of influenza Aviruses (431), although both drugs have been equally effec-tive in treating patients. Amantadine became available in1966; rimantadine is being considered for approval by theU.S. Food and Drug Administration.

It has been known for some time that amantadine inhibitsan early phase of viral replication (207, 232), more specifi-cally, virus uncoating (48, 381). Recent studies of avianinfluenza viruses have also demonstrated a block at a laterstage, virus maturation and assembly (184, 461). Suscepti-bility to amantadine is determined principally by the M2protein (184, 281), a virus-specified matrix protein present onthe surface of infected cells and in the virion in smallamounts (530). It has been proposed (28) that this proteinforms ion channels through which protons pass across themembranes of intracellular endocytic and exocytic vesicles.During virus uncoating, protons are transferred from theendocytic vesicle to the virion, allowing the fall in pH thatreleases free viral nucleoprotein into the cell cytoplasm.During virus assembly, protons are transferred out of theexocytic vesicle, thus maintaining the pH above the level atwhich the viral hemagglutinin, a major surface glycoprotein,would lose its structural integrity and fail to be incorporatedinto the viral envelope. Amantadine appears to block thisM2-mediated transfer of protons and thus inhibits viral

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.150 BEAN

a)Io L

CZ

0

-o

aC

-o

-o

0

z

00

*0

0

0

*0

a)

._

Zz

0

0

0

o

._UU

ca

_ c

.O.>

.0o

._

E

Ai

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 151

5H

ACYCLOVIR GUANOSINE RIBAVIRIN

H2N'

N3OH

GANCICLOVIR ZIDOVUDINE DIDEOXYINOSINE DI DEOXYCYT I DINE

FIG. 5. The purine nucleoside guanosine and some nucleoside analogs currently in use for antiviral therapy.

uncoating or viral maturation or both, depending on thestrain of virus (185). At in vitro concentrations much higherthan those at which these effects occur, adamantanes inhibitother RNA viruses, including influenza B virus, andparamyxoviruses (114). With current drug formulations,these high concentrations cannot be achieved in patients,and thus the adamantanes remain useful only for the man-agement of influenza A virus infections (Table 1).Both drugs are very effective in preventing illness due to

influenza A virus. When given prophylactically during acommunity outbreak, either compound reduces the risk ofacquiring influenzal illness by 50 to 90% ( 68, 110, 161, 317,331, 500). Rimantadine has fewer side effects when used in

NH2 HCI

this manner, although lowering the dose of amantadine alsoreduces adverse effects (378, 419). The efficacy of low-doseamantadine has not been proven in this situation, however,and the higher dose remains the prophylactic regimen ofchoice until rimantadine becomes available. Both drugs aremore effective in preventing illness than in preventing infec-tion with influenza A virus (110, 317). This may be beneficial,however, in that it allows the patient to produce protectiveantibodies without developing frank illness. Currently, theImmunization Practices Advisory Committee recommendsusing amantadine prophylactically to protect those at risk ofinfluenza complications who cannot or have not been vacci-nated (60). The drug can also be given at the same time as

NH2 *HCIH-C-CH3

[AMANTADINE RIMANTADINE

0

-0-p-clgI 30°0-

3 Na

FOSCARNETFIG. 6. Nonnucleoside antiviral agents.

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

TABLE 1. Antiviral agents currently available and their uses

Agent Route of administration Use Adult dosage

Acyclovir Oral Initial genital herpes 200 mg 5 times daily for 10 days

Ophthalmic ointment

Recurrent genital herpesSuppression, genital herpesSuppression, mucocutaneous herpes

in immunocompromised hostTreatment, mucocutaneous herpes inimmunocompromised host

Zoster in immunocompetent host

Herpes encephalitisNeonatal herpesaSevere genital herpesTreatment, mucocutaneous herpes,immunocompromised host

Suppression, mucocutaneous herpesin immunocompromised hosta

Zoster or varicella in immunocom-promised host

Initial genital herpesCutaneous herpes in immunocompro-

mised host

CMV retinitis in AIDS

HIV infection, CD4 cells <500/mm3,or symptomatic

Herpes encephalitisNeonatal herpesZoster in immunocompromised host

Herpes keratitis

Influenza A: treatmentInfluenza A: prophylaxis

Severe RSV, infants and children

Lassa fever'

200 mg 5 times daily for 5 days400 mg twice daily for up to 1 yr200 mg 3 to 5 times daily

200-400 mg 5 times daily until healed

800 mg 5 times daily for 7-10 days

10 mg/kg 3 times daily for 10-21 days500 mg/mTh 3 times daily for 10 days5 mg/kg 3 times daily for 5 days5 mg/kg 3 times daily for 7 days

5 mg/kg 3 times daily

Adult: 10 mg/kg 3 times daily for 7 days

Child: 500 mg/mrn 3 times daily for 7 days

Apply every 3 h up to 6 times daily for 7 daysApply every 3 h up to 6 times daily for 7 days

Induction: 5 mg/kg twice daily for 14-21 daysMaintenance: 5 mg/kg daily

100 mg 5 times daily

15 mg/kg daily over 12-24 h for 10 days15 mg/kg daily over 12-24 h for 10 days15 mg/kg daily over 12-24 h for 5 days

0.5 in. [1.27 cm] to eye 5 times daily for 7-21days

200 mg daily for 5-7 days200 mg daily

12-18 h daily for 3-7 days (concn, 20 mg/ml)

2-g load, then 0.5-1 g 3-4 times daily for 10days

Ophthalmic solution

Ophthalmic solution

Hepatitis B-chronic active liver dis-ease'

Hepatitis C-chronic liver disease

Condyloma acuminata (warts)

CMV retinitis

Acyclovir-resistant HSV or VZVa

Zidovudine intolerance or treatmentfailure in HIV-infected host

Herpes keratitis

Herpes keratitis

5 x 106 U daily for 4 mo

2 x 106-3 x 106 U 3 times weekly for 6 mo

1 x 106 U per wart in up to 5 warts 3 timesweekly

Induction: 60 mg/kg 3 times daily for 2-3 wkMaintenance: 90-120 mg/kg daily40-60 mg/kg 3 times daily

Tablet: 125-300 mg (as 2 tablets) 2 times daily

Powder: 167-375 mg 2 times daily

1 drop every 2 h up to 9 drops daily untilhealed

1 drop/h during day and every 2 h at night un-

til healed

a The package insert is not approved for this indication.b Square meters of body surface area.

Intravenous

Topical

Ganciclovir

Zidovudine

Vidarabine

Intravenous

Oral

Intravenous

Amantadine

Ribavirin

Oral

Aerosol

Intravenous

Interferon Subcutaneous

Intralesional

IntravenousFoscarnet

Didanosine

Trifluridine

Idoxuridine

Oral

152 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 153

influenza virus vaccine to protect the patient until an anti-body response to the vaccine occurs.Both amantadine and rimantadine have been used for the

treatment of influenza. They are most effective when givenwithin the first 48 h of illness and may allow the patient toreturn to routine daily activities 1 to 2 days earlier than notreatment would (191, 475, 483, 514). Amantadine alsoappears superior to aspirin in this regard. In one comparativestudy, patients taking 3.25 g of aspirin daily became afebrilesooner but had more side effects (insomnia, nausea, ringingin the ears) than amantadine recipients, who experienced anearlier reduction in symptoms and fewer adverse reactions(528). The peripheral pulmonary airway dysfunction accom-panying influenza is also improved by amantadine (279), andthis may be important for earlier resolution in normal hostsand for minimizing disease in those with underlying pulmo-nary or cardiac conditions. The effects of therapy on thecomplications of influenza, such as pneumonia and myo-carditis, are unknown.Although drug resistance does not occur naturally among

influenza viruses (27), it can be induced by therapy, andthese resistant viruses can cause influenzal illness. Whenpatients ill with influenza were treated with rimantadine,they improved symptomatically, but 50% of them beganshedding rimantadine-resistant viruses within 4 to 6 days(177, 188). In one study, these resistant viruses were appar-ently transmitted to family members, who also became illwith influenza (188). Resistance was mapped to the M2protein (29), and subsequent studies of similarly resistantavian viruses have shown that these resistant strains aregenetically stable and equal in virulence to wild-type viruses(25). These findings raise the possibility that widespread useof adamantanes for treatment of influenza might result inantiviral pressure and selection of predominantly resistantvirus populations. To prevent the spread of resistant virus, ithas been suggested that patients be isolated or have limitedcontact with others while they are being treated for influenza(186). The effects of this and other methods to controltransmission are unknown, however. In view of these prob-lems and of the unknown effect of therapy in preventing orrelieving complications, no recommendation for therapy ofinfluenza is currently made by the Centers for DiseaseControl (60), although amantadine is approved for thispurpose (package insert).

Ribavirin

Ribavirin (1-beta-D-ribofuranosyl-1,2,4-triazole-3-carbox-amide) was first synthesized in the early 1970s as part of anintensive effort to identify new antiviral agents (517). At firstglance, it appears to be a nucleoside analog with an openpyrimidine ring (Fig. 5). Structurally (362) and functionally(458), however, it most closely resembles guanosine. Riba-virin is active in vitro against a wide variety of RNA andDNA viruses, including adenoviruses, herpesviruses, influ-enza A and B viruses (429, 517), respiratory syncytial virus(RSV) (212), bunyaviruses, arenaviruses (214), reoviruses(367), and HIV (292).The mode of action has not been clearly defined. Three

mechanisms have been proposed, and all may operate tosome extent in cells infected by different viruses. This mayalso account for the wide spectrum of activity. Ribavirinreadily diffuses into eukaryotic cells, where it is convertedby cellular enzymes to the mono-, di-, and triphosphateforms (437, 458). The monophosphate is a potent competi-tive inhibitor of IMP dehydrogenase, an enzyme essential for

the synthesis of GTP. This inhibition results in a decrease inthe cellular pools of guanine nucleotide necessary for bothcellular and viral replication (458, 459). Ribavirin inhibitscapping of viral mRNA (170, 493), a critical step in thereplication of most viruses. Its most important effect, how-ever, appears to be on the early events of viral replication.Ribavirin directly inhibits viral RNA-dependent RNA poly-merase of influenza viruses (129), and it can retard bothinitiation (54, 338, 367, 521) and elongation (129, 521) ofmRNA transcripts. It is not incorporated into the growingchain of viral nucleic acid and does not cause chain termi-nation (129, 477). Only one strain of virus, a mutant of fowlplague virus, is known to be resistant to ribavirin (215).

In addition to its antiviral properties, ribavirin has im-munoregulatory effects. In cell culture and in animals, itinhibits macromolecular synthesis and cell division (270),lymphocyte proliferation, and nucleic acid synthesis (341). Italso suppresses B lymphocytes and subsequent antibodyproduction (358) and tumor growth (357). To date, however,none of these effects has been shown to be of consequence inpatients treated with ribavirin.The major toxicities of ribavirin are anemia and embryo-

toxicity. Anemia occurs when ribavirin diffuses into eryth-rocytes and accumulates, because erythrocytes lack phos-phatases and are unable to hydrolyze (dephosphorylate) thedrug. The erythrocytes then become damaged as they ageand are removed prematurely from the circulation (52, 334).At very high doses of ribavirin in animals, bone marrowsuppression of erythroid precursors also occurs (52). All ofthese effects are reversible upon removal of the drug, but thehalf-life of ribavirin in human erythrocytes is 40 days, so theeffect is prolonged. In pregnant rodents treated with ribavi-rin, skeletal defects in the developing embryo as well as fetalresorption have been observed (242). Because of this, thedrug is contraindicated during pregnancy, and safety mea-sures must be taken by pregnant health care workers whoadminister ribavirin to patients (see below).

Ribavirin became commercially available for use as anaerosol in 1986. Both the oral and intravenous forms havebeen studied, but the drug was found to be most effectivewhen given to animals by pulmonary aerosol, so it wasdeveloped for that purpose. The drug is diluted in a reser-voir, nebulized by a small-particle aerosol generator, anddelivered to the patient via face mask, ventilator, or infantoxygen hood. Aerosolized drug can leak from the systemduring administration and may be inhaled by health careworkers. This may represent a hazard to pregnant workers,although it is not known whether the small amounts ab-sorbed into the bloodstream will damage the fetus (57).

In college students with uncomplicated influenza, aerosolribavirin reduced the duration of fever and symptoms anddecreased viral shedding (239, 291, 513). This has not beentrue of all trials and for all influenza variants, however (33,167). Because of this inconsistency and because aerosoltherapy is an expensive and cumbersome mode of therapyfor a self-limited illness, ribavirin is not often used in thissetting.

In infants with bronchiolitis due to RSV, ribavirin hasshown some effect in decreasing viral shedding, improvingoxygenation, and reducing the duration of illness (178, 179,388, 469), although questions have been raised about themethods used in treatment trials (494). In a recent trial,ribavirin decreased the duration of oxygen therapy, mechan-ical ventilation, and hospital stay in otherwise healthy in-fants with severe RSV illness (439). Nevertheless, importantquestions remain unanswered, including duration of therapy,

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

response of children with underlying cardiac and pulmonarydisease, and efficacy of the drug when administered late inthe course of illness (368). Ribavirin is extremely expensive($400 per day in drug costs alone), and all children withself-limited RSV cannot be hospitalized and treated. Alongwith the possible toxicity for health care workers, theseunanswered questions indicate the great need for additionalstudies to define the optimum use of this drug for respiratoryviral disease.

Because of its in vitro activity against bunyaviruses andarenaviruses, intravenous ribavirin has been investigated fortreatment of viral hemorrhagic fevers (214). Most notablehas been the success with Lassa fever. When patients at highrisk of death from Lassa fever were treated intravenouslywithin 6 days of fever onset, the death rate of 55 to 76% inplacebo-treated patients decreased to 5 to 9% in ribavirinrecipients (293). Oral ribavirin is also beneficial againstLassa fever and is given as prophylaxis to high-risk contactsof Lassa fever patients (58). Recent small-scale studies haveshown that ribavirin crosses the blood-brain barrier well,giving drug levels in the cerebrospinal fluid that are 50 to100% of those in serum (82, 330). For this reason, it may alsobe of use in the treatment of common viral encephalitidescaused by bunyaviruses, such as La Crosse encephalitis (54,214).

Ribavirin has recently been investigated for possible use inHIV-infected patients. Although the drug given orally in highdoses appeared to delay progression to AIDS (384), manyquestions have been raised about the study methodology(39). In addition, no effects on surrogate markers of HIVprogression (CD4 counts, p24 antigenemia, viremia, or im-mune function) were found, and it is not clear that clinicallyachievable drug levels are inhibitory for viral replication(292, 385-387). In view of these problems, licensure ofribavirin for treatment of HIV is no longer being pursued inthe United States, although the drug is available in othercountries.The place of ribavirin in our armamentarium of antiviral

agents is not yet clear. Though useful in aerosol form, it iscumbersome, expensive, and perhaps toxic. Intravenousribavirin is very useful for life-threatening illness, such asLassa fever, but toxicity limits the use of this form in lesssevere diseases. Liposome-encapsulated ribavirin has beenused successfully in animal models for treatment of RiftValley fever and influenza (163, 236), and it may represent away to achieve improved drug delivery to target organswhile minimizing toxicity. Analogs of ribavirin havebeen synthesized and tested for antiviral activity. One com-pound, 5-ethynyl-1-beta-ribofuranosylimidazole-4-carboxa-mide (EICAR), was originally studied as an antileukemicagent but has now been shown to be 10 to 30 times as activeas ribavirin against RSV and influenza viruses (98). Whetherit will be clinically useful remains to be seen.

Vidarabine

Vidarabine (9-beta-D-arabinofuranosyladenine) was thefirst antiviral agent licensed for intravenous use in the UnitedStates, in 1978. It is an analog of the nucleoside adenosine(Fig. 1) and was originally synthesized and studied in theearly 1960s as an antitumor agent. Subsequently, it wasfound to be active in vitro against HSV, varicella-zostervirus (VZV), cytomegalovirus (CMV), vaccinia virus, andsome RNA tumor viruses. In animal models, it was activeagainst HSV and vaccinia virus (409).The mechanism of action has not been completely delin-

eated, but it involves phosphorylation of the nucleoside bycellular enzymes (44, 355) and incorporation into the grow-ing chains of both cellular and viral DNAs, with resultantslowing of DNA synthesis (343). Vidarabine triphosphate isalso a competitive inhibitor of DNA polymerase, both cel-lular and viral. It is more potent, however, against viralenzymes, and this potency may account for its selectiveinhibition of viral DNA synthesis (117, 417).

Vidarabine inhibits other steps in nucleic acid synthesis,such as RNA polyadenylation (393) and terminal deoxynu-cleotidyl transferase (108), but whether or not these contrib-ute to its antiviral activity is unknown. It also inhibitsS-adenosylhomocysteine hydrolase (398), an enzyme neces-sary for methylation of tRNA and mRNA and for othercellular transmethylation reactions. This inhibition results incytotoxicity for mononuclear cells and in the possible accu-mulation of biogenic amines (117), both of which maycontribute to the toxicity of this compound.

Vidarabine became available as an eye ointment in 1977and remains useful for the treatment of herpes keratitis(Table 1). Although it is also useful for the treatment of HSVand VZV in immunocompromised patients (502, 504, 507,510) and for herpes encephalitis (508, 509) and neonatalherpes (506, 511), it has been almost entirely replaced byacyclovir for these diseases. Vidarabine is difficult to use. Itis poorly soluble in water and rapidly deaminated in vivo toarahypoxanthine, a much less active compound. It musttherefore be administered continuously by the intravenousroute in large volumes of fluid. Toxicities, including nauseaand vomiting, weakness, weight loss, megaloblastosis ofbone marrow erythroid precursors, tremors, and myoclonushave been seen in a high proportion of treated patients (287,394). Acyclovir is easier to administer, less toxic, and moreactive against herpesviruses. Although vidarabine does notrequire a virus-coded thymidine kinase for activation, andthymidine kinase-deficient HSV isolates are susceptible tothe drug in vitro, vidarabine has not proven useful fortreatment of acyclovir-resistant HSV in HIV-infected pa-tients, and foscarnet is preferred (402).

Acyclovir

Acyclovir [9-(2-hydroxyethoxymethyl)guanine] has thebest therapeutic index of available antiviral agents, and it iswidely used for the treatment of herpesvirus infections. Itwas "discovered" in the mid-1970s by Elion and colleagueswhile they were screening for antiviral activity of nucleosideanalogs (410). The compound is an analog of guanosine (Fig.5), but instead of a complete ribose ring attached to thepurine base, it has an acyclic side chain. Acyclovir diffusesfreely into cells but is activated and accumulates only in cellsinfected with herpesviruses because the first step in activa-tion, phosphorylation at the 5' position of the side chain, iscatalyzed only by a virus-specified thymidine kinase and notto any appreciable extent by cellular kinases (Fig. 4) (122,159). Further phosphorylations to the diphosphate and theactive triphosphate forms are carried out by cellular en-

zymes (122, 312). During DNA replication, acyclovirtriphosphate competes with the natural substrate, dGTP, forthe viral DNA polymerase. It has a higher affinity for theenzyme, however, and is preferentially incorporated into thegrowing chain of viral DNA (122, 453). Because the 3'hydroxyl is missing from the attached acyclovir molecule,the next nucleotide cannot be attached, and DNA replicationis terminated (103, 156, 294). Furthermore, when acyclovir isbound to the DNA template in the presence of the next

154 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 155

nucleotide, a tight irreversible complex is formed and thepolymerase is completely inactivated, further slowing viralDNA synthesis (103, 158, 369). Taken together, these prop-erties make acyclovir highly selective in inhibiting viralreplication while having little effect on host cell function.There is, for instance, a 3,000-fold difference between theconcentration of acyclovir needed to inhibit HSV type 1(HSV-1) and that needed to inhibit the cells in which it isgrown (122).The herpesviruses vary in their susceptibilities to acyclo-

vir. HSV-1 and HSV-2 are exquisitely susceptible, requiringmean in vitro concentrations of 0.04 and 0.4 ,ug/ml, respec-tively, for 50% inhibition of viral replication (86, 329). Thethymidine kinase of VZV does not phosphorylate acycloviras effectively, and the virus is 8 to 10 times less susceptibleto the drug than HSV (35, 86, 329). Although its DNApolymerase is very susceptible to acyclovir (452), CMV doesnot encode a thymidine kinase (133), and high drug levels (5to 25 ,ug/ml) are required for inhibition (86, 353). It is notclear that Epstein-Barr virus (EBV) encodes a thymidinekinase capable of activating acyclovir. Its DNA polymeraseis, however, very susceptible to the drug (91), and the 50%inhibitory concentrations are low (73, 278). Acyclovir has noeffect on the latent phase of any of the herpesviruses.

Acyclovir is available in three formulations: an ointment,pills or capsules, and an intravenous suspension. Systemicabsorption and other side effects of the ointment are mini-mal, but it is not very effective (see below) and has largelybeen replaced by oral formulations. Levels achieved inserum after intravenous administration are well above theinhibitory concentrations of HSV, VZV, EBV, and somestrains of CMV. With oral dosing, however, only 15 to 20%of the medication is absorbed, and levels in serum are notconsistently above the inhibitory concentrations for any ofthe herpesviruses except HSV (100).

Acyclovir is remarkably free of serious toxicity. Whengiven intravenously in high doses, it can cause a transientrise in serum creatinine that is apparently due to obstructedrenal tubules (22, 407). A reversible neurologic syndromeconsisting of confusion, lethargy, and occasionally halluci-nations and coma has also been described. This syndrome isusually seen in very ill, immunocompromised patients re-ceiving high-dose intravenous therapy (137, 253, 491). Withoral therapy, nausea, diarrhea, and headache have occasion-ally been reported. Acyclovir is not carcinogenic and ismitogenic or teratogenic only at very high levels in animals(479). It does cross the placenta, however, and thereforemust be used with caution in pregnant patients (45, 172).Acyclovir is very effective for the treatment of HSV in

otherwise healthy patients (Table 1). When given orally orintravenously, it shortens the course of initial genital herpesby one-third to one-half (46, 79, 314, 323). Recurrent genitalherpes does not respond as dramatically, even when patientsinitiate therapy themselves at the first sign of disease (323,374). This may be because recurrences normally last only afew days, and it is difficult to demonstrate a shortening of thecourse with drug therapy. Acyclovir is, however, very usefulin preventing recurrent disease. When given daily in lowdoses to patients with frequent eruptions, it suppresses themalmost completely and causes no side effects even withprolonged use (112, 230, 306, 313). It does not preventasymptomatic viral shedding (457), and transmission hasoccurred when patients were free of genital lesions (392).Topical acyclovir, a 5% ointment, is useful, though not aseffective as oral therapy, for treatment of initial genitalherpes (79, 80, 472). In recurrent disease it offers no benefit

(282, 373). Topical therapy is occasionally used in pregnantpatients who have mild genital herpes and should not receivesystemic acyclovir.

Oral treatment of herpes labialis (the common cold sore)shortens the course of illness in those patients who are proneto more severe eruptions and are able to initiate therapythemselves at the first sign of an eruption (451). When givenprophylactically to patients exposed to the sun or to artificialUV light, oral acyclovir decreased the frequency of herpeticlesions appearing within the next 2 or 7 days, respectively(448, 449) but did not affect the lesions which appearedimmediately (within 48 h) after the exposure (448). Topicaltherapy is of little benefit in otherwise healthy patients withcold sores (448, 520). In the treatment of herpes encephalitis,acyclovir has proven both more effective and less toxic thanvidarabine (434, 503). It reduces morbidity to 14% (vidara-bine reduces it to 38%), and it is now the treatment of choicefor that disease. A recent trial of acyclovir versus vidarabinefor the treatment of neonatal herpes showed neither drug tobe superior (501). Given the sample size, however, as muchas a 25% difference in morbidity and mortality could havebeen present and gone undetected.

Acyclovir has also been very useful for management ofmucocutaneous HSV in immunocompromised patients. Ad-ministered orally or intravenously, it decreases pain andviral shedding and accelerates healing (310, 425). Though notas effective, topical therapy can also be useful in this regard(505). When given suppressively to HSV-seropositive pa-tients, acyclovir greatly reduces the chances of a recurrenceduring subsequent chemotherapy (405) or transplantation(406, 418, 423, 492).VZV infection responds better to intravenous than to oral

acyclovir, probably because levels in serum after oral ad-ministration are often below the 50% inhibitory dose for thevirus. In immunocompromised patients with chickenpox orzoster, intravenous acyclovir has been very effective indecreasing dissemination and other complications (15, 326,361, 424). It can also be given orally in the post-bone marrowtransplantation period and appears to decrease the fre-quency of zoster during that time (346, 421). In otherwisehealthy patients with zoster, intravenous acyclovir de-creases acute pain and accelerates lesion healing (23, 350),but oral therapy has had only a modest effect against thisdisease (213, 296). Neither form of therapy reduces posther-petic neuralgia. Of importance, oral acyclovir decreases thefrequency of eye complications in patients with ophthalmiczoster (69, 70). In one study of otherwise healthy childrenwith chickenpox, oral acyclovir reduced the duration ofdisease by 1 day but did not influence the complication rate,days off from school, or transmission rate within the family(18). In otherwise healthy adults with chickenpox pneumo-nia, intravenous acyclovir appears to hasten recovery (175).

Despite the reduced susceptibility of CMV to acyclovir,the drug may yet prove useful in the management of CMVdisease in immunocompromised patients. Two studies, oneof bone marrow recipients (309) and one of renal allograftrecipients (17), have suggested that high-dose intravenousacyclovir can reduce the frequency of CMV infection anddisease when given suppressively in the peritransplantationperiod. The reasons for this are not clear, but may relate tothe fact that only low numbers of viruses are present earlyafter reactivation, and the small amount of active acyclovirpresent may be sufficient to limit viral replication enough toprevent clinical disease. Acyclovir therapy of establishedCMV infections in immunocompromised patients has notbeen successful (16, 489).

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

Treatment of diseases due to EBV has also been disap-pointing, despite the apparent susceptibility of the virus toacyclovir. The course and symptoms of mononucleosis arenot affected by treatment, although viral shedding from theoropharynx is decreased (3, 481). On the other hand, acy-clovir does appear to be effective for treating hairy leuko-plakia, an EBV-associated condition characterized by pain-ful tongue lesions in patients infected with HIV (377).Improvement is transient, however, and the lesions recurwhen the drug is stopped.

Resistance of HSV to acyclovir has been extensivelystudied. It results from changes in the virus-specified thymi-dine kinase and/or the DNA polymerase and occurs by oneof three mechanisms: a deletion or mutation conferringinability or reduced ability to express thymidine kinase,resulting in lack of acyclovir phosphorylation (a TK- orthymidine kinase-deficient mutant) (72, 413); expression of aDNA polymerase with reduced affinity for acyclovir triphos-phate (72, 265, 413); or expression of a thymidine kinase withaltered substrate binding properties for the drug (90, 154).Both of the last two mechanisms result in lack of recognitionof acyclovir as a substrate for the enzyme. Among patientisolates, thymidine kinase-deficient mutants are most com-mon, but viruses expressing the other mechanisms have alsobeen found (49, 76, 123, 399, 426, 490).

In nature, HSV populations consist of a mixture of TK-and normal (TK+) virus variants (337). They are predomi-nantly TK+, however, because these viruses have a selec-tive advantage in being more capable of establishing latent,reactivatable infections in host sensory ganglia. They arealso more neurovirulent (121, 142, 143, 471). Under pressureof antiviral drug therapy, a population can become predom-inantly TK-, as the TK+ variants are unable to replicate.When the drug is withdrawn, the reverse occurs. In practice,resistance develops when patients are treated for prolongedperiods with acyclovir in doses that do not suppress viralreplication completely but allow "breakthrough" replicationto occur. To date, this has been seen almost entirely inimmunocompromised patients, (19, 24, 131, 220, 335). Treat-ment consists of withdrawing acyclovir and, when neces-sary, administering a drug with a different mode of action,such as foscarnet (see below). The importance of resistancefor the future of acyclovir therapy is not yet clear. Thymi-dine kinase-deficient mutants are generally not as hardy astheir TK+ counterparts. This not true, however, of mostthymidine kinase-altered and DNA polymerase-altered vi-ruses (90, 141, 142, 265). Whether resistant virus populationswill be able to establish themselves and compete withwild-type viruses remains to be seen.

Because of acyclovir's poor oral availability and lack ofeffect against some herpesviruses, other drugs are beingsought for treatment of herpesvirus infections. Famcicloviris the diacetyl ester of penciclovir, an acyclic nucleosideanalog with a long intracellular half-life (484) and goodactivity against HSV and VZV but poor oral absorption(485). Famciclovir is well absorbed and rapidly converted topenciclovir (485) and is very promising for treatment ofherpesvirus infections. 1-Beta-D-arabinofurasonyl-E-5-(2-bromovinyl)uracil, or BV-ara-U, is a thymidine analogwhich is 1,000 times more active against VZV than acyclovir(283). Cellular toxicity is seen at concentrations 1 million-fold higher than those needed to inhibit viral replication,making this drug highly selective and very promising fortreatment of VZV infections. It is currently in patient trials.

Ganciclovir

Ganciclovir, 9-(1,3-dihydroxy-2-propoxymethyl)guanine,is a nucleoside analog similar to acyclovir except that itcontains a carbon with attached hydroxyl group at the 3'position of the ribose ring (Fig. 5). It is active against all thehuman herpesviruses (139) and is up to 100 times more activethan acyclovir against human CMV (436, 474). It is also moretoxic than acyclovir and for this reason has been used onlyfor treatment of serious CMV disease in immunocompro-mised patients.

It is not clear why ganciclovir is so much more effectivethan acyclovir against CMV. In HSV-infected cells, ganci-clovir, like acyclovir, is activated by a virus-specified thy-midine kinase (37, 139, 436). In CMV-infected cells, in whichno viral thymidine kinase is induced, ganciclovir is alsophosphorylated and accumulates to much higher levels thanacyclovir (37, 151). It is removed from these cells veryslowly, persisting in stable form for several days (37). Themolecular mechanisms of activation, accumulation, and deg-radation are not known but probably account to some extentfor the difference in activity, as ganciclovir is actually a lesspotent inhibitor of viral DNA polymerase than acyclovir is(151, 286). Ganciclovir triphosphate is incorporated into thegrowing viral DNA chain and markedly slows DNA synthe-sis (285). Because the 3' hydroxyl is present on the molecule,the next incoming nucleotide can be bound to the nascentDNA chain, and ganciclovir is thus not a chain terminator(65, 285). Despite the fact that ganciclovir, like acyclovir, isa more potent inhibitor of viral than cellular polymerases(151, 436), it is more toxic than acyclovir. In vitro inhibitionof the myeloid elements of bone marrow occurs at levels inserum seen with intravenous administration of ganciclovir(443, 444). The drug is also mitogenic in mammalian cells andcarcinogenic and embryotoxic in animals (467). In humans,ganciclovir causes a reversible bone marrow toxicity inapproximately 25% of patients, a factor that frequently limitsits use (62, 208, 422).There have been no placebo-controlled trials to evaluate

the efficacy of ganciclovir. Historically controlled trials haveshown the drug to be generally useful in the management ofCMV disease in immunocompromised patients, althoughefficacy varies among different patient groups. AIDS pa-tients with CMV retinitis respond well to intravenous ther-apy, with 80% or more achieving stabilization or improve-ment in their vision (47, 74, 138, 197, 208, 217, 271). Almostall relapse, however, unless maintenance therapy is insti-tuted, and even then many patients experience breakthroughdisease or cannot tolerate the medication. These problemsand the difficulties of long-term intravenous therapy haveprompted the investigation of other modes of treatment.Intravitreal administration of ganciclovir avoids myelosup-pression and has stabilized vision in some patients (195,198). Oral therapy is also being investigated. While the oralbioavailability of ganciclovir is very poor (6 to 8% [96]),levels in serum may be high enough to inhibit some strains ofCMV (222). AIDS patients with CMV pneumonia may havea response rate as high as 60 to 80% when given intravenousganciclovir (47, 271), although strict criteria for diagnosiswere not applied in published studies. On the other hand,only 10 to 40% of bone marrow transplant patients respond,and their mortality remains high despite suppression of viralreplication and shedding by the drug (47, 83, 422). Onehypothesis for this difference is that, in bone marrow recip-ients, CMV pneumonia is an immune-mediated diseaseresulting from T-cell cytotoxic responses to viral antigens

156 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 157

expressed on the surface of infected lung cells, whereas inAIDS patients, whose cellular cytotoxic responses are poor,the disease is due to direct damage to the lungs (174). Toneutralize viral antigens and prevent recognition by immuneeffector cells, high-titered CMV immunoglobulin has beenadministered with ganciclovir in bone marrow transplantpatients. Such combined antiviral and immunomodulatortherapy has increased the survival rate to 50 to 70% (125,371). In addition, it may permit a decrease in ganciclovirdosage, with an attendant decrease in bone marrow toxicity.In one recent trial, suppressive ganciclovir therapy was verysuccessful in preventing clinical disease in bone marrowrecipients who had CMV in their respiratory secretions buthad not yet developed pneumonia (411). An additionalplacebo-controlled trial has confirmed and extended thesefindings; early ganciclovir therapy in bone marrow recipientsexcreting CMV but not yet ill reduced the frequency ofCMVdisease by 93% (from 15 of 35 to 1 of 37 patients) andsignificantly improved patient survival at 6 months post-transplantation (163). In uncontrolled trials of AIDS patientswith CMV gastrointestinal disease, ganciclovir appeared toreduce diarrhea and the pain and dysphagia of esophagitis(62, 74, 109). A recent, well-controlled study of bone marrowrecipients, however, indicated no benefit in this patientgroup (372). In patients who have undergone solid-organtransplantation and developed CMV disease, there is someevidence that ganciclovir therapy improves the recovery rate(119, 180, 193) and may improve graft survival (119).

Ganciclovir resistance has been reported in three immu-nocompromised patients being treated for CMV disease(127) and was due to lack of phosphorylation of the drug bythe CMV-infected cells (452). This finding is consistent withphosphorylation of ganciclovir by a virus-induced cellularenzyme or by an enzyme that is virus encoded. In addition,a CMV strain produced in the laboratory has been found topossess two mutations which confer ganciclovir-resistance:one in the DNA polymerase gene giving rise to an alteredDNA polymerase and one at an unknown site giving rise toan inability to phosphorylate the drug (463). An HSV strainresistant to ganciclovir because of an altered DNA polymer-ase has been identified (84). A recent prospective surveyestimated that 8% of patients receiving ganciclovir for morethan 3 months developed resistant CMV (118), suggestingthat prolonged therapy may facilitate the emergence ofresistant strains. Although the importance of ganciclovirresistance is presently unknown, the three reported patientsall had progressive CMV disease, and this suggests thatresistance can be associated with therapeutic failure.

Foscarnet

Foscarnet (trisodium phosphonoformate) is a pyrophos-phate analog unlike any other antiviral agent currently in use(Fig. 6). It was developed in the late 1970s as a less toxic,more effective alternative to phosphonoacetic acid, anotherpyrophosphate analog with antiviral properties (196). TheFood and Drug Administration recently granted an indica-tion for treatment of CMV retinitis in AIDS patients. Inclinical trials, foscarnet has also proven useful for treatmentof acyclovir-resistant herpesvirus infections.

Unlike nucleoside analogs, foscarnet does not need to beactivated by cellular or viral kinases. Instead, the moleculebinds directly to the pyrophosphate-binding sites of RNAand DNA polymerases, inhibiting them in a noncompetitivefashion with respect to nucleotide substrates (328). While themechanism of inhibition is not well understood, one hypoth-

esis is that polymerase-bound foscamet forms an unstableintermediate with nucleoside monophosphates, resulting indegradation of the nucleic acids (275, 328). Both cellular andviral polymerases are affected by foscarnet, but some viralenzymes are inhibited by concentrations 1/10 to 1/2 thoseneeded for cellular enzyme inhibition. These include thepolymerases of some strains of influenza A virus, the humanherpesviruses, hepatitis B virus, and the reverse tran-scriptase of HIV (269, 328, 404, 488, 493). Foscarnet isdifficult to use. Oral bioavailability is poor (433), althoughefforts are being made to increase absorption and prolong thehalf-life by conjugation to fatty alcohols (322). Currently, thedrug is available only in intravenous formulations, and itmust be given frequently by means of an infusion pump, asthe half-life in plasma is very short (9, 433). At physiologicpH, foscarnet is highly ionized and has limited cell penetra-tion (328, 433). Levels in spinal fluid are usually about 40%of those in plasma but may vary by a wide margin (7). Up to30% of the drug may be deposited in bone, with a subsequenthalf-life of several months (432). The importance of suchdeposition, especially for bone development in children, isnot known. The major adverse effects of foscarnet are renalimpairment (present in most patients and dose limiting in 10to 23% of them), imbalances of electrolytes such as calcium,phosphate, potassium, and magnesium (101, 134, 221, 224,382, 495), and seizures (7). It appears that foscamet chelatesdivalent metal ions such as calcium, resulting in a dose-related but reversible drop in serum calcium that may besevere or even fatal (223). Penile ulcers have also been seenwith systemic therapy and may result from the localizedirritant effect of high levels of ionized foscamet in the urine(136, 168, 276, 319, 482).

In AIDS patients with CMV retinitis, foscarnet and gan-ciclovir appear to be equally effective in preventing progres-sion of disease, as assessed in clinical trials with historicalcontrols. Vision stabilized or improved in 80 to 100% ofpatients, but maintenance therapy was required, and evenso, 70% or more relapsed (134, 224, 274, 336, 495). Thepotential advantage of foscarnet in this setting is that it hasless bone marrow toxicity than ganciclovir, and it may bepossible to use it in conjunction with zidovudine. Theadditive bone marrow toxicities of ganciclovir and zidovu-dine preclude their simultaneous administration. A con-trolled trial directly comparing ganciclovir and foscamet forCMV retinitis was recently completed. Preliminary analysisconfirmed that they are equally effective in halting progres-sion and suggested that foscarnet may prolong survival by afew months in some patients. Other uncontrolled trials haveindicated that some bone marrow and renal transplant pa-tients with severe CMV pneumonia or with fever and leuko-penia may improve with foscarnet therapy. Many died,however, and in the absence of a control group, it is difficultto assess the value of therapy in these patients (4, 238, 382).

In AIDS patients who develop acyclovir-resistant HSV orVZV infections after prolonged courses of therapy, foscar-net has proven very useful (Table 1). About 80% of patientsrespond with ulcer healing and symptom resolution (63, 130,400, 401). In this setting, foscarnet is also superior tovidarabine; the rate of lesion healing is higher and there isless toxicity (402). Maintenance therapy with foscarnet isneeded, as with other antiherpesvirus agents, and is success-ful in about 70% of patients. Apparent resistance to foscar-net has developed, however, after several courses of therapyin HIV-infected patients (34) and in one bone marrowallograft recipient treated for CMV pneumonia (241). Themechanisms of resistance have not yet been determined for

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

158 BEAN

these viruses but probably involve altered DNA polymer-ases, as seen when viruses are exposed to foscarnet in vitro(102). When patients are infected with thymidine kinase-deficient, acyclovir-resistant HSV or VZV mutants, as ismost commonly the case, foscarnet is useful for therapy, asit does not require activation by thymidine kinases. Whenacyclovir resistance is due to changes in the viral DNApolymerase, however, it may be accompanied by resistanceto foscarnet, and in these cases the drug may not be useful.

Foscarnet is effective against HIV at levels achieved inpatients' serum (404, 488). In preliminary studies, it de-creased viral (p24) antigenemia and raised helper lympho-cyte (CD4) counts (30, 221, 328), but the effect was transient,and patients relapsed when the drug was stopped. The needfor intravenous administration has made it difficult to studyfoscarnet for long-term maintenance therapy in HIV infec-tion and may preclude its use in favor of more easilyadministered drugs.

Zidovudine

Zidovudine (3'-azido-3'-deoxythymidine; formerly knownas azidothymidine, or AZT), is a synthetic dideoxynucleo-side, one of a family of nucleoside analogs in which the 2'and 3' hydroxyls of the ribose ring have been replaced byhydrogens or other moieties. It was first studied as anantitumor compound and then as a drug for feline leukemiavirus, and in 1985, it was found to inhibit HIV (316). Thedrug became commercially available in 1987 and is now usedfor management of HIV infections.

Zidovudine is an analog of deoxythymidine in which the 3'hydroxyl has been replaced by an azido (N3) group (Fig. 5).The drug is activated to its mono- di-, and triphosphateforms by cellular enzymes in both HIV-infected and unin-fected cells (155). The triphosphate is a competitive inhibitorof HIV reverse transcriptase, the RNA-dependent DNApolymerase needed for transcription of viral genomic RNAinto double-stranded DNA that can be incorporated into hostcell DNA (Fig. 2). In fact, zidovudine binds to reversetranscriptase much better than does its natural competitordTTP and is thus the preferred substrate (66, 155, 454). Italso inhibits reverse transcriptase at about 1/100 the concen-tration needed for inhibition of cellular DNA polymerasealpha (66, 155, 316). Zidovudine triphosphate is incorporatedinto the growing chain of DNA, and because the azido groupoccupies the 3' hydroxyl site, additional nucleotides cannotattach and the chain is terminated (155, 454).

Zidovudine is active in vitro against mammalian and avianretroviruses at low concentrations (153) and against EBV atconcentrations 10 to 100 times those needed to inhibit HIV(277). It inhibits some bacteria of the Enterobacteriaceaefamily when they are actively dividing (124, 234).

Zidovudine is available in both oral and intravenousforms. It is absorbed rapidly and completely from thegastrointestinal tract and so is well suited to oral use (38). Italso penetrates well into the cerebrospinal fluid and is usefulfor treating central nervous system manifestations of AIDS.The drug is metabolized primarily by hepatic glucuronida-tion, and thus its metabolism may be inhibited by other drugsthat share this pathway, such as sulfa-containing com-pounds, nonsteroidal anti-inflammatory agents, and narcoticanalgesics. Probenecid, another compound that inhibitsglucuronidation, can also be used to prolong the half-life ofzidovudine (247). One of the major drawbacks to the suc-cessful use of zidovudine has been its rapid elimination andshort plasma half-life, necessitating frequent administration.

However, its intracellular half-life is considerably longer (8,155), and more convenient dosing schedules have recentlybeen proven effective. Zidovudine also crosses the placenta(497) and has recently been associated with fetal resorptionwhen administered to mice prior to and early in pregnancy(476). The importance of this for patients remains to be seen.Despite the fact that mammalian cells do not have reversetranscriptases to be inhibited by zidovudine, it is nonethelessa moderately toxic drug. This may be explained by recentobservations. Zidovudine monophosphate competitively in-hibits thymidylate kinase, a cellular enzyme that catalyzesthe conversion of nucleoside monophosphates to diphos-phates (155). As a consequence, when cells are exposed tothe drug, intracellular pools of nucleoside di- and triphos-phates decrease, and cellular DNA synthesis is slowedaccordingly (8). In addition, when zidovudine is incorpo-rated into a growing chain of cellular DNA, it causes chaintermination (8). In patients, zidovudine can cause anemiasevere enough to require transfusion or discontinuation oftherapy. Neutropenia is less frequent, but other reactionssuch as nausea and headache are common, especially athigher doses and in patients with more advanced AIDS (166,379). A myopathy characterized clinically by muscle weak-ness and anatomically by abnormal mitochondria in musclefibers has also been described (88). Patients on zidovudineappear to have a high risk of non-Hodgkin lymphoma (318,354), although whether this susceptibility is due to the drugor simply to prolonged survival in the face of profoundimmunosuppression is not yet clear. The bone marrow-suppressive effects of zidovudine are at least additive (206)and may be synergistic with those of ganciclovir (359). Fewpatients can tolerate concurrent use of these drugs.Zidovudine is useful for treatment of all stages of HIV

infection (Table 1). In patients with AIDS or AIDS-relatedcomplex, high-dose zidovudine (1,200 to 1,500 mg daily)prolongs survival, reduces the frequency of opportunisticinfections, and improves functional capacity, weight gain,and immunologic function (116, 146). Viremia or HIV (p24)antigenemia also decreases during therapy (218, 446). Al-though the survival benefit remains after 1 to 2 years oftherapy (81, 145), the other benefits tend to disappear, witha return of antigenemia and opportunistic infections and adeterioration of immunologic function. This is partly be-cause drug toxicity requires dose reductions or discontinu-ation of therapy in many patients. A recent study has shownthat a lower daily dose, 600 mg, is as beneficial as the higherdose and much less toxic, at least in nonadvanced AIDS(144). It is now the recommended dosage. As little as 300 mgdaily may be beneficial in patients with AIDS-related com-plex (75), although it is not recommended unless patients areunable to tolerate higher doses. High-dose zidovudine is stillused for treatment of HIV neurologic disease (412, 523, 527).The bone marrow-suppressive effects of zidovudine sparethe megakaryocytes and platelets, and the drug can be usedto treat HIV-induced thrombocytopenia (332, 466). Althoughzidovudine reaches high concentrations in semen (199), itdoes not appear to decrease shedding from this site (252),and this lack may have important implications for sexualtransmission of disease.

In asymptomatic or mildly symptomatic HIV patients withCD4 (helper) T-lymphocyte counts of less than 500/mm3,both high- and low-dose zidovudine delayed disease progres-sion and transiently improved immunologic function andantigenemia (147, 487). Adverse effects were also less com-mon in these patients than in those with more advanceddisease. These patients have not been followed long enough

CLIN. MICROBIOL. REV.

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 159

to determine whether zidovudine prolongs survival in earlyHIV infection. A recent cost effectiveness analysis, how-ever, indicates that zidovudine therapy in early HIV infec-tion compares favorably with other well-accepted medicalinterventions. The cost per year of life saved, estimated at$6,553 (1989 dollars), is very similar to that of counselingpatients to quit smoking (415).

Resistance of HIV to zidovudine has been seen in patientsreceiving therapy for prolonged periods (258, 267, 391).Resistance appears to develop more rapidly and to higherlevels in patients with more advanced disease and greaterviral burdens (380), a situation somewhat analogous to thedevelopment of isoniazid resistance in patients with cavitarytuberculosis and high burdens of mycobacteria. The clinicalimportance of resistance is not known; thus far, there hasbeen no correlation with individual patient responses totherapy. At the molecular level, resistance is due to specificchanges in three or four amino acid residues of the reversetranscriptase molecule (267). In patients, high-level resis-tance is associated with accumulation of multiple mutations,and it appears to develop in a stepwise fashion over time.The only cross-resistance observed has been to other 3'-azido-containing nucleosides, suggesting decreased recogni-tion of this moiety by the altered reverse transcriptase (264).

Despite rapid and remarkable advances in the develop-ment and use of zidovudine, many questions remain unan-swered. The minimum effective dose is not yet known, norare the effects of resistance and cumulative toxicity.Whether the drug prolongs survival when given early indisease remains to be seen, and whether it will prevent HIVinfection if given prophylactically after exposure is un-known, though anecdotal experience suggests that this is notthe case (263).

Didanosine

Like zidovudine, didanosine (2',3'-dideoxyinosine [ddI];Fig. 5) is a dideoxynucleoside. It became available formanagement of HIV infections in late 1991, before definitivepatient trials were completed. It is the first compound tofollow a "fast track" to Food and Drug Administrationapproval and be made available prior to completion of safetyand efficacy studies in an attempt to make promising newdrugs quickly available to desperately ill patients.

Didanosine is an analog of deoxyadenosine. It is con-verted by cellular enzymes to ddl monophosphate, then todideoxyadenosine (ddA) monophosphate, to ddA diphos-phate, and finally to ddA triphosphate (ddATP), the activeform (1, 78, 227). Like the triphosphates of other dideoxy-nucleosides, ddATP competitively inhibits HIV reversetranscriptase and acts as a chain terminator for proviralDNA (524). Human DNA polymerase alpha is relativelyresistant to ddATP, but polymerases beta and gamma aremore sensitive to it (496), perhaps accounting for some of thetoxicity seen in patients. In vitro, ddl is less potent againstHIV than dideoxycytidine (ddC) (315), but intracellularlyddATP is as active as zidovudine (344), and it has anintracellular half-life of 8 to 12 h (compared with about 3 hfor zidovudine [1, 227]). Didanosine is also active againstzidovudine-resistant HIV isolates (264).

Didanosine is available in oral form but is somewhatdifficult for patients to take correctly. At acid pH, hydrolysisoccurs at the glycosidic bond between the sugar and the basemoieties of the nucleoside, inactivating the compound. Thedrug is thus supplied as both a buffered powder to be madeinto solution and as buffered chewable tablets which must be

taken at least two at a time to provide adequate bufferingcapacity in the stomach.The major side effects have been a painful peripheral

neuropathy in 16 to 34% of patients treated to date; pancre-atitis, seen in 8 to 9% of patients and occasionally fatal (77,257, 525); and diarrhea in 18 to 34% of patients, possiblyrelated to buffering agents used in the powder (182).Didanosine is avoided in patients at high risk for pancreati-tis, such as those with a history of heavy alcohol consump-tion or previous pancreatitis, and it increases the risk ofpancreatitis when given simultaneously with intravenouspentamidine or ganciclovir. Development of pancreatitis orperipheral neuropathy requires discontinuing the drug.

In phase I uncontrolled dose escalation studies of patientspreviously treated with zidovudine, didanosine therapy re-sulted in significant rises in CD4 cell counts and decreases inp24 antigenemia (50, 77, 257, 525). Phase II controlled trialsare under way to determine whether ddI has any effect onpatient survival, time to progression of disease, or frequencyof opportunistic infections. In view of the desperate need foradditional anti-HIV therapies and because rising CD4 cellcounts are a surrogate marker for disease improvement,didanosine was approved for use in both adult and pediatricHIV-infected patients who are unable to tolerate zidovudine(usually because of bone marrow toxicity) or who experi-ence clinical or immunologic deterioration while receiving it.Zidovudine is still regarded as first-line HIV therapy. Theplace of ddl, both by itself and in combination with otheragents for HIV therapy, will need to be determined in futuretrials.

Investigational Antiretroviral Agents

Many compounds are under investigation as candidatedrugs for the management of HIV infection. It is not possibleto discuss them all, and this section will concentrate on thosewhich are especially promising or represent a new approachto therapy and for which there is adequate description in thescientific literature.The in vitro antiretroviral activity of dideoxynucleosides

other than zidovudine was first described in 1986 (315). ddC(Fig. 5) protected cells against the cytopathic effect of HIVat 1/20 to 1/50 the concentrations needed for ddA, ddl, anddideoxyguanosine, and it was the first of these to beginclinical trials. ddC is well absorbed orally, has a shorthalf-life, and penetrates the central nervous system, al-though not as well as zidovudine (526). Its phosphorylationpathway is different from that of zidovudine, and, unlikezidovudine, it is excreted by the kidneys (526). Its toxicityprofile is also substantially different from that of zidovudine.In early trials, patients were given high doses and many ofthem developed a painful peripheral neuropathy, necessitat-ing discontinuance of the drug. Decreases in p24 antigenemiaand rises in CD4 counts were also seen, however, and nobone marrow toxicity was observed (304, 526). Currently,trials are in progress to determine the efficacy of lower dosesand of dosage schedules that employ zidovudine and ddCtogether, given either simultaneously or in an alternatingfashion (43, 351, 435). Approval of ddC by the Food andDrug Administration is expected in the near future.Two other dideoxynucleosides are currently in clinical

trials. Dideoxythymidinine, also known as D4T, has goodactivity against HIV but is phosphorylated differently fromzidovudine and has less bone marrow toxicity (205, 284).Azidouridine is slightly less active against HIV than zidovu-dine, but is also less toxic to bone marrow (67, 531), and it

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

OHHO oAOH

K No CH2OHICH2CH2CH" CH3

N-BUTYL-DNJ

4H o"t.0

PROTEASE INHIBITOR XVIIFIG. 7. Investigational antiretroviral agents. Ph, phenyl; tBu,

tertiary butyl; N-butyl-DNJ, N-butyl-deoxynojirimycin.

inhibits the virus very well in peripheral blood mononuclearcells. It is currently in phase I trials.

Several nonnucleoside compounds have shown excellentactivity in vitro against HIV reverse transcriptase and arecandidates for patient trials. Among the most potent andspecific are the tetrahydroimidazobenzodiazepinone (TIBO)compounds, benzodiazepine derivatives that inhibit HIVtype 1 (HIV-1) but not HIV-2 or other retroviruses. In vitro,the selectivity index of TIBO R82150 (Fig. 7) is greater than31,000, compared with 6,200 for zidovudine and 191 fordidanosine in the same cell system (339). These agents donot require phosphorylation and inhibit HIV-1 reverse tran-scriptase by an as yet unidentified mechanism. B1-RG-587(nevirapine) is a dipyridodiazepinone with properties similarto those of the TIBO compounds (249, 305). It is beginningphase I trials in the near future. A group of six-substitutedacylouridine derivatives, the HEPT [1-(2-hydroxyethoxy-methyl)-6-phenylthiothymine] congeners, are also highly po-tent and specific for HIV-1, do not require phosphorylation,and appear to interfere with reverse transcriptase by amechanism different from that of zidovudine (10, 12). Onecompound, HEPT-S, also has pharmacokinetic propertiessuitable for oral administration.

Reverse transcriptase inhibitors can prevent new DNAfrom being made in newly infected cells but they cannotprevent the reactivation of HIV from previously infectedcells, as reverse transcriptase is not involved in this process(Fig. 2). Identification of agents capable of interfering withHIV at later points in the replication cycle, and perhapspreventing reactivation, would represent a major advance inthe management of HIV infection. One such group of agentsis the HIV protease inhibitors. HIV reverse transcriptaseand other gag and gag-pol gene products are synthesized asprecursor polypeptides and must be cleaved and processedbefore they can be packaged into virions. HIV protease, anaspartic proteinase encoded by the virus, is responsible forthis processing and is essential for the proper assembly andmaturation of fully infectious HIV-1 virions. It can also be

inhibited by protease inhibitors, small peptide molecules thatmimic the natural substrate for the reaction (97) (Fig. 7).Several such compounds have been identified and are beingdeveloped for clinical use (128, 248, 383). They are highlypotent and selective inhibitors of HIV in vitro, and at leastone has been shown to have antiviral activity when added tocell cultures late (18 to 20 h) after HIV infection and inchronically infected cells (162).Another promising compound, which acts later in the HIV

replication cycle, is N-butyldeoxynojirimycin (Fig. 7). Itreduces infectious virus in vitro in acutely infected cells andeliminates HIV entirely from chronically infected cells (231).The compound works by inhibiting glycosylation of theenvelope glycoprotein gp120, thereby reducing its ability tobind to CD4 receptors and initiate new viral infection.Although glycosylation of viral glycoproteins is nonspecificand can be carried out by cellular enzymes, N-butyl-deoxy-nojirimycin is highly specific for HIV-infected cells in vitro.Phase I trials are under way to determine its clinical utility.Another approach is interference with the viral mRNA

itself. This was first proposed for Rous sarcoma virus in 1978(529) and has been vigorously pursued for the treatment ofmany diverse diseases in addition to viral infections. Itconsists of constructing small oligonucleotides that are com-plementary to specific genes or mRNA sequences (antisenseoligonucleotides). These bind to the nucleic acid, ultimatelypreventing gene expression and protein synthesis. A mole-cule antisense to the mRNA of rev, a critical regulatoryprotein of HIV, has been synthesized, and it inhibits proteinsynthesis in vitro in chronically infected cells (289). Whilethis approach is extremely exciting, several problems witholigonucleotides, including degradation by RNases, cellularpermeability, specificity, and large-scale production, mustbe solved before they can be used as therapeutic agents. Allof these problems are under active investigation (397).

It is also possible to interfere with HIV infection at its firststep, the binding of HIV to CD4 lymphocytes (Fig. 2).Recombinant, soluble CD4 (rsCD4) is the synthetic gpl20binding portion of the complete CD4 molecule (the trans-membrane and cytoplasmic portions are missing). In vitro,rsCD4 binds to gpl20 on the virion surface and preventsattachment of the virion to CD4 lymphocytes (438). Inpatients, no toxicities were seen when the compound wasadministered in phase I trials, but the half-life in serum wasvery short, and sustained changes in p24 antigenemia andCD4 cell counts were not seen (229, 414). Currently, effortsare being directed toward the construction of rsCD4-immu-noglobulin G (IgG) and rsCD4-IgM hybrid molecules thathave longer half-lives (53), may cross the placenta (53), havegreater activity (478), and may recruit antibody-dependent(51) or complement-mediated (478) immune functions todestroy infected cells. Other efforts have resulted in thecoupling of rsCD4 to toxins such as Pseudomonas exotoxinA (64) and the plant toxin ricin (473). When bound to theHIV-infected lymphocyte and internalized, these com-pounds result in cell death. The use of Pseudomonas exo-toxin has been especially effective in vitro and "cures" cellcultures of HIV infection when used in conjunction withreverse transcriptase inhibitors (6).

IMMUNOGLOBULINS

Immunoglobulins are available in three different formula-tions: an intramuscular preparation termed immune globulinor IG; an intravenous form termed IVIG; and several differ-ent intravenous preparations with high titers of antibodies to

H

CH3TIBO R82 150

160 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 161

individual viruses. All are made from pooled human plasmaand contain predominantly IgG, though small amounts ofother globulins are also present. High-titered (hyperimmune)globulins are derived from pooled units of plasma selectedfor high antibody titers to specific viruses. IG has beenavailable for many years, but it cannot be administeredintravenously because of the tendency for IgG to formaggregates and fix complement in vivo. Recent advances inthe cold fractionation procedure used, however, have madeit possible to prevent this aggregation, and newer prepara-tions can be given intravenously. Large volumes of fluidcannot be administered intramuscularly, so the amount ofimmunoglobulin that can be given by this route has also beenlimited. It is possible to give much larger amounts by theintravenous route, although these preparations are ex-tremely costly. Regardless of route of administration, immu-noglobulins are quite safe and are associated with very fewside effects. The mechanism of action is unknown, but in alllikelihood it involves the antibodies present.As will be discussed below, immunoglobulins are more

effective when used prophylactically than therapeutically.For viruses, this is probably because the administered anti-bodies are present during the extracellular phase of infec-tion, when it is possible to neutralize or opsonize virionsbefore they enter cells and are protected from humoralimmunity.

Currently, IG is used primarily for prevention of hepatitisA. It is administered to patients within a few days ofexposure to contaminated food or to an infected person andprevents infection or chemical illness in 80 to 95% ofrecipients (176, 255). It is also given at regular intervals totravelers who will be spending prolonged periods in condi-tions of poor or uncertain sanitation (61, 518). Although IGhas not been unequivocally shown to decrease infection withnon-A, non-B hepatitis, it may be "reasonable" to adminis-ter it to persons who sustain percutaneous exposures toinfected blood (61). IG has not been studied for prevention ofhepatitis C. For prevention of measles, IG should be given topersons who have not had the disease or been vaccinatedagainst it and who are household contacts of measles pa-tients, to those who have been exposed while pregnant, andto those who are immunocompromised when exposed (59).IG does not replace vaccine for control of measles out-breaks.Hyperimmune globulins are used for postexposure pre-

vention of hepatitis B, chickenpox, and rabies. Hepatitis BIG prevents disease in 75% of persons with needlestickexposures (420). It also prevents hepatitis B surface antigencarriage in 85 to 95% of babies born to e-antigen-positivemothers (26, 61). Along with hepatitis B vaccine, hepatitis BIG is given within a few hours of birth to babies of hepatitisB surface antigen-positive mothers. Varicella-zoster IGmodifies the severity of chickenpox but does not preventinfection when given to exposed susceptible persons. Inimmunocompromised children of less than 15 years of age(many of whom have not already had chickenpox and thusare not immune), it is administered as soon as possible afterexposure to chickenpox. It is also given to babies who haveno maternal antibody, including newborns whose mothersdevelop chickenpox within 5 days before or 2 days afterdelivery, premature babies whose mothers are seronegative,and all premature babies of less than 28 weeks of gestation.Varicella-zoster IG is very expensive, the supply is limited,and its use in adults, including exposed seronegative preg-nant women, is limited to those situations in which thephysician seeks to reduce disease severity. It has not been

shown to decrease the frequency of congenital or neonatalvaricella when administered to exposed pregnant womenand, in fact, may do the opposite by making the disease lesssevere in the mother while allowing transmission to the fetus(56). Human rabies IG became available in 1975 and is madefrom the plasma of hyperimmunized human donors. It isused in conjunction with human rabies vaccine to decreasetransmission and death after exposure to rabid animals (55).CMV IG was recently approved for management of CMVinfections in renal transplant patients. When given prophy-lactically, it reduced the frequency of serious CMV diseasein seronegative recipients of seropositive kidneys, althoughit did not decrease transmission of the virus in this setting(442). In bone marrow transplant patients the use of prophy-lactic CMV IG is controversial. In two studies it decreasedinfection rates in seronegative recipients (41, 308), in one itdid not (42), and in one it decreased CMV disease severitybut not infection rates (516). These discrepancies may be dueto differences in antibody titers of the immunoglobulins or tothe fact that small numbers of patients were studied.

Intravenous immunoglobulins were originally developedfor the treatment of primary immune deficiency states. Theyhave, however, been studied and used for many otherpurposes, including treatment and prophylaxis of viral infec-tions. Preparations are available from several different man-ufacturers, and while they are frequently considered equiv-alent, both manufacturing processes and donor populationsdiffer considerably and may result in substantially differentend products. Moreover, manufacturers are not permitted topublish information about antibody titers of their products,so it is difficult to know whether preparations have equalefficacy unless they are directly compared in patient trials.As discussed earlier, IVIG improves survival when used

in conjunction with ganciclovir for treatment of CMV pneu-monia in bone marrow recipients (125, 371). In a recent trial,IVIG also decreased the frequency of graft-versus-hostdisease and interstitial pneumonia when given alone toseropositive bone marrow recipients (462). Although CMVwas not confirmed as the cause of pneumonia in this study,it is the most common cause of interstitial pneumoniafollowing bone marrow transplantation and it is associatedwith graft-versus-host disease. This suggests that IVIG maybe beneficial in this setting.Another important use of IVIG is in the treatment of

chronic enteroviral meningoencephalitis in children withagammaglobulinemia. Given intravenously, immunoglobulinin one study did not reach very high levels in the cerebro-spinal fluid, and children almost always relapsed, thoughthey had responded initially (297). When immunoglobulinwas given through an intraventricular catheter, however,50% of the children responded, and a few appear to havebeen cured of their infection (120, 297). Further studies needto be done, but intraventricular immunoglobulin appears tobe a major advance in the treatment of this devastatingillness.There has been a great deal of interest in the use of IVIG

for the management of HIV infection and its complications.IVIG itself does not contain antibodies to HIV, but it mayprovide functioning antibodies to other infectious agents inpatients who are functionally hypogammaglobulinemic de-spite having elevated serum globulin levels (261). Children,unlike adults, do not have protective antibodies from previ-ous illnesses, and they may benefit from exogenous immu-noglobulins. One large multicenter trial of IVIG therapyrecently demonstrated that symptomatic HIV-infected chil-dren with CD4 counts of greater than 200 per mm3 had

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

significantly fewer serious bacterial infections and fewerhospital days than those treated with placebo (320). A similarbenefit was not seen among those with fewer CD4 cells, butthis may have been because of the small number of childrenstudied. A second trial is under way to further investigatethese findings.No preparation of hyperimmune HIV globulin is available.

A recent trial using a high-titered preparation made from theplasma of two donors with high anti-p24 antibody titersshowed that AIDS patients given the plasma had immediateclearing of p24 antigenemia, fewer symptoms, transient risesin T lymphocytes, fewer opportunistic infections, and lowerrates of HIV isolation from blood (219). Such results areintriguing but have not been confirmed.

IMMUNOMODULATORS

In the strict sense of the word, immunomodulators aresubstances that modify the response of immune competentcells through signaling mechanisms (519). Many discussions,however, including this one, include immune-based thera-pies with a direct effect on cells such as the use of specificantibodies, exogenous immune effector cells, and proce-dures such as bone marrow transplantation. Immunomodu-lators are administered in the hope of augmenting or restor-ing host immune responses to infectious agents ormalignancies.A few older agents, for instance, levamisole, isoprinosine,

and transfer factor, have been used for several years inpatients with viral infections (455). Their effects have neverbeen convincingly demonstrated, however, and they remainlargely investigational. The AIDS epidemic has rekindledinterest in immunomodulators, and with the use of recombi-nant DNA technology it has become possible to manufacturethese compounds in large quantities and to study their effectsin patients, animal models, and in vitro systems. Immuno-modulation in HIV infection (and other viral infections) canbe a two-edged sword, however. Potent immunomodulatorssuch as cytokines have the potential for increasing viralreplication and perhaps triggering the transition from latentto active infection by stimulating target cells and makingthem more capable of supporting viral infection. They mayalso induce new targets by stimulating cells otherwise non-permissive for viral infection. Last but not least, they mayworsen or cause opportunistic infections by stimulating thetarget cells for these organisms. If some of the features ofHIV disease are autoimmune in nature, it is possible thatimmunomodulators will also worsen them (519).Tumor necrosis factor alpha, interleukin 6, and granulo-

cyte-macrophage colony-stimulating factor (GM-CSF) havebeen shown in vitro to induce expression of HIV fromchronically infected cells. Interleukins 1, 2, 3, and 4 andgamma interferon do not, while alpha interferon and trans-forming growth factor beta actually down-regulate HIVexpression (135). Only two immunomodulators, alpha inter-feron and GM-CSF, are commercially available, and thisdiscussion will concentrate on them. Many other agents arein the early phases of investigation for treatment of HIVinfection. Three of these, inosine pranobex (isoprinosine),ditiocarb sodium (diethyldithiocarbamate), and IMREG-1,have been administered to patients in controlled trials andhave shown some possible short-term effects in decreasingprogression of the disease (171, 200, 342). More and betterstudies are needed, however, to establish their utility intreatment of HIV infection. Other agents such as thymichormones (21, 430), enkephalins (333), interleukin 2 (416),

and activated natural killer cells (301) and bone marrowtransplantation (262) are in earlier stages of investigation.One other intriguing approach is that proposed by Salk: theuse of inactivated HIV vaccine or its components in HIV-infected asymptomatic individuals to stimulate cytotoxicresponses to infected cells and eliminate them, preventingprogression of disease and development of full-blown AIDS(403). An early, phase I trial of this approach has shown it tobe safe and immunogenic (370). Patients were followed forless than 1 year, however, and further trials will be neces-sary to determine the effect of postinfection immunization onthe course of HIV disease.

Interferons are natural products, small proteins, or glyco-proteins that are elaborated by eukaryotic cells in responseto viral infection. In turn, they induce in other noninfectedcells biochemical changes that render these cells resistant tosubsequent viral infection. The "antiviral state" induced byinterferons is not specific with respect to infecting virus butis relatively specific with respect to species of host cell oranimal. Interferons were first described by Isaacs and Lin-demann in 1957 (216) and have subsequently been found tohave potent antiproliferative and immunomodulating effectsin addition to their antiviral properties (349). Their primaryfunction is, in fact, related to the regulation of cell growth,differentiation, and immune function (228). Interferons arecurrently being used and actively investigated for the treat-ment of malignancies as well as viral diseases. In naturethere are three types, alpha, beta, and gamma, classified bycell of origin, primary function, and other chemical andphysical properties. Alpha and beta interferons are secretedin response to viral infection and are involved in the re-sponse to it, while gamma interferon is produced by acti-vated T lymphocytes and is involved in the response toantigens and mitogens. While gamma interferon is the pri-mary immunomodulator, all interferons affect the immunesystem to some extent. The only products available com-mercially are alpha interferons, and this review will concen-trate on them.

Interferons are now known to be cytokines, hormonelikepolypeptides that play an important regulatory role in theearly nonspecific phases of the host response to microbialinvasion. They are elaborated by almost all nucleated cellsafter exposure to a variety of stimuli, including viruses, viralantigens, and double-stranded RNA. They are then secretedfrom the infected cells and attach to receptors on other,uninfected cells. After internalization, they trigger the for-mation of enzymes that, upon exposure to double-strandedRNA, degrade viral mRNA and inhibit protein synthesis,resulting in an aborted viral infection. The best studied ofthese enzymes are 2,5-oligo(A), a family of oligonucleotidesthat synthesize an activator of RNase L, an enzyme thatcleaves RNA; and protein kinase, which phosphorylates andthereby inactivates eIF-2, a protein initiation factor (228).These are not the only mechanisms by which virus infectionsare halted, however. The multiplication of HIV, for in-stance, is directly inhibited by interferons, apparently at alate stage of replication involving release of virus particlesfrom the cell membrane (356). While these mechanisms areclear-cut in vitro, the in vivo actions of interferons areundoubtedly more complex because of the effects of inter-ferons on immune functions, including stimulation of naturalkiller cells and of B-lymphocyte proliferation, inhibition oflymphocyte blastogenesis, and enhanced expression of ma-jor histocompatibility class I receptors on cell surfaces (240).Interferons are responsible for the fever, chills, arthralgias,

162 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 163

and myalgias frequently seen with viral infections, and theymay actually cause immune-mediated tissue damage.Most interferons in use today are made by using recom-

binant DNA technology. They are not absorbed from thegastrointestinal tract when given orally and thus are admin-istered by intramuscular or subcutaneous injection (512).The serum half-life is short, but the antiviral state induced incells can last several days (228).The most common early adverse effect is an influenzalike

illness characterized by fever, headache, myalgias, andarthralgias. These symptoms can usually be controlled withmedication, and tolerance soon develops, so that therapycan be continued. Later, however, fatigue, anorexia, andweight loss can develop, and these often limit the use of thedrug. Other adverse effects have been described, thoughthey are much less common: bone marrow toxicity, psycho-logical abnormalities (376), and cardiac toxicities (445).Interferon neutralizing antibodies are produced in 2 to 3% ofpatients receiving systemic therapy (447). While the signifi-cance of these antibodies is not completely understood, theirpresence has not yet been associated with a diminishedresponse in patients being treated for viral infections.

Interferons were first used clinically for the managementof herpesvirus infections in immunocompromised patients.Although they were useful in controlling the complicationsof chickenpox and zoster (5, 303, 515) and in preventing thereactivation of CMV after renal transplantation (203), theyhad no effect on prevention of CMV after bone marrowtransplantation (307) and were not as effective as acyclovirfor treatment of genital herpes (256, 340). In addition, thefrequency of adverse effects was high, and they were soonreplaced by acyclovir and ganciclovir.Because the common cold is caused by a variety of

different viruses, a nonspecific agent such as interferonwould seem an ideal mode of treatment and/or prophylaxis.Leukocyte interferon was first administered intranasally forthis purpose in 1973 (302). Unfortunately, it produced a veryhigh frequency of local side effects such as nasal stuffiness,dryness, and bleeding and actually prolonged cold symptomsrather than shortening them (190). When given prophylacti-cally to exposed family members, intranasal recombinantinterferon decreased the frequency of rhinovirus colds, but ithas little effect on colds due to other viruses (115, 187), andit was again associated with very bothersome side effects. Ithas now been abandoned for this purpose.

Anogenital warts (condyloma acuminata) lend themselvesto local, intralesional therapy, and interferons may be usefulfor this purpose. Compared with placebo, they promotefaster healing and fewer recurrences both in untreatedpatients and in those with refractory warts (132, 152, 375).Side effects were less common and less severe than inpatients receiving systemic therapy, although intralesionalinterferon is absorbed systemically. Interferons used inconjunction with other local therapies such as podophyllin(113) and laser vaporization (480) also appear superior tothese agents alone. The role of intralesional interferon ther-apy in genital warts is not yet clear. Such therapy is veryexpensive, and 20 to 30% of patients still suffer recurrences.Intralesional injections are painful, limiting the number ofwarts than can be treated. Cervical warts are very difficult toinject and may not be treatable by this mode.

In adults with chronic hepatitis B, a lack of endogenousinterferon may play a role in failure to clear the acute viralinfection and in progression to chronic disease (94). Treat-ment with exogenous interferons has been under investiga-tion for several years. Earlier trials suggested that the best

results were achieved by pretreating patients with pred-nisone, stopping this treatment, and then initiating interferontherapy during the rebound from corticosteroid therapy(347). A recent well-controlled trial, however, shows thatthis approach is effective in only a small subset of hepatitispatients with minimal liver dysfunction (348). Another largersubset of sicker patients responded to long-term (4-month)treatment with interferon alone, showing improvements inliver function and in abnormal histology and disappearanceof hepatitis B e antigen, surface antigen, and DNA from theserum (348). A recent long-term follow-up study of treatedpatients indicated that many of these remissions were sus-tained for 3 to 7 years and may, in fact, represent cures of thedisease (209, 245). In total, however, only 30 to 40% ofchronic hepatitis B patients respond to interferon, and whenlower doses are given, the response rate is lower (280).Further studies are needed to better determine which pa-tients will benefit from interferon therapy and whether it willhave any effect on long-term progression of disease, onsurvival, and on the frequency of subsequent hepatocellularcarcinoma.

In recent studies of interferon for chronic hepatitis C,about 50% of treated patients responded with decreases inliver function abnormalities, hepatic inflammation, and viralRNA in serum (93, 107, 427). Six months later, 50% of thoseresponding had relapsed, resulting in a sustained responserate of only 20 to 25%. Although alpha interferon 2b has beenapproved for treatment of chronic hepatitis C (Table 1),many important questions remain about optimum dose andduration of treatment and the need for maintenance therapy(106). Better ways to identify patients likely to respond arealso needed. In view of the cost and side effects of inter-feron, it should not be routinely used in hepatitis C untilthese problems are solved.

Interferons have been known for some time to inhibitanimal retroviruses, and they were shown in 1985 to inhibitHIV (204). In latently infected cells, interferons appear toact at a late point in virus replication to inhibit virionassembly and/or release (356). In acute infection, they pre-vent new infection of cells (204) and may act at an earlierpoint in replication. In mature macrophages, interferonsappear to limit the formation of proviral DNA (246). Whenadministered to AIDS patients for treatment of Kaposisarcoma, interferon resulted in decreased p24 antigenemiaand increased CD4 cells, although there was a "rebound"increase in p24 in some patients when the drug was stopped(105, 260). A subsequent placebo-controlled trial in asymp-tomatic HIV patients confirmed the ability of interferon todecrease viremia and perhaps opportunistic infections in thispopulation (259). It also appears that interferons work bestwhen given in early infection, while the immune system isstill reasonably intact. The role of interferons, includingthose produced endogenously in HIV infection, is complex,however, and not completely understood. It may involvemechanisms by which the virus is able to avoid the effects ofthe drug, such as down-regulation of alpha interferon recep-tors on cell surfaces after prolonged exposure to these agents(272). As noted earlier, interferons also fail to cross theblood-brain barrier and can be associated with intolerableside effects, especially in those asymptomatic patients whostand to benefit the most from them (259). For these reasons,use of interferons alone for HIV infection may be proble-matic, and they are probably best used in combination withother agents such as zidovudine and other dideoxynucleo-sides that inhibit viral replication at a different stage. Invitro, alpha interferon and zidovudine are synergistic in

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

inhibiting HIV (183). In early, dose-finding patient trials, ithas been possible to administer both agents together in dosesthat appear to have an antiviral effect, although bone marrowtoxicity has been dose limiting in many cases (31, 250, 254).Much work remains to be done in this area, includinginvestigation of gamma interferons for therapy of AIDS anduse of additional agents, such as CSFs to alleviate thedose-limiting toxicities of such combinations as zidovudineand interferon.CSFs are naturally occurring cytokines that stimulate the

reproduction and differentiation of granulocytes (G-CSF) orgranulocytes, monocytes, and macrophages (GM-CSF) frombone marrow precursor cells. They also enhance the effec-tiveness of mature cells by, for instance, preventing neutro-phil migration, potentiating antigen processing by macro-phages, and stimulating phagocytosis of yeast cells andbacteria (499). Both G-CSF and GM-CSF are availablecommercially and are produced in Escherichia coli or inyeast cells by using recombinant DNA techniques. Theywere originally developed for prevention and treatment ofneutropenias induced by cancer chemotherapy and bonemarrow transplantation. A recent study indicates that theyare extremely useful for this purpose, even reducing thenumber of hospital days required following autologous mar-row transplantation (321).A great deal of interest has also developed in using them to

control the complications of HIV infection, especially theneutropenias resulting from antiretroviral therapy. CSFshave no direct effect on HIV and do not affect HIV-infectedlymphocytes. In monocytes, however, GM-CSF stimulatesvirus replication in vitro (251, 345), an effect consistent withthe cell stimulatory properties of cytokines discussed earlier.If zidovudine is added to these in vitro systems, its potencyis enhanced, and much lower concentrations are required toinhibit HIV than in the absence of GM-CSF (345). This effectis probably due to enhanced drug phosphorylation by thestimulated cells, and while it may be useful in the treatmentof AIDS patients, it also raises the possibility of increasedzidovudine-induced bone marrow toxicity when both drugsare administered simultaneously. These issues have not beenresolved, but they have not prevented substantial progress inthe use of CSFs.Groopman and colleagues first showed that GM-CSF was

biologically active in raising granulocyte counts in leuko-penic AIDS patients (173) and went on to demonstrate thatthe induced neutrophils functioned normally in killing Staph-ylococcus aureus. The same investigators also identified twopatients with defective bacterial killing mechanisms (one inphagocytosis and one in intracellular killing), but these, too,appeared to be corrected by GM-CSF (14). Subsequentstudies have shown both G-CSF and GM-CSF to be veryeffective in correcting the neutropenias seen with zidovudinetherapy alone (311) or combined with interferon, and the twoCSFs have allowed treatment to continue in patients other-wise unable to tolerate it because of bone marrow toxicity(92, 408). This has been true of patients with advanced (408)as well as early (92) HIV disease. There is also preliminaryevidence that GM-CSF may ameliorate the neutropeniaassociated with ganciclovir therapy (181). The doses of CSFsneeded in AIDS patients are usually much lower than thoseused in cancer patients, and the side effects are more closelyrelated to the use of zidovudine and interferon. CSFs are,however, prohibitively expensive. In the dosages used forcancer patients, the cost is approximately $200 per day (300).Whether or not it will be lower in AIDS patients remains tobe seen. In the face of this, it is important for future studies

to identify the patients most likely to benefit from theseexpensive agents.

PROBLEMS OF ANTWIRAL THERAPY

ResistanceUnlike bacteria, viruses do not have a wide variety of

mechanisms for developing resistance to chemotherapeuticagents. They are genetically simple and metabolically inert,and changes like those seen in bacteria, such as decreaseduptake of antibiotics, acquisition of resistance plasmids, orinduction of inactivating enzymes, have not been seen.Viruses do, however, have two properties that allow them toevade potential inhibitors: they have the ability to replicateto high titers in host cells, and they can mutate very rapidly.As a consequence, most viral resistance arises from geneticmutation, giving rise to changes in either enzymes or struc-tural components of the virion. The success of this mecha-nism can be seen in cell culture, where viruses readilydevelop resistance after only a few passes in the presence ofan inhibitory agent (for a review, see reference 140). Incontrast, very few resistant viruses have been isolated fromimmunocompetent patients being treated for viral diseases(19, 126). Resistant herpesviruses are isolated from patientsimmunocompromised by cancer chemotherapy or transplan-tation, although this is relatively infrequent. In addition,most of these resistant viruses appear to have reducedvirulence and to cause predominantly indolent, non-life-threatening infections (19, 85, 428). There are however,increasing reports of AIDS patients with life-threatening orvery serious illnesses due to resistant herpesviruses (127,131, 164, 288), suggesting that severely immunocompro-mised patients may provide a milieu in which mutant virusesare not at a selective disadvantage and can prosper. Inaddition, a larger viral load creates a larger gene pool fromwhich mutations can emerge. Under these circumstances,selective pressure from antiviral agents could result in thepropagation and perhaps the transmission of resistant vi-ruses, eventually replacing wild-type susceptible popula-tions with more resistant ones.

Despite what is known about viral resistance in vitro, agreat deal remains to be learned about its importance andabout the management of patients infected with resistantviruses. A summary of unanswered questions follows.Under what circumstances does resistance arise? In gen-

eral, it seems that the greatest risk is in severely immuno-compromised patients receiving prolonged courses of anti-viral therapy, but the details of the relationship amongresistance, severity of underlying disease, and treatment areunknown. Although the herpesviruses do not become resis-tant easily in immunocompetent patients (325), influenzaviruses do (188). The reasons for this difference are notknown.What are the properties of the resistant viruses? Virulence

may be reduced, but this appears to depend on the host, atleast among herpesviruses. Rimantadine-resistant influenzaA virus strains, on the other hand, are as virulent for ferrets(464) and for people (188) as their wild-type counterparts.Whether resistant viruses can establish latency is anotherimportant but unanswered question. This ability may alsodepend on the host and the mechanism of resistance. Like-wise, little is known about transmissibility of resistant vi-ruses. To date, transmission of resistant herpesviruses hasnot been documented, but it may only be a matter of time,unless resistance is accompanied by reduced transmissibil-

164 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 165

ity. Resistant influenza viruses, on the other hand, appear tobe readily transmitted, at least in a household setting (188).Another very important issue is cross-resistance amongantiviral drugs. A great deal is known about in vitro cross-resistance, and a few clinically useful patterns haveemerged, such as cross-resistance of zidovudine-resistantHIV to other azido-containing nucleosides and susceptibilityof thymidine kinase-deficient HSV to drugs not needingphosphorylation or not requiring a virus-induced enzyme forphosphorylation. A great deal remains to be learned, how-ever. At least 20 different DNA polymerase mutants havebeen found in vitro among HSV variants, for instance, andmost of them possess altered drug-binding properties. Resis-tance to one compound is usually accompanied by decreasedsusceptibility to other drugs of the same class, but predict-able cross-resistance among different classes of drugs hasalso been found and is thought to indicate overlap ofdrug-binding sites (71). Exceptions to known patterns alsoexist, further confirming the complexity of resistance due toDNA polymerase changes. Consequently, predicting cross-resistance in DNA polymerase mutants may not be possibleunless the viruses are carefully characterized.What is the result of the development of drug resistance in

the patient? As discussed above, the appearance of zidovu-dine-resistant HIV has not yet been associated with clinicaldeterioration in individual patients, although it may play arole in the reduced effectiveness of zidovudine after manymonths of therapy. Failure of herpes lesions to heal and evenserious illness are frequently due to acyclovir-resistantherpesviruses in AIDS patients, but this is not true in otherimmunocompromised patients in whom failure to heal mayor may not be due to acyclovir resistance and in whomlesions due to resistant virus may heal despite the lack ofeffective therapy (470). Rimantadine prophylaxis againstinfluenza A was ineffective in persons with household expo-sures to rimantadine-treated patients, but the influenzalillness itself (due to rimantadine-resistant influenza) wastypical and resolved in the usual self-limited fashion (188).Thus, viral resistance is clinically important in some, but notall, situations and much remains to be learned.What are the most reliable ways of identifying resistance

in the laboratory? Traditional research methods that involvepropagation of viruses in cell culture, exposure to inhibitoryagents, and measurement of subsequent changes in host cellsor in amount of virus produced are time-consuming andexpensive. They may also be unreliable in that they canselect for virus strains that grow well in cell culture but arenot necessarily representative of the virus populationpresent in the patient. It is increasingly appreciated thatviruses isolated from patients are not clonal derivativeshomogeneous in their properties but instead are heteroge-neous populations of viruses with differing drug susceptibil-ities and other properties, including the ability to grow indifferent cell lines (123, 149). As a result, new methods ofmeasuring resistance are being developed for the researchlaboratory, including use of the polymerase chain reactionfor detection of resistance genes, cell lines which allowgrowth of a broader spectrum of viral strains, and improvedmethods of nucleic acid hybridization (226, 264, 266). It ishoped that these can be adapted to the clinical laboratory sothat patients can benefit directly from the improved meth-ods.What can be done to minimize problems due to drug

resistance? As with tuberculosis, it is hoped that combina-tion therapy in HIV disease will forestall or even preventclinical manifestations of drug resistance (see below). Other

measures that have been successful in herpesvirus-infectedpatients include avoiding therapy with prolonged courses ofdrugs at "subinhibitory" levels and discontinuation of ther-apy whenever possible.

LatencyWith the possible exception of a new agent that may

"cure'" cell cultures of HIV infection (see N-butyl-deoxyno-jirimycin, above), antiviral agents are virustatic rather thanvirucidal. In vitro, such agents are most active againstactively proliferating viruses, and when these agents areremoved from cell cultures, virus replication resumes. Inpatients, a course of treatment with acyclovir does notprevent future recurrences of HSV, nor does zidovudineprevent progression of AIDS, indicating that these drugs donot eliminate these viruses when they are in the latent state.Furthermore, the presence of latent virus in treated patientscould afford the opportunity for prolonged contact betweenvirus and drug, with attendant opportunities for develop-ment of drug resistance (140). The successful treatment oflatent viral infections will depend, as it does in acute viralinfections, on the identification of specific viral processesthat can be specifically attacked by antiviral agents. Re-cently, a great deal of progress has been made in defining themolecular events associated with latent HSV infections. Agroup of RNAs, termed latency-associated transcript, hasbeen described and possibly linked to reactivation of latentHSV (201); for a review, see reference 456. Advances suchas these should eventually make it possible to identify agentscapable of interfering with viruses in the latent state.

Immunosuppression by Antiviral Agents

Because antiviral agents are virustatic but not virucidal,host immune responses remain critical to the recovery ofpatients from viral infections. As discussed above, manyantiviral agents have antiproliferative activity against rapidlydividing cells, and inhibition of host immune responses hasalways been a concern in antiviral therapy. A recent directcomparison of the effects of several antiviral agents on theproliferative responses of peripheral blood mononuclearcells from healthy individuals indicated that zidovudine,ganciclovir, and ribavirin decreased mitogenesis, while acy-clovir and didanosine had no effect (192). Another studyshowed that zidovudine, didanosine, and ddC had no inhib-itory effect on the bactericidal activity of polymorphonuclearleukocytes and possibly enhanced it (389). Further studiesare needed, but these trials demonstrate the need for iden-tifying the immunosuppressive properties of individual anti-viral agents, establishing their importance in patients, andusing this information to individualize patient therapy.

PROSPECTS FOR THE FUTURE

LiposomesLiposomes are synthetic phospholipid vesicles that can be

used as carriers of many biologic molecules, includingantitumor and antimicrobial agents, immunomodulators,toxins, and enzymes. When administered to animals orhumans, they are naturally distributed to the reticuloendo-thelial organs and cleared by phagocytic cells, especiallyblood mononuclear cells and tissue macrophages. Lipo-somes can also be targeted to specific cells by incorporatingthe appropriate antibodies into them. For these reasons,

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

they are ideal agents for delivery of drugs to intracellularorganisms such as viruses. Although no patient trials ofantiviral therapy have yet been reported, animal and in vitrostudies are intriguing and encouraging. Liposomes havebeen used to increase the cellular permeability of highlyionized drugs such as foscarnet (468) and to lengthen thehalf-life of ddC in the central nervous system of rats (237).HSV-infected cells can be selectively destroyed by lipo-somes encapsulated with acyclovir and tagged with anti-HSV antibodies (324) or by mononuclear phagocytes acti-vated by liposomes encapsulated with immunomodulatorssuch as gamma interferon or macrophage-activating factor(243). Of importance, CD4-bearing liposomes interact withHIV-infected lymphocytes and deliver their contents to theinterior of these cells (87). This raises the possibility ofloading these liposomes with antiviral agents such as reversetranscriptase inhibitors and using them to deliver the drugsto the interior of HIV-infected cells. HIV-1 also fusesdirectly with negatively charged liposomes, and its infectiv-ity is thereby decreased (244), another promising approachto treatment of HIV.

Combination Therapy

Prompted by the success of combination chemotherapyfor cancer and stimulated by the need for effective treatmentof HIV infection, combination therapy has come to theforefront of antiviral treatment. The purpose is threefold: toenhance the efficacy of single-agent therapy, to minimizetoxicity by reducing individual drug dosages, and to preventor forestall the development of drug resistance. There aremany reports of drug combinations synergistic in vitroagainst HIV and other viruses. Many patient trials are alsounder way, but few have been completed and most data arepreliminary. The most promising combinations consist of anonnucleoside and a nucleoside inhibitor of HIV reversetranscriptase or of two drugs active at different sites of viralreplication. Different toxicities are also very important. InAIDS patients with Kaposi sarcoma, for instance, zidovu-dine and alpha interferon together showed early evidence ofsuppressing antigenemia more effectively than either drugalone and, possibly, of delaying drug resistance (31, 148).ddC is a reverse transcriptase inhibitor like zidovudine, butit is phosphorylated by a different mechanism and hasdifferent toxicities. When ddC is administered with zidovu-dine in an alternating regimen, less toxicity is seen than witheither agent alone at full doses (43, 351, 435). Very excitingresults have been obtained by giving GM-CSF to patientswho develop neutropenia on zidovudine-interferon regi-mens. GM-CSF rapidly alleviated the neutropenia and al-lowed patients to remain on this very effective regimen whenthey would otherwise have been unable to tolerate it (92,408). One recent trial demonstrated what appears to be anadditive effect of zidovudine and foscarnet in suppressingp24 antigenemia (225). Although it has no activity againstretroviruses, acyclovir can be added to zidovudine to controlconcomitant HSV infections. If herpesviruses, especiallyCMV, can transactivate HIV or induce cellular receptors forit, as has been shown in vitro (95, 295; for a review, seereference 273), control of herpesvirus infections may be-come an important part of HIV management. Despite theenthusiasm for combination therapy, adverse interactionsmust be closely monitored, and prospective combinations ofagents must be tested in vitro and in animals before they aregiven to patients. Ribavirin, for instance, is phosphorylatedby the same enzymes as zidovudine and in vitro inhibits its

effectiveness against HIV (11, 486). In vitro, when didanos-ine was added to zidovudine, the combination synergisticallyinhibited HIV, but at high concentrations it also additivelyinhibited bone marrow progenitor cells (111), indicating thatcareful dosage adjustment will be needed in treating patientswith this combination.

Computer-Aided Drug Design

Traditionally, the search for antiviral agents has consistedof randomly screening compounds for viral inhibitory activ-ity in cell culture, identifying a few, making structuralmodifications in them, and retesting for changes in activity.Computer-aided drug design uses X-ray crystallography todetermine the three-dimensional structures of viral macro-molecules and then employs computer simulation and so-phisticated thermodynamic computations to study atomicinteractions with other molecules and to delineate the struc-tures of those with favorable binding characteristics (290). Inthis way, the effect of structural modifications can be deter-mined and promising compounds can be identified before aninvestment is made in synthesizing and testing them. Thefirst human virus to be studied in this fashion was the humanrhinovirus. Its three-dimensional atomic structure was de-scribed by Rossmann and colleagues in 1985 (396). In 1986,the interaction of the virus with a known picornavirusinhibitor, WIN 51711, was described: the drug was boundwithin a deep depression on the virion surface and inducedconformational changes that increased stability and pre-vented uncoating of the virus (150, 441). A series of com-pounds with similar bridging properties were subsequentlysynthesized and studied (13), and some of them are currentlybeing tested in patients for the treatment of rhinovirus colds.The structure of the influenza virus hemagglutinin and itsinteraction with its cellular receptor, sialic acid, have alsobeen described and suggest that agents that would interferewith attachment of the influenza virus to cells could bedesigned (498). It has been proposed that HIV has a surfacesimilar to that of rhinoviruses and thus might also beinhibited at the uncoating step (395). DesJarlais and col-leagues (104) have recently used the structure of HIV-1protease and a computer-assisted searching program toidentify and study potential inhibitors of this enzyme. Inaddition, the structure of HIV reverse transcriptase itself isbeing studied by X-ray crystallography in the hope ofidentifying specific inhibitory compounds (268). Computer-assisted drug design is still in its infancy, and no antiviralagents have yet been designed de novo by this technique.Nevertheless, it is advancing rapidly and holds great promisefor the efficient, selective identification of new drugs.

Role of the Clinical Microbiology LaboratoryMost antiviral agents in use have a narrow spectrum of

activity, and many of them cause substantial toxicity at theusual dosages. Consequently, empiric patient therapy withantiviral agents is seldom possible, although it is a commonpractice with antibacterial agents. Identification of a specificviral pathogen is usually needed, and although antiviralsusceptibility testing is currently only necessary in referralcenters, the rising number of HIV-infected patients and theincreasing use of antiviral agents will eventually demand thatit be available in general and community hospitals as well.

Recent advances in diagnostic methodology have made itpossible to identify many common viral pathogens frompatient specimens within a few hours to a few days (440).

166 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 167

Standard cell culture is still necessary for identification of allpossible pathogens if no specific virus is suspected of caus-ing a given illness. When only a single or a small number ofviruses is implicated, however, immunofluorescent or en-zyme immunoassays can frequently be performed directlyon patient specimens and can detect the presence of viralantigens within a few hours. RSV is commonly identified inthis way.

In other cases, shell vials are used to allow a period ofviral amplification before identification is attempted. Thespecimen is centrifuged onto a monolayer of cells grown ona coverslip in the bottom of a 1-dram (3.7-ml; shell) vial.Nutrient medium is added and the vial is incubated, usuallyfor 18 to 72 h. The coverslip is then removed, the cells arefixed, and, most commonly, a fluorescein-conjugated anti-body is added to detect the presence of viral antigen in thecells. HSV, VZV, CMV, and influenza viruses, as well assome others, can be identified in 1 to 3 days by using thismethod.Almost no convenient, practical methods for antiviral

susceptibility testing are available to the clinical laboratory.The plaque reduction assay, a method commonly used bythe research laboratory to measure antiviral activity (86,189), is time-consuming, cumbersome, and difficult to repro-duce. In addition, it depends on the ability of the virus togrow in a given cell line and thus may select virus popula-tions capable of replicating well in vitro but not necessarilyrepresentative of the population infecting the patient. Manyother methods have been used to quantitate viral replicationin the presence of inhibitory agents including nucleic acidhybridization (160), shell vials with immunofluorescentstaining (470), immunoperoxidase staining (211), enzymeimmunoassay (366), dye uptake assay (299), and yield reduc-tion assay (360). None of these methods has been standard-ized among laboratories, however. One method of nucleicacid hybridization for HSV susceptibility has been madecommercially available in kit form. It compares well with theplaque reduction assay and involves wicking of lysed, virus-infected cells onto membranes followed by hybridizationwith a radioiodinated DNA probe (465). Although thismethod is more adaptable to clinical laboratories than manyothers and is in use in some, 3 to 4 days are required forsusceptibility testing after the virus is isolated in cell cultureand radioisotopes, frequently a disposal problem for micro-biology laboratories, are still employed. As the demand forantiviral susceptibility testing grows, it is hoped that practi-cal diagnostic techniques will also become available.

CONCLUSIONS

Although early progress in antiviral therapy was slow,rapid advances have been made in the last decade. As ourunderstanding of viral replication increases, more and moreviral processes are being identified as possible targets forinhibition by antiviral compounds. Immunomodulators arebeing used increasingly to augment the immune response inthe treatment of viral diseases. Resistance to antiviral drugshas been noted among diverse groups of viruses but thus farhas caused treatment failure primarily in highly immunosup-pressed patients, such as those with AIDS. It is hoped thatcombination antiviral therapy, in addition to improving theeffectiveness of individual drugs, will limit the impact ofresistance on patient care. Other new approaches, such asthe use of liposomes to deliver antiviral drugs intracellularlyand the use of computers to identify potential new agentsrapidly and inexpensively, should result in increasingly safe

and effective antiviral drugs. With advances in laboratorytechnology making it possible to quickly identify virusesand, in the near future, perform susceptibility tests on them,antiviral therapy will rapidly become a routine part ofmedical care.

ACKNOWLEDGMENTS

I am indebted to Doris Hart and Sherry Hall for manuscriptpreparation; to Bruce Coary, Lourdes Aquino, and Eutiquio Chavezfor bibliographic assistance; and to Herb Comess for manuscript,graphics, and bibliographic preparation.

REFERENCES

1. Ahluwalia, G., D. A. Cooney, H. Mitsuya, A. Fridland, K. P.Flora, Z. Hao, M. Dalal, S. Broder, and D. G. Johns. 1987.Initial studies on the cellular pharmacology of 2',3'-dideoxyi-nosine, an inhibitor of HIV infectivity. Biochem. Pharmacol.36:3797-3800.

2. Ahluwalia, G., M. A. Johnson, A. Fridland, D. A. Cooney, S.Broder, and D. G. Johns. 1988. Cellular pharmacology of theanti-HIV agent 2',3'-dideoxyadenosine. Proc. Am. Assoc.Cancer Res. 29:349.

3. Andersson, J., S. Britton, I. Ernberg, U. Andersson, W. Henle,B. Skoldenberg, and A. Tisell. 1986. Effect of acyclovir oninfectious mononucleosis: double-blind, placebo-controlledstudy. J. Infect. Dis. 153:283-290.

4. Apperley, J. F., R. E. Marcus, J. M. Goldman, D. G. Wardle,P. J. Gravett, and A. Chanas. 1985. Foscarnet for cytomega-lovirus pneumonitis. Lancet i:1151.

5. Arvin, A. M., J. H. Kushner, S. Feldman, R. L. Baehner, D.Hammond, and T. C. Merigan. 1982. Human leukocyte inter-feron for the treatment of varicella in children with cancer. N.Engl. J. Med. 306:761-765.

6. Ashorn, P., B. Moss, J. N. Weinstein, V. K. Chaudhary, D. J.FitzGerald, I. Pastan, and E. A. Berger. 1990. Elimination ofinfectious human immunodeficiency virus from human T-cellcultures by synergistic action of CD4-pseudomonas exotoxinand reverse transcriptase inhibitors. Proc. Natl. Acad. Sci.USA 87:8889-8893.

7. Astra Pharmaceutical Products, Inc. 1991. Foscavir. Packageinsert. Astra Pharmaceutical Products, Inc., Westborough,Mass.

8. Avramis, V. I., W. Markson, R. L. Jackson, and E. Gomperts.1989. Biochemical pharmacology of zidovudine in humanT-lymphoblastoid cells (CEM). AIDS 3:417-422.

9. Aweeka, F., J. Gambertoglio, J. Mills, and M. A. Jacobson.1989. Pharmacokinetics of intermittently administered intrave-nous foscarnet in the treatment of acquired immunodeficiencysyndrome patients with serious cytomegalovirus retinitis. An-timicrob. Agents Chemother. 33:742-745.

10. Baba, M., E. DeClercq, S. lida, H. Tanaka, I. Nitta, M.Ubasawa, H. Takashima, K. Sekiya, K. Umezu, H. Nakashima,S. Shigeta, R. T. Walker, and T. Miyasaka. 1990. Anti-humanimmunodeficiency virus type I activities and pharmacokineticsof novel 6-substituted acyclouridine derivatives. Antimicrob.Agents Chemother. 34:2358-2363.

11. Baba, M., R. Pauwels, J. Balzarini, P. Herdewijn, E. DeClercq,and J. Desmyter. 1987. Ribavirin antagonizes inhibitory effectsof pyrimidine 2',3'-dideoxynucleosides but enhances inhibi-tory effects of purine 2',3'-dideoxynucleosides on replicationof human immunodeficiency virus in vitro. Antimicrob. AgentsChemother. 31:1613-1617.

12. Baba, M., H. Tanaka, E. DeClercq, R. Pauwels, J. Balzarini, D.Schols, H. Nakashima, C. F. Perno, R. T. Walker, and T.Miyasaka. 1989. Highly specific inhibition of human immuno-deficiency virus type I by a novel 6-substituted acyclouridinederivative. Biochem. Biophys, Res. Commun. 165:1375-1381.

13. Badger, J., I. Minor, M. J. Kremer, M. A. Oliveira, T. J.Smith, J. P. Griffith, D. M. A. Gujerin, S. Krishnaswamy, M.Luo, M. G. Rossmann, M. A. McKinlay, G. D. Dina, F. J.

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

Dutko, M. Fancher, R. R. Rueckert, and B. A. Heinz. 1988.Structural analysis of a series of antiviral agents complexedwith human rhinovirus 14. Proc. Natl. Acad. Sci. USA 85:3304-3308.

14. Baldwin, G. C., J. C. Gasson, S. G. Quan, J. Fleischmann, R.Weisbart, D. Oette, R. T. Mitsuyasu, and D. W. Golde. 1988.Granulocyte-macrophage colony-stimulating factor enhancesneutrophil function in acquired immunodeficiency syndromepatients. Proc. Natl. Acad. Sci. USA 85:2763-2766.

15. Balfour, H. H., B. Bean, 0. L. Laskin, R. F. Ambinder, J. D.Meyers, J. C. Wade, J. A. Zaia, D. Aeppli, L. E. Kirk, A. C.Segreti, R. E. Keeney, and the Burroughs Wellcome Collabora-tive Acyclovir Study Group. 1983. Acyclovir halts progressionof herpes zoster in immunocompromised patients. N. Engl. J.Med. 308:1448-1453.

16. Balfour, H. H., B. Bean, C. D. Mitchell, G. W. Sachs, J. R.Boen, and C. K. Edelman. 1982. Acyclovir in immunocompro-mised patients with cytomegalovirus disease. Am. J. Med.73(Suppl. 1A):241-248.

17. Balfour, H. H., B. A. Chace, J. T. Stapleton, R. L. Simmons,and D. S. Fryd. 1989. A randomized, placebo-controlled trial oforal acyclovir for the prevention of cytomegalovirus disease inrecipients of renal allografts. N. Engl. J. Med. 320:1381-1387.

18. Balfour, H. H., J. M. Kelly, C. S. Suarez, R. C. Heussner, J. A.Englund, D. D. Crane, P. V. McGuirt, A. F. Clemmer, andD. M. Aeppli. 1990. Acyclovir treatment of varicella in other-wise healthy children. J. Pediatr. 116:633-639.

19. Barry, D. W., S. Nusinoff-Lehrman, M. N. Ellis, K. K. Biron,and P. A. Furman. 1985. Viral resistance, clinical experience.Scand. J. Infect. Dis. 47:155-164.

20. Bauer, D. J., L. St. Vincent, C. H. Kempe, and A. W. Downie.1963. Prophylactic treatment of smallpox contacts with N-me-thylisatin beta-thiosemicarbazone. Lancet ii:494-496.

21. Beall, G., S. Kruger, F. Morales, D. Imagawa, J. A. Goldsmith,D. Fisher, J. Steinberg, J. Phair, S. Whaling, and J. Bitran.1990. A double-blind, placebo-controlled trial of thymostimulinin symptomatic HIV-infected patients. AIDS 4:679-681.

22. Bean, B., and D. Aeppli. 1985. Adverse effects of high-doseintravenous acyclovir in ambulatory patients with acute herpeszoster. J. Infect. Dis. 151:362-363.

23. Bean, B., C. Braun, and H. H. Balfour. 1982. Acyclovirtherapy for acute herpes zoster. Lancet ii:118-121.

24. Bean, B., C. Fletcher, J. Englund, S. NusinoffLehrman, and M.Ellis. 1987. Progressive mucocutaneous herpes simplex infec-tion due to acyclovir-resistant virus in an immunocompro-mised patient: correlation of viral susceptibilities and plasmalevels with response to therapy. Diagn. Microbiol. Infect. Dis.7:199-204.

25. Bean, W. J., S. C. Threlkeld, and R. G. Webster. 1989. Biologicpotential of amantadine-resistant influenza A virus in an avianmodel. J. Infect. Dis. 159:1050-1056.

26. Beasley, R. P., C. C. Lin, K. Y. Wang, F. J. Hsieh, L. Y.Hwang, C. E. Stevens, T. S. Sun, and W. Szmuness. 1981.Hepatitis B immune globulin (HBIG) efficacy in the interrup-tion of perinatal transmission of hepatitis B virus carrier state.Lancet ii:388-393.

27. Belshe, R. B., B. Burk, F. Newman, R. L. Cerruti, and I. S.Sim. 1989. Resistance of influenza A virus to amantadine andrimantadine: results of one decade of surveillance. J. Infect.Dis. 159:430-435.

28. Belshe, R. B., and A. J. Hay. 1989. Drug resistance andmechanisms of action on influenza A viruses. J. Respir. Dis.10(Suppl.):S52-S61.

29. Belshe, R. B., M. H. Smith, C. B. Hall, R. Betts, and A. J. Hay.1988. Genetic basis of resistance to rimantadine emergingduring treatment of influenza virus infection. J. Virol. 62:1508-1512.

30. Bergdahl, S., A. Sonnerborg, A. Larsson, and 0. I. Stranne-gard. 1988. Declining levels of HIV P24 antigen in serumduring treatment with foscarnet. Lancet i:1052.

31. Berglund, O., K. Engman, A. Ehrnst, J. Andersson, K. Lid-man, B. Akerlund, A. Sonnerborg, and 0. Strannegard. 1990.Combined treatment of symptomatic human immunodeficiency

virus type I infection with native interferon-alfa and zidovu-dine. J. Infect. Dis. 163:710-715.

32. Berkman, S. A., M. L. Lee, and R. P. Gale. 1990. Clinical usesof intravenous immunoglobulins. Ann. Intern. Med. 112:278-292.

33. Bernstein, D. I., P. D. Reuman, J. R. Sherwood, E. C. Young,and G. M. Schiff. 1988. Ribavirin small-particle-aerosol treat-ment of influenza B virus infection. Antimicrob. Agents Che-mother. 32:761-764.

34. Birch, C. J., G. Tachedjian, R. R. Doherty, K. Hayes, and I. D.Gust. 1990. Altered sensitivity to antiviral drugs of herpessimplex virus isolates from a patient with the acquired immu-nodeficiency syndrome. J. Infect. Dis. 162:731-734.

35. Biron, K. K., and G. B. Elion. 1980. In vitro susceptibility ofvaricella-zoster virus to acyclovir. Antimicrob. Agents. Che-mother. 18:443-447.

36. Biron, K. K., J. A. Fyfe, S. C. Stanat, L. K. Leslie, J. B. Sorrell,C. U. Lambe, and D. M. Coen. 1986. A human cytomegalovirusmutant resistant to the nucleoside analog 9-(2-hydroxy-1-(hy-droxymethyl)ethoxy)methyl)guanine (BW B759U) induces re-duced levels of BW B759U triphosphate. Proc. Natl. Acad.Sci. USA 83:8769-8773.

37. Biron, K. K., S. C. Stanat, J. B. Sorrell, J. A. Fyfe, P. M.Keller, C. U. Lambe, and D. J. Nelson. 1985. Metabolicactivation of the nucleoside analog 9-(2-hydroxy-1-(hydroxy-methyl)ethoxy)methyl)guanine in human diploid fibroblastsinfected with human cytomegalovirus. Proc. Natl. Acad. Sci.USA 82:2473-2477.

38. Blum, M. R., S. H. T. Liao, S. S. Good, and P. de Miranda.1988. Pharmakokinetics and bioavailability of zidovudine inhumans. Am. J. Med. 85(Suppl. 2A):189-194.

39. Bodsworth, N., and D. A. Cooper. 1990. Ribavirin: a role inHIV infection? J. Acquired Immune Defic. Syndr. 3:893-895.

40. Boston Interhospital Virus Study Group and the NIAID-Spon-sored Cooperative Antiviral Clinical Study. 1975. Failure of highdose 5-iodo-2'-deoxyuridine in the therapy of herpes simplexvirus encephalitis. Evidence of unacceptable toxicity. N. Engl.J. Med. 292:599-603.

41. Bowden, R. A., L. D. Fisher, K. Rogers, M. Cays, and J. D.Meyers. 1991. Cytomegalovirus (CMV)-specific intravenousimmunoglobulin for the prevention of primary CMV infectionand disease after marrow transplant. J. Infect. Dis. 164:483-487.

42. Bowden, R. A., M. Sayers, N. Flournoy, B. Newton, M. Banaji,E. D. Thomas, and J. D. Meyers. 1986. Cytomegalovirusimmune globulin and seronegative blood products to preventprimary cytomegalovirus infection after marrow transplanta-tion. N. Engl. J. Med. 314:1006-1110.

43. Bozzette, S. A., and D. D. Richman. 1990. Salvage therapy forzidovudine-intolerant HIV-infected patients with alternatingand intermittent regimens of zidovudine and dideoxycytidine.Am. J. Med. 88(Suppl. 5B):24S-26S.

44. Brink, J. J., and G. A. LePage. 1964. Metabolic effects of9-D-arabinosylpurines in ascites tumor cells. Cancer Res. 24:312-318.

45. Brown, Z. A., and D. A. Baker. 1989. Acyclovir therapy duringpregnancy. Obstet. Gynecol. 73:526-531.

46. Bryson, Y. J., M. Dillon, M. Lovett, G. Acuna, S. Taylor, J. D.Cherry, B. L. Johnson, E. Wiesmeier, W. Growdon, T. Creagh-Kirk, and R. Keeney. 1983. Treatment of first episodes ofgenital herpes simplex virus infection with oral acyclovir. N.Engl. J. Med. 308:916-921.

47. Buhles, W. C., B. J. Mastre, A. J. Tinker, V. Strand, S. H.Koretz, and the Syntex Collaborative Ganciclovir TreatmentStudy Group. 1988. Ganciclovir treatment of life- or sight-threatening cytomegalovirus infection: experience in 314 im-munocompromised patients. Rev. Infect. Dis. 10(Suppl. 3):S495-S506.

48. Bukrinskaya, A. G., N. K. Vorkunova, and R. A. Narman-betova. 1980. Rimantadine hydrochloride blocks the secondstep of influenza virus uncoating. Arch. Virol. 66:275-282.

49. Burns, W. H., R. Saral, G. W. Santos, 0. L. Laskin, P. S.Lietman, C. McLaren, and D. W. Barry. 1982. Isolation and

168 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 169

characterization of resistant herpes simplex virus after acyclo-vir therapy. Lancet i:421-423.

50. Butler, K. M., R. N. Husson, F. M. Balis, P. Brouwers, J. Eddy,D. El-Amin, J. Gress, M. Hawkins, P. Jarosinski, H. Moss, D.Poplack, S. Santacroce, D. Venzon, L. Wiener, P. Wolters, andP. A. Pizzo. 1991. Dideoxyinosine in children with sympto-matic human immunodeficiency virus infection. N. Engl. J.Med. 324:137-144.

51. Byrn, R. A., J. Mordenti, C. Lucas, D. Smith, S. A. Marsters,J. S. Johnson, P. Cossum, S. M. Chamow, F. M. Wurm, T.Gregory, J. E. Groopman, and D. J. Capon. 1990. Biologicalproperties of a CD4 immunoadhesin. Nature (London) 344:667-670.

52. Canonico, P. G., M. Kende, and J. W. Huggins. 1984. Thetoxicology and pharmacology of ribavirin in experimentalanimals, p. 65-77. In R. A. Smith, V. Knight, and J. A. D.Smith (ed.), Clinical applications of ribavirin. Academic Press,Inc., New York.

53. Capon, D. J., S. M. Chamow, J. Mordenti, S. A. Marsters, T.Gregory, H. Mitsuya, R. A. Byrn, C. Lucas, F. M. Wurm, J. E.Groopman, S. Broder, and D. H. Smith. 1989. Designing CD4immunoadhesins for AIDS therapy. Nature (London) 337:525-531.

54. Cassidy, L. F., and J. L. Patterson. 1989. Mechanism of LaCrosse virus inhibition by ribavirin. Antimicrob. Agents Che-mother. 33:2009-2011.

55. Centers for Disease Control. 1984. Rabies prevention-UnitedStates, 1984. Morbid. Mortal. Weekly Rep. 33:393-408.

56. Centers for Disease Control. 1984. Varicella-zoster immuneglobulin for the prevention of chickenpox: recommendations ofthe immunization practices advisory committee. Ann. Intern.Med. 100:859-865.

57. Centers for Disease Control. 1988. Assessing exposures ofhealth-care personnel to aerosols of ribavirin-California.Morbid. Mortal. Weekly Rep. 37:560-563.

58. Centers for Disease Control. 1988. Management of patients withsuspected viral hemorrhagic fever. Morbid. Mortal. WeeklyRep. 37(S-3):1-16.

59. Centers for Disease Control. 1989. Measles prevention: recom-mendations of the immunization practices advisory committee(ACIP). Morbid. Mortal. Weekly Rep. 38(S-9):1-18.

60. Centers for Disease Control. 1990. Prevention and control ofinfluenza. Morbid. Mortal. Weekly Rep. 39(RR-7):1-15.

61. Centers for Disease Control. 1990. Protection against viralhepatitis. Morbid. Mortal. Weekly Rep. 39(RR-2):1-26.

62. Chachoua, A., D. Dieterich, K. Krasinski, J. Greene, L.Laubenstein, J. Wernz, W. Buhles, and S. Koretz. 1987. 9-(1,3-dihydroxy-2-propoxymethyl)guanine (ganciclovir) in the treat-ment of cytomegalovirus gastrointestinal disease with theacquired immunodeficiency syndrome. Ann. Inter. Med. 107:133-137.

63. Chatis, P. A., C. H. Miller, L. E. Schrager, and C. S. Crump-acker. 1989. Successful treatment with foscarnet of an acyclo-vir-resistant mucocutaneous infection with herpes simplexvirus in a patient with acquired immunodeficiency syndrome.N. Engl. J. Med. 320:297-300.

64. Chaudhary, V. K., T. Mizukami, T. R. Fuerst, D. J. FitzGer-aid, B. Moss, I. Pastan, and E. A. Berger. 1988. Selective killingof HIV-infected cells by recombinant human CD4-Pseudomo-nas exotoxin hybrid protein. Nature (London) 335:369-372.

65. Chen, Y.-C., S. P. Grill, G. E. Dutschman, K. Nakayama, andK. F. Bastow. 1983. Metabolism of 9-(1,3-dihydroxy-2-propoxymethyl-guanine, a new anti-herpes virus compound, inherpes simplex virus-infected cells. J. Biol. Chem. 258:12460-12464.

66. Cheng, Y., G. E. Dutschman, K. F. Bastow, M. G. Sarngadha-ran, and R. Y. C. Ting. 1987. Human immunodeficiency virusreverse transcriptase. J. Biol. Chem. 262:2187-2189.

67. Chu, C. K., R. F. Schinazi, M. K. Ahn, G. V. Ullas, and Z. P.Gu. 1989. Structure-activity relationships of pyrimidine nucle-osides as antiviral agents for human immunodeficiency virustype I in peripheral blood mononuclear cells. J. Med. Chem.32:612-617.

68. Clover, R. D., S. A. Crawford, T. D. Abell, C. N. Ramsey, Jr.,W. P. Glezen, and R. B. Couch. 1986. Effectiveness of riman-tadine prophylaxis of children within families. Am. J. Dis.Child. 140:706-709.

69. Cobo, L. M., G. N. Foulks, T. Liesegang, J. Lass, J. Sutphin, K.Wilhelmus, D. B. Jones, S. Chapman, and A. Segreti. 1985. Oralacyclovir in the therapy of acute herpes zoster ophthalmicus.Ophthalmology 92:1574-1583.

70. Cobo, L. M., G. N. Foulks, T. Liesegang, J. Lass, J. E. Sutphin,K. Wilhelmus, D. B. Jones, S. Chapman, A. C. Segreti, andD. H. King. 1986. Oral acyclovir in the treatment of acuteherpes zoster ophthalmicus. Ophthalmology 93:763-770.

71. Coen, D. M. 1986. General aspects of virus drug resistancewith special reference to herpes simplex virus. J. Antimicrob.Chemother. 18(Suppl. B):1-10.

72. Coen, D. M., and P. A. Schaffer. 1980. Two distinct loci conferresistance to acycloguanosine in herpes simplex virus type 1.Proc. Natl. Acad. Sci. USA 77:2265-2269.

73. Colby, B. M., J. E. Shaw, G. B. Elion, and J. S. Pagano. 1980.Effect of acyclovir [9-(2-hydroxyethoxymethyl) guanine] onEpstein-Barr virus DNA replication. J. Virol. 34:560-568.

74. Collaborative DHPG Treatment Study Group. 1986. Treatmentof serious cytomegalovirus infections with 9-(1,3-dihydroxy-2-propoxymethyl)guanine in patients with AIDS and other im-munodeficiencies. N. Engl. J. Med. 314:801-805.

75. Collier, A. C., S. Bozzette, R. W. Coombs, D. M. Causey, D. A.Schoenfeld, S. A. Spector, C. B. Pettinelli, G. Davies, D. D.Richman, J. M. Leedom, P. Kidd, and L. Corey. 1990. A pilotstudy of low-dose zidovudine in human immunodeficiencyvirus infection. N. Engl. J. Med. 373:1015-1021.

76. Collins, P., B. A. Larder. N. M. Oliver, S. Kemp, I. W. Smith,and G. Darby. 1989. Characterization of a DNA polymerasemutant of herpes simplex virus from a severely immunocom-promised patient receiving acyclovir. J. Gen. Virol. 70:375-382.

77. Cooley, T. P., L. M. Kunches, C. A. Saunders, J. K. Ritter,C. J. Perkins, C. McLaren, R. P. McCaffrey, and H. A.Liebman. 1990. Once-daily administration of 2',3'-dideoxyi-nosine (ddl) in patients with the acquired immunodeficiencysyndrome or AIDS-related complex. N. Engl. J. Med. 322:1340-1345.

78. Cooney, D. A., G. Ahluwalia, H. Mitsuya, A. Fridland, M.Johnson, Z. Hao, M. Dalal, J. Balzarini, S. Broder, and D. G.Johns. 1987. Initial studies on the cellular pharmacology of2',3'-dideoxyadenosine, an inhibitor of HTLV-III infectivity.Biochem. Pharmacol. 36:1765-1768.

79. Corey, L., J. Benedetti, C. Critchlow, G. Mertz, J. Douglas, K.Fife, A. Fahnlander, M. L. Remington, C. Winter, and J.Dragavon. 1983. Treatment of primary first-episode genitalherpes simplex virus infections with acyclovir: results oftopical, intravenous and oral therapy. J. Antimicrob. Chemo-ther. 12(Suppl. B):79-88.

80. Corey, L., A. J. Nahmias, M. E. Guinan, J. K. Benedetti,C. W. Critchlow, and K. K. Holmes. 1982. A trial of topicalacyclovir in genital herpes simplex virus infections. N. Engl. J.Med. 306:1313-1320.

81. Creagh-Kirk, T., P. Doi, E. Andrews, S. Nusinoff-Lehrman, H.Tilson, D. Hoth, and D. W. Barry. 1988. Survival experienceamong patients with AIDS receiving zidovudine. JAMA 260:3009-3015.

82. Crumpacker, C., B. Glenn, D. Lucey, S. Hussey, and J.Connor. 1986. Ribavirin enters cerebrospinal fluid. Lancetii:45-46.

83. Crumpacker, C., S. Marlowe, J. L. Zhang, S. Abrams, P.Watkins, and the Ganciclovir Bone Marrow Transplant Treat-ment Group. 1988. Treatment of cytomegalovirus pneumonia.Rev. Infect. Dis. 10(Suppl. 3):S538-S546.

84. Crumpacker, C. S., P. N. Kowalsky, S. A. Oliver, L. E.Schnipper, and A. K. Field. 1984. Resistance of herpes simplexvirus to 9-(2-hydroxy-1-hydroxymethyl)ethoxy)methyl)gua-nine: physical mapping of drug synergism within the viral DNApolymerase locus. Proc. Natl. Acad. Sci. USA 81:1556-1560.

85. Crumpacker, C. S., L. E. Schnipper, S. I. Marlowe, P. N.

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

Kowalsky, B. J. Hershey, and M. J. Levin. 1982. Resistance toantiviral drugs of herpes simplex virus isolated from a patienttreated with acyclovir. N. Engl. J. Med. 306:343-346.

86. Crumpacker, C. S., L. E. Schnipper, J. A. Zaia, and M. J.Levin. 1979. Growth inhibition by acycloguanosine of herpes-viruses isolated from human infections. Antimicrob. AgentsChemother. 15:642-645.

87. Cudd, A., C. A. Noonan, P. F. Tosi, J. L. Melnick, and C.Nicolau. 1990. Specific interaction of CD4-bearing liposomeswith HIV-infected cells. J. Acquired Immune Defic. Syndr.3:109-114.

88. Dalakas, M. C., I. Ilia, G. H. Pezeshkpour, J. P. Laukaitis, B.Cohen, and J. L. Griffin. 1990. Mitochondrial myopathy causedby long-term zidovudine therapy. N. Engl. J. Med. 322:1098-1105.

89. Daniels, R. S., J. C. Downie, A. J. Hay, M. Knossow, J. J.Skehel, M. L. Wang, and D. C. Wiley. 1985. Fusion mutants ofthe influenza virus hemagglutinin glycoprotein. Cell 40:431-439.

90. Darby, G., H. J. Field, and S. A. Salisbury. 1981. Alteredsubstrate specificity of herpes simplex virus thymidine kinaseconfers acyclovir-resistance. Nature (London) 289:81-83.

91. Datta, A. K., B. M. Colby, J. E. Shaw, and J. S. Pagano. 1980.Acyclovir inhibition of Epstein-Barr Virus replication. Proc.Natl. Acad. Sci. USA 77:5163-5166.

92. Davey, R. T., V. J. Davey, J. A. Metcalf, J. J. Zurlo, J. A.Kovacs, J. Falloon, M. A. Polis, K. M. Zunich, H. Masur, andH. C. Lane. 1991. A phase I/II trial of zidovudine, interferon-alpha, and granulocyte-macrophage colony-stimulating factorin the treatment of human immunodeficiency virus type Iinfection. J. Infect. Dis. 164:43-52.

93. Davis, G. L., L. A. Balart, E. R. Schiff, K. Lindsay, H. C.IBodenheimer, R. P. Perrillo, W. Carey, I. M. Jacobson, J.Payne, J. L. Dienstag, D. H. VanThiel, C. Tamburro, J.Lefkowitch, J. Albrecht, C. Meschievitz, T. J. Ortego, A. Gibas,and the Hepatitis Interventional Therapy Group. 1989. Treat-ment of chronic hepatitis C with recombinant interferon alfa.N. Engl. J. Med. 321:1501-1506.

94. Davis, G. L., and J. H. Hoofnagle. 1986. Interferon in viralhepatitis: role in pathogenesis and treatment. Hepatol. 6:1038-1041.

95. Davis, M. G., S. C. Kenney, J. Kamine, J. S. Pagano, and E. S.Huang. 1987. Immediate-early gene region of human cytomeg-alovirus trans-activates the promoter of human immunodefi-ciency virus. Proc. Natl. Acad. Sci. USA 84:8642-8646.

96. DeArmond, B. 1991. Future directions in the management ofcytomegalovirus infections. J. Acquired Immune Defic. Syndr.4(Suppl. I):S53-S56.

97. DeClercq, E. 1991. Basic approaches to anti-retroviral treat-ment. J. Acquired Immune Defic. Syndr. 4:207-218.

98. DeClercq, E., M. Cools, J. Balzarini, R. Snoeck, G. Andrei, M.Hosoya, S. Shigeta, T. Ueda, N. Minakawa, and A. Matsuda.1991. Antiviral activities of 5-ethynyl-1-13-D-ribofuranosylimi-dazole-4-carboxamide and related compounds. Antimicrob.Agents Chemother. 35:679-684.

99. DeClercq, E., J. Descamps, G. Verhelst, R. T. Walker, A. S.Jones, P. F. Torrence, and D. Shugar. 1980. Comparativeefficacy of antiherpes drugs against different strains of herpessimplex virus. J. Infect. Dis. 141:563-574.

100. De Miranda, P., and M. R. Blum. 1983. Pharmacokinetics ofacyclovir after intravenous and oral administration. J. Antimi-crob. Chemother. 12(Suppl. B):29-37.

101. Deray, G., F. Martinez, C. Katlama, B. Levaltier, H. Beaufils,M. Danis, M. Rozenheim, A. Baumelou, E. Dohin, M. Gentilini,and C. Jacobs. 1989. Foscarnet nephrotoxicity: mechanism,incidence and prevention. Am. J. Nephrol. 9:316-321.

102. Derse, D., K. F. Bastow, and Y.-C. Cheng. 1982. Characteriza-tion of the DNA polymerases induced by a group of herpessimplex virus type I variants selected for growth in thepresence of phosphonoformic acid. J. Biol. Chem. 257:10251-10260.

103. Derse, D., Y.-C. Cheng, P. A. Furman, M. H. St. Clair, andG. B. Elion. 1981. Inhibition of purified human and herpes

simplex virus-induced DNA polymerases by 9-(2-hydrox-yethoxymethyl) guanine triphosphate. J. Biol. Chem. 256:11447-11451.

104. DesJarlais, R. L., G. L. Seibel, I. D. Kuntz, P. S. Furth, J. C.Alvarez, P. R. Ortiz de Montellano, D. L. DeCamp, L. M. Babe,and C. S. Craik. 1990. Structure-based design of nonpeptideinhibitors specific for the human immunodeficiency virus Iprotease. Proc. Natl. Acad. Sci. USA 87:6644-6648.

105. DeWit, R., C. A. B. Boucher, K. H. N. Veenhof, J. K. M. E.Schattenkerk, P. J. M. Bakker, and S. A. Danner. 1988. Clinicaland virological effects of high-dose recombinant interferon-alpha in disseminated AIDS-related Kaposi's sarcoma. Lancetii:1214-1217.

106. Di Bisceglie, A. M., and J. H. Hoofnagle. 1991. Therapy ofchronic hepatitis C with alpha-interferon: the answer? Or morequestions? Hepatology 13:601-603.

107. Di Bisceglie, A. M., P. Martin, C. Kassianides, M. Lisker-Melman, L. Murray, J. Waggoner, Z. Goodman, S. M. Banks,and J. H. Hoofnagle. 1989. Recombinant interferon alfa therapyfor chronic hepatitis C. N. Engl. J. Med. 321:1506-1510.

108. Dicioccio, R. A., and B. I. S. Srivastava. 1977. Kinetics ofinhibition of deoxynucleotide-polymerizing enzyme activitiesfrom normal and leukemic human cells by 9-beta-D-arabino-furanosyladenine 5'-triphosphate and 1-beta-D-arabinofurano-sylcytosine 5'-triphosphate. Eur. J. Biochem. 79:411-418.

109. Dieterich, D. T., A. Chachoua, F. Lafleur, and C. Worrell.1988. Ganciclovir treatment of gastrointestinal infectionscaused by cytomegalovirus in patients with AIDS. Rev. Infect.Dis. 10(Suppl. 3):S532-S537.

110. Dolin, R., R. C. Reichman, H. P. Madore, R. Maynard, P. N.Linton, and J. Webber-Jones. 1982. A controlled trial ofamantadine and rimantadine in the prophylaxis of influenza Ainfection. N. Engl. J. Med. 307:580-584.

111. Dornsife, R. E., M. H. St. Clair, A. T. Huang, T. J. Panella,G. W. Koszalka, C. L. Burns, and D. R. Averett. 1991. Anti-human immunodeficiency virus synergism by zidovudine (3'-azidothymidine) and didanosine (dideoxyinosine) contrastswith their additive inhibition of normal human marrow progen-itor cells. Antimicrob. Agents Chemother. 35:322-328.

112. Douglas, J. M., C. Critchlow, J. Benedetti, G. J. Mertz, J. D.Connor, M. A. Hintz, A. Fahnlander, M. Remington, C.Winter, and L. Corey. 1984. A double-blind study of oralacyclovir for suppression of recurrences of genital herpessimplex virus infection. N. Engl. J. Med. 310:1551-1556.

113. Douglas, J. M., L. J. Eron, F. N. Judson, M. Rogers, M. B.Alder, E. Taylor, D. Tanner, and E. Peets. 1990. A randomizedtrial of combination therapy with intralesional interferon alpha2b and podophyllin versus podophyllin alone for the therapy ofanogenital warts. J. Infect. Dis. 162:52-59.

114. Douglas, R. G., Jr. 1990. Prophylaxis and treatment of influ-enza. N. Engl. J. Med. 322:443-450.

115. Douglas, R. M., B. W. Moore, H. B. Miles, L. M. Davies,N. M. H. Graham, P. Ryan, D. A. Worswick, and J. K.Albrecht. 1986. Prophylactic efficacy of intranasal alpha2-interferon against rhinovirus infections in the family setting. N.Engl. J. Med. 314:65-70.

116. Dournon, E., W. Rozenbaum, C. Michon, C. Perronne, P.DeTruchis, E. Bouvet, M. Levacher, S. Matheron, S. Ghara-khanian, P. M. Girard, D. Salmon, C. Leport, M. C. Dazza, B.Regnier, and the Claude Bernard Hospital AZT Study Group.1988. Effects of zidovudine in 365 consecutive patients withAIDS or AIDS-related complex. Lancet ii:1297-1302.

117. Drach, J. C. 1983. Targets for the design of antiviral agents, p.231-257. In E. DeClercq and R. T. Walker (ed.), Purinenucleoside analogs as antiviral agents. Plenum Press, NewYork.

118. Drew, W. L., R. C. Miner, D. F. Busch, S. E. Follansbee, J.Gullett, S. G. Mehalko, S. M. Gordon, W. F. Owen, Jr., T. R.Matthews, W. C. Buhles, and B. DeArmond. 1991. Prevalenceof resistance in patients receiving ganciclovir for serious cyto-megalovirus infection. J. Infect. Dis. 163:716-719.

119. Dunn, D. L., J. L. Mayoral, K. J. Gillingham, C. M. Loeffler,K. L. Brayman, M. A. Kramer, A. Erice, H. H. Balfour, C. V.

170 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 171

Fletcher, R. M. Bolman, A. J. Matas, W. D. Payne, D. E. R.Sutherland, and J. S. Najarian. 1991. Treatment of invasivecytomegalovirus disease in solid organ transplant patients withganciclovir. Transplantation 51:98-106.

120. Dwyer, J. M., and K. Erlendsson. 1988. Intraventricular gam-ma-globulin for the management of enterovirus encephalitis.Pediatr. Infect. Dis. J. 7:S30-S33.

121. Efstathiou, S., S. Kemp, G. Darby, and A. C. Minson. 1989.The role of herpes simplex virus type I thymidine kinase inpathogenesis. J. Gen. Virol. 70:869-879.

122. Elion, G. B., P. A. Furman, J. A. Fyfe, P. de Miranda, L.Beauchamp, and H. J. Schaeffer. 1977. Selectivity of action ofan antiherpetic agent, 9-(2-hydroxyethoxymethyl) guanine.Proc. Natl. Acad. Sci. USA 74:5716-5720.

123. Ellis, M. N., P. M. Keller, J. A. Fyfe, J. L. Martin, J. F.Rooney, S. E. Straus, S. Nusinoff Lehrman, and D. W. Barry.1987. Clinical isolate of herpes simplex virus type 2 thatinduces a thymidine kinase with altered substrate specificity.Antimicrob. Agents Chemother. 31:1117-1125.

124. Elwell, L. P., R. Ferone, G. A. Freeman, J. A. Fyfe, J. A. Hill,P. H. Ray, C. A. Richards, S. C. Singer, V. B. Knick, J. L.Rideout, and T. P. Zimmerman. 1987. Antibacterial activityand mechanism of action of 3'-azido-3'-deoxythymidine (BWA509U). Antimicrob. Agents Chemother. 31:274-280.

125. Emanuel, D., I. Cunningham, K. Jules-Elysee, J. A. Brochstein,N. A. Kernan, J. Laver, D. Stover, D. A. White, A. Fels, B.Polsky, H. Castro-Malaspina, J. R. Peppard, P. Bartus, U.Hammerling, and R J. O'Reilly. 1988. Cytomegalovirus pneu-monia after bone marrow transplantation successfully treatedwith the combination of ganciclovir and high-dose intravenousimmune globulin. Ann. Intern. Med. 109:777-782.

126. Englund, J. A., M. E. Zimmerman, E. M. Swierkosz, J. L.Goodman, D. R. Scholl, and H. H. Balfour. 1990. Herpessimplex virus resistant to acyclovir. A study in a tertiary carecenter. Ann. Intern. Med. 112:416-422.

127. Erice, A., S. Chou, K. K. Biron, S. C. Stanat, H. H. Balfour,and M. Jordan. 1989. Progressive disease due to ganciclovir-resistant cytomegalovirus in immunocompromised patients. N.Engl. J. Med. 320:289-293.

128. Erickson, J., D. J. Neidhart, J. VanDrie, D. J. Kempf, X. C.Wang, D. W. Norbeck, J. J. Plattner, J. W. Rittenhouse, M.Turon, N. Wideburg, W. E. Kohlbrenner, R. Simmer, R.Helfrich, D. A. Paul, and M. Knigge. 1990. Design, activity,and 2.8 A crystal structure of a C2 symmetric inhibitor com-plexed to HIV-I protease. Science 249:527-533.

129. Eriksson, B., E. Helgstrand, N. G. Johansson, A. Larsson, A.Misiorny, J. 0. Noren, L. Philipson, K. Stenberg, G. Stening, S.Stridh, and B. Oberg. 1977. Inhibition of influenza virusribonucleic acid polymerase by ribavirin triphosphate. Antimi-crob. Agents. Chemother. 11:946-951.

130. Erlich, K. S., M. A. Jacobson, J. E. Koehler, S. E. Follansbee,D. P. Drennan, L. Gooze, S. Safrin, and J. Mills. 1989.Foscarnet therapy for severe acyclovir-resistant herpes sim-plex virus type-2 infections in patients with the acquiredimmunodeficiency syndrome (AIDS). Ann. Intern. Med. 110:710-713.

131. Erlich, K. S., J. Mills, P. Chatis, G. J. Mertz, D. F. Busch,S. E. Follansbee, R. M. Grant, and C. S. Crumpacker. 1989.Acyclovir-resistant herpes simplex virus infections in patientswith the acquired immunodeficiency syndrome. N. Engl. J.Med. 320:293-296.

132. Eron, L. J., F. Judson, S. Tucker, S. Prawer, J. Mills, K.Murphy, M. Hickey, M. Rogers, S. Flannigan, N. Hien, H. I.Katz, S. Goldman, A. Gottlieb, K. Adams, P. Burton, D.Tanner, E. Taylor, and E. Peets. 1986. Interferon therapy forcondylomata acuminata. N. Engl. J. Med. 315:1059-1064.

133. Estes, J. E., and E.-S. Huang. 1977. Stimulation of cellularthymidine kinases by human cytomegalovirus. J. Virol. 24:13-21.

134. Fanning, M. M., S. E. Read, M. Benson, S. Vas, A. Rachlis, V.Kozousek, C. Mortimer, P. Harvey, C. Schwartz, E. Chew, J.Brunton, A. Matlow, I. Salit, H. Vellend, and S. Walmsley.1990. Foscarnet therapy of cytomegalovirus retinitis in AIDS.

J. Acquired Immune Defic. Syndr. 3:472-479.135. Fauci, A. S., S. M. Schnittman, G. Poli, S. Koenig, and G.

Pantaleo. 1991. Immunopathogenic mechanisms in human im-munodeficiency virus (HIV) infection. Ann. Intern. Med. 114:678-693.

136. Fegueux, S., D. Salmon, C. Picard, C. Sauvage, P. Longuet, andT. Desmoulins. 1990. Penile ulcerations with foscarnet. Lancet335:547.

137. Feldman, S., J. Rodman, and B. Gregory. 1988. Excessiveserum concentrations of acyclovir and neurotoxicity. J. Infect.Dis. 157:385-388.

138. Felsenstein, D., D. J. D'amico, M. S. Hirsch, D. A. Neumeyer,D. M. Cederberg, P. deMiranda, and R. T. Schooley. 1985.Treatment of cytomegalovirus retinitis with 9-(2-hydroxy-1-(hydroxymethyl) ethoxymethyl)guanine. Ann. Intern. Med.103:377-380.

139. Field, A. K., M. E. Davies, C. DeWitt, H. C. Perry, R. Liou, J.Germershausen, J. D. Karkas, W. T. Ashton, D. B. R.Johnston, and R. L. Tolman. 1983. 9-(2-Hydroxy-1-(hydroxy-methyl)ethoxy)methyl)guanine: a selective inhibitor of herpesgroup virus replication. Proc. Natl. Acad. Sci. USA 80:4139-4143.

140. Field, H. J. 1989. Persistent herpes simplex virus infection andmechanisms of virus drug resistance. Eur. J. Clin. Microbiol.Infect. Dis. 8:671-680.

141. Field, H. J., and D. M. Coen. 1986. Pathogenicity of herpessimplex virus mutants containing drug resistance mutations inthe viral DNA polymerase gene. J. Virol. 60:286-289.

142. Field, H. J., and G. Darby. 1980. Pathogenicity in mice ofstrains of herpes simplex virus which are resistant to acyclovirin vitro and in vivo. Antimicrob. Agents Chemother. 17:209-216.

143. Field, H. J., and P. Wildy. 1978. The pathogenicity of thymi-dine kinase-deficient mutants of herpes simplex virus in mice.J. Hyg. Camb. 81:2167-77.

144. Fischl, M. A., C. B. Parker, C. Pettinelli, M. Wulfsohn, M. S.Hirsch, A. C. Collier, D. Antoniskis, M. Ho, D. D. Richman, E.Fuchs, T. C. Merigan, R. C. Reichman, J. Gold, N. Steigbigel,G. S. Leoung, S. Rasheed, A. Tsiatis, and the AIDS ClinicalTrials Group. 1990. A randomized controlled trial of a reduceddaily dose of zidovudine in patients with the acquired immu-nodeficiency syndrome. N. Engl. J. Med. 323:1009-1014.

145. Fischl, M. A., D. D. Richman, D. M. Causey, M. H. Grieco, Y.Bryson, D. Mildvan, 0. L. Laskin, J. E. Groopman, P. A.Volberding, R. T. Schooley, G. G. Jackson, D. T. Durack, J. C.Andrews, S. Nusinoff-Lehrman, D. W. Barry, and the AZTCollaborative Working Group. 1989. Prolonged zidovudinetherapy in patients with AIDS and advanced AIDS-relatedcomplex. JAMA 262:2405-2410.

146. Fischl, M. A., D. D. Richman, M. H. Grieco, M. S. Gottlieb,P. A. Volberding, 0. L. Laskin, J. M. Leedom, J. E. Groop-man, D. Mildvan, R. T. Schooley, G. G. Jackson, D. T. Durack,D. King, and the AZT Collaborative Working Group. 1987. Theefficacy of azidothymidine (AZT) in the treatment of patientswith AIDS and AIDS-related complex. N. Engl. J. Med.317:185-191.

147. Fischl, M. A., D. D. Richman, N. Hansen, A. C. Collier, J. T.Carey, M. F. Para, W. D. Hardy, R. Dolin, W. G. Powderly,J. D. Allan, B. Wong, T. C. Merigan, V. J. McAuliffe, N. E.Hyslop, F. S. Rhame, H. H. Balfour, Jr., S. A. Spector, P.Volberding, C. Pettinelli, J. Anderson, and the AIDS ClinicalTrials Group. 1990. The safety and efficacy of zidovudine(AZT) in the treatment of subjects with mildly symptomatichuman immunodeficiency virus type I (HIV) infection. Ann.Intern. Med. 112:727-737.

148. Fischl, M. A., R. B. Uttamchandani, L. Resnick, R. Agarwal,M. A. Fletcher, J. Patrone-Reese, L. Dearmas, J. Chidekel, M.McCann, and M. Myers. 1991. A phase I study of recombinanthuman interferon-alpha2a or human lymphoblastoid interferon-alphan, and concomitant zidovudine in patients with AIDS-related Kaposi's sarcoma. J. Acquired Immune Defic. Syndr.4:1-10.

149. Fisher, A. G., B. Ensoli, D. Looney, A. Rose, R. C. Gallo, M. S.

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

Saag, G. M. Shaw, B. H. Hahn, and F. Wong-Staal. 1988.Biologically diverse molecular variants within a single HIV-Iisolate. Nature (London) 334:444 447.

150. Fox, M. P., M. J. Otto, and M. A. McKinlay. 1986. Preventionof rhinovirus and poliovirus uncoating by WIN 51711, a newantiviral drug. Antimicrob. Agents Chemother. 30:110-116.

151. Freitas, V. R., D. F. Smee, M. Chernow, R. Boehme, and T. R.Matthews. 1985. Activity of 9-(1,3-dihydroxy-2-propoxymeth-yl)guanine compared with that of acyclovir against human,monkey, and rodent cytomegaloviruses. Antimicrob. AgentsChemother. 28:240-245.

152. Friedman-Kien, A. E., L. J. Eron, M. Conant, W. Growdon, H.Badiak, P. W. Bradstreet, D. Fedorczyk, J. R. Trout, and T. F.Plasse. 1988. Natural interferon alfa for treatment of condylo-mata acuminata. JAMA 259:533-538.

153. Furman, P. A., and D. W. Barry. 1988. Spectrum of antiviralactivity and mechanism of action of zidovudine. Am. J. Med.85(Suppl. 2A):176-181.

154. Furman, P. A., D. M. Coen, M. H. St. Clair, and P. A. Schaffer.1981. Acyclovir-resistant mutants of herpes simplex virus typeI express altered DNA polymerase or reduced acyclovir phos-phorylating activities. J. Virol. 40:936-941.

155. Furman, P. A., J. A. Fyfe, M. H. St. Clair, K. Weinhold, J. L.Rideout, G. A. Freeman, S. Nusinoff Lehrman, D. P. Bolognesi,S. Broder, H. Mitsuya, and D. W. Barry. 1986. Phosphoryla-tion of 3'-azido-3'-deoxythymidine and selective interaction ofthe 5'-triphosphate with human immunodeficiency virus re-verse transcriptase. Proc. Natl. Acad. Sci. USA 83:8333-8337.

156. Furman, P. A., P. V. McGuirt, P. M. Keller, J. A. Fyfe, andG. B. Elion. 1980. Inhibition by acyclovir of cell growth andDNA synthesis of cells biochemically transformed with her-pesvirus genetic information. Virology 102:420-430.

157. Furman, P. A., M. H. St. Clair, J. A. Fyfe, J. L. Rideout, P. M.Keller, and G. B. Elion. 1979. Inhibition of herpes simplexvirus-induced DNA polymerase activity and viral DNA repli-cation by 9-(2-hydroxyethoxymethyl)guanine and its triphos-phate. J. Virol. 32:72-77.

158. Furman, P. A., M. H. St. Clair, and T. Spector. 1984. Acyclo-vir triphosphate is a suicide inactivator of the herpes simplexvirus DNA polymerase. J. Biol. Chem. 259:9575-9579.

159. Fyfe, J. A., P. M. Keller, P. A. Furman, R. L. Miller, and G. B.Elion. 1978. Thymidine kinase from herpes simplex virusphosphorylates the new antiviral compound, 9-(2-hydrox-yethoxymethyl) guanine. J. Biol. Chem. 253:8721-8727.

160. Gadler, H., A. Larsson, and E. Solver. 1984. Nucleic acidhybridization, a method to determine effects of antiviral com-pounds on herpes simplex virus type 1 DNA synthesis. Anti-viral Res. 4:63-70.

161. Galbraith, A. W., J. S. Oxford, G. C. Schild, and G. I. Watson.1969. Protective effect of 1-adamantanamine hydrochloride oninfluenza A2 infections in the family environment. Lancetii:1026-1028.

162. Galpin, S., J. Martin, T. O'Connor, S. Redshaw, N. Roberts,and D. Kinchington. 1991. Further studies on the HIV protein-ase inhibitor RO 31-8959. Antiviral Res. 15(Suppl 1):96.

163. Gangemi, J. D., M. Nachtigal, D. Barnhart, L. Krech, and P.Jani. Therapeutic efficacy of liposome-encapsulated ribavirinand muramyl tripeptide in experimental infection with influ-enza or herpes simplex virus. J. Infect. Dis. 155:510-517.

164. Gately, A., R. M. Gander, P. C. Johnson, S. Kit, H. Otsuka,and S. Kohl. 1990. Herpes simplex virus type 2 meningoen-cephalitis resistant to acyclovir in a patient with AIDS. J.Infect. Dis. 161:711-715.

165. Gelmon, K. J., S. G. Montaner, M. Fanning, J. R. M. Smith, J.Falutz, C. Tsoukas, J. Gill, G. Wells, M. O'Shaughnessy, M.Wainberg, and J. Ruedy. 1989. Nature, time course and dosedependence of zidovudine-related side effects: results from themulticenter Canadian azidothymidine trial. AIDS 3:555-561.

166. Gershon, A. A., S. Steinberg, and P. A. Brunell. 1974. Zosterimmune globulin. A further assessment. N. Engl. J. Med.290:243-245.

167. Gilbert, B. E., S. Z. Wilson, V. Knight, R. B. Couch, J. M.Quarles, L. Dure, N. Hayes, and G. Willis. 1985. Ribavirin

small-particle aerosol treatment of infections caused by influ-enza virus strains A/Victoria/7/83 (HlNl) and B/Texas/1/84.Antimicrob. Agents Chemother. 27:309-313.

168. Gilquin, J., L. Weiss, and M. D. Kazatchkine. 1990. Genital andoral erosions induced by foscarnet. Lancet 335:287.

169. Goodrich, J. M., M. Mori, C. A. Gleaves, C. Du Mond, M.Cays, D. F. Ebeling, W. C. Buhles, B. DeArmond, and J. D.Meyers. 1991. Early treatment with ganciclovir to preventcytomegalovirus disease after allogenic bone marrow trans-plantation. N. Engl. J. Med. 325:1601-1607.

170. Goswami, B. B., J. Fujitaki, and R. A. Smith. 1979. The broadspectrum antiviral agent ribavirin inhibits capping of mRNA.Biochem. Biophys. Res. Commun. 89:830-836.

171. Gottlieb, M. S., R. A. Zackin, M. Fiala, D. H. Henry, A. J.Marcel, K. L. Combs, J. Viera, H. A. Liebman, L. A. Cone,K. S. Hillman, and A. A. Gottlieb. 1991. Response to treatmentwith the leukocyte-derived immunomodulator IMREG-1 inimmunocompromised patients with AIDS-related complex.Ann. Intern. Med. 115:84-91.

172. Greffe, B. S., S. L. Dooley, R. B. Deddish, and H. C. Krasny.1986. Transplacental passage of acyclovir J. Pediatr. 108:1020-1021.

173. Groopman, J. E., R. T. Mitsuyasu, M. J. DeLeo, D. H. Oette,and D. W. Golde. 1987. Effect of recombinant human granulo-cyte-macrophage colony-stimulating factor on myelopoiesis inthe Acquired Immunodeficiency Syndrome. N. Engl. J. Med.317:593-598.

174. Grundy, J. E., J. D. Shanley, and P. D. Griffiths. 1987. Iscytomegalovirus interstitial pneumonitis in transplant recipi-ents an immunopathological condition? Lancet ii:996-999.

175. Haake, D. A., P. C. Zakowski, D. L. Haake, and Y. J. Bryson.1990. Early treatment with acyclovir for varicella pneumonia inotherwise healthy adults: retrospective controlled study andreview. Rev. Infect. Dis. 12:788-798.

176. Hadler, S. C., J. J. Erben, D. Matthews, K. Starko, D. P.Francis, and J. E. Maynard. 1983. Effect of immunoglobulin on

hepatitis A in day-care centers. JAMA 249:48-53.177. Hall, C. B., R. Dolin, C. L. Gala, D. M. Markovitz, Y. Q.

Zhang, P. H. Madore, F. A. Disney, W. B. Ta, J. L. Green,A. B. Francis, and M. E. Pichichero. 1987. Children withinfluenza A infection: treatment with rimantadine. Pediatrics80:275-282.

178. Hall, C. B., J. T. McBride, C. L. Gala, S. W. Hildreth, andK. C. Schnabel. 1985. Ribavirin treatment of respiratory syn-cytial viral infection in infants with underlying cardiopulmo-nary disease. JAMA 254:3047-3051.

179. Hall, C. B., J. T. McBride, E. E. Walsh, D. M. Bell, C. L. Gala,S. Hildret, L. G. Ten Eyck, and W. J. Hall. 1983. Aerosolizedribavirin treatment of infants with respiratory syncytial viralinfection. N. Engl. J. Med. 308:1443-1447.

180. Harbison, M. A., P. C. DeGirolami, R. L. Jenkins, and S. M.Hammer. 1988. Ganciclovir therapy of severe cytomegalovirusinfections in solid-organ transplant recipients. Transplantation46:82-88.

181. Hardy, W. D. 1991. Combined ganciclovir and recombinanthuman granulocyte-macrophage colony-stimulating factor inthe treatment of cytomegalovirus retinitis in AIDS patients. J.Acquired Immune Defic. Syndr. 4(Suppl. I):S22-S28.

182. Hartman, N. R., R. Yarchoan, J. M. Pluda, R. V. Thomas,K. S. Marczyk, S. Broder, and D. G. Johns. 1990. Pharmaco-kinetics of 2',3'-dideoxyadenosine and 2',3'-dideoxyinosine inpatients with severe human immunodeficiency virus infection.Clin. Pharmacol. Ther. 47:647-654.

183. Hartshorn, K. L., M. W. Vogt, T. Chou, R. S. Blumberg, R.Byington, R. T. Schooley, and M. S. Hirsch. 1987. Synergisticinhibition of human immunodeficiency virus in vitro by azido-thymidine and recombinant alpha interferon. Antimicrob.Agents Chemother. 31:168-172.

184. Hay, A. J., A. J. Wolstenholme, J. J. Skehel, and M. H. Smith.1985. The molecular basis of the specific anti-influenza actionof amantadine. EMBO J. 4:3021-3024.

185. Hay, A. J., M. C. Zambon, A. J. Wolstenholme, J. J. Skehel,and M. H. Smith. 1986. Molecular basis of resistance of

172 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 173

influenza A viruses to amantadine. J. Antimicrob. Chemother.18(Suppl. B):19-29.

186. Hayden, F. G. 1989. Significance of rimantadine resistance ininfluenza A viruses. J. Respir. Dis. 10(Suppl.):562-567.

187. Hayden, F. G., J. K. Albrecht, D. L. Kaiser, and J. M.Gwaltney. 1986. Prevention of natural colds by contact prophy-laxis with intranasal alpha2-interferon. N. Engl. J. Med. 314:71-75.

188. Hayden, F. G., R. B. Beishe, R. D. Clover, A. J. Hay, M. G.Oakes, and W. Soo. 1989. Emergence and apparent transmis-sion of rimantadine-resistant influenza A virus in families. N.Engl. J. Med. 321:1696-1702.

189. Hayden, F. G., K. M. Cote, and R. G. Douglas. 1980. Plaqueinhibition assay for drug susceptibility testing of influenzaviruses. Antimicrob. Agents Chemother. 17:865-870.

190. Hayden, F. G., D. L. Kaiser, and J. K. Albrecht. 1988.Intranasal recombinant alfa-2b interferon treatment of natu-rally occurring common colds. Antimicrob. Agents Chemo-ther. 32:224-230.

191. Hayden, F. G., and A. S. Monto. 1986. Oral rimantadinehydrochloride therapy of influenza A virus H3N2 subtypeinfection in adults. Antimicrob. Agents Chemother. 29:339-341.

192. Heagy, W., C. Crumpacker, P. A. Lopez, and R. W. Finberg.1991. Inhibition of immune functions by antiviral drugs. J.Clin. Invest. 87:1916-1924.

193. Hecht, D. W., D. R. Snydman, C. S. Crumpacker, B. G.Werner, B. Heinze-Lacey, and the Boston Renal TransplantStudy Group. 1988. Ganciclovir for treatment of renal trans-plant-associated primary cytomegalovirus pneumonia. J. In-fect. Dis. 157:187-190.

194. Heidelberger, C., and D. H. King. 1979. Trifluorothymidine.Pharmacol. Ther. 6:427-442.

195. Heinemann, M. H. 1989. Long-term intravitreal ganciclovirtherapy for cytomegalovirus retinopathy. Arch. Ophthalmol.107:1767-1772.

196. Helgstrand, E., B. Eriksson, N. G. Johansson, B. Lannero, A.Larsson, A. Misiorny, J. 0. Noren, B. Sjoberg, K. Stenberg, G.Stening, S. Stridh, B. Oberg, S. Alenius, and L. Philipson. 1979.Trisodium phosphonoformate, a new antiviral compound. Sci-ence 201:819-821.

197. Henderly, D. E., W. R. Freeman, D. M. Causey, and N. A. Rao.1987. Cytomegalovirus retinitis and response to therapy withganciclovir. Ophthalmology 94:425-434.

198. Henry, K., H. Cantrill, C. Fletcher, B. J. Chinnock, and H. H.Balfour. 1987. Use of intravitreal ganciclovir (dihydroxypropoxymethyl guanine) for cytomegalovirus retinitis in apatient with AIDS. Am. J. Ophthalmol. 103:17-23.

199. Henry, K., B. J. Chinnock, R. P. Quinn, C. V. Fletcher, P.deMiranda, and H. H. Balfour. 1988. Concurrent zidovudinelevels in semen and serum determined by radioimmunosassayin patients with AIDS or AIDS-related complex. JAMA 259:3023-3026.

200. Hersh, E. M., G. Brewton, D. Abrams, J. Bartlett, J. Galpin, P.Gill, R. Gorter, M. Gottlieb, J. J. Jonikas, S. Landesman, A.Levin, A. Marcel, E. A. Petersen, M. Whiteside, J. Zahradnik,C. Negron, F. Boutitie, J. Caraux, J. M. Dupuy, and L. R.Salmi. 1991. Ditiocarb sodium (diethyldithiocarbamate) ther-apy in patients with symptomatic HIV infection and AIDS.JAMA 265:1538-1544.

201. Hill, J. M., F. Sedarati, R. T. Javier, E. K. Wagner, and J. G.Stevens. 1990. Herpes simplex virus latent phase transcriptionfacilitates in vivo reactivation. Virology 174:117-125.

202. Hirsch, M. S. 1990. Chemotherapy of human immunodefi-ciency virus infections: current practice and future prospects.J. Infect. Dis. 161:845-847.

203. Hirsch, M. S., R. T. Schooley, A. B. Cosimi, P. S. Russell, F. L.Delmonico, N. E. Tolkoff-Rubin, J. T. Herrin, K. Cantell, M. L.Farrell, T. R. Rota, and R. H. Rubin. 1983. Effects of interfer-on-alpha on cytomegalovirus reactivation syndromes in renal-transplant recipients. N. Engl. J. Med. 308:1489-1493.

204. Ho, D. D., T. R. Rota, J. C. Kaplan, K. L. Hartshorn, C. A.Andrews, R. T. Schooley, and M. S. Hirsch. 1985. Recombinant

human interferon alfa-a suppresses HTLV-III replication invitro. Lancet i:602-604.

205. Ho, H., and M. J. M. HitchcocL 1989. Cellular pharmacologyof 2',3'-dideoxy-2',3'-didehydrothymidine, a nucleoside ana-log active against human immunodeficiency virus. Antimicrob.Agents Chemother. 33:844-849.

206. Hochster, H., D. Dieterich, S. Bozzette, R. C. Reichman, J. D.Connor, L. Liebes, R. L. Sonke, S. A. Spector, F. Valentine, C.Pettinelli, and D. D. Richman. 1990. Toxicity of combinedganciclovir and zidovudine for cytomegalovirus disease asso-ciated with AIDS. Ann. Intern. Med. 113:111-117.

207. Hoffmann, C. E., E. M. Neumayer, R. F. Huff, and R. A.Coldsby. 1965. Mode of action of the antiviral activity ofamantadine in tissue culture. J. Bacteriol. 90:623-628.

208. Holland, G. N., M. J. Sakamoto, D. Hardy, Y. Sidikaro, A. E.Kreiger, L. M. Frenkel, and the UCLA CMV Retinopathy StudyGroup. 1986. Treatment of cytomegalovirus retinopathy inpatients with acquired immunodeficiency syndrome. Arch.Ophthalmol. 104:1794-1800.

209. Hoofnagle, J. H., M. Peters, K. D. Mullen, D. B. Jones, V.Rustgi, A. DiBisceglie, C. Hallahan, Y. Park, C. Meschievitz,and E. A. Jones. 1988. Randomized, controlled trial of recom-binant human alpha-interferon in patients with chronic hepati-tis B. Gastroenterology 95:1318-1325.

210. Hovi, T. 1988. Successful selective inhibitors of viruses. p.1-21. In H. J. Field (ed.), Antiviral agents: the developmentand assessment of antiviral chemotherapy. CRC Press, BocaRaton, Fla.

211. Howell, C. L., and M. J. Miller. 1984. Rapid method fordetermining the susceptibility of herpes simplex virus to acy-clovir. Diagn. Microbiol. Infect. Dis. 2:77-84.

212. Hruska, J. F., J. M. Bernstein, R. G. Douglas, Jr., and C. B.Hall. 1990. Effects of ribavirin on respiratory syncytial virus invitro. Antimicrob. Agents Chemother. 17:770-775.

213. Huff, J. C., B. Bean, H. H. Balfour, 0. L. Laskin, J. D.Connor, L. Corey, J. Bryson, and P. McGuirt. 1988. Therapy ofherpes zoster with oral acyclovir. Am. J. Med. 85(Suppl.2A):84-89.

214. Huggins, J. W. 1989. Prospects for treatment of viral hemor-rhagic fevers with ribavirin, a broad-spectrum antiviral drug.Rev Infect Dis. 11(Suppl. 4):5750-5761.

215. Indulen, M. K., and R. L. Feldblum. 1982. Obtaining of avirazole-resistant fowl plague virus mutant. Acta Virol. 26:109.

216. Isaacs, A., and J. Lindenmann. 1957. Virus interference. I. Theinterferon. Proc. R. Soc. London Ser. B 147:258-267.

217. Jabs, D. A., C. Enger, and J. C. Bartlett. 1989. Cytomegalovi-rus retinitis and acquired immunodeficiency syndrome. Arch.Ophthalmol. 107:75-80.

218. Jackson, G. G., D. A. Paul, L. A. Falk, M. Rubenis, J. C.Despotes, D. Mack, M. Knigge, and E. E. Emeson. 1988.Human immunodeficiency virus (HIV) antigenemia (p24) in theacquired immunodeficiency syndrome (AIDS) and the effect oftreatment with zidovudine (AZT). Ann. Intern. Med. 108:175-180.

219. Jackson, G. G., M. Rubenis, M. Knigge, J. T. Perkins, D. A.Paul, J. C. Despotes, and P. Spencer. 1988. Passive immunon-eutralisation of human immunodeficiency virus in patients withadvanced AIDS. Lancet ii:647-652.

220. Jacobson, M. A., T. G. Berger, S. Fikrig, P. Becherer, J. W.Moohr, S. C. Stanat, and K. K. Biron. 1990. Acyclovir-resistant varicella zoster virus infection after chronic oralacyclovir therapy in patients with the acquired immunodefi-ciency syndrome (AIDS). Ann. Intern. Med. 112:187-191.

221. Jacobson, M. A., S. Crowe, J. Levy, F. Aweeka, J. Gamberto-glio, N. McManus, and J. Mills. 1988. Effect of foscarnettherapy on infection with human immunodeficiency virus inpatients with AIDS. J. Infect. Dis. 158:862-865.

222. Jacobson, M. A., P. de Miranda, D. M. Cederberg, T. Burnette,E. Cobb, H. R. Brodie, and J. Mills. 1987. Human pharmaco-kinetics and tolerance of oral ganciclovir. Antimicrob. AgentsChemother. 31:1251-1254.

223. Jacobson, M. A., J. G. Gambertoglio, F. T. Aweeka, D. M.Causey, and A. A. Portale. 1991. Foscarnet-induced hypocal-

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

cemia and effects of foscarnet on calcium metabolism. J. Clin.Endocrinol. Metab. 72:1130-1135.

224. Jacobson, M. A., J. J. O'Donnell, and J. Mills. 1989. Foscarnettreatment of cytomegalovirus retinitis in patients with theacquired immunodeficiency syndrome. Antimicrob. AgentsChemother. 33:736-741.

225. Jacobson, M. A., C. van der Horst, D. M. Causey, M. Deh-linger, R. Hafner, and J. Mills. 1991. In vivo additive antiret-roviral effect of combined zidovudine and foscarnet therapy forhuman immunodeficiency virus infection (ACTG Protocol053). J. Infect. Dis. 163:1219-1222.

226. Japour, A. J., P. A. Chatis, H. E. Eigenrauch, and C. S.Crumpacker. 1991. Detection of human immunodeficiencyvirus type I clinical isolates with reduced sensitivity to zidovu-dine and dideoxyinosine by RNA-RNA hybridization. Proc.Natl. Acad. Sci. USA 88:3092-3096.

227. Johnson, M. A., G. Ahluwalia, M. C. Connelly, D. A. Cooney,S. Broder, D. G. Johns, and A. Fridland. 1988. Metabolicpathways for the activation of the antiretroviral agent 2',3'-dideoxyadenosine in human lymphoid cells. J. Biol. Chem.263:15354-15357.

228. Joklik, W. K. 1990. Interferons, p. 383-410. In B. W. Fields,D. M. Knipe, R. M. Chanock, M. S. Hirsch, J. L. Melnick,T. P. Monath, and B. Roizman (ed.), Fields virology, 2nd ed.Raven Press, New York.

229. Kahn, J. O., J. D. Allan, T. L. Hodges, L. D. Kaplan, C. J.Arri, H. F. Fitch, A. E. Izu, J. Mordenti, S. A. Sherwin, J. E.Groopman, and P. A. Volberding. 1990. The safety and phar-macokinetics of recombinant soluble CD4 (rCD4) in subjectswith the acquired immunodeficiency syndrome (AIDS) andAIDS-related complex. Ann. Intern. Med. 112:254-261.

230. Kaplowitz, L. G., D. Baker, L. Gelb, J. Blythe, R. Hale, P.Frost, C. Crumpacker, S. Rabinovich, J. E. Peacocki, J. Hern-don, L. G. Davis, and the Acyclovir Study Group. 1991.Prolonged continuous acyclovir treatment of normal adultswith frequently recurring genital herpes simplex virus infec-tion. JAMA 265:747-751.

231. Karpas, A. G., W. J. Fleet, R. A. Dwek, S. Petursson, S. K.Namgoong, N. G. Ramsden, G. S. Jacob, and T. W. Radema-cher. 1988. Aminosugar derivatives as potential anti-humanimmunodeficiency virus agents. Proc. Natl. Acad. Sci. USA85:9229-9233.

232. Kato, N., and H. J. Eggers. 1969. Inhibition of uncoating offowl plague virus by 1-adamantanamine hydrochloride. Virol-ogy 37:632-641.

233. Kaufman, H. E., E. L. Martola, and C. Dohlman. 1962. Use of5-iodo-2'-deoxyuridine (IDU) in treatment of herpes simplexkeratitis. Arch. Ophthalmol. 68:235-239.

234. Keith, B. R., G. White, and H. R. Wilson. 1989. In vivo efficacyof zidovudine (3Y-azido-3'-deoxythymidine) in experimentalgram-negative bacterial infections. Antimicrob. Agents Che-mother. 33:479-483.

235. Keller, P. M., J. A. Fyfe, L. Beauchamp, C. M. Lubbers, P. A.Furman, H. J. Schaeffer, and G. B. Elion. 1981. Enzymaticphosphorylation of acyclic nucleoside analogs and correlationswith antiherpetic activities. Biochem. Pharmacol. 30:3071-3077.

236. Kende, M., C. R. Alving, W. L. Rill, G. M. Swartz, Jr., andP. G. Canonico. 1985. Enhanced efficacy of liposome-encapsu-lated ribavirin against Rift Valley Fever virus infection in mice.Antimicrob. Agents Chemother. 27:903-907.

237. Kim, S., S. Scheerer, M. A. Geyer, and S. B. Howell. 1990.Direct cerebrospinal fluid delivery of an antiretroviral agentusing multivesicular liposomes. J. Infect. Dis. 162:750-752.

238. Klintmalm, G., B. Lonnqvist, B. Oberg, G. Gahrton, J. Ler-nestedt, G. Lundgren, 0. Ringden, K. Robert, B. Wahren, andC. Groth. 1985. Intravenous foscarnet for the treatment ofsevere cytomegalovirus infection in allograft recipients. Scand.J. Infect. Dis. 17:157-163.

239. Knight, V., S. Z. Wilson, J. M. Quarles, S. E. Greggs, H. W.McClung, B. K. Waters, R. W. Cameron, J. M. Zerwas, andR. B. Couch. 1981. Ribavirin small-particle aerosol treatmentof influenza. Lancet ii:945-949.

240. Knop, J. 1990. Immunologic effects of interferon. J. Invest.Dermatol. 95(Suppl.):725-745.

241. Knox, K. K., W. R. Drobyski, and D. R. Carrigan. 1991.Cytomegalovirus isolate resistant to ganciclovir and foscarnetfrom a marrow transplant patient. Lancet 337:1292-1293.

242. Kochhar, D. M., J. D. Penner, and T. B. Knudsen. 1980.Embryotoxic, teratogenic, and metabolic effects of ribavirin inmice. Toxicol. Appl. Pharmacol. 52:99-112.

243. Koff, W. C., I. J. Fidler, S. D. Showalter, M. K. Chakrabarty,B. Hampar, L. M. Ceccorulli, and E. S. Kleinerman. 1984.Human monocytes activated by immunomodulators in lipo-somes lyse herpesvirus-infected but not normal cells. Science224:1007-1009.

244. Konopka, K., B. R. Davis, C. E. Larsen, D. R. Alford, R. J.Debs, and N. Duzgunes. 1990. Liposomes modulate humanimmunodeficiency virus infectivity. J. Gen. Virol. 71:2899-2907.

245. Korenman, J., B. Baker, J. Waggoner, J. E. Everhart, A. M. DiBisceglie, and J. H. Hoofnagle. 1991. Long-term remission ofchronic hepatitis B after alpha-interferon therapy. Ann. Intern.Med. 114:629-634.

246. Kornbluth, R. S., P. S. Oh, J. R. Munis, P. H. Cleveland, andD. D. Richman. 1990. The role of interferons in the control ofHIV replication in macrophages. Clin. Immunol. Immu-nopathol. 54:200-219.

247. Kornhauser, D. M., C. W. Hendrix, L. J. Nerhood, B. G. Petty,A. S. Woods, J. G. Bartlett, and P. S. Lietman. 1989. Proben-ecid and zidovudine metabolism. Lancet ii:473-475.

248. Kotler, M., R. A. Katz, W. Danho, J. Leis, and A. M. Skalka.1988. Synthetic peptides as substrates and inhibitors of aretroviral protease. Proc. Natl. Acad. Sci. USA 85:4185-4189.

249. Koup, R. A., V. J. Merluzzi, K. D. Hargrave, J. Adams, K.Grozinger, R. J. Eckner, and J. L. Sullivan. 1991. Inhibition ofhuman immunodeficiency virus type I (HIV-I) replication bythe dipyridodiazepinone B1-RG-587. J. Infect. Dis. 163:966-970.

250. Kovacs, J. A., L. Deyton, R. Davey, J. Falloon, K. Zunich, D.Lee, J. A. Metcalf, J. W. Bigley, L. A. Sawyer, K. C. Zoon, H.Masur, A. S. Fauci, and II. C. Lane. 1989. Combined zidovu-dine and interferon-alpha therapy in patients with KaposiSarcoma and the Acquired Immunodeficiency Syndrome(AIDS). Ann. Intern. Med. 111:280-287.

251. Koyanagi, Y., W. A. O'Brien, J. Q. Zhao, D. W. Golde, J. C.Gasson, and I. S. Y. Chen. 1988. Cytokines alter production ofHIV-I from primary mononuclear phagocytes. Science 241:1673-1675.

252. Krieger, J. N., R. W. Coombs, A. C. Collier, S. 0. Ross, K.Chaloupka, D. K. Cummings, V. L. Murphy, and L. Corey.1991. Recovery of human immunodeficiency virus type I fromsemen: minimal impact of stage of infection and currentantiviral chemotherapy. J. Infect. Dis. 163:386-388.

253. Krigel, R. L. 1986. Reversible neurotoxicity due to oral acy-clovir in a patient with chronic lymphocytic leukemia. J.Infect. Dis. 154:189.

254. Krown, S. E., J. W. M. Gold, D. Niedzwiecki, D. Bundow, N.Flomenberg, B. Gansbacher, and B. J. Brew. 1990. Interferon-alpha with zidovudine: safety, tolerance, and clinical andvirologic effects in patients with Kaposi Sarcoma associatedwith the Acquired Immunodeficiency Syndrome (AIDS). Ann.Intern. Med. 112:812-821.

255. Krugman, S., R. Ward, J. P. Giles, and A. M. Jacobs. 1960.Infectious hepatitis. Studies on the effect of gamma globulinand on the incidence of inapparent infection. JAMA 174:883-890.

256. Kuhls, T. L., J. Sacher, E. Pineda, D. Santomauro, E. Wies-meier, W. A. Growdon, and Y. J. Bryson. 1986. Suppression ofrecurrent genital herpes simplex virus infection with recombi-nant alpha 2 interferon. J. Infect. Dis. 154:437-442.

257. Lambert, J. S., M. Seidlin, R. C. Reichman, C. S. Plank, M.Laverty, G. D. Morse, C. Knupp, C. McLaren, C. Pettinelli,F. T. Valentine, and R. Dolin. 1990. 2',3'-Dideoxyinosine (ddl)in patients with the Acquired Immunodeficiency Syndrome orAIDS-related complex. N. Engl. J. Med. 322:1333-1340.

174 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 175

258. Land, S., G. Treloar, D. McPhee, C. Birch, R. Doherty, D.Cooper, and I. Gust. 1990. Decreased in vitro susceptibility tozidovudine of HIV isolates obtained from patients with AIDS.J. Infect. Dis. 161:326-329.

259. Lane, H. C., V. Davey, J. A. Kovacs, J. Feinberg, J. A. Metcalf,B. Herpin, R. Walker, L. Deyton, R. T. Davey, J. Falloon,M. A. Polis, N. P. Salzman, M. Baseler, H. Masur, and A. S.Fauci. 1990. Interferon-alfa in patients with asymptomatichuman immunodeficiency virus (HIV) infection. Ann. Intern.Med. 112:805-811.

260. Lane, H. C., J. Feinberg, V. Davey, L. Deyton, M. Baseler, J.Manischewitz, H. Masur, J. A. Kovacs, B. Herpin, R. Walker,J. A. Metcalf, N. Salzman, G. Quinnan, and A. S. Fauci. 1988.Anti-retroviral effects of interferon-alpha in AIDS-associatedKaposi's sarcoma. Lancet ii:1218-1222.

261. Lane, H. C., H. Masur, L. C. Edgar, G. Whalen, A. H. Rook,and A. S. Fauci. 1983. Abnormalities of B-cell activation andimmunoregulation in patients with the acquired immunodefi-ciency syndrome. N. Engl. J. Med. 309:453-458.

262. Lane, H. C., K. M. Zunich, W. Wilson, F. Cefali, M. Easter,J. A. Kovacs, H. Masur, S. F. Leitman, H. G. Klein, R. G. Steis,D. L. Longo, and A. S. Fauci. 1990. Syngeneic bone marrow

transplantation and adoptive transfer of peripheral blood lym-phocytes combined with zidovudine in human immunodefi-ciency virus (HIV) infection. Ann. Intern. Med. 113:512-519.

263. Lange, J. M. A., C. A. B. Boucher, C. E. M. Hollak, E. H. H.Wiltink, P. Reiss, E. A. van Royen, M. Roos, S. A. Danner, andJ. Goudsmit. 1990. Failure of zidovudine prophylaxis afteraccidental exposure to HIV-I. N. Engl. J. Med. 322:1375-1377.

264. Larder, B. A., B. Chesebro, and D. D. Richman. 1990. Suscep-tibilities of zidovudine-susceptible and -resistant human immu-nodeficiency virus isolates to antiviral agents determined byusing a quantitative plaque reduction assay. Antimicrob.Agents Chemother. 34:436-441.

265. Larder, B. A., and G. Darby. 1985. Selection and characteri-zation of acyclovir-resistant herpes simplex virus type I mu-tants inducing altered DNA polymerase activities. Virology146:262-271.

266. Larder, B. A., G. Darby, and D. D. Richman. 1989. HIV withreduced sensitivity to zidovudine (AZT) isolated during pro-longed therapy. Science 243:1731-1734.

267. Larder, B. A., and S. D. Kemp. 1989. Multiple mutations inHIV-I reverse transcriptase confer high-level resistance tozidovudine (AZT). Science 246:1155-1158.

268. Larder, B. A., D. J. M. Purifoy, K. L. Powell, C. Bradley, S.Kemp, M. Tisdale, P. Ertl, G. K. Darby, and D. Stammers.1988. Structural studies of the acquired immunodeficiencysyndrome virus reverse transcriptase. Am. J. Med. 85(Suppl.2A):173-175.

269. Larsson, A., and B. Oberg. 1981. Selective inhibition of her-pesvirus DNA synthesis by foscarnet. Antiviral Res. 1:55-62.

270. Larsson, A., K. Stenberg, and B. Oberg. 1978. Reversibleinhibition of cellular metabolism by ribavirin. Antimicrob.Agents Chemomther. 13:154-158.

271. Laskin, 0. L., D. M. Cederberg, J. Mills, L. J. Eron, D.Mildvan, S. A. Spector, and the Ganciclovir Study Group. 1987.Ganciclovir for the treatment and suppression of serious infec-tions caused by cytomegalovirus. Am. J. Med. 83:201-207.

272. Lau, A. S., G. E. Hannigan, M. H. Freedman, and B. R. G.Williams. 1986. Regulation of interferon receptor expression inhuman blood lymphocytes in vitro and during interferon ther-apy. J. Clin. Invest. 77:1632-1638.

273. Laurence, J. 1990. Molecular interactions among herpesvirusesand human immunodeficiency viruses. J. Infect. Dis. 162:338-346.

274. Lehoang, P., B. Girard, M. Robinet, P. Marcel, L. Zazoun, S.Matheron, W. Rozenbaum, C. Katlama, I. Morer, J. 0. Ler-nestedt, H. Saraux, Y. Pouliquen, M. Gentilini, and F. Rousse-lie. 1989. Foscarnet in the treatment of cytomegalovirus retini-tis in acquired immune deficiency syndrome. Ophthalmology96:865-874.

275. Leinbach, S. S., J. M. Reno, L. F. Lee, A. F. Isbell, and J. A.Boezi. 1976. Mechanism of phosphonoacetate inhibition of

herpesvirus-induced DNA polymerase. Biochemistry 15:426-430.

276. Lernstedt, J. O., and A. C. Chanas. 1990. Penile ulcerationswith foscarnet. Lan-et 335:548.

277. Lin, J., Z. Zhang, M. C. Smith, K. Biron, and J. S. Pagano.1988. Anti-human immunodeficiency virus agent 3'-azido-3'-deoxythymidine inhibits replication of Epstein-Barr virus. An-timicrob. Agents Chemother. 32:265-267.

278. Lin, J.-C., M. C. Smith, and J. S. Pagano. 1984. Prolongedinhibitory effect of 9-(1,3-dihydroxy-2-propoxymethyl)guanineagainst replication of Epstein-Barr virus. J. Virol. 50:50-55.

279. Little, J. W., W. J. Hall, R. G. Douglas, Jr., R. W. Hyde, andD. M. Speers. 1976. Amantadine effect on peripheral airwaysabnormalities in influenza. Ann. Intern. Med. 85:177-182.

280. Lok, A. S. F., P. C. Wu, C. L. Lai, and E. K. Y. Leung. 1988.Long-term follow-up in a randomised controlled trial of recom-binant alpha 2-interferon in Chinese patients with chronichepatitis B infection. Lancet ii:298-302.

281. Lubeck, M.-D., J. L. Schulman, and P. Palese. 1978. Suscep-tibility of influenza A viruses to amantadine is influenced bythe gene coding for M protein. J. Virol. 28:710-716.

282. Luby, J. P., J. W. Gnann, W. J. Alexander, V. A. Hatcher,A. E. Friedman-Kien, R. J. Klein, H. Keyserling, A. Nahmias,J. Mills, J. Schachter, J. M. Douglas, L. Corey, and S. L. Sacks.1984. A collaborative study of patient-initiated treatment ofrecurrent genital herpes with topical acyclovir or placebo. J.Infect. Dis. 150:1-6.

283. Machida, H., A. Kuninaka, and H. Yoshino. 1982. Inhibitoryeffects of antiherpesviral thymidine analogs against varicella-zoster virus. Antimicrob. Agents Chemother. 21:358-361.

284. Mansuri, M., J. E. Starrett, I. Ghazzouli, M. J. M. Hitchcock,R. Z. Sterzycki, V. Brankovan, T. S. Lin, E. M. August, W. H.Prusoff, J. Sommadossi, and J. C. Martin. 1989. (1-(2,3-Dide-oxy-beta-D-glycero-pent-2-enofuranosyl) thymine. A highlypotent and selective anti-HIV agent. J. Med. Chem. 32:461-466.

285. Mar, E. C., Y. C. Cheng, and E. S. Huang. 1983. Effect of9-(1,3-dihydroxy-2-propoxymethyl)guanine on human cyto-megalovirus replication in vitro. Antimicrob. Agents Chemo-ther. 24:518-521.

286. Mar, E. C., J. F. Chiou, Y. C. Cheng, and E. S. Huang. 1985.Inhibition of cellular DNA polymerase and human cytomega-lovirus-induced DNA polymerase by the triphosphates of9-(2-hydroxyethoxymethyl)guanine and 9-(1,3-dihydroxy-2-propoxymethyl)guanine. J. Virol. 53:776-780.

287. Marker, S. C., R. J. Howard, K. E. Groth, A. R. Mastri, R. L.Simmons, and H. H. Balfour, Jr. 1980. A trial of vidarabine forcytomegalovirus infection in renal transplant patients. Arch.Intern. Med. 140:1441-1444.

288. Marks, G. L., P. E. Nolan, K. S. Erlich, and M. N. Ellis. 1989.Mucocutaneous dissemination of acyclovir-resistant herpessimplex virus in a patient with AIDS. Rev. Infect. Dis.11:474-476.

289. Matsukura, M., G. Zon, K. Shinozuka, M. R. Guroff, T.Shimada, C. A. Stein, H. Mitsuya, F. Wong-Staal, J. S. Cohen,and S. Broder. 1989. Regulation of viral expression of humanimmunodeficiency virus in vitro by an antisense phospho-rothioate oligodeoxynucleotide against rev (art/trs) in chroni-cally infected cells. Proc. Natl. Acad. Sci. USA 86:4244-4248.

290. McCammon, J. A. 1987. Computer-aided molecular design.Science 238:486-491.

291. McClung, H. W., V. Knight, B. E. Gilbert, S. Z. Wilson, J. M.Quarles, and G. W. Divine. 1983. Ribavirin aerosol treatmentof influenza B virus infection. JAMA 249:2671-2674.

292. McCormick, J. B., J. P. Getchell, S. W. Mitchell, and D. R.Hicks. 1984. Ribavirin suppresses replication of lymphadenop-athy-associated virus in cultures of human adult T lympho-cytes. Lancet ii:1367-1369.

293. McCormick, J. B., I. J. King, P. A. Webb, C. L. Scribner,R. B. Craven, K. M. Johnson, L. H. Elliott, and R. B. Williams.1986. Lassa fever. Effective therapy with ribavirin. N. Engl. J.Med. 314:20-26.

294. McGuirt, P. V., J. E. Shaw, G. B. Elion, and P. A. Furman.

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

1984. Identification of small DNA fragments synthesized inherpes simplex virus-infected cells in the presence of acyclo-vir. Antimicrob. Agents. Chemother. 25:507-509.

295. McKeating, J. A., P. D. Griffiths, and R. A. Weiss. 1990. HIVsusceptibility conferred to human fibroblasts by cytomegalovi-rus-induced Fc receptor. Nature (London) 343:659-661.

296. McKendrick, M. W., J. I. McGill, J. E. White, and M. J. Wood.1986. Oral acyclovir in acute herpes zoster. Br. Med. J.293:1529-1532.

297. McKinney, R. E., S. L. Katz, and C. M. Wilfert. 1987. Chronicenteroviral meningoencephalitis in agammaglobulinemic pa-tients. Rev. Infect. Dis. 9:334-356.

298. McKinney, R. E., M. A. Maha, E. M. Connor, J. Feinberg,G. B. Scott, M. Wulfsohn, K. McIntosh, W. Borkowsky, J. F.Modlin, P. Weintrub, K. O'Donnell, R. D. Gelbert, G. K.Rogers, S. Nusinoff Lehrman, C. M. Wilfert, and the Protocol043 Study Group. 1991. A multicenter trial of oral zidovudine inchildren with advanced human immunodeficiency virus dis-ease. N. Engl. J. Med. 324:1018-1025.

299. McLaren, C., M. N. Ellis, and G. A. Hunter. 1983. A colori-metric assay for the measurement of the sensitivity of herpessimplex viruses to antiviral agents. Antiviral Res. 3:223-234.

300. Medical Letter. 1991. Granulocyte colony-stimulating factors.Med. Lett. 33:61-63.

301. Melder, R. J., R. Balachandran, C. R. Rinaldo, P. Gupta, T. L.Whiteside, and R. B. Herberman. 1990. Cytotoxic activityagainst HIV-infected monocytes by recombinant interleukin2-activated natural killer cells. AIDS Res. Hum. Retroviruses6:1011-1015.

302. Merigan, T. C., T. S. Hall, S. E. Reed, and D. A. J. Tyrrell.1973. Inhibition of respiratory virus infection by locally appliedinterferon. Lancet i:563-567.

303. Merigan, T. C., K. H. Rand, R. B. Pollard, P. S. Abdallah,G. W. Jordan, and R. P. Fried. 1978. Human leukocyte inter-feron for the treatment of herpes zoster in patients with cancer.N. Engl. J. Med. 298:981-987.

304. Merigan, T. C., G. Skowron, S. A. Bozzette, D. Richman, R.Uttamchandani, M. Fischl, R. Schooley, M. Hirsch, W. Soo, C.Pettinelli, H. Schaumburg, and the ddC Study Group of theAIDS Clinical Trials Group. 1989. Circulating p24 antigenlevels and responses to dideoxycytidine in human immunode-ficiency virus (HIV) infections. Ann. Intern. Med. 110:189-194.

305. Merluzzi, V. J., K. D. Hargrave, M. Labadia, K. Grozinder, M.Skoog, J. C. Wu, C. K. Shih, K. Eckner, S. Hattox, J. Adams,A. S. Rosenthal, R. Faanes, R. J. Eckner, R. A. Koup, and J. L.Sullivan. 1990. Inhibition of HIV-I replication by a nonnucle-oside reverse transcriptase inhibitor. Science 250:1411-1413.

306. Mertz, G. J., C. C. Jones, J. Mills, K. H. Fife, S. M. Lemon,J. T. Stapleton, E. L. Hill, L. G. Davis, and the Acyclovir StudyGroup. 1988. Long-term acyclovir suppression of frequentlyrecurring genital herpes simplex virus infection. JAMA 260:201-206.

307. Meyers, J. D., N. Flournoy, J. E. Sanders, R. W. McGuffin,B. A. Newton, L. D. Fisher, L. G. Lum, F. R. Appelbaum, K.Doney, K. M. Sullivan, R. Storb, C. D. Buckner, and E. D.Thomas. 1987. Prophylactic use of human leukocyte interferonafter allogenic marrow transplantation. Ann. Intern. Med.107:809-816.

308. Meyers, J. D., J. Leszcyzynski, J. A. Zaia, N. Flournoy, B.Newton, D. R. Snydman, G. G. Wright, M. J. Levin, and E. D.Thomas. 1973. Prevention of cytomegalovirus infection bycytomegalovirus immune globulin after marrow transplanta-tion. Ann. Intern. Med. 98:442-446.

309. Meyers, J. D., E. C. Reed, D. H. Schepp, M. Thornquist, P. S.Dandliker, C. A. Vicary, N. Flournoy, L. E. Kirk, J. H. Kersey,E. D. Thomas, and H. H. Balfour. 1988. Acyclovir for preven-tion of cytomegalovirus infection and disease after allogeneicmarrow transplantation. N. Engl. J. Med. 318:70-75.

310. Meyers, J. D., J. C. Wade, C. D. Mitchell, R. Saral, P. S.Lietman, D. T. Durack, M. J. Levin, A. C. Segreti, and H. H.Balfour. 1982. Multicenter collaborative trial of intravenousacyclovir for treatment of mucocutaneous herpes simplex virus

infection in the immunocompromised host. Am. J. Med.73(Suppl. 1A):229-235.

311. Miles, S. A., R. T. Mitsuyasu, J. Moreno, G. Baldwin, N. K.Alton, L. Souza, and J. A. Glaspy. 1991. Combined therapywith recombinant granulocyte colony-stimulating factor anderythropoietin decreases hematologic toxicity from zidovu-dine. Blood 77:2109-2117.

312. Miller, W. H., and R. L. Miller. 1990. Phosphorylation ofacyclovir (acycloguanosine) monophosphate by GMP kinase.J. Biol. Chem. 255:7204-7207.

313. Mindel, A., A. Faherty, D. Hindley, I. V. D. Weller, S.Sutherland, A. P. Fiddian, and M. W. Adler. 1984. Prophylac-tic oral acyclovir in recurrent genital herpes. Lancet ii:57-59.

314. Mindel, A. M., W. Adler, S. Sutherland, and A. P. Fiddian.1982. Intravenous acyclovir treatment for primary genitalherpes. Lancet i:697-700.

315. Mitsuya, H., and S. Broder. 1986. Inhibition of the in vitroinfectivity and cytopathic effect of human T-lymphotrophicvirus type III/lymphadenopathy-associated virus (HTLV-III/LAV) by 2',3'-dideoxynucleosides. Proc. Natl. Acad. Sci.USA 83:1911-1915.

316. Mitsuya, H., K. J. Weinhold, P. A. Furman, M. H. St. Clair, S.NusinoffLehrman, R. C. Gallo, D. Bolognesi, D. W. Barry, andS. Broder. 1985. 3'-Azido-3'-deoxythymidine (BW A509U): anantiviral agent that inhibits the infectivity and cytopathic effectof human T-lymphotropic virus type III/lymphadenopathy-associated virus in vitro. Proc. Natl. Acad. Sci. USA 82:7096-7100.

317. Monto, A. S., R. A. Gunn, M. G. Bandyk, and C. L. King.1979. Prevention of Russian influenza by amantadine. JAMA241:1003-1007.

318. Moore, R. D., H. Kessler, D. D. Richman, C. Flexner, and R. E.Chaisson. 1991. Non-Hodgkin's lymphoma in patients withadvanced HIV infection treated with zidovudine. JAMA 265:2208-2211.

319. Moyle, G., S. E. Barton, A. Balestrini, M. Nelson, M. Youle,D. A. Hawkins, and B. G. Gazzard. 1990. Penile ulcerationswith foscarnet. Lancet 335:547-548.

320. National Institute of Child Health and Human DevelopmentIntravenous Immunoglobulin Study Group. 1991. Intravenousimmune globulin for the prevention of bacterial infections inchildren with symptomatic human immunodeficiency virusinfection. N. Engl. J. Med. 325:73-80.

321. Nemunaitis, J., S. N. Rabinowe, J. W. Singer, P. J. Bierman,J. M. Vose, A. S. Freedman, N. Onetto, S. Gillis, D. Oette, M.Gold, C. D. Buckner, J. A. Hansen, J. Ritz, F. R. Applebaum,J. 0. Armitage, and L. M. Nadler. 1991. Recombinant granu-locyte-macrophage colony-stimulating factor after autologousbone marrow transplantation for lymphoid cancer. N. Engl. J.Med. 324:1773-1778.

322. Neto, C. C., J. M. Steim, P. S. Sarin, D. K. Sun, N. N. Bhongle,R. K. Piratla, and J. G. Turcotte. 1990. Lipid conjugates ofantiretroviral agents. II. Disodium palmityl phosphonofor-mate: anti-HIV activity, physical properties, and interactionwith plasma proteins. Biochem. Biophys. Res. Commun. 171:458-464.

323. Nilsen, A. E., T. Aasen, A. M. Halsos, B. R. Kinge, E. A. L.Tjotta, K. Wikstrom, and A. P. Fiddian. 1982. Efficacy of oralacyclovir in the treatment of initial and recurrent genitalherpes. Lancet ii:571-573.

324. Norley, S. G., L. Huang, and B. T. Rouse. 1986. Targeting ofdrug loaded immunoliposomes to herpes simplex virus infectedcorneal cells: an effective means of inhibiting virus replicationin vitro. J. Immunol. 136:681-685.

325. Nusinoff Lehrman, S., J. M. Douglas, L. Corey, and D. W.Barry. 1986. Recurrent genital herpes and suppressive oralacyclovir therapy. Relation between clinical outcome andin-vitro drug sensitivity. Ann. Intern. Med. 104:786-790.

326. Nyerges, G., Z. Meszner, E. Gyarmati, and S. Kerpel-Fronius.1988. Acyclovir prevents dissemination of varicella in immu-nocompromised children. J. Infect. Dis. 157:309-313.

327. Oberg, B. 1983. Inhibitors of virus-specific enzymes, p. 35-69.In C. H. Stuart-Harris and J. Oxford (ed.), Problems of

176 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 177

antiviral therapy. Academic Press, London.328. Oberg, B. 0. 1989. Antiviral effects of phosphonoformate

(PFA, foscarnet sodium). Pharmacol. Ther. 40:213-285.329. O'Brien, J. J., and D. M. Campoli-Richards. 1989. Acyclovir.

An updated review of its antiviral activity, pharmacokineticproperties and therapeutic efficacy. Drugs 37:233-309.

330. Ogle, J. W., P. Toetzis, W. D. Parker, N. Alvarez, K. McIntosh,M. J. Levin, and B. A. Lauer. 1989. Oral ribavirin therapy forsubacute sclerosing panencephalitis. J. Infect. Dis. 159:748-750.

331. Oker-Blom, N., T. Hovi, P. Leinikki, T. Palosuo, R. Pettersson,and J. Suni. 1970. Protection of man from natural infectionwith influenza A2 Hong Kong virus by amantadine: a con-trolled field trial. Br. Med. J. 3:676-678.

332. Oksenhendler, E., P. Bierling, F. Ferchal, J. P. Clauvel, and M.Seligmann. 1989. Zidovudine for thrombocytopenic purpurarelated to human immunodeficiency virus (HIV) infection.Ann. Intern. Med. 110:365-368.

333. Oleson, D., H. Grierson, J. Goldsmith, D. T. Purtilo, and D.Johnson. 1989. Augmentation of natural cytotoxicity by leucineenkephalin in cultured peripheral blood mononuclear cellsfrom patients infected with human immunodeficiency virus.Clin. Immunol. Immunopathol. 51:386-395.

334. Page, T., and J. D. Connor. 1990. The metabolism of ribavirinin erythrocytes and nucleated cells. Int. J. Biochem. 22:379-383.

335. Pahwa, S., K. Biron, W. Lim, P. Swenson, M. H. Kaplan, N.Sadick, and R. Pahwa. 1988. Continous varicella-zoster infec-tion associated with acyclovir resistance in a child with AIDS.JAMA 260:2879-2882.

336. Palestine, A. G., M. A. Polis, M. D. De Smet, B. F. Baird, J.Falloon, J. A. Kovacs, R. T. Davey, J. J. Zurlo, K. M. Zunich,M. Davis, L. Hubbard, R. Brothers, F. L. Ferris, E. Chew, J. L.Davis, B. I. Rubin, S. D. Mellow, J. A. Metcalf, J. Manis-chewitz, J. R. Minor, R. B. Nussenblatt, H. Masur, and H. C.Lane. 1991. A randomized, controlled trial of foscarnet in thetreatment of cytomegalovirus retinitis in patients with AIDS.Ann. Intern. Med. 115:665-673.

337. Parris, D. S., and J. E. Harrington. 1982. Herpes simplex virusvariants resistant to high concentrations of acyclovir exist inclinical isolates. Antimicrob. Agents Chemother. 22:71-77.

338. Patterson, J. L., and R. Fernandez-Larsson. 1990. Molecularmechanisms of action of ribavirin. Rev. Infect. Dis. 12:1139-1146.

339. Pauwels, R., K. Andries, J. Desmyter, D. Schols, M. J. Kukla,H. J. Breslin, A. Raeymaeckers, J. V. Gelder, R. Woesten-borghs, J. Heykants, K. Schellekens, M. A. C. Janssen, E.DeClercq, and P. A. J. Janseen. 1990. Potent and selectiveinhibition of HIV-I replication in vitro by a novel series ofTIBO derivatives. Nature (London) 343:470-474.

340. Pazin, G. J., J. H. Harger, J. A. Armstrong, M. K. Breinig,R. J. Caplan, K. Cantell, and M. Ho. 1987. Leukocyte inter-feron for treating first episodes of genital herpes in women. J.Infect. Dis. 156:891-898.

341. Peavy, D. L., W. C. Koff, D. S. Hyman, and V. Knight. 1980.Inhibition of lymphocyte proliferative responses by ribavirin.Infect. Immun. 29:583-589.

342. Pedersen, C., E. Sandstrom, C. S. Petersen, G. Norkrans, J.Gerstoft, A. Karlsson, K. C. Christensen, C. Hakansson, P. 0.Pehrson, J. 0. Nielsen, H. J. Jurgensen, and the ScandinavianIsoprinosine Study Group. 1990. The efficacy of inosine pra-nobex in preventing the acquired immunodeficiency syndromein patients with human immunodeficiency virus infection. N.Engl. J. Med. 322:1757-1763.

343. Pelling, J. C., J. C. Drach, and C. Shipman, Jr. 1981. Internu-cleotide incorporation of arabinosyladenine into herpes sim-plex virus and mammalian cell DNA. Virology 109:323-334.

344. Perno, C. F., R. Yarchoan, D. A. Cooney, N. R. Hartman, S.Gartner, M. Popovic, Z. Hao, T. L. Gerrard, Y. A. Wilson,D. G. Johns, and S. Broder. 1988. Inhibition of human immu-nodeficiency virus (HIV-I/HTLV-III/Ba-L) replication in freshand cultured human peripheral blood monocytes/macrophagesby azidothymidine and related 2',3'-dideoxynucleosides. J.

Exp. Med. 168:1111-1125.345. Perno, C. F., R. Yarchoan, D. A. Cooney, N. R. Hartman,

D. S. A. Webb, Z. Hao, H. Mitsuya, D. G. Johns, and S.Broder. 1989. Replication of human immunodeficiency virus inmonocytes. J. Exp. Med. 169:933-951.

346. Perren, T. J., R. L. Powles, D. Easton, K. Stolle, and P. J.Selby. 1988. Prevention of herpes zoster in patients by long-term oral acyclovir after allogeneic bone marrow transplanta-tion. Am. J. Med. 85(Suppl. 2A):99-101.

347. Perrillo, R. P., F. G. Regenstein, M. G. Peters, K. DeSchryver-Kecskemeti, C. J. Bodicky, C. R. Campbell, and M. C. Kuhns.1988. Prednisone withdrawal followed by recombinant alphainterferon in the treatment of chronic type B hepatitis. Ann.Intern. Med. 109:95-100.

348. Perrillo, R. P., E. R. Schiff, G. L. Davis, H. C. Bodenheimer, K.Lindsay, J. Payne, J. L. Dienstag, C. O'Brien, C. Tamburro,I. M. Jacobson, R. Sampliner, D. Feit, J. Lefkowitch, M.Kuhns, C. Meschievitz, B. Sanghvi, J. Albrecht, A. Gibas, andthe Hepatitis Interventional Therapy Group. 1990. A random-ized, controlled trial of interferon alfa-2b alone and afterprednisone withdrawal for the treatment of chronic hepatitis B.N. Engl. J. Med. 323:295-301.

349. Pestka, S., and J. A. Langer. 1987. Interferons and theiractions. Annu. Rev. Biochem. 56:727-777.

350. Peterslund, N. A., J. Ipsen, H. Schonheyder, K. Seyer-Hansen,V. Esmann, and H. Juhl. 1981. Acyclovir in herpes zoster.Lancet ii:827-830.

351. Pizzo, P. A., K. Butler, F. Balis, E. Brouwers, M. Hawkins, J.Eddy, M. Einloth, J. Falloon, R. Husson, P. Jarosinski, J. Meer,H. Moss, D. G. Poplack, S. Santacroce, L. Wiener, and P.Wolters. 1990. Dideoxycytidine alone and in an alternatingschedule with zidovudine in children with symptomatic humanimmunodeficiency virus infection. J. Pediatr. 117:799-808.

352. Pizzo, P. A., J. Eddy, J. Falloon, F. M. Balis, R. F. Murphy, H.Moss, P. Wolters, P. Brouwers, P. Jarosinski, M. Rubin, S.Broder, R. Yarchoan, A. Brunetti, M. Maha, S. Nusinoff-Lehrman, and D. G. Poplack. 1988. Effect of continuousintravenous infusion of zidovudine (AZT) in children withsymptomatic HIV infection. N. Engl. J. Med. 319:889-896.

353. Plotkin, S. A., S. E. Starr, and C. K. Bryan. 1982. In vitro andin vivo responses of cytomegalovirus to acyclovir. Am. J.Med. 73(Suppl. 1A):257-261.

354. Pluda, J. M., R. Yarchoan, E. S. Jaffe, I. M. Feuerstein, D.Solomon, S. M. Steinberg, K. M. Wyvill, A. Raubitschek, D.Katz, and S. Broder. 1990. Development of non-Hodgkinlymphoma in a cohort of patients with severe human immuno-deficiency virus (HIV) infection on long-term antiretroviraltherapy. Ann. Intern. Med. 113:276-282.

355. Plunkett, W., and S. S. Cohen. 1975. Metabolism of 9-p-D-arabinofuranosyladenine by mouse fibroblasts. Cancer Res.35:415-422.

356. Poli, G., J. M. Orenstein, A. Kinter, T. M. Folks, and A. S.Fauci. 1989. Interferon-alpha but not AZT suppresses HIVexpression in chronically infected cell lines. Science 244:575-577.

357. Potter, C. W., J. P. Phair, L. Vodinelich, R. Fenton, and R.Jennings. 1976. Antiviral, immunosuppressive and antitumoreffects of ribavirin. Nature (London) 259:496-497.

358. Powers, C. N., D. L. Peavy, and V. Knight. 1982. Selectiveinhibition of functional lymphocyte subpopulations by ribavi-rin. Antimicrob. Agents Chemother. 22:108-114.

359. Prichard, M. N., L. E. Prichard, W. A. Baguley, M. R. Nassiri,and C. Shipman. 1991. Three-dimensional analysis of thesynergistic cytotoxicity of ganciclovir and zidovudine. Antimi-crob. Agents Chemother. 35:1060-1065.

360. Prichard, M. N., S. R. Turk, L. A. Coleman, S. L. Engelhardt,C. Shipman, and J. C. Drach. 1990. A microtiter virus yieldreduction assay for the evaluation of antiviral compoundsagainst human cytomegalovirus and herpes simplex virus. J.Virol. Methods 28:101-106.

361. Prober, C. G., L. E. Kirk, and R. E. Keeney. 1982. Acyclovirtherapy of chickenpox in immunosuppressed children-a col-laborative study. J. Pediatr. 101:622-625.

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

362. Prusiner, P., and M. Sundaralingham. 1973. A new class ofsynthetic nucleoside analogues with broad-spectrum antiviralproperties. Nature (London) New Biol. 244:116-117.

363. Prusoff, W. H. 1959. Synthesis and biological activities ofiododeoxyuridine, an analog of thymidine. Biochim. Biophys.Acta 32:295-296.

364. Prusoff, W. H. 1963. A review of some aspects of 5-iododeox-yuridine and azauridine. Cancer Res. 23:1246-1259.

365. Prusoff, W. H., W. R. Mancini, T.-S. Lin, J. J. Lee, S. A.Siegel, and M. J. Otto. 1984. Physical and biological conse-quences of incorporation of antiviral agents into virus DNA.Antiviral Res. 4:303-315.

366. Rabalais, G. P., M. J. Levin, and F. E. Berkowitz. 1987. Rapidherpes simplex virus susceptibility testing using an enzyme-linked immunosorbent assay performed in situ on fixed virus-infected monolayers. Antimicrob. Agents Chemother. 31:946-948.

367. Rankin, J. T., Jr., S. B. Eppes, J. B. Antczak, and W. K. Jokik.1989. Studies on the mechanism of the antiviral activity ofribavirin against reovirus. Virology 168:147-158.

368. Ray, C. G. 1988. Ribavirin: ambivalence about an antiviralagent. Am. J. Dis. Child. 142:488-489.

369. Reardon, J. E., and T. Spector. 1989. Herpes simplex virustype I DNA polymerase: mechanism of inhibition by acyclovirtriphosphate. J. Biol. Chem. 264:7406-7411.

370. Redfield, R. R., D. L. Birx, N. Ketter, E. Tramont, V. Polonis,C. Davis, J. F. Brundage, G. Smith, S. Johnson, A. Fowler, T.Wierzba, A. Shafferman, F. Volvovitz, C. Oster, D. S. Burke,and the Military Medical Consortium for Applied RetroviralResearch. 1991. A phase I evaluation of the safety and immu-nogenicity of vaccination with recombinant gpl60 in patientswith early human immunodeficiency virus infection. N. Engl.J. Med. 324:1677-1684.

371. Reed, E. C., R. A. Bowden, P. S. Dandliker, K. E. Lilleby, andJ. D. Meyers. 1988. Treatment of cytomegalovirus pneumoniawith ganciclovir and intravenous cytomegalovirus immuno-globulin in patients with bone marrow transplants. Ann. In-tern. Med. 109:783-788.

372. Reed, E. C., J. L. Wolford, K. J. Kopecky, K. E. Lilleby, P. S.Dandliker, J. L. Todaro, G. B. McDonald, and J. D. Meyers.1990. Ganciclovir for the treatment of cytomegalovirus gastro-enteritis in bone marrow transplant patients. Ann. Intern.Med. 112:505-510.

373. Reichman, R. C., G. J. Badger, M. E. Guinan, A. J. Nahmias,R. E. Keeney, L. G. Davis, T. Ashikaga, and R. Dolin. 1983.Topically administered acyclovir in the treatment of recurrentherpes simplex genitalis: a controlled trial. J. Infect. Dis.147:336-340.

374. Reichman, R. C., G. J. Badger, G. J. Mertz, L. Corey, D. D.Richman, J. D. Connor, D. Redfield, M. C. Savoia, M. N.Oxman, Y. Bryson, D. L. Tyrrell, J. Portnoy, T. Creigh-Kirk,R. E. Keeney, T. Ashikage, and R. Dolin. 1984. Treatment ofrecurrent genital herpes simplex infections with oral acyclovir.JAMA 251:2103-2107.

375. Reichman, R. C., D. Oakes, W. Bonnez, C. Greisberger, S.Tyring, L. Miller, R. Whitley, H. Carveth, M. Weidner, G.Krueger, L. Yorkey, N. J. Roberts, and R. Dolin. 1988. Treat-ment of condyloma acuminatum with three different interfer-ons administered intralesionally. Ann. Intern. Med. 108:675-679.

376. Renault, P. F., J. H. Hoofnagle, Y. Park, K. D. Mullen, M.Peters, D. B. Jones, V. Rustgi, and E. A. Jones. 1987. Psychi-atric complications of long-term interferon alfa therapy. Arch.Intern. Med. 147:1577-1580.

377. Resnick, L., J. S. Herbst, D. V. Ablashi, S. Atherton, B. Frank,L. Rosen, and S. N. Horwitz. 1988. Regression of oral hairyleukoplakia after orally administered acyclovir therapy. JAMA259:384-388.

378. Reuman, P. D., D. I. Bernstein, M. C. Keefer, E. C. Young,J. R. Sherwood, and G. M. Schiff. 1989. Efficacy and safety oflow dosage amantadine hydrochloride as prophylaxis for influ-enza A. Antiviral Res. 11:27-40.

379. Richman, D., M. A. Fischl, M. H. Grikeco, M. S. Gottlieb, P. A.

Volberding, 0. L. Laskin, J. M. Leedom, J. E. Groopman, D.Mildvan, M. S. Hirsch, G. G. Jackson, D. T. Durak, S.Nusinoff-Lehrman, and the AZT Collaborative Working Group.1987. The toxicity of azidothymidine (AZT) in the treatment ofpatients with AIDS and AIDS-related complex. N. Engl. J.Med. 317:192-197.

380. Richman, D. D., J. M. Grimes, and S. W. Lagakos. 1990. Effectof stage of disease and drug dose on zidovudine susceptibilitiesof isolates of human immunodeficiency virus. J. AcquiredImmune Defic. Syndr. 3:743-746.

381. Richman, D. D., K. Y. Hostetler, P. J. Yazaki, and S. Clark.1986. Fate of influenza A virion proteins after entry intosubcellular fractions of LLC cells and the effect of amantadine.Virology 151:200-210.

382. Ringden, O., B. Lonnqvist, T. Paulin, J. Ahlmen, G. Klint-maim, B. Wahren, and J. 0. Lernestedt. 1986. Pharmacokinet-ics, safety and preliminary clinical experiences using foscarnetin the treatment of cytomegalovirus infections in bone marrowand renal transplant recipients. J. Antimicrob. Chemother.17:373-387.

383. Roberts, N. A., J. A. Martin, D. Kinchington, A. V. Broad-hurst, J. C. Craig, J. B. Duncan, S. A. Galpin, B. K. Handa, J.Kay, A. Krohn, R. W. Lambert, J. H. Merrett, J. S. Mills,K. E. B. Parkes, S. Redshaw, A. J. Ritchie, D. L. Taylor,G. J. Thomas, and P. J. Machin. 1990. Rational design ofpeptide-based HIV proteinase inhibitors. Science 248:358-361.

384. Roberts, R. B., G. M. Dickinson, P. N. R. Heseltine, J. M. Lee,P. W. A. Mansell, S. Rodriguez, K. M. Johnson, J. A. Lubina,R. W. Makuch, and the Ribavirin-LAS Collaborative Group.1990. A multicenter clinical trial of oral ribavirin in HIV-infected patients with lymphadenopathy. J. Acquired Immun.Defic. Syndr. 3:884-892.

385. Roberts, R. B., F. B. Hollinger, W. P. Parks, S. Rasheed, J.Laurence, P. N. R. Heseltine, R. W. Makuch, J. A. Lubina,K. M. Johnson, and the Ribavirin-LAS Collaborative Group.1990. A multicenter clinical trial of oral ribavirin in HIV-infected people with lymphadenopathy: virologic observa-tions. AIDS 4:67-72.

386. Roberts, R. B., K. Jurica, W. A. Meyer III, H. Paxton, andR. W. Makuch. 1990. A Phase I study of ribavirin in humanimmunodeficiency virus-infected patients. J. Infect. Dis. 162:638-642.

387. Roberts, R. B., 0. L. Laskin, J. Laurence, D. Scavuzzo, H. W.Murray, Y. T. Kim, and J. D. Connor. 1987. Ribavirin phar-macodynamics in high-risk patients for acquired immunodefi-ciency syndrome. Pharmacol. Ther. 42:365-373.

388. Rodriguez, W. J., H. W. Kim, C. D. Brandt, R. J. Fink, P. R.Getson, J. Arrobio, T. M. Murphy, V. McCarthy, and R. H.Parrott. 1987. Aerosolized ribavirin in the treatment of patientswith respiratory syncytial virus disease. Pediatr. Infect. Dis. J.6:159-163.

389. Roilides, E., D. Venzon, P. A. Pizzo, and M. Rubin. 1990.Effects of antiretroviral dideoxynucleosides on polymorphonu-clear leukocyte function. Antimicrob. Agents Chemother. 34:1672-1677.

390. Roizman, B. 1990. Multiplication of viruses. An overview, p.87-94. In B. N. Fields and D. M. Knipe (ed.), Fields virology,2nd ed. Raven Press, New York.

391. Rooke, R., M. Tremblay, H. Soudeyns, L. DeStephano, X.-J.Yao, M. Fanning, J. S. G. Montaner, M. O'Shaughnessy, K.Gelmon, C. Tsoukas, J. Gill, J. Ruedy, M. A. Wainberg, andthe Canadian Zidovudine Multi-Centre Study Group. 1989.Isolation of drug-resistant variants of HIV-I from patients on

long-term zidovudine therapy. AIDS 3:411-415.392. Rooney, J. F., J. M. Felser, J. M. Ostrove, and S. E. Straus.

1986. Acquisition of genital herpes from an asymptomaticsexual partner. N. Engl. J. Med. 314:1561-1564.

393. Rose, K. M., and S. T. Jacob. 1978. Selective inhibition ofRNA polyadenylation by Ara-ATP in vitro: a possible mecha-nism for antiviral action of ARA-A. Biochem. Biophys. Res.Commun. 81:1418-1424.

394. Ross, A. H., A. Julia, and C. Balakrishnan. 1976. Toxicity of

178 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 179

adenine arabinoside in humans. J. Infect. Dis. 133(Suppl.):A192-A198.

395. Rossmann, M. G. 1988. Antiviral agents targeted to interactwith viral capsid proteins and a possible application to humanimmunodeficiency virus. Proc. Natl. Acad. Sci. USA 85:4625-4627.

396. Rossmann, M. G., E. Arnold, J. W. Erickson, E. A. Franken-berger, J. P. Griffith, H.-J. Hecht, J. E. Johnson, G. Kamer, M.Luo, A. G. Mosser, R. R. Rueckert, B. Sherry, and G. Vriend.1985. Structure of a human common cold virus and functionalrelationship to other picornaviruses. Nature (London) 317:145-153.

397. Rothenberg, M., G. Johnson, C. Laughlin, I. Green, J. Cra-dock, N. Sarver, and J. S. Cohen. 1989. Oligodeoxynucleotidesas anti-sense inhibitors of gene expression: therapeutic impli-cations. J. Natl. Cancer Inst. 81:1539-1544.

398. Sacks, S. L., T. C. Merigan, J. Kaminska, and I. H. Fox. 1982.Inactivation of S-adenosylhomocysteine hydrolase during ad-enine arabinoside therapy. J. Clin. Invest. 69:226-30.

399. Sacks, S. L., R. J. Wanklin, D. E. Reece, K. A. Hicks, K. L.Tyler, and D. M. Coen. 1989. Progressive esophagitis fromacyclovir-resistant herpes simplex. Ann. Intern. Med. 111:893-899.

400. Safrin, S., T. Assaykeen, S. Follansbee, and J. Mills. 1990.Foscarnet therapy for acyclovir-resistant mucocutaneous her-pes simplex virus infection in 26 AIDS patients: preliminarydata. J. Infect. Dis. 161:1078-1084.

401. Safron, S., T. G. Berger, I. Gilson, P. R. Wolfe, C. B. Wofsy, J.Mills, and K. K. Biron. 1991. Foscarnet therapy in five patientswith AIDS and acyclovir-resistant varicella-zoster virus infec-tion. Ann. Intern. Med. 115:19-21.

402. Safrin, S., C. Crumpacker, P. Chatis, R. Davis, R. Hafner, J.Rush, H. A. Kessler, B. Landry, J. Mills, and other members ofthe AIDS Clinical Trials Group. 1991. A controlled trial com-paring foscarnet with vidarabine for acyclovir-resistant muco-cutaneous herpes simplex in the acquired immunodeficiencysyndrome. N. Engl. J. Med. 325:551-555.

403. Salk, J. 1987. Prospects for the control of AIDS by immunizingseropositive individuals. Nature (London) 327:473-476.

404. Sandstrom, E. G., R. E. Byington, J. C. Kaplan, and M. S.Hirsch. 1985. Inhibition of human T-cell lymphotropic virustype III in vitro by phosphonoformate. Lancet i:1480-1482.

405. Saral, R., R. F. Ambinder, W. H. Burns, C. M. Angelopulos,D. E. Griffin, P. J. Burke, and P. S. Lietman. 1983. Acyclovirprophylaxis against herpes simplex virus infection in patientswith leukemia. Ann. Intern. Med. 99:773-776.

406. Saral, R., W. H. Burns, 0. L. Laskin, G. W. Santos, and P. S.Lietman. 1981. Acyclovir prophylaxis of herpes-simplex-virusinfections. N. Engl. J. Med. 305:63-67.

407. Sawyer, M. H., D. E. Webb, J. E. Balow, and S. E. Straus.1988. Acyclovir-induced renal failure. Am. J. Med. 84:1067-71.

408. Scadden, D. T., H. A. Bering, J. D. Levine, J. Bresnahan, L.Evans, C. Epstein, and J. E. Groopman. 1991. Granulocyte-macrophage colony-stimulating factor mitigates the neutrope-nia of combined interferon alfa and zidovudine treatment ofacquired immune deficiency syndrome-associated Kaposi'sSarcoma. J. Clin. Oncol. 9:802-808.

409. Schabel, F. M., Jr. 1968. The antiviral activity of 9-,-D-Arabinofuranosyladenine. (ARA-A). Chemotherapy 13:321-338.

410. Schaeffer, H. J., L. Beauchamp, P. de Miranda, and G. B.Elion, D. J. Bauer, and P. Collins. 1978. 9-(2-Hydroxyethoxy-methyl) guanine activity against viruses of the herpes group.Nature (London) 272:583-585.

411. Schmidt, G. M., D. A. Horak, J. C. Niland, S. R. Duncan, S. J.Forman, J. A. Zaia, and the City of Hope-Stanford-Syntex CMVStudy Group. 1991. A randomized, controlled trial of prophy-lactic ganciclovir for cytomegalovirus pulmonary infection inrecipients of allogeneic bone marrow transplants. N. Engl. J.Med. 324:1005-1011.

412. Schmitt, F. A., J. W. Bigley, R. McKinnis, P. E. Logue, R. W.Evans, J. L. Drucker, and the AZT Collaborative Working

Group. 1988. Neuropsychological outcome of zidovudine(AZT) treatment of patients with AIDS and AIDS-relatedcomplex. N. Engl. J. Med. 319:1573-1578.

413. Schnipper, L. E., and C. S. Crumpacker. 1980. Resistance ofherpes simplex virus to acycloguanosine: role of viral thymi-dine kinase and DNA polymerase loci. Proc. Natl. Acad. Sci.USA 77:2270-2273.

414. Schooley, R. T., T. C. Merigan, P. Gaut, M. S. Hirsch, M.Holodniy, T. Flynn, S. Liu, R. E. Byington, S. Henochowicz, E.Gubish, D. Kufe, J. Schindler, A. Dawson, D. Thomas, D. G.Hanson, B. Letwin, T. Liu, J. Gulinello, S. Kennedy, R. Fisher,and D. D. Ho. 1990. Recombinant soluble CD4 therapy inpatients with the Acquired Immunodeficiency Syndrome(AIDS) and AIDS-related complex. Ann. Intern. Med. 112:247-253.

415. Schulman, K. A., L. A. Lynn, H. A. Glick, and J. M. Eisenberg.1991. Cost effectiveness of low-dose zidovudine therapy forasymptomatic patients with human immunodeficiency virus(HIV) infection. Ann. Intern. Med. 114:798-802.

416. Schwartz, D. H., G. Skowron, and T. C. Merigan. 1991. Safetyand effects of interleukin-2 plus zidovudine in asymptomaticindividuals infected with human immunodeficiency virus. J.Acquired Immune Defic. Syndr. 4:11-23.

417. Schwartz, P. M., C. Shipman, Jr., and J. C. Drach. 1976.Antiviral activity of arabinosyladenine and arabinosylhypox-anthine in herpes simplex virus-infected KB cells: selectiveinhibition of viral deoxyribonucleic acid synthesis in the pres-ence of an adenosine deaminase inhibitor. Antimicrob. AgentsChemother. 10:64-74.

418. Seale, L., C. J. Jones, S. Kathpalia, G. G. Jackson, M. Mozes,M. S. Maddux, and D. Packham. 1985. Prevention of herpes-virus infections in renal allograft recipients by low-dose oralacyclovir. JAMA 254:3435-3438.

419. Sears, S. D., and M. L. Clements. 1987. Protective efficacy oflow-dose amantadine in adults challenged with wild-type influ-enza A virus. Antimicrob. Agents Chemother. 31:1470-1473.

420. Seeff, L. B., E. C. Wright, H. J. Zimmerman, H. J. Alter, A. A.Dietz, B. F. Felsher, J. D. Finkelstein, P. Garcia-Pont, J. L.Gerin, H. B. Greenlee, J. Hamilton, P. V. Holland, P. M.Kaplan, T. Kiernan, R. S. Koff, C. M. Leevy, V. J. McAuliffe,N. Nath, R. H. Purcell, E. R. Schiff, C. C. Schwartz, C. H.Tamburro, Z. Vlahcevic, R. Zemel, and D. S. Zimmon. 1978.Type B hepatitis after needle-stick exposure: prevention withhepatitis B immune globulin. Ann. Intern. Med. 88:285-293.

421. Selby, P. J., R. L. Powles, D. Easton, T. J. Perren, K. Stolle, B.Jameson, A. P. Fiddian, Y. Tryhorn, and H. Stern. 1989. Theprophylactic role of intravenous and long-term oral acyclovirafter allogeneic bone marrow transplantation. Br. J. Cancer59:434-438.

422. Shepp, D. H., P. S. Dandliker, P. deMiranda, T. C. Burnette,D. M. Cederberg, L. E. Kirk, and J. D. Meyers. 1985. Activityof 9-(2-hydroxy-1-(hydroxymethyl)ethoxymethyl)guanine inthe treatment of cytomegalovirus pneumonia. Ann. Intern.Med. 103:368-373.

423. Shepp, D. H., P. S. Dandliker, N. Flournoy, and J. D. Meyers.1987. Sequential intravenous and twice-daily oral acyclovir forextended prophylaxis of herpes simplex virus infection inmarrow transplant patients. Transplantation 43:654-658.

424. Shepp, D. H., P. S. Dandliker, and J. D. Meyers. 1986.Treatment of varicella-zoster virus infection in severely immu-nocompromised patients. N. Engl. J. Med. 314:208-212.

425. Shepp, D. H., B. A. Newton, P. S. Dandliker, N. Flournoy, andJ. D. Meyers. 1985. Oral acyclovir therapy for mucocutaneousherpes simplex virus infections in immunocompromised mar-row transplant recipients. Ann. Intern. Med. 102:783-785.

426. Shigeta, S., S. Mori, T. Yokota, K. Konno, and E. De Clercq.1986. Characterization of a varicella-zoster virus variant withaltered thymidine kinase activity. Antimicrob. Agents Chemo-ther. 29:1053-1058.

427. Shindo, M., A. M. Di Bisceglie, L. Cheung, J. W.-K. Shih, K.Cristiano, S. M. Feinstone, and J. H. Hoofnagle. 1991. Decreasein serum hepatitis C viral RNA duirng alpha-interferon therapyfor chronic hepatitis C. Ann. Intern. Med. 115:700-704.

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

428. Sibrack, C. D., L. T. Gutman, C. M. Wilfert, C. McLaren,M. H. St. Clair, P. M. Keller, and D. W. Barry. 1982. Patho-genicity of acyclovir-resistant herpes simplex virus type I froman immunodeficient child. J. Infect. Dis. 146:673-682.

429. Sidwell, R. W., J. H. Huffman, G. P. Khare, L. B. Allen, J. T.Witkowski, and R. K. Robins. 1972. Broad spectrum antiviralactivity of virazole: ribofuranosyl-1,2,4-triazole-3-carbox-amide. Science 177:705-706.

430. Silvestris, F., A. Gernone, M. A. Frassanito, and F. Dammacco.1989. Immunologic effects of long-term thymopentin treatmentin patients with HIV-induced lymphadenopathy syndrome. J.Lab. Clin. Med. 113:139-144.

431. Sim, I. S., R. L. Cerruti, and E. V. Connell. 1989. Anti-influenza virus activity of rimantadine in cells in culture. J.Respir. Dis. 10(Suppl.):546-551.

432. Sjovall, J., S. Bergdahl, G. Movin, S. Ogenstad, and M.Saarimaki. 1989. Pharmacokinetics of foscarnet and distribu-tion to cerebrospinal fluid after intravenous infusion in patientswith human immunodeficiency virus infection. Antimicrob.Agents Chemother. 33:1023-1031.

433. Sjovall, J., A. Karlsson, S. Ogenstad, E. Sandstrom, and M.Saarimaki. 1988. Pharmacokinetics and absorpition of foscar-net after intravenous and oral administration to patients withhuman immunodeficiency virus. Clin. Pharmacol. Ther. 44:65-73.

434. Skoldenberg, B., K. Alestig, L. Burman, A. Korkman, K.Lovgren, R. Norrby, G. Stiernstedt, M. Forsgren, T. Berg-strom, E. Dahlqvist, A. Fryden, K. Norlin, E. Olding-Stenkvist,I. Uhnoo, and K. deVahl. 1984. Acyclovir versus vidarabine inherpes simplex encephalitis. Lancet ii:707-711.

435. Skowron, G., and T. M. Merigan. 1990. Alternating andintermittent regimens of zidovudine (3'-azido-3'-deoxythymi-dine) and dideoxycytidine (2',3'-dideoxycytidine) in the treat-ment of patients with acquired immunodeficiency syndrome(AIDS) and AIDS-related complex. Am. J. Med. 88(Suppl.5B):20S-23S.

436. Smee, D. F., J. C. Martin, J. P. H. Verheyden, and T. R.Matthews. 1983. Anti-herpesvirus activity of the acyclic nucle-oside 9-(1,3-dihyhdroxy-2-propoxymethyl) guanine. Antimi-crob. Agents Chemother. 23:676-682.

437. Smee, D. F., and T. R. Matthews. 1986. Metabolism of ribavirinin respiratory syncytial virus-infected and uninfected cells.Antimicrob. Agents Chemother. 30:117-121.

438. Smith, D. H., R. A. Byrn, S. A. Marsters, T. Gregory, J. E.Groopman, and D. J. Capon. 1987. Blocking of HIV-I infectiv-ity by a soluble, secreted form of the CD4 antigen. Science238:1704-1707.

439. Smith, D. W., L. R. F. Frankel, L. H. Mathers, A. T. S. Tang,R. L. Ariagno, and C. G. Prober. 1991. A controlled trial ofaerosolized ribavirin in infants receiving mechanical ventila-tion for severe respiratory syncytial virus infection. N. Engl. J.Med. 325:24-29.

440. Smith, T. F. 1987. Rapid methods for the diagnosis of viralinfections. Lab. Med. 18:16-20.

441. Smith, T. J., M. J. Kremer, M. Luo, G. Vriend, E. Arnold, G.Kamer, M. G. Rossmann, M. A. McKinlay, G. D. Diana, andM. J. Otto. 1986. The site of attachment in human rhinovirus 14for antiviral agents that inhibit uncoating. Science 233:1286-1293.

442. Snydman, D. R., B. G. Werner, B. Heinze-Lacey, V. P.Bernardi, N. L. Tilney, R. L. Kirkman, E. L. Milford, S. I. Cho,H. L. Bush, A. S. Levey, T. B. Strom, C. B. Carpenter, R. H.Levey, W. E. Harmon, C. E. Zimmerman, M. E. Shapiro, T.Steinman, F. LoGerfo, B. Idelson, G. P. J. Schroter, M. J.Levin, J. McIver, J. Leszcynski, and G. F. Grady. 1987. Use ofcytomegalovirus immune globulin to prevent primary cytomeg-alovirus disease in renal-transplant recipients. N. Engl. J.Med. 317:1049-1054.

443. Sommadossi, J. P., R. Bevan, T. Ling, F. Lee, B. Mastre, M. D.Chaplin, C. Nerenberg, S. Koretz, and W. C. Buhles, Jr. 1988.Clinical pharmacokinetics of ganciclovir in patients with nor-mal and impared renal function. Rev. Infect. Dis. 10(Suppl.3):S507-S514.

444. Sommadossi, J.-P., and R. Carlisle. 1987. Toxicity of 3-azido-3'deoxythymidine and 9-(1,3-dihydroxy-2 propoxymethyl)guanine for normal human hematopoietic progenitor cells invitro. Antimicrob. Agents Chemother. 31:452-454.

445. Sonnenblick, M., and A. Rosin. 1991. Cardiotoxicity of inter-feron. A review of 44 cases. Chest. 99:557-561.

446. Spear, J. B., C. A. Benson, J. C. Pottage, D. A. Paul, A. L.Landay, and H. A. Kessler. 1988. Rapid rebound of serumhuman immunodeficiency virus antigen after discontinuingzidovudine therapy. J. Infect. Dis. 158:1132-1133.

447. Spiegel, R. J., J. R. Spicehandler, S. L. Jacobs, and E. M.Oden. 1986. Low incidence of serum neutralizing factors inpatients receiving recombinant alfa-2b interferon (Intron A).Am. J. Med. 80:223-228.

448. Spruance, S. L., D. J. Freeman, J. C. B. Stewart, M. B.McKeough, L. G. Wenerstrom, G. G. Krueger, M. W. Piep-korn, W. G. Stroop, and N. H. Rowe. 1991. The natural historyof ultraviolet radiation-induced herpes simplex labialis andresponse to therapy with peroral and topical formulations ofacyclovir. J. Infect. Dis. 163:728-734.

449. Spruance, S. L., M. L. Hamill, W. S. Hoge, L. G. Davis, and J.Mills. 1988. Acyclovir iprevents reactivation of herpes simplexlabialis in skiers. JAMA 260:1597-1599.

450. Spruance, S. L., J. C. B. Stewart, D. J. Freeman, V. J.Brightman, J. L. Cox, G. Wenerstrom, M. B. McKeough, andN. H. Rowe. 1990. Early application of topical 15% idoxuridinein dimethyl sulfoxide shortens the course of herpes simplexlabialis: a multicenter placebo-controlled trial. J. Infect. Dis.161:191-197.

451. Spruance, S. L., J. C. B. Stewart, N. H. Rowe, M. B. McKe-ough, G. Wenerstrom, and D. J. Freeman. 1990. Treatment ofrecurrent herpes simplex labialis with oral acyclovir. J. Infect.Dis. 161:185-190.

452. Stanat, S. C., J. E. Reardon, A. Erice, M. C. Jordon, W. L.Drew, and K. K. Biron. 1991. Ganciclovir-resistant cytomega-lovirus clinical isolates: mode of resistance to ganciclovir.Antimicrob. Agents Chemother. 35:2191-2197.

453. St. Clair, M. H., P. A. Furman, C. M. Lubbers, and G. B.Elion. 1980. Inhibition of cellular alpha and virally induceddeoxyribonucleic acid polymerase by the triphosphate of acy-clovir. Antimicrob. Agents Chemother. 18:741-745.

454. St. Clair, M. H., C. A. Richards, T. Spector, K. J. Weinhold,W. H. Miller, A. J. Langlois, and P. A. Furman. 1987.3'-Azido-3'-deoxythymidine triphosphate as an inhibitor andsubstrate of purified human immunodeficiency virus reversetranscriptase. Antimicrob. Agents Chemother. 31:1972-1977.

455. Steele, R. W., and R. K. Charlton. 1987. Immune modulatorsas antiviral agents, p. 911-924. In W. L. Drew (ed.), Clinics inlaboratory medicine. The W. B. Saunders Co., Philadelphia.

456. Stevens, J. G. 1989. Human herpesviruses: a consideration ofthe latent state. Microbiol. Rev. 53:318-332.

457. Straus, S. E., M. Seidlin, H. E. Takiff, J. F. Rooney, J. M.Felser, H. A. Smith, P. Roane, F. Johnson, C. Hallahan, J. M.Ostrove, and S. Nusinoff-Lehrman. 1989. Effect of oral acyclo-vir treatment of symptomatic and asymptomatic virus sheddingin recurrent genital herpes. Sex. Transm. Dis. 16:107-113.

458. Streeter, D. G., J. T. Witkowski, G. P. Khare, R. W. Sidwell,R. J. Bauer, R. K. Robins, and L. N. Simon. 1973. Mechanismof action of 1-p-D-ribofuranosyl-1,2,4-triazole-3-carboxamide(Virazole), a new broad-spectrum antiviral agent. Proc. Natl.Acad. Sci. USA 70:1174-1178.

459. Stridh, S. 1983. Determination of ribonucleoside triphosphatepools in influenza A virus-infected MDCK cells. Arch. Virol.77:223-229.

460. Stuart-Harris, C. 1983. Antiviral chemotherapy; an introduc-tion and apology for the slow progress, p. 1-12. In C. H.Stuart-Harris and J. Oxford (ed.), Problems of antiviral che-motherapy. Academic Press, Ltd., London.

461. Sugrue, R. J., G. Bahadur, M. C. Zambon, M. Hall-Smith,A. R. Douglas, and A. J. Hay. 1990. Specific structural altera-tion of the influenza hemagglutinin by amantadine. EMBO J.9:3469-3476.

462. Sullivan, K. M., K. J. Kopecky, J. Jocom, L. Fisher, C. D.

180 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

ANTIVIRAL THERAPY 181

Buckner, J. D. Meyers, G. W. Counts, R. A. Bowden, F. B.Petersen, R. P. Witherspoon, M. D. Budinger, R. S. Schwartz,F. R. Applebaum, R. A. Clift, J. A. Hansen, J. E. Sanders,E. D. Thomas, and R. Storb. 1990. Immunomodulatory andantimicrobial efficacy of intravenous immunoglobulin in bonemarrow transplantation. N. Engl. J. Med. 323:705-712.

463. Sullivan, V., K. Biron, M. Davis, S. Stanat, C. Talarico, and D.Coen. 1991. Resistance of HCMV to DHPG, HPMPC andHPMPA mapped to the viral DNA polymerase gene. AntiviralRes. S1:84.

464. Sweet, C., F. G. Hayden, K. J. Jakeman, S. Grambas, and A. J.Hay. 1991. Virulence of Rimantadine-resistant human influ-enza A (H3N2) viruses in ferrets. J. Infect. Dis. 164:969-972.

465. Swierkosz, E. M., D. R. Scholl, J. L. Brown, J. D. Jollick, andC. A. Gleaves. 1987. Improved DNA hybridization method fordetection of acyclovir-resistant herpes simplex virus. Antimi-crob. Agents Chemother. 31:1465-1469.

466. Swiss Group for Clinical Studies on the Acquired Immunodefi-ciency Syndrome (AIDS). 1988. Zidovudine for the treatment ofthrombocytopenia associated with human immunodeficiencyvirus (HIV). Ann. Intern. Med. 109:718-721.

467. Syntex Laboratories. 1990. Cytovene (package insert). SyntexLaboratories, Inc., Palo Alto, Calif.

468. Szoka, F. C., and C. J. Chu. 1988. Increased efficacy ofphosphonoformate and phosphonoacetate inhibition of herpessimplex virus type 2 replication by encapsulation in liposomes.Antimicrob. Agents Chemother. 32:858-864.

469. Taber, L. H., V. Knight, B. E. Gilbert, H. W. McClung, S. Z.Wilson, J. Norton, J. M. Thurson, W. H. Gordon, R. L. Atmar,and W. R. Schlaudt. 1983. Ribavirin aerosol treatment ofbronchiolitis associated with respiratory syncytial virus infec-tion in infants. Pediatrics 72:613-618.

470. Telenti, A., and T. F. Smith. 1989. Screening with a shell vialassay for antiviral activity against cytomegalovirus. Diagn.Microbiol. Infect. Dis. 12:5-8.

471. Tenser, R. B., and W. A. Edris. 1987. Trigeminal ganglioninfection by thymidine kinase-negative mutants of herpes sim-plex virus after in vivo complementation. J. Virol. 61:2171-2174.

472. Thin, R. N., J. M. Nabarro, J. D. Parker, and A. P. Fiddian.1983. Topical acyclovir in the treatment of initial genitalherpes. Br. J. Vener. Dis. 59:116-119.

473. Till, M. A., V. Ghetie, T. Gregory, E. J. Patzer, J. P. Porter,J. W. Uhr, D. J. Capon, and E. S. Vitetta. 1988. HIV-infectedcells are killed by rCD4-ricin A chain. Science 242:1166-1168.

474. Tocci, M. J., T. J. Livcelli, H. C. Perry, C. S. Crumpacker, andA. K. Field. 1984. Effects of the nucleoside analog 2'-nor-2'-deoxyguanosine on human cytomegalovirus replication. Anti-microb. Agents Chemother. 25:247-252.

475. Togo, Y., R. B. Hornick, J. J. Felitti, V. J. Felitti, M. I.Kaufman, A. T. Dawkins, Jr., V. E. Kilpe, and J. L. Glaghorn.1970. Evaluation of therapeutic efficacy of amantadine inpatients with naturally occurring A2 influenza. JAMA 211:1149-1156.

476. Toltzis, P., C. M. Marx, N. Kleinman, E. M. Levine, and E. V.Schmidt. 1991. Zidovudine-associated embryonic toxicity inmice. J. Infect. Dis. 163:1212-1218.

477. Toltzis, P., K. O'Connell, and J. L. Patterson. 1988. Effect ofphosphorylated ribavirin on vesicular stomatitis virus tran-scription. Antimicrob. Agents Chemother. 32:492-497.

478. Traunecker, A., J. Schneider, H. Kiefer, and K. Karjalainen.1989. Highly efficient neutralization of HIV with recombinantCD4-immunoglobulin molecules. Nature (London) 339:68-70.

479. Tucker, W. E. 1982. Preclinical toxicology profile of acyclovir:an overview. Am. J. Med. 73(Suppl. 1A):27-30.

480. Vance, J. C., and D. Davis. 1990. Interferon alpha-2b injectionsused as an adjuvant therapy to carbon dioxide laser vaporiza-tion of recalcitrant ano-genital condylomata acuminata. J.Invest. Dermatol. 95(Suppl.):146S-148S.

481. Van der Horst, C., J. Joncas, G. Ahronheim, N. Gustafson, G.Stein, M. Gurwith, G. Fleisher, J. Sullivan, J. Sixbey, S.Roland, J. Fryer, K. Champney, R. Schooley, C. Sumaya, andJ. S. Pagano. 1991. Lack of effect of peroral acyclovir for the

treatment of acute infectious mononucleosis. J. Infect. Dis.164:788-792.

482. Van Der Pijl, J. W., P. H. J. Frissen, P. Reiss, H. J. Hulsebosch,J. G. Van Den Tweel, J. M. A. Lange, and S. A. Danner. 1990.Foscarnet and penil ulcertation. Lancet 335:286.

483. Van Voris, L. P., R. F. Betts, F. G. Hayden, W. A. Christmas,and R. G. Douglas, Jr. 1981. Successful treatment of naturallyoccurring influenza A/USSR/77 HlNl. JAMA 245:1128-1131.

484. Vere Hodge, R. A., and R. M. Perkins. 1989. Mode of action of9-(4-hydroxy-3-hydroxymethylbut-1-yl)guanine (BRL 39123)against herpes simplex virus in MRC-5 cells. Antimicrob.Agents Chemother. 33:223-229.

485. Vere Hodge, R. A., D. Sutton, M. R. Boyd, M. R. Harnden, andR. L. Jarvest. 1989. Selection of an oral prodrug (BRL 42810;Famciclovir) for the antiherpesvirus agent BRL 39123 [9-(4-hydroxy-3-hydroymethylbut-1-yl)guanine; penciclovir]. Anti-microb. Agents Chemother. 33:1765-1773.

486. Vogt, M. W., K. L. Hartshorn, P. A. Furman, T. C. Chou,J. A. Fyfe, L. A. Coleman, C. Crumpacker, R. T. Schooley, andM. S. Hirsch. 1987. Ribavirin antagonizes the effect of azido-thymidine on HIV replication. Science 235:1376-1379.

487. Volberding, P. A., S. W. Lagakos, M. A. Koch, C. Pettinelli,M. W. Myers, D. K. Booth, H. H. Balfour, R. C. Reichman,J. A. Bartlet, M. S. Hirsch, R. L. Murphy, W. D. Hardy, R.Soeiro, M. A. Fischl, J. G. Bartlett, T. C. Merigan, N. E.Hyslop, D. D. Richman, F. T. Valentine, L. Corey, and theAIDS Clinical Trials Group of the National Institute of Allergyand Infectious Diseases. 1990. Zidovudine in asymptomatichuman immunodeficiency virus infection. N. Engl. J. Med.322:941-949.

488. Vrang, L., and B. Oberg. 1986. PPi analogs as inhibitors ofhuman T-lymphotropic virus type III reverse transcriptase.Antimicrob. Agents Chemother. 29:867-872.

489. Wade, J. C., R. W. McGuffin, S. C. Springmeyer, B. Newton,J. W. Singer, and J. D. Meyers. 1983. Treatment of cytomeg-aloviral pneumonia with high-dose acyclovir and human leu-kocyte interferon. J. Infect. Dis. 148:557-562.

490. Wade, J. C., C. McLaren, and J. D. Meyers. 1983. Frequencyand significance of acyclovir-resistant herpes simplex virusisolated from marrow transplant patients receiving multiplecourses of treatment with acyclovir. J. Infect. Dis. 148:1077-1082.

491. Wade, J. C., and J. D. Meyers. 1983. Neurologic symptomsassociated with parenteral acyclovir treatment after marrowtransplantation. Ann. Intern. Med. 98:921-925.

492. Wade, J. C., B. Newton, N. Flournoy, and J. D. Meyers. 1984.Oral acyclovir for prevention of herpes simplex virus reactiva-tion after marrow transplantation. Ann. Intern. Med. 100:823-828.

493. Wahren, B., and B. Oberg. 1980. Reversible inhibition ofcytomegalovirus replication by phosphonoformate. Intervirol-ogy 14:7-15.

494. Wald, E. R., D. Dashefsky, and M. Green. 1988. In re ribavirin:a case of premature adjudication? J. Pediatr. 112:154-158.

495. Walmsley, S. L., E. Chew, S. E. Read, H. Vellend, I. Salit, A.Rachlis, and M. M. Fanning. 1988. Treatment of cytomegalo-virus retinitis with trisodium phosphonoformate hexahydrate.J. Infect. Dis. 137:569-572.

496. Waqar, M., M. J. Evans, K. F. Manly, R. C. Hughes, and J. A.Huberman. 1984. Effects of 2',3'-dideoxynucleosides on mam-malian cells and viruses. J. Cell Physiol. 121:402-408.

497. Watts, D. H., Z. A. Brown, T. Tartaglione, S. K. Burchett, K.Opheim, R. Coombs, and L. Corey. 1991. Pharmacokineticdisposition of zidovudine during pregnancy. J. Infect. Dis.163:226-232.

498. Weis, W., J. H. Brown, S. Cusack, J. C. Paulson, J. J. Skehel,and D. C. Wiley. 1988. Structure of the influenza virus haemag-glutinin complexed with its receptor, sialic acid. Nature (Lon-don) 333:426-431.

499. Weisbart, R. H., J. C. Gasson, and D. W. Golde. 1989.Colony-stimulating factors and host defense. Ann. Intern.Med. 110:297-303.

500. Wendel, H. A. 1964. Clinical and serologic effects in influenza

VOL. 5, 1992

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from

CLIN. MICROBIOL. REV.

of 1-adamantanamine HCL-a double-blind study. Fed. Proc.23:387.

501. Whitley, R., A. Arvin, C. Prober, S. Burchett, L. Corey, D.Powell, S. Plotkin, S. Starr, C. Alford, J. Connor, R. Jacobs, A.Nahmias, S. Soong, and the National Institute of Allergy andInfectious Diseases Collaborative Antiviral Study Group. 1991.A controlled trial comparing vidarabine with acyclovir inneonatal herpes simplex virus infection. N. Engl. J. Med.324:444-449.

502. Whitley, R., M. Hilty, R. Haynes, Y. Bryson, J. D. Connor,S.-J. Soong, C. A. Alford, and the National Institute of Allergyand Infectious Diseases Collaborative Antiviral Study Group.1982. Vidarabine therapy of varicella in immunosuppressedpatients. J. Pediatr. 101:125-131.

503. Whitley, R. J., C. A. Alford, M. S. Hirsch, R. T. Schooley, J. P.Luby, F. Y. Aoki, D. Hanley, A. J. Nahmias, S. Soong, and theNIA1D Collaborative Antiviral Study Group. 1986. Vidarabineversus acyclovir therapy in herpes simplex encephalitis. N.Engl. J. Med. 314:144-149.

504. Whitley, R. J., L. T. Ch'ien, R. Dolin, G. J. Galasso, and C. A.Alford, Editors, and the Collaborative Study Group. 1976.Adenine arabinoside therapy of herpes zoster in the immuno-suppressed. N. Engl. J. Med. 294:1193-1199.

505. Whitley, R. J., M. Levin, N. Barton, B. J. Hershey, G. Davisd,R. E. Keeney, J. Whelchel, A. G. Diethelm, P. Kartus, and S. J.Soong. 1984. Infections caused by herpes simplex virus in theimmunocompromised host: natural history and topical acyclo-vir therapy. J. Infect. Dis. 150:323-329.

506. Whitley, R. J., A. J. Nahmias, S. J. Soong, G. G. Galasso, C. L.Fleming, and C. A. Alford. 1980. Vidarabine therapy of neo-natal herpes simplex virus infection. Pediatrics 66:495-501.

507. Whitley, R. J., S. J. Soong, R. Dolin, R. Betts, C. Linnemann,Jr., C. A. Alford, Jr., and the NIAID Collaborative AntiviralStudy Group. 1982. Early vidarabine therapy to control thecomplications of herpes zoster in immunosuppressed patients.N. Engl. J. Med. 307:971-975.

508. Whitley, R. J., S. J. Soong, R. Dolin, G. J. Galasso, L. T.Ch'ien, C. A. Alford, and the Collaborative Study Group. 1977.Adenine arabinoside therapy of biopsy-proved herpes simplexencephalitis. N. Engl. J. Med. 297:289-294.

509. Whitley, R. J., S. J. Soong, M. S. Hirsch, A. W. Karchmer, R.Dolin, G. Galasso, J. K. Dunnick, C. A. Alford, and the NLAIDCollaborative Antiviral Study Group. 1981. Herpes simplexencephalitis. N. Engl. J. Med. 304:313-318.

510. Whitley, R. J., S. Spruance, F. G. Hayden, J. Overall, C. A.Alford, Jr., J. M. Gwaltney, Jr., S. J. Soong, and the NIAIDCollaborative Antiviral Study Group. 1984. Vidarabine therapyfor mucocutaneous herpes simplex virus infections in theimmunocompromised host. J. Infect. Dis. 149:1-8.

511. Whitley, R. J., A. Yeager, P. Kartus, Y. Bryson, J. D. Conner,C. A. Alford, A. Nahmias, and S.-J. Soong. 1983. Neonatalherpes simplex virus infection: follow-up evaluation of vidar-abine therapy. Pediatrics 72:778-785.

512. Wills, R. J. 1990. Clinical pharmacokinetics of interferons.Clin. Pharmacokinet. 19:390-399.

513. Wilson, S. Z., B. E. Gilbert, J. M. Quarles, V. Knight, H. W.McClung, R. V. Moore, and R. B. Couch. 1984. Treatment ofinfluenza A (H1N1) virus infection with ribavirin aerosol.Antimicrob. Agents Chemother. 26:200-203.

514. Wingfield, W. L., D. Pollack, and R. R. Grunert. 1969. Ther-apeutic efficacy of amantadine HCI and rimantadine HCI innaturally occurring influenza A2 respiratory illness in man. N.Engl. J. Med. 281:579-584.

515. Winston, D. J., L. J. Eron, M. Ho, G. Pazin, H. Kessler, J. C.Pottage, J. Gallagher, G. Sartiano, W. G. Ho, R. E. Champlin,L. Bernhardt, J. Bigley, L. Kanitra, P. I. Nadler, and theHoffmann-La Roche Herpes Zoster Study Group. 1988. Recom-binant interferon alpha-2a for treatment of herpes zoster inimmunosuppressed patients with cancer. Am. J. Med. 85:147-151.

516. Winston, D. J., W. G. Ho, C. H. Lin, K. Bartoni, M. D.Budinger, R. P. Gale, and R. E. Champlin. 1987. Intravenousimmune globulin for prevention of cytomegalovirus infectionand interstitial pneumonia after bone marrow transplantation.Ann. Intern. Med. 106:12-18.

517. Witkowski, J. T., R. K. Robins, R. W. Sidwell, and L. N.Simon. 1972. Design, synthesis and broad spectrum antiviralactivity of 1-fi-D-ribofuranosyl-1,2,4-triazole-3-carboxamideand related nucleosides. J. Med. Chem. 15:1150-1154.

518. Woodson, R. D., and J. J. Clinton. 1976. Hepatitis prophylaxisabroad. JAMA 209:1053-1059.

519. World Health Organization. 1989. Report of a WHO informalconsultation on preclinical and clinical aspects of the use ofimmunomodulators in HIV infection. AIDS 4:WHO 1-WHO14.

520. Worral, G. 1991. Topical acyclovir for recurrent herpes labialisin primary care. Can. Fam. Physician 37:92-98.

521. Wray, S. K., B. E. Gilbert, and V. Knight. 1985. Effect ofribavirin triphoshate on primer generation and elongation dur-ing influenza virus transcription in vitro. Antiviral Res. 5:39-48.

522. Wray, S. K., B. E. Gilbert, M. W. Noall, and V. Knight. 1985.Mode of action of ribavirin: effect of nucleotide pool alter-ations on influenza virus ribonucleoprotein synthesis. AntiviralRes. 5:29-37.

523. Yarchoan, R., P. Brouwers, A. R. Spitzer, J. Grafman, B.Safai, C. F. Perno, S. M. Larson, G. Berg, M. A. Fischl, A.Wichman, R. V. Thomas, A. Brunetti, P. J. Schmidt, C. E.Myers, and S. Broder. 1987. Response of human-immunodefi-ciency-virus-associated neurological disease to 3'-azido-3'-deoxythymidine. Lancet i:132-135.

524. Yarchoan, R., H. Mitsuya, C. E. Myers, and S. Broder. 1989.Clinical pharmacology of 3'-azido-2',3'-dideoxythymidine (zi-dovudine) and related dideoxynucleosides. N. Engl. J. Med.321:726-738.

525. Yarchoan, R., J. M. Pluda, R. V. Thomas, H. Mitsuya, P.Brouwers, K. M. Wyvill, N. Hartman, D. G. Johns, and S.Broder. 1990. Long-term toxicity/activity profile of 2',3'-dideoxyinosine in AIDS or AIDS-related complex. Lancet336:526-529.

526. Yarchoan, R., R. V. Thomas, J. Allain, N. McAtee, R. Dubin-sky, H. Mitsuya, T. J. Lawley, B. Safai, C. E. Myers, C. F.Perno, R. W. Klecker, R. J. Willis, M. A. Fischl, M. C.McNeely, J. M. Pluda, M. Leuther, J. Collins, and S. Broder.1988. Phase I studies of 2',3'-dideoxycytidine in severe humanimmunodeficiency virus infection as a single agent and alter-nating with zidovudine (AZT). Lancet i:76-81.

527. Yarchoan, R., R. V. Thomas, J. Grafman, A. Wichman, M.Dalakas, N. McAtee, G. Berg, M. Fischl, C. F. Perno, R. W.Klecker, A. Buchbinder, S. Tay, S. M. Larson, C. E. Myers,and S. Broder. 1988. Long-term administration of 3-azido-2',3'-dideoxythymidine to patients with AIDS-related neuro-logical disease. Ann. Neurol. 23(Suppl.):S82-S87.

528. Younkin, S. W., R. F. Betts, F. K. Roth, and R. G. Douglas, Jr.1983. Reduction in fever and symptoms in young adults withinfluenza A/Brazil/78 HlNl infection after treatment withaspirin or amantadine. Antimicrob. Agents Chemother. 23:577-582.

529. Zamecnik, P. C., and M. L. Stephenson. 1978. Inhibition ofRous sarcoma virus replication and cell transformation by aspecific oligodeoxynucleotide. Proc. Natl. Acad. Sci. USA75:280-284.

530. Zebedee, S. L., and R. A. Lamb. 1988. Influenza A virus M2protein: monoclonal antibody restriction of virus growth anddetection of M2 in virions. J. Virol. 62:2762-2772.

531. Zhu, Z., R. F. Schinazi, C. Chu, G. J. Williams, C. B. Colby,and J. Sommadossi. 1990. Cellular metabolism of 3'-azido-2',3'-dideoxyuridine with formation of 5'-O-diphosphohexosederivatives by previously unrecognized metabolic pathwaysfor 2'-deoxyuridine analogs. Mol. Pharmacol. 38:929-938.

182 BEAN

on May 24, 2020 by guest

http://cmr.asm

.org/D

ownloaded from