当前位置: 首页 > 期刊 > 《新英格兰医药杂志》 > 2004年第10期 > 正文
编号:11307499
HIV Drug Resistance
http://www.100md.com 《新英格兰医药杂志》
     The use of combinations of antiretroviral drugs has proven remarkably effective in controlling the progression of human immunodeficiency virus (HIV) disease and prolonging survival,1 but these benefits can be compromised by the development of drug resistance.2,3 Resistance is the consequence of mutations that emerge in the viral proteins targeted by antiretroviral agents. In the United States, as many as 50 percent of patients receiving antiretroviral therapy are infected with viruses that express resistance to at least one of the available antiretroviral drugs.4 Consequently, the transmission of drug-resistant strains is also a growing concern.5,6,7 Because drug-resistant HIV often exhibits resistance to several classes of antiretroviral drugs8 and because cross-resistance between drugs within a class is frequent,9,10,11,12 the emergence of resistance always complicates further efforts to control viral replication. This review focuses on the mechanisms underlying the selection of drug-resistant HIV and on the consequences of viral resistance with respect to the evolution of HIV infection.

    Principles of Drug Therapy for HIV

    The drugs currently used to treat HIV type 1 (HIV-1) infection (Table 1) belong to four distinct classes: nucleoside and nucleotide analogues, which act as DNA-chain terminators and inhibit reverse transcription of the viral RNA genome into DNA, a crucial event occurring at an early stage of the viral life cycle; nonnucleoside reverse-transcriptase inhibitors, which bind and inhibit reverse transcriptase, the viral enzyme that conducts reverse transcription; protease inhibitors, which target the viral protease, the enzyme required for the cleavage of precursor proteins (gag and gag–pol), permitting the final assembly of the inner core of viral particles; and entry inhibitors, which block the penetration of HIV virions into their target cells. Combinations of antiretroviral drugs are now used for the treatment of HIV infection — so-called highly active antiretroviral therapy (HAART). Current HAART regimens generally comprise three antiretroviral drugs, usually two nucleoside analogues and either a protease inhibitor or a nonnucleoside reverse-transcriptase inhibitor.13 The use of agents from different classes is instrumental in controlling the development of resistance, but whereas some drug combinations have been shown to be antagonistic, there is no evidence that any combinations of currently available drugs are strongly synergistic in vitro.

    Table 1. Antiretroviral Agents Used in the Treatment of HIV Infection.

    Development of Resistance

    Induced Resistance

    Two concepts are important to an understanding of the development of drug resistance. First, HIV infection is characterized by high levels of virus production and turnover. In most untreated patients, the total number of productively infected cells in the lymphoid tissue has been estimated to be approximately 107 to 108 cells.14 During the chronic phase of HIV infection, this number is relatively stable, reflecting the balance between the infection of new target cells and their clearance. Because the half-life of infected cells is remarkably short (one to two days), the maintenance of this steady state requires that HIV infect new target cells at a very high rate.15,16,17 Second, the viral population in an infected person is highly heterogeneous.18 The reverse transcription of viral RNA into DNA is notoriously prone to error,19,20 introducing on average one mutation for each viral genome transcribed. Most of these errors are base substitutions, but duplications, insertions, and recombination can also occur. The high rate of HIV infection, combined with the high mutation rate that occurs during each cycle of infection, ensures that patients have a complex and diverse mixture of viral quasispecies, each differing by one or more mutations.

    Under these circumstances, it is easy to understand why if any of these mutations can confer some selective advantage to the virus, such as a decrease in its susceptibility to an antiretroviral agent, the corresponding quasispecies will overtake the others, following a simple darwinian selection process. The rapidity of this process depends on the level of the selective advantage conferred by the mutation, the prevalence of the mutant within the virus population, and the level of drug at the site of HIV replication. In some cases, substitutions of single amino acids can produce high levels of resistance. Since minority viral quasispecies carrying any single mutation are believed to exist even before treatment is started,21 the emergence of these highly resistant single mutants can occur in a matter of weeks. For other agents, only low-level resistance can be induced by single mutations. In these cases, high levels of resistance or complete resistance requires the gradual accumulation of additional mutations.

    Combination therapy can block this selection process for two reasons. First, multiple mechanisms (each requiring different mutations) are required for resistance to occur to all drugs in the regimen. Even if a small number of variants with the potential for resistance to individual agents exist before treatment, it is highly unlikely that some of these variants are able to resist all the drugs. Second, multiple drugs suppress viral replication more effectively than single agents.22,23 In the absence of ongoing viral replication, the generation of new variants is also arrested. In patients who receive HAART as a first line of antiretroviral therapy, the emergence of viral resistance is possible only if HIV continues to replicate in the presence of levels of drugs that are insufficient to block viral replication but sufficient to exert a positive selective pressure on variants with decreased drug susceptibility. Under these conditions, viruses with resistance to all the components of the regimen will gradually emerge. Thus, it is worth emphasizing that in the context of HAART, resistance is most often the consequence — not the cause — of initial treatment failure. Once resistance begins to develop, however, a vicious circle of increasing treatment failure and increasing levels of resistance can lead to situations in which it becomes impossible to control viral replication with currently available drugs.

    Primary Resistance

    Patients primarily infected with HIV strains that exhibit resistance to zidovudine were identified as early as 1993, six years after zidovudine was introduced as the first antiretroviral drug active against HIV.24 Since HAART became available in 1996, a number of reports have described the transmission of HIV strains with resistance to single or multiple antiretroviral drugs.5,6,7 The prevalence of primary HIV resistance appears to be quite variable in different communities, but there is a trend toward an increase.6,7 Although most such cases involve the transmission of strains of HIV-1 from patients in whom resistance has developed during therapy, some strains of HIV are naturally resistant to some antiretroviral drugs. For example, HIV type 2 (HIV-2) is intrinsically resistant to most nonnucleoside reverse-transcriptase inhibitors.25 Similarly, some subtypes of HIV-1 can be less susceptible to protease inhibitors or nonnucleoside reverse-transcriptase inhibitors than the subtype B strains that are prevalent in the United States and Europe.26,27,28,29 More information is needed on the natural susceptibility of non-B subtypes and on the patterns of resistance mutations that occur in these strains, which can differ from those observed with subtype B. Because non-B subtypes are dominant in Africa and Asia, particular care in the choice of antiretroviral regimens must be exercised in those regions.

    Mechanisms of Resistance

    Considerable progress has been made in identifying mutations associated with drug resistance (Table 2) and in understanding the mechanisms through which they confer resistance (Table 1). A variety of mechanisms have been identified that differ both for different classes of drugs and for drugs of a given class. The locations of drug-resistance mutations in reverse transcriptase and in protease are shown in Figure 1 and Figure 3A, respectively.

    Table 2. Mutations Involved in Resistance of HIV to Nucleoside Analogues, Nonnucleoside Reverse-Transcriptase Inhibitors (NNRTIs), and Protease Inhibitors.

    Figure 1. Complex of HIV-1 Reverse Transcriptase with an RNA–DNA Duplex.

    Reverse transcriptase is a heterodimer comprising two subunits: p66, which includes the catalytic site, which has three key aspartic acid residues (D110, D185, and D186), and p51, which does not include domains that are directly involved in the polymerization reaction. NNRTI denotes nonnucleoside reverse-transcriptase inhibitors. Modified from a figure generated by Dr. David Stammers, Oxford University, Oxford, United Kingdom.

    Figure 3. HIV-1 Protease Dimer Binding with a Protease Inhibitor (Panel A) and a Drug-Sensitive (Wild-Type) Protease Juxtaposed against a Drug-Resistant Protease (Panel B).

    Panel A shows the amino acid chains of both subunits of the protease. The protease inhibitor (ritonavir) occupies the central substrate-binding domain of the enzyme. The sites of amino acid residues most frequently involved in resistance to protease inhibitors are shown as red beads. The designation of the corresponding residues is written in red for the first subunit of the protease and in blue for the other subunit. In Panel B, the substrate-binding cavity contains a protease inhibitor (lopinavir) in the context of either the sensitive protease (green) or the resistant protease (red). Modified from figures generated by Dr. Ladislau Kovari, Wayne State University School of Medicine, Detroit.

    Resistance to Nucleoside and Nucleotide Analogues

    Nucleoside analogues and nucleotide analogues (Table 1) arrest the synthesis of viral DNA by reverse transcriptase. After phosphorylation by cellular kinases, these compounds are incorporated by reverse transcriptase into the nascent chain of viral DNA. Because these drugs lack a 3' hydroxyl group, no additional nucleotides can be attached to them, and the synthesis of viral DNA is arrested. Two distinct mechanisms are involved in HIV resistance to these drugs: impairment of the incorporation of the analogue into DNA and removal of the analogue from the prematurely terminated DNA chain.

    Impairment of Analogue Incorporation

    Several mutations or groups of mutations in reverse transcriptase can promote resistance by selectively impairing the ability of reverse transcriptase to incorporate an analogue into DNA (Figure 2A). They essentially include the M184V mutation, the Q151M complex of mutations, and the K65R mutation (Table 2).

    Figure 2. The Two Principal Mechanisms of Resistance of HIV to Nucleoside Analogues.

    In Panel A, the incorporation of a nucleoside analogue into drug-sensitive viruses results in the termination of the viral DNA chain. Mutations in drug-resistant viruses prevent the incorporation of the nucleoside analogue into the growing viral DNA chain. In Panel B, ATP in drug-sensitive viruses does not have access to a reverse transcriptase that has formed a complex with a nucleoside analogue. Mutations that cause resistance to nucleoside analogues, referred to as thymidine analogue mutations, allow ATP to bind reverse transcriptase near the 3' end of viral DNA terminated by the incorporation of a nucleoside analogue. ATP then excises the analogue from viral DNA, allowing reverse transcription to proceed normally.

    The M184V mutation involves the replacement of methionine by valine at position 184 of the reverse transcriptase and is the main mutation that confers resistance to lamivudine.31 Methionine 184 is located at the heart of the catalytic site of reverse transcriptase, and its replacement by a valine, which has a different side chain, interferes with the proper positioning of lamivudine triphosphate within the catalytic site (Figure 1 and Figure 2A).32 The M184V mutation induces very high levels of resistance to lamivudine. When lamivudine is used as a single agent, resistant strains overtake wild-type virus in a few weeks,33 and when lamivudine is used as part of a failing regimen of HAART, the M184V mutation is almost always the first mutation to emerge.34,35

    The group of mutations referred to as the Q151M complex36 (Table 2) is most often selected for in the course of the failure of regimens containing stavudine and didanosine. This pathway always starts with the Q151M substitution, a residue located in the immediate vicinity of the nucleotide-binding site of reverse transcriptase, and is followed by the gradual accumulation of secondary mutations that enhance resistance and increase the activity of the enzyme.37 The Q151M complex is relatively rare in HIV-1 (fewer than 5 percent of all HIV strains with resistance to nucleoside analogues) but can confer high-level resistance to most — but not all (e.g., lamivudine and tenofovir) — analogues.36 Interestingly, the Q151M complex is markedly more frequent in HIV-2 than in HIV-1.

    The K65R mutation is seen with increasing frequency in patients in whom therapy with nucleoside or nucleotide analogues fails, especially when the regimen includes tenofovir or abacavir. This mutation appears to confer resistance to most analogues, with the exception of zidovudine.

    Removal of the Analogue from the Terminated DNA Chain

    Removal of the nucleoside analogue from the terminated DNA chain is associated with a group of mutations commonly termed "thymidine analogue mutations" (Table 2). Mutations from this group are most frequently selected for after the failure of drug combinations that include thymidine analogues, such as zidovudine and stavudine, but they can promote resistance to almost all nucleoside and nucleotide analogues, including tenofovir.38,39,40,41 These mutations occur gradually, and their order of emergence can vary.10,42 Thymidine analogue mutations promote resistance by fostering ATP- or pyrophosphate-mediated removal of nucleoside analogues from the 3' end of the terminated DNA strand (Figure 2B). 43,44 ATP and pyrophosphate, which are abundant in normal lymphocytes, do not participate in the DNA-polymerization reaction, but the structure of a reverse transcriptase expressing thymidine analogue mutations facilitates their entry into a site adjacent to the incorporated analogue.45,46 In this position, ATP or pyrophosphate can attack the phosphodiester bond that links the analogue to DNA, resulting in removal of the analogue (Figure 2B). Interestingly, the efficiency of this process, also known as "primer rescue," can be significantly decreased by the presence of other mutations in reverse transcriptase, a phenomenon that has been best described in the case of the M184V mutation.47 As a consequence, M184V slows the selection of thymidine analogue mutations by thymidine analogues41 and may slightly increase the residual antiviral activity of some nucleoside analogues in spite of the presence of thymidine analogue mutations.

    Resistance to Nonnucleoside Reverse-Transcriptase Inhibitors

    Nonnucleoside reverse-transcriptase inhibitors are small molecules that have a strong affinity for a hydrophobic pocket located close to the catalytic domain of the reverse transcriptase (Figure 1 and Figure 4). The binding of the inhibitors affects the flexibility of the enzyme, thereby blocking its ability to synthesize DNA.48 The mutations that are selected for after the failure of treatment with nonnucleoside reverse-transcriptase inhibitors are all located in the pocket targeted by these compounds, and they reduce the affinity of the drug.48,49,50,51,52,53 Because of subtle differences in the interaction between various nonnucleoside reverse-transcriptase inhibitors and the hydrophobic pocket,51 however, the mutations that emerge most frequently are somewhat drug-dependent (Table 2). Resistance to nevirapine is often associated with the Y181C mutation, but other mutations, such as Y188C, K103N, G190A, and V106A, also occur. Initial resistance to efavirenz is generally characterized by the K103N mutation, but the Y188L mutation is also seen.

    Figure 4. Mechanism of Resistance of HIV to Nonnucleoside Reverse-Transcriptase Inhibitors.

    Nonnucleoside reverse-transcriptase inhibitors (NNRTIs) have a strong affinity for a pocket next to the active site of reverse transcriptase. In drug-sensitive viruses, NNRTIs bind this pocket and block the polymerization of DNA by reverse transcriptase. In drug-resistant viruses, mutations prevent the binding of NNRTIs, allowing DNA polymerization to proceed normally.

    Resistance to Protease Inhibitors

    The HIV protease cleaves large polyprotein precursors at specific sites, releasing the structural proteins and enzymes necessary for the assembly of infectious viral particles. In the absence of a functional protease, viral particles are produced, but they are immature and are not infectious. The protease of HIV is a symmetrically assembled homodimer with a central, symmetric, substrate-binding cavity (Figure 3A). Detailed knowledge of the structure of this domain and of the structure of the natural protein substrates of the enzyme has led to the design of specific inhibitors whose chemical structure mimics that of the viral peptides that are normally recognized and cleaved by the protease.54,55 These compounds display a strong affinity for the active site of the HIV protease and inhibit the catalytic activity of the enzyme in a highly selective manner.

    Resistance to protease inhibitors is the consequence of amino acid substitutions that emerge either inside the substrate-binding domain of the enzyme or at distant sites56,57,58 (Table 2 and Figure 3A). Directly or indirectly, these amino acid changes modify the number and the nature of the points of contact between the inhibitors and the protease, thereby reducing their affinity for the enzyme.59,60,61,62 As an example, the common resistance mutation V82A reduces the size of an amino acid residue in the protease that is more important for binding most inhibitors than for binding the natural viral protein substrate (Figure 3A). 62 Protease inhibitors have been designed to bind the protease with maximal affinity and tend to occupy more space inside the active site cavity than do natural substrates. Unlike the inhibitors, the natural substrates of the protease have a variable, but generally less tight, interaction with the catalytic site, a phenomenon that promotes the ordered sequential cleavage of the polyproteins required for proper assembly of the viral particle. Resistance mutations in the protease, which result in an overall enlargement of the catalytic site of the enzyme (Figure 3B), would thus be predicted to have a greater effect on the binding of inhibitors than the natural templates.

    Some mutations are selected for only by certain protease inhibitors (Table 2), reflecting particularities in the chemical structure of the inhibitors that influence their interaction with the substrate-binding domain of the enzyme. However, there is considerable overlap between the combinations of mutations in HIV strains that develop resistance to protease inhibitors. This overlap explains the wide cross-resistance that is generally observed within this drug class.9,12,63 Remarkably, resistance to protease inhibitors can also be promoted by mutations in some of the natural viral substrates of the protease.64,65,66 Characteristic substitutions of amino acids near cleavage sites of the gag polyprotein have been identified that can increase the level of resistance and the replicative capacity of the virus by facilitating cleavage under conditions in which the amount of active enzyme is suboptimal or improving the ability of proteases containing resistance mutations to interact with the substrate.

    Resistance to Fusion Inhibitors

    HIV-1 enters target cells through an intricate sequence of interactions between the HIV envelope glycoprotein (gp) complex (gp120–gp41) and specific cell-surface receptors.67 The early steps in this process allow gp41, the fusogenic component of the complex, to interact with the cell membrane, thereby tethering the virus to its target. The membranes of the virus and target cell are then brought into close proximity, fostering their fusion, by further rearrangement of gp41. In this step, a distal hydrophobic region of gp41, HR2, folds onto a more proximal hydrophobic region, HR1, effectively shortening the molecule. Enfuvirtide, a 36-amino-acid peptide derived from HR2, destabilizes this process by binding to HR1 and blocks the infectivity of HIV-1. Viral resistance to enfuvirtide usually results from mutations located in a stretch of 10 amino acids within HR1.68,69 Interestingly, changes in amino acids in gp41 outside HR1 — and even changes in gp120 — appear to be associated with significant differences in the susceptibility of the virus to enfuvirtide.70,71 These mutations or polymorphisms probably explain the remarkably wide range of natural susceptibility to enfuvirtide among HIV-1 strains70,71,72 and could participate in the evolution of acquired resistance to enfuvirtide.

    Cross-Resistance

    Cross-resistance, defined as resistance to drugs to which a virus has never been exposed, results from mutations that have been selected for by the use of another drug. Cross-resistance is always restricted to drugs within a given class of antiretroviral agents, but all three classes of antiretroviral drugs are affected. Early in the evolution of resistance to nucleoside analogues or protease inhibitors, viruses may have only a low level of cross-resistance to alternative agents within each of these two classes of drugs.10,12 Nevertheless, these strains may need to add only one or a few additional mutations to this preexisting scaffolding for high-level cross-resistance to develop. Therefore, in patients infected with strains that have low levels of cross-resistance, the switch to apparently active alternative drugs can be accompanied by rapid selection for highly resistant variants, at the expense of minimal evolutionary changes.73,74

    Evolution of Resistance

    Resistance is not an all-or-nothing phenomenon and generally increases over time.10,75,76 Single mutations rarely produce complete resistance to antiretroviral drugs, although the M184V mutation in reverse transcriptase, which results in complete resistance to lamivudine, is an exception to this rule. Single mutations in the hydrophobic pocket of reverse transcriptase also provide strong resistance to nonnucleoside reverse-transcriptase inhibitors, but the viral strains in patients in whom regimens using these drugs are failing often incur additional mutations, suggesting that the level of resistance provided by single mutations is not optimal.50,77

    Resistance to reverse-transcriptase inhibitors through the accumulation of thymidine analogue mutations or Q151M complex mutations and resistance to protease inhibitors are always gradual processes, leading to progressive increases in the level of resistance.58,78 Nonetheless, even after several mutations have accumulated in the HIV protease and reverse transcriptase sufficient to produce patent treatment failure, resistance may not have reached maximal levels, and additional mutations, associated with increases in resistance, can occur even in the absence of any changes in treatment.75 Indeed, the development of complete resistance may represent an exception. Because resistance mutations can impair viral replicative capacity, the solution adopted by the dominant viral population may entail making concessions in terms of resistance. Efforts are being made to evaluate the antiviral activity retained by individual drugs in the face of resistance mutations. Such knowledge may help in the treatment of patients infected with viruses that express resistance to multiple classes of drugs, since keeping HIV under some pharmacologic pressure may reduce its pathogenic potential.

    Effect of Resistance on Viral Replicative and Pathogenic Capacity

    Many resistance mutations impair viral replication. Because these mutations modify key viral proteins, they have deleterious effects of variable extent on protein function. Although some of these deficits can be partially corrected by compensatory mutations,57,79 viruses forced to develop higher levels of resistance under intense and continuous pressure by antiretroviral drugs often have a substantial impairment in their replicative capacity.75 The degree of replicative impairment conferred by resistance mutations is highly variable. The most crippling mutations appear to be those associated with resistance to protease inhibitors,65,80,81,82 but significant replicative defects can be related to mutations in reverse transcriptase that confer resistance to nucleoside analogues or nonnucleoside reverse-transcriptase inhibitors,83,84 as well as envelope mutations associated with resistance to enfuvirtide.

    The clinical effect of resistance-associated loss of viral replicative capacity is the subject of intense investigation. Several observations suggest that resistant viruses have lost some of their virulence. When antiretroviral treatment is interrupted in patients infected with HIV that is resistant to multiple drugs, the resistant strain is more or less rapidly replaced by wild-type virus. This change is accompanied by a drop in CD4 T-cell counts, suggesting that wild-type virus has a greater replicative and pathogenic potential.85 In a significant proportion of patients in whom HAART is failing, the CD4 T-cell counts remain significantly above pretreatment levels, despite the poor control of replication.86,87,88 Whether this apparent immunologic benefit of HAART in spite of patent virologic failure is the direct consequence of reduced viral pathogenicity, a sign of persistent residual activity of some drugs in the regimen, or both, remains to be elucidated. Consequently, therapeutic strategies that take advantage of resistance-associated loss of HIV replicative capacity have yet to be identified.

    Minority and Archived Populations of Virus

    The dominant population of resistant virus in the plasma does not always reflect the complex diversity of viral quasispecies in patients in whom HAART is failing. Like the viral populations present in untreated HIV-infected persons,18 those present in patients in whom therapy is failing remain quite heterogeneous.89,90 Minority populations of virus expressing a variety of distinct combinations of resistance mutations are generally present and can continue to evolve.91 Thus, these populations serve as a reservoir for the generation of novel resistance genotypes that can ultimately supplant the previously dominant population. Furthermore, throughout the natural history of HIV infection, viral genomes are continually being archived as latently integrated proviruses in resting cells.92,93 When patients in whom drug resistance has developed are treated with alternative agents for long periods, the mutations associated with resistance to the initially prescribed drugs are often no longer detectable in viruses obtained from plasma samples. Nevertheless, if therapy with these drugs is later resumed, archived resistant strains can reemerge,94,95 as the cells harboring these viruses become activated.

    Testing for Drug Resistance

    When plasma viral load rises in spite of therapy, it is currently recommended that antiretroviral treatment be changed quickly.96,97 Because cross-resistance between antiretroviral drugs within each class is frequent, changes in treatment cannot be based on the simple assumption that HIV will remain susceptible to alternative drugs within a class that the patient has previously received. In an effort to facilitate the selection of efficacious alternative regimens, resistance tests evaluating the susceptibility of HIV to individual antiretroviral drugs have been developed. Two types of assays are currently available: genotypic assays, which detect the presence of resistance mutations, and phenotypic assays, which measure the susceptibility of the virus to various drugs in tissue-culture systems. A number of retrospective studies have attempted to validate the clinical relevance of the results of resistance tests.97,98 In general, resistance to a drug has been found to be associated with reduced responsiveness of HIV to the drug in vivo, but the finding of apparent susceptibility on resistance testing is a somewhat less reliable predictor of a good response. This common observation can be explained by a number of factors, including the imprecision of some of the assays, the short evolutionary distance required for some initially susceptible viruses to develop full cross-resistance to the new agents if viral replication is not completely suppressed,73,74 the existence of confounding variables such as the pharmacokinetics of individual drugs, and the presence of species of archived or minority viruses with alternative resistance profiles,89,90,91 which are usually not detected by the assays. Prospective, randomized studies have examined the clinical benefit of drug-resistance testing.97,98 These studies evaluated patients infected with viruses in different stages of the evolution of resistance and have yielded somewhat conflicting results, but most have demonstrated that genotypic testing provides at least a moderate benefit.

    Perspectives

    As the inevitable consequence of the incomplete suppression of HIV-1 replication by antiretroviral drugs, resistance is a permanent threat for patients who are undergoing antiretroviral treatment, and transmission of resistant viruses is becoming an important concern. Prevention of resistance is a priority that requires unrelenting patient education regarding the risks of resistance and the use of improved drug regimens that ensure optimal tolerance, adherence, and potency. Once established, resistance evolves, diversifies, and may become irreversible. Nonetheless, new drugs are becoming available that appear to retain substantial antiviral activity against HIV-1 strains that are resistant to multiple drugs. These are either drugs from existing classes that have increased potency and improved pharmacokinetic properties or drugs from new classes that are not susceptible to cross-resistance. Although preliminary data indicate that viral resistance to these new drugs can also develop,99,100 the lessons learned about the development of viral resistance to the currently available antiretroviral drugs may prove helpful in devising treatment strategies with optimized antiviral potency that can minimize the development of resistance to these new agents.

    Dr. Clavel reports having served as a consultant for GlaxoSmithKline, Bristol-Myers Squibb, and Bioalliance Pharma; holding stock in Bioalliance Pharma; and serving as a lecturer for GlaxoSmithKline, Bristol-Myers Squibb, Roche, and Gilead.

    Source Information

    From the Unité de Recherche Antivirale, Institut National de la Santé et de la Recherche Médicale, Unité 552, H?pital Bichat–Claude Bernard, Paris.

    Address reprint requests to Dr. Clavel at INSERM, U552, IMEA, H?pital Bichat–Claude Bernard, 46 rue Henri Huchard, 75018 Paris, France, or at clavel@bichat.inserm.fr.

    References

    Palella FJ Jr, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. N Engl J Med 1998;338:853-860.

    DeGruttola V, Dix L, D'Aquila R, et al. The relation between baseline HIV drug resistance and response to antiretroviral therapy: re-analysis of retrospective and prospective studies using a standardized data analysis plan. Antivir Ther 2000;5:41-48.

    Ledergerber B, Egger M, Erard V, et al. AIDS-related opportunistic illnesses occurring after initiation of potent antiretroviral therapy: the Swiss HIV Cohort Study. JAMA 1999;282:2220-2226.

    Richman D, Bozette S, Morton S, et al. The prevalence of antiretroviral drug resistance in the US. In: Program and abstracts of the Interscience Conference on Antimicrobial Agents and Chemotherapy. Chicago, December 16–19, 2001. Washington, D.C.: American Society for Microbiology, 2001. abstract.

    Yerly S, Kaiser L, Race E, Bru JP, Clavel F, Perrin L. Transmission of antiretroviral-drug-resistant HIV-1 variants. Lancet 1999;354:729-733.

    Little SJ, Holte S, Routy J-P, et al. Antiretroviral-drug resistance among patients recently infected with HIV. N Engl J Med 2002;347:385-394.

    Grant RM, Hecht FM, Warmerdam M, et al. Time trends in primary HIV-1 drug resistance among recently infected persons. JAMA 2002;288:181-188.

    Shafer RW, Winters MA, Palmer S, Merigan TC. Multiple concurrent reverse transcriptase and protease mutations and multidrug resistance of HIV-1 isolates from heavily treated patients. Ann Intern Med 1998;128:906-911.

    Hertogs K, Bloor S, Kemp SD, et al. Phenotypic and genotypic analysis of clinical HIV-1 isolates reveals extensive protease inhibitor cross-resistance: a survey of over 6000 samples. AIDS 2000;14:1203-1210.

    Richman DD. Susceptibility to nucleoside analogues of zidovudine-resistant isolates of human immunodeficiency virus. Am J Med 1990;88:8S-10S.

    Miller V, Larder BA. Mutational patterns in the HIV genome and cross-resistance following nucleoside and nucleotide analogue drug exposure. Antivir Ther 2001;6:Suppl 3:25-44.

    Race E, Dam E, Obry V, Paulous S, Clavel F. Analysis of HIV cross-resistance to protease inhibitors using a rapid single-cycle recombinant virus assay for patients failing on combination therapies. AIDS 1999;13:2061-2068.

    Yeni PG, Hammer SM, Carpenter CC, et al. Antiretroviral treatment for adult HIV infection in 2002: updated recommendations of the International AIDS Society-USA Panel. JAMA 2002;288:222-235.

    Haase AT. Population biology of HIV-1 infection: viral and CD4+ T cell demographics and dynamics in lymphatic tissues. Annu Rev Immunol 1999;17:625-656.

    Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, Markowitz M. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 1995;373:123-126.

    Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho DD. HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span, and viral generation time. Science 1996;271:1582-6.

    Wei X, Ghosh SK, Taylor ME, et al. Viral dynamics in human immunodeficiency virus type 1 infection. Nature 1995;373:117-122.

    Meyerhans A, Cheynier R, Albert J, et al. Temporal fluctuations in HIV quasispecies in vivo are not reflected by sequential HIV isolations. Cell 1989;58:901-910.

    Roberts JD, Bebenek K, Kunkel TA. The accuracy of reverse transcriptase from HIV-1. Science 1988;242:1171-1173.

    Preston BD, Poiesz BJ, Loeb LA. Fidelity of HIV-1 reverse transcriptase. Science 1988;242:1168-1171.

    Coffin JM. HIV population dynamics in vivo: implications for genetic variation, pathogenesis, and therapy. Science 1995;267:483-489.

    Gulick RM, Mellors JW, Havlir D, et al. Treatment with indinavir, zidovudine, and lamivudine in adults with human immunodeficiency virus infection and prior antiretroviral therapy. N Engl J Med 1997;337:734-739.

    Hammer SM, Squires KE, Hughes MD, et al. A controlled trial of two nucleoside analogues plus indinavir in persons with human immunodeficiency virus infection and CD4 cell counts of 200 per cubic millimeter or less. N Engl J Med 1997;337:725-733.

    Erice A, Mayers DL, Strike DG, et al. Primary infection with zidovudine-resistant human immunodeficiency virus type 1. N Engl J Med 1993;328:1163-1165.

    Witvrouw M, Pannecouque C, Van Laethem K, Desmyter J, De Clercq E, Vandamme AM. Activity of non-nucleoside reverse transcriptase inhibitors against HIV-2 and SIV. AIDS 1999;13:1477-1483.

    Shafer RW, Eisen JA, Merigan TC, Katzenstein DA. Sequence and drug susceptibility of subtype C reverse transcriptase from human immunodeficiency virus type 1 seroconverters in Zimbabwe. J Virol 1997;71:5441-5448.

    Descamps D, Collin G, Letourneur F, et al. Susceptibility of human immunodeficiency virus type 1 group O isolates to antiretroviral agents: in vitro phenotypic and genotypic analyses. J Virol 1997;71:8893-8898.

    Palmer S, Alaeus A, Albert J, Cox S. Drug susceptibility of subtypes A, B, C, D, and E human immunodeficiency virus type 1 primary isolates. AIDS Res Hum Retroviruses 1998;14:157-162.

    Descamps D, Apetrei C, Collin G, Damond F, Simon F, Brun-Vezinet F. Naturally occurring decreased susceptibility of HIV-1 subtype G to protease inhibitors. AIDS 1998;12:1109-1111.

    HIV drug resistance mutations. San Francisco: International AIDS Society-USA, 2002. (Accessed February 9, 2004, at http://www.iasusa.org/resistance_mutations/index.html.)

    Boucher CA, Cammack N, Schipper P, et al. High-level resistance to (-) enantiomeric 2'-deoxy-3'-thiacytidine in vitro is due to one amino acid substitution in the catalytic site of human immunodeficiency virus type 1 reverse transcriptase. Antimicrob Agents Chemother 1993;37:2231-2234.

    Sarafianos SG, Das K, Clark AD Jr, et al. Lamivudine (3TC) resistance in HIV-1 reverse transcriptase involves steric hindrance with beta-branched amino acids. Proc Natl Acad Sci U S A 1999;96:10027-10032.

    Schuurman R, Nijhuis M, van Leeuwen R, et al. Rapid changes in human immunodeficiency virus type 1 RNA load and appearance of drug-resistant virus populations in persons treated with lamivudine (3TC). J Infect Dis 1995;171:1411-1419.

    Descamps D, Flandre P, Calvez V, et al. Mechanisms of virologic failure in previously untreated HIV-infected patients from a trial of induction-maintenance therapy. JAMA 2000;283:205-211.

    Havlir DV, Hellmann NS, Petropoulos CJ, et al. Drug susceptibility in HIV infection after viral rebound in patients receiving indinavir-containing regimens. JAMA 2000;283:229-234.

    Iversen AK, Shafer RW, Wehrly K, et al. Multidrug-resistant human immunodeficiency virus type 1 strains resulting from combination antiretroviral therapy. J Virol 1996;70:1086-1090.

    Kosalaraksa P, Kavlick MF, Maroun V, Le R, Mitsuya H. Comparative fitness of multi-dideoxynucleoside-resistant human immunodeficiency virus type 1 (HIV-1) in an in vitro competitive HIV-1 replication assay. J Virol 1999;73:5356-5363.

    Larder BA, Kemp SD. Multiple mutations in HIV-1 reverse transcriptase confer high-level resistance to zidovudine (AZT). Science 1989;246:1155-1158.

    Coakley EP, Gillis JM, Hammer SM. Phenotypic and genotypic resistance patterns of HIV-1 isolates derived from individuals treated with didanosine and stavudine. AIDS 2000;14:F9-F15.

    Shafer RW, Winters MA, Jellinger RM, Merigan TC. Zidovudine resistance reverse transcriptase mutations during didanosine monotherapy. J Infect Dis 1996;174:448-449.

    Picard V, Angelini E, Maillard A, et al. Comparison of genotypic and phenotypic resistance patterns of human immunodeficiency virus type 1 isolates from patients treated with stavudine and didanosine or zidovudine and lamivudine. J Infect Dis 2001;184:781-784.

    Boucher CA, O'Sullivan E, Mulder JW, et al. Ordered appearance of zidovudine resistance mutations during treatment of 18 human immunodeficiency virus-positive subjects. J Infect Dis 1992;165:105-110.

    Meyer PR, Matsuura SE, Mian AM, So AG, Scott WA. A mechanism of AZT resistance: an increase in nucleotide-dependent primer unblocking by mutant HIV-1 reverse transcriptase. Mol Cell 1999;4:35-43.

    Arion D, Kaushik N, McCormick S, Borkow G, Parniak MA. Phenotypic mechanism of HIV-1 resistance to 3'-azido-3'-deoxythymidine (AZT): increased polymerization processivity and enhanced sensitivity to pyrophosphate of the mutant viral reverse transcriptase. Biochemistry 1998;37:15908-15917.

    Boyer PL, Sarafianos SG, Arnold E, Hughes SH. Selective excision of AZTMP by drug-resistant human immunodeficiency virus reverse transcriptase. J Virol 2001;75:4832-4842.

    Chamberlain PP, Ren J, Nichols CE, et al. Crystal structures of zidovudine- or lamivudine-resistant human immunodeficiency virus type 1 reverse transcriptases containing mutations at codons 41, 184, and 215. J Virol 2002;76:10015-10019.

    Larder BA, Kemp SD, Harrigan PR. Potential mechanism for sustained antiretroviral efficacy of AZT-3TC combination therapy. Science 1995;269:696-699.

    Esnouf RM, Ren J, Hopkins AL, et al. Unique features in the structure of the complex between HIV-1 reverse transcriptase and the bis(heteroaryl)piperazine (BHAP) U-90152 explain resistance mutations for this nonnucleoside inhibitor. Proc Natl Acad Sci U S A 1997;94:3984-3989.

    Richman DD, Havlir D, Corbeil J, et al. Nevirapine resistance mutations of human immunodeficiency virus type 1 selected during therapy. J Virol 1994;68:1660-1666.

    Bacheler LT, Anton ED, Kudish P, et al. Human immunodeficiency virus type 1 mutations selected in patients failing efavirenz combination therapy. Antimicrob Agents Chemother 2000;44:2475-2484.

    Boyer PL, Currens MJ, McMahon JB, Boyd MR, Hughes SH. Analysis of nonnucleoside drug-resistant variants of human immunodeficiency virus type 1 reverse transcriptase. J Virol 1993;67:2412-2420.

    Ren J, Nichols C, Bird L, et al. Structural mechanisms of drug resistance for mutations at codons 181 and 188 in HIV-1 reverse transcriptase and the improved resilience of second generation non-nucleoside inhibitors. J Mol Biol 2001;312:795-805.

    Hsiou Y, Ding J, Das K, et al. The Lys103Asn mutation of HIV-1 RT: a novel mechanism of drug resistance. J Mol Biol 2001;309:437-445.

    Roberts NA, Martin JA, Kinchington D, et al. Rational design of peptide-based HIV proteinase inhibitors. Science 1990;248:358-361.

    Erickson J, Kempf D. Structure-based design of symmetric inhibitors of HIV-1 protease. Arch Virol Suppl 1994;9:19-29.

    Condra JH, Schleif WA, Blahy OM, et al. In vivo emergence of HIV-1 variants resistant to multiple protease inhibitors. Nature 1995;374:569-571.

    Kaplan AH, Michael SF, Wehbie RS, et al. Selection of multiple human immunodeficiency virus type 1 variants that encode viral proteases with decreased sensitivity to an inhibitor of the viral protease. Proc Natl Acad Sci U S A 1994;91:5597-5601.

    Molla A, Korneyeva M, Gao Q, et al. Ordered accumulation of mutations in HIV protease confers resistance to ritonavir. Nat Med 1996;2:760-766.

    Chen Z, Li Y, Schock HB, Hall D, Chen E, Kuo LC. Three-dimensional structure of a mutant HIV-1 protease displaying cross-resistance to all protease inhibitors in clinical trials. J Biol Chem 1995;270:21433-21436.

    Ridky TW, Kikonyogo A, Leis J, et al. Drug-resistant HIV-1 proteases identify enzyme residues important for substrate selection and catalytic rate. Biochemistry 1998;37:13835-13845.

    Hong L, Zhang XC, Hartsuck JA, Tang J. Crystal structure of an in vivo HIV-1 protease mutant in complex with saquinavir: insights into the mechanisms of drug resistance. Protein Sci 2000;9:1898-1904.

    Prabu-Jeyabalan M, King N, Nalivaika E, Scott W, Schiffer C. Drug resistance and substrate recognition in HIV-1 protease. Antiviral Ther 2002;7:Suppl 1:S36. abstract.

    Schapiro JM, Winters MA, Lawrence J, Merigan TC. Clinical cross-resistance between the HIV-1 protease inhibitors saquinavir and indinavir and correlations with genotypic mutations. AIDS 1999;13:359-365.

    Doyon L, Croteau G, Thibeault D, Poulin F, Pilote L, Lamarre D. Second locus involved in human immunodeficiency virus type 1 resistance to protease inhibitors. J Virol 1996;70:3763-3769.

    Mammano F, Petit C, Clavel F. Resistance-associated loss of viral fitness in human immunodeficiency virus type 1: phenotypic analysis of protease and gag coevolution in protease inhibitor-treated patients. J Virol 1998;72:7632-7637.

    Zhang Y, Imamichi H, Imamichi T, et al. Drug resistance during indinavir therapy is caused by mutations in the protease gene and in its Gag substrate cleavage sites. J Virol 1997;71:6662-6670.

    Kilby JM, Eron JJ. Novel therapies based on mechanisms of HIV-1 cell entry. N Engl J Med 2003;348:2228-2238.

    Rimsky LT, Shugars DC, Matthews TJ. Determinants of human immunodeficiency virus type 1 resistance to gp41-derived inhibitory peptides. J Virol 1998;72:986-993.

    Wei X, Decker JM, Liu H, et al. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob Agents Chemother 2002;46:1896-1905.

    Derdeyn CA, Decker JM, Sfakianos JN, et al. Sensitivity of human immunodeficiency virus type 1 to fusion inhibitors targeted to the gp41 first heptad repeat involves distinct regions of gp41 and is consistently modulated by gp120 interactions with the coreceptor. J Virol 2001;75:8605-8614.

    Reeves JD, Gallo SA, Ahmad N, et al. Sensitivity of HIV-1 to entry inhibitors correlates with envelope/coreceptor affinity, receptor density, and fusion kinetics. Proc Natl Acad Sci U S A 2002;99:16249-16254.

    Labrosse B, Labernardiere JL, Dam E, et al. Baseline susceptibility of primary human immunodeficiency virus type 1 to entry inhibitors. J Virol 2003;77:1610-1613.

    Dulioust A, Paulous S, Guillemot L, Delavalle AM, Boue F, Clavel F. Constrained evolution of human immunodeficiency virus type 1 protease during sequential therapy with two distinct protease inhibitors. J Virol 1999;73:850-854.

    Droz C, Morand-Joubert L, Raguin G, et al. Impact and evolution of resistance in patients treated by a salvage regimen combining amprenavir, lopinavir and ritonavir (the puzzle1 study). Antiviral Ther 2002;7:Suppl 1:S111. abstract.

    Barbour JD, Wrin T, Grant RM, et al. Evolution of phenotypic drug susceptibility and viral replication capacity during long-term virologic failure of protease inhibitor therapy in human immunodeficiency virus-infected adults. J Virol 2002;76:11104-11112.

    Ross L, Johnson M, DeMasi R, et al. Viral genetic heterogeneity in HIV-1-infected individuals is associated with increasing use of HAART and higher viremia. AIDS 2000;14:813-819.

    Hanna GJ, Johnson VA, Kuritzkes DR, et al. Patterns of resistance mutations selected by treatment of human immunodeficiency virus type 1 infection with zidovudine, didanosine, and nevirapine. J Infect Dis 2000;181:904-911.

    Mammano F, Trouplin V, Zennou V, Clavel F. Retracing the evolutionary pathways of human immunodeficiency virus type 1 resistance to protease inhibitors: virus fitness in the absence and in the presence of drug. J Virol 2000;74:8524-8531.

    Nijhuis M, Schuurman R, de Jong D, et al. Increased fitness of drug resistant HIV-1 protease as a result of acquisition of compensatory mutations during suboptimal therapy. AIDS 1999;13:2349-2359.

    Croteau G, Doyon L, Thibeault D, McKercher G, Pilote L, Lamarre D. Impaired fitness of human immunodeficiency virus type 1 variants with high-level resistance to protease inhibitors. J Virol 1997;71:1089-1096.

    Martinez-Picado J, Savara AV, Sutton L, D'Aquila RT. Replicative fitness of protease inhibitor-resistant mutants of human immunodeficiency virus type 1. J Virol 1999;73:3744-3752.

    Zennou V, Mammano F, Paulous S, Mathez D, Clavel F. Loss of viral fitness associated with multiple Gag and Gag-Pol processing defects in human immunodeficiency virus type 1 variants selected for resistance to protease inhibitors in vivo. J Virol 1998;72:3300-3306.

    Back NK, Nijhuis M, Keulen W, et al. Reduced replication of 3TC-resistant HIV-1 variants in primary cells due to a processivity defect of the reverse transcriptase enzyme. EMBO J 1996;15:4040-4049.

    Bleiber G, Munoz M, Ciuffi A, Meylan P, Telenti A. Individual contributions of mutant protease and reverse transcriptase to viral infectivity, replication, and protein maturation of antiretroviral drug-resistant human immunodeficiency virus type 1. J Virol 2001;75:3291-3300.

    Deeks SG, Wrin T, Liegler T, et al. Virologic and immunologic consequences of discontinuing combination antiretroviral-drug therapy in HIV-infected patients with detectable viremia. N Engl J Med 2001;344:472-480.

    Kaufmann D, Pantaleo G, Sudre P, Telenti A. CD4-cell count in HIV-1-infected individuals remaining viraemic with highly active antiretroviral therapy (HAART): Swiss HIV Cohort Study. Lancet 1998;351:723-724.

    Piketty C, Castiel P, Belec L, et al. Discrepant responses to triple combination antiretroviral therapy in advanced HIV disease. AIDS 1998;12:745-750.

    Deeks SG, Barbour JD, Martin JN, Swanson MS, Grant RM. Sustained CD4+ T cell response after virologic failure of protease inhibitor-based regimens in patients with human immunodeficiency virus infection. J Infect Dis 2000;181:946-953.

    Resch W, Parkin N, Stuelke EL, Watkins T, Swanstrom R. A multiple-site-specific heteroduplex tracking assay as a tool for the study of viral population dynamics. Proc Natl Acad Sci U S A 2001;98:176-181.

    Hance AJ, Lemiale V, Izopet J, et al. Changes in human immunodeficiency virus type 1 populations after treatment interruption in patients failing antiretroviral therapy. J Virol 2001;75:6410-6417.

    Charpentier C, Dwyer D, Lecossier D, Clavel F, Hance A. Co-evolution and competition of viral populations with distinct resistance genotypes in patients failing treatment with protease inhibitors. Antiviral Ther 2002;7:Suppl 1:S42. abstract.

    Finzi D, Hermankova M, Pierson T, et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 1997;278:1295-1300.

    Wong JK, Hezareh M, Gunthard HF, et al. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 1997;278:1291-1295.

    Izopet J, Souyris C, Hance A, et al. Evolution of human immunodeficiency virus type 1 populations after resumption of therapy following treatment interruption and shift in resistance genotype. J Infect Dis 2002;185:1506-1510.

    Verhofstede C, Wanzeele FV, Van Der Gucht B, De Cabooter N, Plum J. Interruption of reverse transcriptase inhibitors or a switch from reverse transcriptase to protease inhibitors resulted in a fast reappearance of virus strains with a reverse transcriptase inhibitor-sensitive genotype. AIDS 1999;13:2541-2546.

    Dybul M, Fauci AS, Bartlett JG, Kaplan JE, Pau AK. Guidelines for using antiretroviral agents among HIV-infected adults and adolescents: recommendations of the Panel on Clinical Practices for Treatment of HIV. MMWR Recomm Rep 2002;51:1-55.

    Hirsch MS, Brun-Vezinet F, Clotet B, et al. Antiretroviral drug resistance testing in adults infected with human immunodeficiency virus type 1: 2003 recommendations of an International AIDS Society-USA Panel. Clin Infect Dis 2003;37:113-128.

    Haubrich R, Demeter L. International perspectives on antiretroviral resistance: clinical utility of resistance testing: retrospective and prospective data supporting use and current recommendations. J Acquir Immune Defic Syndr 2001;26:Suppl 1:S51-S59.

    Hazuda DJ, Felock P, Witmer M, et al. Inhibitors of strand transfer that prevent integration and inhibit HIV-1 replication in cells. Science 2000;287:646-650.

    Trkola A, Kuhmann SE, Strizki JM, et al. HIV-1 escape from a small molecule, CCR5-specific entry inhibitor does not involve CXCR4 use. Proc Natl Acad Sci U S A 2002;99:395-400.

    Related Letters:

    HIV Drug Resistance

    Calmy A., Pascual F., Ford N.(Fran?ois Clavel, M.D., an)