Clinical Research Using 3'-Azido-2'3'-Dideoxythyrnidine (AZT) and Related Dideoxynucleosides in the Therapy of AIDS
 
S. Broder     Hämatol. Bluttransf. Vol 35

National Cancer Institute, Bethesda, Maryland 20892, USA.


Introduction

Pathogenic retroviruses play an etiologic role in the acquired immune deficiency syndrome (AIDS) and its related disorders. While no cure for diseases caused by these agents is available, we are now in an era in which therapy against pathogenic retroviruses is a practical reality. Therapies against the etiologic agent of AIDS [13] were made possible by the discovery that a retrovirus, now called the human immunodeficiency virus (HIV), caused the disorder [4-6]. This discovery and the ability to grow the virus in large quantities enabled the development of in vitro techniques to find drugs that inhibit the replication of HIV [7, 8]. Substances that acted against HIV in vitro could then be identified for further research, and the orderly development of drugs thus proceeded. The work was in part a outgrowth of early research on animal retroviral systems in a number of laboratories [9-12]. No one person or group can take full credit for these discoveries, and a great debt is owed to many scientists who pioneered this research. More recently, our group and other groups have observed that certain members of a class of compounds called dideoxynucleosides are potent inhibitors in vitro of the replication of HIV in human T cells [8, 13-26]. In all the compounds, the hydroxy (-OH) group in the 3'-position on the sugar ring is replaced by a hydrogen atom ( -H) or National Cancer Institute, Bethesda, Maryland 20892, USA. another group that cannot form phosphodiester linkages. 3'-Azido-2',3'-dideoxythyrnidine (also called zidovudine, 3'-azido-3'-deoxythyrnidine, azidothyrnidine, or AZT), the first of these compounds to be tested clinically, reduced the morbidity and mortality associated with severe HIV infection [27, 28]. Volberding et al. [29] reported that AZT is effective in delaying progression to fulminant AIDS in asymptomatic patients infected with HIV. Other dideoxynucleosides are now in various stages of clinical testing. Other substances that act at various stages of HIV's replicative cycle also have been shown to block replication in vitro, and some are undergoing clinical testing [7, 31-59]. This article reviews certain clinical applications of one antiretroviral agent, AZT, and discusses the status of several related compounds. It also addresses other approaches to antiretroviral treatment. While an ultimate cure for AIDS will require further basic research, the knowledge already at hand might make a major impact against the death and suffering frorn this disease in the coming decade. During the decade of the nineties, AIDS is expected to increase, and the disease is likely to become a major cause of death in men, women, and children. In some parts of the world, infant and child mortality could be as much as 30% greater than what one would have expected [60].


Potential Mechanisms of Action Against HIV

Although there are substantial differences among the dideoxynucleosides, we can make several general comments about their mechanisms of action. Each drug likely inhibits reverse transcriptase but must first be activated to a 5'-triphosphate form by various enzymes of a target cell [61-65]. It is the triphosphate form that is active against HIV [61, 66]. The activation process, called anabolic phosphorylation, involves aseries of enzymes (kinases) [61-65, 67]. AZT and other dideoxynucleosides, as triphosphates, exert their antiretroviral activity at the level of reverse transcriptase (viral DNA polymerase) [12, 61, 66, 68, 69]. Reverse transcriptase is essential in the replicative cycle of HIV. A great deal is now known about the overall structure of the polymerase domain of HIV reverse transcriptase, about the active site, and about the secondary structure within the active site (e.g., see [69]. When HIV enters a target cell, this enzyme makes a complementary strand DNA copy of the viral genomic RNA and then catalyzes the production of a second, positive strand DNA copy. The genetic information of HIV is thus encoded in a double-stranded form of DNA. Two mechanisms may contribute to the effect of AZT and other dideoxynucleosides on reverse transcriptase. First, as triphosphates, they compete with the cellular deoxynucleoside-5'triphosphates that are essential substrates for the formation of proviral DNA by reverse transcriptase with inhibition constants generally in the range of 0.005-0.2 µM [61, 70- 73]. These concentrations can be attained in cells exposed to the drugs [61- 65]. Second, such 5'triphosphatases act as chain terminators in the synthesis of proviral DNA. Because of the 3' modification of these compounds, once viral reverse transcriptase adds them to a growing chain of viral DNA, the DNA is elongated by exactly one residue and then terminated [66]. In contrast to HIV reverse transcriptase, LXXXIV mammalian DNA polymerase alpha is relatively resistant to the effects of these drugs (inhibition constants, 100-230 µM) [12, 61, 63, 70], which is one reason for their selective antiretroviral activity in cells that can phosphorylate them. However, mammalian DNA polymerase gamma, found in mitochondria, and DNA polymerase beta are also sensitive to these compounds (inhibition constants, 0.016-0.µM and 2.6-70 µM, respectively), and this may be a basis for drug toxicity [12,61,63,70]. lt should always be borne in mind that additional mechanisms of activity and toxicity might be at work. It is possible that AZT works through an as yet unidentified intermediate. AZT and related dideoxynucleosides have activity against certain retroviruses, including HIV type 2, human T -celllymphotropic virus type I, animal lentiviruses, and murine retroviruses [9 -12, 68,74- 78]. (Some congeners also have in vitro activity in an animal model of hepatitis B virus [79] that, although a DNA virus, replicates through an RNA intermediate using areverse transcriptase-like DNA polymerase [80].) This conservation of activity suggests that the sensitivity of reverse transcriptase to these agents (as triphosphates) is linked to an essential feature of the viral enzyme. Nevertheless, two groups recently reported that certain isolates of HIV from patients who had taken AZT for 6 months or longer had reduced sensitivity to AZT in vitro [81, 82]. Some of the known mutations would be likely to affect the charge or alpha helix content of the catalytic site, or at least the probable catalytic site, of reverse transcriptase. Larder et al. [83] previously showed that the induction in vitro of certain mutations in reverse transcriptase by site directed mutagenesis could make the enzyme less sensitive to inhibition of AZT triphosphate and phosphonoformate (but also reduce enzymatic activity). However, HIV isolated from patients receiving long-term AZT therapy retains its in vitro sensitivity to inhibition by most other dideoxynucleosides (such as 2',3'-dideoxycytidine) and phosphonoformate [82]. Also, preliminary studies suggest that the sensitivity of the reverse transcriptase obtained from these resistant viral isolates to 3'-azido-2',3'dideoxythymidine- 5' -triphosphate (AZT- TP) did not change [82]. These preliminary findings suggest that while changes in reverse transcriptase may account for the loss of sensitivity to AZT, additional studies are needed to clarify these issues. Many clinical investigators believe that the emergence of AZT -insensitive isolates is a marker of impending clinical progression, but the clinical importance of the reduced viral sensitivity to AZT is not fully known. The potential problem, however, underscores the urgency for additional experimental therapeutic agents and for regimens that employ multiple agents.


Biochemical Pharmacology of AZT

Unlike most nucleosides that enter cells by specialized transport systems, AZT can enter mammalian cells by passive, nonfacilitated diffusion [84]. Once inside, the drug is phosphorylated to a triphosphate form by aseries of kinases that usually phosphorylate thymidine [51].3'Azido- 2' ,3 ' -dideoxyth ymidine- 5' -mono phosphate (AZT -MP) is first produced by thymidine kinase, and two additional phosphates are then added by the sequential action of thymidylate kinase and nucleotide diphosphate kinase to form 3'-azido-2',3'-dideoxythymidine5'-triphosphate (AZT -TP), the active moiety [61, 72, 73]. The addition of a second phosphate group to AZT-MP by thymidylate kinase is probably the ratelimiting step for this process [61]. The reaction occurs much more slowly (relative maximal velocity, 0.3% ) than the phosphorylation of thymidine-5'-monophosphate, the usual substrate for this enzyme [61], and human cells exposed to AZT accumulate relatively high levels of AZT -MP , but low levels of AZT- TP [61]. AZT-MP binds efficiently to thymidylate kinase, but comes off slowly, thus tying up the enzyme. In certain T -cell lines exposed to very high concentrations of AZT (50-200 uuM), decreased phosphorylation of thymidine and decreased levels of thymidine-5'-triphosphate (the normal DNA building block that competes with AZT -TP for reverse transcriptase) have been reported [61, 85]. This may result from the inhibition of thymidylate kinase by AZT -MP (inhibition constant, 8.6 µM) [61]. However, decreased concentrations of thymidine5'-triphosphate have not been found in other cell lines or with lower concentrations of AZT [85, 86], and whether thymidine kinase inhibition contributes to the bone marrow toxicity induced by AZT is uncertain. Furman et al. [61] originally reported that the levels of deoxycytidine-5'-triphosphate, another building block of DNA, fell in the presence of AZT, but later they concluded that the finding was due to a technical error [87]. It should be stressed that murine cells handle AZT in a very different way from human cells.


Pharmacokinetics of AZT

In culture, AZT inhibits new HIV infection of Iymphocytes at levels of 1- 5 !lM ( even under conditions of high multiplicity of infection) [8]. The initial pharmacokinetic studies demonstrated that AZT absorbed weIl orally (average oral bioavailability, 63% ) and that peak plasma levels of 3-4 µM are attained 30-90 min after an oral dose of 200 mg is ingested [27, 88, 89]. The peak plasma concentration is proportional to the amount administered, over a wide range of doses [89]. The serum half-life is only 1.1 h [27,89,90]. For this reason, a dosing every 4 h was chosen for phase II testing in patients with advanced disease and then adopted as the recommended schedule. It should be stressed, however, that there is not yet enough information to determine the optimal dose or dosing schedules for AZT. Less frequent dosing schedules and total daily doses work, but further research is needed. It is likely that total doses of approximately 600 mg per day using an interval of eight hours between doses would be adequate in most adults. The levels of intracellular AZT -TP (the activity moiety of AZT) have a half life of about 3 h [61]. Thus, effective anti HIV activity may theoretically be attained with an oral dosing interval of 8 or 12 h. Alternatively, for the optimal therapeutic benefit, it may be necessary to maintain constant plasma drug levels. Resolving these issues will require careful controlled trials. Approximately 15% -20% of an administered dose of AZT is excreted unchanged in the urine, and 75% is metabolized by hepatic glucuronidation to form 3'-azido-2',3'-dideoxy-5'-glucuronylthyrnidine, an apparently inert metabolite that is also excreted into the urine [90]. The enzyme or enzymes responsible for the glucuronidation of AZT may be inhibited by other compounds that share this pathway, and such compounds may prolong the half-life of the drug [91]. In this regard, probenecid inhibits both hepatic glucuronidation and renal excretion and thus reduces the total body clearance of AZT by 65 %. Other drugs that undergo hepatic glucuronidation and may, theoretically, inhibit the metabolism of AZT include nonsteroidal anti-inflammatory agents, narcotic analgesics, and sulfonamide antibiotics. Until the interactions of such drugs with AZT have been carefully investigated, clinicians should be aware that they may affect the metabolism of AZT, and AZT may affect theirs. Finally, the metabolism of AZT could be slower in patients with severe hepatic disease. It should be stressed that different nucleoside analogs will show individual pharmacokinetic profiles [27- 30, 32, 63, 89-98]. Because HIV can infect cells in the central nervous system and cause dementia, antiretroviral agents used in the treatment of AIDS should be capable of penetrating the brain. Three to four hours after a dose of AZT has been administered, levels in cerebrospinal fluid are approximately 60% of those in plasma (range, 10%-156%) [27, 38, 89, 99], which indicates that AZT can enter the brain by diffusion from the cerebrospinal fluid or possibly through capillaries in the brain. The clinical improvernent that occurs in patients with HIV dementia who are given AZT suggests that the drug reaches the site of viral replication in the central nervous system [28,99]. However, AZT may not cross the blood-brain barrier in all species [100]. Evidence suggests that cells belonging to the monocyte-rnacrophage series are the most important target cells of HIV infection in the brain [101, 102]. These nonproliferating cells have lower levels of kinases than Iyrnphocytes [103, 104] and dideoxynucleosides may not be efficiently phosphorylated in them. One study indicated that AZT is poorly phosphorylated in peripheral blood monocytes and macrophages and does not protect these cells against infection by the Iyrnphadenopathy-associated virus strain of HIV in vitro [104]. This study is not correct. Two subsequent studies demonstrated that low concentrations of AZT and other dideoxynucleosides profoundIy inhibited the replication of a monocytotropic strain of HIV in monocytes and macrophages [105, 106]. These results are consistent with the observation that dementia induced by HIV may be at least ternporarily reversed by AZT [21, 99, 107]. The potent activity of AZT in monocytes and macrophages can perhaps be explained by the observation that such cells have very low levels ofthyrnidine-5'triphosphate, the normal nucleotide that competes with AZT -TP at the level of reverse transcriptase [105]. Thus, the ratio of AZT -TP to thyrnidine-5'-triphosphate may actually be higher in monocytes than it is in T cells [105]. Additional studies indicate that granulocyte-rnonocyte colony-stimulating factor, which stimulates the replication of HIV in monocytes [108, 109], increases the entry of AZT into these cells and potentiates its activity against HIV [109]. However, it is important to use caution in extrapolating these data to clinical applications.


Clinical Application of AZT

In the initial clinical studies of AZT at the National Cancer Institute and Duke University Medical Center, patients with AIDS or AIDS-related complex had immunologic, virologic, and clinical improvement during 6weeks of therapy [27,88]. (Three of the 19 patients from the original phase I study, each of whom had AIDSrelated complex or Kaposi's sarcoma when they entered the trial, were alive 3.5 years after the initiation of therapy .) Also, several patients with HIV dementia who were given AZT had substantial improvement in their intellectual function, accompanied in some by a normalization in the pattern of use of cerebral glucose (as assessed by positron emission tomography) [99, 100]. On the basis of these results, Wellcome Research Laboratories began a multicenter, randomized, controlled trial of AZT in February 1986 among 282 patients with AIDS (after their first episode of Pneumocystis carinii pneumonia) or severe AIDS-related complex [29,111]. The trial demonstrated a reduced mortality in the patients receiving AZT. By September 1986,19 patients taking placebo but only one taking the drug had died [28]. After 36 weeks, 39.3% of those taking placebo had died compared with 6.2% of those taking AZT, and after 52 weeks the cumulative mortality in the patients treated with AZT was still only 10.3 % (no comparable figure is available for the placebo group, because most were given AZT after September 1986, thus ending the control arm) [28]. Suppressive prophylaxis for Pneumocystis pneumonia was not a formal protocol option in the 7 month randomized trial, although a few patients randomly distributed between the two arms received it. Also, 19 of the 144 originally assigned to AZT received more than 6 weeks of such prophylaxis during the 52 weeks after their entry into the study. Eliminating these patients from the analysis does not affect the basis conclusion of the trial. Subsequent studies have suggested that patients with AIDS who receive AZT in conjunction with prophylactic therapy for Pneumocystis pneumonia may have a lower mortality than those who receive AZT alone [112]. However, it is important to stress that the precise role of Pneumocystis pneumonia prophylaxis in patients who are receiving antiretroviral therapy has not been defined. The phase II study also showed that patients receiving AZT had a temporary increase in their CD4 + Iymphocyte counts (average, 80/mm3), fewer opportunistic infections, and an average weight gain of about 0.5 kg, as compared with those receiving placebo [28]. Furthermore, the results confirmed an observation in the phase I study; some patients with cognitive dysfunction induced by HIV improved when given AZT [99,107], Finally, patients who received AZT had a decreased viral load as compared with the placebo group in assessments made by measuring levels of serum HIV p 24 antigen [113]. The ability to isolate HIV from cultured Iymphocytes, however, was not affected, although there was a delay in the appearance of HIV in cultures [114]. Although serum HIV p 24 antigen levels are an experimental clinical measure of HIV replication, they may be affected by antibodies to p 24 and other factors. Better methods of assessing viral load are urgently needed. The polymerase chain reaction will likely prove useful in this regard [115]. On the basis of this trial's results, AZT was approved for the treatment of severe HIV infection in most countries, In the United States, it was approved in March 1987 for patients who have had P. carinii pneumonia or whose CD4+ cell count is below 200/mm³. Evidence from the New York State Department of Health indicates that the survival of patients given a diagnosis of AIDS in 1987 increased substantially over that of patients whose disease was diagnosed in previous years. It is exceedingly likely that the widespread use of AZT contributed to this trend [116]. The dose used in the phase II study is now known to be higher than is necessary for optimal effects. In both the phase I and phase II trials, it became apparent that the increase in the number of CD4+ Iymphocytes induced by AZT may be transient [27, 29, 117], particularly in patients with fulminant AIDS, whose CD4 count often returns to baseline after 16- 20 weeks of therapy [28]. In this trend, the contributions of direct drug toxicity, altered host defense mechanisms, and changes in viral sensitivity to AZT are unclear. It is highly probable that no antiretroviral agent will work to maximal advantage if the host immune response is severely damaged. Some patients have had late increases in levels of HIV p 24 antigen even while receiving a constant dose of AZT [118]. In addition, AZT has a number of toxic effects; the most frequent is suppression of bone marrow cells, and anemia is its most frequent manifestation [27,28, 111,117]. An increase in the mean corpuscular volume of erythrocytes often occurs before frank anemia [27, 111 ], but the dose should not be modified on the basis of this measure. Patients receiving AZT may have megaloblastic changes in bone marrow, maturational arrest of erythrocyte lineage, or hypoplastic (rarely aplastic) changes. Hypoplastic changes can occur without an increase in mean corpuscular volume [119,120]. Bone marrow toxicity occurs more frequently in patients with established AIDS, and in the phase II study, 45% of the patients who had P. carinii pneumonia required transfusions or a reduction in dose during the first 6 months of AZT therapy [111]. In a subsequent open trial, only 21 % of the patients with AIDS could complete 6 months of full-dose AZT treatment without a reduction in dose or the interruption of therapy [121]. Marrow toxicity is also more frequent in patients with underlying anemia, low CD4+ cell counts, or low (or low-normal) serum folic acid or vitamin B12 levels before therapy begins [27, 67, 111]. Pending further study, vitamin-replacement therapy may be useful in patients with low levels of these vitamins. The platelet count is generally spared until late in the course of AZT therapy. In fact, the drug can actually induce increased platelet counts in patients with thrombocytopenia induced by HIV [122, 123]. Other toxic effects of AZT include nausea, vomiting, myalgias, myositis (particularly in patients who receive the drug for more than a year), headaches, abnormalities of liver function, and bluish nail pigmentation [27,111,124126]. Very high doses can cause anxiety, confusion, and tremulousness [27, 117, 127]. These symptoms occasionally develop in patients receiving the current recommended dosage. Finally, a few patients have had seizure, an encephalopathy similar to Wernicke's or Stevens Johnson syndrome [128-130]. Thus, although AZT decreases morbidity and mortality among patients with severe HIV infection, its use can be associated with substantial toxicity, particularly in those with advanced disease. Slightly more than 2 years after AZT was first observed to inhibit the replication of HIV in vitro, it was approved by the Food and Drug Administration for the treatment of AIDS. Because of this extraordinarily rapid development, a number of questions regarding its use remain unanswered. In AIDS, as in perhaps no other condition, the line between approved and experimental therapy is difficult to draw. Physicians frequently ask whether AZT should be administered early in the course of HIV infection. The drug appears to be relatively weil tolerated at this stage [131], and recent results of a randomized trial have indicated that HIVseropositive patients with less than 500 CD4 cells/mm3 who were given AZT had less frequent progression to severe AIDS-related complex or AIDS than those given placebo [29]. As we learn more about the factors that make a progression to AIDS highly likely (e.g., high serum HIV p 24 antigen levels or ß2-microglobulinernia) [132, 133], it may be possible to target AZT therapy to patients who are at high risk. At present, we cannot say that starting AZT early in an asymptomatic phase of HIV infection provides a survival advantage over waiting until more symptomatic disease supervlenes. In considering early intervention with AZT, it is of particular concern that the drug may be carcinogenic or mutagenic [134]. Its long-term effects are unknown. Rodents exposed to high doses of AZT for long periods can develop vaginal neoplasms (principally nonmetastasizing squamous cell carcinomas). Male rodents did not develop tumors. The implications of these sex- and site-specific tumorigenic effects in rodents are not clear at this time, but the results provide a warning against complacency when using this class of drug. It is worth ernphasizing again that AIDS itself makes the development of certain cancers more likely, and AZT may be associated with the higher incidence of cancers in patients whose immunosurveillance mechanisms are disturbed, simply because it increases their longevity. This has occurred in certain immunodeficiency disorders of childhood, in which advances in the treatment of infections have allowed patients to survive longer [135]. Lymphomas have developed in a significant subset of the original phase I patients between 1 and 3 years after AZT therapy began. The use of AZT in children with AIDS is an area that is only now being investigated. The high incidence in certain cities of cord blood sampIes that are seropositive for HIV (for example, 1 in 80 newborns in New York Cityare seropositive [116,136]) indicates that the number of children with AIDS will grow in the near future. AIDS is dramatically altering the landscape of obstetrical and pediatric care in many countries. The manifestations of HIV infection in children can differ from those in adults. Neurologic dysfunction and high-grade bacterial infections are much more evident, for example [137], and the patterns of drug toxicity may differ. Ongoing studies at the National Cancer Institute suggest that administration of AZT by continuous intravenous infusion can reverse certain neurologic symptoms associated with HIV in children with AIDS [138]. In some patients, the intelligence quotients returned to what they had been before the disease developed [138]. However, the problems of bone marrow suppression limit that treatment. In both adults and children who have dementia associated with AIDS, considerable bone marrow suppression may be tolerable if antiretroviral therapy can reverse major neurologic deficits. Another unresolved issue is whether AZT can prevent new HIV infection if it is given at the time of viral exposure. Kittens can be protected against feline leukernia virus (a retrovirus) by the administration of AZT at the time of infection [78]. Also, fetal mice can be protected against retroviral infection by the administration of AZT to their mothers [139]. A short course of AZT at the time of exposure (e.g., after a serious needle-stick or laboratory accident) may therefore be useful. However, because it is mutagenic (and carcinogenic in rodents) and can induce chromosomal abnormalities [134], its use in such a setting cannot be recommended except in an approved protocol. Finally, even in patients for whom AZT is recommended, there is much to learn. The available evidence suggests but does not prove that patients should continue to receive therapeutic doses for as long as they can tolerate the drug. AZT was determined to be effective because it lowered the risk of opportunistic infections and prolonged life. The late decline in the CD4+ cell counts is thus not an indication to stop therapy. However, it is not yet clear how patients who have hernatologic toxicity, evidence of clinical progression, or increased levels of serum HIV p 24 antigen while receiving AZT should be managed. Certain factors that stimulate bone marrow, such as erythropoietin, granulocyte-stimulating factors, or granulocyte-monocyte colony-stimulating factors [109, 110], may reduce the suppression of bone marrow associated with AZT, and these approaches need further research in an academic center .


Biochemical Pharmacology of Other Antiretroviral Dideoxynucleosides Including 2',3'-Dideoxyinosine (Didanosine)

As noted, a number of dideoxynucleosides other than AZT have antiretroviral activity in vitro [13- 26], and several studies of such agents are now enrolling patients. There are substantial differences in the rate at which human cells phosphorylate these compounds and in their enzymatic pathways. These differences are profoundly important to their antiretroviral activities. 2',3'-Dideoxycytidine (ddC) is, for example much more potent than 2',3'-dideoxythymidine (ddT) in most human cells because of differences in its phosphorylation [31,61-65,67]. Since their rates of phosphorylation differ between species [68], one cannot draw conclusions about their activities in human cells on the basis of their performance in animal cells. 2',3'-Dideoxyadenosine (ddA), 2',3'-dideoxyinosine (ddI), and ddC are three compounds with potent activity against HIV in human T cells and monocytes in vitro under study [13,14,105] in clinical trials. One of the most active dideoxynucleosides is ddC, a pyrimidine analog [13, 14]. Unlike many cytidine analogs, it is resistant to deamination by the ubiquitous enzyme cytidine deaminase [62, 63]. It is, therefore, stable in plasma and bioavailable after oral administration. After entering a cell, ddC is phosphorylated by a set of enzymes that usually phosphorylate deoxycytidine [62,63,141, 142]. Thus, ddC is activated by a different pathway than AZT. Also, ddC does not affect the levels of its competing dideoxynucleoside triphosphate, deoxycytidine 5'-triphosphate [14]. However, its anabolic phosphorylation and activity may be affected by other nucleosides (e.g. thymidine) [143]. ddC is exceptionally potent and the optimal dose to avoid neuropathy is still under study. Total doses of 2 mg per day or less are being tested in adults. It is worth emphasizing that this entire class of nucleoside analogs represents a new area of clinical research. These drugs have antiretroviral activity; however, they also have considerable potential for side effects. These drugs should be administered only by physicians who are well versed in their properties. ddA and its immediate metabolite ddI are purine analogs with in vitro activity against HIV [13] and, unlike AZT, relatively little toxicity against bone marrow precursor cells [144]. Within cells, ddA can be phosphorylated to its active 5'triphosphate moiety [64]. It is also susceptible to deamination by adenosine deaminase and forms ddI [64]. In human plasma and cell extracts, this conversion occurs almost instantaneously [26, 64]. As mentioned, ddI has potent in vitro activity against HIV [13] because it can be metabolized in human cells to form ddA5'-triphosphate through a complex series of reactions [65]. Interestingly, ddI uses the enzyme 5'-nucleotidase to undergo the initial phosphorylation it needs for activation and ultimate salvage back to ddA TP .Thus, for many purposes the two drugs can be considered identical. Once ddA and ddI are converted to ddA-5'triphosphate in cells, they rernain there for a relatively long time their intra cellular half-life is more than 12 h [145]. Thus, even with their short plasma halflife, they may be clinically effective when administered relatively infrequently (e.g., every 8-24 h). Unlike AZT or ddC, ddA and ddI undergo solvolysis (cleavage) in acid reactions to form a purine base and dideoxyribose [64]. This may lower their capacity for oral absorption, and they must be used with antacids or buffers. High concentrations of the free purine base of ddA, adenine, have been reported to cause renal damage [146]. The free base of ddI, hypoxanthine, does not have similar toxicity, and it may, therefore, be preferable for oral administration. Preliminary results from a phase I trial and ddI suggest that it is an active antiretroviral drug [9]. Some patients have now received this drug for more than 2 years. The major side effects to data have been a reversible peripheral neuropathy and acute pancreatitis. In some cases, the pancreatitis may be lethai. It appears that a prior history of pancreatic disease is risk factor for this complication. Significant diarrhea and hypokalernia may occur. These side effects seem to be dose related. Patients who have advanced disease or who are debilitated have an increased risk for toxicity. At doses less than 8 mg/kg per day, serious side effects are significantly less common than at higher doses. In an average adult, total doses should not exceed 500 mg per day. It is possible that even lower doses eventually will be found active. The use of alcohol is contraindicated in patients receiving ddI due to the possibility of pancreatitis. Certain analogs of ddA (e.g., 2',3'dideoxy-2'-fluoro-ara-adenosine) are resistant to acid hydrolysis [21] and, therefore, may have better bioavailability than ddA. Also, certain 2-halogen-substituted forms of ddA are resistant to deamination [26], and they may be directIy phosphorylated and not follow the ddI pathway. Whether clinical studies with such forms will produce compounds superior to ddA or ddI is not known; ddI has potent in vitro activity against HIV in its own right [13], and the issue of acid instability can be addressed by simple measures such as the buffering of gastric secretion. Like AZT, anumber of analogs of ddT have been tested for activity against HIV in vitro. Many were inactive, but a few blocked the replication of HIV in human T cells [14, 16, 17,22, 147]. An unsaturated form of ddT called 2',3'-didehydro2',3'-dideoxythyrnidine is about as active as AZT on a molar basis [16, 17]. Unlike AZT, it does not affect the activity of thyrnidylate kinase [148], whether it induces bone marrow toxicity rernains undetermined. The dose-Iirniting toxicity is peripheral neuropathy. Finally, a 3'substituted uridine analog, 3'-azido-2',3'deoxyuridine, which appears to be activated by the same enzymes that phosphorylate AZT, has some anti-HIV activity in vitro [19, 149]. All these dideoxynucleosides have an intact oxacyclopentane (sugar) ring. However, several acyclic compounds ( adenallene, cytallene, and a phosphonyl-rnethylethyl purine derivative) also have activity against HIV in vitro as single agents. Such compounds provide new relation between structure and activity and may be of value in developing a new class of anti-HIV agents.


Clinical Research with Dideoxycytidine

Although AZT can prolong the lives of patients with AIDS, there are some limitations to its use. The hernatologic toxicity of AZT is not inextricably linked to its antiviral effect, and we can expect that other agents will be worth exploring or have different patterns of toxicity. In vitro testing and studies of animal toxicology can provide clues as to which drugs are likely to have favorable therapeutic results. Ultimately, however, the issues can be resolved only by testing in patients, and an effort is now under way to test several of these agents in patients with AIDS or related conditions. The first to be studied clinically (after AZT) was ddC, which has potent activity against HIV in vitro at concentrations of 0.01-0.5 µM, depending on the viral dose used in the assay system [13,69, 70]. It is weil absorbed when given orally, and peak levels of 0.1-0.2 µM can be attained after the oral administration of 0.03 mg/kg body weight [30]. Like AZT, ddC has a half-life of slightly more than 1 h. lt differs from AZT in that it is excreted by the kidneys [30,150]. Finally, ddC penetrates at least partially the cerebrospinal fluid [30, 150]. Both the initial study of ddC [30] and a subsequent trial [1-51] found evidence of clinical activity against HIV. Nearly all the patients who received daily doses of between 0.06 and 0.54 mg/kg had decreased levels of serum HIV p24 antigen [30,151], and most had small increases in the number ofCD4+ cells by week 2 [30]. Furthermore, some had an increase in antigen-induced T -cell proliferation in vitro [30]. The decrease in levels of p24 antigen persisted in some patients for at least several weeks after the drug was withdrawn. In others, however, the immunologic and virologic values moved toward baseline after several weeks despite the continued administration of ddC [30]. One purpose of these studies was to define the dose-limiting toxic effects of ddC. In a number of patients, particularly those receiving higher doses, maculovesicular eutaneous eruptions, aphthous oral ulcerations, fever, and malaise developed after 1-4 weeks of therapy [30, 151, 152]. These symptoms usually resolved in 1-2 weeks even with continued therapy. However, after several months of continuous therapy with daily doses of 0.06 mg/kg or more, most patients had a painful sensory motor peripheral neuropathy (involving mainly the feet) that became the doselimiting toxic effect [30, 151]. This neuropathy appeared earlier, was more severe, and lasted longer when the highest doses were tested; some patients receiving the highest doses still had persistent, moderate sensory loss and pain a year after the drug was discontinued [151, 153]. Neurotoxicity resolved much more quickly, however, in patients receiving lower doses [30, 151, 153]. One metabolic product of ddC in human cells is dideoxycytidine diphosphate choline [62], which could conceivably contribute to the neuropathy. Alternatively, the neuropathy may result from an inhibitory effect of ddC-5'triphosphate on mitochondrial DNA polymerase gamma (inhibition constant, 0.16 µM) [63, 154]. Thus, a search for ddC congeners which would not affect mitochondrial DNA synthesis is under way. Scientists at Hoffmann-LaRoche have begun studying a fluorinated version of ddC which may spare mitochondrial DNA polymerase. A continuation of the study of Merigan etal. [151] and a separate study organized by M. Gottlieb and W. Soo (personal communication) have shown that many patients can tolerate lower doses of ddC (0.03 mg/kg per day) for 6 months or more; mild, readily reversible neuropathy developed in a minority of patients. This dose of ddC was associated with a decline in HIV p24 antigen levels and an increase in the number ofCD4+ lymphocytes in most patients. Since the toxicity of ddC is strikingly different from that of AZT, combining the two agents may reduce overall toxicity. To test this approach, a group of patients with AIDS or AIDS related complex followed a regimen alternating AZT (200 mg every 4 h) and ddC (0.09 or 0.18 mg/kg per day) therapy in 7day periods [30, 155]. It was hoped that neuropathy would not occur or would occur later with the intermittent administration of ddC. Preliminary results suggest that the toxicity of both agents can be significantly reduced. Some patients have now tolerated the regimen for more than 36 months [155] (unpublished data). Overall, the patients had an average increase of more than 70 CD4+ cells/mm³ at week 22, sustained decreases in serum p24 antigen levels, and a mean weight gain of 5 kg (not caused by fluid retention) [30, 155]. It is interesting to note that on low-dose or intermittent dosing regimens, once patients pass the 6month mark without neuropathy, they may have a significant probability of avoiding serious neuropathy on continued administration of ddC. Next to AZT, ddC has been given to patients longer than any other dideoxynucleoside. lt is probable that ddC will find its best use as part of a combination regimen with AZT.


Other Anti-HIV Agents in Preclinical and Clinical Development

This article has focused on the use of dideoxynucleosides as antiviral agents, in part because they are bioavailable after oral administration and because data from several studies support their virustatic activity in vitro. This is, however, by no means the only approach being investigated for the treatment of AIDS. The genome and replicative cycle of HIV are very complex, and several stages of replication may, therefore, be potential targets for antiretroviral therapy [31, 67, 156, 157]. Already, anumber of agents that may ac t at various stages have been defined. Although an extensive review of these other approaches is beyond the scope of this article, a few points are worth stressing. Certain agents under study appear to ac t by inhibiting the initial binding of HIV to its CD4 glycoprotein receptor on target cells [36- 38, 41, 42, 52-56, 158-160]. Using molecular biologic techniques, several groups recently reported truncated soluble forms ofCD4 that lack the transmembrane and cytoplasmic domains [52-56]. At concentrations of 2- 20 µg/ml, these forms inhibited the binding of HIV to T cells, the formation of syncytia, and the infection of T cells [52-56]. A potential advantage of this approach is that soluble CD4 is likely to inhibit, to some degree, all forms of HIV that use CD4 as the cell receptor. Also, agents that ac t at the cell surface may block cell death induced by syncytia, which can occur even when the target cell is not infected by HIV [38, 161, 162]. Phase I trials of recombinant CD4 are now under way. Second-generation versions of CD4 (such as CD4-immunoglobulin hybrid proteins) retain their activity against HIV in vitro, but may gain other desirable properties, such as a longer circulating half-life [163]. A phase 1 trial of such CD4-immunoglobulin hybrids is now under way at the National Cancer Institute and at other academic centers. Also, forms of recombinant CD4 linked to Pseudo monas endotoxin or ricin selectivity kill cells expressing HIV envelope proteins in vitro [164, 165]. In patients, such agents might selectively kill cells that can replicate HIV without being killed by the virus (e.g., macrophages). Unfortunately, CD4 does not necessarily bind to the gp 120 of primary isolates (as opposed to laboratory isolates) with high affinity. This may mean that very high doses ofCD4 need to be used. Recently, low-molecular-weight dextran sulfate (7000-8000) was found to inhibit the infectivity of T cells by HIV [36- 38]. This polyanionic polysaccharide also appears to inhibit the initial binding step [38]. A phase I/II trial of orally administered dextran sulfate suggested that it had little toxicity but also little effect on the number of CD4+ cells or serum p24 antigen levels [166]. However, dextran sulfate has since been found to be very poorly absorbed when given orally, and studies of intravenous dextran sulfate are needed in order to assess this agent. Other molecules in this class are worth studying by parenteral administration. Recent advances in our understanding of the biochemistry of HIV replication have made the testing of new approaches to therapy possible. For example, antisense phosphorothioate oligodeoxynucleotides, which can bind to specific segments of the HIV genome, have sequence-specific inhibitory effects that may result from the arrest of translation after its hybridization to messenger RNA [48]. Interestingly, such compounds may also inhibit the replication of HIV in a manner that is not sequence specific [47]. Alteration of the sugar moiety of viral glycoproteins (e.g., by inhibitors of trimming glucosidases) reduces the infectivity of the resulting viruses [57,58]. In addition to dideoxynucleosides, other agents may ac t at the level of reverse transcriptase. In particular, phosphonoformate, a pyrophosphate analog with activity against herpesvirus, has activity against HIV in vitro. Several pilot trials suggest that this drug can reduce serum HIV p24 antigen levels in patients with HIV infection [167, 168]. However, no reliable oral formulation is available, and this remains one drawback of this drug. There is a growing interest in developing drugs that inhibit the protease of HIV. During the next few years, it is likely that several protease inhibitors will enter clinical trials. Several agents that act at different stages of viral replication (e.g., interferon-alfa) have synergy with AZT in vitro [34, 36, 40] and this could theoretically result in better treatment in patients. Interferon-alfa may be particularly interesting in this regard, because it also has a direct antitumor effect against cutaneous Kaposi's sarcoma [169-172]. In a similar vein, the antiherpes drug acyclovir, which has little activity against HIV alone, can potentiate the anti-HIV activity of AZT in vitro [14, 31]. In a pilot clinical trial, patients with AIDS or AIDS-related complex tolerated these drugs together for 10-30 weeks [32]. A theoretical advantage of the regimen is that suppressing the replication of herpesvirus may secondarily reduce the replication of HIV since a product of herpesvirus, ICPO (infected cell protein), can increase the initiation of HIV transcription [173, 174]. The possible suppression of human herpesvirus 6 (human B-cell lymphotropic virus), which can infect lymphoid cells [175], may also be relevant. (In a related fashion, adenovirus enzyme-immunoassay product can also amplify HIV transcription [173]. Certain dideoxynucleosides can inhibit the replication of adenovirus [176] and thus may conceivably reduce the replication of HIV.) Acyclovir has been reported to be at least additive with AZT in inhibiting the replication of Epstein-Barr virus, and it could theoretically benefit patients infected with both that virus and HIV [177 -179]. Whether AZT and acyclovir together offer a therapeutic advantage over AZT alone is not yet clear. Only properly controlled clinical trials can answer this point. Not all combinations of anti-HIV drugs have synergistic or even additive effects. For example, the nucleoside analog ribavirin inhibits the phosphorylation of AZT in vitro and blocks its activity against HIV [50]. Ribavirin, however, increases the phosphorylation of purine analogs such as ddA through complex mechanisms involving its ability to inhibit inosine monophosphate dehydrogenase [180]. Ribavirin can be given orally and may in theory potentiate the antiHIV effects of ddA or ddl. Unfortunately, one cannot predict from first principles whether this kind of potentiation would be good or bad. Once again, only carefully controlled clinical trials can resolve this issue. These in vitro observations should alert clinicians to the possibility of unexpected interactions among agents, and they are an argument against ad hoc experimentation with antiretroviral therapies outside approved clinical trials.


Conclusion

In this article a number of new therapeutic agents and strategies have been discussed. We now have at hand a number of approaches that can inhibit the replication of HIV in vitro. These approaches, as well as a number of additional developments which are in the offing, can be expected to induce clinical improvement and prolong life even in patients with advanced AIDS. The progress against the mortality caused by AIDS is noteworthy in its own right, but there have been a number of advances that have improved the quality of life. For example, the incidence of dementias ascribable to AIDS has been noted to have decreased after the introduction of AZT [181]. HIV infection is probably a lifelong process. It now appears highly likely that a complete latency phase does not exist. Rather, many, if not all patients, have circulating infectious HIV particles present in their plasma even when the disease is clinically quiescent. Thus, it is perhaps unrealistic to expect a single drug to provide therapy for all patients. The experiences with cancer therapy, as well as the experiences with other serious infections, suggest that a combination of drugs will produce superior clinical out come and less toxicity than any single therapy used alone [182]. Combination therapy may also delay or prevent the emergence of viral resistance. Just as in the treatment of certain leukemias or advanced bacterial diseases, optimal therapy against HIV may require at least three different phases: induction, consolidation, and maintenance. It is worth noting that the drugs and biological agents, as well as the relevant doses of such drugs and agents, may vary in each phase. At present, the only formally approved antiretroviral agents are AZT and ddl. AZT has been proven to reduce morbidity and mortality above and beyond any effect of aerosolized pentarnidine in severe cases of AIDS [183]. Nevertheless, several virustatic drugs in the same general family are being tested in patients, and it seems highly probable that AZT is not the only agent which eventually will prove effective against HIV. As with a number of other therapies used in life-threatening disorders, AZT may have a relatively low therapeutic index in some patients. Therefore, it is very important that clinicians pay close attention to its.clinical pharmacology and to the specific patient responses that occur following initial therapy. As new experimental agents are tested and become more widely available, it is important that careful adherence to the principles of clinical trials be a major priority if we are to succeed in the mission of developing better therapeutic options. As simpler assays to measure plasma drug levels become available [184, 185], their results conceivably may provide useful data in the optimal management of HIV infections. A number of studies are now under way to test whether various agents should be administered to patients with early HIV infections and to explore other therapeutic regimens. In the coming decade, it seems highly probable that major advances will occur against the death and suffering caused by HIV, but this progress can be ensured only if the principles of scientific drug development and controlled trials are maintained.


References

I. Gottlicb MS, Schroff R, Schankcr HM, ct al. (1981) Pneumocy.stis carinii pneu monia and mucosal candidiasis in previously healthy homosexual men: evidence of a ncw acquired cellular immunodeficiency. N Eng] M Med 305:1425-1431
2. Masur H, Michelis MA, Greene JB, et al. (1981) An outbreak of community acquired Pneumocysti carinii pneumonia: initial manifestation of cellular immune dysfunction. N Eng] Med 305:1431 1438
3. Siegal FP, Lopez C, Hammer GS, et al. (1981) Severe acquired immunodeficiency in male hornosexuals, manifested by chronic perjanal ulcerative herpes simplex lesions. N Engl J Med 305 : 1439-1444
4. Barre-Sinoussi F, Chermann JC, Rey F, et al. (1983) Isolation of a T-lymphotropic retrovirus frorn a patjent at rjsk for acquired immune deficjency syndrome (AIDS). Science 220:868-871
5. Gallo RC, Salahuddin SZ, Popovic M, et al. (1984) Frequent detection and isolation of cytopathic retroviruses (HTL V III) from patients with AIDS and at risk for AIDS. Science 224. 500- 503
6. Popovic M, Sarngadharan MG, Read E, Gallo RC (1984) Detection isolation, and continuous production of cytopathic retroviruscs (HTL V -III) frorn patients with AIDS and pre-AIDS. Science 224:497-500
7. Mitsuya H, Popovic M, Yarchoan R, Matsushita S, Gallo RC, Broder S (1984) Suramin protection of T -cells in vitro against infectjvity and cytopathjc effect of HTLV-III. Science 226:172-174
8. Mitsuya H, Weinhold KJ, Furman PA, et al. (1985) 3'-Azjdo-3'-deoxythyrnidine (BW A509U). an antiviral agent that inhibits the infectivity and cytopathic effect of human T -lymphotropic virus type III/lymphadenopathy-associated virus in vitro. Proc Natl Acad Sci USA 82:7096-7100
9. Ostertag W, Roesler G, Krieg CJ, et al. (1974) Induction of endogenous virus and of thyrnidine kinase by bromodeoxy-uridjne in cell cultures transform ed by Friend virus. Proc Natl Acad Sci USA 71:4980-4985
10. De Clercq E (1979) Suramin: a potent inhibitor of the reverse transcriptase of RNA tumor viruses. Cancer Lett 8 :9-22
11. Furmanski P, Bourguignon GJ, Bolles CS, Corombos JD, Das MR (1980) Inhibition by 2',3'-dideoxythyrnidine of retroviral infection of mouse and human cells. Cancer Lett 8.307-315
12. Waqar MA, Evans MJ, Manly KF, Hughes RG, Huberman JA (1984) Effects of 2',3'-dideoxynucleosides on mammalian cells and viruses. J Cell Physiol 121 :402-408
13. Mitsuya H, Broders S (1986) Inhibition of the in vitro infectivity and cytopathic effect on human T -lymphotropic virus type III/lymphadenopathy-associated virus (HTL V -III/LA V) by 2',3'-dideoxynucleosides. Proc Natl Acad Sci USA 83.1911-1915
14. Mitsuya H, Matsukura M, Broder S (1987) Rapid in vitro screening systems for assessing activity of agents against HTLV-III/LA V. In: Broder S (ed) AIDS: modern concepts and therapeutic challenges. Dekker, New York, pp 303-333
15. Balzarini J, Pauwels R, Herdewijn P, et al. (1986) Potent and selective antiHTL V -III/LA V activity of 2',3'-dideoxycytidine, the 2',3' unsaturated derivative of 2',3'-dedeoxycytidine. Biochern Biophys Res Commun 140:735-742
16. Hamamoto Y, Nakashima H, Matsui T, Matsuda A, Ueda T, Yamamoto N (1987) Inhibitory effect of 2',3'-didehydro-2',3'-dideoxynucleosides on infectivity, cytopathic effects, and replication of human immunodeficiency virus. Antimicrob Agents Chernother 31:907-910
17. Balzarini J, Kang G-J, Dalal M, et al. (1987) The anti-HTLV-III (anti-HIV) and cytotoxjc activity of 2',3'-didehydro2',3'-dideoxyribonucleosides: a comparison with their parenteral 2',3'-dideoxyribonucleosides. Mol Pharmacol 32 : 162-167
18. Lin T -S, Schinazi RF, PrusoffWH (1987) Potent and selective in vitro activity of 3'deoxythyrnidjn-2'-ene (3-dcoxy-2',3'-didehydrothyrnidine) against human immunodeficjency virus. Biochern Phar macoI36:2713-2718
19. Schinazi RF, Chu C-K, Ahn M-K, et al. (1987) Selective in vitro inhibition of human immunodeficiency virus (HIV) replication by 3'-azido-2',3'-dideoxyuridine (CS-87) [Abstract]. J Clin Biochern [Suppl] 11D:74
20. Kirn C-H, Marquez VE, Broder S, Mitsuya H, Drjscoll JS (1987) Potentjal anti AIDS drugs: 2',3'-dideoxycytjdine analogues. J Med Chern 30.862-866
21. Marqucz VE, Tscng CK, Kelly JA, et al. (1987) 2',3'-Dideoxy-2'-fluoro-ara-A: an acid-stable purine nucleoside active against human immunodeficicncy vjrus (HIV). Biochern Pharmacol 36:2719-2722
22. Herdewijn P, Balzarini J, dc Clercq E, et al. (1987) 3'-Substituted 2',3'-dideoxynucleoside analogues as potential antiHIV (HTL V -III/LA V) agents. J Med Chern 30:1270-1278
23. Balzarini J, Robins MJ, Zou RM, Herdewijn P, de Clercq E (1987) The 2',3'-dideoxyriboside of 2,6-diaminopurine and its 2',2'-didehydro derivative inhibit the deamination of 2', 3'-dideoxyadenosine, an inhibitor of human immunodeficiency virus (HIV) replication. Biochern Biophys Res Commun 145.277-283
24. Baba M, Pauwels R, Balzarinj J, Herdewijn P, de Clercq E (1987) Selective inhibition of human immunodeficiency virus (HIV) by 3'-azido-2',3'-dideoxyguanosine in vitro. Biochern Biophys Res Commun 145:1080-1086
25. Herdewijn P, Pauwels R, Baba M, Balzarini J, de Clercq E (1987) Synthesis and anti-HIV activity of various 2'- and 3'substituted 2',3'-didcoxyadenosines: a structure-activity analysis. J Med Chern 30:2131-2137
26. Haertle T, Carrera CJ, Wasson DB, Sowers LC, Richman DD, Carson DA (19-8) Metabolism and anti-human immunodeficiency vjrus-1 activation of 2halo-2',3'-dideoxyadenosine dcrivatives. J Biol Chem 263:5870-5875
27. Yarchoan R, KleckerRW,Weinhold KJ, et al. (1986) Administration of 3'-azido 3'-deoxythyrnidine, an inhibitor of HTL V -III/LA V replication to patients with AIDS or AIDS-related complex. Lancet 1: 575- 580
28. Fischi MA, Rjchman DD, Grieco MH, et al. (1987) The efficacy of azidothymidine (AZT) in the treatment of patients with AIDS and AIDS-related complex: a double-blind, placebo-controlled trial. N Engl J Med 317.185-191
29. Volberdjng PA, Lagakos SW, Koch MA, et al. (1990) Zidovudine in asymptomatic human immunodeficiency virus infection: a controlled trial in persons with fewer than 500 CD4-positive cells per cubic mjllimeter. N Engl J Med 322.941-949
30. Yarchoan R, Perno CF, Thomas RV, et al. (1988) Phase I studies of 2',3'djdeoxycytidine jn severe human immunodeficiency virus infection as a single agcnt and alternatjng with zidovudinc (AZT). Lancet 1:76-81
31. Mitsuya H, Broders S (1987) Strategies for antiviral therapy in AIDS. Nature 325: 773-778
32. Surbone A, Yarchoan R, McAtee N, et al. (1988) Treatment of the acquired immunodeficicncy syndrome (AIDS) and AIDS-related complex with a regimen of 3'-azido-2',3'-dideoxythyrnidine (azidothyrnidine or zidovudine) and acyclovjr: a pilot study. Ann Intern Med 108.534-540
33. Montefiori DC, Mitchell WM (1987) Antivjral activity of mjsmatched doublestranded RNA against human immunodeficiency virus in vitro. Proc Natl Acad Sci USA 84: 2985- 2989
34. Mitchell WM, Montefiori DC, Robinson WE Jr, Strayer DR, Carter WA (1987) Mismatched double-sizcd RNA (ampligen) reduces concentration of zjdovudine (azidothyrnidine) required for in vitro inhibition of human jmmunodeficiency virus. Lancet 1 :890-892
35. Carter W A, Strayer DR, Brodsky I, et al. (1987) Clinical, immunological, and virological effects of ampligen, a mismatched double-stranded RNA, in patients with AIDS or AIDS-related complex. Lancet 1:1286-1292
36. Ueno R, Kuno S (1987) Dextran sulphate, a potent anti-HIV agent in vitro having synergism with zjdovudine. Lancet 1.1379
37. Ito M, Baba M, Sato A, Pauwels R, de Clercq E, Shigeta S (1987) Inhibitory effect of dextran sulfate and heparjn on the replicatjon of human immunodeficiency virus (HIV) in vitro. Antiviral Res 7.361-367
38. Mitsuya H, Looney DJ, Kuno S, Ueno R, Wong-Staal F, Broder S (1988) Dextran sulfate suppression of viruses in the HIV family: jnhibition of virion binding to CD4 + cells. Science 240 : 646-649
39. Ho DD, Hartshorn KL, Rota TR, et al. (1985) Recombinant human interferon alfa-A suppresses HTL V -III replication in vitro. Lancet 1. 602-604
40. Hartshorn KL, Vogt MW, Chou T-C, et al. (1987) Synergistic inhibitjon ofhuman jmmunodeficiency vjrus in vitro by azjdothyrnidine and recombinant A interferon. Antjmjcrob Agents Chernother 31.168-172
41. Matsushita S, Robert-Guroff M, Rusche J, et al. (1988) Characterjzation of a human immunodeficiency virus neutralizing monoclonal antibody and mapping of the neutralizing epitope. J ViroI62.2107-2141
42. Pert CB, HilI JM, Ruff MR, et al. (1986) Octapeptides deduced from the neuropeptjde receptor-like pattern of antigen T 4 in brain potcntly inhibit human immunodeficiency virus receptor binding and T -cell infectivity. Proc Natl Acad Scj USA 83.9254-9258
43. Wetterberg L, Alexius B, Sääf J, Sönnerborg A, Britton S, Pert C (1987) Peptide T in treatment of AIDS. Lancet 1 :159
44. Barnes DH (1987) Debate over potential AIDS drug. Science 237.128-130
45. Sandstrom EG, Kaplan JC, Byington RE, Hirsch MS (1985) Inhibitjon of human T cell lymphotropic virus type II in vitro by phosphonoformate. Lancet 1:1480-1482
46. Sarin PS, Taguchi Y, Sun D, Thornton A, Gallo RC, Öberg B (1985) Inhibition of HTL V-III/LA V replication by foscarnet. Biochern Pharmacol 34:4075-4079
47. Matsukura M, Shinozuka K, Zon G, et al. (1987) Phosphorothioate analogs of oligodeoxynucleotides: inhibitors of replication and cytopathic effects of human immunodeficiency virus. Proc Natl Acad Sci USA 84:7706- 7710
48. Matsukura M, Shinozuka K, Zon G, ct al. ( 1988) Phosphorothioate arlalogs of oligodeoxynucleotide inhibil viral replication of human jmmunodeficiency virus (HIV): jnhibitjon of de novo infection in uninfected cells and regulation of viral expression in chronically infected cells (Abstr). Clin Res 36: 463 A
49. McCormick JB, Getchell JP, Mitchell SW, Hicks DR (1984) Ribavirin suppresses replication of lymphadenopathyassociated virus in cultures of human adult T lymphocytes. Lancet 2.1367 1369 XCVIl
50. Vogt MW, Hartshorn KL, Furman PA, et al. (1987) Ribavirin antagonizes the effect of azidothymidine on HIV replication. Science 235:1367-1379
51. Anand R, Moore J, Feorino P, Curran J, Srinivasan A (1986) Rifabutine inhibits HTLV-III. Lancet 1 :97-98
52. Smith DH, Byrn RA, Marsters SA, Gregory T, Groopman JE, Capon DJ (1987) Blocking of Hrv I infectivity by a soluble, secreted form of CD4 antigen. Science 238.1704-1709
53. Fisher RA, Bertonis JM, Meier W, et al. (1988) HIV infection is blocked in vitro by recombinant soluble CD4. Nature 331:76-78
54. Hussey RE, Richardson NE, Kowalski M, et al. (1988) A soluble CD4 protein selectivcly inhibits HIV replication and syncytium formation. Nature 331 :78-81
55. Deen KC, McDougal JS, Inacker R, et al. (1988) A soluble form of CD4 (T4) protein inhibits AIDS virus infection. Nature 331 :82-84
56. Traunecker A, Lukc W, Karjalainen K (1988) Soluble CD4 molecules neutralize human immunodeficiency virus type 1. Nature 331.84-86
57. Walker BD, Kowalski M, Goh WC, et al. (1987) Inhibition of human immunodeficiency virus syncytium formation and virus replication by castanospermine. Proc Natl Acad Sci USA 84:8120-8124
58. Gruters RA, Neefjes JJ, Tersmette M, et al. (1987) interference with HIV -induced syncytium formation and viral infectivity by inhibitors of trimming glucosidase. Nature 330:74-77
59. Hayashi S, Phadtare S, Zemlicka J, Matsukura M, Mitsuya H, Broder S (1988) Adenallene and cytallene: acyclic-nuclcoside analogues that inhibit replication and cytopathic effect of human immunodeficiency virus (HIV) in vitro. Proc Natl Acad Sci USA 85:6127-6131
60. Chin J (1990) Current and future dimensions of the HIV /AIDS pandemic in women and children. Lancet 336:221-224
61. Furman PA, Fyfe JA, St Clair MH, et al. (1986) Phosphorylation of 3'-azido-3'deoxythymidine and selective interaction of the 5'-triphosphatc with human immunodeficiency virus reverse transcriptase. Proc Natl Acad Sci USA 83:8333-8337
62. Cooney DA, Dalal M, Mitsuya H, et al. (1986) rnitial studies on the cellular pharmacology of 2',3'-dideoxycytidine, an inhibitor ofHTL V -III infectivity. Biochem Pharmacol 35. 2065- 2068
63. Starncs MC, Chcng YC (1987) Cellular metabolism of 2',3'-dideoxycytidine, a compound active against human immunodeficiency virus in vitro. J Biol Chem 262:988-991
64. Cooney DA, Ahluwalia G, Mitsuya H, et al. (1987) Initial studies on the cellular pharmacology of 2',3'-dideoxyadenosine, an inhibitor of HTL V -III infectivity. Biochem Pharmacol 36: 1765 -1768
65. Ah[uwalia G, Cooney DA, Mitsuya H, ct al. (1987) Initial studies on the cellular pharmacology of 2',3'-dideoxyinosine, an inhibitor of HIV infectivity. Biochcm Pharmaco[ 36:3797-3800
66. Mitsuya H, Jarrett RF, Matsukura M, et al. (1987) Long-term inhibitor of human T lymphotropic virus type 1I1/lymphadenopathy-associated virus (human immunodeficiency virus) DNA synthesis and RNA expression in T cells protected by 2',3'-dideoxynucleosides in vitro. Proc Natl Acad Sci USA 84:20332037
67. Yarchoan R, Broder S (1987) Development of antiretroviral therapy for the acquired immunodeficiency syndrome and rclatcd disordcrs: a progress rcport. N Engl J Med 316.557-564
68. Dahlberg JE, Mitsuya H, Blam SB, Broder S, Aaronson SA (1987) Broad spectrum antiretroviral activity of 2',3'dideoxynucleosides. Proc Natl Acad Sci USA 84.2469-2473
69. Barber AM, Hizi A, Maizel JV, Hughes SH (1990) Hrv-1 reverse transcriptase: structure predictions for the polymerase domain. AIDS Res Hum Retroviruses 6:1061-1072
70. Mitsuya H, Dahlberg JE, Spigelman Z, ct al. (1988) 2',3'-Didcoxynuclcosidcs: broad spectrum antiretroviral activity and mechanism of action. In: Bolognesi D (ed) Human retroviruses, cancer, and ArDS. approachcs to prcvcntion and therapy. Liss, New York, pp 407-421
71. Hoa Z, Dalal M, Cooney DA, et al. (1987) A comparison of 2',3'-dideoxynucleoside 5'-triphosphates as inhibitors of retroviral reverse transcriptases (Abstr). Proc Am Assoc Cancer Res 28:323
72. Vrang L, Bazin H, Remaud G, Chattopadhyaya J, Oberg B (1987) Inhibition of the reverse transeriptase from HIV by 3'azido-3'-deoxythymidine triphosphate and its threo analogue. Antiviral Res 7.139-149
73. Cheng YC, Dutschman GE, Bastow KF, Sarngadharan MG, Ting RY (1987) Human immunode/iciency virus reverse transcriptase. general properties and its interactions with nucleoside triphosphate analogs. J Biol Chem 262:2187-2189
74. Richman DD (1987) Dideoxynucleosides are less inhibitory in vitro against human immunode/iciency virus type 2 (HIV -2) than against HIV-1. Antimicrob Agents Chemother 31.1879-1881
75. Mitsuya H, Broder S (1988) Inhibition of infectivity and replication of HIV -2 and SIV in helper T -cells by 2',3'-dideoxynucleosides in vitro. AIDS Res Hum Retroviruses 4: 107 -113
76. Matsushita S, Mitsuya H, Reitz MS, Broder S (1987) Pharmacological inhibition of in vitro infectivity of human T lymphotropic virus type I. J Clin Invest 80:394-400
77. Ruprecht RM, O'Brien LG, Rossoni LD, Nusinoff-Lehrman S (1986) Suppression of mouse viraemia and retroviral disease by 3'-azido-3'-deoxythymidine. Nature 323:467-469
78. Tavares L, Roneker C, Johnston K, Lehrman SN, de Noronha F (1987) 3'Azido 3'-deoxythymidine in feline leukemia virus-infected cats: a model for therapy and prophylaxis of AIDS. Cancer Res 47:3190-3194
79. Kassianides C, Hoofnagle J, Miller RH, et al. (1989) Inhibition of duck hepatitis B virus replication by 2',3'-dideoxycytjdine: a potent inhibitor of reverse transcriptase. Gastroenterology 97.1275-1280
80. Summers J, Mason WS (1982) Replication of the genome of a hepatitis B-Iike virus by reverse transcription of an RNA intermediate. Ce]] 29:403-415
81. Zimmerman F, Biessert L, von Briesen H, et al. (1988) Development of HIVvariants with higher resistance against HIV under treatment with AZT (Abstr). 4th International Conference on AIDS, June 12-16, Stockholm, vol2. Swedish Ministry of Health and Social Affairs, Stockholm, p 180
82. Larder BA, Darby G, Richman DD (1989) H1V with reduced sensitivjty to zidovudine (AZT) isolated during prolonged therapy. Science 243:1731-1734
83. Larder BA, Purifoy DJM, Powell KL, Darby G (1987) Site-speci/ic mutagenesis of AIDS virus reverse transcriptase. Nature 327.716- 717
84. Zimmerman TP, Mahony WB, Prus KL (1987) 3'-Azido-3'-deoxythymjdine: an unusual nucleoside analogue that permeates the membrane of human erythrocytes and lymphocytes by nonfacilitated diffusion. J Biol Chem 262:5748-5754
85. Frick LW, Nelson DJ, St Clair MH, Furman PA, Krenitsky TA (1988) Effects of 3'-azido-3'-deoxythymjdine on the deoxynucleotide triphosphate pools of cultured human cells. Biochem Biophys Rcs Commun 154.124-129
86. Hao Z, Cooney DA, Hartman NR, et al. (1988) Factors determining the activity of 2',3'-dideoxynucleosides in suppressing human immunodeticiency virus in vitro. Mol Pharmacol 34:431-435 87. Harrington JA, Miller WH, Spector T (1987) Effector studies of 3'-azidothymidine nucleotides with human ribonucleotide reductase. Biochem Pharmaco136.3757-3761
88. Yarchoan R, Broder S (1987) Strategies for the pharmacological intervention against HTL V -III/LA V. In: Broder S (ed) AIDS. modern concepts and therapeutic challenges. Dekker, New York, pp 335-360
89. Klecker RW Jr, Collins JM, Yarchoan R, et al. (1987) Plasma and cerebrospinal f1uid pharmacokinetics of 3'-azido-3'deoxythymidine. a novel pyrimidine analog with potential application for the treatment of patients with AIDS and related diseases. Clin Pharmacol Ther 41:407-412
90. Blum MR, Liao SH, Good SS, de Miranda P (1988) Pharmacokinetics and bioavailability of zidovudine in humans. Am J Med [SuppI2A] 85.189-194
91. De Miranda P, Good SS, Blum MR, Thomas RV, Yarchoan R, Broder S (1986) The effect of probenecid on the pharmacokinetic disposition of azidothymidine (AZT) (Abstr). 2nd International Conference on AIDS, June 23-25, Paris
92. Klecker RW Jr, Colljns JM, Yarchoan RC, et al. (1988) Pharmacokinetics of 2',3'-dideoxycytidine in patients with AIDS and related disorders. J Clin PharmaeoI28:837-842
93. Blum MR, Liao SH, de Miranda P (1982) Overview of acyclovir pharmacokinetic disposition in adults and children. Am J Med [Suppll A] 73:186-192
94. Furman PA, de Miranda P, St Clair MH, Elion GB (1981) Metabolism ofacyclovir in virus-infected and uninfected cells. Antimicrob Agents Chemother 20.518-524
95. Laskin OL, Longstreth JA, Hart CC, ct al. (1987) Ribavirin disposition in highrisk patients for acquired immunodeficiency syndrome. Clin Pharmacol Ther 41:546-555
96. Crumpacker C, Bubley G, Lucey D, Hussey S, Connor J (1986) Ribavirin enters cerebrospinal fluid. Lancet 2. 45-46
97. Yarchoan R, Mitsuya H, Thomas RV, et al. (1989) In vivo activity against HIV and favorable toxicity profile of 2',3'dideoxyinosine. Science 245.412-415
98. Hartman NR, Yarchoan R, Kelly JA, Cooncy DA, Broder S, Johns DG (1989) Cellular and clinical pharmacokinetics of 2',3'-dideoxyadenosine and 2',3'-dideoxyinosinc (Abstr). 5th International Conference on AIDS, June 4-9, Montreal
99. Yarchoan R, Thomas RV, Grafman J, et al. (1988) Long-term administration of 3'-azido-2',3'-dideoxythymidine to patients with AIDS-related neurological disease. Ann Ncurol [Suppl] 23:S82S87
100. Ellison S, Terasaki T, Pardridge WM (1988) AZT and dideoxynuclcosides do not cross the bJood-brain barrier (Abstr). Clin Res 36.117A 101. Gartner S, Markovits P, Markovitz DM, Kaplan MH, Gallo RC, Popovic M (1986) The role of mononuclcar phagocytes in HTLV-III/LAV infection. Science 233:215-219
102. Koenig S, Gendeiman HE, Orenstein JM, et al. (1986) Detection of AIDs virus in macrophages in brain tissue from AIDS paticnts with encephalopathy. Science 233.1089-1093
103. Stadecker MJ, Unanue ER (1979) The regulation ofthymidine secrction by macrophages. J ImmunoI 123:568-571
104. Richman DD, Kornbluth RS, Carson DA (1987) Failure of dideoxynucleosides to inhibit human immunodeficiency virus replication in cultured human macrophages J Exp Med 166.1144- 1149
105. Perno C-F, Yarchoan R, Cooney DA, et al. (1988) Inhibition of human immunodeficiency virus (HIV -1 /HTL V IIIBa -J replication in fresh and cultured human peripheraJ blood monocytes/macrophages by azidothymidine and related 2',3'-dideoxynucleosidcs. J Exp Med 168: 1111-1125
106. Skjnner MA, Matthews TJ, Greenwall TK, Bolognesi DP, Hebdon M (1988) AZT inhibits HIV -1 replication in monocytes. J AIDS 1:162-163 107. Schmitt FA, Bigley JW, McKinnis R, et al. (1988) NeuropsychoJogical outcome of zidovudine (AZT) treatment of patients wjth AIDS and AIDS-related complex. N EngJ J Med 319:1573-1578
108. Koyanagi Y, O'Brien WA, Zhao JQ, Golde DW, Gasson JC, Chen ISY (1988) Cytokines alter production of HIV-1 from primary mononuclear phagocytes. Science 241.1673-1675
109. Perno C-F, Yarchoan R, Cooney DA, et al. (1989) Replication of human immunodeficiency virus in monocytes: granulocyte/macrophage colony-stimulating factor (GM-CSF) potentiates viral production yet enhances the antiviral effect mediated by 3'-azido-2',3'djdeoxythymidine AZT and other dideoxynucleoside congeners of thymjdine. J Exp Med 169:933-951
110. Brunetti A, Berg C, di Chiro G, et al. (1989) Reversal of brain metabolic abnormalities following treatment of AIDS dementia complex with 3'-azido-2',3'dideoxythymidjne (AZT, zidovudine). A PET-FDG study. J Nucl Med 30:581 -590
111. Richman DD, Fischi MA, Grieco MH, et al. (1987) The toxjcity of azidothymidine (AZT) jn the treatment of patients with AIDS and AIDS-relatcd complex. a double-bJind, placebo-controlled trial. N Engl J Med 317:192-197
112. Andrews JC, McManus M, Rogers G (1988) AZT Collaborative Working Group. Clinical benefits of concurrent administration of zidovudine and therapy for the suppression of Pneumocystis ('arinii. 4th Internatjonal Conference on AIDS, June 12~ 16, Stockholm, voll. Swedjsh Ministry of Health and Social Affairs, Stockholm, p 412
113. Chaisson RE, Allain J-P, Leuther M, Volberding PA (1986) Significant changes in HIV antigen level in the serum of patients treated with azidothymidine. N Eng] J Med 315:1610-1611
114. Makuch RW, Parks WP (1988) Statistical methods for the analysis of HIV-I core polypeptide antigen data in clinical studies. AIDS Res Hurn Retroviruses 4:305-316
115. Kwok S, Mack DH, Mullis KB, et al. (1987) Identification of human immunodeficiency virus sequences by using in vitro enzymatic amplication and oligomer cleavage detection. J Virol 1 : 1690-1694
116. AIDS in New York State: through 1988. State of New York Department of Health, Albany
117. Yarchoan R, Broder S (1987) Preliminary results on the use of dideoxynucleosides in the therapy of AIDS. In: Chanock RM, Lerner RA, Brown F, Ginsberg HS (eds) Vaccine 87. Cold Spring Harbor Press, Cold Spring Harbor, pp 214-224
118. Reiss P, Langc JMA, Boucher CA, Danner SA, Goudsmit J (1988) Resumption of HIV antigen production during continuous zidovudine trcatment. Lancet 1.421
119. Gill PS, Rarick M, Brynes RK, Causey D, Loureiro C, Levine AM (1987) Azidothyrnidine associated with bonc marrow failure in the acquired immunodeficiency syndrome (AIDS). Ann Intern Med 107:502-505
120. Walker RE, Parker RI, Kovacs JA, et al. (1988) Anernia and erythropoiesis in patients with the acquired immunodeficiency syndrome (AIDS) and Kaposi sarcoma treated with zirdovudine. Ann Intern Med 108.372-376
121. Dournon E, Matheron S, Rozenbaurn W, et al. (1988) Effects ofzidovudine in 365 consecutive patients with AIDS or AIDS-rclated complex. Lancet 2:1297-1302
122. Gottlieb MS, Wolfe PR, Chafey S (1987) Response of AIDS-related thrombocytopenia to intravenous and oral azidothyrnidine (3'-azido-3'-deoxythyrnidine). AIDS Res Hurn Retroviruses 3 .109-114
123. Hyrncs KB, Greene JB, Karpatkin S (1988) The effect of azidothyrnidine on HIV -related thrombocytopenia. N Eng] J Med 318:516-517 124. Bessen LJ, Greene JB, Louie E, Seitzman P, Weinberg H (1988) Severe polymyositis-like syndrome associated with zidovudine therapy of AIDs and ARC. N Eng] J Med 318:708
125. Furth PA, Kazakis AM (1987) Nail pigmentation changes associated with azidothyrnidine (zidovudine). Ann Intern Med 107:350
126. Melamed AJ, Muller RJ, Gold JWM, et al. (1987) Possible zidovudine-induced hepatotoxicity. JAMA 258.2063
127. Bach MC (1987) Possible drug interaction during therapy with azidothyrnidine and acyclovir for AIDS. N Engl J Med 316.547
128. Hagler DN, Frame PT (1986) Azidothymidine neurotoxicity. Lancet 2: 1392-1393
129. Davtyan DG, Vinters HV (1987) Wernicke's encephalopathy in AIDS patient treated with zidovudine. Lancet 1.919-920
130. Creagh-Kirk T, Doi P, Andrews E, et al. (1988) Survival experience among patients with AIDS receiving zidovudine: follow-up of patients in a compassionate plea program. JAMA 260:3009-3015
131. De Wolf F, Lange JMA, Goudsmit J, et al. (1988) Effect ofzidovudine on serum human immunodeficiency virus antigen levels in symptom-free subjects. Lancet 1:372-376
132. AIlain J-P, Laurian Y, Paul DA, et al. (1987) Long-terrn evaluation of HIV antigen and antibodies to p24 and gp41 in patients with hernophilia. potential clinical importance. N Engl J Med 317.1114-1121
133. Moss AR, Bacchetti P, Osmond D, et al. (1988) Seropositivity for HIV and the development of AIDS or AIDS-related condition: three-year follow up of the San Francisco General Hospital cohort. Br Med J 296.745-750
134. Retrovir Capsules Package Insert (Zidovudine). Burroughs-Wellcome, Research Triangle Park, N.C. 135. Waldmann TA, Misiti J, Nelson DL, Kraerner KH (1983) Ataxia telangiectasis. a multisystem disease with immunodeficiency impaired organ maturation x-ray hypersensitivity, and a high incidence of neoplasia. Ann Intern Med 99.367-379
136. Hoff R, Berardi VP, Weiblen BJ, Mahoney- Trout L, Mitchell ML, Grady GF (1988) Seroprevalence of human immunodeficiency virus among childbearing women: estimation by testing samples of blood from newborns- N Engl J Med 318:525-530
137. Parks WP, Scott GB (1985) Pediatric AIDS- a disease spectrum causally associated with HTL V -ITI infection- Cancer Res 45-4659s-4661s 138. Pizzo PA, Eddyl, Falloon J, etal- (1988) Errect of continuous intravenous jnfusion of zidovudine (AZT) in children with symptomatic HTV infection- N Engl J Med 319:889-896
139. Sharpe AH, Jaenisch R, Ruprecht RM (1987) Retroviruses and mouse embryos: a rapid model for neurovirulence and transplacental antiviral therapy- Science 236:1671-1674
140. Groopman JE, Mitsuyasu RT, DeLeo MJ, Oette DH, Golde DW (1987) Errect of recombinant human granulocytemacrophage colony-stimulating factor on myelopoiesis in the acquired immunodeficiency syndrome- N Eng] J Med 317-593-598
141. Kierdaszuk B, Bohman C, Eriksson S (1988) Phosphorylation of the antiviral nucleoside 2',3'-dideoxycytidine by pure human deoxycytidine kinase (Abstr)- 4th International Conterence on AIDS, June 12-16, Stockholm, voll- Swedish Ministry of Health and Social Arrairs, Stockholm, p 223
142. Balzarini J, Pauwels R, Baba M, et al(1988) The in vitro and in vivo anliretrovirus activity, and intracellular metabolism of 3'-azido-2',3'-dideoxythyrnidine and 2',3'-dideoxycytidine are highly dependent on the ce!l speciesBiochern Pharmacol 37- 897 -903
143. Balzarini J, Cooney DA, Dalal M, el al(1987) 2',3'-Dideoxycytidine- regulation of its metabolism and anti-retroviral potency by natural pyrimidine nucleosides and by inhibitors of pyrimidine nucleotide synthesis- Mol Pharmacol 32-798-806
144. Ganser A, Greher J, Volkers B, Staszewski A, Hoelzer D (1988) Azidothymidine in the trealment of ATDS- N Engl J Med 318:250-251
145. Ahluwalia G, Johnson MA, Fridland A, Cooney DA, Broder S, Johns DG (1988) Ce!lular pharmacology of the anti-HTV agent 2',3'-dideoxyadenosine (Abstr)Proc- Am Assoc Cancer Res 29: 349
146- Lindblad G, Jonsson G, Falk 1(1973) Adenine toxicity: a three week inlravenous study in dogs- Acta Pharmacol Toxicol (Copenh) 32-246-256
147. Greengrass CW, Hoople DW, Street SD, et al (1989) 1-(3-Cyano-2,3-deoxy-beta-D-erythro-pentofuranosyl)thymine ( cyanothyrnidine)- synthesis and antiviral evaluation against human immunodeficiency virus- J Med Chern 32-618622 148- August EM, Lin T -S, Marongiu ME, Gao YS, Qian H-Y, PrusorrWH (1988) Cellular pharmacology of 2',3'-dideoxy-2',3'-didehydrothyrnidine and 2',3'dideoxy-2,3'-didehydrocytidine in H 9 cells- 4th International Conference on AIDS, June 12-16, Stockholm, vol. 1 Swedish Ministry of Health and Social Arrairs, Stockholm, p 220 149. Balzarini J, Baba M, Pauwels R, Herdewijn P, de Clercq E (1988) Anti-retrovirus activity of 3'-fluoro- and 3'-azidosubstituted pyrimidine 2',3'-dideoxynucleoside analogues- Biochern Pharmacol 37:2847-2856
150. Ke!ly JA, Littersl CL, Roth JS, et al(1987) The djsposition and metabolism of 2',3'-dideoxycytidine, an in vjtro inhibitor of human T -lymphotropic virus type TII intectivity, in mice and monkeysDrug Metab Dispos 15- 595-601
151. Merigan TC, Skowron G, Bozzette S, et al- (1989) Circulating p24 antigen levels and responses to dideoxycytidine in human immunodeficiency virus (HIV) jntcctions: a phase I and II study- Ann Intern Med 110-189-194
152. McNeely MC, Yarchoan R, Broder S, Lawley Tl (1989) Dermatologic complications associated with administration of 2',3'-dideoxycytidine in patients with human immunodeficiency virus infection- J Am Acad Dermatol 21-1213-1217
153- Dubinsky RM, Dalakas M, Yarchoan R, Broders S (1988) Follow-up of ncuropathy of 2',3'-dideoxycytidine- Lancet 1-832
154. Chen CH, Cheng YC (1989) Delayed cytotoxicity and selectivc loss of mitochondrial DNA in cells treated with antihuman immunodeficiency virus (HIV) compound 2',3'-dideoxycytjdine (ddc) (Abstr)- FASEB J 3-A1282
155- Yarchoan R, Thomas RV, Pluda J, et al(1988) Long term treatment of severe human jmmunodeficiency virus (HIV) infection with an alternating regimen of two dideoxynucleosides (Abstr)- Clin Res 36-450A
156. Hirsch MS, Kaplan JC (1987) Treatment of human immunodeficiency virus infections. Antimicrob Agents Chernother 31:839-843
157. De Clercq E (1987) Perspectives for the chernotherapy of AIDS. Anticancer Res 7.1023-1038
158. Dalgleish AG, Beverley PC, Clapharn PR, Crawford DH, Greaves MF, Weiss RA (1984) The CD4 (T 4) antigen is an cssential component of the receptor for the AIDS retrovirus. Nature 312:763-767
159. Klatzmann D, Champagnc E, Chamaret S, et al. (1984) T -lymphocyte T 4 molecule behaves as the receptor for human retrovirus LAV. Nature 312:767-768
160. Ashkenazi A, Presta LG, Marsters SA, Camarrato TR, RosenthaI KA, Fandly BM, Capon DJ (1990) Mapping thc CD4 binding site for human immunodeficiency virus by alanine-scanning mutagenesis. Proc Natl Acad Sci USA 87. 7150-7154
161. Lifson JD, Reyes GR, McGrath MS, Stein BS, Englernan EG (1986) AIDS retrovirus induced cytopathology: giant cell formation and involvernent of CD4 antigen. Science 232:1123-1127
162. Lifson JD, Feinberg MB, Reyes GR, et al. (1986) Induction of CD4 dependent cell fusion by the HTL V III/LA V envelopc glycoprotcin. Nature 323:725- 728
163. Capon JD, Chamow SM, Mordenti J, et al. (1989) Designing CD4 immunoadhesins for AIDS therapy. Nature 337 : 525-531
164. Chaudhary VK, Mizukami T, Fuerst TR, et al. (1988) Selective killing of HIV infected cells by recombinant human CD4 Pseudomonas exotoxin hybrid protein. Nature 335:369-372
165. Till MA, Ghetie V, Gregory T, et al. (1988) HIV-infcctcd cc!ls are killed by rCD4-ricin A chain. Scicncc 242:1166-1168
166. Abrams Dr, Kuno S, Wong R, et al. (1989) Oral dextran sulfate (UAO01) in the treatment ofthe acquired immunodeficiency syndrome (ArDS) and AIDSrelated complex. Ann rntern Med 110: 183-188
167. Jacobson MA, Crowe S, Levy J, et al. (1988) Beneficial effect of intermittent intravenous (IV) foscarnet (PF A) therapy on HrV infection in patients with AIDS. 4th International Conference on ArDS, June 12-16, Stockholm, vol2. Swedish Ministry of Health and Social Affairs, Stockholm, p 163
168. Bergdahl S, Sönnerborg A, Albert J, et al. (1988) Antiviral effect against Hrv in patients with AIDS-related complex given intermittent r. v. foscarnet. 4th International Conference on AIDS, June 12-16, Stockholm, vol 2. Swedish Ministry of Health and Social Affairs, Stockholm, p 163 169. Krown SE, Real FX, Cunningham RundIes S, et al. (1983) Preliminary observations on the effect of recombinant leukocyte A interferon in homosexual men with Kaposi's sarcoma. N Engl J Med 308.1071-1076
170. Groopman JE, Gottlieb MS, Goodman J, et al. (1984) Recombinant alpha 2 interferon therapy for Kaposi's sarcoma associated with the acquired immunodeficiency syndrome. Ann intern Med 100:671-676
171. Gelmann EP, Longo D, Lane HC, et al. (1987) Combination chemotherapy of disseminated Kaposi's sarcoma in patients with acquired immunodeficiency syndrome. Am J Med 82:456-462
172. Lane HC, Kovacs JA, Feinberg J, et al. (1988) Anti-retroviral effects of interferon-alpha in AIDS-associated Kaposi's sarcoma. Lancct 2.1218-1222
173. Nabel GJ, Rice SA, Knipe DM, Baltimore D (1988) Alternative mechanisms for activation of human immunodeficiency virus cnhanccr in T cclls. Science 239:1299-1302
174. Mosca JD, Bednarik DP, Raj NB, et al. (1987) Activation of human immunodeficiency virus by herpesvirus infcction: identification of a region within the long terminal repeat that responds to a transacting factor encoded by herpes simplcx virus 1. Proc Natl Acad Sci USA 84. 7408-7412
175. Salahuddin SZ, Ablashi DV, Markharn PD, et al. (1986) Isolation of a new virus, HBL V, in patients with Iyrnphoproliferative disordcrs. Scicnce 234.596-601
176. Van der Vliet PC, Kwant MM (1981) Role ofDNA polymerasc / in adenovirus DNA replication: mechanism of inhibition by 2',3'-dideoxynucleosidc 5'triphosphates. Biochernistry 20.2628-2632
177. Lin J-C, Zhang Z-X, Smith MC, Biron K, Pagano JS (1988) Anti-human im munodeficiency virus agent 3'-azido-3'deoxythyrnidine inhibits replication of Epstein-Barr virus. Antimicrob Agents Chernother 32:265-267
178. Birx DL, Redfield RR, Tosato G (1986) Defective regulation of Epstein- Barr virus infection in patients with acquired immunodeficiency syndrome (A1DS) or A1DS-related disorders. N Engl J Med 314.874-879
179. Yarchoan R, Redfield RR, Broder S (1986) Mechanisms of B cell activation in patients with acquired immunodeficiency syndrome and related disorders. Contribution of antibody-producing B cells, of Epstein-Barr virus-infected B cells, and of immunoglobulin production induced by human T cell lymphotropic virus, type III/lymphadenopathy-associated virus. J Clin Invest 78.439-447
180. Baba M, Pauwels R, Balzarini J, Herdewijn R, de Clercq E, Desmyter J (1987) Ribavirin antagonizes inhibitory effects of pyrimidine 2',3'-dideoxynucleosides but enhances inhibitory effects of purine 2',3'-dideoxynucleosides on replication of human immunodcficicncy virus in vitro. Antimicrob Agents Chernother 31:1613-1617
181. Portegies P, de Gans J, Lange JMA, et al. (1989) Declining incidence of A1DS dementia complex after introduction of zidovudine treatment. Br Med J 299. 819-821
182. De Vita VT, Schein PS (1973) The use of drugs in combination for the treatment of cancer rationale and results. N Eng] J Med 288:998-1006
183. Montgomery AB, Leoung GS, Wardlaw LA, ct al. (1989) Effect of zidovudine on mortality rates and Pneumocystis carinii (PCP) incidence in A1DS and ARC patients on acrosol pentamidine (Abstr). Am Rev Respir Dis [Suppl] 139:A250
184. Henry K, Chinnock BJ, Quinn RP, Fletcher CV, de Miranda P, Balfour HH Jr (1988) Concurrent zidovudine levels in semen and serum determined by radioimmunoassay in patients with AIDS or A1DS-related complcx. JAMA 259. 3023-3026
185. Puckett LD, Orf JW, Hostager BS, Goldensoph CR, Donner JE, Heinke CJ (1988) Characteristics of a two hour RIA for monitoring of the levels of AZT in patients' serum (Abstr). 4th International Conference on AIDS, June 1216, Stockholm, vol 2. Swedish Ministry of Health and Social Affairs, Stockholm, p 177