Antiviral Therapy: Current Concepts and Practices

Size: px
Start display at page:

Download "Antiviral Therapy: Current Concepts and Practices"

Transcription

1 CLINICAL MICROBIOLOGY REVIEWS, Apr. 1992, p Vol. 5, No /92/ $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 INTRODUCTION CELLULAR AND VIRAL REPLICATION ANTIVIRAL AGENTS Amantadine and Rimantadine Ribavirin Vidarabine Acyclovir Ganciclovir Foscarnet Zidovudine Didanosine Investigational Antiretroviral Agents IMMUNOGLOBULINS IMMUNOMODULATORS PROBLEMS OF ANTWIRAL THERAPY Resistance Latency Immunosuppression by Antiviral Agents PROSPECTS FOR THE FUTURE Liposomes Combination Therapy Computer-Aided Drug Design Role of the Clinical Microbiology Laboratory CONCLUSIONS ACKNOWLEDGMENTS REFERENCES 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 administered to patients in the 1960s, when Bauer prevented disease by giving thiosemicarbazone (methisazone) to patients exposed 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 development 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 146 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 trifluorothymidine (5-trifluoromethyl-2'-deoxyuridine) (Fig. 1). Idoxuridine 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 systemically to patients, however, it did not decrease the risk of death from herpes encephalitis and it caused myelosuppression 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

2 VOL. 5, 1992 ANTIVIRAL THERAPY 147 OH IDOXURIDINE TRIFLUOROTHYMIDINE VIl FIG. 1. Some nucleoside analogs first used as antiviral agents. that they demonstrated the feasibility of antiviral therapy in patients. They also sustained interest in a continuing search for more selective agents. By the late 1970s, it was known that most human pathogenic viruses possess enzymes coded by the viruses themselves and not present in uninfected cells (327). Most of these enzymes are involved in viral nucleic acid synthesis, as discussed below. Their discovery represented a major advance in antiviral therapy by making it possible to direct efforts at finding specific inhibitors of these enzymes rather than nonspecific inhibitors of viral growth in cell culture. Replication of viruses can be divided into several steps: (i) attachment to the cell, (ii) penetration, (iii) uncoating of nucleic acid, (iv) transcription and translation of early (regulatory) proteins, (v) nucleic acid synthesis, (vi) synthesis of late (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, assembly, and release closely resemble normal cellular processes and are thought to be carried out, in many instances, by cellular enzymes (210). It is at the point of nucleic acid synthesis that viral processes diverge most from their cellular counterparts and are most likely to require virus-specified enzymes. This is because eukaryotic cells contain doublestranded DNA and, when they replicate, make new DNA from the parental DNA template in the cell nucleus (Fig. 3). In addition, they transcribe mrna from DNA and transport it into the cytoplasm, where proteins are translated. Viruses, on the other hand, may contain DNA or RNA as their genomic material, and the nucleic acid may be double or single stranded, circular or linear, and segmented or in one continuous piece. RNA may be either positive or negative stranded (having the same sense or direction as mrna, and thus directly translatable, or having the opposite sense and not directly translatable). Viruses may replicate in the nucleus, in the cytoplasm, or in both. Despite these differences from their cellular hosts, viruses must be able to fit into host cell synthetic pathways if they are to replicate successfully. They must present to the cell either a form of DNA that can be transcribed directly into mrna or a form of mrna that the cell can recognize and translate into proteins (390). Many different pathways have evolved by which viruses accomplish this; some examples are shown in Fig. 3. Retroviruses, for example, carry single-stranded duplex RNA as their genomic material and, when infecting cells, must supply a virus-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 enteroviruses carry a positive single-stranded RNA that can be directly translated by host cell enzymes, but they must encode an RNA replicase which allows use of the genomic RNA as a template for new negative-stranded RNA. Orthomyxoviruses (influenza viruses), on the other hand, carry a negative-stranded RNA, and thus must supply an RNAdependent RNA polymerase from which a plus-stranded mrna can be made. Such unique enzymes, critical to viral replication but unnecessary for cellular function, offer very good targets for selective inhibition by antiviral agents. They are not the only enzyme targets, however. Other enzymes such as nucleoside kinases and DNA polymerases are encoded by both cells and viruses (Fig. 4). The properties of such enzymes can differ greatly with respect to substrate specificity, binding affinity for substrates, and susceptibility to inhibition by various compounds. These differences can be exploited in developing antiviral compounds. For example, thymidine kinases catalyze the first step in the preparation of pyrimidine nucleosides for incorporation into DNA, phosphorylation of the 5' carbon atom of the pentose ring (Fig. 4). Thymidine kinase specified by HSV binds acyclovir much better than cellular thymidine kinase does and phosphorylates it 3 million times faster (235). DNA polymerases are responsible for incorporating nucleotide triphosphates into growing DNA chains, and again, herpesvirus-specified DNA polymerases differ from their cellular counterparts: they are 30 times more susceptible to inhibition by acyclovir triphosphate than are the alpha DNA polymerases of human origin (122). Differences like these make it possible to identify compounds that selectively inhibit viral functions. What advantage is there to viruses in encoding and carrying enzymes also made by cells? The answer is survival. Viruses carrying the extra enzymes have a wider host range and can infect cells that do not encode a critical enzyme or that express it only at a certain point in the cell cycle. HSV mutants that do not encode thymidine kinase, for example, are less capable of establishing reactivatable latent infections in neurons (which are nondividing cells) than are thymidine kinase-encoding strains (121, 142, 471). Many antiviral agents are nucleoside analogs, structures that closely resemble the natural nucleosides used as building blocks for DNA synthesis (Fig. 1 and 5). As such, these drugs are phosphorylated by cellular nucleoside kinases and incorporated into growing DNA chains by DNA polymer-

3 So nu (a *a 0 _ z O Ck, 0. oc Ji IL A ni I *X la _ ~%> r%o V %, o z z IC z a a~~~~._ 0. IL O 0 CZ CZ u E 0 C Downloaded from icx z 1- z o z z o ~~~~o X I I <~~~~~C on October 22, 2018 by guest At OM44, v ba 0 pq a *0 0 21, z 4d i bo a CL 0 CL 0 00 In 148

4 VOL. 5, 1992 ANTIVIRAL THERAPY 149 RETROVIRUSES as du RNA ORTHOMYXOVIRUSES as RNA (-) cdna * REOVIRUSES ds RNA ; % // * PICORNAVIRUSES a / t ss RNA(+) *J crdsia mrna 4 proteins" DNA supercolled HEPATITIS B VIRUS FIG. 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 substrates for both enzymes (Fig. 4). Many of the early antiviral agents, such as idoxuridine and trifluorothymidine, were equally good substrates for both viral and cellular kinases. Even though they were slightly better inhibitors of viral than of cellular polymerases (194, 365), they were activated and capable of decreasing DNA synthesis in both infected and uninfected cells. Many of the newer nucleosides are less toxic, in part because there is at least one point in their functional pathways which is specific for virus-infected cells. For example, acyclovir is activated only in virus-infected cells, and zidovudine inhibits an enzyme, human immunodeficiency virus (HIV) reverse transcriptase, not found in normal cells. Although the greatest success has been achieved by using inhibitors of nucleic acid synthesis as antiviral agents, additional enzymes are necessary for other steps in the viral replication cycle and should be amenable to inhibition by putative antiviral compounds. Special interest in such compounds has been generated by the AIDS epidemic and the increasingly obvious need to treat HIV infection with combinations of drugs active at different sites of viral replication. Such combination therapy, it is hoped, will decrease drug toxicity and reduce the chances of antiviral resistance developing during treatment. Unfortunately, steps such as viral attachment and penetration and assembly and release are catalyzed by cellular enzymes (210), and it has been difficult to find compounds that interfere specifically with these events. Nevertheless, some success has been achieved with amantadine (an inhibitor of uncoating) for management of influenza, and recombinant soluble CD4 may yet prove effective at inhibiting attachment of HIV to host lymphocytes. Interferons, which inhibit viral mrna transcription and protein synthesis, are also useful as antiviral agents. In addition, HIV proteases, which cleave precursor polypeptides to form functional reverse transcriptases, are very promising targets for selective inhibition. ANTIVIRAL AGENTS Amantadine and Rimantadine The adamantanes, amantadine (1-aminoadamantane hydrochloride) and its alpha-methyl derivative, rimantadine, are used for management of influenza A virus infections. They are cyclic amines with bulky, cagelike structures unlike those of other known antiviral agents (Fig. 6). Their mechanisms of action and spectra of antiviral activity are identical, but rimantadine is metabolized differently from amantadine, and it causes fewer central nervous system side effects such as insomnia and difficulty concentrating (110, 514). It is also a more potent in vitro inhibitor of influenza A viruses (431), although both drugs have been equally effective in treating patients. Amantadine became available in 1966; rimantadine is being considered for approval by the U.S. Food and Drug Administration. It has been known for some time that amantadine inhibits an early phase of viral replication (207, 232), more specifically, virus uncoating (48, 381). Recent studies of avian influenza viruses have also demonstrated a block at a later stage, virus maturation and assembly (184, 461). Susceptibility to amantadine is determined principally by the M2 protein (184, 281), a virus-specified matrix protein present on the surface of infected cells and in the virion in small amounts (530). It has been proposed (28) that this protein forms ion channels through which protons pass across the membranes of intracellular endocytic and exocytic vesicles. During virus uncoating, protons are transferred from the endocytic vesicle to the virion, allowing the fall in ph that releases free viral nucleoprotein into the cell cytoplasm. During virus assembly, protons are transferred out of the exocytic vesicle, thus maintaining the ph above the level at which the viral hemagglutinin, a major surface glycoprotein, would lose its structural integrity and fail to be incorporated into the viral envelope. Amantadine appears to block this M2-mediated transfer of protons and thus inhibits viral

5 150 BEAN CLIN. MICROBIOL. REV. Io L a) -o -o ac -o z CZ 0._ 0 0 *0 a)._ U U 0 Zz o ca *0 _ c.o.>.0o 0._ E Ai

6 VOL. 5, 1992 ANTIVIRAL THERAPY 151 5H ACYCLOVIR GUANOSINE RIBAVIRIN H2N' OH N3 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 the strain of virus (185). At in vitro concentrations much higher than those at which these effects occur, adamantanes inhibit other RNA viruses, including influenza B virus, and paramyxoviruses (114). With current drug formulations, these high concentrations cannot be achieved in patients, and thus the adamantanes remain useful only for the management of influenza A virus infections (Table 1). Both drugs are very effective in preventing illness due to influenza A virus. When given prophylactically during a community outbreak, either compound reduces the risk of acquiring influenzal illness by 50 to 90% ( 68, 110, 161, 317, 331, 500). Rimantadine has fewer side effects when used in NH2 HCI NH2 *HCI H-C-CH3 this manner, although lowering the dose of amantadine also reduces adverse effects (378, 419). The efficacy of low-dose amantadine has not been proven in this situation, however, and the higher dose remains the prophylactic regimen of choice until rimantadine becomes available. Both drugs are more effective in preventing illness than in preventing infection with influenza A virus (110, 317). This may be beneficial, however, in that it allows the patient to produce protective antibodies without developing frank illness. Currently, the Immunization Practices Advisory Committee recommends using amantadine prophylactically to protect those at risk of influenza complications who cannot or have not been vaccinated (60). The drug can also be given at the same time as [ 0-0-p-cl gi Na AMANTADINE RIMANTADINE FOSCARNET FIG. 6. Nonnucleoside antiviral agents.

7 152 BEAN 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 Recurrent genital herpes 200 mg 5 times daily for 5 days Suppression, genital herpes 400 mg twice daily for up to 1 yr Suppression, mucocutaneous herpes 200 mg 3 to 5 times daily in immunocompromised host Treatment, mucocutaneous herpes in mg 5 times daily until healed immunocompromised host Zoster in immunocompetent host 800 mg 5 times daily for 7-10 days Ganciclovir Zidovudine Vidarabine Amantadine Ribavirin Interferon Foscarnet Didanosine Intravenous Topical Intravenous Oral Intravenous Ophthalmic ointment Oral Aerosol Intravenous Subcutaneous Intralesional Intravenous Oral Herpes encephalitis Neonatal herpesa Severe genital herpes Treatment, mucocutaneous herpes, immunocompromised host Suppression, mucocutaneous herpes in immunocompromised hosta Zoster or varicella in immunocompromised host Initial genital herpes Cutaneous herpes in immunocompromised host CMV retinitis in AIDS HIV infection, CD4 cells <500/mm3, or symptomatic Herpes encephalitis Neonatal herpes Zoster in immunocompromised host Herpes keratitis Influenza A: treatment Influenza A: prophylaxis Severe RSV, infants and children Lassa fever' Hepatitis B-chronic active liver disease' Hepatitis C-chronic liver disease Condyloma acuminata (warts) CMV retinitis Acyclovir-resistant HSV or VZVa Zidovudine intolerance or treatment failure in HIV-infected host 10 mg/kg 3 times daily for days 500 mg/mth 3 times daily for 10 days 5 mg/kg 3 times daily for 5 days 5 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 days Apply every 3 h up to 6 times daily for 7 days Induction: 5 mg/kg twice daily for days Maintenance: 5 mg/kg daily 100 mg 5 times daily 15 mg/kg daily over h for 10 days 15 mg/kg daily over h for 10 days 15 mg/kg daily over h for 5 days 0.5 in. [1.27 cm] to eye 5 times daily for 7-21 days 200 mg daily for 5-7 days 200 mg daily h daily for 3-7 days (concn, 20 mg/ml) 2-g load, then g 3-4 times daily for 10 days 5 x 106 U daily for 4 mo 2 x x 106 U 3 times weekly for 6 mo 1 x 106 U per wart in up to 5 warts 3 times weekly Induction: 60 mg/kg 3 times daily for 2-3 wk Maintenance: mg/kg daily mg/kg 3 times daily Tablet: mg (as 2 tablets) 2 times daily Powder: mg 2 times daily Trifluridine Ophthalmic solution Herpes keratitis 1 drop every 2 h up to 9 drops daily until healed Idoxuridine Ophthalmic solution a The package insert is not approved for this indication. b Square meters of body surface area. Herpes keratitis 1 drop/h during day and every 2 h at night until healed

8 VOL. 5, 1992 influenza virus vaccine to protect the patient until an antibody response to the vaccine occurs. Both amantadine and rimantadine have been used for the treatment of influenza. They are most effective when given within the first 48 h of illness and may allow the patient to return to routine daily activities 1 to 2 days earlier than no treatment would (191, 475, 483, 514). Amantadine also appears superior to aspirin in this regard. In one comparative study, patients taking 3.25 g of aspirin daily became afebrile sooner but had more side effects (insomnia, nausea, ringing in the ears) than amantadine recipients, who experienced an earlier reduction in symptoms and fewer adverse reactions (528). The peripheral pulmonary airway dysfunction accompanying influenza is also improved by amantadine (279), and this may be important for earlier resolution in normal hosts and for minimizing disease in those with underlying pulmonary or cardiac conditions. The effects of therapy on the complications of influenza, such as pneumonia and myocarditis, are unknown. Although drug resistance does not occur naturally among influenza viruses (27), it can be induced by therapy, and these resistant viruses can cause influenzal illness. When patients ill with influenza were treated with rimantadine, they improved symptomatically, but 50% of them began shedding rimantadine-resistant viruses within 4 to 6 days (177, 188). In one study, these resistant viruses were apparently transmitted to family members, who also became ill with influenza (188). Resistance was mapped to the M2 protein (29), and subsequent studies of similarly resistant avian viruses have shown that these resistant strains are genetically stable and equal in virulence to wild-type viruses (25). These findings raise the possibility that widespread use of adamantanes for treatment of influenza might result in antiviral pressure and selection of predominantly resistant virus populations. To prevent the spread of resistant virus, it has been suggested that patients be isolated or have limited contact with others while they are being treated for influenza (186). The effects of this and other methods to control transmission are unknown, however. In view of these problems and of the unknown effect of therapy in preventing or relieving complications, no recommendation for therapy of influenza is currently made by the Centers for Disease Control (60), although amantadine is approved for this purpose (package insert). Ribavirin Ribavirin (1-beta-D-ribofuranosyl-1,2,4-triazole-3-carboxamide) was first synthesized in the early 1970s as part of an intensive effort to identify new antiviral agents (517). At first glance, it appears to be a nucleoside analog with an open pyrimidine ring (Fig. 5). Structurally (362) and functionally (458), however, it most closely resembles guanosine. Ribavirin is active in vitro against a wide variety of RNA and DNA viruses, including adenoviruses, herpesviruses, influenza 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 to some extent in cells infected by different viruses. This may also account for the wide spectrum of activity. Ribavirin readily diffuses into eukaryotic cells, where it is converted by cellular enzymes to the mono-, di-, and triphosphate forms (437, 458). The monophosphate is a potent competitive inhibitor of IMP dehydrogenase, an enzyme essential for ANTIVIRAL THERAPY 153 the synthesis of GTP. This inhibition results in a decrease in the cellular pools of guanine nucleotide necessary for both cellular and viral replication (458, 459). Ribavirin inhibits capping of viral mrna (170, 493), a critical step in the replication of most viruses. Its most important effect, however, appears to be on the early events of viral replication. Ribavirin directly inhibits viral RNA-dependent RNA polymerase of influenza viruses (129), and it can retard both initiation (54, 338, 367, 521) and elongation (129, 521) of mrna transcripts. It is not incorporated into the growing chain of viral nucleic acid and does not cause chain termination (129, 477). Only one strain of virus, a mutant of fowl plague virus, is known to be resistant to ribavirin (215). In addition to its antiviral properties, ribavirin has immunoregulatory effects. In cell culture and in animals, it inhibits macromolecular synthesis and cell division (270), lymphocyte proliferation, and nucleic acid synthesis (341). It also suppresses B lymphocytes and subsequent antibody production (358) and tumor growth (357). To date, however, none of these effects has been shown to be of consequence in patients treated with ribavirin. The major toxicities of ribavirin are anemia and embryotoxicity. Anemia occurs when ribavirin diffuses into erythrocytes and accumulates, because erythrocytes lack phosphatases and are unable to hydrolyze (dephosphorylate) the drug. The erythrocytes then become damaged as they age and are removed prematurely from the circulation (52, 334). At very high doses of ribavirin in animals, bone marrow suppression of erythroid precursors also occurs (52). All of these effects are reversible upon removal of the drug, but the half-life of ribavirin in human erythrocytes is 40 days, so the effect is prolonged. In pregnant rodents treated with ribavirin, skeletal defects in the developing embryo as well as fetal resorption have been observed (242). Because of this, the drug is contraindicated during pregnancy, and safety measures must be taken by pregnant health care workers who administer ribavirin to patients (see below). Ribavirin became commercially available for use as an aerosol in Both the oral and intravenous forms have been studied, but the drug was found to be most effective when given to animals by pulmonary aerosol, so it was developed for that purpose. The drug is diluted in a reservoir, nebulized by a small-particle aerosol generator, and delivered to the patient via face mask, ventilator, or infant oxygen hood. Aerosolized drug can leak from the system during administration and may be inhaled by health care workers. This may represent a hazard to pregnant workers, although it is not known whether the small amounts absorbed into the bloodstream will damage the fetus (57). In college students with uncomplicated influenza, aerosol ribavirin reduced the duration of fever and symptoms and decreased viral shedding (239, 291, 513). This has not been true of all trials and for all influenza variants, however (33, 167). Because of this inconsistency and because aerosol therapy is an expensive and cumbersome mode of therapy for a self-limited illness, ribavirin is not often used in this setting. In infants with bronchiolitis due to RSV, ribavirin has shown some effect in decreasing viral shedding, improving oxygenation, and reducing the duration of illness (178, 179, 388, 469), although questions have been raised about the methods used in treatment trials (494). In a recent trial, ribavirin decreased the duration of oxygen therapy, mechanical ventilation, and hospital stay in otherwise healthy infants with severe RSV illness (439). Nevertheless, important questions remain unanswered, including duration of therapy,

9 154 BEAN response of children with underlying cardiac and pulmonary disease, and efficacy of the drug when administered late in the course of illness (368). Ribavirin is extremely expensive ($400 per day in drug costs alone), and all children with self-limited RSV cannot be hospitalized and treated. Along with the possible toxicity for health care workers, these unanswered questions indicate the great need for additional studies to define the optimum use of this drug for respiratory viral disease. Because of its in vitro activity against bunyaviruses and arenaviruses, intravenous ribavirin has been investigated for treatment of viral hemorrhagic fevers (214). Most notable has been the success with Lassa fever. When patients at high risk of death from Lassa fever were treated intravenously within 6 days of fever onset, the death rate of 55 to 76% in placebo-treated patients decreased to 5 to 9% in ribavirin recipients (293). Oral ribavirin is also beneficial against Lassa fever and is given as prophylaxis to high-risk contacts of Lassa fever patients (58). Recent small-scale studies have shown that ribavirin crosses the blood-brain barrier well, giving drug levels in the cerebrospinal fluid that are 50 to 100% of those in serum (82, 330). For this reason, it may also be of use in the treatment of common viral encephalitides caused by bunyaviruses, such as La Crosse encephalitis (54, 214). Ribavirin has recently been investigated for possible use in HIV-infected patients. Although the drug given orally in high doses appeared to delay progression to AIDS (384), many questions have been raised about the study methodology (39). In addition, no effects on surrogate markers of HIV progression (CD4 counts, p24 antigenemia, viremia, or immune function) were found, and it is not clear that clinically achievable drug levels are inhibitory for viral replication (292, ). In view of these problems, licensure of ribavirin for treatment of HIV is no longer being pursued in the United States, although the drug is available in other countries. The place of ribavirin in our armamentarium of antiviral agents is not yet clear. Though useful in aerosol form, it is cumbersome, expensive, and perhaps toxic. Intravenous ribavirin is very useful for life-threatening illness, such as Lassa fever, but toxicity limits the use of this form in less severe diseases. Liposome-encapsulated ribavirin has been used successfully in animal models for treatment of Rift Valley fever and influenza (163, 236), and it may represent a way to achieve improved drug delivery to target organs while minimizing toxicity. Analogs of ribavirin have been synthesized and tested for antiviral activity. One compound, 5-ethynyl-1-beta-ribofuranosylimidazole-4-carboxamide (EICAR), was originally studied as an antileukemic agent but has now been shown to be 10 to 30 times as active as ribavirin against RSV and influenza viruses (98). Whether it will be clinically useful remains to be seen. CLIN. MICROBIOL. REV. Vidarabine Vidarabine (9-beta-D-arabinofuranosyladenine) was the first antiviral agent licensed for intravenous use in the United States, in It is an analog of the nucleoside adenosine (Fig. 1) and was originally synthesized and studied in the early 1960s as an antitumor agent. Subsequently, it was found to be active in vitro against HSV, varicella-zoster virus (VZV), cytomegalovirus (CMV), vaccinia virus, and some RNA tumor viruses. In animal models, it was active against HSV and vaccinia virus (409). The mechanism of action has not been completely delineated, but it involves phosphorylation of the nucleoside by cellular enzymes (44, 355) and incorporation into the growing chains of both cellular and viral DNAs, with resultant slowing of DNA synthesis (343). Vidarabine triphosphate is also a competitive inhibitor of DNA polymerase, both cellular and viral. It is more potent, however, against viral enzymes, and this potency may account for its selective inhibition of viral DNA synthesis (117, 417). Vidarabine inhibits other steps in nucleic acid synthesis, such as RNA polyadenylation (393) and terminal deoxynucleotidyl transferase (108), but whether or not these contribute to its antiviral activity is unknown. It also inhibits S-adenosylhomocysteine hydrolase (398), an enzyme necessary for methylation of trna and mrna and for other cellular transmethylation reactions. This inhibition results in cytotoxicity for mononuclear cells and in the possible accumulation of biogenic amines (117), both of which may contribute to the toxicity of this compound. Vidarabine became available as an eye ointment in 1977 and remains useful for the treatment of herpes keratitis (Table 1). Although it is also useful for the treatment of HSV and VZV in immunocompromised patients (502, 504, 507, 510) and for herpes encephalitis (508, 509) and neonatal herpes (506, 511), it has been almost entirely replaced by acyclovir for these diseases. Vidarabine is difficult to use. It is poorly soluble in water and rapidly deaminated in vivo to arahypoxanthine, a much less active compound. It must therefore be administered continuously by the intravenous route in large volumes of fluid. Toxicities, including nausea and vomiting, weakness, weight loss, megaloblastosis of bone marrow erythroid precursors, tremors, and myoclonus have been seen in a high proportion of treated patients (287, 394). Acyclovir is easier to administer, less toxic, and more active against herpesviruses. Although vidarabine does not require a virus-coded thymidine kinase for activation, and thymidine kinase-deficient HSV isolates are susceptible to the drug in vitro, vidarabine has not proven useful for treatment of acyclovir-resistant HSV in HIV-infected patients, and foscarnet is preferred (402). Acyclovir Acyclovir [9-(2-hydroxyethoxymethyl)guanine] has the best therapeutic index of available antiviral agents, and it is widely used for the treatment of herpesvirus infections. It was "discovered" in the mid-1970s by Elion and colleagues while they were screening for antiviral activity of nucleoside analogs (410). The compound is an analog of guanosine (Fig. 5), but instead of a complete ribose ring attached to the purine base, it has an acyclic side chain. Acyclovir diffuses freely into cells but is activated and accumulates only in cells infected with herpesviruses because the first step in activation, phosphorylation at the 5' position of the side chain, is catalyzed only by a virus-specified thymidine kinase and not to any appreciable extent by cellular kinases (Fig. 4) (122, 159). Further phosphorylations to the diphosphate and the active triphosphate forms are carried out by cellular enzymes (122, 312). During DNA replication, acyclovir triphosphate competes with the natural substrate, dgtp, for the viral DNA polymerase. It has a higher affinity for the enzyme, however, and is preferentially incorporated into the growing 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 replication is terminated (103, 156, 294). Furthermore, when acyclovir is bound to the DNA template in the presence of the next

10 VOL. 5, 1992 nucleotide, a tight irreversible complex is formed and the polymerase is completely inactivated, further slowing viral DNA synthesis (103, 158, 369). Taken together, these properties make acyclovir highly selective in inhibiting viral replication while having little effect on host cell function. There is, for instance, a 3,000-fold difference between the concentration of acyclovir needed to inhibit HSV type 1 (HSV-1) and that needed to inhibit the cells in which it is grown (122). The herpesviruses vary in their susceptibilities to acyclovir. HSV-1 and HSV-2 are exquisitely susceptible, requiring mean in vitro concentrations of 0.04 and 0.4,ug/ml, respectively, for 50% inhibition of viral replication (86, 329). The thymidine kinase of VZV does not phosphorylate acyclovir as effectively, and the virus is 8 to 10 times less susceptible to the drug than HSV (35, 86, 329). Although its DNA polymerase is very susceptible to acyclovir (452), CMV does not encode a thymidine kinase (133), and high drug levels (5 to 25,ug/ml) are required for inhibition (86, 353). It is not clear that Epstein-Barr virus (EBV) encodes a thymidine kinase capable of activating acyclovir. Its DNA polymerase is, however, very susceptible to the drug (91), and the 50% inhibitory concentrations are low (73, 278). Acyclovir has no effect on the latent phase of any of the herpesviruses. Acyclovir is available in three formulations: an ointment, pills or capsules, and an intravenous suspension. Systemic absorption and other side effects of the ointment are minimal, but it is not very effective (see below) and has largely been replaced by oral formulations. Levels achieved in serum after intravenous administration are well above the inhibitory concentrations of HSV, VZV, EBV, and some strains of CMV. With oral dosing, however, only 15 to 20% of the medication is absorbed, and levels in serum are not consistently above the inhibitory concentrations for any of the herpesviruses except HSV (100). Acyclovir is remarkably free of serious toxicity. When given intravenously in high doses, it can cause a transient rise in serum creatinine that is apparently due to obstructed renal tubules (22, 407). A reversible neurologic syndrome consisting of confusion, lethargy, and occasionally hallucinations and coma has also been described. This syndrome is usually seen in very ill, immunocompromised patients receiving high-dose intravenous therapy (137, 253, 491). With oral therapy, nausea, diarrhea, and headache have occasionally been reported. Acyclovir is not carcinogenic and is mitogenic or teratogenic only at very high levels in animals (479). It does cross the placenta, however, and therefore must 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 or intravenously, it shortens the course of initial genital herpes by one-third to one-half (46, 79, 314, 323). Recurrent genital herpes does not respond as dramatically, even when patients initiate therapy themselves at the first sign of disease (323, 374). This may be because recurrences normally last only a few days, and it is difficult to demonstrate a shortening of the course with drug therapy. Acyclovir is, however, very useful in preventing recurrent disease. When given daily in low doses to patients with frequent eruptions, it suppresses them almost completely and causes no side effects even with prolonged use (112, 230, 306, 313). It does not prevent asymptomatic viral shedding (457), and transmission has occurred when patients were free of genital lesions (392). Topical acyclovir, a 5% ointment, is useful, though not as effective as oral therapy, for treatment of initial genital herpes (79, 80, 472). In recurrent disease it offers no benefit ANTIVIRAL THERAPY 155 (282, 373). Topical therapy is occasionally used in pregnant patients who have mild genital herpes and should not receive systemic acyclovir. Oral treatment of herpes labialis (the common cold sore) shortens the course of illness in those patients who are prone to more severe eruptions and are able to initiate therapy themselves at the first sign of an eruption (451). When given prophylactically to patients exposed to the sun or to artificial UV light, oral acyclovir decreased the frequency of herpetic lesions appearing within the next 2 or 7 days, respectively (448, 449) but did not affect the lesions which appeared immediately (within 48 h) after the exposure (448). Topical therapy is of little benefit in otherwise healthy patients with cold sores (448, 520). In the treatment of herpes encephalitis, acyclovir has proven both more effective and less toxic than vidarabine (434, 503). It reduces morbidity to 14% (vidarabine reduces it to 38%), and it is now the treatment of choice for that disease. A recent trial of acyclovir versus vidarabine for the treatment of neonatal herpes showed neither drug to be superior (501). Given the sample size, however, as much as a 25% difference in morbidity and mortality could have been present and gone undetected. Acyclovir has also been very useful for management of mucocutaneous HSV in immunocompromised patients. Administered orally or intravenously, it decreases pain and viral shedding and accelerates healing (310, 425). Though not as effective, topical therapy can also be useful in this regard (505). When given suppressively to HSV-seropositive patients, acyclovir greatly reduces the chances of a recurrence during 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 administration are often below the 50% inhibitory dose for the virus. In immunocompromised patients with chickenpox or zoster, intravenous acyclovir has been very effective in decreasing dissemination and other complications (15, 326, 361, 424). It can also be given orally in the post-bone marrow transplantation period and appears to decrease the frequency of zoster during that time (346, 421). In otherwise healthy patients with zoster, intravenous acyclovir decreases acute pain and accelerates lesion healing (23, 350), but oral therapy has had only a modest effect against this disease (213, 296). Neither form of therapy reduces postherpetic neuralgia. Of importance, oral acyclovir decreases the frequency of eye complications in patients with ophthalmic zoster (69, 70). In one study of otherwise healthy children with chickenpox, oral acyclovir reduced the duration of disease 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 pneumonia, 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 CMV disease in immunocompromised patients. Two studies, one of bone marrow recipients (309) and one of renal allograft recipients (17), have suggested that high-dose intravenous acyclovir can reduce the frequency of CMV infection and disease when given suppressively in the peritransplantation period. The reasons for this are not clear, but may relate to the fact that only low numbers of viruses are present early after reactivation, and the small amount of active acyclovir present may be sufficient to limit viral replication enough to prevent clinical disease. Acyclovir therapy of established CMV infections in immunocompromised patients has not been successful (16, 489).

11 156 BEAN Treatment of diseases due to EBV has also been disappointing, despite the apparent susceptibility of the virus to acyclovir. The course and symptoms of mononucleosis are not affected by treatment, although viral shedding from the oropharynx is decreased (3, 481). On the other hand, acyclovir does appear to be effective for treating hairy leukoplakia, an EBV-associated condition characterized by painful tongue lesions in patients infected with HIV (377). Improvement is transient, however, and the lesions recur when the drug is stopped. Resistance of HSV to acyclovir has been extensively studied. It results from changes in the virus-specified thymidine kinase and/or the DNA polymerase and occurs by one of three mechanisms: a deletion or mutation conferring inability or reduced ability to express thymidine kinase, resulting in lack of acyclovir phosphorylation (a TK- or thymidine kinase-deficient mutant) (72, 413); expression of a DNA polymerase with reduced affinity for acyclovir triphosphate (72, 265, 413); or expression of a thymidine kinase with altered substrate binding properties for the drug (90, 154). Both of the last two mechanisms result in lack of recognition of acyclovir as a substrate for the enzyme. Among patient isolates, thymidine kinase-deficient mutants are most common, but viruses expressing the other mechanisms have also been found (49, 76, 123, 399, 426, 490). In nature, HSV populations consist of a mixture of TKand normal (TK+) virus variants (337). They are predominantly TK+, however, because these viruses have a selective advantage in being more capable of establishing latent, reactivatable infections in host sensory ganglia. They are also more neurovirulent (121, 142, 143, 471). Under pressure of antiviral drug therapy, a population can become predominantly 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 prolonged periods with acyclovir in doses that do not suppress viral replication completely but allow "breakthrough" replication to occur. To date, this has been seen almost entirely in immunocompromised patients, (19, 24, 131, 220, 335). Treatment consists of withdrawing acyclovir and, when necessary, administering a drug with a different mode of action, such as foscarnet (see below). The importance of resistance for the future of acyclovir therapy is not yet clear. Thymidine kinase-deficient mutants are generally not as hardy as their TK+ counterparts. This not true, however, of most thymidine kinase-altered and DNA polymerase-altered viruses (90, 141, 142, 265). Whether resistant virus populations will be able to establish themselves and compete with wild-type viruses remains to be seen. Because of acyclovir's poor oral availability and lack of effect against some herpesviruses, other drugs are being sought for treatment of herpesvirus infections. Famciclovir is the diacetyl ester of penciclovir, an acyclic nucleoside analog with a long intracellular half-life (484) and good activity against HSV and VZV but poor oral absorption (485). Famciclovir is well absorbed and rapidly converted to penciclovir (485) and is very promising for treatment of herpesvirus infections. 1-Beta-D-arabinofurasonyl-E-5-(2- bromovinyl)uracil, or BV-ara-U, is a thymidine analog which is 1,000 times more active against VZV than acyclovir (283). Cellular toxicity is seen at concentrations 1 millionfold higher than those needed to inhibit viral replication, making this drug highly selective and very promising for treatment of VZV infections. It is currently in patient trials. Ganciclovir CLIN. MICROBIOL. REV. Ganciclovir, 9-(1,3-dihydroxy-2-propoxymethyl)guanine, is a nucleoside analog similar to acyclovir except that it contains a carbon with attached hydroxyl group at the 3' position of the ribose ring (Fig. 5). It is active against all the human herpesviruses (139) and is up to 100 times more active than acyclovir against human CMV (436, 474). It is also more toxic than acyclovir and for this reason has been used only for treatment of serious CMV disease in immunocompromised patients. It is not clear why ganciclovir is so much more effective than acyclovir against CMV. In HSV-infected cells, ganciclovir, like acyclovir, is activated by a virus-specified thymidine kinase (37, 139, 436). In CMV-infected cells, in which no viral thymidine kinase is induced, ganciclovir is also phosphorylated and accumulates to much higher levels than acyclovir (37, 151). It is removed from these cells very slowly, persisting in stable form for several days (37). The molecular mechanisms of activation, accumulation, and degradation are not known but probably account to some extent for the difference in activity, as ganciclovir is actually a less potent inhibitor of viral DNA polymerase than acyclovir is (151, 286). Ganciclovir triphosphate is incorporated into the growing viral DNA chain and markedly slows DNA synthesis (285). Because the 3' hydroxyl is present on the molecule, the next incoming nucleotide can be bound to the nascent DNA chain, and ganciclovir is thus not a chain terminator (65, 285). Despite the fact that ganciclovir, like acyclovir, is a more potent inhibitor of viral than cellular polymerases (151, 436), it is more toxic than acyclovir. In vitro inhibition of the myeloid elements of bone marrow occurs at levels in serum seen with intravenous administration of ganciclovir (443, 444). The drug is also mitogenic in mammalian cells and carcinogenic and embryotoxic in animals (467). In humans, ganciclovir causes a reversible bone marrow toxicity in approximately 25% of patients, a factor that frequently limits its use (62, 208, 422). There have been no placebo-controlled trials to evaluate the efficacy of ganciclovir. Historically controlled trials have shown the drug to be generally useful in the management of CMV disease in immunocompromised patients, although efficacy varies among different patient groups. AIDS patients with CMV retinitis respond well to intravenous therapy, with 80% or more achieving stabilization or improvement in their vision (47, 74, 138, 197, 208, 217, 271). Almost all relapse, however, unless maintenance therapy is instituted, and even then many patients experience breakthrough disease or cannot tolerate the medication. These problems and the difficulties of long-term intravenous therapy have prompted the investigation of other modes of treatment. Intravitreal administration of ganciclovir avoids myelosuppression and has stabilized vision in some patients (195, 198). Oral therapy is also being investigated. While the oral bioavailability of ganciclovir is very poor (6 to 8% [96]), levels in serum may be high enough to inhibit some strains of CMV (222). AIDS patients with CMV pneumonia may have a response rate as high as 60 to 80% when given intravenous ganciclovir (47, 271), although strict criteria for diagnosis were 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 viral replication and shedding by the drug (47, 83, 422). One hypothesis for this difference is that, in bone marrow recipients, CMV pneumonia is an immune-mediated disease resulting from T-cell cytotoxic responses to viral antigens

12 VOL. 5, 1992 expressed on the surface of infected lung cells, whereas in AIDS patients, whose cellular cytotoxic responses are poor, the disease is due to direct damage to the lungs (174). To neutralize viral antigens and prevent recognition by immune effector cells, high-titered CMV immunoglobulin has been administered with ganciclovir in bone marrow transplant patients. Such combined antiviral and immunomodulator therapy has increased the survival rate to 50 to 70% (125, 371). In addition, it may permit a decrease in ganciclovir dosage, with an attendant decrease in bone marrow toxicity. In one recent trial, suppressive ganciclovir therapy was very successful in preventing clinical disease in bone marrow recipients who had CMV in their respiratory secretions but had not yet developed pneumonia (411). An additional placebo-controlled trial has confirmed and extended these findings; early ganciclovir therapy in bone marrow recipients excreting CMV but not yet ill reduced the frequency of CMV disease by 93% (from 15 of 35 to 1 of 37 patients) and significantly improved patient survival at 6 months posttransplantation (163). In uncontrolled trials of AIDS patients with CMV gastrointestinal disease, ganciclovir appeared to reduce diarrhea and the pain and dysphagia of esophagitis (62, 74, 109). A recent, well-controlled study of bone marrow recipients, however, indicated no benefit in this patient group (372). In patients who have undergone solid-organ transplantation and developed CMV disease, there is some evidence that ganciclovir therapy improves the recovery rate (119, 180, 193) and may improve graft survival (119). Ganciclovir resistance has been reported in three immunocompromised patients being treated for CMV disease (127) and was due to lack of phosphorylation of the drug by the CMV-infected cells (452). This finding is consistent with phosphorylation of ganciclovir by a virus-induced cellular enzyme or by an enzyme that is virus encoded. In addition, a CMV strain produced in the laboratory has been found to possess two mutations which confer ganciclovir-resistance: one in the DNA polymerase gene giving rise to an altered DNA polymerase and one at an unknown site giving rise to an inability to phosphorylate the drug (463). An HSV strain resistant to ganciclovir because of an altered DNA polymerase has been identified (84). A recent prospective survey estimated that 8% of patients receiving ganciclovir for more than 3 months developed resistant CMV (118), suggesting that prolonged therapy may facilitate the emergence of resistant strains. Although the importance of ganciclovir resistance is presently unknown, the three reported patients all had progressive CMV disease, and this suggests that resistance can be associated with therapeutic failure. Foscarnet Foscarnet (trisodium phosphonoformate) is a pyrophosphate 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, another pyrophosphate analog with antiviral properties (196). The Food and Drug Administration recently granted an indication for treatment of CMV retinitis in AIDS patients. In clinical trials, foscarnet has also proven useful for treatment of acyclovir-resistant herpesvirus infections. Unlike nucleoside analogs, foscarnet does not need to be activated by cellular or viral kinases. Instead, the molecule binds directly to the pyrophosphate-binding sites of RNA and DNA polymerases, inhibiting them in a noncompetitive fashion with respect to nucleotide substrates (328). While the mechanism of inhibition is not well understood, one hypoth- ANTIVIRAL THERAPY 157 esis is that polymerase-bound foscamet forms an unstable intermediate with nucleoside monophosphates, resulting in degradation of the nucleic acids (275, 328). Both cellular and viral polymerases are affected by foscarnet, but some viral enzymes are inhibited by concentrations 1/10 to 1/2 those needed for cellular enzyme inhibition. These include the polymerases of some strains of influenza A virus, the human herpesviruses, hepatitis B virus, and the reverse transcriptase of HIV (269, 328, 404, 488, 493). Foscarnet is difficult to use. Oral bioavailability is poor (433), although efforts are being made to increase absorption and prolong the half-life by conjugation to fatty alcohols (322). Currently, the drug is available only in intravenous formulations, and it must be given frequently by means of an infusion pump, as the half-life in plasma is very short (9, 433). At physiologic ph, foscarnet is highly ionized and has limited cell penetration (328, 433). Levels in spinal fluid are usually about 40% of those in plasma but may vary by a wide margin (7). Up to 30% of the drug may be deposited in bone, with a subsequent half-life of several months (432). The importance of such deposition, especially for bone development in children, is not known. The major adverse effects of foscarnet are renal impairment (present in most patients and dose limiting in 10 to 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 chelates divalent metal ions such as calcium, resulting in a doserelated but reversible drop in serum calcium that may be severe or even fatal (223). Penile ulcers have also been seen with systemic therapy and may result from the localized irritant effect of high levels of ionized foscamet in the urine (136, 168, 276, 319, 482). In AIDS patients with CMV retinitis, foscarnet and ganciclovir appear to be equally effective in preventing progression of disease, as assessed in clinical trials with historical controls. Vision stabilized or improved in 80 to 100% of patients, but maintenance therapy was required, and even so, 70% or more relapsed (134, 224, 274, 336, 495). The potential advantage of foscarnet in this setting is that it has less bone marrow toxicity than ganciclovir, and it may be possible to use it in conjunction with zidovudine. The additive bone marrow toxicities of ganciclovir and zidovudine preclude their simultaneous administration. A controlled trial directly comparing ganciclovir and foscamet for CMV retinitis was recently completed. Preliminary analysis confirmed that they are equally effective in halting progression and suggested that foscarnet may prolong survival by a few months in some patients. Other uncontrolled trials have indicated that some bone marrow and renal transplant patients with severe CMV pneumonia or with fever and leukopenia may improve with foscarnet therapy. Many died, however, and in the absence of a control group, it is difficult to assess the value of therapy in these patients (4, 238, 382). In AIDS patients who develop acyclovir-resistant HSV or VZV infections after prolonged courses of therapy, foscarnet has proven very useful (Table 1). About 80% of patients respond with ulcer healing and symptom resolution (63, 130, 400, 401). In this setting, foscarnet is also superior to vidarabine; the rate of lesion healing is higher and there is less toxicity (402). Maintenance therapy with foscarnet is needed, as with other antiherpesvirus agents, and is successful in about 70% of patients. Apparent resistance to foscarnet has developed, however, after several courses of therapy in HIV-infected patients (34) and in one bone marrow allograft recipient treated for CMV pneumonia (241). The mechanisms of resistance have not yet been determined for

13 158 BEAN these viruses but probably involve altered DNA polymerases, as seen when viruses are exposed to foscarnet in vitro (102). When patients are infected with thymidine kinasedeficient, acyclovir-resistant HSV or VZV mutants, as is most commonly the case, foscarnet is useful for therapy, as it does not require activation by thymidine kinases. When acyclovir resistance is due to changes in the viral DNA polymerase, however, it may be accompanied by resistance to foscarnet, and in these cases the drug may not be useful. Foscarnet is effective against HIV at levels achieved in patients' serum (404, 488). In preliminary studies, it decreased viral (p24) antigenemia and raised helper lymphocyte (CD4) counts (30, 221, 328), but the effect was transient, and patients relapsed when the drug was stopped. The need for intravenous administration has made it difficult to study foscarnet for long-term maintenance therapy in HIV infection and may preclude its use in favor of more easily administered drugs. Zidovudine Zidovudine (3'-azido-3'-deoxythymidine; formerly known as azidothymidine, or AZT), is a synthetic dideoxynucleoside, one of a family of nucleoside analogs in which the 2' and 3' hydroxyls of the ribose ring have been replaced by hydrogens or other moieties. It was first studied as an antitumor compound and then as a drug for feline leukemia virus, and in 1985, it was found to inhibit HIV (316). The drug became commercially available in 1987 and is now used for 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 triphosphate forms by cellular enzymes in both HIV-infected and uninfected cells (155). The triphosphate is a competitive inhibitor of HIV reverse transcriptase, the RNA-dependent DNA polymerase needed for transcription of viral genomic RNA into double-stranded DNA that can be incorporated into host cell DNA (Fig. 2). In fact, zidovudine binds to reverse transcriptase much better than does its natural competitor dttp and is thus the preferred substrate (66, 155, 454). It also inhibits reverse transcriptase at about 1/100 the concentration needed for inhibition of cellular DNA polymerase alpha (66, 155, 316). Zidovudine triphosphate is incorporated into the growing chain of DNA, and because the azido group occupies the 3' hydroxyl site, additional nucleotides cannot attach and the chain is terminated (155, 454). Zidovudine is active in vitro against mammalian and avian retroviruses at low concentrations (153) and against EBV at concentrations 10 to 100 times those needed to inhibit HIV (277). It inhibits some bacteria of the Enterobacteriaceae family when they are actively dividing (124, 234). Zidovudine is available in both oral and intravenous forms. It is absorbed rapidly and completely from the gastrointestinal tract and so is well suited to oral use (38). It also penetrates well into the cerebrospinal fluid and is useful for treating central nervous system manifestations of AIDS. The drug is metabolized primarily by hepatic glucuronidation, and thus its metabolism may be inhibited by other drugs that share this pathway, such as sulfa-containing compounds, nonsteroidal anti-inflammatory agents, and narcotic analgesics. Probenecid, another compound that inhibits glucuronidation, can also be used to prolong the half-life of zidovudine (247). One of the major drawbacks to the successful use of zidovudine has been its rapid elimination and short plasma half-life, necessitating frequent administration. CLIN. MICROBIOL. REV. However, its intracellular half-life is considerably longer (8, 155), and more convenient dosing schedules have recently been proven effective. Zidovudine also crosses the placenta (497) and has recently been associated with fetal resorption when 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 reverse transcriptases to be inhibited by zidovudine, it is nonetheless a moderately toxic drug. This may be explained by recent observations. Zidovudine monophosphate competitively inhibits thymidylate kinase, a cellular enzyme that catalyzes the conversion of nucleoside monophosphates to diphosphates (155). As a consequence, when cells are exposed to the drug, intracellular pools of nucleoside di- and triphosphates decrease, and cellular DNA synthesis is slowed accordingly (8). In addition, when zidovudine is incorporated into a growing chain of cellular DNA, it causes chain termination (8). In patients, zidovudine can cause anemia severe enough to require transfusion or discontinuation of therapy. Neutropenia is less frequent, but other reactions such as nausea and headache are common, especially at higher doses and in patients with more advanced AIDS (166, 379). A myopathy characterized clinically by muscle weakness and anatomically by abnormal mitochondria in muscle fibers has also been described (88). Patients on zidovudine appear to have a high risk of non-hodgkin lymphoma (318, 354), although whether this susceptibility is due to the drug or simply to prolonged survival in the face of profound immunosuppression is not yet clear. The bone marrowsuppressive effects of zidovudine are at least additive (206) and may be synergistic with those of ganciclovir (359). Few patients 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-related complex, high-dose zidovudine (1,200 to 1,500 mg daily) prolongs survival, reduces the frequency of opportunistic infections, and improves functional capacity, weight gain, and immunologic function (116, 146). Viremia or HIV (p24) antigenemia also decreases during therapy (218, 446). Although the survival benefit remains after 1 to 2 years of therapy (81, 145), the other benefits tend to disappear, with a return of antigenemia and opportunistic infections and a deterioration of immunologic function. This is partly because drug toxicity requires dose reductions or discontinuation of therapy in many patients. A recent study has shown that a lower daily dose, 600 mg, is as beneficial as the higher dose and much less toxic, at least in nonadvanced AIDS (144). It is now the recommended dosage. As little as 300 mg daily may be beneficial in patients with AIDS-related complex (75), although it is not recommended unless patients are unable to tolerate higher doses. High-dose zidovudine is still used for treatment of HIV neurologic disease (412, 523, 527). The bone marrow-suppressive effects of zidovudine spare the megakaryocytes and platelets, and the drug can be used to treat HIV-induced thrombocytopenia (332, 466). Although zidovudine reaches high concentrations in semen (199), it does not appear to decrease shedding from this site (252), and this lack may have important implications for sexual transmission of disease. In asymptomatic or mildly symptomatic HIV patients with CD4 (helper) T-lymphocyte counts of less than 500/mm3, both high- and low-dose zidovudine delayed disease progression and transiently improved immunologic function and antigenemia (147, 487). Adverse effects were also less common in these patients than in those with more advanced disease. These patients have not been followed long enough

14 VOL. 5, 1992 to determine whether zidovudine prolongs survival in early HIV infection. A recent cost effectiveness analysis, however, indicates that zidovudine therapy in early HIV infection compares favorably with other well-accepted medical interventions. The cost per year of life saved, estimated at $6,553 (1989 dollars), is very similar to that of counseling patients to quit smoking (415). Resistance of HIV to zidovudine has been seen in patients receiving therapy for prolonged periods (258, 267, 391). Resistance appears to develop more rapidly and to higher levels in patients with more advanced disease and greater viral burdens (380), a situation somewhat analogous to the development of isoniazid resistance in patients with cavitary tuberculosis and high burdens of mycobacteria. The clinical importance of resistance is not known; thus far, there has been no correlation with individual patient responses to therapy. At the molecular level, resistance is due to specific changes in three or four amino acid residues of the reverse transcriptase molecule (267). In patients, high-level resistance 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 recognition of this moiety by the altered reverse transcriptase (264). Despite rapid and remarkable advances in the development and use of zidovudine, many questions remain unanswered. The minimum effective dose is not yet known, nor are the effects of resistance and cumulative toxicity. Whether the drug prolongs survival when given early in disease remains to be seen, and whether it will prevent HIV infection if given prophylactically after exposure is unknown, though anecdotal experience suggests that this is not the case (263). Didanosine Like zidovudine, didanosine (2',3'-dideoxyinosine [ddi]; Fig. 5) is a dideoxynucleoside. It became available for management of HIV infections in late 1991, before definitive patient trials were completed. It is the first compound to follow a "fast track" to Food and Drug Administration approval and be made available prior to completion of safety and efficacy studies in an attempt to make promising new drugs quickly available to desperately ill patients. Didanosine is an analog of deoxyadenosine. It is converted by cellular enzymes to ddl monophosphate, then to dideoxyadenosine (dda) monophosphate, to dda diphosphate, and finally to dda triphosphate (ddatp), the active form (1, 78, 227). Like the triphosphates of other dideoxynucleosides, ddatp competitively inhibits HIV reverse transcriptase and acts as a chain terminator for proviral DNA (524). Human DNA polymerase alpha is relatively resistant to ddatp, but polymerases beta and gamma are more sensitive to it (496), perhaps accounting for some of the toxicity seen in patients. In vitro, ddl is less potent against HIV than dideoxycytidine (ddc) (315), but intracellularly ddatp is as active as zidovudine (344), and it has an intracellular half-life of 8 to 12 h (compared with about 3 h for zidovudine [1, 227]). Didanosine is also active against zidovudine-resistant HIV isolates (264). Didanosine is available in oral form but is somewhat difficult for patients to take correctly. At acid ph, hydrolysis occurs at the glycosidic bond between the sugar and the base moieties of the nucleoside, inactivating the compound. The drug is thus supplied as both a buffered powder to be made into solution and as buffered chewable tablets which must be ANTIVIRAL THERAPY 159 taken at least two at a time to provide adequate buffering capacity in the stomach. The major side effects have been a painful peripheral neuropathy in 16 to 34% of patients treated to date; pancreatitis, seen in 8 to 9% of patients and occasionally fatal (77, 257, 525); and diarrhea in 18 to 34% of patients, possibly related to buffering agents used in the powder (182). Didanosine is avoided in patients at high risk for pancreatitis, such as those with a history of heavy alcohol consumption or previous pancreatitis, and it increases the risk of pancreatitis when given simultaneously with intravenous pentamidine or ganciclovir. Development of pancreatitis or peripheral neuropathy requires discontinuing the drug. In phase I uncontrolled dose escalation studies of patients previously treated with zidovudine, didanosine therapy resulted in significant rises in CD4 cell counts and decreases in p24 antigenemia (50, 77, 257, 525). Phase II controlled trials are under way to determine whether ddi has any effect on patient survival, time to progression of disease, or frequency of opportunistic infections. In view of the desperate need for additional anti-hiv therapies and because rising CD4 cell counts are a surrogate marker for disease improvement, didanosine was approved for use in both adult and pediatric HIV-infected patients who are unable to tolerate zidovudine (usually because of bone marrow toxicity) or who experience clinical or immunologic deterioration while receiving it. Zidovudine is still regarded as first-line HIV therapy. The place of ddl, both by itself and in combination with other agents for HIV therapy, will need to be determined in future trials. Investigational Antiretroviral Agents Many compounds are under investigation as candidate drugs for the management of HIV infection. It is not possible to discuss them all, and this section will concentrate on those which are especially promising or represent a new approach to therapy and for which there is adequate description in the scientific 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 HIV at 1/20 to 1/50 the concentrations needed for dda, ddl, and dideoxyguanosine, and it was the first of these to begin clinical trials. ddc is well absorbed orally, has a short half-life, and penetrates the central nervous system, although not as well as zidovudine (526). Its phosphorylation pathway is different from that of zidovudine, and, unlike zidovudine, it is excreted by the kidneys (526). Its toxicity profile is also substantially different from that of zidovudine. In early trials, patients were given high doses and many of them developed a painful peripheral neuropathy, necessitating discontinuance of the drug. Decreases in p24 antigenemia and rises in CD4 counts were also seen, however, and no bone marrow toxicity was observed (304, 526). Currently, trials are in progress to determine the efficacy of lower doses and of dosage schedules that employ zidovudine and ddc together, given either simultaneously or in an alternating fashion (43, 351, 435). Approval of ddc by the Food and Drug Administration is expected in the near future. Two other dideoxynucleosides are currently in clinical trials. Dideoxythymidinine, also known as D4T, has good activity against HIV but is phosphorylated differently from zidovudine and has less bone marrow toxicity (205, 284). Azidouridine is slightly less active against HIV than zidovudine, but is also less toxic to bone marrow (67, 531), and it

15 160 BEAN H CH3 TIBO R HO 4H 0 OH oaoh K No CH2OH I CH2CH2CH" CH3 N-BUTYL-DNJ o"t. PROTEASE INHIBITOR XVII FIG. 7. Investigational antiretroviral agents. Ph, phenyl; tbu, tertiary butyl; N-butyl-DNJ, N-butyl-deoxynojirimycin. inhibits the virus very well in peripheral blood mononuclear cells. It is currently in phase I trials. Several nonnucleoside compounds have shown excellent activity in vitro against HIV reverse transcriptase and are candidates for patient trials. Among the most potent and specific are the tetrahydroimidazobenzodiazepinone (TIBO) compounds, benzodiazepine derivatives that inhibit HIV type 1 (HIV-1) but not HIV-2 or other retroviruses. In vitro, the selectivity index of TIBO R82150 (Fig. 7) is greater than 31,000, compared with 6,200 for zidovudine and 191 for didanosine in the same cell system (339). These agents do not require phosphorylation and inhibit HIV-1 reverse transcriptase by an as yet unidentified mechanism. B1-RG-587 (nevirapine) is a dipyridodiazepinone with properties similar to those of the TIBO compounds (249, 305). It is beginning phase I trials in the near future. A group of six-substituted acylouridine derivatives, the HEPT [1-(2-hydroxyethoxymethyl)-6-phenylthiothymine] congeners, are also highly potent and specific for HIV-1, do not require phosphorylation, and appear to interfere with reverse transcriptase by a mechanism different from that of zidovudine (10, 12). One compound, HEPT-S, also has pharmacokinetic properties suitable for oral administration. Reverse transcriptase inhibitors can prevent new DNA from being made in newly infected cells but they cannot prevent the reactivation of HIV from previously infected cells, as reverse transcriptase is not involved in this process (Fig. 2). Identification of agents capable of interfering with HIV at later points in the replication cycle, and perhaps preventing reactivation, would represent a major advance in the management of HIV infection. One such group of agents is the HIV protease inhibitors. HIV reverse transcriptase and other gag and gag-pol gene products are synthesized as precursor polypeptides and must be cleaved and processed before they can be packaged into virions. HIV protease, an aspartic proteinase encoded by the virus, is responsible for this processing and is essential for the proper assembly and maturation of fully infectious HIV-1 virions. It can also be CLIN. MICROBIOL. REV. inhibited by protease inhibitors, small peptide molecules that mimic the natural substrate for the reaction (97) (Fig. 7). Several such compounds have been identified and are being developed for clinical use (128, 248, 383). They are highly potent and selective inhibitors of HIV in vitro, and at least one has been shown to have antiviral activity when added to cell cultures late (18 to 20 h) after HIV infection and in chronically infected cells (162). Another promising compound, which acts later in the HIV replication cycle, is N-butyldeoxynojirimycin (Fig. 7). It reduces infectious virus in vitro in acutely infected cells and eliminates HIV entirely from chronically infected cells (231). The compound works by inhibiting glycosylation of the envelope glycoprotein gp120, thereby reducing its ability to bind to CD4 receptors and initiate new viral infection. Although glycosylation of viral glycoproteins is nonspecific and can be carried out by cellular enzymes, N-butyl-deoxynojirimycin 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 of many diverse diseases in addition to viral infections. It consists of constructing small oligonucleotides that are complementary to specific genes or mrna sequences (antisense oligonucleotides). These bind to the nucleic acid, ultimately preventing gene expression and protein synthesis. A molecule antisense to the mrna of rev, a critical regulatory protein of HIV, has been synthesized, and it inhibits protein synthesis in vitro in chronically infected cells (289). While this approach is extremely exciting, several problems with oligonucleotides, including degradation by RNases, cellular permeability, specificity, and large-scale production, must be solved before they can be used as therapeutic agents. All of these problems are under active investigation (397). It is also possible to interfere with HIV infection at its first step, the binding of HIV to CD4 lymphocytes (Fig. 2). Recombinant, soluble CD4 (rscd4) is the synthetic gpl20 binding portion of the complete CD4 molecule (the transmembrane and cytoplasmic portions are missing). In vitro, rscd4 binds to gpl20 on the virion surface and prevents attachment of the virion to CD4 lymphocytes (438). In patients, no toxicities were seen when the compound was administered in phase I trials, but the half-life in serum was very short, and sustained changes in p24 antigenemia and CD4 cell counts were not seen (229, 414). Currently, efforts are being directed toward the construction of rscd4-immunoglobulin G (IgG) and rscd4-igm hybrid molecules that have longer half-lives (53), may cross the placenta (53), have greater activity (478), and may recruit antibody-dependent (51) or complement-mediated (478) immune functions to destroy infected cells. Other efforts have resulted in the coupling of rscd4 to toxins such as Pseudomonas exotoxin A (64) and the plant toxin ricin (473). When bound to the HIV-infected lymphocyte and internalized, these compounds result in cell death. The use of Pseudomonas exotoxin has been especially effective in vitro and "cures" cell cultures of HIV infection when used in conjunction with reverse transcriptase inhibitors (6). IMMUNOGLOBULINS Immunoglobulins are available in three different formulations: an intramuscular preparation termed immune globulin or IG; an intravenous form termed IVIG; and several different intravenous preparations with high titers of antibodies to

16 VOL. 5, 1992 individual viruses. All are made from pooled human plasma and contain predominantly IgG, though small amounts of other globulins are also present. High-titered (hyperimmune) globulins are derived from pooled units of plasma selected for high antibody titers to specific viruses. IG has been available for many years, but it cannot be administered intravenously because of the tendency for IgG to form aggregates and fix complement in vivo. Recent advances in the cold fractionation procedure used, however, have made it possible to prevent this aggregation, and newer preparations can be given intravenously. Large volumes of fluid cannot be administered intramuscularly, so the amount of immunoglobulin that can be given by this route has also been limited. It is possible to give much larger amounts by the intravenous route, although these preparations are extremely costly. Regardless of route of administration, immunoglobulins are quite safe and are associated with very few side effects. The mechanism of action is unknown, but in all likelihood 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 antibodies are present during the extracellular phase of infection, when it is possible to neutralize or opsonize virions before they enter cells and are protected from humoral immunity. Currently, IG is used primarily for prevention of hepatitis A. It is administered to patients within a few days of exposure to contaminated food or to an infected person and prevents infection or chemical illness in 80 to 95% of recipients (176, 255). It is also given at regular intervals to travelers who will be spending prolonged periods in conditions of poor or uncertain sanitation (61, 518). Although IG has not been unequivocally shown to decrease infection with non-a, non-b hepatitis, it may be "reasonable" to administer it to persons who sustain percutaneous exposures to infected blood (61). IG has not been studied for prevention of hepatitis C. For prevention of measles, IG should be given to persons who have not had the disease or been vaccinated against it and who are household contacts of measles patients, to those who have been exposed while pregnant, and to those who are immunocompromised when exposed (59). IG does not replace vaccine for control of measles outbreaks. Hyperimmune globulins are used for postexposure prevention of hepatitis B, chickenpox, and rabies. Hepatitis B IG prevents disease in 75% of persons with needlestick exposures (420). It also prevents hepatitis B surface antigen carriage in 85 to 95% of babies born to e-antigen-positive mothers (26, 61). Along with hepatitis B vaccine, hepatitis B IG is given within a few hours of birth to babies of hepatitis B surface antigen-positive mothers. Varicella-zoster IG modifies the severity of chickenpox but does not prevent infection when given to exposed susceptible persons. In immunocompromised children of less than 15 years of age (many of whom have not already had chickenpox and thus are not immune), it is administered as soon as possible after exposure to chickenpox. It is also given to babies who have no maternal antibody, including newborns whose mothers develop chickenpox within 5 days before or 2 days after delivery, 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 pregnant women, is limited to those situations in which the physician seeks to reduce disease severity. It has not been ANTIVIRAL THERAPY 161 shown to decrease the frequency of congenital or neonatal varicella when administered to exposed pregnant women and, in fact, may do the opposite by making the disease less severe in the mother while allowing transmission to the fetus (56). Human rabies IG became available in 1975 and is made from the plasma of hyperimmunized human donors. It is used in conjunction with human rabies vaccine to decrease transmission and death after exposure to rabid animals (55). CMV IG was recently approved for management of CMV infections in renal transplant patients. When given prophylactically, it reduced the frequency of serious CMV disease in seronegative recipients of seropositive kidneys, although it did not decrease transmission of the virus in this setting (442). In bone marrow transplant patients the use of prophylactic CMV IG is controversial. In two studies it decreased infection rates in seronegative recipients (41, 308), in one it did not (42), and in one it decreased CMV disease severity but not infection rates (516). These discrepancies may be due to differences in antibody titers of the immunoglobulins or to the fact that small numbers of patients were studied. Intravenous immunoglobulins were originally developed for the treatment of primary immune deficiency states. They have, however, been studied and used for many other purposes, including treatment and prophylaxis of viral infections. Preparations are available from several different manufacturers, and while they are frequently considered equivalent, both manufacturing processes and donor populations differ considerably and may result in substantially different end products. Moreover, manufacturers are not permitted to publish information about antibody titers of their products, so it is difficult to know whether preparations have equal efficacy unless they are directly compared in patient trials. As discussed earlier, IVIG improves survival when used in conjunction with ganciclovir for treatment of CMV pneumonia in bone marrow recipients (125, 371). In a recent trial, IVIG also decreased the frequency of graft-versus-host disease and interstitial pneumonia when given alone to seropositive bone marrow recipients (462). Although CMV was not confirmed as the cause of pneumonia in this study, it is the most common cause of interstitial pneumonia following bone marrow transplantation and it is associated with graft-versus-host disease. This suggests that IVIG may be beneficial in this setting. Another important use of IVIG is in the treatment of chronic enteroviral meningoencephalitis in children with agammaglobulinemia. Given intravenously, immunoglobulin in one study did not reach very high levels in the cerebrospinal fluid, and children almost always relapsed, though they had responded initially (297). When immunoglobulin was given through an intraventricular catheter, however, 50% of the children responded, and a few appear to have been cured of their infection (120, 297). Further studies need to be done, but intraventricular immunoglobulin appears to be a major advance in the treatment of this devastating illness. 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 may provide functioning antibodies to other infectious agents in patients who are functionally hypogammaglobulinemic despite having elevated serum globulin levels (261). Children, unlike adults, do not have protective antibodies from previous illnesses, and they may benefit from exogenous immunoglobulins. One large multicenter trial of IVIG therapy recently demonstrated that symptomatic HIV-infected children with CD4 counts of greater than 200 per mm3 had

17 162 BEAN significantly fewer serious bacterial infections and fewer hospital days than those treated with placebo (320). A similar benefit was not seen among those with fewer CD4 cells, but this may have been because of the small number of children studied. A second trial is under way to further investigate these findings. No preparation of hyperimmune HIV globulin is available. A recent trial using a high-titered preparation made from the plasma of two donors with high anti-p24 antibody titers showed that AIDS patients given the plasma had immediate clearing of p24 antigenemia, fewer symptoms, transient rises in T lymphocytes, fewer opportunistic infections, and lower rates of HIV isolation from blood (219). Such results are intriguing but have not been confirmed. IMMUNOMODULATORS In the strict sense of the word, immunomodulators are substances that modify the response of immune competent cells through signaling mechanisms (519). Many discussions, however, including this one, include immune-based therapies with a direct effect on cells such as the use of specific antibodies, exogenous immune effector cells, and procedures such as bone marrow transplantation. Immunomodulators are administered in the hope of augmenting or restoring host immune responses to infectious agents or malignancies. A few older agents, for instance, levamisole, isoprinosine, and transfer factor, have been used for several years in patients with viral infections (455). Their effects have never been convincingly demonstrated, however, and they remain largely investigational. The AIDS epidemic has rekindled interest in immunomodulators, and with the use of recombinant DNA technology it has become possible to manufacture these compounds in large quantities and to study their effects in patients, animal models, and in vitro systems. Immunomodulation in HIV infection (and other viral infections) can be a two-edged sword, however. Potent immunomodulators such as cytokines have the potential for increasing viral replication and perhaps triggering the transition from latent to active infection by stimulating target cells and making them more capable of supporting viral infection. They may also induce new targets by stimulating cells otherwise nonpermissive for viral infection. Last but not least, they may worsen or cause opportunistic infections by stimulating the target cells for these organisms. If some of the features of HIV disease are autoimmune in nature, it is possible that immunomodulators will also worsen them (519). Tumor necrosis factor alpha, interleukin 6, and granulocyte-macrophage colony-stimulating factor (GM-CSF) have been shown in vitro to induce expression of HIV from chronically infected cells. Interleukins 1, 2, 3, and 4 and gamma interferon do not, while alpha interferon and transforming growth factor beta actually down-regulate HIV expression (135). Only two immunomodulators, alpha interferon and GM-CSF, are commercially available, and this discussion will concentrate on them. Many other agents are in the early phases of investigation for treatment of HIV infection. Three of these, inosine pranobex (isoprinosine), ditiocarb sodium (diethyldithiocarbamate), and IMREG-1, have been administered to patients in controlled trials and have shown some possible short-term effects in decreasing progression of the disease (171, 200, 342). More and better studies are needed, however, to establish their utility in treatment of HIV infection. Other agents such as thymic hormones (21, 430), enkephalins (333), interleukin 2 (416), CLIN. MICROBIOL. REV. and activated natural killer cells (301) and bone marrow transplantation (262) are in earlier stages of investigation. One other intriguing approach is that proposed by Salk: the use of inactivated HIV vaccine or its components in HIVinfected asymptomatic individuals to stimulate cytotoxic responses to infected cells and eliminate them, preventing progression of disease and development of full-blown AIDS (403). An early, phase I trial of this approach has shown it to be safe and immunogenic (370). Patients were followed for less than 1 year, however, and further trials will be necessary to determine the effect of postinfection immunization on the course of HIV disease. Interferons are natural products, small proteins, or glycoproteins that are elaborated by eukaryotic cells in response to viral infection. In turn, they induce in other noninfected cells biochemical changes that render these cells resistant to subsequent viral infection. The "antiviral state" induced by interferons is not specific with respect to infecting virus but is relatively specific with respect to species of host cell or animal. Interferons were first described by Isaacs and Lindemann in 1957 (216) and have subsequently been found to have potent antiproliferative and immunomodulating effects in addition to their antiviral properties (349). Their primary function is, in fact, related to the regulation of cell growth, differentiation, and immune function (228). Interferons are currently being used and actively investigated for the treatment of malignancies as well as viral diseases. In nature there are three types, alpha, beta, and gamma, classified by cell of origin, primary function, and other chemical and physical properties. Alpha and beta interferons are secreted in response to viral infection and are involved in the response to it, while gamma interferon is produced by activated T lymphocytes and is involved in the response to antigens and mitogens. While gamma interferon is the primary immunomodulator, all interferons affect the immune system to some extent. The only products available commercially are alpha interferons, and this review will concentrate on them. Interferons are now known to be cytokines, hormonelike polypeptides that play an important regulatory role in the early nonspecific phases of the host response to microbial invasion. They are elaborated by almost all nucleated cells after exposure to a variety of stimuli, including viruses, viral antigens, and double-stranded RNA. They are then secreted from the infected cells and attach to receptors on other, uninfected cells. After internalization, they trigger the formation of enzymes that, upon exposure to double-stranded RNA, degrade viral mrna and inhibit protein synthesis, resulting in an aborted viral infection. The best studied of these enzymes are 2,5-oligo(A), a family of oligonucleotides that synthesize an activator of RNase L, an enzyme that cleaves RNA; and protein kinase, which phosphorylates and thereby inactivates eif-2, a protein initiation factor (228). These are not the only mechanisms by which virus infections are halted, however. The multiplication of HIV, for instance, is directly inhibited by interferons, apparently at a late stage of replication involving release of virus particles from the cell membrane (356). While these mechanisms are clear-cut in vitro, the in vivo actions of interferons are undoubtedly more complex because of the effects of interferons on immune functions, including stimulation of natural killer cells and of B-lymphocyte proliferation, inhibition of lymphocyte blastogenesis, and enhanced expression of major histocompatibility class I receptors on cell surfaces (240). Interferons are responsible for the fever, chills, arthralgias,

18 VOL. 5, 1992 and myalgias frequently seen with viral infections, and they may actually cause immune-mediated tissue damage. Most interferons in use today are made by using recombinant DNA technology. They are not absorbed from the gastrointestinal tract when given orally and thus are administered by intramuscular or subcutaneous injection (512). The serum half-life is short, but the antiviral state induced in cells can last several days (228). The most common early adverse effect is an influenzalike illness characterized by fever, headache, myalgias, and arthralgias. These symptoms can usually be controlled with medication, and tolerance soon develops, so that therapy can be continued. Later, however, fatigue, anorexia, and weight loss can develop, and these often limit the use of the drug. Other adverse effects have been described, though they are much less common: bone marrow toxicity, psychological abnormalities (376), and cardiac toxicities (445). Interferon neutralizing antibodies are produced in 2 to 3% of patients receiving systemic therapy (447). While the significance of these antibodies is not completely understood, their presence has not yet been associated with a diminished response in patients being treated for viral infections. Interferons were first used clinically for the management of herpesvirus infections in immunocompromised patients. Although they were useful in controlling the complications of chickenpox and zoster (5, 303, 515) and in preventing the reactivation of CMV after renal transplantation (203), they had no effect on prevention of CMV after bone marrow transplantation (307) and were not as effective as acyclovir for treatment of genital herpes (256, 340). In addition, the frequency of adverse effects was high, and they were soon replaced by acyclovir and ganciclovir. Because the common cold is caused by a variety of different viruses, a nonspecific agent such as interferon would seem an ideal mode of treatment and/or prophylaxis. Leukocyte interferon was first administered intranasally for this purpose in 1973 (302). Unfortunately, it produced a very high frequency of local side effects such as nasal stuffiness, dryness, and bleeding and actually prolonged cold symptoms rather than shortening them (190). When given prophylactically to exposed family members, intranasal recombinant interferon decreased the frequency of rhinovirus colds, but it has little effect on colds due to other viruses (115, 187), and it was again associated with very bothersome side effects. It has now been abandoned for this purpose. Anogenital warts (condyloma acuminata) lend themselves to local, intralesional therapy, and interferons may be useful for this purpose. Compared with placebo, they promote faster healing and fewer recurrences both in untreated patients and in those with refractory warts (132, 152, 375). Side effects were less common and less severe than in patients receiving systemic therapy, although intralesional interferon is absorbed systemically. Interferons used in conjunction with other local therapies such as podophyllin (113) and laser vaporization (480) also appear superior to these agents alone. The role of intralesional interferon therapy in genital warts is not yet clear. Such therapy is very expensive, and 20 to 30% of patients still suffer recurrences. Intralesional injections are painful, limiting the number of warts than can be treated. Cervical warts are very difficult to inject and may not be treatable by this mode. In adults with chronic hepatitis B, a lack of endogenous interferon may play a role in failure to clear the acute viral infection and in progression to chronic disease (94). Treatment with exogenous interferons has been under investigation for several years. Earlier trials suggested that the best ANTIVIRAL THERAPY 163 results were achieved by pretreating patients with prednisone, stopping this treatment, and then initiating interferon therapy during the rebound from corticosteroid therapy (347). A recent well-controlled trial, however, shows that this approach is effective in only a small subset of hepatitis patients with minimal liver dysfunction (348). Another larger subset of sicker patients responded to long-term (4-month) treatment with interferon alone, showing improvements in liver function and in abnormal histology and disappearance of hepatitis B e antigen, surface antigen, and DNA from the serum (348). A recent long-term follow-up study of treated patients indicated that many of these remissions were sustained for 3 to 7 years and may, in fact, represent cures of the disease (209, 245). In total, however, only 30 to 40% of chronic hepatitis B patients respond to interferon, and when lower doses are given, the response rate is lower (280). Further studies are needed to better determine which patients will benefit from interferon therapy and whether it will have any effect on long-term progression of disease, on survival, and on the frequency of subsequent hepatocellular carcinoma. In recent studies of interferon for chronic hepatitis C, about 50% of treated patients responded with decreases in liver function abnormalities, hepatic inflammation, and viral RNA in serum (93, 107, 427). Six months later, 50% of those responding had relapsed, resulting in a sustained response rate of only 20 to 25%. Although alpha interferon 2b has been approved for treatment of chronic hepatitis C (Table 1), many important questions remain about optimum dose and duration of treatment and the need for maintenance therapy (106). Better ways to identify patients likely to respond are also needed. In view of the cost and side effects of interferon, it should not be routinely used in hepatitis C until these problems are solved. Interferons have been known for some time to inhibit animal retroviruses, and they were shown in 1985 to inhibit HIV (204). In latently infected cells, interferons appear to act at a late point in virus replication to inhibit virion assembly and/or release (356). In acute infection, they prevent new infection of cells (204) and may act at an earlier point in replication. In mature macrophages, interferons appear to limit the formation of proviral DNA (246). When administered to AIDS patients for treatment of Kaposi sarcoma, interferon resulted in decreased p24 antigenemia and 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 asymptomatic HIV patients confirmed the ability of interferon to decrease viremia and perhaps opportunistic infections in this population (259). It also appears that interferons work best when given in early infection, while the immune system is still reasonably intact. The role of interferons, including those produced endogenously in HIV infection, is complex, however, and not completely understood. It may involve mechanisms by which the virus is able to avoid the effects of the drug, such as down-regulation of alpha interferon receptors on cell surfaces after prolonged exposure to these agents (272). As noted earlier, interferons also fail to cross the blood-brain barrier and can be associated with intolerable side effects, especially in those asymptomatic patients who stand to benefit the most from them (259). For these reasons, use of interferons alone for HIV infection may be problematic, and they are probably best used in combination with other agents such as zidovudine and other dideoxynucleosides that inhibit viral replication at a different stage. In vitro, alpha interferon and zidovudine are synergistic in

19 164 BEAN inhibiting HIV (183). In early, dose-finding patient trials, it has been possible to administer both agents together in doses that appear to have an antiviral effect, although bone marrow toxicity has been dose limiting in many cases (31, 250, 254). Much work remains to be done in this area, including investigation of gamma interferons for therapy of AIDS and use of additional agents, such as CSFs to alleviate the dose-limiting toxicities of such combinations as zidovudine and interferon. CSFs are naturally occurring cytokines that stimulate the reproduction and differentiation of granulocytes (G-CSF) or granulocytes, monocytes, and macrophages (GM-CSF) from bone marrow precursor cells. They also enhance the effectiveness of mature cells by, for instance, preventing neutrophil migration, potentiating antigen processing by macrophages, and stimulating phagocytosis of yeast cells and bacteria (499). Both G-CSF and GM-CSF are available commercially and are produced in Escherichia coli or in yeast cells by using recombinant DNA techniques. They were originally developed for prevention and treatment of neutropenias induced by cancer chemotherapy and bone marrow transplantation. A recent study indicates that they are extremely useful for this purpose, even reducing the number of hospital days required following autologous marrow transplantation (321). A great deal of interest has also developed in using them to control the complications of HIV infection, especially the neutropenias resulting from antiretroviral therapy. CSFs have no direct effect on HIV and do not affect HIV-infected lymphocytes. In monocytes, however, GM-CSF stimulates virus replication in vitro (251, 345), an effect consistent with the cell stimulatory properties of cytokines discussed earlier. If zidovudine is added to these in vitro systems, its potency is enhanced, and much lower concentrations are required to inhibit HIV than in the absence of GM-CSF (345). This effect is probably due to enhanced drug phosphorylation by the stimulated cells, and while it may be useful in the treatment of AIDS patients, it also raises the possibility of increased zidovudine-induced bone marrow toxicity when both drugs are administered simultaneously. These issues have not been resolved, but they have not prevented substantial progress in the use of CSFs. Groopman and colleagues first showed that GM-CSF was biologically active in raising granulocyte counts in leukopenic AIDS patients (173) and went on to demonstrate that the induced neutrophils functioned normally in killing Staphylococcus aureus. The same investigators also identified two patients with defective bacterial killing mechanisms (one in phagocytosis and one in intracellular killing), but these, too, appeared to be corrected by GM-CSF (14). Subsequent studies have shown both G-CSF and GM-CSF to be very effective in correcting the neutropenias seen with zidovudine therapy alone (311) or combined with interferon, and the two CSFs have allowed treatment to continue in patients otherwise 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 preliminary evidence that GM-CSF may ameliorate the neutropenia associated with ganciclovir therapy (181). The doses of CSFs needed in AIDS patients are usually much lower than those used in cancer patients, and the side effects are more closely related to the use of zidovudine and interferon. CSFs are, however, prohibitively expensive. In the dosages used for cancer patients, the cost is approximately $200 per day (300). Whether or not it will be lower in AIDS patients remains to be seen. In the face of this, it is important for future studies CLIN. MICROBIOL. REV. to identify the patients most likely to benefit from these expensive agents. PROBLEMS OF ANTWIRAL THERAPY Resistance Unlike bacteria, viruses do not have a wide variety of mechanisms for developing resistance to chemotherapeutic agents. They are genetically simple and metabolically inert, and changes like those seen in bacteria, such as decreased uptake of antibiotics, acquisition of resistance plasmids, or induction of inactivating enzymes, have not been seen. Viruses do, however, have two properties that allow them to evade potential inhibitors: they have the ability to replicate to high titers in host cells, and they can mutate very rapidly. As a consequence, most viral resistance arises from genetic mutation, giving rise to changes in either enzymes or structural components of the virion. The success of this mechanism can be seen in cell culture, where viruses readily develop resistance after only a few passes in the presence of an inhibitory agent (for a review, see reference 140). In contrast, very few resistant viruses have been isolated from immunocompetent patients being treated for viral diseases (19, 126). Resistant herpesviruses are isolated from patients immunocompromised by cancer chemotherapy or transplantation, although this is relatively infrequent. In addition, most of these resistant viruses appear to have reduced virulence and to cause predominantly indolent, non-lifethreatening infections (19, 85, 428). There are however, increasing reports of AIDS patients with life-threatening or very serious illnesses due to resistant herpesviruses (127, 131, 164, 288), suggesting that severely immunocompromised patients may provide a milieu in which mutant viruses are not at a selective disadvantage and can prosper. In addition, a larger viral load creates a larger gene pool from which mutations can emerge. Under these circumstances, selective pressure from antiviral agents could result in the propagation and perhaps the transmission of resistant viruses, eventually replacing wild-type susceptible populations with more resistant ones. Despite what is known about viral resistance in vitro, a great deal remains to be learned about its importance and about the management of patients infected with resistant viruses. A summary of unanswered questions follows. Under what circumstances does resistance arise? In general, it seems that the greatest risk is in severely immunocompromised patients receiving prolonged courses of antiviral therapy, but the details of the relationship among resistance, severity of underlying disease, and treatment are unknown. Although the herpesviruses do not become resistant easily in immunocompetent patients (325), influenza viruses do (188). The reasons for this difference are not known. What are the properties of the resistant viruses? Virulence may be reduced, but this appears to depend on the host, at least among herpesviruses. Rimantadine-resistant influenza A 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 another important but unanswered question. This ability may also depend on the host and the mechanism of resistance. Likewise, little is known about transmissibility of resistant viruses. To date, transmission of resistant herpesviruses has not been documented, but it may only be a matter of time, unless resistance is accompanied by reduced transmissibil-

20 VOL. 5, 1992 ity. Resistant influenza viruses, on the other hand, appear to be readily transmitted, at least in a household setting (188). Another very important issue is cross-resistance among antiviral drugs. A great deal is known about in vitro crossresistance, and a few clinically useful patterns have emerged, such as cross-resistance of zidovudine-resistant HIV to other azido-containing nucleosides and susceptibility of thymidine kinase-deficient HSV to drugs not needing phosphorylation or not requiring a virus-induced enzyme for phosphorylation. A great deal remains to be learned, however. At least 20 different DNA polymerase mutants have been found in vitro among HSV variants, for instance, and most of them possess altered drug-binding properties. Resistance to one compound is usually accompanied by decreased susceptibility to other drugs of the same class, but predictable cross-resistance among different classes of drugs has also been found and is thought to indicate overlap of drug-binding sites (71). Exceptions to known patterns also exist, further confirming the complexity of resistance due to DNA polymerase changes. Consequently, predicting crossresistance in DNA polymerase mutants may not be possible unless the viruses are carefully characterized. What is the result of the development of drug resistance in the patient? As discussed above, the appearance of zidovudine-resistant HIV has not yet been associated with clinical deterioration in individual patients, although it may play a role in the reduced effectiveness of zidovudine after many months of therapy. Failure of herpes lesions to heal and even serious illness are frequently due to acyclovir-resistant herpesviruses in AIDS patients, but this is not true in other immunocompromised patients in whom failure to heal may or may not be due to acyclovir resistance and in whom lesions due to resistant virus may heal despite the lack of effective therapy (470). Rimantadine prophylaxis against influenza A was ineffective in persons with household exposures to rimantadine-treated patients, but the influenzal illness itself (due to rimantadine-resistant influenza) was typical and resolved in the usual self-limited fashion (188). Thus, viral resistance is clinically important in some, but not all, situations and much remains to be learned. What are the most reliable ways of identifying resistance in the laboratory? Traditional research methods that involve propagation of viruses in cell culture, exposure to inhibitory agents, and measurement of subsequent changes in host cells or in amount of virus produced are time-consuming and expensive. They may also be unreliable in that they can select for virus strains that grow well in cell culture but are not necessarily representative of the virus population present in the patient. It is increasingly appreciated that viruses isolated from patients are not clonal derivatives homogeneous in their properties but instead are heterogeneous populations of viruses with differing drug susceptibilities and other properties, including the ability to grow in different cell lines (123, 149). As a result, new methods of measuring resistance are being developed for the research laboratory, including use of the polymerase chain reaction for detection of resistance genes, cell lines which allow growth of a broader spectrum of viral strains, and improved methods of nucleic acid hybridization (226, 264, 266). It is hoped that these can be adapted to the clinical laboratory so that patients can benefit directly from the improved methods. What can be done to minimize problems due to drug resistance? As with tuberculosis, it is hoped that combination therapy in HIV disease will forestall or even prevent clinical manifestations of drug resistance (see below). Other ANTIVIRAL THERAPY 165 measures that have been successful in herpesvirus-infected patients include avoiding therapy with prolonged courses of drugs at "subinhibitory" levels and discontinuation of therapy whenever possible. Latency With the possible exception of a new agent that may "cure'" cell cultures of HIV infection (see N-butyl-deoxynojirimycin, above), antiviral agents are virustatic rather than virucidal. In vitro, such agents are most active against actively proliferating viruses, and when these agents are removed from cell cultures, virus replication resumes. In patients, a course of treatment with acyclovir does not prevent future recurrences of HSV, nor does zidovudine prevent progression of AIDS, indicating that these drugs do not eliminate these viruses when they are in the latent state. Furthermore, the presence of latent virus in treated patients could afford the opportunity for prolonged contact between virus and drug, with attendant opportunities for development of drug resistance (140). The successful treatment of latent viral infections will depend, as it does in acute viral infections, on the identification of specific viral processes that can be specifically attacked by antiviral agents. Recently, a great deal of progress has been made in defining the molecular events associated with latent HSV infections. A group of RNAs, termed latency-associated transcript, has been described and possibly linked to reactivation of latent HSV (201); for a review, see reference 456. Advances such as these should eventually make it possible to identify agents capable 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 of patients from viral infections. As discussed above, many antiviral agents have antiproliferative activity against rapidly dividing cells, and inhibition of host immune responses has always been a concern in antiviral therapy. A recent direct comparison of the effects of several antiviral agents on the proliferative responses of peripheral blood mononuclear cells from healthy individuals indicated that zidovudine, ganciclovir, and ribavirin decreased mitogenesis, while acyclovir and didanosine had no effect (192). Another study showed that zidovudine, didanosine, and ddc had no inhibitory effect on the bactericidal activity of polymorphonuclear leukocytes and possibly enhanced it (389). Further studies are needed, but these trials demonstrate the need for identifying the immunosuppressive properties of individual antiviral agents, establishing their importance in patients, and using this information to individualize patient therapy. PROSPECTS FOR THE FUTURE Liposomes Liposomes are synthetic phospholipid vesicles that can be used as carriers of many biologic molecules, including antitumor and antimicrobial agents, immunomodulators, toxins, and enzymes. When administered to animals or humans, they are naturally distributed to the reticuloendothelial organs and cleared by phagocytic cells, especially blood mononuclear cells and tissue macrophages. Liposomes can also be targeted to specific cells by incorporating the appropriate antibodies into them. For these reasons,

21 166 BEAN they are ideal agents for delivery of drugs to intracellular organisms such as viruses. Although no patient trials of antiviral therapy have yet been reported, animal and in vitro studies are intriguing and encouraging. Liposomes have been used to increase the cellular permeability of highly ionized drugs such as foscarnet (468) and to lengthen the half-life of ddc in the central nervous system of rats (237). HSV-infected cells can be selectively destroyed by liposomes encapsulated with acyclovir and tagged with anti- HSV antibodies (324) or by mononuclear phagocytes activated by liposomes encapsulated with immunomodulators such as gamma interferon or macrophage-activating factor (243). Of importance, CD4-bearing liposomes interact with HIV-infected lymphocytes and deliver their contents to the interior of these cells (87). This raises the possibility of loading these liposomes with antiviral agents such as reverse transcriptase inhibitors and using them to deliver the drugs to the interior of HIV-infected cells. HIV-1 also fuses directly with negatively charged liposomes, and its infectivity is thereby decreased (244), another promising approach to treatment of HIV. Combination Therapy Prompted by the success of combination chemotherapy for cancer and stimulated by the need for effective treatment of HIV infection, combination therapy has come to the forefront of antiviral treatment. The purpose is threefold: to enhance the efficacy of single-agent therapy, to minimize toxicity by reducing individual drug dosages, and to prevent or forestall the development of drug resistance. There are many reports of drug combinations synergistic in vitro against HIV and other viruses. Many patient trials are also under way, but few have been completed and most data are preliminary. The most promising combinations consist of a nonnucleoside and a nucleoside inhibitor of HIV reverse transcriptase or of two drugs active at different sites of viral replication. Different toxicities are also very important. In AIDS patients with Kaposi sarcoma, for instance, zidovudine and alpha interferon together showed early evidence of suppressing antigenemia more effectively than either drug alone and, possibly, of delaying drug resistance (31, 148). ddc is a reverse transcriptase inhibitor like zidovudine, but it is phosphorylated by a different mechanism and has different toxicities. When ddc is administered with zidovudine in an alternating regimen, less toxicity is seen than with either agent alone at full doses (43, 351, 435). Very exciting results have been obtained by giving GM-CSF to patients who develop neutropenia on zidovudine-interferon regimens. GM-CSF rapidly alleviated the neutropenia and allowed patients to remain on this very effective regimen when they would otherwise have been unable to tolerate it (92, 408). One recent trial demonstrated what appears to be an additive effect of zidovudine and foscarnet in suppressing p24 antigenemia (225). Although it has no activity against retroviruses, acyclovir can be added to zidovudine to control concomitant HSV infections. If herpesviruses, especially CMV, can transactivate HIV or induce cellular receptors for it, as has been shown in vitro (95, 295; for a review, see reference 273), control of herpesvirus infections may become an important part of HIV management. Despite the enthusiasm for combination therapy, adverse interactions must be closely monitored, and prospective combinations of agents must be tested in vitro and in animals before they are given to patients. Ribavirin, for instance, is phosphorylated by the same enzymes as zidovudine and in vitro inhibits its CLIN. MICROBIOL. REV. effectiveness against HIV (11, 486). In vitro, when didanosine was added to zidovudine, the combination synergistically inhibited HIV, but at high concentrations it also additively inhibited bone marrow progenitor cells (111), indicating that careful dosage adjustment will be needed in treating patients with this combination. Computer-Aided Drug Design Traditionally, the search for antiviral agents has consisted of randomly screening compounds for viral inhibitory activity in cell culture, identifying a few, making structural modifications in them, and retesting for changes in activity. Computer-aided drug design uses X-ray crystallography to determine the three-dimensional structures of viral macromolecules and then employs computer simulation and sophisticated thermodynamic computations to study atomic interactions with other molecules and to delineate the structures of those with favorable binding characteristics (290). In this way, the effect of structural modifications can be determined and promising compounds can be identified before an investment is made in synthesizing and testing them. The first human virus to be studied in this fashion was the human rhinovirus. Its three-dimensional atomic structure was described by Rossmann and colleagues in 1985 (396). In 1986, the interaction of the virus with a known picornavirus inhibitor, WIN 51711, was described: the drug was bound within a deep depression on the virion surface and induced conformational changes that increased stability and prevented uncoating of the virus (150, 441). A series of compounds with similar bridging properties were subsequently synthesized and studied (13), and some of them are currently being tested in patients for the treatment of rhinovirus colds. The structure of the influenza virus hemagglutinin and its interaction with its cellular receptor, sialic acid, have also been described and suggest that agents that would interfere with attachment of the influenza virus to cells could be designed (498). It has been proposed that HIV has a surface similar to that of rhinoviruses and thus might also be inhibited at the uncoating step (395). DesJarlais and colleagues (104) have recently used the structure of HIV-1 protease and a computer-assisted searching program to identify and study potential inhibitors of this enzyme. In addition, the structure of HIV reverse transcriptase itself is being studied by X-ray crystallography in the hope of identifying specific inhibitory compounds (268). Computerassisted drug design is still in its infancy, and no antiviral agents have yet been designed de novo by this technique. Nevertheless, it is advancing rapidly and holds great promise for the efficient, selective identification of new drugs. Role of the Clinical Microbiology Laboratory Most antiviral agents in use have a narrow spectrum of activity, and many of them cause substantial toxicity at the usual dosages. Consequently, empiric patient therapy with antiviral agents is seldom possible, although it is a common practice with antibacterial agents. Identification of a specific viral pathogen is usually needed, and although antiviral susceptibility testing is currently only necessary in referral centers, the rising number of HIV-infected patients and the increasing use of antiviral agents will eventually demand that it be available in general and community hospitals as well. Recent advances in diagnostic methodology have made it possible to identify many common viral pathogens from patient specimens within a few hours to a few days (440).

22 VOL. 5, 1992 Standard cell culture is still necessary for identification of all possible pathogens if no specific virus is suspected of causing a given illness. When only a single or a small number of viruses is implicated, however, immunofluorescent or enzyme immunoassays can frequently be performed directly on patient specimens and can detect the presence of viral antigens within a few hours. RSV is commonly identified in this way. In other cases, shell vials are used to allow a period of viral amplification before identification is attempted. The specimen is centrifuged onto a monolayer of cells grown on a coverslip in the bottom of a 1-dram (3.7-ml; shell) vial. Nutrient medium is added and the vial is incubated, usually for 18 to 72 h. The coverslip is then removed, the cells are fixed, and, most commonly, a fluorescein-conjugated antibody is added to detect the presence of viral antigen in the cells. HSV, VZV, CMV, and influenza viruses, as well as some others, can be identified in 1 to 3 days by using this method. Almost no convenient, practical methods for antiviral susceptibility testing are available to the clinical laboratory. The plaque reduction assay, a method commonly used by the research laboratory to measure antiviral activity (86, 189), is time-consuming, cumbersome, and difficult to reproduce. In addition, it depends on the ability of the virus to grow in a given cell line and thus may select virus populations capable of replicating well in vitro but not necessarily representative of the population infecting the patient. Many other methods have been used to quantitate viral replication in the presence of inhibitory agents including nucleic acid hybridization (160), shell vials with immunofluorescent staining (470), immunoperoxidase staining (211), enzyme immunoassay (366), dye uptake assay (299), and yield reduction assay (360). None of these methods has been standardized among laboratories, however. One method of nucleic acid hybridization for HSV susceptibility has been made commercially available in kit form. It compares well with the plaque reduction assay and involves wicking of lysed, virusinfected cells onto membranes followed by hybridization with a radioiodinated DNA probe (465). Although this method is more adaptable to clinical laboratories than many others and is in use in some, 3 to 4 days are required for susceptibility testing after the virus is isolated in cell culture and radioisotopes, frequently a disposal problem for microbiology laboratories, are still employed. As the demand for antiviral susceptibility testing grows, it is hoped that practical 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 our understanding of viral replication increases, more and more viral processes are being identified as possible targets for inhibition by antiviral compounds. Immunomodulators are being used increasingly to augment the immune response in the treatment of viral diseases. Resistance to antiviral drugs has been noted among diverse groups of viruses but thus far has caused treatment failure primarily in highly immunosuppressed patients, such as those with AIDS. It is hoped that combination antiviral therapy, in addition to improving the effectiveness of individual drugs, will limit the impact of resistance on patient care. Other new approaches, such as the use of liposomes to deliver antiviral drugs intracellularly and the use of computers to identify potential new agents rapidly and inexpensively, should result in increasingly safe ANTIVIRAL THERAPY 167 and effective antiviral drugs. With advances in laboratory technology making it possible to quickly identify viruses and, in the near future, perform susceptibility tests on them, antiviral therapy will rapidly become a routine part of medical care. ACKNOWLEDGMENTS I am indebted to Doris Hart and Sherry Hall for manuscript preparation; to Bruce Coary, Lourdes Aquino, and Eutiquio Chavez for 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 Initial studies on the cellular pharmacology of 2',3'-dideoxyinosine, an inhibitor of HIV infectivity. Biochem. Pharmacol. 36: Ahluwalia, G., M. A. Johnson, A. Fridland, D. A. Cooney, S. Broder, and D. G. Johns Cellular pharmacology of the anti-hiv agent 2',3'-dideoxyadenosine. Proc. Am. Assoc. Cancer Res. 29: Andersson, J., S. Britton, I. Ernberg, U. Andersson, W. Henle, B. Skoldenberg, and A. Tisell Effect of acyclovir on infectious mononucleosis: double-blind, placebo-controlled study. J. Infect. Dis. 153: Apperley, J. F., R. E. Marcus, J. M. Goldman, D. G. Wardle, P. J. Gravett, and A. Chanas Foscarnet for cytomegalovirus pneumonitis. Lancet i: Arvin, A. M., J. H. Kushner, S. Feldman, R. L. Baehner, D. Hammond, and T. C. Merigan Human leukocyte interferon for the treatment of varicella in children with cancer. N. Engl. J. Med. 306: Ashorn, P., B. Moss, J. N. Weinstein, V. K. Chaudhary, D. J. FitzGerald, I. Pastan, and E. A. Berger Elimination of infectious human immunodeficiency virus from human T-cell cultures by synergistic action of CD4-pseudomonas exotoxin and reverse transcriptase inhibitors. Proc. Natl. Acad. Sci. USA 87: Astra Pharmaceutical Products, Inc Foscavir. Package insert. Astra Pharmaceutical Products, Inc., Westborough, Mass. 8. Avramis, V. I., W. Markson, R. L. Jackson, and E. Gomperts Biochemical pharmacology of zidovudine in human T-lymphoblastoid cells (CEM). AIDS 3: Aweeka, F., J. Gambertoglio, J. Mills, and M. A. Jacobson Pharmacokinetics of intermittently administered intravenous foscarnet in the treatment of acquired immunodeficiency syndrome patients with serious cytomegalovirus retinitis. Antimicrob. Agents Chemother. 33: 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 Anti-human immunodeficiency virus type I activities and pharmacokinetics of novel 6-substituted acyclouridine derivatives. Antimicrob. Agents Chemother. 34: Baba, M., R. Pauwels, J. Balzarini, P. Herdewijn, E. DeClercq, and J. Desmyter Ribavirin antagonizes inhibitory effects of pyrimidine 2',3'-dideoxynucleosides but enhances inhibitory effects of purine 2',3'-dideoxynucleosides on replication of human immunodeficiency virus in vitro. Antimicrob. Agents Chemother. 31: Baba, M., H. Tanaka, E. DeClercq, R. Pauwels, J. Balzarini, D. Schols, H. Nakashima, C. F. Perno, R. T. Walker, and T. Miyasaka Highly specific inhibition of human immunodeficiency virus type I by a novel 6-substituted acyclouridine derivative. Biochem. Biophys, Res. Commun. 165: 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.

23 168 BEAN Dutko, M. Fancher, R. R. Rueckert, and B. A. Heinz Structural analysis of a series of antiviral agents complexed with human rhinovirus 14. Proc. Natl. Acad. Sci. USA 85: Baldwin, G. C., J. C. Gasson, S. G. Quan, J. Fleischmann, R. Weisbart, D. Oette, R. T. Mitsuyasu, and D. W. Golde Granulocyte-macrophage colony-stimulating factor enhances neutrophil function in acquired immunodeficiency syndrome patients. Proc. Natl. Acad. Sci. USA 85: 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 Collaborative Acyclovir Study Group Acyclovir halts progression of herpes zoster in immunocompromised patients. N. Engl. J. Med. 308: Balfour, H. H., B. Bean, C. D. Mitchell, G. W. Sachs, J. R. Boen, and C. K. Edelman Acyclovir in immunocompromised patients with cytomegalovirus disease. Am. J. Med. 73(Suppl. 1A): Balfour, H. H., B. A. Chace, J. T. Stapleton, R. L. Simmons, and D. S. Fryd A randomized, placebo-controlled trial of oral acyclovir for the prevention of cytomegalovirus disease in recipients of renal allografts. N. Engl. J. Med. 320: Balfour, H. H., J. M. Kelly, C. S. Suarez, R. C. Heussner, J. A. Englund, D. D. Crane, P. V. McGuirt, A. F. Clemmer, and D. M. Aeppli Acyclovir treatment of varicella in otherwise healthy children. J. Pediatr. 116: Barry, D. W., S. Nusinoff-Lehrman, M. N. Ellis, K. K. Biron, and P. A. Furman Viral resistance, clinical experience. Scand. J. Infect. Dis. 47: Bauer, D. J., L. St. Vincent, C. H. Kempe, and A. W. Downie Prophylactic treatment of smallpox contacts with N-methylisatin beta-thiosemicarbazone. Lancet ii: Beall, G., S. Kruger, F. Morales, D. Imagawa, J. A. Goldsmith, D. Fisher, J. Steinberg, J. Phair, S. Whaling, and J. Bitran A double-blind, placebo-controlled trial of thymostimulin in symptomatic HIV-infected patients. AIDS 4: Bean, B., and D. Aeppli Adverse effects of high-dose intravenous acyclovir in ambulatory patients with acute herpes zoster. J. Infect. Dis. 151: Bean, B., C. Braun, and H. H. Balfour Acyclovir therapy for acute herpes zoster. Lancet ii: Bean, B., C. Fletcher, J. Englund, S. NusinoffLehrman, and M. Ellis Progressive mucocutaneous herpes simplex infection due to acyclovir-resistant virus in an immunocompromised patient: correlation of viral susceptibilities and plasma levels with response to therapy. Diagn. Microbiol. Infect. Dis. 7: Bean, W. J., S. C. Threlkeld, and R. G. Webster Biologic potential of amantadine-resistant influenza A virus in an avian model. J. Infect. Dis. 159: Beasley, R. P., C. C. Lin, K. Y. Wang, F. J. Hsieh, L. Y. Hwang, C. E. Stevens, T. S. Sun, and W. Szmuness Hepatitis B immune globulin (HBIG) efficacy in the interruption of perinatal transmission of hepatitis B virus carrier state. Lancet ii: Belshe, R. B., B. Burk, F. Newman, R. L. Cerruti, and I. S. Sim Resistance of influenza A virus to amantadine and rimantadine: results of one decade of surveillance. J. Infect. Dis. 159: Belshe, R. B., and A. J. Hay Drug resistance and mechanisms of action on influenza A viruses. J. Respir. Dis. 10(Suppl.):S52-S Belshe, R. B., M. H. Smith, C. B. Hall, R. Betts, and A. J. Hay Genetic basis of resistance to rimantadine emerging during treatment of influenza virus infection. J. Virol. 62: Bergdahl, S., A. Sonnerborg, A. Larsson, and 0. I. Strannegard Declining levels of HIV P24 antigen in serum during treatment with foscarnet. Lancet i: Berglund, O., K. Engman, A. Ehrnst, J. Andersson, K. Lidman, B. Akerlund, A. Sonnerborg, and 0. Strannegard Combined treatment of symptomatic human immunodeficiency CLIN. MICROBIOL. REV. virus type I infection with native interferon-alfa and zidovudine. J. Infect. Dis. 163: Berkman, S. A., M. L. Lee, and R. P. Gale Clinical uses of intravenous immunoglobulins. Ann. Intern. Med. 112: Bernstein, D. I., P. D. Reuman, J. R. Sherwood, E. C. Young, and G. M. Schiff Ribavirin small-particle-aerosol treatment of influenza B virus infection. Antimicrob. Agents Chemother. 32: Birch, C. J., G. Tachedjian, R. R. Doherty, K. Hayes, and I. D. Gust Altered sensitivity to antiviral drugs of herpes simplex virus isolates from a patient with the acquired immunodeficiency syndrome. J. Infect. Dis. 162: Biron, K. K., and G. B. Elion In vitro susceptibility of varicella-zoster virus to acyclovir. Antimicrob. Agents. Chemother. 18: Biron, K. K., J. A. Fyfe, S. C. Stanat, L. K. Leslie, J. B. Sorrell, C. U. Lambe, and D. M. Coen A human cytomegalovirus mutant resistant to the nucleoside analog 9-(2-hydroxy-1-(hydroxymethyl)ethoxy)methyl)guanine (BW B759U) induces reduced levels of BW B759U triphosphate. Proc. Natl. Acad. Sci. USA 83: Biron, K. K., S. C. Stanat, J. B. Sorrell, J. A. Fyfe, P. M. Keller, C. U. Lambe, and D. J. Nelson Metabolic activation of the nucleoside analog 9-(2-hydroxy-1-(hydroxymethyl)ethoxy)methyl)guanine in human diploid fibroblasts infected with human cytomegalovirus. Proc. Natl. Acad. Sci. USA 82: Blum, M. R., S. H. T. Liao, S. S. Good, and P. de Miranda Pharmakokinetics and bioavailability of zidovudine in humans. Am. J. Med. 85(Suppl. 2A): Bodsworth, N., and D. A. Cooper Ribavirin: a role in HIV infection? J. Acquired Immune Defic. Syndr. 3: Boston Interhospital Virus Study Group and the NIAID-Sponsored Cooperative Antiviral Clinical Study Failure of high dose 5-iodo-2'-deoxyuridine in the therapy of herpes simplex virus encephalitis. Evidence of unacceptable toxicity. N. Engl. J. Med. 292: Bowden, R. A., L. D. Fisher, K. Rogers, M. Cays, and J. D. Meyers Cytomegalovirus (CMV)-specific intravenous immunoglobulin for the prevention of primary CMV infection and disease after marrow transplant. J. Infect. Dis. 164: Bowden, R. A., M. Sayers, N. Flournoy, B. Newton, M. Banaji, E. D. Thomas, and J. D. Meyers Cytomegalovirus immune globulin and seronegative blood products to prevent primary cytomegalovirus infection after marrow transplantation. N. Engl. J. Med. 314: Bozzette, S. A., and D. D. Richman Salvage therapy for zidovudine-intolerant HIV-infected patients with alternating and intermittent regimens of zidovudine and dideoxycytidine. Am. J. Med. 88(Suppl. 5B):24S-26S. 44. Brink, J. J., and G. A. LePage Metabolic effects of 9-D-arabinosylpurines in ascites tumor cells. Cancer Res. 24: Brown, Z. A., and D. A. Baker Acyclovir therapy during pregnancy. Obstet. Gynecol. 73: 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 Treatment of first episodes of genital herpes simplex virus infection with oral acyclovir. N. Engl. J. Med. 308: Buhles, W. C., B. J. Mastre, A. J. Tinker, V. Strand, S. H. Koretz, and the Syntex Collaborative Ganciclovir Treatment Study Group Ganciclovir treatment of life- or sightthreatening cytomegalovirus infection: experience in 314 immunocompromised patients. Rev. Infect. Dis. 10(Suppl. 3): S495-S Bukrinskaya, A. G., N. K. Vorkunova, and R. A. Narmanbetova Rimantadine hydrochloride blocks the second step of influenza virus uncoating. Arch. Virol. 66: Burns, W. H., R. Saral, G. W. Santos, 0. L. Laskin, P. S. Lietman, C. McLaren, and D. W. Barry Isolation and

24 VOL. 5, 1992 characterization of resistant herpes simplex virus after acyclovir therapy. Lancet i: 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, and P. A. Pizzo Dideoxyinosine in children with symptomatic human immunodeficiency virus infection. N. Engl. J. Med. 324: 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 Biological properties of a CD4 immunoadhesin. Nature (London) 344: Canonico, P. G., M. Kende, and J. W. Huggins The toxicology and pharmacology of ribavirin in experimental animals, p 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 Designing CD4 immunoadhesins for AIDS therapy. Nature (London) 337: Cassidy, L. F., and J. L. Patterson Mechanism of La Crosse virus inhibition by ribavirin. Antimicrob. Agents Chemother. 33: Centers for Disease Control Rabies prevention-united States, Morbid. Mortal. Weekly Rep. 33: Centers for Disease Control Varicella-zoster immune globulin for the prevention of chickenpox: recommendations of the immunization practices advisory committee. Ann. Intern. Med. 100: Centers for Disease Control Assessing exposures of health-care personnel to aerosols of ribavirin-california. Morbid. Mortal. Weekly Rep. 37: Centers for Disease Control Management of patients with suspected viral hemorrhagic fever. Morbid. Mortal. Weekly Rep. 37(S-3): Centers for Disease Control Measles prevention: recommendations of the immunization practices advisory committee (ACIP). Morbid. Mortal. Weekly Rep. 38(S-9): Centers for Disease Control Prevention and control of influenza. Morbid. Mortal. Weekly Rep. 39(RR-7): Centers for Disease Control Protection against viral hepatitis. Morbid. Mortal. Weekly Rep. 39(RR-2): Chachoua, A., D. Dieterich, K. Krasinski, J. Greene, L. Laubenstein, J. Wernz, W. Buhles, and S. Koretz (1,3- dihydroxy-2-propoxymethyl)guanine (ganciclovir) in the treatment of cytomegalovirus gastrointestinal disease with the acquired immunodeficiency syndrome. Ann. Inter. Med. 107: Chatis, P. A., C. H. Miller, L. E. Schrager, and C. S. Crumpacker Successful treatment with foscarnet of an acyclovir-resistant mucocutaneous infection with herpes simplex virus in a patient with acquired immunodeficiency syndrome. N. Engl. J. Med. 320: Chaudhary, V. K., T. Mizukami, T. R. Fuerst, D. J. FitzGeraid, B. Moss, I. Pastan, and E. A. Berger Selective killing of HIV-infected cells by recombinant human CD4-Pseudomonas exotoxin hybrid protein. Nature (London) 335: Chen, Y.-C., S. P. Grill, G. E. Dutschman, K. Nakayama, and K. F. Bastow Metabolism of 9-(1,3-dihydroxy-2- propoxymethyl-guanine, a new anti-herpes virus compound, in herpes simplex virus-infected cells. J. Biol. Chem. 258: Cheng, Y., G. E. Dutschman, K. F. Bastow, M. G. Sarngadharan, and R. Y. C. Ting Human immunodeficiency virus reverse transcriptase. J. Biol. Chem. 262: Chu, C. K., R. F. Schinazi, M. K. Ahn, G. V. Ullas, and Z. P. Gu Structure-activity relationships of pyrimidine nucleosides as antiviral agents for human immunodeficiency virus type I in peripheral blood mononuclear cells. J. Med. Chem. 32: ANTIVIRAL THERAPY Clover, R. D., S. A. Crawford, T. D. Abell, C. N. Ramsey, Jr., W. P. Glezen, and R. B. Couch Effectiveness of rimantadine prophylaxis of children within families. Am. J. Dis. Child. 140: Cobo, L. M., G. N. Foulks, T. Liesegang, J. Lass, J. Sutphin, K. Wilhelmus, D. B. Jones, S. Chapman, and A. Segreti Oral acyclovir in the therapy of acute herpes zoster ophthalmicus. Ophthalmology 92: Cobo, L. M., G. N. Foulks, T. Liesegang, J. Lass, J. E. Sutphin, K. Wilhelmus, D. B. Jones, S. Chapman, A. C. Segreti, and D. H. King Oral acyclovir in the treatment of acute herpes zoster ophthalmicus. Ophthalmology 93: Coen, D. M General aspects of virus drug resistance with special reference to herpes simplex virus. J. Antimicrob. Chemother. 18(Suppl. B): Coen, D. M., and P. A. Schaffer Two distinct loci confer resistance to acycloguanosine in herpes simplex virus type 1. Proc. Natl. Acad. Sci. USA 77: Colby, B. M., J. E. Shaw, G. B. Elion, and J. S. Pagano Effect of acyclovir [9-(2-hydroxyethoxymethyl) guanine] on Epstein-Barr virus DNA replication. J. Virol. 34: Collaborative DHPG Treatment Study Group Treatment of serious cytomegalovirus infections with 9-(1,3-dihydroxy-2- propoxymethyl)guanine in patients with AIDS and other immunodeficiencies. N. Engl. J. Med. 314: 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 A pilot study of low-dose zidovudine in human immunodeficiency virus infection. N. Engl. J. Med. 373: Collins, P., B. A. Larder. N. M. Oliver, S. Kemp, I. W. Smith, and G. Darby Characterization of a DNA polymerase mutant of herpes simplex virus from a severely immunocompromised patient receiving acyclovir. J. Gen. Virol. 70: Cooley, T. P., L. M. Kunches, C. A. Saunders, J. K. Ritter, C. J. Perkins, C. McLaren, R. P. McCaffrey, and H. A. Liebman Once-daily administration of 2',3'-dideoxyinosine (ddl) in patients with the acquired immunodeficiency syndrome or AIDS-related complex. N. Engl. J. Med. 322: Cooney, D. A., G. Ahluwalia, H. Mitsuya, A. Fridland, M. Johnson, Z. Hao, M. Dalal, J. Balzarini, S. Broder, and D. G. Johns Initial studies on the cellular pharmacology of 2',3'-dideoxyadenosine, an inhibitor of HTLV-III infectivity. Biochem. Pharmacol. 36: Corey, L., J. Benedetti, C. Critchlow, G. Mertz, J. Douglas, K. Fife, A. Fahnlander, M. L. Remington, C. Winter, and J. Dragavon Treatment of primary first-episode genital herpes simplex virus infections with acyclovir: results of topical, intravenous and oral therapy. J. Antimicrob. Chemother. 12(Suppl. B): Corey, L., A. J. Nahmias, M. E. Guinan, J. K. Benedetti, C. W. Critchlow, and K. K. Holmes A trial of topical acyclovir in genital herpes simplex virus infections. N. Engl. J. Med. 306: Creagh-Kirk, T., P. Doi, E. Andrews, S. Nusinoff-Lehrman, H. Tilson, D. Hoth, and D. W. Barry Survival experience among patients with AIDS receiving zidovudine. JAMA 260: Crumpacker, C., B. Glenn, D. Lucey, S. Hussey, and J. Connor Ribavirin enters cerebrospinal fluid. Lancet ii: Crumpacker, C., S. Marlowe, J. L. Zhang, S. Abrams, P. Watkins, and the Ganciclovir Bone Marrow Transplant Treatment Group Treatment of cytomegalovirus pneumonia. Rev. Infect. Dis. 10(Suppl. 3):S538-S Crumpacker, C. S., P. N. Kowalsky, S. A. Oliver, L. E. Schnipper, and A. K. Field Resistance of herpes simplex virus to 9-(2-hydroxy-1-hydroxymethyl)ethoxy)methyl)guanine: physical mapping of drug synergism within the viral DNA polymerase locus. Proc. Natl. Acad. Sci. USA 81: Crumpacker, C. S., L. E. Schnipper, S. I. Marlowe, P. N.

25 170 BEAN Kowalsky, B. J. Hershey, and M. J. Levin Resistance to antiviral drugs of herpes simplex virus isolated from a patient treated with acyclovir. N. Engl. J. Med. 306: Crumpacker, C. S., L. E. Schnipper, J. A. Zaia, and M. J. Levin Growth inhibition by acycloguanosine of herpesviruses isolated from human infections. Antimicrob. Agents Chemother. 15: Cudd, A., C. A. Noonan, P. F. Tosi, J. L. Melnick, and C. Nicolau Specific interaction of CD4-bearing liposomes with HIV-infected cells. J. Acquired Immune Defic. Syndr. 3: Dalakas, M. C., I. Ilia, G. H. Pezeshkpour, J. P. Laukaitis, B. Cohen, and J. L. Griffin Mitochondrial myopathy caused by long-term zidovudine therapy. N. Engl. J. Med. 322: Daniels, R. S., J. C. Downie, A. J. Hay, M. Knossow, J. J. Skehel, M. L. Wang, and D. C. Wiley Fusion mutants of the influenza virus hemagglutinin glycoprotein. Cell 40: Darby, G., H. J. Field, and S. A. Salisbury Altered substrate specificity of herpes simplex virus thymidine kinase confers acyclovir-resistance. Nature (London) 289: Datta, A. K., B. M. Colby, J. E. Shaw, and J. S. Pagano Acyclovir inhibition of Epstein-Barr Virus replication. Proc. Natl. Acad. Sci. USA 77: 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, and H. C. Lane A phase I/II trial of zidovudine, interferonalpha, and granulocyte-macrophage colony-stimulating factor in the treatment of human immunodeficiency virus type I infection. J. Infect. Dis. 164: 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 Treatment of chronic hepatitis C with recombinant interferon alfa. N. Engl. J. Med. 321: Davis, G. L., and J. H. Hoofnagle Interferon in viral hepatitis: role in pathogenesis and treatment. Hepatol. 6: Davis, M. G., S. C. Kenney, J. Kamine, J. S. Pagano, and E. S. Huang Immediate-early gene region of human cytomegalovirus trans-activates the promoter of human immunodeficiency virus. Proc. Natl. Acad. Sci. USA 84: DeArmond, B Future directions in the management of cytomegalovirus infections. J. Acquired Immune Defic. Syndr. 4(Suppl. I):S53-S DeClercq, E Basic approaches to anti-retroviral treatment. J. Acquired Immune Defic. Syndr. 4: DeClercq, E., M. Cools, J. Balzarini, R. Snoeck, G. Andrei, M. Hosoya, S. Shigeta, T. Ueda, N. Minakawa, and A. Matsuda Antiviral activities of 5-ethynyl-1-13-D-ribofuranosylimidazole-4-carboxamide and related compounds. Antimicrob. Agents Chemother. 35: DeClercq, E., J. Descamps, G. Verhelst, R. T. Walker, A. S. Jones, P. F. Torrence, and D. Shugar Comparative efficacy of antiherpes drugs against different strains of herpes simplex virus. J. Infect. Dis. 141: De Miranda, P., and M. R. Blum Pharmacokinetics of acyclovir after intravenous and oral administration. J. Antimicrob. Chemother. 12(Suppl. B): Deray, G., F. Martinez, C. Katlama, B. Levaltier, H. Beaufils, M. Danis, M. Rozenheim, A. Baumelou, E. Dohin, M. Gentilini, and C. Jacobs Foscarnet nephrotoxicity: mechanism, incidence and prevention. Am. J. Nephrol. 9: Derse, D., K. F. Bastow, and Y.-C. Cheng Characterization of the DNA polymerases induced by a group of herpes simplex virus type I variants selected for growth in the presence of phosphonoformic acid. J. Biol. Chem. 257: Derse, D., Y.-C. Cheng, P. A. Furman, M. H. St. Clair, and G. B. Elion Inhibition of purified human and herpes CLIN. MICROBIOL. REV. simplex virus-induced DNA polymerases by 9-(2-hydroxyethoxymethyl) guanine triphosphate. J. Biol. Chem. 256: 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 Structure-based design of nonpeptide inhibitors specific for the human immunodeficiency virus I protease. Proc. Natl. Acad. Sci. USA 87: DeWit, R., C. A. B. Boucher, K. H. N. Veenhof, J. K. M. E. Schattenkerk, P. J. M. Bakker, and S. A. Danner Clinical and virological effects of high-dose recombinant interferonalpha in disseminated AIDS-related Kaposi's sarcoma. Lancet ii: Di Bisceglie, A. M., and J. H. Hoofnagle Therapy of chronic hepatitis C with alpha-interferon: the answer? Or more questions? Hepatology 13: Di Bisceglie, A. M., P. Martin, C. Kassianides, M. Lisker- Melman, L. Murray, J. Waggoner, Z. Goodman, S. M. Banks, and J. H. Hoofnagle Recombinant interferon alfa therapy for chronic hepatitis C. N. Engl. J. Med. 321: Dicioccio, R. A., and B. I. S. Srivastava Kinetics of inhibition of deoxynucleotide-polymerizing enzyme activities from normal and leukemic human cells by 9-beta-D-arabinofuranosyladenine 5'-triphosphate and 1-beta-D-arabinofuranosylcytosine 5'-triphosphate. Eur. J. Biochem. 79: Dieterich, D. T., A. Chachoua, F. Lafleur, and C. Worrell Ganciclovir treatment of gastrointestinal infections caused by cytomegalovirus in patients with AIDS. Rev. Infect. Dis. 10(Suppl. 3):S532-S Dolin, R., R. C. Reichman, H. P. Madore, R. Maynard, P. N. Linton, and J. Webber-Jones A controlled trial of amantadine and rimantadine in the prophylaxis of influenza A infection. N. Engl. J. Med. 307: Dornsife, R. E., M. H. St. Clair, A. T. Huang, T. J. Panella, G. W. Koszalka, C. L. Burns, and D. R. Averett Antihuman immunodeficiency virus synergism by zidovudine (3'- azidothymidine) and didanosine (dideoxyinosine) contrasts with their additive inhibition of normal human marrow progenitor cells. Antimicrob. Agents Chemother. 35: 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 A double-blind study of oral acyclovir for suppression of recurrences of genital herpes simplex virus infection. N. Engl. J. Med. 310: Douglas, J. M., L. J. Eron, F. N. Judson, M. Rogers, M. B. Alder, E. Taylor, D. Tanner, and E. Peets A randomized trial of combination therapy with intralesional interferon alpha 2b and podophyllin versus podophyllin alone for the therapy of anogenital warts. J. Infect. Dis. 162: Douglas, R. G., Jr Prophylaxis and treatment of influenza. N. Engl. J. Med. 322: 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 Prophylactic efficacy of intranasal alpha2- interferon against rhinovirus infections in the family setting. N. Engl. J. Med. 314: Dournon, E., W. Rozenbaum, C. Michon, C. Perronne, P. DeTruchis, E. Bouvet, M. Levacher, S. Matheron, S. Gharakhanian, P. M. Girard, D. Salmon, C. Leport, M. C. Dazza, B. Regnier, and the Claude Bernard Hospital AZT Study Group Effects of zidovudine in 365 consecutive patients with AIDS or AIDS-related complex. Lancet ii: Drach, J. C Targets for the design of antiviral agents, p In E. DeClercq and R. T. Walker (ed.), Purine nucleoside analogs as antiviral agents. Plenum Press, New York 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 Prevalence of resistance in patients receiving ganciclovir for serious cytomegalovirus infection. J. Infect. Dis. 163: 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.

26 VOL. 5, 1992 Fletcher, R. M. Bolman, A. J. Matas, W. D. Payne, D. E. R. Sutherland, and J. S. Najarian Treatment of invasive cytomegalovirus disease in solid organ transplant patients with ganciclovir. Transplantation 51: Dwyer, J. M., and K. Erlendsson Intraventricular gamma-globulin for the management of enterovirus encephalitis. Pediatr. Infect. Dis. J. 7:S30-S Efstathiou, S., S. Kemp, G. Darby, and A. C. Minson The role of herpes simplex virus type I thymidine kinase in pathogenesis. J. Gen. Virol. 70: Elion, G. B., P. A. Furman, J. A. Fyfe, P. de Miranda, L. Beauchamp, and H. J. Schaeffer Selectivity of action of an antiherpetic agent, 9-(2-hydroxyethoxymethyl) guanine. Proc. Natl. Acad. Sci. USA 74: 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 Clinical isolate of herpes simplex virus type 2 that induces a thymidine kinase with altered substrate specificity. Antimicrob. Agents Chemother. 31: 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 Antibacterial activity and mechanism of action of 3'-azido-3'-deoxythymidine (BW A509U). Antimicrob. Agents Chemother. 31: 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 Cytomegalovirus pneumonia after bone marrow transplantation successfully treated with the combination of ganciclovir and high-dose intravenous immune globulin. Ann. Intern. Med. 109: Englund, J. A., M. E. Zimmerman, E. M. Swierkosz, J. L. Goodman, D. R. Scholl, and H. H. Balfour Herpes simplex virus resistant to acyclovir. A study in a tertiary care center. Ann. Intern. Med. 112: Erice, A., S. Chou, K. K. Biron, S. C. Stanat, H. H. Balfour, and M. Jordan Progressive disease due to ganciclovirresistant cytomegalovirus in immunocompromised patients. N. Engl. J. Med. 320: 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 Design, activity, and 2.8 A crystal structure of a C2 symmetric inhibitor complexed to HIV-I protease. Science 249: 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 Inhibition of influenza virus ribonucleic acid polymerase by ribavirin triphosphate. Antimicrob. Agents. Chemother. 11: Erlich, K. S., M. A. Jacobson, J. E. Koehler, S. E. Follansbee, D. P. Drennan, L. Gooze, S. Safrin, and J. Mills Foscarnet therapy for severe acyclovir-resistant herpes simplex virus type-2 infections in patients with the acquired immunodeficiency syndrome (AIDS). Ann. Intern. Med. 110: Erlich, K. S., J. Mills, P. Chatis, G. J. Mertz, D. F. Busch, S. E. Follansbee, R. M. Grant, and C. S. Crumpacker Acyclovir-resistant herpes simplex virus infections in patients with the acquired immunodeficiency syndrome. N. Engl. J. Med. 320: 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 Interferon therapy for condylomata acuminata. N. Engl. J. Med. 315: Estes, J. E., and E.-S. Huang Stimulation of cellular thymidine kinases by human cytomegalovirus. J. Virol. 24: 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 Foscarnet therapy of cytomegalovirus retinitis in AIDS. ANTIVIRAL THERAPY 171 J. Acquired Immune Defic. Syndr. 3: Fauci, A. S., S. M. Schnittman, G. Poli, S. Koenig, and G. Pantaleo Immunopathogenic mechanisms in human immunodeficiency virus (HIV) infection. Ann. Intern. Med. 114: Fegueux, S., D. Salmon, C. Picard, C. Sauvage, P. Longuet, and T. Desmoulins Penile ulcerations with foscarnet. Lancet 335: Feldman, S., J. Rodman, and B. Gregory Excessive serum concentrations of acyclovir and neurotoxicity. J. Infect. Dis. 157: Felsenstein, D., D. J. D'amico, M. S. Hirsch, D. A. Neumeyer, D. M. Cederberg, P. demiranda, and R. T. Schooley Treatment of cytomegalovirus retinitis with 9-(2-hydroxy-1- (hydroxymethyl) ethoxymethyl)guanine. Ann. Intern. Med. 103: 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 (2-Hydroxy-1-(hydroxymethyl)ethoxy)methyl)guanine: a selective inhibitor of herpes group virus replication. Proc. Natl. Acad. Sci. USA 80: Field, H. J Persistent herpes simplex virus infection and mechanisms of virus drug resistance. Eur. J. Clin. Microbiol. Infect. Dis. 8: Field, H. J., and D. M. Coen Pathogenicity of herpes simplex virus mutants containing drug resistance mutations in the viral DNA polymerase gene. J. Virol. 60: Field, H. J., and G. Darby Pathogenicity in mice of strains of herpes simplex virus which are resistant to acyclovir in vitro and in vivo. Antimicrob. Agents Chemother. 17: Field, H. J., and P. Wildy The pathogenicity of thymidine kinase-deficient mutants of herpes simplex virus in mice. J. Hyg. Camb. 81: 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 Clinical Trials Group A randomized controlled trial of a reduced daily dose of zidovudine in patients with the acquired immunodeficiency syndrome. N. Engl. J. Med. 323: 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 AZT Collaborative Working Group Prolonged zidovudine therapy in patients with AIDS and advanced AIDS-related complex. JAMA 262: Fischl, M. A., D. D. Richman, M. H. Grieco, M. S. Gottlieb, P. A. Volberding, 0. L. Laskin, J. M. Leedom, J. E. Groopman, D. Mildvan, R. T. Schooley, G. G. Jackson, D. T. Durack, D. King, and the AZT Collaborative Working Group The efficacy of azidothymidine (AZT) in the treatment of patients with AIDS and AIDS-related complex. N. Engl. J. Med. 317: 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 Clinical Trials Group The safety and efficacy of zidovudine (AZT) in the treatment of subjects with mildly symptomatic human immunodeficiency virus type I (HIV) infection. Ann. Intern. Med. 112: 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 A phase I study of recombinant human interferon-alpha2a or human lymphoblastoid interferonalphan, and concomitant zidovudine in patients with AIDSrelated Kaposi's sarcoma. J. Acquired Immune Defic. Syndr. 4: Fisher, A. G., B. Ensoli, D. Looney, A. Rose, R. C. Gallo, M. S.

27 172 BEAN Saag, G. M. Shaw, B. H. Hahn, and F. Wong-Staal Biologically diverse molecular variants within a single HIV-I isolate. Nature (London) 334: Fox, M. P., M. J. Otto, and M. A. McKinlay Prevention of rhinovirus and poliovirus uncoating by WIN 51711, a new antiviral drug. Antimicrob. Agents Chemother. 30: Freitas, V. R., D. F. Smee, M. Chernow, R. Boehme, and T. R. Matthews Activity of 9-(1,3-dihydroxy-2-propoxymethyl)guanine compared with that of acyclovir against human, monkey, and rodent cytomegaloviruses. Antimicrob. Agents Chemother. 28: 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 Natural interferon alfa for treatment of condylomata acuminata. JAMA 259: Furman, P. A., and D. W. Barry Spectrum of antiviral activity and mechanism of action of zidovudine. Am. J. Med. 85(Suppl. 2A): Furman, P. A., D. M. Coen, M. H. St. Clair, and P. A. Schaffer Acyclovir-resistant mutants of herpes simplex virus type I express altered DNA polymerase or reduced acyclovir phosphorylating activities. J. Virol. 40: 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 Phosphorylation of 3'-azido-3'-deoxythymidine and selective interaction of the 5'-triphosphate with human immunodeficiency virus reverse transcriptase. Proc. Natl. Acad. Sci. USA 83: Furman, P. A., P. V. McGuirt, P. M. Keller, J. A. Fyfe, and G. B. Elion Inhibition by acyclovir of cell growth and DNA synthesis of cells biochemically transformed with herpesvirus genetic information. Virology 102: Furman, P. A., M. H. St. Clair, J. A. Fyfe, J. L. Rideout, P. M. Keller, and G. B. Elion Inhibition of herpes simplex virus-induced DNA polymerase activity and viral DNA replication by 9-(2-hydroxyethoxymethyl)guanine and its triphosphate. J. Virol. 32: Furman, P. A., M. H. St. Clair, and T. Spector Acyclovir triphosphate is a suicide inactivator of the herpes simplex virus DNA polymerase. J. Biol. Chem. 259: Fyfe, J. A., P. M. Keller, P. A. Furman, R. L. Miller, and G. B. Elion Thymidine kinase from herpes simplex virus phosphorylates the new antiviral compound, 9-(2-hydroxyethoxymethyl) guanine. J. Biol. Chem. 253: Gadler, H., A. Larsson, and E. Solver Nucleic acid hybridization, a method to determine effects of antiviral compounds on herpes simplex virus type 1 DNA synthesis. Antiviral Res. 4: Galbraith, A. W., J. S. Oxford, G. C. Schild, and G. I. Watson Protective effect of 1-adamantanamine hydrochloride on influenza A2 infections in the family environment. Lancet ii: Galpin, S., J. Martin, T. O'Connor, S. Redshaw, N. Roberts, and D. Kinchington Further studies on the HIV proteinase inhibitor RO Antiviral Res. 15(Suppl 1): Gangemi, J. D., M. Nachtigal, D. Barnhart, L. Krech, and P. Jani. Therapeutic efficacy of liposome-encapsulated ribavirin and muramyl tripeptide in experimental infection with influenza or herpes simplex virus. J. Infect. Dis. 155: Gately, A., R. M. Gander, P. C. Johnson, S. Kit, H. Otsuka, and S. Kohl Herpes simplex virus type 2 meningoencephalitis resistant to acyclovir in a patient with AIDS. J. Infect. Dis. 161: 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 Nature, time course and dose dependence of zidovudine-related side effects: results from the multicenter Canadian azidothymidine trial. AIDS 3: Gershon, A. A., S. Steinberg, and P. A. Brunell Zoster immune globulin. A further assessment. N. Engl. J. Med. 290: Gilbert, B. E., S. Z. Wilson, V. Knight, R. B. Couch, J. M. Quarles, L. Dure, N. Hayes, and G. Willis Ribavirin CLIN. MICROBIOL. REV. small-particle aerosol treatment of infections caused by influenza virus strains A/Victoria/7/83 (HlNl) and B/Texas/1/84. Antimicrob. Agents Chemother. 27: Gilquin, J., L. Weiss, and M. D. Kazatchkine Genital and oral erosions induced by foscarnet. Lancet 335: 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 Early treatment with ganciclovir to prevent cytomegalovirus disease after allogenic bone marrow transplantation. N. Engl. J. Med. 325: Goswami, B. B., J. Fujitaki, and R. A. Smith The broad spectrum antiviral agent ribavirin inhibits capping of mrna. Biochem. Biophys. Res. Commun. 89: 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 Response to treatment with the leukocyte-derived immunomodulator IMREG-1 in immunocompromised patients with AIDS-related complex. Ann. Intern. Med. 115: Greffe, B. S., S. L. Dooley, R. B. Deddish, and H. C. Krasny Transplacental passage of acyclovir J. Pediatr. 108: Groopman, J. E., R. T. Mitsuyasu, M. J. DeLeo, D. H. Oette, and D. W. Golde Effect of recombinant human granulocyte-macrophage colony-stimulating factor on myelopoiesis in the Acquired Immunodeficiency Syndrome. N. Engl. J. Med. 317: Grundy, J. E., J. D. Shanley, and P. D. Griffiths Is cytomegalovirus interstitial pneumonitis in transplant recipients an immunopathological condition? Lancet ii: Haake, D. A., P. C. Zakowski, D. L. Haake, and Y. J. Bryson Early treatment with acyclovir for varicella pneumonia in otherwise healthy adults: retrospective controlled study and review. Rev. Infect. Dis. 12: Hadler, S. C., J. J. Erben, D. Matthews, K. Starko, D. P. Francis, and J. E. Maynard Effect of immunoglobulin on hepatitis A in day-care centers. JAMA 249: 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 Children with influenza A infection: treatment with rimantadine. Pediatrics 80: Hall, C. B., J. T. McBride, C. L. Gala, S. W. Hildreth, and K. C. Schnabel Ribavirin treatment of respiratory syncytial viral infection in infants with underlying cardiopulmonary disease. JAMA 254: 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 Aerosolized ribavirin treatment of infants with respiratory syncytial viral infection. N. Engl. J. Med. 308: Harbison, M. A., P. C. DeGirolami, R. L. Jenkins, and S. M. Hammer Ganciclovir therapy of severe cytomegalovirus infections in solid-organ transplant recipients. Transplantation 46: Hardy, W. D Combined ganciclovir and recombinant human granulocyte-macrophage colony-stimulating factor the treatment of cytomegalovirus retinitis in AIDS patients. J. Acquired Immune Defic. Syndr. 4(Suppl. I):S22-S Hartman, N. R., R. Yarchoan, J. M. Pluda, R. V. Thomas, K. S. Marczyk, S. Broder, and D. G. Johns Pharmacokinetics of 2',3'-dideoxyadenosine and 2',3'-dideoxyinosine in patients with severe human immunodeficiency virus infection. Clin. Pharmacol. Ther. 47: Hartshorn, K. L., M. W. Vogt, T. Chou, R. S. Blumberg, R. Byington, R. T. Schooley, and M. S. Hirsch Synergistic inhibition of human immunodeficiency virus in vitro by azidothymidine and recombinant alpha interferon. Antimicrob. Agents Chemother. 31: Hay, A. J., A. J. Wolstenholme, J. J. Skehel, and M. H. Smith The molecular basis of the specific anti-influenza action of amantadine. EMBO J. 4: Hay, A. J., M. C. Zambon, A. J. Wolstenholme, J. J. Skehel, and M. H. Smith Molecular basis of resistance of in

28 VOL. 5, 1992 influenza A viruses to amantadine. J. Antimicrob. Chemother. 18(Suppl. B): Hayden, F. G Significance of rimantadine resistance in influenza A viruses. J. Respir. Dis. 10(Suppl.): Hayden, F. G., J. K. Albrecht, D. L. Kaiser, and J. M. Gwaltney Prevention of natural colds by contact prophylaxis with intranasal alpha2-interferon. N. Engl. J. Med. 314: Hayden, F. G., R. B. Beishe, R. D. Clover, A. J. Hay, M. G. Oakes, and W. Soo Emergence and apparent transmission of rimantadine-resistant influenza A virus in families. N. Engl. J. Med. 321: Hayden, F. G., K. M. Cote, and R. G. Douglas Plaque inhibition assay for drug susceptibility testing of influenza viruses. Antimicrob. Agents Chemother. 17: Hayden, F. G., D. L. Kaiser, and J. K. Albrecht Intranasal recombinant alfa-2b interferon treatment of naturally occurring common colds. Antimicrob. Agents Chemother. 32: Hayden, F. G., and A. S. Monto Oral rimantadine hydrochloride therapy of influenza A virus H3N2 subtype infection in adults. Antimicrob. Agents Chemother. 29: Heagy, W., C. Crumpacker, P. A. Lopez, and R. W. Finberg Inhibition of immune functions by antiviral drugs. J. Clin. Invest. 87: Hecht, D. W., D. R. Snydman, C. S. Crumpacker, B. G. Werner, B. Heinze-Lacey, and the Boston Renal Transplant Study Group Ganciclovir for treatment of renal transplant-associated primary cytomegalovirus pneumonia. J. Infect. Dis. 157: Heidelberger, C., and D. H. King Trifluorothymidine. Pharmacol. Ther. 6: Heinemann, M. H Long-term intravitreal ganciclovir therapy for cytomegalovirus retinopathy. Arch. Ophthalmol. 107: 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 Trisodium phosphonoformate, a new antiviral compound. Science 201: Henderly, D. E., W. R. Freeman, D. M. Causey, and N. A. Rao Cytomegalovirus retinitis and response to therapy with ganciclovir. Ophthalmology 94: Henry, K., H. Cantrill, C. Fletcher, B. J. Chinnock, and H. H. Balfour Use of intravitreal ganciclovir (dihydroxy propoxymethyl guanine) for cytomegalovirus retinitis in a patient with AIDS. Am. J. Ophthalmol. 103: Henry, K., B. J. Chinnock, R. P. Quinn, C. V. Fletcher, P. demiranda, and H. H. Balfour Concurrent zidovudine levels in semen and serum determined by radioimmunosassay in patients with AIDS or AIDS-related complex. JAMA 259: 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 Ditiocarb sodium (diethyldithiocarbamate) therapy in patients with symptomatic HIV infection and AIDS. JAMA 265: Hill, J. M., F. Sedarati, R. T. Javier, E. K. Wagner, and J. G. Stevens Herpes simplex virus latent phase transcription facilitates in vivo reactivation. Virology 174: Hirsch, M. S Chemotherapy of human immunodeficiency virus infections: current practice and future prospects. J. Infect. Dis. 161: 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 Effects of interferon-alpha on cytomegalovirus reactivation syndromes in renaltransplant recipients. N. Engl. J. Med. 308: Ho, D. D., T. R. Rota, J. C. Kaplan, K. L. Hartshorn, C. A. Andrews, R. T. Schooley, and M. S. Hirsch Recombinant ANTIVIRAL THERAPY 173 human interferon alfa-a suppresses HTLV-III replication in vitro. Lancet i: Ho, H., and M. J. M. HitchcocL Cellular pharmacology of 2',3'-dideoxy-2',3'-didehydrothymidine, a nucleoside analog active against human immunodeficiency virus. Antimicrob. Agents Chemother. 33: 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 Toxicity of combined ganciclovir and zidovudine for cytomegalovirus disease associated with AIDS. Ann. Intern. Med. 113: Hoffmann, C. E., E. M. Neumayer, R. F. Huff, and R. A. Coldsby Mode of action of the antiviral activity of amantadine in tissue culture. J. Bacteriol. 90: Holland, G. N., M. J. Sakamoto, D. Hardy, Y. Sidikaro, A. E. Kreiger, L. M. Frenkel, and the UCLA CMV Retinopathy Study Group Treatment of cytomegalovirus retinopathy in patients with acquired immunodeficiency syndrome. Arch. Ophthalmol. 104: 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 Randomized, controlled trial of recombinant human alpha-interferon in patients with chronic hepatitis B. Gastroenterology 95: Hovi, T Successful selective inhibitors of viruses. p In H. J. Field (ed.), Antiviral agents: the development and assessment of antiviral chemotherapy. CRC Press, Boca Raton, Fla Howell, C. L., and M. J. Miller Rapid method for determining the susceptibility of herpes simplex virus to acyclovir. Diagn. Microbiol. Infect. Dis. 2: Hruska, J. F., J. M. Bernstein, R. G. Douglas, Jr., and C. B. Hall Effects of ribavirin on respiratory syncytial virus in vitro. Antimicrob. Agents Chemother. 17: Huff, J. C., B. Bean, H. H. Balfour, 0. L. Laskin, J. D. Connor, L. Corey, J. Bryson, and P. McGuirt Therapy of herpes zoster with oral acyclovir. Am. J. Med. 85(Suppl. 2A): Huggins, J. W Prospects for treatment of viral hemorrhagic fevers with ribavirin, a broad-spectrum antiviral drug. Rev Infect Dis. 11(Suppl. 4): Indulen, M. K., and R. L. Feldblum Obtaining of a virazole-resistant fowl plague virus mutant. Acta Virol. 26: Isaacs, A., and J. Lindenmann Virus interference. I. The interferon. Proc. R. Soc. London Ser. B 147: Jabs, D. A., C. Enger, and J. C. Bartlett Cytomegalovirus retinitis and acquired immunodeficiency syndrome. Arch. Ophthalmol. 107: Jackson, G. G., D. A. Paul, L. A. Falk, M. Rubenis, J. C. Despotes, D. Mack, M. Knigge, and E. E. Emeson Human immunodeficiency virus (HIV) antigenemia (p24) in the acquired immunodeficiency syndrome (AIDS) and the effect of treatment with zidovudine (AZT). Ann. Intern. Med. 108: Jackson, G. G., M. Rubenis, M. Knigge, J. T. Perkins, D. A. Paul, J. C. Despotes, and P. Spencer Passive immunoneutralisation of human immunodeficiency virus in patients with advanced AIDS. Lancet ii: Jacobson, M. A., T. G. Berger, S. Fikrig, P. Becherer, J. W. Moohr, S. C. Stanat, and K. K. Biron Acyclovirresistant varicella zoster virus infection after chronic oral acyclovir therapy in patients with the acquired immunodeficiency syndrome (AIDS). Ann. Intern. Med. 112: Jacobson, M. A., S. Crowe, J. Levy, F. Aweeka, J. Gambertoglio, N. McManus, and J. Mills Effect of foscarnet therapy on infection with human immunodeficiency virus in patients with AIDS. J. Infect. Dis. 158: Jacobson, M. A., P. de Miranda, D. M. Cederberg, T. Burnette, E. Cobb, H. R. Brodie, and J. Mills Human pharmacokinetics and tolerance of oral ganciclovir. Antimicrob. Agents Chemother. 31: Jacobson, M. A., J. G. Gambertoglio, F. T. Aweeka, D. M. Causey, and A. A. Portale Foscarnet-induced hypocal-

29 174 BEAN cemia and effects of foscarnet on calcium metabolism. J. Clin. Endocrinol. Metab. 72: Jacobson, M. A., J. J. O'Donnell, and J. Mills Foscarnet treatment of cytomegalovirus retinitis in patients with the acquired immunodeficiency syndrome. Antimicrob. Agents Chemother. 33: Jacobson, M. A., C. van der Horst, D. M. Causey, M. Dehlinger, R. Hafner, and J. Mills In vivo additive antiretroviral effect of combined zidovudine and foscarnet therapy for human immunodeficiency virus infection (ACTG Protocol 053). J. Infect. Dis. 163: Japour, A. J., P. A. Chatis, H. E. Eigenrauch, and C. S. Crumpacker Detection of human immunodeficiency virus type I clinical isolates with reduced sensitivity to zidovudine and dideoxyinosine by RNA-RNA hybridization. Proc. Natl. Acad. Sci. USA 88: Johnson, M. A., G. Ahluwalia, M. C. Connelly, D. A. Cooney, S. Broder, D. G. Johns, and A. Fridland Metabolic pathways for the activation of the antiretroviral agent 2',3'- dideoxyadenosine in human lymphoid cells. J. Biol. Chem. 263: Joklik, W. K Interferons, p 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 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 The safety and pharmacokinetics of recombinant soluble CD4 (rcd4) in subjects with the acquired immunodeficiency syndrome (AIDS) and AIDS-related complex. Ann. Intern. Med. 112: Kaplowitz, L. G., D. Baker, L. Gelb, J. Blythe, R. Hale, P. Frost, C. Crumpacker, S. Rabinovich, J. E. Peacocki, J. Herndon, L. G. Davis, and the Acyclovir Study Group Prolonged continuous acyclovir treatment of normal adults with frequently recurring genital herpes simplex virus infection. JAMA 265: Karpas, A. G., W. J. Fleet, R. A. Dwek, S. Petursson, S. K. Namgoong, N. G. Ramsden, G. S. Jacob, and T. W. Rademacher Aminosugar derivatives as potential anti-human immunodeficiency virus agents. Proc. Natl. Acad. Sci. USA 85: Kato, N., and H. J. Eggers Inhibition of uncoating of fowl plague virus by 1-adamantanamine hydrochloride. Virology 37: Kaufman, H. E., E. L. Martola, and C. Dohlman Use of 5-iodo-2'-deoxyuridine (IDU) in treatment of herpes simplex keratitis. Arch. Ophthalmol. 68: Keith, B. R., G. White, and H. R. Wilson In vivo efficacy of zidovudine (3Y-azido-3'-deoxythymidine) in experimental gram-negative bacterial infections. Antimicrob. Agents Chemother. 33: Keller, P. M., J. A. Fyfe, L. Beauchamp, C. M. Lubbers, P. A. Furman, H. J. Schaeffer, and G. B. Elion Enzymatic phosphorylation of acyclic nucleoside analogs and correlations with antiherpetic activities. Biochem. Pharmacol. 30: Kende, M., C. R. Alving, W. L. Rill, G. M. Swartz, Jr., and P. G. Canonico Enhanced efficacy of liposome-encapsulated ribavirin against Rift Valley Fever virus infection in mice. Antimicrob. Agents Chemother. 27: Kim, S., S. Scheerer, M. A. Geyer, and S. B. Howell Direct cerebrospinal fluid delivery of an antiretroviral agent using multivesicular liposomes. J. Infect. Dis. 162: Klintmalm, G., B. Lonnqvist, B. Oberg, G. Gahrton, J. Lernestedt, G. Lundgren, 0. Ringden, K. Robert, B. Wahren, and C. Groth Intravenous foscarnet for the treatment of severe cytomegalovirus infection in allograft recipients. Scand. J. Infect. Dis. 17: Knight, V., S. Z. Wilson, J. M. Quarles, S. E. Greggs, H. W. McClung, B. K. Waters, R. W. Cameron, J. M. Zerwas, and R. B. Couch Ribavirin small-particle aerosol treatment of influenza. Lancet ii: CLIN. MICROBIOL. REV Knop, J Immunologic effects of interferon. J. Invest. Dermatol. 95(Suppl.): Knox, K. K., W. R. Drobyski, and D. R. Carrigan Cytomegalovirus isolate resistant to ganciclovir and foscarnet from a marrow transplant patient. Lancet 337: Kochhar, D. M., J. D. Penner, and T. B. Knudsen Embryotoxic, teratogenic, and metabolic effects of ribavirin in mice. Toxicol. Appl. Pharmacol. 52: Koff, W. C., I. J. Fidler, S. D. Showalter, M. K. Chakrabarty, B. Hampar, L. M. Ceccorulli, and E. S. Kleinerman Human monocytes activated by immunomodulators in liposomes lyse herpesvirus-infected but not normal cells. Science 224: Konopka, K., B. R. Davis, C. E. Larsen, D. R. Alford, R. J. Debs, and N. Duzgunes Liposomes modulate human immunodeficiency virus infectivity. J. Gen. Virol. 71: Korenman, J., B. Baker, J. Waggoner, J. E. Everhart, A. M. Di Bisceglie, and J. H. Hoofnagle Long-term remission of chronic hepatitis B after alpha-interferon therapy. Ann. Intern. Med. 114: Kornbluth, R. S., P. S. Oh, J. R. Munis, P. H. Cleveland, and D. D. Richman The role of interferons in the control of HIV replication in macrophages. Clin. Immunol. Immunopathol. 54: Kornhauser, D. M., C. W. Hendrix, L. J. Nerhood, B. G. Petty, A. S. Woods, J. G. Bartlett, and P. S. Lietman Probenecid and zidovudine metabolism. Lancet ii: Kotler, M., R. A. Katz, W. Danho, J. Leis, and A. M. Skalka Synthetic peptides as substrates and inhibitors of a retroviral protease. Proc. Natl. Acad. Sci. USA 85: Koup, R. A., V. J. Merluzzi, K. D. Hargrave, J. Adams, K. Grozinger, R. J. Eckner, and J. L. Sullivan Inhibition of human immunodeficiency virus type I (HIV-I) replication by the dipyridodiazepinone B1-RG-587. J. Infect. Dis. 163: 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 Combined zidovudine and interferon-alpha therapy in patients with Kaposi Sarcoma and the Acquired Immunodeficiency Syndrome (AIDS). Ann. Intern. Med. 111: Koyanagi, Y., W. A. O'Brien, J. Q. Zhao, D. W. Golde, J. C. Gasson, and I. S. Y. Chen Cytokines alter production of HIV-I from primary mononuclear phagocytes. Science 241: Krieger, J. N., R. W. Coombs, A. C. Collier, S. 0. Ross, K. Chaloupka, D. K. Cummings, V. L. Murphy, and L. Corey Recovery of human immunodeficiency virus type I from semen: minimal impact of stage of infection and current antiviral chemotherapy. J. Infect. Dis. 163: Krigel, R. L Reversible neurotoxicity due to oral acyclovir in a patient with chronic lymphocytic leukemia. J. Infect. Dis. 154: Krown, S. E., J. W. M. Gold, D. Niedzwiecki, D. Bundow, N. Flomenberg, B. Gansbacher, and B. J. Brew Interferonalpha with zidovudine: safety, tolerance, and clinical and virologic effects in patients with Kaposi Sarcoma associated with the Acquired Immunodeficiency Syndrome (AIDS). Ann. Intern. Med. 112: Krugman, S., R. Ward, J. P. Giles, and A. M. Jacobs Infectious hepatitis. Studies on the effect of gamma globulin and on the incidence of inapparent infection. JAMA 174: Kuhls, T. L., J. Sacher, E. Pineda, D. Santomauro, E. Wiesmeier, W. A. Growdon, and Y. J. Bryson Suppression of recurrent genital herpes simplex virus infection with recombinant alpha 2 interferon. J. Infect. Dis. 154: 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 ',3'-Dideoxyinosine (ddl) in patients with the Acquired Immunodeficiency Syndrome or AIDS-related complex. N. Engl. J. Med. 322:

30 VOL. 5, Land, S., G. Treloar, D. McPhee, C. Birch, R. Doherty, D. Cooper, and I. Gust Decreased in vitro susceptibility to zidovudine of HIV isolates obtained from patients with AIDS. J. Infect. Dis. 161: 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 Interferon-alfa in patients with asymptomatic human immunodeficiency virus (HIV) infection. Ann. Intern. Med. 112: 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 Anti-retroviral effects of interferon-alpha in AIDS-associated Kaposi's sarcoma. Lancet ii: Lane, H. C., H. Masur, L. C. Edgar, G. Whalen, A. H. Rook, and A. S. Fauci Abnormalities of B-cell activation and immunoregulation in patients with the acquired immunodeficiency syndrome. N. Engl. J. Med. 309: 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 Syngeneic bone marrow transplantation and adoptive transfer of peripheral blood lymphocytes combined with zidovudine in human immunodeficiency virus (HIV) infection. Ann. Intern. Med. 113: 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, and J. Goudsmit Failure of zidovudine prophylaxis after accidental exposure to HIV-I. N. Engl. J. Med. 322: Larder, B. A., B. Chesebro, and D. D. Richman Susceptibilities of zidovudine-susceptible and -resistant human immunodeficiency virus isolates to antiviral agents determined by using a quantitative plaque reduction assay. Antimicrob. Agents Chemother. 34: Larder, B. A., and G. Darby Selection and characterization of acyclovir-resistant herpes simplex virus type I mutants inducing altered DNA polymerase activities. Virology 146: Larder, B. A., G. Darby, and D. D. Richman HIV with reduced sensitivity to zidovudine (AZT) isolated during prolonged therapy. Science 243: Larder, B. A., and S. D. Kemp Multiple mutations in HIV-I reverse transcriptase confer high-level resistance to zidovudine (AZT). Science 246: Larder, B. A., D. J. M. Purifoy, K. L. Powell, C. Bradley, S. Kemp, M. Tisdale, P. Ertl, G. K. Darby, and D. Stammers Structural studies of the acquired immunodeficiency syndrome virus reverse transcriptase. Am. J. Med. 85(Suppl. 2A): Larsson, A., and B. Oberg Selective inhibition of herpesvirus DNA synthesis by foscarnet. Antiviral Res. 1: Larsson, A., K. Stenberg, and B. Oberg Reversible inhibition of cellular metabolism by ribavirin. Antimicrob. Agents Chemomther. 13: Laskin, 0. L., D. M. Cederberg, J. Mills, L. J. Eron, D. Mildvan, S. A. Spector, and the Ganciclovir Study Group Ganciclovir for the treatment and suppression of serious infections caused by cytomegalovirus. Am. J. Med. 83: Lau, A. S., G. E. Hannigan, M. H. Freedman, and B. R. G. Williams Regulation of interferon receptor expression in human blood lymphocytes in vitro and during interferon therapy. J. Clin. Invest. 77: Laurence, J Molecular interactions among herpesviruses and human immunodeficiency viruses. J. Infect. Dis. 162: Lehoang, P., B. Girard, M. Robinet, P. Marcel, L. Zazoun, S. Matheron, W. Rozenbaum, C. Katlama, I. Morer, J. 0. Lernestedt, H. Saraux, Y. Pouliquen, M. Gentilini, and F. Rousselie Foscarnet in the treatment of cytomegalovirus retinitis in acquired immune deficiency syndrome. Ophthalmology 96: Leinbach, S. S., J. M. Reno, L. F. Lee, A. F. Isbell, and J. A. Boezi Mechanism of phosphonoacetate inhibition of ANTIVIRAL THERAPY 175 herpesvirus-induced DNA polymerase. Biochemistry 15: Lernstedt, J. O., and A. C. Chanas Penile ulcerations with foscarnet. Lan-et 335: Lin, J., Z. Zhang, M. C. Smith, K. Biron, and J. S. Pagano Anti-human immunodeficiency virus agent 3'-azido-3'- deoxythymidine inhibits replication of Epstein-Barr virus. Antimicrob. Agents Chemother. 32: Lin, J.-C., M. C. Smith, and J. S. Pagano Prolonged inhibitory effect of 9-(1,3-dihydroxy-2-propoxymethyl)guanine against replication of Epstein-Barr virus. J. Virol. 50: Little, J. W., W. J. Hall, R. G. Douglas, Jr., R. W. Hyde, and D. M. Speers Amantadine effect on peripheral airways abnormalities in influenza. Ann. Intern. Med. 85: Lok, A. S. F., P. C. Wu, C. L. Lai, and E. K. Y. Leung Long-term follow-up in a randomised controlled trial of recombinant alpha 2-interferon in Chinese patients with chronic hepatitis B infection. Lancet ii: Lubeck, M.-D., J. L. Schulman, and P. Palese Susceptibility of influenza A viruses to amantadine is influenced by the gene coding for M protein. J. Virol. 28: 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 A collaborative study of patient-initiated treatment of recurrent genital herpes with topical acyclovir or placebo. J. Infect. Dis. 150: Machida, H., A. Kuninaka, and H. Yoshino Inhibitory effects of antiherpesviral thymidine analogs against varicellazoster virus. Antimicrob. Agents Chemother. 21: 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 (1-(2,3-Dideoxy-beta-D-glycero-pent-2-enofuranosyl) thymine. A highly potent and selective anti-hiv agent. J. Med. Chem. 32: Mar, E. C., Y. C. Cheng, and E. S. Huang Effect of 9-(1,3-dihydroxy-2-propoxymethyl)guanine on human cytomegalovirus replication in vitro. Antimicrob. Agents Chemother. 24: Mar, E. C., J. F. Chiou, Y. C. Cheng, and E. S. Huang Inhibition of cellular DNA polymerase and human cytomegalovirus-induced DNA polymerase by the triphosphates of 9-(2-hydroxyethoxymethyl)guanine and 9-(1,3-dihydroxy-2- propoxymethyl)guanine. J. Virol. 53: Marker, S. C., R. J. Howard, K. E. Groth, A. R. Mastri, R. L. Simmons, and H. H. Balfour, Jr A trial of vidarabine for cytomegalovirus infection in renal transplant patients. Arch. Intern. Med. 140: Marks, G. L., P. E. Nolan, K. S. Erlich, and M. N. Ellis Mucocutaneous dissemination of acyclovir-resistant herpes simplex virus in a patient with AIDS. Rev. Infect. Dis. 11: 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 Regulation of viral expression of human immunodeficiency virus in vitro by an antisense phosphorothioate oligodeoxynucleotide against rev (art/trs) in chronically infected cells. Proc. Natl. Acad. Sci. USA 86: McCammon, J. A Computer-aided molecular design. Science 238: McClung, H. W., V. Knight, B. E. Gilbert, S. Z. Wilson, J. M. Quarles, and G. W. Divine Ribavirin aerosol treatment of influenza B virus infection. JAMA 249: McCormick, J. B., J. P. Getchell, S. W. Mitchell, and D. R. Hicks Ribavirin suppresses replication of lymphadenopathy-associated virus in cultures of human adult T lymphocytes. Lancet ii: 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 Lassa fever. Effective therapy with ribavirin. N. Engl. J. Med. 314: McGuirt, P. V., J. E. Shaw, G. B. Elion, and P. A. Furman.

31 176 BEAN Identification of small DNA fragments synthesized in herpes simplex virus-infected cells in the presence of acyclovir. Antimicrob. Agents. Chemother. 25: McKeating, J. A., P. D. Griffiths, and R. A. Weiss HIV susceptibility conferred to human fibroblasts by cytomegalovirus-induced Fc receptor. Nature (London) 343: McKendrick, M. W., J. I. McGill, J. E. White, and M. J. Wood Oral acyclovir in acute herpes zoster. Br. Med. J. 293: McKinney, R. E., S. L. Katz, and C. M. Wilfert Chronic enteroviral meningoencephalitis in agammaglobulinemic patients. Rev. Infect. Dis. 9: 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 Protocol 043 Study Group A multicenter trial of oral zidovudine in children with advanced human immunodeficiency virus disease. N. Engl. J. Med. 324: McLaren, C., M. N. Ellis, and G. A. Hunter A colorimetric assay for the measurement of the sensitivity of herpes simplex viruses to antiviral agents. Antiviral Res. 3: Medical Letter Granulocyte colony-stimulating factors. Med. Lett. 33: Melder, R. J., R. Balachandran, C. R. Rinaldo, P. Gupta, T. L. Whiteside, and R. B. Herberman Cytotoxic activity against HIV-infected monocytes by recombinant interleukin 2-activated natural killer cells. AIDS Res. Hum. Retroviruses 6: Merigan, T. C., T. S. Hall, S. E. Reed, and D. A. J. Tyrrell Inhibition of respiratory virus infection by locally applied interferon. Lancet i: Merigan, T. C., K. H. Rand, R. B. Pollard, P. S. Abdallah, G. W. Jordan, and R. P. Fried Human leukocyte interferon for the treatment of herpes zoster in patients with cancer. N. Engl. J. Med. 298: 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 the AIDS Clinical Trials Group Circulating p24 antigen levels and responses to dideoxycytidine in human immunodeficiency virus (HIV) infections. Ann. Intern. Med. 110: 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 Inhibition of HIV-I replication by a nonnucleoside reverse transcriptase inhibitor. Science 250: 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 Study Group Long-term acyclovir suppression of frequently recurring genital herpes simplex virus infection. JAMA 260: 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 Prophylactic use of human leukocyte interferon after allogenic marrow transplantation. Ann. Intern. Med. 107: 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 Prevention of cytomegalovirus infection by cytomegalovirus immune globulin after marrow transplantation. Ann. Intern. Med. 98: 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 Acyclovir for prevention of cytomegalovirus infection and disease after allogeneic marrow transplantation. N. Engl. J. Med. 318: 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 Multicenter collaborative trial of intravenous acyclovir for treatment of mucocutaneous herpes simplex virus CLIN. MICROBIOL. REV. infection in the immunocompromised host. Am. J. Med. 73(Suppl. 1A): Miles, S. A., R. T. Mitsuyasu, J. Moreno, G. Baldwin, N. K. Alton, L. Souza, and J. A. Glaspy Combined therapy with recombinant granulocyte colony-stimulating factor and erythropoietin decreases hematologic toxicity from zidovudine. Blood 77: Miller, W. H., and R. L. Miller Phosphorylation of acyclovir (acycloguanosine) monophosphate by GMP kinase. J. Biol. Chem. 255: Mindel, A., A. Faherty, D. Hindley, I. V. D. Weller, S. Sutherland, A. P. Fiddian, and M. W. Adler Prophylactic oral acyclovir in recurrent genital herpes. Lancet ii: Mindel, A. M., W. Adler, S. Sutherland, and A. P. Fiddian Intravenous acyclovir treatment for primary genital herpes. Lancet i: Mitsuya, H., and S. Broder Inhibition of the in vitro infectivity and cytopathic effect of human T-lymphotrophic virus type III/lymphadenopathy-associated virus (HTLV-III/ LAV) by 2',3'-dideoxynucleosides. Proc. Natl. Acad. Sci. USA 83: Mitsuya, H., K. J. Weinhold, P. A. Furman, M. H. St. Clair, S. NusinoffLehrman, R. C. Gallo, D. Bolognesi, D. W. Barry, and S. Broder '-Azido-3'-deoxythymidine (BW A509U): an antiviral agent that inhibits the infectivity and cytopathic effect of human T-lymphotropic virus type III/lymphadenopathyassociated virus in vitro. Proc. Natl. Acad. Sci. USA 82: Monto, A. S., R. A. Gunn, M. G. Bandyk, and C. L. King Prevention of Russian influenza by amantadine. JAMA 241: Moore, R. D., H. Kessler, D. D. Richman, C. Flexner, and R. E. Chaisson Non-Hodgkin's lymphoma in patients with advanced HIV infection treated with zidovudine. JAMA 265: Moyle, G., S. E. Barton, A. Balestrini, M. Nelson, M. Youle, D. A. Hawkins, and B. G. Gazzard Penile ulcerations with foscarnet. Lancet 335: National Institute of Child Health and Human Development Intravenous Immunoglobulin Study Group Intravenous immune globulin for the prevention of bacterial infections in children with symptomatic human immunodeficiency virus infection. N. Engl. J. Med. 325: 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 Recombinant granulocyte-macrophage colony-stimulating factor after autologous bone marrow transplantation for lymphoid cancer. N. Engl. J. Med. 324: Neto, C. C., J. M. Steim, P. S. Sarin, D. K. Sun, N. N. Bhongle, R. K. Piratla, and J. G. Turcotte Lipid conjugates of antiretroviral agents. II. Disodium palmityl phosphonoformate: anti-hiv activity, physical properties, and interaction with plasma proteins. Biochem. Biophys. Res. Commun. 171: Nilsen, A. E., T. Aasen, A. M. Halsos, B. R. Kinge, E. A. L. Tjotta, K. Wikstrom, and A. P. Fiddian Efficacy of oral acyclovir in the treatment of initial and recurrent genital herpes. Lancet ii: Norley, S. G., L. Huang, and B. T. Rouse Targeting of drug loaded immunoliposomes to herpes simplex virus infected corneal cells: an effective means of inhibiting virus replication in vitro. J. Immunol. 136: Nusinoff Lehrman, S., J. M. Douglas, L. Corey, and D. W. Barry Recurrent genital herpes and suppressive oral acyclovir therapy. Relation between clinical outcome and in-vitro drug sensitivity. Ann. Intern. Med. 104: Nyerges, G., Z. Meszner, E. Gyarmati, and S. Kerpel-Fronius Acyclovir prevents dissemination of varicella in immunocompromised children. J. Infect. Dis. 157: Oberg, B Inhibitors of virus-specific enzymes, p In C. H. Stuart-Harris and J. Oxford (ed.), Problems of

32 VOL. 5, 1992 antiviral therapy. Academic Press, London Oberg, B Antiviral effects of phosphonoformate (PFA, foscarnet sodium). Pharmacol. Ther. 40: O'Brien, J. J., and D. M. Campoli-Richards Acyclovir. An updated review of its antiviral activity, pharmacokinetic properties and therapeutic efficacy. Drugs 37: Ogle, J. W., P. Toetzis, W. D. Parker, N. Alvarez, K. McIntosh, M. J. Levin, and B. A. Lauer Oral ribavirin therapy for subacute sclerosing panencephalitis. J. Infect. Dis. 159: Oker-Blom, N., T. Hovi, P. Leinikki, T. Palosuo, R. Pettersson, and J. Suni Protection of man from natural infection with influenza A2 Hong Kong virus by amantadine: a controlled field trial. Br. Med. J. 3: Oksenhendler, E., P. Bierling, F. Ferchal, J. P. Clauvel, and M. Seligmann Zidovudine for thrombocytopenic purpura related to human immunodeficiency virus (HIV) infection. Ann. Intern. Med. 110: Oleson, D., H. Grierson, J. Goldsmith, D. T. Purtilo, and D. Johnson Augmentation of natural cytotoxicity by leucine enkephalin in cultured peripheral blood mononuclear cells from patients infected with human immunodeficiency virus. Clin. Immunol. Immunopathol. 51: Page, T., and J. D. Connor The metabolism of ribavirin in erythrocytes and nucleated cells. Int. J. Biochem. 22: Pahwa, S., K. Biron, W. Lim, P. Swenson, M. H. Kaplan, N. Sadick, and R. Pahwa Continous varicella-zoster infection associated with acyclovir resistance in a child with AIDS. JAMA 260: 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. Manischewitz, J. R. Minor, R. B. Nussenblatt, H. Masur, and H. C. Lane A randomized, controlled trial of foscarnet in the treatment of cytomegalovirus retinitis in patients with AIDS. Ann. Intern. Med. 115: Parris, D. S., and J. E. Harrington Herpes simplex virus variants resistant to high concentrations of acyclovir exist in clinical isolates. Antimicrob. Agents Chemother. 22: Patterson, J. L., and R. Fernandez-Larsson Molecular mechanisms of action of ribavirin. Rev. Infect. Dis. 12: Pauwels, R., K. Andries, J. Desmyter, D. Schols, M. J. Kukla, H. J. Breslin, A. Raeymaeckers, J. V. Gelder, R. Woestenborghs, J. Heykants, K. Schellekens, M. A. C. Janssen, E. DeClercq, and P. A. J. Janseen Potent and selective inhibition of HIV-I replication in vitro by a novel series of TIBO derivatives. Nature (London) 343: Pazin, G. J., J. H. Harger, J. A. Armstrong, M. K. Breinig, R. J. Caplan, K. Cantell, and M. Ho Leukocyte interferon for treating first episodes of genital herpes in women. J. Infect. Dis. 156: Peavy, D. L., W. C. Koff, D. S. Hyman, and V. Knight Inhibition of lymphocyte proliferative responses by ribavirin. Infect. Immun. 29: 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 Scandinavian Isoprinosine Study Group The efficacy of inosine pranobex in preventing the acquired immunodeficiency syndrome in patients with human immunodeficiency virus infection. N. Engl. J. Med. 322: Pelling, J. C., J. C. Drach, and C. Shipman, Jr Internucleotide incorporation of arabinosyladenine into herpes simplex virus and mammalian cell DNA. Virology 109: 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 Inhibition of human immunodeficiency virus (HIV-I/HTLV-III/Ba-L) replication in fresh and cultured human peripheral blood monocytes/macrophages by azidothymidine and related 2',3'-dideoxynucleosides. J. ANTIVIRAL THERAPY 177 Exp. Med. 168: 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 Replication of human immunodeficiency virus in monocytes. J. Exp. Med. 169: Perren, T. J., R. L. Powles, D. Easton, K. Stolle, and P. J. Selby Prevention of herpes zoster in patients by longterm oral acyclovir after allogeneic bone marrow transplantation. Am. J. Med. 85(Suppl. 2A): Perrillo, R. P., F. G. Regenstein, M. G. Peters, K. DeSchryver- Kecskemeti, C. J. Bodicky, C. R. Campbell, and M. C. Kuhns Prednisone withdrawal followed by recombinant alpha interferon in the treatment of chronic type B hepatitis. Ann. Intern. Med. 109: 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, and the Hepatitis Interventional Therapy Group A randomized, controlled trial of interferon alfa-2b alone and after prednisone withdrawal for the treatment of chronic hepatitis B. N. Engl. J. Med. 323: Pestka, S., and J. A. Langer Interferons and their actions. Annu. Rev. Biochem. 56: Peterslund, N. A., J. Ipsen, H. Schonheyder, K. Seyer-Hansen, V. Esmann, and H. Juhl Acyclovir in herpes zoster. Lancet ii: 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 Dideoxycytidine alone and in an alternating schedule with zidovudine in children with symptomatic human immunodeficiency virus infection. J. Pediatr. 117: 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 Effect of continuous intravenous infusion of zidovudine (AZT) in children with symptomatic HIV infection. N. Engl. J. Med. 319: Plotkin, S. A., S. E. Starr, and C. K. Bryan In vitro and in vivo responses of cytomegalovirus to acyclovir. Am. J. Med. 73(Suppl. 1A): 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 Development of non-hodgkin lymphoma in a cohort of patients with severe human immunodeficiency virus (HIV) infection on long-term antiretroviral therapy. Ann. Intern. Med. 113: Plunkett, W., and S. S. Cohen Metabolism of 9-p-Darabinofuranosyladenine by mouse fibroblasts. Cancer Res. 35: Poli, G., J. M. Orenstein, A. Kinter, T. M. Folks, and A. S. Fauci Interferon-alpha but not AZT suppresses HIV expression in chronically infected cell lines. Science 244: Potter, C. W., J. P. Phair, L. Vodinelich, R. Fenton, and R. Jennings Antiviral, immunosuppressive and antitumor effects of ribavirin. Nature (London) 259: Powers, C. N., D. L. Peavy, and V. Knight Selective inhibition of functional lymphocyte subpopulations by ribavirin. Antimicrob. Agents Chemother. 22: Prichard, M. N., L. E. Prichard, W. A. Baguley, M. R. Nassiri, and C. Shipman Three-dimensional analysis of the synergistic cytotoxicity of ganciclovir and zidovudine. Antimicrob. Agents Chemother. 35: Prichard, M. N., S. R. Turk, L. A. Coleman, S. L. Engelhardt, C. Shipman, and J. C. Drach A microtiter virus yield reduction assay for the evaluation of antiviral compounds against human cytomegalovirus and herpes simplex virus. J. Virol. Methods 28: Prober, C. G., L. E. Kirk, and R. E. Keeney Acyclovir therapy of chickenpox in immunosuppressed children-a collaborative study. J. Pediatr. 101:

33 178 BEAN 362. Prusiner, P., and M. Sundaralingham A new class of synthetic nucleoside analogues with broad-spectrum antiviral properties. Nature (London) New Biol. 244: Prusoff, W. H Synthesis and biological activities of iododeoxyuridine, an analog of thymidine. Biochim. Biophys. Acta 32: Prusoff, W. H A review of some aspects of 5-iododeoxyuridine and azauridine. Cancer Res. 23: Prusoff, W. H., W. R. Mancini, T.-S. Lin, J. J. Lee, S. A. Siegel, and M. J. Otto Physical and biological consequences of incorporation of antiviral agents into virus DNA. Antiviral Res. 4: Rabalais, G. P., M. J. Levin, and F. E. Berkowitz Rapid herpes simplex virus susceptibility testing using an enzymelinked immunosorbent assay performed in situ on fixed virusinfected monolayers. Antimicrob. Agents Chemother. 31: Rankin, J. T., Jr., S. B. Eppes, J. B. Antczak, and W. K. Jokik Studies on the mechanism of the antiviral activity of ribavirin against reovirus. Virology 168: Ray, C. G Ribavirin: ambivalence about an antiviral agent. Am. J. Dis. Child. 142: Reardon, J. E., and T. Spector Herpes simplex virus type I DNA polymerase: mechanism of inhibition by acyclovir triphosphate. J. Biol. Chem. 264: 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 Retroviral Research A phase I evaluation of the safety and immunogenicity of vaccination with recombinant gpl60 in patients with early human immunodeficiency virus infection. N. Engl. J. Med. 324: Reed, E. C., R. A. Bowden, P. S. Dandliker, K. E. Lilleby, and J. D. Meyers Treatment of cytomegalovirus pneumonia with ganciclovir and intravenous cytomegalovirus immunoglobulin in patients with bone marrow transplants. Ann. Intern. Med. 109: 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 Ganciclovir for the treatment of cytomegalovirus gastroenteritis in bone marrow transplant patients. Ann. Intern. Med. 112: Reichman, R. C., G. J. Badger, M. E. Guinan, A. J. Nahmias, R. E. Keeney, L. G. Davis, T. Ashikaga, and R. Dolin Topically administered acyclovir in the treatment of recurrent herpes simplex genitalis: a controlled trial. J. Infect. Dis. 147: 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 Treatment of recurrent genital herpes simplex infections with oral acyclovir. JAMA 251: 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 Treatment of condyloma acuminatum with three different interferons administered intralesionally. Ann. Intern. Med. 108: Renault, P. F., J. H. Hoofnagle, Y. Park, K. D. Mullen, M. Peters, D. B. Jones, V. Rustgi, and E. A. Jones Psychiatric complications of long-term interferon alfa therapy. Arch. Intern. Med. 147: Resnick, L., J. S. Herbst, D. V. Ablashi, S. Atherton, B. Frank, L. Rosen, and S. N. Horwitz Regression of oral hairy leukoplakia after orally administered acyclovir therapy. JAMA 259: Reuman, P. D., D. I. Bernstein, M. C. Keefer, E. C. Young, J. R. Sherwood, and G. M. Schiff Efficacy and safety of low dosage amantadine hydrochloride as prophylaxis for influenza A. Antiviral Res. 11: Richman, D., M. A. Fischl, M. H. Grikeco, M. S. Gottlieb, P. A. CLIN. MICROBIOL. REV. 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 The toxicity of azidothymidine (AZT) in the treatment of patients with AIDS and AIDS-related complex. N. Engl. J. Med. 317: Richman, D. D., J. M. Grimes, and S. W. Lagakos Effect of stage of disease and drug dose on zidovudine susceptibilities of isolates of human immunodeficiency virus. J. Acquired Immune Defic. Syndr. 3: Richman, D. D., K. Y. Hostetler, P. J. Yazaki, and S. Clark Fate of influenza A virion proteins after entry into subcellular fractions of LLC cells and the effect of amantadine. Virology 151: Ringden, O., B. Lonnqvist, T. Paulin, J. Ahlmen, G. Klintmaim, B. Wahren, and J. 0. Lernestedt Pharmacokinetics, safety and preliminary clinical experiences using foscarnet in the treatment of cytomegalovirus infections in bone marrow and renal transplant recipients. J. Antimicrob. Chemother. 17: Roberts, N. A., J. A. Martin, D. Kinchington, A. V. Broadhurst, 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 Rational design of peptide-based HIV proteinase inhibitors. Science 248: 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 A multicenter clinical trial of oral ribavirin in HIVinfected patients with lymphadenopathy. J. Acquired Immun. Defic. Syndr. 3: 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 A multicenter clinical trial of oral ribavirin in HIVinfected people with lymphadenopathy: virologic observations. AIDS 4: Roberts, R. B., K. Jurica, W. A. Meyer III, H. Paxton, and R. W. Makuch A Phase I study of ribavirin in human immunodeficiency virus-infected patients. J. Infect. Dis. 162: Roberts, R. B., 0. L. Laskin, J. Laurence, D. Scavuzzo, H. W. Murray, Y. T. Kim, and J. D. Connor Ribavirin pharmacodynamics in high-risk patients for acquired immunodeficiency syndrome. Pharmacol. Ther. 42: 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 Aerosolized ribavirin in the treatment of patients with respiratory syncytial virus disease. Pediatr. Infect. Dis. J. 6: Roilides, E., D. Venzon, P. A. Pizzo, and M. Rubin Effects of antiretroviral dideoxynucleosides on polymorphonuclear leukocyte function. Antimicrob. Agents Chemother. 34: Roizman, B Multiplication of viruses. An overview, p In B. N. Fields and D. M. Knipe (ed.), Fields virology, 2nd ed. Raven Press, New York 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, and the Canadian Zidovudine Multi-Centre Study Group Isolation of drug-resistant variants of HIV-I from patients long-term zidovudine therapy. AIDS 3: on 392. Rooney, J. F., J. M. Felser, J. M. Ostrove, and S. E. Straus Acquisition of genital herpes from an asymptomatic sexual partner. N. Engl. J. Med. 314: Rose, K. M., and S. T. Jacob Selective inhibition of RNA polyadenylation by Ara-ATP in vitro: a possible mechanism for antiviral action of ARA-A. Biochem. Biophys. Res. Commun. 81: Ross, A. H., A. Julia, and C. Balakrishnan Toxicity of

34 VOL. 5, 1992 adenine arabinoside in humans. J. Infect. Dis. 133(Suppl.): A192-A Rossmann, M. G Antiviral agents targeted to interact with viral capsid proteins and a possible application to human immunodeficiency virus. Proc. Natl. Acad. Sci. USA 85: Rossmann, M. G., E. Arnold, J. W. Erickson, E. A. Frankenberger, J. P. Griffith, H.-J. Hecht, J. E. Johnson, G. Kamer, M. Luo, A. G. Mosser, R. R. Rueckert, B. Sherry, and G. Vriend Structure of a human common cold virus and functional relationship to other picornaviruses. Nature (London) 317: Rothenberg, M., G. Johnson, C. Laughlin, I. Green, J. Cradock, N. Sarver, and J. S. Cohen Oligodeoxynucleotides as anti-sense inhibitors of gene expression: therapeutic implications. J. Natl. Cancer Inst. 81: Sacks, S. L., T. C. Merigan, J. Kaminska, and I. H. Fox Inactivation of S-adenosylhomocysteine hydrolase during adenine arabinoside therapy. J. Clin. Invest. 69: Sacks, S. L., R. J. Wanklin, D. E. Reece, K. A. Hicks, K. L. Tyler, and D. M. Coen Progressive esophagitis from acyclovir-resistant herpes simplex. Ann. Intern. Med. 111: Safrin, S., T. Assaykeen, S. Follansbee, and J. Mills Foscarnet therapy for acyclovir-resistant mucocutaneous herpes simplex virus infection in 26 AIDS patients: preliminary data. J. Infect. Dis. 161: Safron, S., T. G. Berger, I. Gilson, P. R. Wolfe, C. B. Wofsy, J. Mills, and K. K. Biron Foscarnet therapy in five patients with AIDS and acyclovir-resistant varicella-zoster virus infection. Ann. Intern. Med. 115: Safrin, S., C. Crumpacker, P. Chatis, R. Davis, R. Hafner, J. Rush, H. A. Kessler, B. Landry, J. Mills, and other members of the AIDS Clinical Trials Group A controlled trial comparing foscarnet with vidarabine for acyclovir-resistant mucocutaneous herpes simplex in the acquired immunodeficiency syndrome. N. Engl. J. Med. 325: Salk, J Prospects for the control of AIDS by immunizing seropositive individuals. Nature (London) 327: Sandstrom, E. G., R. E. Byington, J. C. Kaplan, and M. S. Hirsch Inhibition of human T-cell lymphotropic virus type III in vitro by phosphonoformate. Lancet i: Saral, R., R. F. Ambinder, W. H. Burns, C. M. Angelopulos, D. E. Griffin, P. J. Burke, and P. S. Lietman Acyclovir prophylaxis against herpes simplex virus infection in patients with leukemia. Ann. Intern. Med. 99: Saral, R., W. H. Burns, 0. L. Laskin, G. W. Santos, and P. S. Lietman Acyclovir prophylaxis of herpes-simplex-virus infections. N. Engl. J. Med. 305: Sawyer, M. H., D. E. Webb, J. E. Balow, and S. E. Straus Acyclovir-induced renal failure. Am. J. Med. 84: Scadden, D. T., H. A. Bering, J. D. Levine, J. Bresnahan, L. Evans, C. Epstein, and J. E. Groopman Granulocytemacrophage colony-stimulating factor mitigates the neutropenia of combined interferon alfa and zidovudine treatment of acquired immune deficiency syndrome-associated Kaposi's Sarcoma. J. Clin. Oncol. 9: Schabel, F. M., Jr The antiviral activity of 9-,-D- Arabinofuranosyladenine. (ARA-A). Chemotherapy 13: Schaeffer, H. J., L. Beauchamp, P. de Miranda, and G. B. Elion, D. J. Bauer, and P. Collins (2-Hydroxyethoxymethyl) guanine activity against viruses of the herpes group. Nature (London) 272: 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 CMV Study Group A randomized, controlled trial of prophylactic ganciclovir for cytomegalovirus pulmonary infection in recipients of allogeneic bone marrow transplants. N. Engl. J. Med. 324: Schmitt, F. A., J. W. Bigley, R. McKinnis, P. E. Logue, R. W. Evans, J. L. Drucker, and the AZT Collaborative Working ANTIVIRAL THERAPY 179 Group Neuropsychological outcome of zidovudine (AZT) treatment of patients with AIDS and AIDS-related complex. N. Engl. J. Med. 319: Schnipper, L. E., and C. S. Crumpacker Resistance of herpes simplex virus to acycloguanosine: role of viral thymidine kinase and DNA polymerase loci. Proc. Natl. Acad. Sci. USA 77: 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 Recombinant soluble CD4 therapy in patients with the Acquired Immunodeficiency Syndrome (AIDS) and AIDS-related complex. Ann. Intern. Med. 112: Schulman, K. A., L. A. Lynn, H. A. Glick, and J. M. Eisenberg Cost effectiveness of low-dose zidovudine therapy for asymptomatic patients with human immunodeficiency virus (HIV) infection. Ann. Intern. Med. 114: Schwartz, D. H., G. Skowron, and T. C. Merigan Safety and effects of interleukin-2 plus zidovudine in asymptomatic individuals infected with human immunodeficiency virus. J. Acquired Immune Defic. Syndr. 4: Schwartz, P. M., C. Shipman, Jr., and J. C. Drach Antiviral activity of arabinosyladenine and arabinosylhypoxanthine in herpes simplex virus-infected KB cells: selective inhibition of viral deoxyribonucleic acid synthesis in the presence of an adenosine deaminase inhibitor. Antimicrob. Agents Chemother. 10: Seale, L., C. J. Jones, S. Kathpalia, G. G. Jackson, M. Mozes, M. S. Maddux, and D. Packham Prevention of herpesvirus infections in renal allograft recipients by low-dose oral acyclovir. JAMA 254: Sears, S. D., and M. L. Clements Protective efficacy of low-dose amantadine in adults challenged with wild-type influenza A virus. Antimicrob. Agents Chemother. 31: 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 Type B hepatitis after needle-stick exposure: prevention with hepatitis B immune globulin. Ann. Intern. Med. 88: Selby, P. J., R. L. Powles, D. Easton, T. J. Perren, K. Stolle, B. Jameson, A. P. Fiddian, Y. Tryhorn, and H. Stern The prophylactic role of intravenous and long-term oral acyclovir after allogeneic bone marrow transplantation. Br. J. Cancer 59: Shepp, D. H., P. S. Dandliker, P. demiranda, T. C. Burnette, D. M. Cederberg, L. E. Kirk, and J. D. Meyers Activity of 9-(2-hydroxy-1-(hydroxymethyl)ethoxymethyl)guanine in the treatment of cytomegalovirus pneumonia. Ann. Intern. Med. 103: Shepp, D. H., P. S. Dandliker, N. Flournoy, and J. D. Meyers Sequential intravenous and twice-daily oral acyclovir for extended prophylaxis of herpes simplex virus infection in marrow transplant patients. Transplantation 43: Shepp, D. H., P. S. Dandliker, and J. D. Meyers Treatment of varicella-zoster virus infection in severely immunocompromised patients. N. Engl. J. Med. 314: Shepp, D. H., B. A. Newton, P. S. Dandliker, N. Flournoy, and J. D. Meyers Oral acyclovir therapy for mucocutaneous herpes simplex virus infections in immunocompromised marrow transplant recipients. Ann. Intern. Med. 102: Shigeta, S., S. Mori, T. Yokota, K. Konno, and E. De Clercq Characterization of a varicella-zoster virus variant with altered thymidine kinase activity. Antimicrob. Agents Chemother. 29: Shindo, M., A. M. Di Bisceglie, L. Cheung, J. W.-K. Shih, K. Cristiano, S. M. Feinstone, and J. H. Hoofnagle Decrease in serum hepatitis C viral RNA duirng alpha-interferon therapy for chronic hepatitis C. Ann. Intern. Med. 115:

35 180 BEAN 428. Sibrack, C. D., L. T. Gutman, C. M. Wilfert, C. McLaren, M. H. St. Clair, P. M. Keller, and D. W. Barry Pathogenicity of acyclovir-resistant herpes simplex virus type I from an immunodeficient child. J. Infect. Dis. 146: Sidwell, R. W., J. H. Huffman, G. P. Khare, L. B. Allen, J. T. Witkowski, and R. K. Robins Broad spectrum antiviral activity of virazole: ribofuranosyl-1,2,4-triazole-3-carboxamide. Science 177: Silvestris, F., A. Gernone, M. A. Frassanito, and F. Dammacco Immunologic effects of long-term thymopentin treatment in patients with HIV-induced lymphadenopathy syndrome. J. Lab. Clin. Med. 113: Sim, I. S., R. L. Cerruti, and E. V. Connell Antiinfluenza virus activity of rimantadine in cells in culture. J. Respir. Dis. 10(Suppl.): Sjovall, J., S. Bergdahl, G. Movin, S. Ogenstad, and M. Saarimaki Pharmacokinetics of foscarnet and distribution to cerebrospinal fluid after intravenous infusion in patients with human immunodeficiency virus infection. Antimicrob. Agents Chemother. 33: Sjovall, J., A. Karlsson, S. Ogenstad, E. Sandstrom, and M. Saarimaki Pharmacokinetics and absorpition of foscarnet after intravenous and oral administration to patients with human immunodeficiency virus. Clin. Pharmacol. Ther. 44: Skoldenberg, B., K. Alestig, L. Burman, A. Korkman, K. Lovgren, R. Norrby, G. Stiernstedt, M. Forsgren, T. Bergstrom, E. Dahlqvist, A. Fryden, K. Norlin, E. Olding-Stenkvist, I. Uhnoo, and K. devahl Acyclovir versus vidarabine in herpes simplex encephalitis. Lancet ii: Skowron, G., and T. M. Merigan Alternating and intermittent regimens of zidovudine (3'-azido-3'-deoxythymidine) and dideoxycytidine (2',3'-dideoxycytidine) in the treatment of patients with acquired immunodeficiency syndrome (AIDS) and AIDS-related complex. Am. J. Med. 88(Suppl. 5B):20S-23S Smee, D. F., J. C. Martin, J. P. H. Verheyden, and T. R. Matthews Anti-herpesvirus activity of the acyclic nucleoside 9-(1,3-dihyhdroxy-2-propoxymethyl) guanine. Antimicrob. Agents Chemother. 23: Smee, D. F., and T. R. Matthews Metabolism of ribavirin in respiratory syncytial virus-infected and uninfected cells. Antimicrob. Agents Chemother. 30: Smith, D. H., R. A. Byrn, S. A. Marsters, T. Gregory, J. E. Groopman, and D. J. Capon Blocking of HIV-I infectivity by a soluble, secreted form of the CD4 antigen. Science 238: Smith, D. W., L. R. F. Frankel, L. H. Mathers, A. T. S. Tang, R. L. Ariagno, and C. G. Prober A controlled trial of aerosolized ribavirin in infants receiving mechanical ventilation for severe respiratory syncytial virus infection. N. Engl. J. Med. 325: Smith, T. F Rapid methods for the diagnosis of viral infections. Lab. Med. 18: Smith, T. J., M. J. Kremer, M. Luo, G. Vriend, E. Arnold, G. Kamer, M. G. Rossmann, M. A. McKinlay, G. D. Diana, and M. J. Otto The site of attachment in human rhinovirus 14 for antiviral agents that inhibit uncoating. Science 233: 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 Use of cytomegalovirus immune globulin to prevent primary cytomegalovirus disease in renal-transplant recipients. N. Engl. J. Med. 317: Sommadossi, J. P., R. Bevan, T. Ling, F. Lee, B. Mastre, M. D. Chaplin, C. Nerenberg, S. Koretz, and W. C. Buhles, Jr Clinical pharmacokinetics of ganciclovir in patients with normal and impared renal function. Rev. Infect. Dis. 10(Suppl. 3):S507-S514. CLIN. MICROBIOL. REV Sommadossi, J.-P., and R. Carlisle Toxicity of 3-azido- 3'deoxythymidine and 9-(1,3-dihydroxy-2 propoxymethyl) guanine for normal human hematopoietic progenitor cells in vitro. Antimicrob. Agents Chemother. 31: Sonnenblick, M., and A. Rosin Cardiotoxicity of interferon. A review of 44 cases. Chest. 99: Spear, J. B., C. A. Benson, J. C. Pottage, D. A. Paul, A. L. Landay, and H. A. Kessler Rapid rebound of serum human immunodeficiency virus antigen after discontinuing zidovudine therapy. J. Infect. Dis. 158: Spiegel, R. J., J. R. Spicehandler, S. L. Jacobs, and E. M. Oden Low incidence of serum neutralizing factors in patients receiving recombinant alfa-2b interferon (Intron A). Am. J. Med. 80: Spruance, S. L., D. J. Freeman, J. C. B. Stewart, M. B. McKeough, L. G. Wenerstrom, G. G. Krueger, M. W. Piepkorn, W. G. Stroop, and N. H. Rowe The natural history of ultraviolet radiation-induced herpes simplex labialis and response to therapy with peroral and topical formulations of acyclovir. J. Infect. Dis. 163: Spruance, S. L., M. L. Hamill, W. S. Hoge, L. G. Davis, and J. Mills Acyclovir iprevents reactivation of herpes simplex labialis in skiers. JAMA 260: Spruance, S. L., J. C. B. Stewart, D. J. Freeman, V. J. Brightman, J. L. Cox, G. Wenerstrom, M. B. McKeough, and N. H. Rowe Early application of topical 15% idoxuridine in dimethyl sulfoxide shortens the course of herpes simplex labialis: a multicenter placebo-controlled trial. J. Infect. Dis. 161: Spruance, S. L., J. C. B. Stewart, N. H. Rowe, M. B. McKeough, G. Wenerstrom, and D. J. Freeman Treatment of recurrent herpes simplex labialis with oral acyclovir. J. Infect. Dis. 161: Stanat, S. C., J. E. Reardon, A. Erice, M. C. Jordon, W. L. Drew, and K. K. Biron Ganciclovir-resistant cytomegalovirus clinical isolates: mode of resistance to ganciclovir. Antimicrob. Agents Chemother. 35: St. Clair, M. H., P. A. Furman, C. M. Lubbers, and G. B. Elion Inhibition of cellular alpha and virally induced deoxyribonucleic acid polymerase by the triphosphate of acyclovir. Antimicrob. Agents Chemother. 18: St. Clair, M. H., C. A. Richards, T. Spector, K. J. Weinhold, W. H. Miller, A. J. Langlois, and P. A. Furman '-Azido-3'-deoxythymidine triphosphate as an inhibitor and substrate of purified human immunodeficiency virus reverse transcriptase. Antimicrob. Agents Chemother. 31: Steele, R. W., and R. K. Charlton Immune modulators as antiviral agents, p In W. L. Drew (ed.), Clinics in laboratory medicine. The W. B. Saunders Co., Philadelphia Stevens, J. G Human herpesviruses: a consideration of the latent state. Microbiol. Rev. 53: 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 Effect of oral acyclovir treatment of symptomatic and asymptomatic virus shedding in recurrent genital herpes. Sex. Transm. Dis. 16: Streeter, D. G., J. T. Witkowski, G. P. Khare, R. W. Sidwell, R. J. Bauer, R. K. Robins, and L. N. Simon Mechanism of 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: Stridh, S Determination of ribonucleoside triphosphate pools in influenza A virus-infected MDCK cells. Arch. Virol. 77: Stuart-Harris, C Antiviral chemotherapy; an introduction and apology for the slow progress, p In C. H. Stuart-Harris and J. Oxford (ed.), Problems of antiviral chemotherapy. Academic Press, Ltd., London Sugrue, R. J., G. Bahadur, M. C. Zambon, M. Hall-Smith, A. R. Douglas, and A. J. Hay Specific structural alteration of the influenza hemagglutinin by amantadine. EMBO J. 9: Sullivan, K. M., K. J. Kopecky, J. Jocom, L. Fisher, C. D.

36 VOL. 5, 1992 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 Immunomodulatory and antimicrobial efficacy of intravenous immunoglobulin in bone marrow transplantation. N. Engl. J. Med. 323: Sullivan, V., K. Biron, M. Davis, S. Stanat, C. Talarico, and D. Coen Resistance of HCMV to DHPG, HPMPC and HPMPA mapped to the viral DNA polymerase gene. Antiviral Res. S1: Sweet, C., F. G. Hayden, K. J. Jakeman, S. Grambas, and A. J. Hay Virulence of Rimantadine-resistant human influenza A (H3N2) viruses in ferrets. J. Infect. Dis. 164: Swierkosz, E. M., D. R. Scholl, J. L. Brown, J. D. Jollick, and C. A. Gleaves Improved DNA hybridization method for detection of acyclovir-resistant herpes simplex virus. Antimicrob. Agents Chemother. 31: Swiss Group for Clinical Studies on the Acquired Immunodeficiency Syndrome (AIDS) Zidovudine for the treatment of thrombocytopenia associated with human immunodeficiency virus (HIV). Ann. Intern. Med. 109: Syntex Laboratories Cytovene (package insert). Syntex Laboratories, Inc., Palo Alto, Calif Szoka, F. C., and C. J. Chu Increased efficacy of phosphonoformate and phosphonoacetate inhibition of herpes simplex virus type 2 replication by encapsulation in liposomes. Antimicrob. Agents Chemother. 32: 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 Ribavirin aerosol treatment of bronchiolitis associated with respiratory syncytial virus infection in infants. Pediatrics 72: Telenti, A., and T. F. Smith Screening with a shell vial assay for antiviral activity against cytomegalovirus. Diagn. Microbiol. Infect. Dis. 12: Tenser, R. B., and W. A. Edris Trigeminal ganglion infection by thymidine kinase-negative mutants of herpes simplex virus after in vivo complementation. J. Virol. 61: Thin, R. N., J. M. Nabarro, J. D. Parker, and A. P. Fiddian Topical acyclovir in the treatment of initial genital herpes. Br. J. Vener. Dis. 59: Till, M. A., V. Ghetie, T. Gregory, E. J. Patzer, J. P. Porter, J. W. Uhr, D. J. Capon, and E. S. Vitetta HIV-infected cells are killed by rcd4-ricin A chain. Science 242: Tocci, M. J., T. J. Livcelli, H. C. Perry, C. S. Crumpacker, and A. K. Field Effects of the nucleoside analog 2'-nor-2'- deoxyguanosine on human cytomegalovirus replication. Antimicrob. Agents Chemother. 25: 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 Evaluation of therapeutic efficacy of amantadine in patients with naturally occurring A2 influenza. JAMA 211: Toltzis, P., C. M. Marx, N. Kleinman, E. M. Levine, and E. V. Schmidt Zidovudine-associated embryonic toxicity in mice. J. Infect. Dis. 163: Toltzis, P., K. O'Connell, and J. L. Patterson Effect of phosphorylated ribavirin on vesicular stomatitis virus transcription. Antimicrob. Agents Chemother. 32: Traunecker, A., J. Schneider, H. Kiefer, and K. Karjalainen Highly efficient neutralization of HIV with recombinant CD4-immunoglobulin molecules. Nature (London) 339: Tucker, W. E Preclinical toxicology profile of acyclovir: an overview. Am. J. Med. 73(Suppl. 1A): Vance, J. C., and D. Davis Interferon alpha-2b injections used as an adjuvant therapy to carbon dioxide laser vaporization of recalcitrant ano-genital condylomata acuminata. J. Invest. Dermatol. 95(Suppl.):146S-148S 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, and J. S. Pagano Lack of effect of peroral acyclovir for the ANTIVIRAL THERAPY 181 treatment of acute infectious mononucleosis. J. Infect. Dis. 164: 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 Foscarnet and penil ulcertation. Lancet 335: Van Voris, L. P., R. F. Betts, F. G. Hayden, W. A. Christmas, and R. G. Douglas, Jr Successful treatment of naturally occurring influenza A/USSR/77 HlNl. JAMA 245: Vere Hodge, R. A., and R. M. Perkins Mode of action of 9-(4-hydroxy-3-hydroxymethylbut-1-yl)guanine (BRL 39123) against herpes simplex virus in MRC-5 cells. Antimicrob. Agents Chemother. 33: Vere Hodge, R. A., D. Sutton, M. R. Boyd, M. R. Harnden, and R. L. Jarvest Selection of an oral prodrug (BRL 42810; Famciclovir) for the antiherpesvirus agent BRL [9-(4- hydroxy-3-hydroymethylbut-1-yl)guanine; penciclovir]. Antimicrob. Agents Chemother. 33: Vogt, M. W., K. L. Hartshorn, P. A. Furman, T. C. Chou, J. A. Fyfe, L. A. Coleman, C. Crumpacker, R. T. Schooley, and M. S. Hirsch Ribavirin antagonizes the effect of azidothymidine on HIV replication. Science 235: 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 the AIDS Clinical Trials Group of the National Institute of Allergy and Infectious Diseases Zidovudine in asymptomatic human immunodeficiency virus infection. N. Engl. J. Med. 322: Vrang, L., and B. Oberg PPi analogs as inhibitors of human T-lymphotropic virus type III reverse transcriptase. Antimicrob. Agents Chemother. 29: Wade, J. C., R. W. McGuffin, S. C. Springmeyer, B. Newton, J. W. Singer, and J. D. Meyers Treatment of cytomegaloviral pneumonia with high-dose acyclovir and human leukocyte interferon. J. Infect. Dis. 148: Wade, J. C., C. McLaren, and J. D. Meyers Frequency and significance of acyclovir-resistant herpes simplex virus isolated from marrow transplant patients receiving multiple courses of treatment with acyclovir. J. Infect. Dis. 148: Wade, J. C., and J. D. Meyers Neurologic symptoms associated with parenteral acyclovir treatment after marrow transplantation. Ann. Intern. Med. 98: Wade, J. C., B. Newton, N. Flournoy, and J. D. Meyers Oral acyclovir for prevention of herpes simplex virus reactivation after marrow transplantation. Ann. Intern. Med. 100: Wahren, B., and B. Oberg Reversible inhibition of cytomegalovirus replication by phosphonoformate. Intervirology 14: Wald, E. R., D. Dashefsky, and M. Green In re ribavirin: a case of premature adjudication? J. Pediatr. 112: Walmsley, S. L., E. Chew, S. E. Read, H. Vellend, I. Salit, A. Rachlis, and M. M. Fanning Treatment of cytomegalovirus retinitis with trisodium phosphonoformate hexahydrate. J. Infect. Dis. 137: Waqar, M., M. J. Evans, K. F. Manly, R. C. Hughes, and J. A. Huberman Effects of 2',3'-dideoxynucleosides on mammalian cells and viruses. J. Cell Physiol. 121: Watts, D. H., Z. A. Brown, T. Tartaglione, S. K. Burchett, K. Opheim, R. Coombs, and L. Corey Pharmacokinetic disposition of zidovudine during pregnancy. J. Infect. Dis. 163: Weis, W., J. H. Brown, S. Cusack, J. C. Paulson, J. J. Skehel, and D. C. Wiley Structure of the influenza virus haemagglutinin complexed with its receptor, sialic acid. Nature (London) 333: Weisbart, R. H., J. C. Gasson, and D. W. Golde Colony-stimulating factors and host defense. Ann. Intern. Med. 110: Wendel, H. A Clinical and serologic effects in influenza

37 182 BEAN of 1-adamantanamine HCL-a double-blind study. Fed. Proc. 23: 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 and Infectious Diseases Collaborative Antiviral Study Group A controlled trial comparing vidarabine with acyclovir in neonatal herpes simplex virus infection. N. Engl. J. Med. 324: Whitley, R., M. Hilty, R. Haynes, Y. Bryson, J. D. Connor, S.-J. Soong, C. A. Alford, and the National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group Vidarabine therapy of varicella in immunosuppressed patients. J. Pediatr. 101: 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 the NIA1D Collaborative Antiviral Study Group Vidarabine versus acyclovir therapy in herpes simplex encephalitis. N. Engl. J. Med. 314: Whitley, R. J., L. T. Ch'ien, R. Dolin, G. J. Galasso, and C. A. Alford, Editors, and the Collaborative Study Group Adenine arabinoside therapy of herpes zoster in the immunosuppressed. N. Engl. J. Med. 294: 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 Infections caused by herpes simplex virus in the immunocompromised host: natural history and topical acyclovir therapy. J. Infect. Dis. 150: Whitley, R. J., A. J. Nahmias, S. J. Soong, G. G. Galasso, C. L. Fleming, and C. A. Alford Vidarabine therapy of neonatal herpes simplex virus infection. Pediatrics 66: Whitley, R. J., S. J. Soong, R. Dolin, R. Betts, C. Linnemann, Jr., C. A. Alford, Jr., and the NIAID Collaborative Antiviral Study Group Early vidarabine therapy to control the complications of herpes zoster in immunosuppressed patients. N. Engl. J. Med. 307: Whitley, R. J., S. J. Soong, R. Dolin, G. J. Galasso, L. T. Ch'ien, C. A. Alford, and the Collaborative Study Group Adenine arabinoside therapy of biopsy-proved herpes simplex encephalitis. N. Engl. J. Med. 297: Whitley, R. J., S. J. Soong, M. S. Hirsch, A. W. Karchmer, R. Dolin, G. Galasso, J. K. Dunnick, C. A. Alford, and the NLAID Collaborative Antiviral Study Group Herpes simplex encephalitis. N. Engl. J. Med. 304: Whitley, R. J., S. Spruance, F. G. Hayden, J. Overall, C. A. Alford, Jr., J. M. Gwaltney, Jr., S. J. Soong, and the NIAID Collaborative Antiviral Study Group Vidarabine therapy for mucocutaneous herpes simplex virus infections in the immunocompromised host. J. Infect. Dis. 149: Whitley, R. J., A. Yeager, P. Kartus, Y. Bryson, J. D. Conner, C. A. Alford, A. Nahmias, and S.-J. Soong Neonatal herpes simplex virus infection: follow-up evaluation of vidarabine therapy. Pediatrics 72: Wills, R. J Clinical pharmacokinetics of interferons. Clin. Pharmacokinet. 19: Wilson, S. Z., B. E. Gilbert, J. M. Quarles, V. Knight, H. W. McClung, R. V. Moore, and R. B. Couch Treatment of influenza A (H1N1) virus infection with ribavirin aerosol. Antimicrob. Agents Chemother. 26: Wingfield, W. L., D. Pollack, and R. R. Grunert Therapeutic efficacy of amantadine HCI and rimantadine HCI in naturally occurring influenza A2 respiratory illness in man. N. Engl. J. Med. 281: 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 the Hoffmann-La Roche Herpes Zoster Study Group Recombinant interferon alpha-2a for treatment of herpes zoster in immunosuppressed patients with cancer. Am. J. Med. 85: CLIN. MICROBIOL. REV Winston, D. J., W. G. Ho, C. H. Lin, K. Bartoni, M. D. Budinger, R. P. Gale, and R. E. Champlin Intravenous immune globulin for prevention of cytomegalovirus infection and interstitial pneumonia after bone marrow transplantation. Ann. Intern. Med. 106: Witkowski, J. T., R. K. Robins, R. W. Sidwell, and L. N. Simon Design, synthesis and broad spectrum antiviral activity of 1-fi-D-ribofuranosyl-1,2,4-triazole-3-carboxamide and related nucleosides. J. Med. Chem. 15: Woodson, R. D., and J. J. Clinton Hepatitis prophylaxis abroad. JAMA 209: World Health Organization Report of a WHO informal consultation on preclinical and clinical aspects of the use of immunomodulators in HIV infection. AIDS 4:WHO 1-WHO Worral, G Topical acyclovir for recurrent herpes labialis in primary care. Can. Fam. Physician 37: Wray, S. K., B. E. Gilbert, and V. Knight Effect of ribavirin triphoshate on primer generation and elongation during influenza virus transcription in vitro. Antiviral Res. 5: Wray, S. K., B. E. Gilbert, M. W. Noall, and V. Knight Mode of action of ribavirin: effect of nucleotide pool alterations on influenza virus ribonucleoprotein synthesis. Antiviral Res. 5: 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 Response of human-immunodeficiency-virus-associated neurological disease to 3'-azido-3'- deoxythymidine. Lancet i: Yarchoan, R., H. Mitsuya, C. E. Myers, and S. Broder Clinical pharmacology of 3'-azido-2',3'-dideoxythymidine (zidovudine) and related dideoxynucleosides. N. Engl. J. Med. 321: Yarchoan, R., J. M. Pluda, R. V. Thomas, H. Mitsuya, P. Brouwers, K. M. Wyvill, N. Hartman, D. G. Johns, and S. Broder Long-term toxicity/activity profile of 2',3'- dideoxyinosine in AIDS or AIDS-related complex. Lancet 336: Yarchoan, R., R. V. Thomas, J. Allain, N. McAtee, R. Dubinsky, 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 Phase I studies of 2',3'-dideoxycytidine in severe human immunodeficiency virus infection as a single agent and alternating with zidovudine (AZT). Lancet i: 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 Long-term administration of 3-azido- 2',3'-dideoxythymidine to patients with AIDS-related neurological disease. Ann. Neurol. 23(Suppl.):S82-S Younkin, S. W., R. F. Betts, F. K. Roth, and R. G. Douglas, Jr Reduction in fever and symptoms in young adults with influenza A/Brazil/78 HlNl infection after treatment with aspirin or amantadine. Antimicrob. Agents Chemother. 23: Zamecnik, P. C., and M. L. Stephenson Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide. Proc. Natl. Acad. Sci. USA 75: Zebedee, S. L., and R. A. Lamb Influenza A virus M2 protein: monoclonal antibody restriction of virus growth and detection of M2 in virions. J. Virol. 62: Zhu, Z., R. F. Schinazi, C. Chu, G. J. Williams, C. B. Colby, and J. Sommadossi Cellular metabolism of 3'-azido- 2',3'-dideoxyuridine with formation of 5'-O-diphosphohexose derivatives by previously unrecognized metabolic pathways for 2'-deoxyuridine analogs. Mol. Pharmacol. 38:

Antiviral Drugs. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018

Antiviral Drugs. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018 Antiviral Drugs Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018 Viruses Viruses are the smallest infective agents, consisting of nucleic acid (DNA or RNA) enclosed

More information

Structure of viruses

Structure of viruses Antiviral Drugs o Viruses are obligate intracellular parasites. o lack both a cell wall and a cell membrane. o They do not carry out metabolic processes. o Viruses use much of the host s metabolic machinery.

More information

- They come in all sizes. -- General Structure is similar.

- They come in all sizes. -- General Structure is similar. - They come in all sizes. -- General Structure is similar. Centers for Disease Control (CDC) and Prevention. Influenza Prevention and Control. Influenza. Available at: http://www.cdc.gov/ncidod/diseases/flu/fluinfo.htm.

More information

HSV DNA replication. Herpesvirus Latency. Latency and Chemotherapy. Human Herpesviruses - subtypes. Acyclovir (acycloguanosine) {Zovirax}

HSV DNA replication. Herpesvirus Latency. Latency and Chemotherapy. Human Herpesviruses - subtypes. Acyclovir (acycloguanosine) {Zovirax} Human Herpesviruses - subtypes Herpes Simplex I (HSVI) - herpes labialis (cold sores) herpes keratitis (eye infections) HSVII - herpes genitalis (genital herpes) Varicella Zoster virus (VZV) Chicken pox

More information

Antiviral Drugs. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018

Antiviral Drugs. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018 Antiviral Drugs Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018 Viruses Viruses are the smallest infective agents, consisting of nucleic acid (DNA or RNA) enclosed

More information

Steps in viral replication (I)

Steps in viral replication (I) Antiviral agents Steps in viral replication (I) Recognition of the target cell Attachment Penetration Uncoating Macromolecular synthesis Assembly of virus Buddding of enveloped viruses Release of virus

More information

Antiviral Drugs Lecture 5

Antiviral Drugs Lecture 5 Antiviral Drugs Lecture 5 Antimicrobial Chemotherapy (MLAB 366) 1 Dr. Mohamed A. El-Sakhawy 2 Introduction Viruses are microscopic organisms that can infect all living cells. They are parasitic and multiply

More information

Antiviral Agents I. Tutorial 6

Antiviral Agents I. Tutorial 6 Antiviral Agents I Tutorial 6 Viruses, the smallest of pathogens, are unable to conduct metabolic processes on their own, they use the metabolic system of the infected cell to replicate ( intracellular

More information

*viruses have no cell wall and made up of nucleic acid components.

*viruses have no cell wall and made up of nucleic acid components. Anti-viral drugs {Please read these notes together with the slides since I only wrote what the doctor added} Apologies in advance for any mistakes In this sheet we are going to talk about anti-viral drugs,

More information

Antiviral Chemotherapy

Antiviral Chemotherapy Viruses are intimate intracellular parasites and their destruction may cause destruction of infected cells. Many virus infections were considered to be self-limited. Most of the damage to cells in virus

More information

The chemical name of acyclovir, USP is 2-amino-1,9-dihydro-9-[(2-hydroxyethoxy)methyl]-6Hpurin-6-one; it has the following structural formula:

The chemical name of acyclovir, USP is 2-amino-1,9-dihydro-9-[(2-hydroxyethoxy)methyl]-6Hpurin-6-one; it has the following structural formula: Acyclovir Ointment, USP 5% DESCRIPTION Acyclovir, USP, is a synthetic nucleoside analogue active against herpes viruses. Acyclovir ointment, USP 5% is a formulation for topical administration. Each gram

More information

Antiviral Agents DEPARTEMEN FARMAKOLOGI & TERAPEUTIK FK USU. 06 August

Antiviral Agents DEPARTEMEN FARMAKOLOGI & TERAPEUTIK FK USU. 06 August Antiviral Agents Dr. Yunita Sari Pane, MS DEPARTEMEN FARMAKOLOGI & TERAPEUTIK FK USU 06 August 2009 1 VIRUSES Obligate intracellular parasites Consist of a core genome in a protein shell and some are surrounded

More information

Diagnosis of Viral Infections. Antiviral Agents. Herpes Zoster. Challenges to the Development of Effective Antiviral Agents

Diagnosis of Viral Infections. Antiviral Agents. Herpes Zoster. Challenges to the Development of Effective Antiviral Agents Diagnosis of Viral Infections Antiviral Agents Scott M. Hammer, M.D. Clinical suspicion Is syndrome diagnostic of a specific entity? Is viral disease in the differential diagnosis of a presenting syndrome?

More information

Antiviral Chemotherapy

Antiviral Chemotherapy 12 Antiviral Chemotherapy Why antiviral drugs? Vaccines have provided considerable success in preventing viral diseases; However, they have modest or often no therapeutic effect for individuals who are

More information

Antiviral Agents. Scott M. Hammer, M.D. Challenges to the Development of Effective Antiviral Agents

Antiviral Agents. Scott M. Hammer, M.D. Challenges to the Development of Effective Antiviral Agents Antiviral Agents Scott M. Hammer, M.D. Challenges to the Development of Effective Antiviral Agents Myriad number of agents Need knowledge of replication at molecular level to define targets Viruses as

More information

MID 40. Diagnosis of Viral Infections. Antiviral Therapy. Herpes Zoster. Challenges to the Development of Effective Antiviral Agents

MID 40. Diagnosis of Viral Infections. Antiviral Therapy. Herpes Zoster. Challenges to the Development of Effective Antiviral Agents Diagnosis of Viral Infections Antiviral Therapy Scott M. Hammer, M.D. Clinical suspicion Is syndrome diagnostic of a specific entity? Is viral disease in the differential diagnosis of a presenting syndrome?

More information

number Done by Corrected by Doctor

number Done by Corrected by Doctor number 34 Done by حسام ابو عوض Corrected by Waseem Alhaj Doctor مالك الزحلف Antiviral Chemotherapy (chemotherapy is another way of saying drugs بنحب نتفلسف وهيك.(بس When dealing with viruses we are entering

More information

Anti-viral drugs. Certain viruses multiply in the cytoplasm but others do in the nucleus Most multiplication take place before diagnosis is made

Anti-viral drugs. Certain viruses multiply in the cytoplasm but others do in the nucleus Most multiplication take place before diagnosis is made Anti-viral Drugs Viruses have no cell wall and made up of nucleic acid components Viruses containing envelope antigenic in nature Viruses are obligate intracellular parasite They do not have a metabolic

More information

Medicinal Chemistry. Antiviral Agents

Medicinal Chemistry. Antiviral Agents Medicinal Chemistry Antiviral Agents Classical antiviral agents More than 50 years have elapsed since the discovery of the first antiviral agents, i.e. methisazone and 5-iodo-2 -deoxyuridine. In contrast

More information

Human Herpes Viruses (HHV) Mazin Barry, MD, FRCPC, FACP, DTM&H Assistant Professor and Consultant Infectious Diseases KSU

Human Herpes Viruses (HHV) Mazin Barry, MD, FRCPC, FACP, DTM&H Assistant Professor and Consultant Infectious Diseases KSU Human Herpes Viruses (HHV) Mazin Barry, MD, FRCPC, FACP, DTM&H Assistant Professor and Consultant Infectious Diseases KSU HERPES VIRUS INFECTIONS objectives: ØTo know the clinically important HHVs. ØTo

More information

Medical Virology. Herpesviruses, Orthomyxoviruses, and Retro virus. - Herpesviruses Structure & Composition: Herpesviruses

Medical Virology. Herpesviruses, Orthomyxoviruses, and Retro virus. - Herpesviruses Structure & Composition: Herpesviruses Medical Virology Lecture 2 Asst. Prof. Dr. Dalya Basil Herpesviruses, Orthomyxoviruses, and Retro virus - Herpesviruses Structure & Composition: Herpesviruses Enveloped DNA viruses. All herpesviruses have

More information

Clinical Aspect and Application of Laboratory Test in Herpes Virus Infection. Masoud Mardani M.D,FIDSA

Clinical Aspect and Application of Laboratory Test in Herpes Virus Infection. Masoud Mardani M.D,FIDSA Clinical Aspect and Application of Laboratory Test in Herpes Virus Infection Masoud Mardani M.D,FIDSA Shahidhid Bh BeheshtiMdi Medical lui Universityit Cytomegalovirus (CMV), Epstein Barr Virus(EBV), Herpes

More information

Anti-viral drugs. Certain viruses multiply in the cytoplasm but others do in the nucleus Most multiplication take place before diagnosis is made

Anti-viral drugs. Certain viruses multiply in the cytoplasm but others do in the nucleus Most multiplication take place before diagnosis is made Anti-viral Drugs Viruses have no cell wall and made up of nucleic acid components Viruses containing envelope antigenic in nature Viruses are obligate intracellular parasite They do not have a metabolic

More information

Herpesviruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics

Herpesviruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics Herpesviruses Virion Genome Genes and proteins Viruses and hosts Diseases Distinctive characteristics Virion Enveloped icosahedral capsid (T=16), diameter 125 nm Diameter of enveloped virion 200 nm Capsid

More information

Non HIV Anti Virals Prof. Mary Klotman

Non HIV Anti Virals Prof. Mary Klotman Mary Klotman, M.D. Professor of Medicine and Microbiology Mount Sinai School of Medicine New York, NY 1 Nucleoside analogues: first line herpes antivirals Parent Oral prodrug Valganciclovir 2 Phosphorylation

More information

Treatment of respiratory virus infection Influenza A & B Respiratory Syncytial Virus (RSV)

Treatment of respiratory virus infection Influenza A & B Respiratory Syncytial Virus (RSV) Treatment of respiratory virus infection Influenza A & B Respiratory Syncytial Virus (RSV) Amantadine and Rimantadine Use is limited to Influenza A infection. Very effective in preventing infection if

More information

Size nm m m

Size nm m m 1 Viral size and organization Size 20-250nm 0.000000002m-0.000000025m Virion structure Capsid Core Acellular obligate intracellular parasites Lack organelles, metabolic activities, and reproduction Replicated

More information

VZV, EBV, and HHV-6-8

VZV, EBV, and HHV-6-8 VZV, EBV, and HHV-6-8 Anne Gershon Common Features of Herpesviruses Morphology Basic mode of replication Primary infection followed by latency Ubiquitous Ability to cause recurrent infections (reactivation

More information

Chapter 49. Antiviral Agents

Chapter 49. Antiviral Agents Chapter 49 Antiviral Agents Antiviral Drugs 1. Characters of Virus Viruses are obligate intracellular parasites their replication depends primarily on synthetic processes of the host cell. 2.Classification

More information

number Done by Corrected by Doctor مالك الزحلف

number Done by Corrected by Doctor مالك الزحلف number 33 Done by Rawan Alkhabaz & Saja Alhijja Corrected by Doctor مالك الزحلف 1 In the previous lecture, we ve talked about second generation quinolone (ciprofloxacin) which is the drug of choice for

More information

MedChem401 Herpesviridae. Herpesviridae

MedChem401 Herpesviridae. Herpesviridae MedChem401 Herpesviridae Members of the herpesvirus family have been identified in more than 80 different animal species Eight have been identified as human pathogens Herpes viruses are a leading cause

More information

Introduction to Viruses That Infect Humans: The DNA Viruses

Introduction to Viruses That Infect Humans: The DNA Viruses Chapter 24 Introduction to Viruses That Infect Humans: The DNA Viruses Copyright The McGraw-Hill Companies, Inc. Permission required for reproduction or display. 24.1 Viruses in Human Infections and Diseases

More information

ZOVIRAX ORAL FORMULATIONS GlaxoSmithKline

ZOVIRAX ORAL FORMULATIONS GlaxoSmithKline ZOVIRAX ORAL FORMULATIONS GlaxoSmithKline Aciclovir QUALITATIVE AND QUANTITATIVE COMPOSITION Tablets containing either 200 mg, 400 mg or 800 mg aciclovir; Dispersible tablets containing either 200 mg,

More information

Human Herpesviruses. VZV, EBV, and HHV-6-8. The rash of VZV is vesicular. MID 34

Human Herpesviruses. VZV, EBV, and HHV-6-8. The rash of VZV is vesicular. MID 34 VZV, EBV, and HHV-6-8 Anne Gershon Human Herpesviruses Replication (lytic infection) occurs in a cascade Latency occurs when the cascade is interrupted Transcription of viral genome and protein synthesis

More information

Virology Introduction. Definitions. Introduction. Structure of virus. Virus transmission. Classification of virus. DNA Virus. RNA Virus. Treatment.

Virology Introduction. Definitions. Introduction. Structure of virus. Virus transmission. Classification of virus. DNA Virus. RNA Virus. Treatment. DEVH Virology Introduction Definitions. Introduction. Structure of virus. Virus transmission. Classification of virus. DNA Virus. RNA Virus. Treatment. Definitions Virology: The science which study the

More information

Lecture 2: Virology. I. Background

Lecture 2: Virology. I. Background Lecture 2: Virology I. Background A. Properties 1. Simple biological systems a. Aggregates of nucleic acids and protein 2. Non-living a. Cannot reproduce or carry out metabolic activities outside of a

More information

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES 1 of 7 I. Viral Origin. A. Retrovirus - animal lentiviruses. HIV - BASIC PROPERTIES 1. HIV is a member of the Retrovirus family and more specifically it is a member of the Lentivirus genus of this family.

More information

Multiple Choice Questions - Paper 1

Multiple Choice Questions - Paper 1 Multiple Choice Questions - Paper 1 Instructions for candidates The examination consists of 30 multiple choice questions, each divided into 5 different parts. Each part contains a statement which could

More information

11/15/2011. Outline. Structural Features and Characteristics. The Good the Bad and the Ugly. Viral Genomes. Structural Features and Characteristics

11/15/2011. Outline. Structural Features and Characteristics. The Good the Bad and the Ugly. Viral Genomes. Structural Features and Characteristics Chapter 19 - Viruses Outline I. Viruses A. Structure of viruses B. Common Characteristics of Viruses C. Viral replication D. HIV II. Prions The Good the Bad and the Ugly Viruses fit into the bad category

More information

Influenza viruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics

Influenza viruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics Influenza viruses Virion Genome Genes and proteins Viruses and hosts Diseases Distinctive characteristics Virion Enveloped particles, quasi-spherical or filamentous Diameter 80-120 nm Envelope is derived

More information

Understanding Viruses CHAPTER 38. Antiviral Agents. Understanding Viruses (cont'd) Viral Infections (cont'd) Viral Infections.

Understanding Viruses CHAPTER 38. Antiviral Agents. Understanding Viruses (cont'd) Viral Infections (cont'd) Viral Infections. Understanding Viruses CHAPTER 38 Antiviral Agents Viral replication A virus cannot replicate on its own It must attach to and enter a host cell It then uses the host cell s energy to synthesize protein,

More information

ACIVIR DT Tablets (Aciclovir)

ACIVIR DT Tablets (Aciclovir) Published on: 29 Jan 2016 ACIVIR DT Tablets (Aciclovir) Composition ACIVIR-200 DT Each dispersible tablet contains Aciclovir, BP 200 mg (in a flavoured base) ACIVIR-400 DT Each dispersible tablet contains

More information

numbe r Done by Corrected by Doctor

numbe r Done by Corrected by Doctor numbe r 5 Done by Mustafa Khader Corrected by Mahdi Sharawi Doctor Ashraf Khasawneh Viral Replication Mechanisms: (Protein Synthesis) 1. Monocistronic Method: All human cells practice the monocistronic

More information

MedChem 401~ Retroviridae. Retroviridae

MedChem 401~ Retroviridae. Retroviridae MedChem 401~ Retroviridae Retroviruses plus-sense RNA genome (!8-10 kb) protein capsid lipid envelop envelope glycoproteins reverse transcriptase enzyme integrase enzyme protease enzyme Retroviridae The

More information

Bacteriophage Reproduction

Bacteriophage Reproduction Bacteriophage Reproduction Lytic and Lysogenic Cycles The following information is taken from: http://student.ccbcmd.edu/courses/bio141/lecguide/unit3/index.html#charvir Bacteriophage Structure More complex

More information

Human Immunodeficiency Virus. Acquired Immune Deficiency Syndrome AIDS

Human Immunodeficiency Virus. Acquired Immune Deficiency Syndrome AIDS Human Immunodeficiency Virus Acquired Immune Deficiency Syndrome AIDS Sudden outbreak in USA of opportunistic infections and cancers in young men in 1981 Pneumocystis carinii pneumonia (PCP), Kaposi s

More information

Viruses. Poxviridae. DNA viruses: 6 families. Herpesviridae Adenoviridae. Hepadnaviridae Papovaviridae Parvoviridae

Viruses. Poxviridae. DNA viruses: 6 families. Herpesviridae Adenoviridae. Hepadnaviridae Papovaviridae Parvoviridae Viruses DNA viruses: 6 families Poxviridae Herpesviridae Adenoviridae Hepadnaviridae Papovaviridae Parvoviridae Human herpesviruses Three subfamilies (genome structure, tissue tropism, cytopathologic effect,

More information

The head of a pin can hold five hundred million rhinoviruses (cause of the

The head of a pin can hold five hundred million rhinoviruses (cause of the The head of a pin can hold five hundred million rhinoviruses (cause of the common cold). One sneeze can generate an aerosol of enough cold viruses to infect thousands of people! DNA-containing Viruses:

More information

virology MCQs 2- A virus commonly transmitted by use of contaminated surgical tools & needles produces a disease called serum hepatitis.

virology MCQs 2- A virus commonly transmitted by use of contaminated surgical tools & needles produces a disease called serum hepatitis. virology MCQs 1- A virus which causes AIDS is: a- Small pox virus. b- Coxsackie B virus. c- Mumps virus. d- Rubella virus. e- HIV-III virus. 2- A virus commonly transmitted by use of contaminated surgical

More information

Viral reproductive cycle

Viral reproductive cycle Lecture 29: Viruses Lecture outline 11/11/05 Types of viruses Bacteriophage Lytic and lysogenic life cycles viruses viruses Influenza Prions Mad cow disease 0.5 µm Figure 18.4 Viral structure of capsid

More information

It has been estimated that 90% of individuals

It has been estimated that 90% of individuals Famciclovir for Cutaneous Herpesvirus Infections: An Update and Review of New Single-Day Dosing Indications Manju Chacko, MD; Jeffrey M. Weinberg, MD Infections with herpes simplex virus (HSV) types 1

More information

Viral genetics VIRAL GENETICS

Viral genetics VIRAL GENETICS Viral genetics Lecturer Dr Ashraf Khasawneh Department of Biomedical Sciences VIRAL GENETICS VIRUSES GROW RAPIDLY A SINGLE PARTICLE PRODUCES A LOT OF PROGENY DNA VIRUSES SEEM TO HAVE ACCESS TO PROOF READING,

More information

19 Viruses BIOLOGY. Outline. Structural Features and Characteristics. The Good the Bad and the Ugly. Structural Features and Characteristics

19 Viruses BIOLOGY. Outline. Structural Features and Characteristics. The Good the Bad and the Ugly. Structural Features and Characteristics 9 Viruses CAMPBELL BIOLOGY TENTH EDITION Reece Urry Cain Wasserman Minorsky Jackson Outline I. Viruses A. Structure of viruses B. Common Characteristics of Viruses C. Viral replication D. HIV Lecture Presentation

More information

number Done by Corrected by Doctor Ashraf

number Done by Corrected by Doctor Ashraf number 4 Done by Nedaa Bani Ata Corrected by Rama Nada Doctor Ashraf Genome replication and gene expression Remember the steps of viral replication from the last lecture: Attachment, Adsorption, Penetration,

More information

Lab 3: Pathogenesis of Virus Infections & Pattern 450 MIC PRACTICAL PART SECTION (30397) MIC AMAL ALGHAMDI 1

Lab 3: Pathogenesis of Virus Infections & Pattern 450 MIC PRACTICAL PART SECTION (30397) MIC AMAL ALGHAMDI 1 Lab 3: Pathogenesis of Virus Infections & Pattern 450 MIC PRACTICAL PART SECTION (30397) 2018 450 MIC AMAL ALGHAMDI 1 Learning Outcomes The pathogenesis of viral infection The viral disease pattern Specific

More information

Properties of Herpesviruses

Properties of Herpesviruses Herpesviruses Properties of Herpesviruses Structure and Composition Spherical icosahedron, 150-200 nm Double-stranded DNA, linear More than 35 proteins Enveloped Replication from nucleus (budding) Features

More information

PRODUCT INFORMATION H 2

PRODUCT INFORMATION H 2 PRODUCT IFORMATIO ZOVIRAX COLD SORE CREAM APPROVED AME: Aciclovir COMPOSITIO: Aciclovir 5% w/w. DESCRIPTIO: Aciclovir is a synthetic acyclic purine nucleoside analogue. Its chemical name is 9-((2-hydroxyethoxy)methyl)guanine.

More information

Immunodeficiencies HIV/AIDS

Immunodeficiencies HIV/AIDS Immunodeficiencies HIV/AIDS Immunodeficiencies Due to impaired function of one or more components of the immune or inflammatory responses. Problem may be with: B cells T cells phagocytes or complement

More information

Human Immunodeficiency Virus

Human Immunodeficiency Virus Human Immunodeficiency Virus Virion Genome Genes and proteins Viruses and hosts Diseases Distinctive characteristics Viruses and hosts Lentivirus from Latin lentis (slow), for slow progression of disease

More information

Viruses. Properties. Some viruses contain other ingredients (e.g., lipids, carbohydrates), but these are derived from their host cells.

Viruses. Properties. Some viruses contain other ingredients (e.g., lipids, carbohydrates), but these are derived from their host cells. Viruses Properties They are obligate intracellular parasites. Probably there are no cells in nature that escape infection by one or more kinds of viruses. (Viruses that infect bacteria are called bacteriophages.)

More information

Some living things are made of ONE cell, and are called. Other organisms are composed of many cells, and are called. (SEE PAGE 6)

Some living things are made of ONE cell, and are called. Other organisms are composed of many cells, and are called. (SEE PAGE 6) Section: 1.1 Question of the Day: Name: Review of Old Information: N/A New Information: We tend to only think of animals as living. However, there is a great diversity of organisms that we consider living

More information

Antifungals, antivirals, antiprotozoals, and anthelmintics

Antifungals, antivirals, antiprotozoals, and anthelmintics Antifungals, antivirals, antiprotozoals, and anthelmintics Joseph K. Ritter, PhD Asst. Prof Department of Pharmacology and Toxicology MSB Room 530 jritter@hsc.vcu.edu Difficulties associated with treatment

More information

Virus and Prokaryotic Gene Regulation - 1

Virus and Prokaryotic Gene Regulation - 1 Virus and Prokaryotic Gene Regulation - 1 We have discussed the molecular structure of DNA and its function in DNA duplication and in transcription and protein synthesis. We now turn to how cells regulate

More information

Chapter 25. 바이러스 (The Viruses)

Chapter 25. 바이러스 (The Viruses) Chapter 25 바이러스 (The Viruses) Generalized Structure of Viruses 2 2 Virus Classification Classification based on numerous characteristics Nucleic acid type Presence or absence of envelope Capsid symmetry

More information

number Done by Corrected by Doctor Ashraf

number Done by Corrected by Doctor Ashraf number 6 Done by حسام أبو عوض Corrected by Doctor Ashraf 1 Antiviral Drugs When dealing with drugs, it is always important to keep an eye on what you want to target (viruses here), so here is a reminder

More information

Valaciclovir Valtrex 500mg Tablets

Valaciclovir Valtrex 500mg Tablets Valaciclovir Valtrex 500mg Tablets PRODUCT DESCRIPTION Each white to off-white, biconvex, elongated, unscored, film-coated tablet branded with GX CF1 in blue details on one side contains 500mg of Valaciclovir.

More information

Antiviral drugs. E.H.H. Wiltink and R. Janknegt

Antiviral drugs. E.H.H. Wiltink and R. Janknegt Antiviral drugs E.H.H. Wiltink and R. Janknegt ntroduction Viruses have too simple a structure to multiply themselves. For multiplication, a virus invades a cell, using the biochemical mechanisms of this

More information

General Properties of Viruses

General Properties of Viruses 1 I. Viruses as Agents of Disease. V. F. Righthand, Ph.D. August 15, 2001 General Properties of Viruses Viruses can infect every form of life. There are hundreds of different viruses that can produce diseases

More information

Dr. Ahmed K. Ali. Outcomes of the virus infection for the host

Dr. Ahmed K. Ali. Outcomes of the virus infection for the host Lec. 9 Dr. Ahmed K. Ali Outcomes of the virus infection for the host In the previous few chapters we have looked at aspects of the virus replication cycle that culminate in the exit of infective progeny

More information

LESSON 4.6 WORKBOOK. Designing an antiviral drug The challenge of HIV

LESSON 4.6 WORKBOOK. Designing an antiviral drug The challenge of HIV LESSON 4.6 WORKBOOK Designing an antiviral drug The challenge of HIV In the last two lessons we discussed the how the viral life cycle causes host cell damage. But is there anything we can do to prevent

More information

Herpes Simplex Virus Resistance to Acyclovir and Penciclovir after Two Decades of Antiviral Therapy

Herpes Simplex Virus Resistance to Acyclovir and Penciclovir after Two Decades of Antiviral Therapy CLINICAL MICROBIOLOGY REVIEWS, Jan. 2003, p. 114 128 Vol. 16, No. 1 0893-8512/03/$08.00 0 DOI: 10.1128/CMR.16.1.114 128.2003 Copyright 2003, American Society for Microbiology. All Rights Reserved. Herpes

More information

19/06/2013. Viruses are not organisms (do not belong to any kingdom). Viruses are not made of cells, have no cytoplasm, and no membranes.

19/06/2013. Viruses are not organisms (do not belong to any kingdom). Viruses are not made of cells, have no cytoplasm, and no membranes. VIRUSES Many diseases of plants and animals are caused by bacteria or viruses that invade the body. Bacteria and viruses are NOT similar kinds of micro-organisms. Bacteria are classified as living organisms,

More information

MONTGOMERY COUNTY COMMUNITY COLLEGE CHAPTER 13: VIRUSES. 1. Obligate intracellular parasites that multiply in living host cells

MONTGOMERY COUNTY COMMUNITY COLLEGE CHAPTER 13: VIRUSES. 1. Obligate intracellular parasites that multiply in living host cells MONTGOMERY COUNTY COMMUNITY COLLEGE CHAPTER 13: VIRUSES I. CHARACTERISTICS OF VIRUSES A. General Characteristics 1. Obligate intracellular parasites that multiply in living host cells 2. Contain a single

More information

ACIVIR I.V. Injection (Aciclovir)

ACIVIR I.V. Injection (Aciclovir) Published on: 22 Sep 2014 ACIVIR I.V. Injection (Aciclovir) Composition ACIVIR I.V. Each ml contains: Aciclovir BP... 25 mg Water for Injection IP...q.s. Dosage Form Injection for I.V. use Pharmacology

More information

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University Medical Virology Immunology Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University Human blood cells Phases of immune responses Microbe Naïve

More information

Alphaherpesvirinae. Simplexvirus (HHV1&2/ HSV1&2) Varicellovirus (HHV3/VZV)

Alphaherpesvirinae. Simplexvirus (HHV1&2/ HSV1&2) Varicellovirus (HHV3/VZV) Alphaherpesvirinae Simplexvirus (HHV1&2/ HSV1&2) Varicellovirus (HHV3/VZV) HERPES SIMPLEX VIRUS First human herpesvirus discovered (1922) Two serotypes recognised HSV-1 & HSV-2 (1962) HSV polymorphism

More information

Antibacterials and Antivirals

Antibacterials and Antivirals Structure of a Bacterium: Antibacterials and Antivirals Capsule: protective layer made up of proteins, sugars and lipids Cell wall: provides the bacteria with its shape and structure Cell membrane: permeable

More information

Persistent Infections

Persistent Infections Persistent Infections Lecture 17 Biology 3310/4310 Virology Spring 2017 Paralyze resistance with persistence WOODY HAYES Acute vs persistent infections Acute infection - rapid and self-limiting Persistent

More information

VALCIVIR Tablets (Valacyclovir hydrochloride)

VALCIVIR Tablets (Valacyclovir hydrochloride) Published on: 10 Jul 2014 VALCIVIR Tablets (Valacyclovir hydrochloride) Composition VALCIVIR-500 Tablets Each film-coated tablet contains: Valacyclovir Hydrochloride equivalent to Valacyclovir.. 500 mg

More information

INFLUENZA VIRUS. INFLUENZA VIRUS CDC WEBSITE

INFLUENZA VIRUS. INFLUENZA VIRUS CDC WEBSITE INFLUENZA VIRUS INFLUENZA VIRUS CDC WEBSITE http://www.cdc.gov/ncidod/diseases/flu/fluinfo.htm 1 THE IMPACT OF INFLUENZA Deaths: PANDEMICS 1918-19 S p a n is h flu 5 0 0,0 0 0 U S 2 0,0 0 0,0 0 0 w o rld

More information

Herpes Simplex Viruses: Disease Burden. Richard Whitley The University of Alabama at Birmingham Herpes Virus Infection and Immunity June 18-20, 2012

Herpes Simplex Viruses: Disease Burden. Richard Whitley The University of Alabama at Birmingham Herpes Virus Infection and Immunity June 18-20, 2012 Herpes Simplex Viruses: Disease Burden Richard Whitley The University of Alabama at Birmingham Herpes Virus Infection and Immunity June 18-20, 2012 Mucocutaneous HSV Infections Life-Threatening HSV Diseases

More information

Herpes Simplex Virus 1-2

Herpes Simplex Virus 1-2 Yamilet Melendez Microbiology 1 Presentation Herpes Simplex Virus 1-2 Introduction Herpes viruses are a leading cause of human viral diseases, second only to influenza and cold viruses Are capable of causing

More information

Coronaviruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics

Coronaviruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics Coronaviruses Virion Genome Genes and proteins Viruses and hosts Diseases Distinctive characteristics Virion Spherical enveloped particles studded with clubbed spikes Diameter 120-160 nm Coiled helical

More information

BIT 120. Copy of Cancer/HIV Lecture

BIT 120. Copy of Cancer/HIV Lecture BIT 120 Copy of Cancer/HIV Lecture Cancer DEFINITION Any abnormal growth of cells that has malignant potential i.e.. Leukemia Uncontrolled mitosis in WBC Genetic disease caused by an accumulation of mutations

More information

STRUCTURE, GENERAL CHARACTERISTICS AND REPRODUCTION OF VIRUSES

STRUCTURE, GENERAL CHARACTERISTICS AND REPRODUCTION OF VIRUSES STRUCTURE, GENERAL CHARACTERISTICS AND REPRODUCTION OF VIRUSES Introduction Viruses are noncellular genetic elements that use a living cell for their replication and have an extracellular state. Viruses

More information

Chapter 13 Viruses, Viroids, and Prions. Biology 1009 Microbiology Johnson-Summer 2003

Chapter 13 Viruses, Viroids, and Prions. Biology 1009 Microbiology Johnson-Summer 2003 Chapter 13 Viruses, Viroids, and Prions Biology 1009 Microbiology Johnson-Summer 2003 Viruses Virology-study of viruses Characteristics: acellular obligate intracellular parasites no ribosomes or means

More information

Chapter13 Characterizing and Classifying Viruses, Viroids, and Prions

Chapter13 Characterizing and Classifying Viruses, Viroids, and Prions Chapter13 Characterizing and Classifying Viruses, Viroids, and Prions 11/20/2017 MDufilho 1 Characteristics of Viruses Viruses Minuscule, acellular, infectious agent having either DNA or RNA Cause infections

More information

Orthomyxoviridae and Paramyxoviridae. Lecture in Microbiology for medical and dental medical students

Orthomyxoviridae and Paramyxoviridae. Lecture in Microbiology for medical and dental medical students Orthomyxoviridae and Paramyxoviridae Lecture in Microbiology for medical and dental medical students Orthomyxoviridae and Paramyxoviridae are ss RNA containng viruses Insert Table 25.1 RNA viruses 2 SIZE

More information

HIV - Life cycle. HIV Life Cyle

HIV - Life cycle. HIV Life Cyle Human Immunodeficiency Virus Retrovirus - integrated into host genome ne single-strand RA 7,000 bases HIV1 > HIV2 > HIV0 Pathology Destruction of CD4+ T lymphocytes Loss of immune function pportunistic

More information

Antivirals for Avian Influenza Outbreaks

Antivirals for Avian Influenza Outbreaks Antivirals for Avian Influenza Outbreaks Issues in Influenza Pandemic Preparedness 1. Surveillance for pandemic preparedness eg. H5N1 2. Public health intervention eg. efficacy, feasibility and impact

More information

الحترمونا من خري الدعاء

الحترمونا من خري الدعاء الحترمونا من خري الدعاء Instructions for candidates The examination consists of 30 multiple choice questions, each divided into 5 different parts. Each part contains a statement which could be true or

More information

7.012 Quiz 3 Answers

7.012 Quiz 3 Answers MIT Biology Department 7.012: Introductory Biology - Fall 2004 Instructors: Professor Eric Lander, Professor Robert A. Weinberg, Dr. Claudette Gardel Friday 11/12/04 7.012 Quiz 3 Answers A > 85 B 72-84

More information

Respiratory Viruses. Respiratory Syncytial Virus

Respiratory Viruses. Respiratory Syncytial Virus Adam Ratner, MD Respiratory Viruses Respiratory viruses are among the most common causes of disease throughout life. Often mild and self-limited, they are still associated with tremendous economic and

More information

Viruses. CLS 212: Medical Microbiology Miss Zeina Alkudmani

Viruses. CLS 212: Medical Microbiology Miss Zeina Alkudmani Viruses CLS 212: Medical Microbiology Miss Zeina Alkudmani History Through the 1800s, many scientists discovered that something smaller than bacteria could cause disease and they called it virion (Latin

More information

Viral Diseases. Question: 5/17/2011

Viral Diseases. Question: 5/17/2011 Viral Diseases Question: What is the likely reason for the dramatic increase in deaths due to heart disease and cancer in 1997 compared to 1900? 1. poor lifestyle choices (high fat diets, smoking, lack

More information

VIRUSES. 1. Describe the structure of a virus by completing the following chart.

VIRUSES. 1. Describe the structure of a virus by completing the following chart. AP BIOLOGY MOLECULAR GENETICS ACTIVITY #3 NAME DATE HOUR VIRUSES 1. Describe the structure of a virus by completing the following chart. Viral Part Description of Part 2. Some viruses have an envelope

More information

Herpesviruses. -Recurrence: clinically obvious disease due to reactivation. **Reactivation and recurrence are used interchangeably.

Herpesviruses. -Recurrence: clinically obvious disease due to reactivation. **Reactivation and recurrence are used interchangeably. *Herpesviruses: A large group of viruses (100 strains), but we are concerned with only 8 strains as they are the only ones to infect human beings *herpesviruses groups: HSV-1 HSV-2 VZV CMV EBV HHV-6 HHV-7

More information

Antivirals. Lecture 20 Biology 3310/4310 Virology Spring 2017

Antivirals. Lecture 20 Biology 3310/4310 Virology Spring 2017 Antivirals Lecture 20 Biology 3310/4310 Virology Spring 2017 You can t go back and you can t stand still. If the thunder don t get you, then the lightning will. JERRY GARCIA The Wheel (lyrics by Robert

More information

ARV Mode of Action. Mode of Action. Mode of Action NRTI. Immunopaedia.org.za

ARV Mode of Action. Mode of Action. Mode of Action NRTI. Immunopaedia.org.za ARV Mode of Action Mode of Action Mode of Action - NRTI Mode of Action - NNRTI Mode of Action - Protease Inhibitors Mode of Action - Integrase inhibitor Mode of Action - Entry Inhibitors Mode of Action

More information