Antiretroviral Treatment for Adult HIV Infection in 2002  
 
Updated Recommendations of the International AIDS Society-USA Panel 
 
Author Information  Patrick G. Yeni, MD; Scott M. Hammer, MD; Charles C. J. Carpenter, MD; David A. Cooper, MD, DSc; Margaret A. Fischl, MD; Jose M. Gatell, MD, PhD; Brian G. Gazzard, MA, MD; Martin S. Hirsch, MD; Donna M. Jacobsen, BS; David A. Katzenstein, MD; Julio S. G. Montaner, MD; Douglas D. Richman, MD; Michael S. Saag, MD; Mauro Schechter, MD, PhD; Robert T. Schooley, MD; Melanie A. Thompson, MD; Stefano Vella, MD; Paul A. Volberding, MD

Objective  New information warrants updated recommendations for the 4 central issues in antiretroviral therapy: when to start, what drugs to start with, when to change, and what to change to. These updated recommendations are intended to guide practicing physicians actively involved in human immunodeficiency virus (HIV)– and acquired immunodeficiency syndrome (AIDS)–related care.

Participants  In 1995, physicians with specific expertise in HIV-related basic science and clinical research, antiretroviral therapy, and HIV patient care were invited by the International AIDS Society-USA to serve on a volunteer panel. In 1999, others were invited to broaden international representation. The 17-member panel met regularly in closed meetings between its last report in 2000 and April 2002 to review current data. The effort was sponsored and funded by the International AIDS Society-USA, a not-for-profit physician education organization.

Evidence and Consensus Process  The full panel was convened in late 2000 and assigned 7 section committees. A section writer and 3 to 5 section committee members (each panel member served on numerous sections) identified relevant evidence and prepared draft recommendations. Basic science, clinical research, and epidemiologic data from the published literature and abstracts from recent (within 2 years) scientific conferences were considered by strength of evidence. Extrapolations from basic science data and expert opinion of the panel members were included as evidence. Draft sections were combined and circulated to the entire panel and discussed in a series of full-panel conference calls until consensus was reached. Final recommendations represent full consensus agreement of the panel.

Conclusions  Because of increased awareness of the activity and toxicity of current drugs, the threshold for initiation of therapy has shifted to a later time in the course of HIV disease. However, the optimal time to initiate therapy remains imprecisely defined. Availability of new drugs has broadened options for therapy initiation and management of treatment failure, which remains a difficult challenge.

JAMA. 2002;288:222-235

JST20002

Progress in antiretroviral therapy has resulted in achievements as well as new challenges.1 The partial restoration of CD4 and CD8 T cell number and function during suppression of human immunodeficiency virus 1 (HIV-1) replication with potent antiretroviral therapy has resulted in dramatic reductions in morbidity, mortality, and health care utilization.2-4 However, the toxicity of many current regimens, suboptimal activity and tolerability, and the emergence of drug resistance all point to the need for treatment strategies to address these challenges. These strategies include both new antiviral drugs and approaches to enhance host cellular immune control of HIV replication. This evolution in the field prompts the need for a reevaluation of current treatment guidelines, particularly regarding when to initiate therapy. The changing threshold for initiating therapy that has been emerging1, 5 is the result of recognition of limitations of currently available agents and is not necessarily a reflection of a major change in our understanding of disease pathogenesis, nor an indication that more aggressive treatment approaches should not be pursued. This is a constantly evolving field and HIV/AIDS (acquired immunodeficiency syndrome) practitioners will need to keep pace with current knowledge.


 

METHODS

 

The International AIDS Society-USA volunteer antiretroviral panel was convened in 1995 to develop treatment recommendations. The panel's goal was to develop current recommendations for use of antiretroviral therapy in the developed world. The panel included physicians with expertise in HIV-associated basic science, clinical research, and patient care. It was expanded in 1999 to broaden its international representation.

The full panel convened in late 2000 to review new data affecting its previous recommendations1 and assign committees for 7 sections: rationale, when to initiate therapy, initial regimens, treatment interruptions, monitoring therapy, changing therapy, and adjuvant therapy. A section writer and 3 to 5 section contributors were appointed (each panel member served on numerous sections) for each section. Each section committee met to identify relevant data and prepare draft recommendations for the sections, which were reviewed and discussed by the full panel. Data from the published literature and abstracts from recent scientific conferences were considered. Evidence strengths were considered according to parameters such as type of study (eg, randomized prospective trial, cohort data, case reports), number of subjects, duration of follow-up, and publication source. For example, published prospective studies with more than 20 patients and more than 48 weeks of follow-up were given high priority. Evidence from abstracts from scientific meetings that had not been published within 2 years of presentation were generally excluded. Extrapolations from basic science data and expert opinion of panel members were included as evidence. Draft sections with supporting data and preliminary recommendations were combined and circulated to the entire panel and discussed in a series of full-panel conference calls. The panel chair and vice-chair obtained group consensus on recommendations and they or section writers revised the report as necessary after each panel meeting or conference call. Recommendations herein are made by full-panel consensus agreement.


 

WHEN TO INITIATE ANTIRETROVIRAL THERAPY

 

Rationale for Treatment in Established HIV Infection
 
Highly active antiretroviral therapy (HAART) is usually effective in rapidly reducing plasma HIV RNA levels (ie, viral load) in antiretroviral-naive patients, accompanied by a gradual increase in CD4 cell counts, sometimes to normal levels.6 The number of memory CD4 cells increases early after effective treatment, as a result of redistribution from lymphoid tissues to the circulation.7 Naive CD4 cells, which are essential for responses to new antigenic challenges, are restored gradually with ongoing effective suppression of viral replication.8 For many antiretroviral-naive patients, CD4 cell counts increase to levels at which the patients are no longer generally susceptible to serious opportunistic infections.3 Because currently available antiretroviral regimens will not eradicate HIV,9, 10 the goal of therapy is to durably inhibit viral replication so that the patient can attain and maintain an effective immune response to most potential microbial pathogens.11

Considerations in Initiating Therapy in Asymptomatic Infection
 
Recent cohort data have provided support for the CD4 cell count being the major determinant of initiating therapy.12, 13 These studies have shown an increased mortality when antiretroviral therapy is initiated in patients with CD4 cell counts below 200/µL compared with initiation at higher levels. It is clear, therefore, that antiretroviral therapy should not be delayed until the patient is at high risk for serious opportunistic diseases (ie, at a CD4 cell count 200/µL).12-15

The CD4 cell level above 200/µL at which to initiate therapy remains unclear. Some serious illnesses, especially active tuberculosis and bacteremic pneumonia, may occur when the CD4 cell count is above 200/µL.16-20 In addition, the immune reconstitution syndrome and its associated morbidity may be observed in some patients starting antiretroviral therapy at low CD4 cell counts.21 Furthermore, some laboratory markers show lower rates of favorable responses when antiretroviral therapy is delayed until the 200 cells/µL threshold is reached. These include less rapid increase in CD4 cell count and potentially decreased ability to reduce viral load to below the limit of detection.12, 15, 22-25 Finally, the genetic complexity of HIV in persons increases with time, and this may facilitate escape from host immune defenses.26 These considerations support use of a CD4 cell count threshold higher than 200/µL. However, there are no definitive data with clinical end points that define at which level above 200 CD4 cells/µL antiretroviral therapy is best started. The available data from cohort studies, with one exception,27 have not been able to define a CD4 stratum above 200 cells/µL at which patients benefit from initiation of therapy.12, 13 Inherent biases that occur with cohort studies, which are observational and not randomized, and relatively limited follow-up in the studies reported to date (2 to 3 years), mean that conclusions from the analyses must be interpreted with caution. However, these are the best data available, there is general consistency across most studies, and it is questionable whether a randomized trial to study the issue of when to start therapy will ever be feasible.

In persons with CD4 cell counts above 350/µL, risk of 3-year clinical progression is low28 and additional concerns about impact of antiretroviral regimens on quality of life, risk of serious adverse drug effects, and limitations on future treatment options generally outweigh the benefits of durable viral suppression. However, it should be noted that roughly a third or more of persons have no treatment-limiting adverse effects for at least 3 years after initiation of treatment, leaving an option to physicians and patients to initiate therapy at higher CD4 cell counts.29 For persons who have already initiated therapy at higher CD4 cell count thresholds (eg, 400, 450, or 500 cells/µL) and have had durable HIV RNA suppression and no adverse effects over periods of months to years, it is not clear whether it is safe to discontinue therapy. Physicians and patients must thoroughly weigh risks and benefits of starting antiretroviral therapy for CD4 cell counts in the 200/µL to 350/µL range and above, and make individualized informed decisions (Table 1).5, 30, 31 The strength of the recommendation should depend on the immunologic status, as well as the patient's understanding of and commitment to an often complex regimen.

Although available data supporting the use of a specific viral load threshold as an independent indicator for initiating therapy are scarce,31 patients with CD4 cell counts at any level who have a viral load above 50 000 to 100 000 copies/mL31 should be closely monitored, because the CD4 cell count decreases more rapidly in untreated persons with higher viral loads.28, 31 Initiation of therapy may be considered in individuals with a viral load above 50 000 to 100 000 copies/mL,31 or a rapidly declining CD4 cell count even if it is above 350/µL. Some observational studies indicate that viral load early in the course of HIV disease is lower in women than in men, but there are no documented sex differences in the relation of CD4 cell count to risk of opportunistic infections.32, 33 Thus, treatment recommendations are the same for women as for men.

Therapy continues to be recommended in all patients with symptomatic established HIV infection. Immediate treatment, but not prophylaxis, of a serious opportunistic infection in patients with advanced HIV disease may take precedence over starting antiretroviral therapy. If potential for adverse drug-drug interactions exists (eg, protease inhibitors [PIs] and rifampin in treatment of Mycobacterium tuberculosis infection), it is wise to choose drugs with minimal or no interactions, or to delay antiretroviral treatment for a few weeks, until drugs causing the interactions can be discontinued. Clinicians should be aware of risk of immune reconstitution illness associated with initial increase in CD4 cell counts in patients starting antiretroviral therapy when there is a confirmed or suspected opportunistic infection.21


 

ESTABLISHED INFECTION: INITIAL THERAPY

 

Choice of Initial Therapy
 
No drug combination can be defined as the optimal initial regimen in all patients. Therapy should thus be individualized using a number of criteria, including efficacy and durability of antiretroviral activity, tolerability and adverse effects (Table 2),34-84 convenience of the regimen, drug-drug interactions, and potential salvageability of initial regimen.

The differences in "clinical activity" that are observed in clinical trials between regimens that contain 2 nucleoside reverse transcriptase inhibitors (NRTIs) with either a nonnucleoside reverse transcriptase inhibitor (NNRTI) or a single (or boosted) PI are often too small to differentiate relative efficacy. The durability of the first regimen is primarily related to issues of adherence, tolerability, and convenience, and baseline virological or immunologic status. Drugs with long half-lives and those for which regular timing of food or hydration is less crucial are likely to have an advantage. Daily adverse effects, even if minor, may reduce adherence. Finally, concerns for long-term toxic effects may be an important cause of patient reluctance to take certain medications. Regimens that involve drugs taken twice a day are likely to be associated with better adherence than those involving drugs taken 3 or more times a day.85 Once-daily regimens may further facilitate adherence and permit directly observed therapy in individual circumstances.

Many patients will ultimately experience at least one treatment failure. Because the initial regimen affects choices available for subsequent regimens, consideration should be given to such issues as overlapping toxicities (eg, with PIs in the case of lipid abnormalities or with some NRTIs in the case of neuropathy) and intraclass cross-resistance (see "First or Second Failure" section). There are currently no data on preferred sequencing of NRTIs. Stavudine and didanosine in combination should be avoided or used with caution in pregnant women because of increased risks of lactic acidosis.86

There are generally 3 types of initial combination regimens that should be considered: (1) a PI (with or without low-dose ritonavir) with 2 NRTIs; (2) an NNRTI with 2 NRTIs; or (3) 3 NRTIs. Other regimen combinations include a PI (with or without low-dose ritonavir) with an NNRTI plus 1 or 2 NRTIs, which should be reserved for special circumstances; and a PI (with low-dose ritonavir) with an NNRTI (see below).

Protease Inhibitor–Based Regimens
Data from randomized controlled trials with clinical outcomes attest to the effectiveness of PIs in combination with NRTIs.87 However, some regimens containing single PIs are often difficult to adhere to because of 3-times-a-day regimens or food constraints (eg, indinavir), or large pill burdens (eg, nelfinavir and amprenavir). Nelfinavir can be taken twice a day, but it is absorbed best when administered with a fatty meal.88

Data on benefits of PIs, in combination with low-dose ritonavir to provide a pharmacokinetic boosting effect, are appearing in studies in treatment-naive patients.89 Low-dose ritonavir can pharmacologically enhance (or boost) saquinavir, indinavir, amprenavir, or lopinavir (the latter available only in a coformulated capsule).90 Ritonavir inhibits enzymes of the cytochrome P450 system; it may act early on absorption and first-pass metabolism, increasing peak plasma concentrations with a co-administered PI (eg, with indinavir or amprenavir); or it may inhibit subsequent metabolism and extend the half-life of the second PI with an increase in trough level of drug (eg, with lopinavir or saquinavir). Such considerations may be important for drugs for which toxicity is related to peak concentrations and risks of virological failure to low trough levels. Advantages of such regimens include once- or twice-daily dosing and minimization of specific food requirements. Nelfinavir is not sufficiently enhanced by low-dose ritonavir to justify this combination.91

There are no comparative studies to determine which boosted PI regimen has the highest level of activity. Also, optimal dose of the PI or ritonavir in such a combination is often not well defined. Higher doses of ritonavir (eg, >100 mg twice a day) in the combination may result in a higher incidence of gastrointestinal adverse effects and lipid abnormalities.92 Little is known about optimal alternative regimens when initial boosted PIs fail.

Specific Ritonavir-Enhanced Protease Inhibitor–Containing Regimens
Lopinavir/ritonavir is an important new addition to the list of approved agents. In one study (48-week analysis), lopinavir/ritonavir plus 2 NRTIs were superior to nelfinavir plus 2 NRTIs in reducing viral load to below 50 copies/mL.93 Grade 3 or 4 elevations in triglyceride levels are significantly more common with lopinavir/ritonavir than with nelfinavir, and high frequencies of grades 3 and 4 cholesterol and triglyceride abnormalities (up to 30%) have been seen when lopinavir/ritonavir is combined with NRTIs and NNRTIs in salvage regimens.94 There have been few virological failures of lopinavir/ritonavir-containing regimens in studies of treatment-naive patients. Thus, there are few data on the patterns of resistance associated with this drug when used in an initial regimen or to guide recommendations on the preferred alternative regimens after lopinavir/ritonavir has failed. Early failures are associated predominantly with the lamivudine-associated M184V mutation when this agent is part of a lopinavir/ritonavir–containing regimen,95 an observation similar to what has been reported with indinavir and amprenavir.96-98

Indinavir/ritonavir (800 mg/100 mg, respectively, twice a day) may be widely used, but it is unclear whether this is the optimal dose combination, or if, for example, 800 mg/200 mg or 400 mg/400 mg is preferable. Incidence of renal adverse effects including stones, flank pain, and hematuria was higher (up to 20% at 24 weeks) in patients who had been treated previously with indinavir 800 mg 3 times a day and were then switched to indinavir/ritonavir.99

Saquinavir 800 or 1000 mg plus ritonavir 100 mg twice a day may be often used because of the adverse effects that are associated with a higher dose of ritonavir (200 mg twice a day), but there is little published comparative experience with this regimen. The pill burden is high if the soft gel saquinavir formulation is used. Ritonavir/saquinavir 400 mg/400 mg, twice a day, has also been used100; in this regimen, ritonavir provides antiretroviral activity as well as pharmacoenhancement. Whether this increases the likelihood of reducing viral loads to below the limit of detection remains unclear. The 24-week, intent-to-treat analysis of a once-daily regimen containing saquinavir (1600 mg, soft gel formulation) plus ritonavir (100 mg) showed that it was virologically inferior to an efavirenz-containing regimen, but the saquinavir/ritonavir group had a higher rate of gastrointestinal adverse effects.101

Combining amprenavir with ritonavir increases trough levels of amprenavir102 and reduces amprenavir pill burden from 16 to 8 pills a day. Given the tolerance profile and high pill burden of amprenavir, and effectiveness of amprenavir-containing combinations after prior PI regimens have failed, use of this combination may be preferred in the treatment-experienced setting.

Nonnucleoside Reverse Transcriptase Inhibitor–Based Regimens
A triple-drug regimen that includes an NNRTI and 2 NRTIs is one of the preferred combinations for initial therapy of established infection, given the effectiveness and the PI-sparing aspect. Three NNRTIs are currently approved in the United States: nevirapine, delavirdine, and efavirenz. No major trials have yet directly compared these drugs. Delavirdine is less often used because of its high pill burden and 3-times-a-day dosing, but it is the only currently available NNRTI that inhibits cytochrome P450, allowing a reduction in the dose of associated PIs.103, 104

Nevirapine has been studied in the ATLANTIC trial, which compared an NNRTI-based regimen with a PI-based regimen and a triple NRTI-based regimen.105 Although not powered to provide definitive conclusions, the results suggested that activity of nevirapine in a multidrug regimen was similar to that of indinavir. Comparability in viral suppression at 36 weeks between nevirapine-containing and nelfinavir-containing regimens (each with 2 NRTIs) has been reported.106 Nevirapine has been associated with severe rashes more often than has efavirenz.107, 108 Use of corticosteroids has not had a beneficial effect.109 Hepatotoxicity, including elevation of transaminases, jaundice, and rare cases of severe clinical hepatitis, occurs with nevirapine more often than with efavirenz.107, 108 Recently, nevirapine has been shown to be associated with favorable changes in lipid profiles, which include decreases in cholesterol and triglyceride levels and increases in HDL levels in patients who discontinued PIs.110

In an open-label study, zidovudine/lamivudine/efavirenz produced a marker response equivalent or superior to that seen with a similar NRTI backbone given with indinavir for up to 96 weeks of follow-up.111 Efavirenz is commonly prescribed because of its once-daily dosing and safety profile.108 Because of teratogenicity in animals,108 efavirenz should be avoided in women who are (or intend to become) pregnant. Hyperexcitability, vivid dreams, nightmares, and hallucinations are associated with efavirenz.108 Mood disturbances and personality changes may be persistent in some patients.108

Triple-NRTI Regimens
Triple-NRTI regimens have become a viable alternative as initial therapy. The largest body of evidence at present is for regimens that contain abacavir. An abacavir/zidovudine/lamivudine combination produced a decrease in viral load (to <400 copies/mL) at 48 weeks equivalent to that produced by zidovudine/lamivudine/indinavir in a placebo-controlled trial.112, 113 However, more complete viral suppression (assessed with an assay with a limit of detection of 50 copies/mL) was less likely with the 3-NRTI regimen in those patients with a pretreatment viral load above 100 000 copies/mL.112, 113 Thus, these regimens are not routinely recommended as initial treatment for patients with high viral loads (eg, >100 000 copies/mL) or with low CD4 cell counts until more data are available. The twice-daily regimen, low pill burden, few daily toxic effects, and lack of undesirable pharmacokinetic interactions are potential advantages of zidovudine/lamivudine/abacavir regimens. Availability of these 3 drugs in a fixed-dose combination is also likely to facilitate adherence. Severe hypersensitivity reactions to abacavir occur in about 3% of patients,114, 115 and is an important consideration.

A virus mutation at codon 184 substantially reduces sensitivity to lamivudine and somewhat (2- to 4-fold) to abacavir. This mutation is often detected during virological failure with abacavir-containing regimens.116, 117 In most such patients,117 viral mutations associated with resistance to zidovudine are not detected, and in theory other NRTIs would be effective in subsequent regimens. However, continuing treatment with this 3-drug combination, despite the presence of detectable HIV, may be accompanied by development of other NRTI-associated mutations.

Protease Inhibitor/NNRTI–Based Regimens, With or Without NRTI
The combination of a PI with an NNRTI and an NRTI (ie, drugs from all 3 classes), is not routinely recommended because the risks of multiclass drug resistance (thus limiting future treatment options) and toxicity may be high. However, in the setting of a patient with advanced disease and a high near-term mortality risk who has an opportunistic infection for which no effective therapy exists (eg, cryptosporidiosis), a 3-class regimen may be considered to assure a high degree of activity and to try to achieve rapid immunologic restoration. Simplification of such a regimen can be considered subsequently. The second setting in which to consider a 3-class regimen is when the patient is infected with a drug-resistant strain whose in vitro resistance testing profile suggests that such a regimen may be effective.

Protease inhibitor (enhanced with low-dose ritonavir) and NNRTI regimens without an NRTI component may be of interest for certain settings; however, insufficient data exist on when to recommend such NRTI-sparing regimens as initial therapy.


 

TREATMENT INTERRUPTIONS

 

Structured, supervised, or strategic treatment interruptions (STIs) have been considered in very different situations: following early therapy of acute HIV syndrome,118 following suppressive antiretroviral therapy of established infection,119-121 and for facilitating salvage after therapy has failed.122 The rationales for interruption of therapy in these 3 settings are different, and the subject has been extensively reviewed elsewhere.123, 124 Given the paucity of available controlled studies and the potential risks, STIs cannot be recommended for current clinical practice, and should preferably be attempted in the context of cohort studies or clinical trials.


 

MONITORING ANTIRETROVIRAL THERAPY

 

Adherence: Assessment and Reinforcement
 
Incomplete adherence to one or more prescribed medications is a key cause of virological failure of antiretroviral regimens.125 A recent study found a doubling of the viral load for every 10% decrement from complete adherence.126 In another study, the viral load response was greater in inmates in a prison where treatment was directly observed than in patients treated with comparable regimens in an outpatient research clinic (85%-100% vs 50%-80%).127, 128 Factors that limit full adherence are complex and incompletely defined but may include high pill number and large pill size, medication schedule and dietary restrictions, toxic adverse effects, and ineffective education and support of patients regarding adherence. Progress in developing new drug formulations (eg, didanosine without the buffer) and fixed-dose combinations (eg, lamivudine/zidovudine/abacavir, lamivudine/zidovudine, and lopinavir/ritonavir) that can simplify regimens is encouraging.

Ways of improving adherence are being investigated.129 Most remain largely empiric and not evidence-based.129 Effective communication between patient and provider is essential both before and after treatment has begun. Some health care centers may use nonphysicians (pharmacists, nurses, peer educators, and others) to effectively assess and support adherence, but the physician should also be actively involved. Once treatment has begun, weekly contact may be appropriate until the patient has established a consistent daily routine of medication use and has passed the time that any short-term adverse effects would be expected. Reinforcing the need for adherence at every health care provider contact is important.

CD4 Cell Counts and Viral Load
 
In the asymptomatic individual, antiretroviral activity is evaluated by assessing changes in CD4 cell count and viral load. A decrease in viral load (indicating a reduction in virus replication) and increase in CD4 cell count (indicating an improvement in immune competence) in response to antiretroviral drugs are both associated with clinical efficacy.130-132

The CD4 cell count typically increases by more than 50 cells/µL at 4 to 8 weeks after antiretroviral therapy has been started or changed, followed by an additional increase of 50 to 100 cells/µL per year thereafter.133, 134 Once CD4 cell counts exceed 200/µL for 3 to 6 months, there are reduced risks of many opportunistic infections and prophylaxis against certain pathogens can often be stopped.17

Current HIV RNA tests have reliable detection limits of about 50 copies/mL, and quantification using commercial tests is reliable at 200 copies/mL.5 With repeated measurements, differences of 0.2 to 0.3 log10 (30% to 50%) are considered significant evidence of a change in viral load.135 Virologically effective therapy generally reduces viral load by more than 90% (ie, a 1-log10, or 10-fold, reduction) within 8 weeks of treatment.136, 137 Failure to attain a 90% reduction by 4 weeks of therapy suggests poor adherence, viral resistance, or inadequate drug exposure.

On initiation of therapy, sequential measurements of CD4 cell count and viral load at 4, 8 to 12, and 16 to 24 weeks have been used to assess the early response to antiretroviral therapy.138-140 A continued decline in viral load at each measurement, together with an increase in CD4 cell count, indicates a drug combination is appropriately active in vivo, and indirectly indicates patient adherence and baseline viral susceptibility. In some instances CD4 cell counts might not increase, or may decrease, with successful suppression of viral replication (HIV RNA, <50 copies/mL).141 Reasons for this phenomenon are not well understood. Drug-related toxic effects and ongoing opportunistic disease should be investigated. Although no specific intervention has been studied well enough to be recommended at this time, it is important to monitor viral load and CD4 cell counts more frequently in patients with such discordant responses.

Once virological suppression has been achieved (2 sequential measurements below the limit of detection of the most sensitive assay available), viral load and CD4 cell numbers are usually monitored every 8 to 12 weeks.142 More frequent monitoring may be appropriate in case of intercurrent illness, change in antiretroviral therapy, introduction of a new treatment that could interfere pharmacologically with antiretroviral drugs, or if adherence to therapy becomes questionable. Frequent monitoring may show, even in patients with drug-sensitive virus who report optimal adherence, occasional small increases in detectable plasma virus (blips) in the range of 50 to 400 copies/mL. There is no evidence to date that such isolated blips are predictive of subsequent overt treatment failure.143

Drug Resistance Testing
 
Guidelines for use of drug resistance testing in clinical practice have been published.144-146 Randomized, prospective trials have demonstrated variable results with respect to short-term virological benefit of genotype or phenotype testing.147-153 Results of studies showing no difference between study groups may have been affected by the interpretation of the resistance information, particularly an under-recognition of the degree of NRTI cross-resistance, and lack of treatment options for patients in whom several regimens have failed. Despite the limitations of these trial results, the clinical value of drug resistance testing is recognized and it is now considered standard-of-care in the management of treatment failure.145 Data are not yet available on which method or type of resistance testing is superior in any given clinical setting. Resistance testing information can provide guidance when selecting which drugs to exclude or include in a new regimen. Testing should be performed when the selective pressure of the failing regimen is still present because resistance may not be detected following withdrawal of the drugs.122 However, resistance mutations archived at the cell level may persist, with potential to rapidly reemerge if the failing regimen is reintroduced. In treatment-naive persons, drug resistance testing should be strongly considered in those who may have been infected with a resistant viral strain, particularly those with more recent infection, or when the initial response is suboptimal in the face of excellent drug adherence.154

Drug Concentration Monitoring
 
Adjusting doses of a drug to maintain a desired plasma level is common with some drugs and has been suggested for antiretroviral drugs.155 This may be more practical with some classes of antiretroviral drugs than others. The NRTIs are especially problematic, because they require intracellular activation and because intracellular concentrations are more difficult to measure than plasma levels. Additionally, drug concentration data are most meaningful in the context of the phenotypic susceptibility of the patient's viral strain, which can be determined only in the setting of detectable viral loads.

The drug trough concentration ideally should not fall below the level necessary to control HIV replication, and the peak concentration should be below the range at which toxic effects would be expected. Composite data on the Cmin, Cmax, and area under the concentration curve (AUC) are generally known and available in the prescribing information for most available drugs, as is the drug concentration necessary in vitro to suppress the replication of wild-type HIV. However, there is substantial interpatient and intrapatient variability in pharmacokinetics, and suppression may be impossible if resistance has emerged. Adding yet more complexity are the different methods of determining suppression thresholds (eg, using IC90 or IC50) and the effect of protein binding and how this is accounted for in the data reported. Data on the effect of Cmax, AUC, or the shape of the drug-decay curve on drug toxicity or effect may be even less firm.

If monitoring of drug levels is considered, practical issues must be addressed, including adherence problems and assay availability, reliability, and cost. Possible causes of variations in drug levels must be considered, including drug-drug interactions with prescription, over-the-counter, alternative, or recreational drugs, and malabsorption due to coincident gastrointestinal disease or food effects. Finally, drug steady-state should have been reached before concentrations are measured.

Drug-concentration monitoring may be of particular value in cases of treatment failure (eg, initial or subsequent therapy), or when salvage therapy with a ritonavir-enhanced PI–based regimen has been initiated, especially if other drugs with known pharmacological interactions, such as efavirenz, are also prescribed (drug resistance testing should also be considered).

Drug concentration monitoring is commercially available in some areas. However, it is difficult to make clear recommendations, because the therapeutic range of plasma concentrations is not known for most drugs. Moreover, few studies have demonstrated the clinical usefulness of drug-concentration monitoring.156 Thus, decisions about adjusting doses of antiretroviral drugs based on level determinations need to be individualized until more data are available.


 

ESTABLISHED INFECTION: CHANGING THERAPY

 

Changing Drugs Because of Incomplete Adherence, Adverse Effects, or Intolerance
 
In the absence of virological or immunologic failure, a regimen may pose problems with adherence, intolerance, or cumulative (long-term) toxic effects. As long as the antiviral activity of the overall regimen is maintained, exchanging individual components of the regimen is acceptable. Examples of such substitutions are a change of stavudine for zidovudine or nevirapine for efavirenz, or substitution of low-dose ritonavir-boosted PIs for single PI components of a regimen.

Substituting individual antiretroviral agents is frequently indicated because of drug-specific toxic effects. In clinical settings in which the offending agent cannot be easily determined, or in which there are significant safety concerns, treatment should be completely interrupted so that acute adverse effects can resolve while alternative regimens are considered. Among patients with serum lipid abnormalities and lipodystrophy, abacavir or nevirapine can be substituted for the PI component.53 Serum lipid abnormalities improve with these changes but improvements in body composition abnormalities have not been consistently documented in studies reported to date.53 More studies and longer patient follow-up are needed. When a change in a drug class is planned, it is preferable to do so in the setting of successful virological suppression.

Changing Therapy Because of Treatment Failure
 
The definition of "treatment failure" (a term that subsumes virological, immunologic, or clinical failure) depends on the clinical setting and mirrors the objective of ongoing therapy at a given time in the patient's treatment course. The duration of response is predicted by level of viral suppression obtained while receiving therapy within 8 weeks.138, 157-159 In the case of the first or second regimen, when virus is wild type or harbors few resistance mutations, maintaining an undetectable viral load is an achievable goal of therapy; in this setting, treatment failure is best defined as inability to achieve a viral load below assay detection limits (eg, <50 copies/mL) or as any sustained return of the viral load to above the target value (eg, >400 copies/mL). With increasing rounds of treatment failure, the level and spectrum of virus resistance may increase, and it may become more difficult to construct an active combination. In patients for whom several regimens have failed, the virus may become multiply resistant, with fewer than 3 active drugs being available, and the objective of achieving stable undetectable viral load with conventional regimens may be unrealistic. Problems with toxicity may further restrict the number of available drugs. The goal of therapy in this setting is primarily to prevent clinical progression, and failure can be defined as a deterioration of the CD4 cell count or the occurrence of a serious opportunistic infection.

First or Second Failure
Treatment failure occurs within the first year of therapy in a substantial proportion of treatment-naive patients.160, 161 A lower, but definite, rate of treatment failure in successive years has been reported with nearly all regimens.14, 111 Thus, failure should be anticipated as part of the long-term strategy of antiretroviral treatment. In the case of confirmed treatment failure, an intervention should be considered promptly in order to minimize emergence of cumulative drug resistance and cross-resistance that will limit availability of alternative options.162 However, treatment failure is not always associated with viral resistance, particularly during initial viral rebound or if virus remains detectable at a low level after several months of therapy.96 In such circumstances a thorough assessment of adherence, dietary requirements, drug-drug interactions, and bioavailability is the first step. In situations where there is no evidence of resistance or adherence issues, adding a new drug or drugs (ie, regimen intensification) may be considered. However, since intensification may add to the complexity of the treatment regimen, such an approach could aggravate problems with adherence. Some drugs, particularly the NNRTIs and lamivudine, generally should not be used alone as intensification agents because of risk of single-step, high-level resistance. Intensifying with ritonavir as a pharmacological enhancer of another PI may be effective in the short term in patients for whom a regimen with a single PI is failing.152 Addition of abacavir or tenofovir disoproxil fumarate may also be of use in this setting.163

When a decision is made to change therapy because of sustained virological failure, the new regimen should be one with highest likely effectiveness, as predicted by the patient's complete drug history and the resistance test result, as well as highest likelihood of tolerability and adherence. New regimens should contain at least 2, and if possible 3, drugs deemed to be active. Virus that replicates during treatment failure may not be resistant to all of the drugs in the failing regimen.96, 152 However, latently infected lymphocytes may harbor archived virus resistant to drugs used in the past but not detected by routine resistance testing. Shared-resistance mutations conferred by an individual drug may lead to cross-resistance among drugs in the same class, complicating the choice of alternative regimens.146, 164 With currently approved NNRTIs, risk of complete NNRTI-class cross-resistance is high when an NNRTI-containing regimen fails. With PIs, intraclass cross-resistance is not so predictable. Depending on the pattern of resistance, an alternative PI (or a combination of PIs) can often be selected. With NRTIs, extent of class cross-resistance is greater than anticipated previously, and level of resistance to some drugs (eg, stavudine) is more difficult to infer from genotype or phenotype.145 In rare circumstances, multidrug resistance to NRTIs may develop through a unique pathway of resistance (eg, the Q151M complex or a 2–amino acid insertion at codon 69).145 Examples of possible alternative regimens (based on predicted resistance patterns) in the setting of first regimen failure are given in Table 3.165

Tenofovir was recently approved for treatment of HIV infection. In its active diphosphate form, the drug has a prolonged intracellular half-life, which permits once-daily dosing.166 The drug is active against isolates containing certain NRTI-associated resistance mutations, including the Q151M complex, but has diminished activity against isolates with the T69S insertion, the K65R mutation, and those with multiple NRTI-associated mutations (NAMs), particularly M41L and L210W.166, 167 The lamivudine-associated M184V mutation enhances tenofovir's activity in vitro.167 The signature mutation associated with this drug is K65R, but in clinical trials and animal studies, this mutation arises infrequently and is not consistently associated with loss of antiretroviral activity.168 In phase 3 trials in antiretroviral-experienced patients with virological failure, tenofovir has produced a consistent 0.6 log10 decrease from baseline in viral load and modest rises in CD4 cell counts over 24 and 48 weeks or longer.169, 170 Among treatment-naive patients treated with tenofovir monotherapy for 21 days, a 1.5 log10 reduction in HIV RNA was observed.171 The drug is generally well tolerated.169, 170 No serious renal dysfunction or bone density alterations have been reported to date. The available clinical trial results suggest a role for tenofovir in management of treatment-experienced patients. Results from ongoing studies will help define the role of tenofovir in management of drug-naive patients.

Multiple Failures
Treatment histories and results of resistance testing may indicate that 2 or fewer active drugs are available for therapy in patients in whom numerous regimens have failed.

Evidence of low rates of clinical progression in such patients, who continue antiretroviral therapy despite viral replication and presence of resistance, argues for continuing treatment in the face of virological failure.172 Some virus with multidrug-resistance mutations has reduced replicative capacity, or fitness, relative to wild-type virus.122, 173, 174 Stabilization of CD4 cell numbers and absence of clinical progression have been demonstrated when viral load is sustained at a level 3-fold (0.5 log10) below the patient's natural set point (pretreatment value).172, 175 Conversely, when all drugs in a regimen are stopped there may be an apparent reversion to wild-type virus within 8 to 12 weeks, associated with a rapid increase in viral load and marked decrease in CD4 cell count.122, 176 Although there is a risk of accumulating additional resistance mutations, continuing drug regimens that maintain selective pressure on the virus is preferable to discontinuing all antiretroviral therapy, especially in settings in which the CD4 cell counts are maintained despite a rebound in viral replication.

In cases in which a change in the therapy cannot be delayed, adding 1 new drug (eg, a drug available through expanded access) may not result in a profound and durable effect, and the best therapy should be selected, based on treatment history, tolerance, and resistance testing. The regimen may include drugs recycled from previous, failed regimens. Multidrug rescue therapy (6 or more drugs, also called "megaHAART") in this setting may exhibit a substantial degree of antiretroviral activity.177, 178 However, this approach may lead to toxic effects and problems with adherence, adverse drug-drug interactions, and increased cost. Moreover, the effect on CD4 cell count is not entirely predictable.179


 

ADJUVANT THERAPY TO ANTIRETROVIRAL DRUGS

 

The concept of manipulating the immune response for host benefit has received increased emphasis. Approaches include attempts to augment (eg, interleukin 2180-184) or dampen (eg, cyclosporin A, corticosteroids, hydroxyurea, and mycophenolic acid185, 186) the immune response generally, and attempts designed to stimulate (treatment interruption and "therapeutic" vaccination187-189) relevant HIV-specific immune effector responses. At this point, however, insufficient clinical data exist to recommend these approaches outside the setting of clinical trials. In addition, in the case of hydroxyurea, significant toxicities have emerged that have dampened enthusiasm for this agent.190


 

SUMMARY

 

The future of antiretroviral therapy rests with the development of new drugs that will result in simpler, more effective, and less toxic regimens along with development of an improved understanding of innate immune system responses and novel approaches to exploit these responses. Several new agents are currently in development, derived from current drug classes (eg, the NRTIs amdoxovir and emtricitabine, the NNRTIs DPC-083 and TMC-125, and the PIs atazanavir and tipranavir) and new drug classes, including entry inhibitors (eg, enfuvirtide) and integrase inhibitors. Potential advantages of these drugs include once-daily dosing, smaller pill size, lower incidence of adverse effects, new viral targets, and activity against virus that is resistant to other drugs in the respective classes. The benefits of current and future agents will continue to be felt by HIV-infected persons in the developed world. Extending these benefits to those living with HIV in the developing world is a challenge that needs to be met.


 
 
Author/Article Information

 
Author Affiliations: Hôpital Bichat-Claude Bernard, X. Bichat Medical School, Paris, France (Dr Yeni); Columbia University College of Physicians and Surgeons, New York, NY (Dr Hammer); Brown University School of Medicine, Providence, RI (Dr Carpenter); University of New South Wales, Sydney, Australia (Dr Cooper); University of Miami School of Medicine, Miami, Fla (Dr Fischl); Hospital Clinic, University of Barcelona, Barcelona, Spain (Dr Gatell); Chelsea and Westminster Hospital, London, England (Dr Gazzard); Harvard Medical School, Boston, Mass (Dr Hirsch); The International AIDS Society-USA (Ms Jacobsen); Stanford University Medical Center, Stanford, Calif (Dr Katzenstein); University of British Columbia, Canada (Dr Montaner); University of California San Diego and San Diego VA Healthcare System (Dr Richman); The University of Alabama at Birmingham (Dr Saag); Universidade Federal do Rio de Janeiro, Brasil (Dr Schechter); University of Colorado School of Medicine, Denver (Dr Schooley); AIDS Research Consortium of Atlanta, Georgia (Dr Thompson); Istituto Superiore di Sanità, Rome, Italy (Dr Vella); University of California San Francisco and San Francisco Veterans Affairs Medical Center (Dr Volberding).
 
Corresponding Author: Patrick Yeni, MD, Hôpital Bichat-Claude Bernard, Department of Infectious Diseases, 46 Rue Henri-Huchard, Paris, Cedex 18 France 75877 (e-mail: [log in to unmask]
).
Reprints: Patrick Yeni, MD, International AIDS Society–USA, 1001 B O'Reilly Ave, San Francisco, CA 94129.

Financial Disclosure: Dr Yeni: grant support/advisor/speakers bureau (ANRS [French National Agency for AIDS Research], Boehringer Ingelheim, Bristol-Myers Squibb, Gilead, GlaxoSmithKline, Hoffmann-La Roche, Tibotec, Triangle); Dr Hammer: site investigator/consultant/grants (Boehringer Ingelheim, Bristol-Myers Squibb, Gilead, GlaxoSmithKline, NIH grants AI46386, AI48013, AI42848, Roche-Trimeris, Shionogi, Shire BioChem, Tibotec-Virco, Triangle); Dr Carpenter: principal investigator (National Institutes of Health and Centers for Disease Control and Prevention grants); Dr Cooper: grant support/honoraria/consultant (Abbott, Boehringer Ingelheim, Bristol-Myers Squibb, Chiron, Commonwealth Department of Health and Ageing [Canberra ACT Australia], Gilead, GlaxoSmithKline, Merck Sharp & Dohme, NIH, Pfizer, Roche); Dr Fischl: grant support/advisor (Abbott, Agouron, Bristol-Myers Squibb, DuPont, GlaxoSmithKline, Triangle, National Institutes of Health); Dr Gatell: grant support/site investigator/advisor/speakers bureau (Abbott, Boehringer Ingelheim, Bristol-Myers Squibb, DuPont, Gilead Sciences, GlaxoSmithKline, Merck Sharp & Dohme, Novirio, Pharmacia & Upjohn, Roche, Schering-Plough, Vertex, Visible Genetics), and uncompensated expert testimony for the European Union regarding merger of DuPont and Bristol-Myers Squibb; Dr Gazzard: grant support/advisor (Abbott, Bristol-Myers Squibb, DuPont, Gilead, Glaxo Wellcome, Pharmacia & Upjohn); Dr Hirsch: grant support/consultant/lecture sponsorship (Bristol-Myers Squibb, GlaxoSmithKline, Merck, NIH, Schering-Plough, Takeda, Trimeris); Dr Katzenstein: stock/research funding/honoraria/advisor/assay kits or reagents (Boehringer Ingelheim, Bristol-Myers Squibb, Gilead, GlaxoSmithKline, Merck, ViroLogic, Visible Genetics, Doris Duke Charitable Trust Distinguished Clinical Scientist Award, and obtained patent US 5,968,730 "PCR Assays for Monitoring Antiviral Therapy and Making Therapeutic Decisions in the Treatment of AIDS" issued October 15, 1999); Dr Montaner: grant support/ad hoc advisor/speaker at sponsored events (Abbott, Agouron, BioChem Pharma, Boehringer Ingelheim, Bristol-Myers Squibb, DuPont, Gilead, Glaxo Wellcome, Hoffmann-La Roche, Merck Frosst Laboratories, Pfizer, Pharmacia & Upjohn, Province of British Columbia, Canadian Institute for Health Research, Shire, and is a coapplicant on a patent for use of a PCR-based mitochondrial DNA assay); Dr Richman: consultant (Abbott, Bristol-Myers Squibb, Chiron, Department of Veterans Affairs, Gilead, GlaxoSmithKline, Merck, NIH, Novirio, Pfizer, Roche, Takeda, Triangle, ViroLogic); Dr Saag: consultant/speakers bureaus/grant support (Abbott, Agouron, Bristol-Myers Squibb, DuPont, Gilead, GlaxoSmithKline, Hoffman-La Roche, Janssen, Ortho Biotech, Pfizer, Pharmacia & Upjohn, Shire, Tibotec, Triangle, Trimeris, ViroLogic); Dr Schechter: grant support/honoraria/consultant (Abbott, Brazil National Research Council, Brazil Ministry of Health, Boehringer Ingelheim, Bristol-Myers Squibb, Gilead, GlaxoSmithKline, Merck, NIAID, Roche, World Health Organization); Dr Schooley: stock options/grant support/honoraria/consultant/lecture sponsorship (Agouron, AnorMed, Bristol-Myers Squibb, Gilead, GlaxoSmithKline, Merck, Mojave, NIH, Pan Pacific Pharmaceuticals, Roche, Tanox, Triangle Pharmaceuticals, Vertex Pharmaceuticals, ViroLogic); Dr Thompson: grant support/honoraria/consultant/lecture sponsorship/advisor (Abbott Laboratories, Agouron, Agouron/Pfizer, Boehringer Ingelheim, Bristol-Myers Squibb, Centers for Disease Control and Prevention Adult Spectrum of Disease Study, Chiron, DuPont Pharmaceuticals, Gilead Sciences, GlaxoSmithKline, Merck, NIAID Acute Infection and Early Disease Research Network, NIAID Community Programs for Clinical Research on AIDS, Oxo-Chemie, Roche, Serono, Triangle Pharmaceuticals, Trimeris, and ViroLogic); Dr Vella: lecture sponsorship for satellite meetings or continuing medical education programs (Abbott, Agouron, Boehringer Ingelheim, Bristol-Myers Squibb, GlaxoSmithKline, Merck, Roche); and Dr Volberding: honoraria/consultant (Abbott, Boehringer Ingelheim, Bristol-Myers Squibb, Gilead, GlaxoSmithKline, Merck, NIAID [Center for AIDS Research]).

Funding/Support: This work was sponsored by the International AIDS Society-USA (IAS-USA) and funded through a reserve fund independent of commercial companies, funded by private donations and interest income. Of note, the panel members were not compensated for this activity. The IAS-USA received grant support for continuing medical education programs and production of Topics in HIV Medicine from Abbott Laboratories, Agouron Pharmaceuticals, Bristol-Myers Squibb, Boehringer Ingelheim, Department of Health and Human Services/Health Resources and Services Administration [subcontract], DuPont, Gilead Sciences, GlaxoSmithKline, Merck US Human Health, Ortho Biotech, Roche Laboratories, Schering Corporation, Virco, ViroLogic, and Visible Genetics, Inc, as well as substantial gifts in kind from Microsoft Corporation, the Gill Foundation, and Philanthropy by Design. However, these funding sources did not support the project represented herein.

Acknowledgment: We thank Morris Schambelan, MD and Constance A. Benson, MD (Chairs of the IAS-USA Complications of Antiretroviral Therapy Expert Panel) for the review of the manuscript, and Michelle Tayag for administrative support in preparing the manuscript.



 

REFERENCES

 

1.
Carpenter CC, Cooper DA, Fischl MA, et al.
Antiretroviral therapy for HIV infection in adults.
JAMA.
2000;283:381-390.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

2.
Hogg RS, Heath KV, Yip B, et al.
Improved survival among HIV-infected individuals following initiation of antiretroviral therapy.
JAMA.
1998;279:450-454.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

3.
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.
MEDLINE

4.
Tashima KT, Hogan JW, Gardner LI, et al.
A longitudinal analysis of hospitalization and emergency department use among human immunodeficiency virus-infected women reporting protease inhibitor use.
Clin Infect Dis.
2001;33:2055-2060.
MEDLINE

5.
US Public Health Service.
Guidelines for the Use of Antiretroviral Agents in HIV-Infected Adults and Adolescents.
Updated February 4, 2002. Available at: http://www.hivatis.org/trtgdlns.html#Adult. Accessibility verified June 4, 2002.

6.
Rizzardi GP, de Boer RJ, Hoover S, et al.
Predicting the duration of antiviral treatment needed to suppress plasma HIV-1 RNA.
J Clin Invest.
2000;105:777-782.
MEDLINE

7.
Hockett RD, Kilby JM, Derdeyn CA, et al.
Constant mean viral copy number per infected cell in tissues regardless of high, low, or undetectable plasma HIV RNA.
J Exp Med.
1999;189:1545-1559.
MEDLINE

8.
Carcelain G, Li TS, Renaud M, et al.
Antiretroviral therapy and immune reconstitution.
J Soc Biol.
1999;193:5-10.
MEDLINE

9.
Zhang L, Ramratnam B, Tenner-Racz K, et al.
Quantifying residual HIV-1 replication in patients receiving combination antiretroviral therapy.
N Engl J Med.
1999;340:1605-1613.
MEDLINE

10.
Finzi D, Blankson J, Siliciano JD, et al.
Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy.
Nat Med.
1999;5:512-517.
MEDLINE

11.
Li TS, Tubiana R, Katlama C, et al.
Long-lasting recovery in CD4 T-cell function and viral-load reduction after highly active antiretroviral therapy in advanced HIV-1 disease.
Lancet.
1998;351:1682-1686.
MEDLINE

12.
Hogg RS, Yip B, Chan KJ, et al.
Rates of disease progression by baseline CD4 cell count and viral load after initiating triple-drug therapy.
JAMA.
2001;286:2568-2577.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

13.
Sterling TR, Chaisson RE, Bartlett JG, et al.
CD4+ lymphocyte level is better than HIV-1 plasma viral load in determining when to initiate HAART.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 519.

14.
Chen R, Westfall A, Cloud G, et al.
Long-term survival after initiation of antiretroviral therapy.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 341.

15.
Kaplan J, Hanson D, Karon J, et al.
Late initiation of antiretroviral therapy (at CD4+ lymphocyte count <200 cells/µL) is associated with increased risk of death.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 520.

16.
Carr A, Cooper DA.
Adverse effects of antiretroviral therapy.
Lancet.
2000;356:1423-1430.
MEDLINE

17.
USPHS/IDSA Prevention of Opportunistic Infections Working Group.
2001 USPHS/IDSA Guidelines for the Prevention of Opportunistic Infections in Persons Infected With Human Immunodeficiency Virus.
Updated November 28, 2001. Available at: http://www.hivatis.org/trtgdlns.html#opportunistic. Accessibility verified June 4, 2002.

18.
Chaisson RE, Schecter GF, Theuer CP, et al.
Tuberculosis in patients with the acquired immunodeficiency syndrome.
Am Rev Respir Dis.
1987;136:570-574.
MEDLINE

19.
Crowe SM, Carlin JB, Stewart KI, et al.
Predictive value of CD4 lymphocyte numbers for the development of opportunistic infections and malignancies in HIV-infected persons.
J Acquir Immune Defic Syndr.
1991;4:770-776.
MEDLINE

20.
Hirschtick RE, Glassroth J, Jordan MC, et al.
Bacterial pneumonia in persons infected with the human immunodeficiency virus.
N Engl J Med.
1995;333:845-851.
MEDLINE

21.
DeSimone JA, Pomerantz RJ, Babinchak TJ.
Inflammatory reactions in HIV-1-infected persons after initiation of highly active antiretroviral therapy.
Ann Intern Med.
2000;133:447-454.
MEDLINE

22.
Tortajada C, Garcia F, Plana M, et al.
Comparison of T-cell subsets' reconstitution after 12 months of highly active antiretroviral therapy initiated during early versus advanced states of HIV disease.
J Acquir Immune Defic Syndr.
2000;25:296-305.
MEDLINE

23.
Phillips AN, Staszewski S, Weber R, et al.
HIV viral load response to antiretroviral therapy according to the baseline CD4 cell count and viral load.
JAMA.
2001;286:2560-2567.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

24.
Kaufmann GR, Bloch M, Zaunders JJ, et al.
Long-term immunological response in HIV-1-infected subjects receiving potent antiretroviral therapy.
AIDS.
2000;14:959-969.
MEDLINE

25.
Chaisson RE, Keruly JC, Moore RD.
Association of initial CD4 cell count and viral load with response to highly active antiretroviral therapy.
JAMA.
2000;284:3128-3129.
FULL TEXT  |  PDF  |  MEDLINE

26.
Shankarappa R, Margolick JB, Gange SJ, et al.
Consistent viral evolutionary changes associated with the progression of human immunodeficiency virus type 1 infection.
J Virol.
1999;73:10489-10502.
MEDLINE

27.
Opravil M, Ledergerber B, Furrer H, et al.
Clinical benefit of early initiation of HAART in patients with asymptomatic HIV infection and CD4 counts >350/mm3.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract LB6.

28.
Mellors JW, Munoz AM, Giorgi JV, et al.
Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection.
Ann Intern Med.
1997;126:946-954.
MEDLINE

29.
Tarwater PM, Margolick JB, Jin J, et al.
Increase and plateau of CD4 T-cell counts in the 3(1/2) years after initiation of potent antiretroviral therapy.
J Acquir Immune Defic Syndr.
2001;27:168-175.
MEDLINE

30.
Lyles RH, Munoz A, Yamashita TE, et al.
Natural history of human immunodeficiency virus type 1 viremia after seroconversion and proximal to AIDS in a large cohort of homosexual men.
J Infect Dis.
2000;181:872-880.
MEDLINE

31.
Egger M.
Prognosis of HIV-1 infected drug naive patients starting potent antiretroviral therapy.
From: 41st Interscience Conference on Antimicrobial Agents and Chemotherapy; December 16-19, 2001; Chicago, Ill. Abstract LB-18.

32.
Farzadegan H, Hoover DR, Astemborski J, et al.
Sex differences in HIV-1 viral load and progression to AIDS.
Lancet.
1998;352:1510-1514.
MEDLINE

33.
Junghans C, Ledergerber B, Chan P, et al.
Sex differences in HIV-1 viral load and progression to AIDS.
Lancet.
1999;353:589-591.
MEDLINE

34.
Moyle GJ, Datta D, Mandalia S, et al.
Hyperlactataemia and lactic acidosis during antiretroviral therapy.
Antivir Ther.
2001;6(suppl 4):66.

35.
Coghlan ME, Sommadossi JP, Jhala NC, et al.
Symptomatic lactic acidosis in hospitalized antiretroviral-treated patients with human immunodeficiency virus infection.
Clin Infect Dis.
2001;33:1914-1921.
MEDLINE

36.
van Gemen B, De Ronde A, Casula M, et al.
Quantification of mitochondrial DNA relative to nuclear DNA in peripheral blood mononuclear cells.
From: 3rd International Workshop on Adverse Drug Reactions and Lipodystrophy in HIV; October 23-26, 2001; Athens, Greece. Abstract 19.

37.
Fortgang IS, Belitsos PC, Chaisson RE, et al.
Hepatomegaly and steatosis in HIV-infected patients receiving nucleoside analog antiretroviral therapy.
Am J Gastroenterol.
1995;90:1433-1436.
MEDLINE

38.
Lenzo NP, Garas BA, French MA.
Hepatic steatosis and lactic acidosis associated with stavudine treatment in an HIV patient.
AIDS.
1997;11:1294-1296.
MEDLINE

39.
Lewis W, Dalakas MC.
Mitochondrial toxicity of antiviral drugs.
Nat Med.
1995;1:417-422.
MEDLINE

40.
Brivet FG, Nion I, Megarbane B, et al.
Fatal lactic acidosis and liver steatosis associated with didanosine and stavudine treatment.
J Hepatol.
2000;32:364-365.
MEDLINE

41.
Lonergan JT, Behling C, Pfander H, et al.
Hyperlactatemia and hepatic abnormalities in 10 human immunodeficiency virus-infected patients receiving nucleoside analogue combination regimens.
Clin Infect Dis.
2000;31:162-166.
MEDLINE

42.
Meyer D, Behrens G, Schneider A, et al.
Serum lactate in nucleoside analogue treated HIV patients correlates with serum-lipids [abstract].
Antivir Ther.
2000;5(suppl 5):54. Abstract P52.

43.
Staszewski S.
Update on study 006-EFV + AZT + 3TC versus the current 'standard of care' IDV + AZT + 3TC.
Int J Clin Pract Suppl.
1999;103:10-15.
MEDLINE

44.
Moyle GJ, Datta D, Mandalia S, et al.
Hyperlactatemia and lactic acidosis during antiretroviral therapy.
From: 3rd International Workshop on Adverse Drug Reactions and Lipodystrophy in HIV; October 23-26, 2001; Athens, Greece. Abstract 98.

45.
Carr A, Samaras K, Burton S, et al.
A syndrome of peripheral lipodystrophy, hyperlipidaemia and insulin resistance in patients receiving HIV protease inhibitors.
AIDS.
1998;12:F51-F58.
MEDLINE

46.
Behrens G, Dejam A, Schmidt H, et al.
Impaired glucose tolerance, beta cell function and lipid metabolism in HIV patients under treatment with protease inhibitors.
AIDS.
1999;13:F63-F70.
MEDLINE

47.
Mulligan K, Grunfeld C, Tai VW, et al.
Hyperlipidemia and insulin resistance are induced by protease inhibitors independent of changes in body composition in patients with HIV-1 infection.
J Acquir Immune Defic Syndr.
2000;23:35-43.
MEDLINE

48.
Purnell JQ, Zambon A, Knopp RH, et al.
Effect of ritonavir on lipids and post-heparin lipase activities in normal subjects.
AIDS.
2000;14:51-57.
MEDLINE

49.
Periard D, Telenti A, Sudre P, et al.
Atherogenic dyslipidemia in HIV-infected individuals treated with protease inhibitors.
Circulation.
1999;100:700-705.
MEDLINE

50.
Dong KL, Bausserman LL, Flynn MM, et al.
Changes in body habitus and serum lipid abnormalities in HIV-positive women on highly active antiretroviral therapy (HAART).
J Acquir Immune Defic Syndr.
1999;21:107-113.
MEDLINE

51.
Echevarria KL, Hardin TC, Smith JA.
Hyperlipidemia associated with protease inhibitor therapy.
Ann Pharmacother.
1999;33:859-863.
MEDLINE

52.
Roberts AD, Muesing RA, Parenti DM, et al.
Alterations in serum levels of lipids and lipoproteins with indinavir therapy for human immunodeficiency virus-infected patients.
Clin Infect Dis.
1999;29:441-443.
MEDLINE

53.
Saag MS, Powderly WG, Schambelan M, et al.
Switching antiretroviral drugs for treatment of metabolic complications in HIV-1 infection.
Top HIV Med.
2002;10:47-51.

54.
National Cholesterol Education Program.
Second Report of the Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel II).
Circulation.
1994;89:1333-1445.
MEDLINE

55.
National Cholesterol Education Program Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults.
Executive Summary of the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III).
JAMA.
2001;285:2486-2497.
FULL TEXT  |  PDF  |  MEDLINE

56.
Smith SC Jr, Greenland P, Grundy SM.
AHA Conference Proceedings: prevention conference V.
Circulation.
2000;101:111-116.
MEDLINE

57.
Neuvonen PJ, Jalava KM.
Itraconazole drastically increases plasma concentrations of lovastatin and lovastatin acid.
Clin Pharmacol Ther.
1996;60:54-61.
MEDLINE

58.
Neuvonen PJ, Kantola T, Kivisto KT.
Simvastatin but not prevastatin is very susceptible to interaction with the CYP3A4 inhibitor itraconazole.
Clin Pharmacol Ther.
1998;63:332-341.
MEDLINE

59.
Kahn SE, Beard JC, Schwartz MW, et al.
Increased beta-cell secretory capacity as mechanism for islet adaptation to nicotinic acid-induced insulin resistance.
Diabetes.
1989;38:562-568.
MEDLINE

60.
Greenberg BH, Blackwelder WC, Levy RI.
Primary type V hyperlipoproteinemia.
Ann Intern Med.
1977;87:526-534.
MEDLINE

61.
Noor MA, Lo JC, Mulligan K, et al.
Metabolic effects of indinavir in healthy HIV-seronegative men.
AIDS.
2001;15:F11-F18.
MEDLINE

62.
Dubé MP, Qian D, Edmondson-Melancon H, et al.
Prospective study of the effects of amprenavir-based therapy on glucose and lipid metabolism in HIV-infected patients [abstract].
Antivir Ther.
2000;5(suppl 5):26. Abstract P4.

63.
Petit JM, Duong M, Duvillard L, et al.
HIV-1 protease inhibitors induce an increase of triglyceride level in HIV-infected men without modification of insulin sensitivity.
Horm Metab Res.
2000;32:367-372.
MEDLINE

64.
Lo JC, Mulligan K, Tai VW, Algren H, Schambelan M.
"Buffalo hump" in men with HIV-1 infection.
Lancet.
1998;351:867-870.
MEDLINE

65.
Saint-Marc T, Partisani M, Poizot-Martin I, et al.
A syndrome of peripheral fat wasting (lipodystrophy) in patients receiving long-term nucleoside analogue therapy.
AIDS.
1999;13:1659-1667.
MEDLINE

66.
Sulkowski MS, Thomas DL, Chaisson RE, Moore RD.
Hepatotoxicity associated with antiretroviral therapy in adults infected with human immunodeficiency virus and the role of hepatitis C or B virus infection.
JAMA.
2000;283:74-80.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

67.
Centers for Disease Control and Prevention.
Serious adverse events attributed to nevirapine regimens for postexposure prophylaxis after HIV exposuresworldwide, 1997-2000.
MMWR Morb Mortal Wkly Rep.
2001;49:1153-1156.
MEDLINE

68.
Bristol-Myers Squibb.
Healthcare provider important drug warning [letter].
January 5, 2001.

69.
Perinatal HIV Guidelines Working Group.
Public Health Service Task Force Recommendations for the Use of Antiretroviral Drugs in Pregnant HIV-1 Infected Women for Maternal Health and Interventions to Reduce Perinatal HIV-1 Transmission in the United States.
May 4, 2001. Available at: http://www.hivatis.org/trtgdlns.html#Perinatal. Accessibility verified June 4, 2002.

70.
Tebas P, Powderly WG, Claxton S, et al.
Accelerated bone mineral loss in HIV-infected patients receiving potent antiretroviral therapy.
AIDS.
2000;14:F63-F67.
MEDLINE

71.
Lawal A, Engelson E, Wang J, et al.
Equivalent osteopenia in HIV-infected subjects studied before and during the era of HAART.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 627.

72.
McGowan I, Cheng A, Coleman S, et al.
Assessment of bone mineral density (BMD) in HIV-infected antiretroviral therapy-naive patients.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 628.

73.
Knobel H, Guelar A, Valdecillo G, et al.
Osteopenia in HIV-infected patients.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 629.

74.
Arpadi SM, Horlick M, Thornton J, et al.
Bone mineral content is lower in prepubertal HIV-infected children.
J Acquir Immune Defic Syndr.
2002;29:450-454.
MEDLINE

75.
Glesby MJ, Hoover DR, Vaamonde CM.
Osteonecrosis in patients infected with human immunodeficiency virus.
J Infect Dis.
2001;184:519-523.
MEDLINE

76.
Scribner AN, Troia-Cancio PV, Cox BA, et al.
Osteonecrosis in HIV.
J Acquir Immune Defic Syndr.
2000;25:19-25.
MEDLINE

77.
Keruly JC, Chaisson RE, Moore RD.
Increasing incidence of avascular necrosis of the hip in HIV-infected patients.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 637.

78.
Bourezane Y, Salard D, Hoen B, et al.
DRESS (drug rash with eosinophilia and systemic symptoms) syndrome associated with nevirapine therapy.
Clin Infect Dis.
1998;27:1321-1322.
MEDLINE

79.
Bossi P, Colin D, Bricaire F, Caumes E.
Hypersensitivity syndrome associated with efavirenz therapy.
Clin Infect Dis.
2000;30:227-228.
MEDLINE

80.
Rescriptor (delavirdine) [package insert].
La Jolla, Calif: Agouron Pharmaceutical Company; 2001.

81.
Pirmohamed M, Park BK.
HIV and drug allergy.
Curr Opin Allergy Clin Immunol.
2001;1:311-316.
MEDLINE

82.
Agenerase (amprenavir) [package insert].
Research Triangle Park, NY: GlaxoSmithKline; 2001.

83.
Mallal S, Nolan D, Witt C, et al.
Association between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir.
Lancet.
2002;359:727-732.
MEDLINE

84.
Ziagen (abacavir) [package insert].
Research Triangle Park, NY; GlaxoSmithKline; 2001.

85.
Stone VE, Hogan JW, Schuman P, et al.
Antiretroviral regimen complexity, self-reported adherence, and HIV patients' understanding of their regimens.
J Acquir Immune Defic Syndr.
2001;28:124-131.
MEDLINE

86.
US Food and Drug Administration.
FDA/Bristol Myers Squibb issues caution for HIV combination therapy with Zerit and Videx in pregnant women [FDA Talk Paper].
January 5, 2001. Available at: http://www.fda.gov/bbs/topics/answers/ans01063.html. Accessibility verified June 4, 2002.

87.
Gulick R, Mellors J, Havlir D.
Indinavir (IDV), zidovudine (ZDV) and lamivudine (3TC).
From: 1st International AIDS Society Conference on HIV Pathogenesis and Treatment; July 8-11, 2001; Buenos Aires, Argentina. Abstract 215.

88.
Viracept [package insert].
La Jolla, Calif: Agouron Pharmaceutical Company; 2001.

89.
Murphy RL, Brun S, Hicks C, et al.
ABT-378/ritonavir plus stavudine and lamivudine for the treatment of antiretroviral-naive adults with HIV-1 infection.
AIDS.
2001;15:F1-F9.
MEDLINE

90.
Flexner C.
Dual protease inhibitor therapy in HIV-infected patients.
Annu Rev Pharmacol Toxicol.
2000;40:649-674.
MEDLINE

91.
Kurowski M, Kaeser B, Mroziekiewicz A, et al.
The influence of low doses of ritonavir on the pharmacokinetics of nelfinavir 1250 mg BID.
From: 40th Interscience Conference on Antimicrobial Agents and Chemotherapy; September 17-20, 2000; Toronto, Canada. Abstract 1639.

92.
Norvir [package insert].
Abbott Park, Ill: Abbott Laboratories; 2000.

93.
King M, Bernstein B, Kempf D, et al.
Comparison of time to achieve HIV RNA <400 copies/mL and <50 copies/mL in a phase III, blinded, randomized clinical trial of ABT-378/r vs NFV in ARV-naive patients.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 329.

94.
Benson CA, Deeks SG, Brun SC, et al.
Safety and antiviral activity at 48 weeks of lopinavir/ritonavir plus nevirapine and 2 nucleoside reverse-transcriptase inhibitors in human immunodeficiency virus type 1-infected protease inhibitor-experienced patients.
J Infect Dis.
2002;185:599-607.
MEDLINE

95.
Bernstein B, Moseley J, Kempf D, et al.
Absence of resistance to Kaletra (ABT-378/r) observed through 48 weeks of therapy in antiretroviral-naive subjects.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 453.

96.
Havlir DV, Marschner IC, Hirsch MS, et al.
Maintenance antiretroviral therapies in HIV infected patients with undetectable plasma HIV RNA after triple-drug therapy.
N Engl J Med.
1998;339:1261-1268.
MEDLINE

97.
Murphy RL, Gulick RM, DeGruttola V, et al.
Treatment with amprenavir alone or amprenavir with zidovudine and lamivudine in adults with human immunodeficiency virus infection.
J Infect Dis.
1999;179:808-816.
MEDLINE

98.
Pialoux G, Raffi F, Brun-Vezinet F, et al.
Randomized trial of three maintenance regimens given after three months of induction therapy with zidovudine, lamivudine, and indinavir in previously untreated HIV-1-infected patients.
N Engl J Med.
1998;339:1269-1276.
MEDLINE

99.
Cahn P, Casiro A, Puentes T, et al.
Continued indinavir (800 mg tid) versus switching to indinavir + ritonavir (800/100 mg bid) in HIV patients having achieved viral load suppression.
From: 1st International AIDS Society Conference on HIV Pathogenesis and Treatment; July 7-11, 2001; Buenos Aires, Argentina. Abstract 60.

100.
Cameron DW, Japour AJ, Xu Y, et al.
Ritonavir and saquinavir combination therapy for the treatment of HIV infection.
AIDS.
1999;13:213-224.
MEDLINE

101.
Montaner JSG, Saag M, Barylski C, et al.
FOCUS study.
From: 41st Interscience Conference on Antimicrobial Agents and Chemotherapy; December 16-19, 2001; Chicago, Ill. Abstract 670.

102.
Kurowski M.
Influence of 50 mg, 100 mg and 200 mg ritonavir (RTV) on the pharmacokinetics (PK) of amprenavir (APV) after multiple doses in healthy volunteers for once daily (QD) and twice daily (BID) regimens.
From: 1st International AIDS Society Conference on HIV Pathogenesis and Treatment; July 8-11, 2001; Buenos Aires, Argentina. Abstract 351.

103.
Gulick RM, Hu XJ, Fiscus SA, et al.
Randomized study of saquinavir with ritonavir or nelfinavir together with delavirdine, adefovir, or both in human immunodeficiency virus-infected adults with virologic failure on indinavir.
J Infect Dis.
2000;182:1375-1384.
MEDLINE

104.
Fletcher CV, Acosta EP, Cheng H, et al.
Competing drug-drug interactions among multidrug antiretroviral regimens used in the treatment of HIV-infected subjects.
AIDS.
2000;14:2495-2501.
MEDLINE

105.
Murphy RL, Katlama C, Johnson V, et al.
The Atlantic study.
From: 39th Interscience Conference on Antimicrobial Agents and Chemotherapy; September 26-29, 1999; San Francisco, Calif. Abstract LB-22.

106.
Podzamczer D, Ferrer E, Consiglio E, et al.
Final 12-month results from the COMBINE study.
From: 1st International AIDS Society Conference on HIV Pathogenesis and Treatment; July 7-11, 2001; Buenos Aires, Argentina. Abstract 7.

107.
Viramune [product monograph].
Columbus, Ohio: Roxane Laboratories Inc; 2000.

108.
Sustiva [product monograph].
Wilmington, Del: DuPont Pharmaceuticals Company; 2000.

109.
Montaner J, Gigliotti M, Cahn P, et al.
The effects of a short course of prednisone (pred) on the incidence of rash associated with nevirapine (NVP, VIRAMUNE).
From: 13th International Conference on AIDS; July 9-14, 2000; Durban, South Africa. Abstract WePpB1378.

110.
van der Valk M, Kastelein JJ, Murphy RL, et al.
Nevirapine-containing antiretroviral therapy in HIV-1 infected patients results in an anti-atherogenic lipid profile.
AIDS.
2001;15:2407-2414.
MEDLINE

111.
Staszewski S, Morales-Ramirez J, Tashima KT, et al.
Efavirenz plus zidovudine and lamivudine, efavirenz plus indinavir, and indinavir plus zidovudine and lamivudine in the treatment of HIV-1 infection in adults.
N Engl J Med.
1999;341:1865-1873.
MEDLINE

112.
Staszewski S, Keiser P, Montaner J, et al.
Abacavir-lamivudine-zidovudine vs indinavir-lamivudine-zidovudine in antiretroviral-naive HIV-infected adults.
JAMA.
2001;285:1155-1163.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

113.
Fischl M, Burnside A, Farthing C, et al.
Efficacy of combivir (COM) (lamivudine 150 mg/zidovudine 300 mg) plus ziagen (abacavir [ABC] 300 mg) BID compared to trizivir (TZV) (3TC 150 mg/ZDV 300 mg/ABC 300 mg) BID in patients receiving prior COM plus ABC.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 315.

114.
Kessler HA, Johnson J, Follansbee S, et al.
Abacavir expanded access program for adult patients infected with human immunodeficiency virus type 1.
Clin Infect Dis.
2002;34:535-542.
MEDLINE

115.
Easterbrook PJ, King D, Waters A, et al.
Epidemiological risk factors and immunological mechanisms of hypersensitivity reactions to abacavir.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 621.

116.
Lanier ER, Hellman N, Scott J, et al.
Determination of a clinically relevant phenotypic resistance "cutoff" for abacavir using the PhenoSense assay.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 254.

117.
Melby T, Tortell S, Thorborn D, et al.
Time to appearance of NRTI-associated mutations and response to subsequent therapy for patients on failing ABC/COM.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract 448.

118.
Rosenberg ES, Altfeld M, Poon SH, et al.
Immune control of HIV-1 after early treatment of acute infection.
Nature.
2000;407:523-526.
MEDLINE

119.
Garcia F, Plana M, Ortiz GM, et al.
The virological and immunological consequences of structured treatment interruptions in chronic HIV-1 infection.
AIDS.
2001;15:F29-F40.
MEDLINE

120.
Ruiz L, Carcelain G, Martinez-Picado J, et al.
HIV dynamics and T-cell immunity after three structured treatment interruptions in chronic HIV-1 infection.
AIDS.
2001;15:F19-F27.
MEDLINE

121.
Dybul M, Chun TW, Yoder C, et al.
Short-cycle structured intermittent treatment of chronic HIV infection with highly active antiretroviral therapy.
Proc Natl Acad Sci U S A.
2001;98:15161-15166.
MEDLINE

122.
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.
MEDLINE

123.
Haubrich RH.
Structured treatment interruptions.
Top HIV Med.
2000;8:9-13.

124.
Lori F, Lisziewicz J.
Structured treatment interruptions for the management of HIV infection.
JAMA.
2001;286:2981-2987.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

125.
Deeks SG.
Determinants of virological response to antiretroviral therapy.
Clin Infect Dis.
2000;30(suppl 2):S177-S184.
MEDLINE

126.
Bangsberg DR, Hecht FM, Charlebois ED, et al.
Adherence to protease inhibitors, HIV-1 viral load, and development of drug resistance in an indigent population.
AIDS.
2000;14:357-366.
MEDLINE

127.
Fischl M, Rodriguez A, Scerpella E, et al.
Impact of directly observed therapy on outcomes in HIV clinical trials.
From: 7th Conference on Retroviruses and Opportunistic Infections; January 30-February 2, 2000; San Francisco, Calif. Abstract 71.

128.
Stenzel MS, McKenzie M, Mitty JA, et al.
Enhancing adherence to HAART.
AIDS Read.
2001;11:317-318.
MEDLINE

129.
Tuldra A, Fumaz CR, Ferrer MJ, et al.
Prospective randomized two-arm controlled study to determine the efficacy of a specific intervention to improve long-term adherence to highly active antiretroviral therapy.
J Acquir Immune Defic Syndr.
2000;25:221-228.
MEDLINE

130.
Marschner IC, Collier AC, Coombs RW, et al.
Use of changes in plasma levels of human immunodeficiency virus type 1 RNA to assess clinical benefit to antiretroviral therapy.
J Infect Dis.
1998;177:40-47.
MEDLINE

131.
Kim S, Hughes MD, Hammer SM, et al.
Both serum HIV type 1 RNA levels and CD4+ lymphocyte counts predict clinical outcome in HIV type 1-infected subjects with 200 to 500 CD4+ cells per cubic millimeter.
AIDS Res Hum Retroviruses.
2000;16:645-653.
MEDLINE

132.
Hughes MD, Johnson VA, Hirsch MS, et al.
Monitoring plasma HIV-1 RNA levels in addition to CD4+ lymphocyte count improves assessment of antiretroviral therapeutic response.
Ann Intern Med.
1997;126:929-938.
MEDLINE

133.
Yamashita TE, Phair JP, Munoz A, et al.
Immunologic and virologic response to highly active antiretroviral therapy in the Multicenter AIDS Cohort Study.
AIDS.
2001;15:735-746.
MEDLINE

134.
Le Moing V, Thiebaut R, Chene G, et al.
Predictors of long-term increase in CD4(+) cell counts in human immunodeficiency virus-infected patients receiving a protease inhibitor-containing antiretroviral regimen.
J Infect Dis.
2002;185:471-480.
MEDLINE

135.
Brambilla D, Reichelderfer PS, Bremer JW, et al.
The contribution of assay variation and biological variation to the total variability of plasma HIV-1 RNA measurements.
AIDS.
1999;13:2269-2279.
MEDLINE

136.
Schapiro JM, Winters MA, Stewart F, et al.
The effect of high-dose saquinavir on viral load and CD4+ T-cell counts in HIV-infected patients.
Ann Intern Med.
1996;124:1039-1050.
MEDLINE

137.
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.
MEDLINE

138.
Demeter LM, Hughes MD, Coombs RW, et al.
Predictors of virologic and clinical outcomes in HIV-1-infected patients receiving concurrent treatment with indinavir, zidovudine, and lamivudine.
Ann Intern Med.
2001;135:954-964.
MEDLINE

139.
Powderly WG, Saag MS, Chapman S, et al.
Predictors of optimal virological response to potent antiretroviral therapy.
AIDS.
1999;13:1873-1880.
MEDLINE

140.
Lepri AC, Miller V, Phillips AN, et al.
The virological response to highly active antiretroviral therapy over the first 24 weeks of therapy according to the pre-therapy viral load and the weeks 4-8 viral load.
AIDS.
2001;15:47-54.
MEDLINE

141.
Spritzler J, Mildvan D, Russo A, et al.
Immunologic predictors of discordance between CD4+ T-cell and HIV-1 RNA responses during antiretroviral therapy.
From: 9th Conference on Retroviruses and Opportunistic Infections; February 24-28, 2002; Seattle, Wash. Abstract 489-M.

142.
Haubrich RH, Currier JS, Forthal DN, et al.
A randomized study of the utility of human immunodeficiency virus RNA measurement for the management of antiretroviral therapy.
Clin Infect Dis.
2001;33:1060-1068.
MEDLINE

143.
Havlir DV, Bassett R, Levitan D, et al.
Prevalence and predictive value of intermittent viremia with combination HIV therapy.
JAMA.
2001;286:171-179.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

144.
Hirsch MS, Conway B, D'Aquila RT, et al.
Antiretroviral drug resistance testing in adults with HIV infection.
JAMA.
1998;279:1984-1991.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

145.
Hirsch MS, Brun-Vézinet F, D'Aquila RT, et al.
Antiretroviral drug resistance testing in adult HIV-1 infection.
JAMA.
2000;283:2417-2426.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

146.
D'Aquila RT, Shapiro JM, Brun-Vézinet F, et al.
Drug resistance mutations in HIV-1.
Top HIV Med.
2002;10:11-15.

147.
Cohen C, Hunt S, Sension M, et al.
A randomized trial assessing the impact of phenotypic resistance testing on antiretroviral therapy.
AIDS.
2002;16:579-588.
MEDLINE

148.
Tural C, Ruiz L, Holtzer C, et al.
Clinical utility of HIV-1 genotyping and expert advice.
AIDS.
2002;16:209-218.
MEDLINE

149.
DeGruttola V, Dix L, D'Aquila R, et al.
The relation between baseline HIV drug resistance and response to antiretroviral therapy.
Antivir Ther.
2000;5:41-48.
MEDLINE

150.
Kemper CA, Witt MD, Keiser PH, et al.
Sequencing of protease inhibitor therapy.
AIDS.
2001;15:609-615.
MEDLINE

151.
Brun-Vézinet F, Race E, Descamps D, et al.
Differences between genotype and phenotype in the NARVAL trial, ANRS 088.
Antivir Ther.
2000;5(suppl 3):78-79.

152.
Baxter JD, Mayers DL, Wentworth DN, et al.
A randomized study of antiretroviral management based on plasma genotypic antiretroviral resistance testing in patients failing therapy.
AIDS.
2000;14:F83-F93.
MEDLINE

153.
Call SA, Saag MS, Westfall AO, et al.
Phenotypic drug susceptibility testing predicts long-term virologic suppression better than treatment history in patients with human immunodeficiency virus infection.
J Infect Dis.
2001;183:401-408.
MEDLINE

154.
Little SJ, Daar ES, D'Aquila RT, et al.
Reduced antiretroviral drug susceptibility among patients with primary HIV infection.
JAMA.
1999;282:1142-1149.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

155.
Gerber JG, Acosta EP.
The potential role of therapeutic drug monitoring in the treatment of HIV infection.
Top HIV Med.
2002;10:27-32.

156.
Burger DM.
Results from the ATHENA study.
From: 2nd International Workshop on Clinical Pharmacology of HIV Therapy; April 2-4, 2001; Noordwijk, the Netherlands. Abstract 6.2a and 6.2b.

157.
Kempf DJ, Rode RA, Xu Y, et al.
The duration of viral suppression during protease inhibitor therapy for HIV-1 infection is predicted by plasma HIV-1 RNA at the nadir.
AIDS.
1998;12:F9-F14.
MEDLINE

158.
Polis MA, Sidorov IA, Yoder C, et al.
Correlation between reduction in plasma HIV-1 RNA concentration 1 week after start of antiretroviral treatment and longer-term efficacy.
Lancet.
2001;358:1760-1765.
MEDLINE

159.
Raboud JM, Montaner JS, Conway B, et al.
Suppression of plasma viral load below 20 copies/µL is required to achieve a long-term response to therapy.
AIDS.
1998;12:1619-1624.
MEDLINE

160.
Chen RY, Mugavero MJ, Westfall AO, et al.
Durability of HAART regimens over time.
From: 41st Interscience Conference on Antimicrobial Agents and Chemotherapy; December 16-19, 2001; Chicago, Ill. Abstract 1914.

161.
Mocroft A, Devereux H, Kinloch-de-Loes S, et al.
Immunological, virological and clinical response to highly active antiretroviral therapy treatment regimens in a complete clinic population.
AIDS.
2000;14:1545-1552.
MEDLINE

162.
Parkin NT, Deeks SG, Wrin MT, et al.
Loss of antiretroviral drug susceptibility at low viral load during early virological failure in treatment-experienced patients.
AIDS.
2000;14:2877-2887.
MEDLINE

163.
Zolopa AR, Shafer RW, Warford A, et al.
HIV-1 genotype resistance patterns predict response to saquinavir-ritonavir therapy in patients in whom previous protease inhibitor therapy had failed.
Ann Intern Med.
1999;131:813-821.
MEDLINE

164.
International AIDS Society–USA Resistance Mutations Project Panel.
Update on Drug Resistance Mutations in HIV-1.
Available at: http://www.iasusa.org/resistance_mutations/
revisedmutafigures-11.30.01.pdf
. Accessibility verified June 4, 2002.

165.
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.
MEDLINE

166.
Miller V, Margot NA, Lu B.
Effect of baseline nucleoside-associated resistance on response to tenofovir DF (TDF) therapy.
From: 9th Conference on Retroviruses and Opportunistic Infections; February 24-28, 2002; Seattle, Wash. Abstract 43.

167.
Palmer S, Margot N, Gilbert H, et al.
Tenofovir, adefovir, and zidovudine susceptibilities of primary human immunodeficiency virus type 1 isolates with non-B subtypes or nucleoside resistance.
AIDS Res Hum Retroviruses.
2001;17:1167-1173.
MEDLINE

168.
Miller MD, Margot NA, Hertogs K, et al.
Antiviral activity of tenofovir (PMPA) against nucleoside-resistant clinical HIV samples.
Nucleosides Nucleotides Nucleic Acids.
2001;20:1025-1028.
MEDLINE

169.
Schooley R, Ruane P, Myers R, et al.
Tenofovir DF.
From: 41st Interscience Conference on Antimicrobial Agents and Chemotherapy; December 16-19, 2001; Chicago, Ill. Abstract 1929.

170.
Squires K, Pierone G, Berger D, et al.
Tenofovir DF.
From: 41st Interscience Conference on Antimicrobial Agents and Chemotherapy; December 16-19, 2001; Chicago, Ill. Abstract 666.

171.
Louie M, Hogan C, Hurley A, et al.
Determining the relative efficacy of tenofovir DF using frequent measurements of HIV-1 RNA during a short course of monotherapy in antiretroviral drug naive individuals.
From: 9th Conference on Retroviruses and Opportunistic Infections; February 24-28, 2002; Seattle, Wash. Abstract 3.

172.
Deeks SG, Barbour JD, Martin JN, et al.
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.
MEDLINE

173.
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.
MEDLINE

174.
Grant RM, Liegler T, Elkin C, et al.
Protease inhibitor resistant HIV-1 has marked decreased fitness in vivo.
From: 8th Conference on Retroviruses and Opportunistic Infections; February 4-8, 2001; Chicago, Ill. Abstract LB4.

175.
DeHovitz JA, Kovacs A, Feldman JG, et al.
The relationship between virus load response to highly active antiretroviral therapy and change in CD4 cell counts.
J Infect Dis.
2000;182:1527-1530.
MEDLINE

176.
Miller V, Sabin C, Hertogs K, et al.
Virological and immunological effects of treatment interruptions in HIV-1 infected patients with treatment failure.
AIDS.
2000;14:2857-2867.
MEDLINE

177.
Miller V, Cozzi-Lepri A, Hertogs K, et al.
HIV drug susceptibility and treatment response to mega-HAART regimen in patients from the Frankfurt HIV cohort.
Antivir Ther.
2000;5:49-55.
MEDLINE

178.
Montaner JS, Harrigan PR, Jahnke N, et al.
Multiple drug rescue therapy for HIV-infected individuals with prior virologic failure to multiple regimens.
AIDS.
2001;15:61-69.
MEDLINE

179.
Deeks SG, Hoh R, Grant RM, et al.
CD4+ T cell kinetics and activation in human immunodeficiency virus-infected patients who remain viremic despite long-term treatment with protease inhibitor-based therapy.
J Infect Dis.
2002;185:315-323.
MEDLINE

180.
Sereti I, Lane HC.
Immunopathogenesis of human immunodeficiency virus.
Clin Infect Dis.
2001;32:1738-1755.
MEDLINE

181.
Kovacs JA, Baseler M, Dewar RJ, et al.
Increases in CD4 T lymphocytes with intermittent courses of interleukin-2 in patients with human immunodeficiency virus infection.
N Engl J Med.
1995;332:567-575.
MEDLINE

182.
Levy Y, Capitant C, Houhou S, et al.
Comparison of subcutaneous and intravenous interleukin-2 in asymptomatic HIV-1 infection.
Lancet.
1999;353:1923-1929.
MEDLINE

183.
Davey RT, Murphy RL, Graziano FM, et al.
Immunologic and virologic effects of subcutaneous interleukin 2 in combination with antiretroviral therapy.
JAMA.
2000;284:183-189.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

184.
Davey RT, Chaitt DG, Albert JM, et al.
Randomized trial of high versus low-dose subcutaneous interleukin-2 outpatient therapy for early human immunodeficiency virus type 1 infection.
J Infect Dis.
1999;179:849-858.
MEDLINE

185.
Kilby JM, Tabereaux PB, Mulanovich V, et al.
Effects of tapering doses of oral prednisone on viral load among HIV-infected patients with unexplained weight loss.
AIDS Res Hum Retroviruses.
1997;13:1533-1537.
MEDLINE

186.
Lori F.
Hydroxyurea and HIV.
AIDS.
1999;13:1433-1442.
MEDLINE

187.
Macgregor RR, Boyer JD, Ugen KE, et al.
First human trial of a DNA-based vaccine for treatment of human immunodeficiency virus type 1 infection.
J Infect Dis.
1998;178:92-100.
MEDLINE

188.
Barouch DH, Craiu A, Kuroda MJ, et al.
Augmentation of immune responses to HIV-1 and simian immunodeficiency virus DNA vaccines by IL-2/Ig plasmid administration in rhesus monkeys.
Proc Natl Acad Sci U S A.
2000;97:4192-4197.
MEDLINE

189.
Clements-Mann ML, Weinhold K, Matthews TJ, et al.
Immune responses to human immunodeficiency virus (HIV) type 1 induced by canarypox expressing HIV-1MN gp120, HIV-1SF2 recombinant gp120, or both vaccines in seronegative adults.
J Infect Dis.
1998;177:1230-1246.
MEDLINE

190.
Havlir DV, Gilbert PB, Bennett K, et al.
Effects of treatment intensification with hydroxyurea in HIV-infected patients with virologic suppression.
AIDS.
2001;15:1379-1388.
MEDLINE

 

 

Edward E. Rylander, M.D.

Diplomat American Board of Family Practice.

Diplomat American Board of Palliative Medicine.