archived

Evidence Summary

Human Immunodeficiency Virus (HIV) Infection: Screening, 2005

July 05, 2005

Recommendations made by the USPSTF are independent of the U.S. government. They should not be construed as an official position of the Agency for Healthcare Research and Quality or the U.S. Department of Health and Human Services.

Screening for HIV: A Review of the Evidence for the U.S. Preventive Services Task Force

By Roger Chou, MD; Laurie Hoyt Huffman, MS; Rongwei Fu, PhD; Ariel K. Smits, MD, MPH; and P. Todd Korthuis, MD, MPH.

This article was first published in Ann Intern Med 2005:143:55-73.

Return to Table of Contents

Background: HIV infection affects 850,000 to 950,000 persons in the United States. The management and outcomes of HIV infection have changed substantially since the U.S. Preventive Services Task Force issued recommendations in 1996. 

Purpose: To synthesize the evidence on risks and benefits of screening for HIV infection.

Data Sources: MEDLINE, the Cochrane Library, reference lists, and experts.

Study Selection: Studies of screening, risk factor assessment, accuracy of testing, follow-up testing, and efficacy of interventions.

Data Extraction: Data on settings, patients, interventions, and outcomes were abstracted for included studies; quality was graded according to criteria developed by the Task Force.

Data Synthesis: No trials directly link screening for HIV with clinical outcomes. Many HIV-infected persons in the United States currently receive diagnosis at advanced stages of disease, and almost all will progress to AIDS if untreated. Screening based on risk factors could identify persons at substantially higher risk but would miss a substantial proportion of those infected. Screening tests for HIV are extremely (>99%) accurate. Acceptance rates for screening and use of recommended interventions vary widely. Highly active antiretroviral therapy (HAART) substantially reduces the risk for clinical progression or death in patients with immunologically advanced disease. Along with other adverse events, HAART is associated with an increased risk for cardiovascular complications, although absolute rates are low after 3 to 4 years.

Limitations: Data are insufficient to estimate the effects of screening and interventions on transmission rates or in patients with less immunologically advanced disease. Long-term data on adverse events associated with HAART are not yet available.

Conclusions: Benefits of HIV screening appear to outweigh harms. The yield from screening higher-prevalence populations would be substantially higher than that from screening the general population.

Return to Table of Contents

Infection with HIV-1 is estimated to affect 850,000 to 950,000 persons in the United States.1 Of those infected, 25% (180,000 to 280,000) are thought to be unaware of their status.1 Almost all patients with untreated HIV infection eventually develop AIDS.2 In the United States, more than 500,000 patients with AIDS have died; approximately 18,000 died in 2003.3 AIDS is the seventh leading cause of death in persons 15 to 24 years of age, and the fifth leading cause in persons 25 to 44 years of age.4 Since 1992, 40,000 new HIV infections have been diagnosed annually.5 Statistical modeling suggests that approximately half of HIV-infected persons in the United States acquire their infection by 25 years of age.6

Infection with HIV causes immune deficiency to a large extent by decreasing the level and function of CD4 T lymphocytes. In untreated patients with CD4 cell counts less than 0.200 x 109 cells/L, the chance of clinical progression or death over 3 years is approximately 86%.7 A higher HIV-1 viral load also predicts faster disease progression.7-10

To update its 1996 recommendations, the U.S. Preventive Services Task Force (USPSTF) commissioned a new systematic review of the risks and benefits of testing for anti-HIV antibodies in asymptomatic adolescents and adults.11 Another article in this issue reviews screening in pregnant women.12

Return to Table of Contents

The Figure summarizes the analytic framework and key questions for this review. Key question 1 addresses direct evidence on the effects of screening on clinical outcomes. The other key questions address the chain of evidence necessary to estimate the effects of screening on clinical outcomes if direct evidence is insufficient. Appendix A discusses the scope and methods used for this review in more detail.

Briefly, we identified relevant studies from MEDLINE® (1983 through 30 June 2004) and the Cochrane Clinical Trials Registry (2004, issue 2), reference lists, hand searches of relevant journals, and suggestions from experts (Appendix B). We selected studies that provided evidence on the benefits and harms of screening, risk factor assessment, accuracy of testing, followup testing, interventions, acceptability of HIV testing, and cost-effectiveness of screening in outpatient settings in the highly active antiretroviral therapy (HAART) era. For interventions, we focused on studies of HAART.14,15 We also reviewed studies on the effectiveness of counseling on risky behaviors16 and prophylaxis against opportunistic infections.17 A separate report13 reviews the effectiveness of other interventions (immunizations, more frequent Papanicolaou testing, and routine monitoring and followup).

We assessed the internal validity and relevance of included studies using predefined criteria developed by the USPSTF (Appendix C).18,19 We rated the overall body of evidence for each key question using the system developed by the USPSTF.

We used the results of the evidence review to construct an outcomes table estimating the effects of one-time screening for HIV infection in hypothetical cohorts of adolescents and adults. We calculated numbers needed to screen (NNS) and treat (NNT) to prevent 1 case of clinical progression or death or to cause 1 cardiovascular complication for each cohort. The point estimates and 95% CIs for NNS and NNT were based on Monte Carlo simulations. CD4 cell counts are reported as X 109 cells/L; to convert to cells/mm3, multiply by 1000.

This research was funded by the Agency for Healthcare Research and Quality (AHRQ) under a contract to support the work of the USPSTF. Agency staff and USPSTF members participated in the initial design of the study and reviewed interim analyses and the final report. Draft reports were distributed to 25 content experts for review. Agency approval was required before this manuscript could be submitted for publication, but the authors are solely responsible for the content and the decision to submit it for publication.

Return to Table of Contents

Does Screening for HIV Infection in Asymptomatic Adolescents and Adults Reduce Premature Death and Disability or Spread of Disease?

No studies compared clinical outcomes between patients in the general population who were screened or not screened for HIV.

Can Clinical or Demographic Characteristics Identify Subgroups of Asymptomatic Adolescents and Adults at Increased Risk for HIV Infection Compared to the General Population?

A substantial proportion of Americans report behaviors that could put them at risk for HIV infection (Table 1).20 A recent U.S. telephone survey (n = 33,913) found that 11% of sexually active respondents reported multiple partners within the last year, and 4.2% reported other high-risk behaviors.21 Adolescents,22,23 men who have sex with men,24 and persons attending sexually transmitted disease clinics also report high rates of recent risky behaviors.25 Even in settings with good access to health care, high-risk behaviors often remain undetected26 or fail to lead to testing despite identification.27

The largest (n = 1,281,606) U.S. study found that 20% to 26% of HIV-infected people identified at federally funded testing sites reported no risk factors.28 Other studies in a variety of settings indicated that 7% to 51% of HIV-positive patients reported no risk factors.26,29-36 The rate of HIV positivity in patients reporting no risk factors was lower in low-prevalence (0.1% to 2.0%) than in high-prevalence (>5%) sites (0.2% to 0.8% vs. 1.4% to 5.7%).28

One good-quality prospective study in a sexually transmitted disease clinic evaluated different methods of selective screening, such as screening only persons with reported risk factors, those with reported risk factors or those in high-prevalence demographic groups, or screening everybody. In this study, screening only persons who reported risk factors (5.8% of those tested) would have resulted in 74% (79 of 107) missed diagnoses. A broader strategy (70% tested) of also screening persons in high-prevalence demographic groups (black men or persons > 30 years of age) would have resulted in substantially fewer (8%) missed diagnoses.37 Two retrospective studies found that similar selective strategies would have resulted in 33% to 41% of the population being tested and 7% (1 of 14)38 to 13% (192 of 1,474)39 missed diagnoses. Four U.S. studies in high-prevalence (>1%) settings demonstrated an increased yield after the implementation of routine voluntary HIV screening.40-43

What Are the Test Characteristics of HIV Antibody Test Strategies?

The use of repeatedly reactive enzyme immunoassay followed by confirmatory Western blot or immunofluorescent assay remains the standard method for diagnosing HIV-1 infection.44,45 A large study of HIV testing in 752 U.S. laboratories reported a sensitivity of 99.7% and specificity of 98.5% for enzyme immunoassay,45 and studies in U.S. blood donors reported specificities of 99.8% and greater than 99.99%.46,47 With confirmatory Western blot, the chance of a false-positive identification in a low-prevalence setting is about 1 in 250,000 (95% CI, 1 in 173,000 to 1 in 379,000).48

Three rapid (results available in 10 to 30 minutes) HIV tests are in use in the United States; 2 (Uni-Gold Recombigen, Trinity Biotech Plc., Bray, Ireland, and OraQuick Advance, OraSure Technologies, Bethlehem, Pennsylvania) for true point-of-care testing,49 and 1 (Reveal G2, MedMira Laboratories, Inc., Halifax, Nova Scotia, Canada) performed in a laboratory. Three good-quality and 10 fair-quality studies evaluated accuracy of rapid tests on blood specimens against standard HIV testing.50-55 Ten were reported in manufacturer inserts.50-52 Most studies reported the accuracy of rapid tests before confirmatory testing because patients may be notified of results before confirmation is available.56

For the OraQuick test, 3 good-quality studies found sensitivities ranging from 96% to 100% and specificity greater than 99.9%.53-55 Three fair-quality studies found sensitivities ranging from 99.6% to 100%, with specificity 100% in all.50 For the Uni-Gold and Reveal tests, 7 fair-quality studies reported sensitivities ranging from 94% to 100% and specificities greater than 99%.50,52 The positive predictive values for the Reveal and Uni-Gold tests were calculated at 25% to 50% in settings with a prevalence of 0.3%, and at 85% to 95% in settings with a prevalence of 5%.57 One good-quality study among 5744 U.S. pregnant women (prevalence, 0.59%) found a positive predictive value of 90% (4 false-positive results) and a negative predictive value of 100% for the OraQuick test using blood.53

Two large (n = 3,570 and n = 4,442), good-quality studies of the OraSure Oral Specimen Collection Device (Epitope, Inc., Beaverton, Oregon) measured sensitivities of 99.9% and 99.2% and specificities of 99.9% and 99.2%.58,59 Urine HIV tests generally appear less accurate than standard testing and are not in widespread use in the United States.60-63 A good-quality (n = 1,255) study of the only U.S. Food and Drug Administration-approved home collection kit (Home Access, Home Access Health Corp., Hoffman Estates, Illinois) found that the sensitivity and specificity obtained with use of finger-stick blood spot samples were both 100% compared to standard testing.64 More than 98% of participants in 2 studies obtained adequate samples for testing.64,65

No studies have evaluated the optimal frequency of HIV screening, which partly depends on the incidence and the prevalence of undetected HIV infection in the group being tested.66

What Are the Harms Associated with Screening?

Information on the frequency and consequences (anxiety, labeling) of false-positive test results is anecdotal67-69 False- and true-negative results could provide false reassurance if high-risk behaviors are continued.

True-positive HIV test results are associated with important harms, including fears of rejection, abandonment, verbal abuse, and physical assault.70 A substantial proportion (20% to 25%) of Americans continue to agree with stigmatizing statements about HIV.71,72 Four percent of 142 patients with recently diagnosed HIV infection reported losing a job because of their status, 1% had been asked to move, and 1% had been assaulted.73

Notification of a positive HIV test result can lead to emotional and psychological distress. On the other hand, receipt of a negative HIV test result is associated with reduced anxiety in at-risk individuals74 Although earlier studies reported high suicide rates after a positive test result,75-78 no studies have addressed suicide risk after an HIV diagnosis in the HAART era. A large prospective cohort study through 1993 found that suicide rates after routine screening were similar between HIV-positive and HIV-negative military recruits.79 Counseling may reduce distress after a positive test result.80-83

Both HIV-negative and HIV-positive persons appear to have similar rates of intimate partner violence when matched for high-risk behaviors.84-86 One prospective cohort study found that rates of abuse declined after disclosure of HIV status.87 Several small observational studies did not find an increased rate of partnership dissolution after a positive diagnosis.87-89

Is Screening Acceptable to Patients?

In the United States, as of 2002 approximately half (43.5%) of persons age 18 to 64 years had been tested at least once for HIV.90 The proportion of tested female adolescents is substantially lower at 25%.91 Among persons reporting high-risk behaviors, recent studies found that 20% to 30% had never been tested.25,92,93

A good-quality systematic review of 62 studies reported that acceptance rates of voluntary HIV testing varied widely (from 11% to 91%) in the United States, even within similar health care settings.94 In general, low-prevalence settings were associated with lower acceptance rates. Higher acceptance rates were associated with the client's perception of HIV risk, acknowledgment of risk behaviors, confidentiality protections, and the provider's belief that testing would be beneficial.

One United Kingdom study of "opt-out" testing (in which an HIV test is considered routine and is performed unless the patient declines) in nonpregnant persons found that uptake increased from 35% to 65%.95 In several studies, anonymous testing was associated with increased testing rates96-98 or higher mean CD4 cell count at diagnosis,99 although others did not find a clear association.100-102 In Connecticut, testing rates in adolescents doubled after removal of a parental consent requirement.103

No clinical trials have evaluated the incremental acceptability of alternative testing (rapid test, home sampling, or oral sampling) compared with standard testing. A recent observational study found that 29% to 69% of patients in different settings accepted rapid testing.104 Another found that all 150 patients being treated for substance abuse who accepted testing chose an oral fluid test over a blood test.105 In studies of patients who accepted home sample collection106,107 or oral fluid sampling,108 a substantial proportion (22% to 33% for home sampling and 58% for oral fluid sampling) had not been previously tested.

How Many Newly Diagnosed HIV-Positive Patients Meet Criteria for Antiretroviral Treatment or Prophylaxis against Opportunistic Infections?

In asymptomatic HIV-positive patients, viral load and CD4 cell count testing are used to determine eligibility for HAART and opportunistic infection prophylaxis.14,16 Antiretroviral therapy is currently recommended for patients with CD4 cell counts less than 0.200 x 109 cells/L. Antiretroviral therapy can also be considered for other asymptomatic patients at high risk for disease progression (CD4 cell count < 0.350 X cells 109/L or viral load > 100,000 copies/mL). Interventions are generally less effective in persons with advanced immune deficiency,109 although some benefit is seen.110,111

No studies report both CD4 cell count and viral load in newly diagnosed patients. Seven U.S. studies in different settings found that the proportion of patients with CD4 cell counts less than 0.200 X 109 cells /L at diagnosis or when establishing care ranged from 12% to 43%, and the proportion with CD4 cell counts less than 0.500 X 109 cells/L ranged from 46% to 80%.26,41,112-116

Screening could identify a higher proportion of persons whose CD4 cell counts have not decreased below thresholds for interventions. In addition, patients with an adequate response to HAART can safely discontinue prophylaxis against certain opportunistic infections.17 We identified no studies estimating the effects of screening or treatment on the proportion of patients qualifying for different interventions.

How Many HIV-Positive Patients Who Meet Criteria for Interventions Receive Them?

Patients positive for HIV who meet criteria for interventions may not receive them. Ten percent to 44% of tested patients do not have a post-test counseling session or fail to return for test results,117-119 although most (79% to 93%) positive patients are eventually located.30,120 Two recent studies of routine testing in urgent care centers found that 74% to 82% of patients learned of their positive results.40-41

Rapid testing was associated with a higher rate of HIV-positive persons learning their status than was standard testing in an anonymous testing clinic (100% vs. 86%),121 sexually transmitted disease clinic (97% vs. 79%),121 and emergency department setting (73% vs. 62%).122 In noncomparative studies, rapid testing resulted in more than 98% of patients learning their status104,123 Of 174,316 persons submitting home samples, 95% to 96% called for results.106

Patients positive for HIV may delay medical care or not receive care at all. In 1996, 36% to 63% of HIV-positive patients were regularly seeing a non-emergency department provider.124 Studies in the United States found that 17% to 29% of patients had delayed entry into care for at least 3 months,125,126 and 11% to 39% delayed it for at least 1 year.126-128 A study of rapid testing found that entry into care within 6 months ranged from 100% (in a sexually transmitted disease clinic) to 22% (in a jail).104

No prospective studies measured the proportion of newly diagnosed HIV-positive persons who received appropriate treatment. Four large (n = 1,411 to 9,530) U.S. surveys found that 53% to 85% of HIV-positive patients were receiving antiretroviral therapy according to then-current guidelines.129-132

How Effective Are Interventions in Improving Clinical Outcomes?

Antiretroviral Agents

Currently, HAART regimens with 3 or more antiretroviral agents, usually from at least 2 different classes, are the standard of care for HIV-infected persons receiving antiretroviral therapy.14,15 A good-quality systematic review of 54 randomized, controlled trials with 16,684 HIV-infected patients with limited or no antiretroviral experience found that 3-drug therapy was more effective than 2-drug therapy (odds ratio, 0.62 [95% CI, 0.50 to 0.78]).133 Observational studies indicate that HAART can result in sustained (up to 4 to 5 years) improvements in CD4 cell counts and viral loads,134-136 although long-term clinical outcomes data are not yet available.

Large, good-quality cohort studies from the United States137-140 and Europe141-143 parallel the findings of the systematic review regarding the effectiveness of HAART. In addition, studies have consistently found a marked decline in morbidity and mortality among U.S. HIV-infected patients that coincided with the widespread adoption of HAART.138-140,144-149 In 2 U.S. studies, for example, mortality rates declined from 20.2140 and 29.4138 per 100 person-years to 8.4 and 8.8 per 100 person-years, respectively.

Few trials have adequately assessed the effect of HAART on quality of life or functional status (such as ability to work).133 Four fair-quality trials of 3-drug vs. 2-drug regimens reported conflicting results for differences in quality-of-life outcomes.150-153

The use of HAART could decrease the spread of HIV from infected persons by decreasing viral loads.154 On the other hand, increases in risky behaviors by patients receiving HAART could offset the beneficial effects of viral suppression.155-158 A recent good-quality meta-analysis of 25 studies found no association between receipt of HAART or having an undetectable viral load and unprotected sex.159 Among both seronegative and seropositive persons, however, unprotected intercourse was associated with optimistic beliefs about HAART or an undetectable viral load (odds ratio, 1.82 [CI, 1.52 to 2.17]).

No studies have estimated the effects of HAART on horizontal transmission rates. One cohort study found that heterosexual transmission from monogamous zidovudine-treated men was lower than that from untreated men (relative risk, 0.5 [CI, 0.1 to 0.9]).160 An epidemiologic study estimated that the annual HIV transmission rate from HIV-seropositive persons in the United States declined from 13% in 1987 (the year zidovudine was introduced) to 5.5% in 1989, and has remained steady at approximately 4.2% since 1990.161 This study was not designed to assess the relative contribution of antiretroviral therapy, changes in high-risk behaviors, or other factors to changes in transmission rates.

Counseling

Because the incidence of new HIV infections has remained steady while mortality due to AIDS has declined, the number of persons living with HIV infection in the United States continues to increase.3 A substantial proportion of HIV-infected persons report behaviors that increase the risk for transmitting infection.16,24,126,162-164 Data on the link between sexual behaviors and reduced risk for HIV transmission are strongest for consistent use of condoms for prevention of heterosexual transmission.165,166 Good-quality systematic reviews found that testing plus counseling is most effective in reducing risky behaviors among serodiscordant heterosexual couples and those testing HIV-positive, with less evidence for beneficial effects in other populations.167-169 Several recent fair-quality observational studies reported decreased self-reported risky behaviors after patients had HIV testing or received a positive diagnosis.170-173 Some174-178 but not all179-182 fair-quality randomized trials found that targeted (tailored to participant needs) or more intensive counseling was associated with greater reductions in risky behaviors than standard or less intensive counseling, but counseling methods varied greatly across trials.

No clinical trials evaluated the impact of testing and counseling compared to no testing and counseling on HIV transmission rates. One prospective U.S. study of 144 serodiscordant heterosexual couples who received counseling and reported reduced risky behaviors found no seroconversion after 193 couple-years of followup.183 A prospective African study found that the rate of seroconversion among uninfected female partners of HIV-positive men was 6 to 9/100 person-years, compared with 22/100 person-years in women with untested partners.184 Two observational studies found that testing plus counseling was associated with a moderate (about 33%) decrease in sexually transmitted diseases among those who tested positive but that it increased the risk among those who tested negative (relative risk, 1.27 to 2).185,186 Two good-quality randomized, controlled trials found that more interactive counseling was more effective than standard counseling in reducing sexually transmitted disease rates among HIV-positive women176 and seronegative heterosexual persons,187 although there were too few new HIV infections to detect differences in HIV rates.187

No studies have estimated the effects of counseling HIV-positive persons regarding injection drug use behaviors on HIV transmission rates. Although cross-sectional studies found that HIV-positive drug users reported less risky behaviors than those untested or not infected,188-190 1 randomized trial191 and 1 prospective study192 found that testing plus counseling was not associated with decreased drug behaviors. On the other hand, 2 randomized trials found that more intense counseling reduced drug use behaviors more than did standard counseling.174,193

Prophylaxis Against Opportunistic Infections

Table 2 summarizes 2 good-quality systematic reviews194,195 and 3 clinical trials196-198 of primary prophylaxis against Pneumocystis carinii pneumonia. Prophylaxis was associated with a nonsignificant mortality benefit.194 Several medications used for prophylaxis against P. carinii pneumonia are also effective for toxoplasmosis prophylaxis.17,195

Two good-quality systematic reviews199,200 found that isoniazid prophylaxis was effective at preventing tuberculosis (risk reduced by 60% to 86%) and death (risk reduced by 21% to 23%) in HIV-positive patients with a positive tuberculin skin test result.17

Table 3 summarizes 4 good-quality placebo-controlled trials201-203 and 2 head-to-head trials204,205 of primary prophylaxis against disseminated Mycobacterium avium intracellulare complex infection. Only clarithromycin was associated with a significant mortality benefit.202

Two placebo-controlled trials of ganciclovir for cytomegalovirus prophylaxis found mixed results for reducing invasive CMV infection, no mortality benefit, and significant adverse events.206,207

In Asymptomatic Patients with HIV Infection, Does Immediate Antiretroviral Treatment Result in Improvements in Clinical Outcomes Compared to Delayed Treatment until the Patient Is Symptomatic?

Initiation of HAART in asymptomatic patients must be weighed against potential harms, including effects on quality of life, long-term adverse events, and the development of resistance. Current U.S. guidelines recommend that all asymptomatic patients with CD4 cell counts less than 0.200 x 109 cells/L be offered HAART.14 Recommendations for other asymptomatic patients are less firm.

Twelve observational studies evaluated the risk for disease progression or death in asymptomatic patients initiating HAART at different CD4 cell count thresholds above 0.200 x 109 cells/L. All lasted less than 4 years and could underestimate long-term risks for immediate treatment. Other limitations of studies include not controlling for lead-time bias208 and not accounting for important confounders, such as the level of adherence209 or physician experience.110

Four fair-quality observational studies controlled for lead-time bias by identifying cohorts of patients at initial CD4 cell count strata and evaluating outcomes according to when they received HAART.210-213 Three U.S. studies found no significant benefit associated with starting HAART at CD4 cell counts between 0.350 and 0.500 X 109 cells/L versus between 0.200 and 0.350 x 109 cells/L (Table 4).210,212,213 A Swiss study reported a benefit for starting at CD4 cell counts above 0.350 X 109 cells/L but did not stratify results of patients starting at CD4 cell counts above or below 0.200 X 109 cells/L.211 Six109,214-218 of 8209,219 other observational studies that did not control for lead-time bias or used novel methodologic approaches found a benefit or trend toward benefit from initiation of treatment at CD4 counts above versus below 0.350 x 109 cells/L.

A randomized clinical trial (the SMART [Strategies for Management of Anti-Retroviral Therapies] study)220 comparing viral suppression in asymptomatic patients with a CD4 cell count less than 0.350 X 109 cells/L with delay until counts decrease below 0.250 X 109 cells/L is in progress, with preliminary results expected in 5 to 7 years.221

What Are the Harms Associated with Antiretroviral Therapy?

Individual antiretroviral drugs, drug classes, and drug combinations are all associated with specific adverse event profiles.14 Retrospective U.S. cohort studies found that 61% of patients had changed or discontinued their initial HAART regimen by 8 months222 and that the median duration of the initial regimen was less than 2 years;223 40% to 50% discontinued the initial regimen because of adverse events. Many antiretroviral-associated adverse events, however, are short-term or self-limited, and effective alternatives can often be found.15,134 Detailed and regularly updated guidelines review adverse events associated with specific antiretroviral drugs, drug classes, and combinations.14 Certain drugs and combinations are not recommended because of associated adverse events.

A recent good-quality systematic review found that 26 of 54 trials of antiretroviral therapy reported drug-related withdrawals, a marker for intolerable or severe adverse events.133 Among trials comparing 3-drug and 2-drug regimens, dropout rates were similar if both regimens either included protease inhibitors or were protease inhibitor-sparing. In a large (n = 1,160), good-quality Swiss cohort study of adverse events in clinical practice, 47% of patients reported a clinical adverse event that was probably or definitely attributed to HAART within the previous 30 days.224 Among these, 9% were graded as serious or severe.

The use of HAART is associated with metabolic disturbances (lipodystrophy syndrome, hyperlipidemia, and diabetes) that are related to an increased risk for cardiovascular events.225,226 The largest prospective study on the risk for cardiovascular events associated with both protease inhibitor-based and non-protease inhibitor-based combination regimens was a good-quality study of 23,468 patients in 11 cohorts.227 It found that the incidence of myocardial infarction increased with longer exposure (adjusted relative rate per year of exposure, 1.26).227 The relative risk for the combined outcome of myocardial infarction, invasive cardiovascular procedures, or stroke was similarly increased, although the event rate was higher (5.7 events/1000 person-years vs. 3.5 events/1000 person-years for myocardial infarction alone).228 Other studies primarily evaluating the cardiovascular risk associated with protease inhibitors also generally found an increased risk.229-237

Studies evaluating trends over time reported mixed findings regarding the rate of cardiovascular events in HIV-infected patients since the introduction of HAART. These studies are limited by potential confounding from changes in clinical practice and the demographic characteristics of persons surviving with HIV infection.238-241

Estimates of the Numbers Needed To Screen and Treat

Table 5 estimates outcomes after 3 years from 1-time screening for HIV in 3 hypothetical cohorts of 10,000 asymptomatic persons (0.3% prevalence, 1% prevalence, and 5% to 15% prevalence [high risk]) (go to Appendix Table for base-case assumptions). Because no trials directly compare 3-drug regimens to placebo, we indirectly calculated (Appendix A) a relative risk for clinical progression or death of 0.35 (CI, 0.25 to 0.47).133 For all cohorts, the number of cases of clinical progression or deaths that were prevented greatly outweighed the number of cardiovascular adverse events caused by antiretroviral therapy. Evidence was insufficient to estimate the effects of screening on transmission rates.

What Is the Cost-Effectiveness of Screening for HIV Infection?

In 2 good-quality studies, the cost-effectiveness of one-time HIV screening in outpatients with 1% prevalence compared to no screening was $38,000 to $42,000 per quality-adjusted life-year.242,243 One of these studies found that the cost-effectiveness improved to $15,000 per quality-adjusted life-year when secondary transmission benefits were directly incorporated into cost-effectiveness ratios, and they remained less than $50,000 per quality-adjusted life-year even when screened populations had HIV prevalences substantially lower than seen in the general population.242 The other study, which did not directly incorporate secondary transmission benefits into cost-effectiveness ratios, found that the incremental cost-effectiveness of one-time screening in the general population was greater than $100,000 per quality-adjusted life-year.243

Neither study incorporated long-term cardiovascular risks associated with HAART into their models. The study by Sanders and colleagues found that the model was sensitive to the effects of screening on secondary transmission and the benefits of early identification and therapy.

The 1996 USPSTF guidelines recommended screening persons who report high-risk behaviors.11 Neither of the 2 reviewed studies evaluated the incremental cost-effectiveness of a strategy of screening only higher-risk persons compared to broader screening strategies in different populations. One of the studies found that the incremental cost-effectiveness of testing every 5 years compared to one-time screening exceeded $50,000 per quality-adjusted life-year.242

Return to Table of Contents

There is no direct evidence on benefits of screening for HIV infection in the general population. Other evidence obtained for the systematic review (summarized in Table 6) indicates that testing is extremely accurate, a high proportion of patients receive a diagnosis at immunologically advanced stages of disease, and interventions (particularly HAART) are effective in reducing morbidity and mortality in patients with immunologically advanced disease. Although long-term HAART is associated with cardiovascular complications, absolute rates are low.

Reasonable screening strategies might be to screen patients with acknowledged risk factors, all patients in settings with a higher prevalence of HIV infection, or all patients in the general population. Studies that have assessed risk factor assessment to guide screening indicate that targeted screening misses a substantial proportion of HIV-positive patients. On the other hand, universal screening would result in large numbers of patients screened for each clinical outcome prevented.

An important gap in the literature is the inadequate evidence with which to accurately estimate the benefits from identification of HIV-positive patients at earlier stages of disease who do not initially qualify for HAART, particularly since screening could lead to higher rates of earlier diagnosis. In these patients, other interventions, such as counseling to reduce transmission, assume greater relative importance. Despite evidence that knowledge of HIV-positive status reduces some high-risk behaviors, there is insufficient evidence with which to accurately estimate the effects on transmission rates. The relationship between HAART use and beliefs, risky behaviors, and transmission rates also needs to be explored further. The case for screening, particularly in lower-risk populations, would be greatly strengthened by studies showing that identification at earlier stages of disease is associated with decreased transmission rates. When available, results of the SMART trial221 will provide important information about the effectiveness of HAART in asymptomatic patients with higher CD4 cell counts.

Other studies are needed on methods to improve risk assessment, effects of streamlined or targeted counseling, methods to improve entry into medical care and uptake of recommended interventions, and effects of newer testing and sampling methods. In addition, data with which to estimate the magnitude of screening harms and on methods to minimize their risk are limited. Continued attention to adverse events as patients continue receiving HAART will help clarify long-term risks.

Despite continuing HIV education efforts and the availability of effective interventions, incidence of HIV remains steady in the United States, and HIV infection continues to place an enormous burden on the health care system. Further implementation and evaluation of screening programs could have an important impact on the morbidity and mortality associated with this disease.

Return to Table of Contents

Acknowledgments: This review of the evidence was funded by the Agency for Healthcare Research and Quality (AHRQ) for the U.S. Preventive Services Task Force (USPSTF). The investigators acknowledge the contributions of Kim Villemyer for her help in preparing the full evidence report and the manuscript; Christina Bougatsos for her help in preparing the manuscript; and Andrew Hamilton, M.L.S., M.S., for conducting the literature searches. They also thank Mark Helfand, M.D., M.P.H.; Heidi D. Nelson, M.D., M.P.H.; David Lanier, M.D.; members of the U.S. Preventive Services Task Force; and reviewers for their contributions to this project.

Grant Support: This study was conducted by the Oregon Evidence-based Practice Center under contract to the Agency for Healthcare Research and Quality (AHRQ). Contract No. 290-02-0024, Rockville, MD.

Potential Financial Conflicts of Interest: None disclosed.

Return to Table of Contents

1. Fleming P, Byers RH, Sweeney PA, Daniels D, Karon JM, Janssen RS. HIV prevalence in the United States, 2000 [Abstract]. In: Program and Abstracts of the 9th Conference on Retroviruses and Opportunistic Infections, Seattle, Washington, 24-28 February 2002. Alexandria, Virginia: Foundation for Retrovirology and Human Health;2002.
2. 1993 revised classification system for HIV infection and expanded surveillance case definition for AIDS among adolescents and adults. MMWR Recomm Rep 1992;41:1-19.
3. HIV/AIDS Surveillance Report. Atlanta: Centers for Disease Control and Prevention;2003 (vol 15). Accessed at www.cdc.gov/hiv/stats/2003SurveillanceReport.pdf on 21 March 2005.
4. Kochanek KD, Smith BL. Deaths: preliminary data for 2002. National Vital Statistics Reports. Hyattsville, MD: National Center for Health Statistics;2004:52(no. 13).
5. Increases in HIV diagnoses—29 states, 1999-2002. MMWR Morb Mortal Wkly Rep 2003;52:1145-8. [PMID: 14647015].
6. Rosenberg PS, Biggar RJ, Goedert JJ. Declining age at HIV infection in the United States [Letter]. N Engl J Med 1994;330:789-90.
7. Mellors JW, Munoz A, Giorgi JV, Margolick JB, Tassoni CJ, Gupta P, et al. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med 1997;126:946-54.
8. Chene G, Sterne JA, May M, Costagliola D, Ledergerber B, Phillips AN, et al. Prognostic importance of initial response in HIV-1 infected patients starting potent antiretroviral therapy: analysis of prospective studies. Lancet 2003;362:679-86.
9. Phillips A. Short-term risk of AIDS according to current CD4 cell count and viral load in antiretroviral drug-naive individuals and those treated in the monotherapy era. AIDS 2004;18:51-8.
10. Mellors JW, Kingsley LA, Rinaldo CR Jr, Todd JA, Hoo BS, Kokka RP, et al. Quantitation of HIV-1 RNA in plasma predicts outcome after seroconversion. Ann Intern Med 1995;122:573-9.
11. U.S. Preventive Services Task Force. Guide to Clinical Preventive Services. 2nd ed. Alexandria, VA: International Medical Publishing. 1996.
12. Chou R, Smits AK, Huffman LH, Fu R, Korthuis PT. Prenatal screening for HIV: a review of the evidence for the U.S. Preventive Services Task Force. Ann Intern Med 2005;143:000-000.
13. Chou R, Huffman LH, Fu R, Smits AK, Korthuis PT. Screening for human immunodeficiency virus in adolescents and adults: systematic evidence synthesis. Rockville, MD: Agency for Healthcare Research and Quality;2005. 
14. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Bethesda, MD: U.S. Department of Health and Human Services;7 April 2005. Accessed at www.aidsinfo.nih.gov/guidelines/adult/aa_040705.pdf.
15. Yeni PG, Hammer SM, Hirsch MS, Saag MS, Schechter M, Carpenter CC, et al. Treatment for adult HIV infection: 2004 recommendations of the International AIDS Society—USA Panel. JAMA 2004;292:251-65.
16. Revised guidelines for HIV counseling, testing, and referral. MMWR Recomm Rep 2001;50:1-57;quiz CE1-19a1-CE6-19a1.
17. Kaplan JE, Masur H, Holmes KK. Guidelines for preventing opportunistic infections among HIV-infected persons—2002. Recommendations of the U.S. Public Health Service and the Infectious Diseases Society of America. MMWR Recomm Rep 2002;51:1-52.
18. Harris RP, Helfand M, Woolf SH, Lohr KN, Mulrow CD, Teutsch SM, et al. Current methods of the U.S. Preventive Services Task Force: a review of the process. Am J Prev Med 2001;20:21-35.
19. Saha S, Hoerger TJ, Pignone MP, Teutsch SM, Helfand M, Mandelblatt JS, et al. The art and science of incorporating cost effectiveness into evidence-based recommendations for clinical preventive services. Am J Prev Med 2001;20:36-43.
20. HIV transmission risk behavior among men and women living with HIV in 4 cities in the United States. J Acquir Immune Defic Syndr 2004;36:1057-66.
21. Prevalence of risk behaviors for HIV infection among adults—United States, 1997. MMWR Morb Mortal Wkly Rep 2001;50:262-5.
22. Murphy DA, Durako SJ, Moscicki AB, Vermund SH, Ma Y, Schwarz DF, et al. No change in health risk behaviors over time among HIV infected adolescents in care: role of psychological distress. J Adolesc Health 2001;29:57-63.
23. Abma JC, Sonenstein FL. Sexual activity and contraceptive practices among teenagers in the United States, 1988 and 1995. Vital Health Stat 23. 2001:1-79.
24. High-risk sexual behavior by HIV-positive men who have sex with men—16 sites, United States, 2000-2002. MMWR Morb Mortal Wkly Rep 2004;53:891-4.
25. HIV Testing Survey, 2002. HIV/AIDS Special Surveillance Report 5. Atlanta: Centers for Disease Control and Prevention;2004. Accessed at www.cdc.gov/hiv/stats/HIV-Test-Survey2002.pdf on 21 March 2005.
26. Klein D, Hurley LB, Merrill D, Quesenberry CP Jr. Review of medical encounters in the 5 years before a diagnosis of HIV-1 infection: implications for early detection. J Acquir Immune Defic Syndr 2003;32:143-52.
27. Liddicoat RV, Horton NJ, Urban R, Maier E, Christiansen D, Samet JH. Assessing missed opportunities for HIV testing in medical settings. J Gen Intern Med 2004;19:349-56.
28. Peterman TA, Todd KA, Mupanduki I. Opportunities for targeting publicly funded human immunodeficiency virus counseling and testing. J Acquir Immune Defic Syndr Hum Retrovirol 1996;12:69-74.
29. Groseclose SL, Erickson B, Quinn TC, Glasser D, Campbell CH, Hook EW 3rd. Characterization of patients accepting and refusing routine, voluntary HIV antibody testing in public sexually transmitted disease clinics. Sex Transm Dis 1994;21:31-5.
30. Erickson B, Wasserheit JN, Rompalo AM, Brathwaite W, Glasser D, Hook EW 3rd. Routine voluntary HIV screening in STD clinic clients: characterization of infected clients. Sex Transm Dis 1990;17:194-9.
31. Kassler WJ, Zenilman JM, Erickson B, Fox R, Peterman TA, Hook EW 3rd. Seroconversion in patients attending sexually transmitted disease clinics. AIDS 1994;8:351-5.
32. Alpert PL, Shuter J, DeShaw MG, Webber MP, Klein RS. Factors associated with unrecognized HIV-1 infection in an inner-city emergency department. Ann Emerg Med 1996;28:159-64.
33. D'Angelo LJ, Getson PR, Luban NL, Gayle HD. Human immunodeficiency virus infection in urban adolescents: can we predict who is at risk? Pediatrics 1991;88:982-6.
34. Harris RL, Boisaubin EV, Salyer PD, Semands DF. Evaluation of a hospital admission HIV antibody voluntary screening program. Infect Control Hosp Epidemiol 1990;11:628-34.
35. Asch SM, London AS, Barnes PF, Gelberg L. Testing for human immunodeficiency virus infection among tuberculosis patients in Los Angeles. Am J Respir Crit Care Med 1997;155:378-81.
36. Theuer CP, Hopewell PC, Elias D, Schecter GF, Rutherford GW, Chaisson RE. Human immunodeficiency virus infection in tuberculosis patients. J Infect Dis 1990;162:8-12.
37. Chen Z, Branson B, Ballenger A, Peterman TA. Risk assessment to improve targeting of HIV counseling and testing services for STD clinic patients. Sex Transm Dis 1998;25:539-43.
38. Kirkland KB, Meriwether RA, MacKenzie WR, Binz WC, Allen RJ, Veenhuis PE. Clinician judgement as a tool for targeting HIV counseling and testing in North Carolina state mental hospitals, 1994. AIDS Patient Care STDS 1999;13:473-9.
39. Kelen GD, Hexter DA, Hansen KN, Humes R, Vigilance PN, Baskerville M, et al. Feasibility of an emergency department-based, risk-targeted voluntary HIV screening program. Ann Emerg Med 1996;27:687-92.
40. Voluntary HIV testing as part of routine medical care—Massachusetts, 2002. MMWR Morb Mortal Wkly Rep 2004;53:523-6.
41. Routinely recommended HIV testing at an urban urgent-care clinic—Atlanta, Georgia, 2000. MMWR Morb Mortal Wkly Rep 2001;50:538-41.
42. Walensky RP, Losina E, Steger-Craven KA, Freedberg KA. Identifying undiagnosed human immunodeficiency virus: the yield of routine, voluntary inpatient testing. Arch Intern Med 2002;162:887-92.
43. Goggin MA, Davidson AJ, Cantril SV, O'Keefe LK, Douglas JM. The extent of undiagnosed HIV infection among emergency department patients: results of a blinded seroprevalence survey and a pilot HIV testing program. J Emerg Med 2000;19:13-9.
44. From the Centers for Disease Control. Interpretation and use of the western blot assay for serodiagnosis of human immunodeficiency virus type 1 infections. JAMA 1989;262:3395-7.
45. Update: serologic testing for HIV-1 antibody—United States, 1988 and 1989. MMWR Morb Mortal Wkly Rep 1990;39:380-3.
46. Update: serologic testing for antibody to human immunodeficiency virus. MMWR Morb Mortal Wkly Rep 1988;36:833-40, 845.
47. MacDonald KL, Jackson JB, Bowman RJ, Polesky HF, Rhame FS, Balfour HH Jr, et al. Performance characteristics of serologic tests for human immunodeficiency virus type 1 (HIV-1) antibody among Minnesota blood donors. Public health and clinical implications. Ann Intern Med 1989;110:617-21.
48. Kleinman S, Busch MP, Hall L, Thomson R, Glynn S, Gallahan D, et al. False-positive HIV-1 test results in a low-risk screening setting of voluntary blood donation. Retrovirus Epidemiology Donor Study. JAMA 1998;280:1080-5.
49. Donovan BJ, Rublein JC, Leone PA, Pilcher CD. HIV infection: point-of-care testing. Ann Pharmacother 2004;38:670-6.
50. Uni-Gold Recombigen HIV [Package insert;#045-138]. Bray, Ireland: Trinity Biotech Plc.;rev. 03/04.
51. OraQuick Rapid HIV-1 Antibody Test [Package insert;#3001-0951]. Bethlehem, PA: OraSure Technologies;rev. 10/03.
52. Reveal Rapid HIV-1 Antibody Test [Package insert;#FDAINS0065]. Halifax, Nova Scotia, Canada: MedMira Laboratories;rev. 0/1.
53. Bulterys M, Jamieson DJ, O'Sullivan MJ, Cohen MH, Maupin R, Nesheim S, et al. Rapid HIV-1 testing during labor: a multicenter study. JAMA 2004;292:219-23.
54. O'Connell RJ, Merritt TM, Malia JA, VanCott TC, Dolan MJ, Zahwa H, et al. Performance of the OraQuick rapid antibody test for diagnosis of human immunodeficiency virus type 1 infection in patients with various levels of exposure to highly active antiretroviral therapy. J Clin Microbiol 2003;41:2153-5.
55. Reynolds SJ, Ndongala LM, Luo CC, Mwandagalirwa K, Losoma AJ, Mwamba KJ, et al. Evaluation of a rapid test for the detection of antibodies to human immunodeficiency virus type 1 and 2 in the setting of multiple transmitted viral subtypes. Int J STD AIDS 2002;13:171-3.
56. Protocols for confirmation of reactive rapid HIV tests. MMWR Morb Mortal Wkly Rep 2004;53(10):221-2.
57. Rapid HIV antibody testing during labor and delivery for women of unknown HIV status. Atlanta: Centers for Disease Control and Prevention;30 January 2004. Accessed at www.cdc.gov/hiv/rapid_testing/materials/Labor&DeliveryRapidTesting.pdf on 20 July 2004.
58. Gallo D, George JR, Fitchen JH, Goldstein AS, Hindahl MS. Evaluation of a system using oral mucosal transudate for HIV-1 antibody screening and confirmatory testing. OraSure HIV Clinical Trials Group. JAMA 1997;277:254-8.
59. Granade TC, Phillips SK, Parekh B, Gomez P, Kitson-Piggott W, Oleander H, et al. Detection of antibodies to human immunodeficiency virus type 1 in oral fluids: a large-scale evaluation of immunoassay performance. Clin Diagn Lab Immunol 1998;5:171-5.
60. HIV assays: operational characteristics (phase I)— urine and oral fluid (saliva) specimens. Geneva, Switzerland: World Health Organization;January 2002. Available at www.who.int/diagnostics_laboratory/publications/en/hiv_assays_rep_13.pdf.
61. Martinez PM, Torres AR, Ortiz de Lejarazu R, Montoya A, Martin JF, Eiros JM. Human immunodeficiency virus antibody testing by enzyme-linked fluorescent and western blot assays using serum, gingival-crevicular transudate, and urine samples. J Clin Microbiol 1999;37:1100-6.
62. Schopper D, Vercauteren G. Testing for HIV at home: what are the issues? [Editorial] AIDS 1996;10:1455-65.
63. Desai S, Bates H, Michalski FJ. Detection of antibody to HIV-1 in urine [Letter]. Lancet 1991;337:183-4.
64. Frank AP, Wandell MG, Headings MD, Conant MA, Woody GE, Michel C. Anonymous HIV testing using home collection and telemedicine counseling. A multicenter evaluation. Arch Intern Med 1997;157:309-14.
65. Spielberg F, Critchlow C, Vittinghoff E, Coletti AS, Sheppard H, Mayer KH, et al. Home collection for frequent HIV testing: acceptability of oral fluids, dried blood spots and telephone results. HIV Early Detection Study Group. AIDS 2000;14:1819-28.
66. Kaplan EH, Satten GA. Repeat screening for HIV: when to test and why. J Acquir Immune Defic Syndr 2000;23:339-45.
67. Mylonakis E, Paliou M, Greenbough TC, Flaningan TP, Letvin NL, Rich JD. Report of a false-positive HIV test result and the potential use of additional tests in establishing HIV serostatus. Arch Intern Med 2000;160:2386-8.
68. Wai CT, Tambyah PA. False-positive HIV-1 ELISA in patients with hepatitis B [Letter]. Am J Med 2002;112:737.
69. Sayre KR, Dodd RY, Tegtmeier G, Layug L, Alexander SS, Busch MP. False-positive human immunodeficiency virus type 1 western blot tests in noninfected blood donors. Transfusion 1996;36:45-52.
70. Gielen AC, O'Campo P, Faden RR, Eke A. Women's disclosure of HIV status: experiences of mistreatment and violence in an urban setting. Women Health 1997;25:19-31.
71. Herek GM, Capitanio JP, Widaman KF. HIV-related stigma and knowledge in the United States: prevalence and trends, 1991-1999. Am J Public Health 2002;92:371-7.
72. HIV-related knowledge and stigma—United States, 2000. MMWR Morb Mortal Wkly Rep 2000;49:1062-4.
73. Kilmarx PH, Hamers FF, Peterman TA. Living with HIV. Experiences and perspectives of HIV-infected sexually transmitted disease clinic patients after posttest counseling. Sex Transm Dis 1998;25:28-37.
74. Perry SW, Jacobsberg LB, Fishman B, Weiler PH, Gold JW, Frances AJ. Psychological responses to serological testing for HIV. AIDS 1990;4:145-52.
75. Rundell JR, Kyle KM, Brown GR, Thomason JL. Risk factors for suicide attempts in a human immunodeficiency virus screening program. Psychosomatics 1992;33:24-7.
76. Marzuk PM, Tierney H, Tardiff K, Gross EM, Morgan EB, Hsu MA, et al. Increased risk of suicide in persons with AIDS JAMA 1988;259:1333-7.
77. van Haastrecht HJ, Mientjes GH, van den Hoek AJ, Coutinho RA. Death from suicide and overdose among drug injectors after disclosure of first HIV test result. AIDS 1994;8:1721-5.
78. Cote TR, Biggar RJ, Dannenberg AL. Risk of suicide among persons with AIDS A national assessment. JAMA 1992;268:2066-8.
79. Dannenberg AL, McNeil JG, Brundage JF, Brookmeyer R. Suicide and HIV infection. Mortality follow-up of 4147 HIV-seropositive military service applicants. JAMA 1996;276:1743-6.
80. Perry S, Fishman B, Jacobsberg L, Young J, Frances A. Effectiveness of psychoeducational interventions in reducing emotional distress after human immunodeficiency virus antibody testing. Arch Gen Psychiatry 1991;48:143-7.
81. Chesney MA, Chambers DB, Taylor JM, Johnson LM, Folkman S. Coping effectiveness training for men living with HIV: results from a randomized clinical trial testing a group-based intervention. Psychosom Med 2003;65:1038-46.
82. Antoni MH, Cruess DG, Cruess S, Lutgendorf S, Kumar M, Ironson G, et al. Cognitive-behavioral stress management intervention effects on anxiety, 24-hr urinary norepinephrine output, and T-cytotoxic/suppressor cells over time among symptomatic HIV-infected gay men. J Consult Clin Psychol 2000;68:31-45.
83. Cruess DG, Antoni MH, Schneiderman N, Ironson G, McCabe P, Fernandez JB, et al. Cognitive-behavioral stress management increases free testosterone and decreases psychological distress in HIV-seropositive men. Health Psychol 2000;19:12-20.
84. Vlahov D, Wientge D, Moore J, et al. Violence among women with or at risk for HIV infection. AIDS and Behavior 1998;2:53-60.
85. Koenig LJ, Moore J. Women, violence, and HIV: a critical evaluation with implications for HIV services. Matern Child Health J 2000;4:103-9.
86. Cohen M, Deamant C, Barkan S, Richardson J, Young M, Holman S, et al. Domestic violence and childhood sexual abuse in HIV-infected women and women at risk for HIV. Am J Public Health 2000;90:560-5.
87. Kissinger PJ, Niccolai LM, Magnus M, Farley TA, Maher JE, Richardson-Alston G, et al. Partner notification for HIV and syphilis: effects on sexual behaviors and relationship stability. Sex Transm Dis 2003;30:75-82.
88. Schnell DJ, Higgins DL, Wilson RM, Goldbaum G, Cohn DL, Wolitski RJ. Men's disclosure of HIV test results to male primary sex partners. Am J Public Health 1992;82:1675-6.
89. Hoxworth T, Spencer NE, Peterman TA, Craig T, Johnson S, Maher JE. Changes in partnerships and HIV risk behaviors after partner notification. Sex Transm Dis 2003;30:83-8.
90. Number of persons tested for HIV—United States, 2002. MMWR Morb Mortal Wkly Rep 2004;53:1110-3.
91. Abma JC, Chandra A, Mosher W, Peterson LS, Piccinino LJ. Fertility, family planning, and women's health: new data from the 1995 National Survey of Family Growth. Vital Health Stat 1997;May:1-114.
92. Anderson JE, Carey JW, Taveras S. HIV testing among the general US population and persons at increased risk: information from national surveys, 1987-1996. Am J Public Health 2000;90:1089-95.
93. Kellerman SE, Lehman JS, Lansky A, Stevens MR, Hecht FM, Bindman AB, et al. HIV testing within at-risk populations in the United States and the reasons for seeking or avoiding HIV testing. J Acquir Immune Defic Syndr 2002;31:202-10.
94. Irwin KL, Valdiserri RO, Holmberg SD. The acceptability of voluntary HIV antibody testing in the United States: a decade of lessons learned. AIDS 1996;10:1707-17.
95. Stanley B, Fraser J, Cox NH. Uptake of HIV screening in genitourinary medicine after change to "opt-out" consent. BMJ 2003;326:1174.
96. Hirano D, Gellert GA, Fleming K, Boyd D, Englender SJ, Hawks H. Anonymous HIV testing: the impact of availability on demand in Arizona. Am J Public Health 1994;84:2008-10.
97. Fehrs LJ, Fleming D, Foster LR, McAlister RO, Fox V, Modesitt S, et al. Trial of anonymous versus confidential human immunodeficiency virus testing. Lancet 1988;2:379-82.
98. Hertz-Picciotto I, Lee LW, Hoyo C. HIV test-seeking before and after the restriction of anonymous testing in North Carolina. Am J Public Health 1996;86:1446-50.
99. Bindman AB, Osmond D, Hecht FM, Lehman JS, Vranizan K, Keane D, et al. Multistate evaluation of anonymous HIV testing and access to medical care. Multistate Evaluation of Surveillance of HIV (MESH) Study Group. JAMA 1998;280:1416-20.
100. Hoxworth T, Hoffman R, Cohn D, Davidson A. Anonymous HIV testing: does it attract clients who would not seek confidential testing? AIDS Public Policy J 1994;9:182-9.
101. Nakashima AK, Horsley R, Frey RL, Sweeney PA, Weber JT, Fleming PL. Effect of HIV reporting by name on use of HIV testing in publicly funded counseling and testing programs. JAMA 1998;280:1421-6.
102. Castrucci BC, Williams DE, Foust E. The elimination of anonymous HIV testing: a case study in North Carolina. J Public Health Manag Pract 2002;8:30-7.
103. Meehan TM, Hansen H, Klein WC. The impact of parental consent on the HIV testing of minors. Am J Public Health 1997;87:1338-41.
104. Kendrick SR, Kroc KA, Couture E, Weinstein RA. Comparison of point-of-care rapid HIV testing in three clinical venues. AIDS 2004;18:2208-10.
105. Pugatch DL, Levesque BG, Lally MA, Reinert SE, Filippone WJ, Combs CM, et al. HIV testing among young adults and older adolescents in the setting of acute substance abuse treatment. J Acquir Immune Defic Syndr 2001;27:135-42.
106. Branson BM. Home sample collection tests for HIV infection. JAMA 1998;280:1699-701.
107. McQuitty M, McFarland W, Kellogg TA, White E, Katz MH. Home collection versus publicly funded HIV testing in San Francisco: who tests where? J Acquir Immune Defic Syndr 1999;21:417-22.
108. Sy FS, Rhodes SD, Choi ST, Drociuk D, Laurent AA, Naccash RM, et al. The acceptability of oral fluid testing for HIV antibodies. A pilot study in gay bars in a predominantly rural state. Sex Transm Dis 1998;25:211-5.
109. Egger M, May M, Chene G, Phillips AN, Ledergerber B, Dabis F, et al. Prognosis of HIV-1-infected patients starting highly active antiretroviral therapy: a collaborative analysis of prospective studies. Lancet 2002;360:119-29.
110. Wood E, Hogg RS, Yip B, Harrigan PR, O'Shaughnessy MV, Montaner JS. Is there a baseline CD4 cell count that precludes a survival response to modern antiretroviral therapy? AIDS 2003;17:711-20.
111. Kazempour K, Kammerman LA, Farr SS. Survival effects of ZDV, ddI, and ddC in patients with CD4 < or = 50 cells/mm3. J Acquir Immune Defic Syndr Hum Retrovirol 1995;10 Suppl 2:S97-106.
112. Samet JH, Freedberg KA, Savetsky JB, Sullivan LM, Stein MD. Understanding delay to medical care for HIV infection: the long-term non-presenter. AIDS 2001;15:77-85.
113. Katz MH, Bindman AB, Keane D, Chan AK. CD4 lymphocyte count as an indicator of delay in seeking human immunodeficiency virus-related treatment. Arch Intern Med 1992;152:1501-4.
114. Luby S, Jones J, Horan J. Using CD4 counts to evaluate the stages and epidemiology of HIV infection in South Carolina public clinic patients. Am J Public Health 1994;84:377-81.
115. Hutchinson CM, Wilson C, Reichart CA, Marsiglia VC, Zenilman JM, Hook EW 3rd. CD4 lymphocyte concentrations in patients with newly identified HIV infection attending STD clinics. Potential impact on publicly funded health care resources. JAMA 1991;266:253-6.
116. Dybul M, Bolan R, Condoluci D, Cox-Iyamu R, Redfield R, Hallahan CW, et al. Evaluation of initial CD4+ T cell counts in individuals with newly diagnosed human immunodeficiency virus infection, by sex and race, in urban settings. J Infect Dis 2002;185:1818-21.
117. HIV counseling and testing in publicly funded sites. Annual Report 1997 and 1998. Atlanta: Centers for Disease Control and Prevention;2001. Accessed at www.cdc.gov/hiv/pubs/cts98.pdf on 3 March 2005.
118. Failure to return for HIV test results among persons at high risk for HIV infection: results from a multistate interview project. J Acquir Immune Defic Syndr 2004;35:511-518.
119. Molitor F, Bell RA, Truax SR, Ruiz JD, Sun RK. Predictors of failure to return for HIV test result and counseling by test site type. AIDS Educ Prev 1999;11:1-13.
120. Hightow LB, Miller WC, Leone PA, Wohl D, Smurzynski M, Kaplan AH. Failure to return for HIV posttest counseling in an STD clinic population. AIDS Educ Prev 2003;15:282-90.
121. Kassler WJ. Advances in HIV testing technology and their potential impact on prevention. AIDS Educ Prev 1997;9:27-40.
122. Kelen GD, Shahan JB, Quinn TC. Emergency department-based HIV screening and counseling: experience with rapid and standard serologic testing. Ann Emerg Med 1999;33:147-55.
123. Keenan PA, Keenan JM. Rapid hiv testing in urban outreach: a strategy for improving posttest counseling rates. AIDS Educ Prev 2001;13:541-50.
124. Bozzette SA, Berry SH, Duan N, Frankel MR, Leibowitz AA, Lefkowitz D, et al. The care of HIV-infected adults in the United States. HIV Cost and Services Utilization Study Consortium. N Engl J Med 1998;339:1897-904.
125. Turner BJ, Cunningham WE, Duan N, Andersen RM, Shapiro MF, Bozzette SA, et al. Delayed medical care after diagnosis in a US national probability sample of persons infected with human immunodeficiency virus. Arch Intern Med 2000;160:2614-22.
126. Supplement to HIV/AIDS surveillance (SHAS): demographics and behavioral data from a supplemental HIV/AIDS behavioral surveillance project 1997-2000. Special Surveillance Report No. 2. Atlanta: Centers for Disease Control and Prevention;2004:1-27.
127. Osmond DH, Bindman AB, Vranizan K, Lehman JS, Hecht FM, Keane D, et al. Name-based surveillance and public health interventions for persons with HIV infection. Multistate Evaluation of Surveillance for HIV Study Group. Ann Intern Med 1999;131:775-9.
128. Samet JH, Freedberg KA, Stein MD, Lewis R, Savetsky J, Sullivan L, et al. Trillion virion delay: time from testing positive for HIV to presentation for primary care. Arch Intern Med 1998;158:734-40.
129. Stall R, Pollack L, Mills TC, Martin JN, Osmond D, Paul J, et al. Use of antiretroviral therapies among HIV-infected men who have sex with men: a household-based sample of 4 major American cities. Am J Public Health 2001;91:767-73.
130. Cunningham WE, Markson LE, Andersen RM, Crystal SH, Fleishman JA, Golin C, et al. Prevalence and predictors of highly active antiretroviral therapy use in patients with HIV infection in the United States. HCSUS Consortium. HIV Cost and Services Utilization. J Acquir Immune Defic Syndr 2000;25:115-23.
131. Kaplan JE, Parham DL, Soto-Torres L, van Dyck K, Greaves JA, Rauch K, et al. Adherence to guidelines for antiretroviral therapy and for preventing opportunistic infections in HIV-infected adults and adolescents in Ryan White-funded facilities in the United States. J Acquir Immune Defic Syndr 1999;21:228-35.
132. McNaghten AD, Hanson DL, Dworkin MS, Jones JL. Differences in prescription of antiretroviral therapy in a large cohort of HIV-infected patients. J Acquir Immune Defic Syndr 2003;32:499-505.
133. Jordan R, Gold L, Cummins C, Hyde C. Systematic review and meta-analysis of evidence for increasing numbers of drugs in antiretroviral combination therapy. BMJ 2002;324:757.
134. Gulick RM, Meibohm A, Havlir D, Eron JJ, Mosley A, Chodakewitz JA, et al. Six-year follow-up of HIV-1-infected adults in a clinical trial of antiretroviral therapy with indinavir, zidovudine, and lamivudine. AIDS 2003;17:2345-9.
135. Kaufmann GR, Perrin L, Pantaleo G, Opravil M, Furrer H, Telenti A, et al. CD4 T-lymphocyte recovery in individuals with advanced HIV-1 infection receiving potent antiretroviral therapy for 4 years: the Swiss HIV Cohort Study. Arch Intern Med 2003;163:2187-95.
136. Garcia F, De Lazzari E, Plana M, Castro P, Mestre G, Nomdedeu M, et al. Long-term CD4+ T-Cell Response to Highly Active Antiretroviral Therapy According to Baseline CD4+ T-Cell Count. J Acquir Immune Defic Syndr 2004;36:702-13.
137. AIDS cases, deaths, and persons living with AIDS by year, 1985-2002—United States. Atlanta: Centers for Disease Control and Prevention;2002. Accessed at www.cdc.gov/hiv/stats/hasr1402.htm on 2 December 2004.
138. Palella FJ Jr, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998;338:853-60.
139. McNaghten AD, Hanson DL, Jones JL, Dworkin MS, Ward JW. Effects of antiretroviral therapy and opportunistic illness primary chemoprophylaxis on survival after AIDS diagnosis. Adult/Adolescent Spectrum of Disease Group. AIDS 1999;13:1687-95.
140. Moore RD, Chaisson RE. Natural history of HIV infection in the era of combination antiretroviral therapy. AIDS 1999;13:1933-42.
141. Egger M, Hirschel B, Francioli P, Sudre P, Wirz M, Flepp M, et al. Impact of new antiretroviral combination therapies in HIV infected patients in Switzerland: prospective multicentre study. Swiss HIV Cohort Study. BMJ 1997;315:1194-9.
142. Mocroft A, Vella S, Benfield TL, Chiesi A, Miller V, Gargalianos P, et al. Changing patterns of mortality across Europe in patients infected with HIV-1. EuroSIDA Study Group. Lancet 1998;352:1725-30.
143. Pezzotti P, Napoli PA, Acciai S, Boros S, Urciuoli R, Lazzeri V, et al. Increasing survival time after AIDS in Italy: the role of new combination antiretroviral therapies. Tuscany AIDS Study Group. AIDS 1999;13:249-55.
144. Vittinghoff E, Scheer S, O'Malley P, Colfax G, Holmberg SD, Buchbinder SP. Combination antiretroviral therapy and recent declines in AIDS incidence and mortality. J Infect Dis 1999;179:717-20.
145. Detels R, Tarwater P, Phair JP, Margolick J, Riddler SA, Munoz A, et al. Effectiveness of potent antiretroviral therapies on the incidence of opportunistic infections before and after AIDS diagnosis. AIDS 2001;15:347-55.
146. Lee LM, Karon JM, Selik R, Neal JJ, Fleming PL. Survival after AIDS diagnosis in adolescents and adults during the treatment era, United States, 1984-1997. JAMA 2001;285:1308-15.
147. Tarwater PM, Mellors J, Gore ME, Margolick JB, Phair J, Detels R, et al. Methods to assess population effectiveness of therapies in human immunodeficiency virus incident and prevalent cohorts. Am J Epidemiol 2001;154:675-81.
148. Louie JK, Hsu LC, Osmond DH, Katz MH, Schwarcz SK. Trends in causes of death among persons with acquired immunodeficiency syndrome in the era of highly active antiretroviral therapy, San Francisco, 1994-1998. J Infect Dis 2002;186:1023-7.
149. Update: AIDS—United States, 2000. MMWR Morb Mortal Wkly Rep 2002;51:592-5.
150. Revicki DA, Moyle G, Stellbrink HJ, Barker C. Quality of life outcomes of combination zalcitabine-zidovudine, saquinavir-zidovudine, and saquinavir-zalcitabine-zidovudine therapy for HIV-infected adults with CD4 cell counts between 50 and 350 per cubic millimeter. PISCES (SV14604) Study Group. AIDS 1999;13:851-8.
151. Bucciardini R, Wu AW, Floridia M, Fragola V, Ricciardulli D, Tomino C, et al. Quality of life outcomes of combination zidovudine-didanosine-nevirapine and zidovudine-didanosine for antiretroviral-naive advanced HIV-infected patients. AIDS 2000;14:2567-74.
152. Nieuwkerk PT, Gisolf EH, Colebunders R, Wu AW, Danner SA, Sprangers MA. Quality of life in asymptomatic and symptomatic HIV-infected patients in a trial of ritonavir/saquinavir therapy. The Prometheus Study Group. AIDS 2000;14:181-7.
153. Coplan PM, Cook JR, Carides GW, Heyse JF, Wu AW, Hammer SM, et al. Impact of indinavir on the quality of life in patients with advanced HIV infection treated with zidovudine and lamivudine. Clin Infect Dis 2004;39:426-33.
154. Quinn TC, Wawer MJ, Sewankambo N, Serwadda D, Li C, Wabwire-Mangen F, et al. Viral load and heterosexual transmission of human immunodeficiency virus type 1. Rakai Project Study Group. N Engl J Med 2000;342:921-9.
155. Murphy G, Charlett A, Jordan LF, Osner N, Gill ON, Parry JV. HIV incidence appears constant in men who have sex with men despite widespread use of effective antiretroviral therapy. AIDS 2004;18:265-72.
156. Do AN, Hanson DL, Dworkin MS, Jones JL. Risk factors for and trends in gonorrhea incidence among persons infected with HIV in the United States. AIDS 2001;15:1149-55.
157. Katz MH, Schwarcz SK, Kellogg TA, Klausner JD, Dilley JW, Gibson S, et al. Impact of highly active antiretroviral treatment on HIV seroincidence among men who have sex with men: San Francisco. Am J Public Health 2002;92:388-94.
158. Scheer S, Chu PL, Klausner JD, Katz MH, Schwarcz SK. Effect of highly active antiretroviral therapy on diagnoses of sexually transmitted diseases in people with AIDS. Lancet 2001;357:432-5.
159. Crepaz N, Hart TA, Marks G. Highly active antiretroviral therapy and sexual risk behavior: a meta-analytic review. JAMA 2004;292:224-36.
160. Musicco M, Lazzarin A, Nicolosi A, Gasparini M, Costigliola P, Arici C, et al. Antiretroviral treatment of men infected with human immunodeficiency virus type 1 reduces the incidence of heterosexual transmission. Italian Study Group on HIV Heterosexual Transmission. Arch Intern Med 1994;154:1971-6.
161. Holtgrave DR. Estimation of annual HIV transmission rates in the United States, 1978-2000. J Acquir Immune Defic Syndr 2004;35:89-92.
162. Janssen RS, Valdiserri RO. HIV Prevention in the United States: increasing emphasis on working with those living with HIV. J Acquir Immune Defic Syndr 2004;37 Suppl 2:S119-21.
163. Janssen RS, Holtgrave DR, Valdiserri RO, Shepherd M, Gayle HD, De Cock KM. The serostatus approach to fighting the HIV epidemic: prevention strategies for infected individuals. Am J Public Health 2001;91:1019-24.
164. Incorporating HIV prevention into the medical care of persons living with HIV. Recommendations of CDC, the Health Resources and Services Administration, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America. MMWR Recomm Rep 2003;52:1-24.
165. Weller S, Davis K. Condom effectiveness in reducing heterosexual HIV transmission (Cochrane Review). The Cochrane Library. Chichester, United Kingdom: J Wiley;2004.
166. Pinkerton SD, Abramson PR. Effectiveness of condoms in preventing HIV transmission. Soc Sci Med 1997;44:1303-12.
167. Higgins DL, Galavotti C, O'Reilly KR, Schnell DJ, Moore M, Rugg DL, et al. Evidence for the effects of HIV antibody counseling and testing on risk behaviors. JAMA 1991;266:2419-29.
168. Weinhardt LS, Carey MP, Johnson BT, Bickham NL. Effects of HIV counseling and testing on sexual risk behavior: a meta-analytic review of published research, 1985-1997. Am J Public Health 1999;89:1397-405.
169. Wolitski RJ, MacGowan RJ, Higgins DL, Jorgensen CM. The effects of HIV counseling and testing on risk-related practices and help-seeking behavior. AIDS Educ Prev 1997;9:52-67.
170. Adoption of protective behaviors among persons with recent HIV infection and diagnosis—Alabama, New Jersey, and Tennessee, 1997-1998. MMWR Morb Mortal Wkly Rep 2000;49:512-5.
171. Parsons JT, Huszti HC, Crudder SO, Rich L, Mendoza J. Maintenance of safer sexual behaviours: evaluation of a theory-based intervention for HIV seropositive men with haemophilia and their female partners. Haemophilia 2000;6:181-90.
172. Colfax GN, Buchbinder SP, Cornelisse PG, Vittinghoff E, Mayer K, Celum C. Sexual risk behaviors and implications for secondary HIV transmission during and after HIV seroconversion. AIDS 2002;16:1529-35. [PMID: 12131191.
173. Belzer M, Rogers AS, Camarca M, Fuchs D, Peralta L, Tucker D, et al. Contraceptive choices in HIV infected and HIV at-risk adolescent females. J Adolesc Health 2001;29:93-100.
174. Rotheram-Borus MJ, Swendeman D, Comulada WS, Weiss RE, Lee M, Lightfoot M. Prevention for Substance-Using HIV-Positive Young People: Telephone and In-Person Delivery. J Acquir Immune Defic Syndr 2004;37 Suppl 2:S68-77.
175. Rotheram-Borus MJ, Lee MB, Murphy DA, Futterman D, Duan N, Birnbaum JM, et al. Efficacy of a preventive intervention for youths living with HIV. Am J Public Health 2001;91:400-5.
176. Wingood GM, DiClemente RJ, Mikhail I, Lang DL, McCree DH, Davies SL, et al. A randomized controlled trial to reduce HIV transmission risk behaviors and sexually transmitted diseases among women living with HIV: The WiLLOW Program. J Acquir Immune Defic Syndr 2004;37:S58-S67.
177. Fogarty LA, Heilig CM, Armstrong K, Cabral R, Galavotti C, Gielen AC, et al. Long-term effectiveness of a peer-based intervention to promote condom and contraceptive use among HIV-positive and at-risk women. Public Health Rep 2001;116 Suppl 1:103-19.
178. Kalichman SC, Rompa D, Cage M, DiFonzo K, Simpson D, Austin J, et al. Effectiveness of an intervention to reduce HIV transmission risks in HIV-positive people. Am J Prev Med 2001;21:84-92.
179. Coates TJ, McKusick L, Kuno R, Stites DP. Stress reduction training changed number of sexual partners but not immune function in men with HIV. Am J Public Health 1989;79:885-7.
180. Cleary PD, Van Devanter N, Steilen M, Stuart A, Shipton-Levy R, McMullen W, et al. A randomized trial of an education and support program for HIV-infected individuals. AIDS 1995;9:1271-8.
181. Patterson TL, Shaw WS, Semple SJ. Reducing the sexual risk behaviors of HIV+ individuals: outcome of a randomized controlled trial. Ann Behav Med 2003;25:137-45.
182. Richardson JL, Milam J, McCutchan A, Stoyanoff S, Bolan R, Weiss J, et al. Effect of brief safer-sex counseling by medical providers to HIV-1 seropositive patients: a multi-clinic assessment. AIDS 2004;18:1179-86.
183. Padian NS, O'Brien TR, Chang Y, Glass S, Francis DP. Prevention of heterosexual transmission of human immunodeficiency virus through couple counseling. J Acquir Immune Defic Syndr 1993;6:1043-8.
184. Allen S, Tice J, Van de Perre P, Serufilira A, Hudes E, Nsengumuremyi F, et al. Effect of serotesting with counseling on condom use and seroconversion among HIV discordant couples in Africa. BMJ 1992;304:1605-9.
185. Otten MW Jr, Zaidi AA, Wroten JE, Witte JJ, Peterman TA. Changes in sexually transmitted disease rates after HIV testing and posttest counseling, Miami, 1988 to 1989. Am J Public Health 1993;83:529-33.
186. Chamot E, Coughlin SS, Farley TA, Rice JC. Gonorrhea incidence and HIV testing and counseling among adolescents and young adults seen at a clinic for sexually transmitted diseases. AIDS 1999;13:971-9.
187. Kamb ML, Fishbein M, Douglas JM Jr, Rhodes F, Rogers J, Bolan G, et al. Efficacy of risk-reduction counseling to prevent human immunodeficiency virus and sexually transmitted diseases: a randomized controlled trial. Project RESPECT Study Group. JAMA 1998;280:1161-7.
188. Desenclos JC, Papaevangelou G, Ancelle-Park R. Knowledge of HIV serostatus and preventive behaviour among European injecting drug users. The European Community Study Group on HIV in Injecting Drug Users. AIDS 1993;7:1371-7.
189. Schlumberger MG, Desenclos JC, Papaevangelou G, Richardson SC, Ancelle-Park R. Knowledge of HIV serostatus and preventive behaviour among European injecting drug users: second study. European Community Study Group on HIV in Injecting Drug Users. Eur J Epidemiol 1999;15:207-15.
190. Celentano DD, Munoz A, Cohn S, Vlahov D. Dynamics of behavioral risk factors for HIV/AIDS: a 6-year prospective study of injection drug users. Drug Alcohol Depend 2001;61:315-22.
191. Calsyn DA, Saxon AJ, Freeman G Jr, Whittaker S. Ineffectiveness of AIDS education and HIV antibody testing in reducing high-risk behaviors among injection drug users. Am J Public Health 1992;82:573-5.
192. McCusker J, Bigelow C, Stoddard AM, Zorn M. Human immunodeficiency virus type 1 antibody status and changes in risk behavior among drug users. Ann Epidemiol 1994;4:466-71.
193. Margolin A, Avants SK, Warburton LA, Hawkins KA, Shi J. A randomized clinical trial of a manual-guided risk reduction intervention for HIV-positive injection drug users. Health Psychol 2003;22:223-8.
194. Ioannidis JP, Cappelleri JC, Skolnik PR, Lau J, Sacks HS. A meta-analysis of the relative efficacy and toxicity of Pneumocystis carinii prophylactic regimens. Arch Intern Med 1996;156:177-88.
195. Bucher HC, Griffith L, Guyatt GH, Opravil M. Meta-analysis of prophylactic treatments against Pneumocystis carinii pneumonia and toxoplasma encephalitis in HIV-infected patients. J Acquir Immune Defic Syndr Hum Retrovirol 1997;15:104-14.
196. El-Sadr WM, Murphy RL, Yurik TM, Luskin-Hawk R, Cheung TW, Balfour HH Jr, et al. Atovaquone compared with dapsone for the prevention of Pneumocystis carinii pneumonia in patients with HIV infection who cannot tolerate trimethoprim, sulfonamides, or both. Community Program for Clinical Research on AIDS and the AIDS Clinical Trials Group. N Engl J Med 1998;339:1889-95.
197. Payen MC, De Wit S, Sommereijns B, Clumeck N. A controlled trial of dapsone versus pyrimethamine-sulfadoxine for primary prophylaxis of Pneumocystis carinii pneumonia and toxoplasmosis in patients with AIDS. Biomed Pharmacother 1997;51:439-45.
198. Dunne MW, Bozzette S, McCutchan JA, Dube MP, Sattler FR, Forthal D, et al. Efficacy of azithromycin in prevention of Pneumocystis carinii pneumonia: a randomised trial. California Collaborative Treatment Group. Lancet 1999;354:891-5.
199. Bucher HC, Griffith LE, Guyatt GH, Sudre P, Naef M, Sendi P, et al. Isoniazid prophylaxis for tuberculosis in HIV infection: a meta-analysis of randomized controlled trials. AIDS 1999;13:501-7.
200. Wilkinson D. Drugs for preventing tuberculosis in HIV infected persons (Cochrane Review). The Cochrane Library. Chichester, United Kingdom: J Wiley;2003.
201. Oldfield EC 3rd, Fessel WJ, Dunne MW, Dickinson G, Wallace MR, Byrne W, et al. Once weekly azithromycin therapy for prevention of Mycobacterium avium complex infection in patients with AIDS: a randomized, double-blind, placebo-controlled multicenter trial. Clin Infect Dis 1998;26:611-9.
202. Pierce M, Crampton S, Henry D, Heifets L, LaMarca A, Montecalvo M, et al. A randomized trial of clarithromycin as prophylaxis against disseminated Mycobacterium avium complex infection in patients with advanced acquired immunodeficiency syndrome. N Engl J Med 1996;335:384-91.
203. Nightingale SD, Cameron DW, Gordin FM, Sullam PM, Cohn DL, Chaisson RE, et al. Two controlled trials of rifabutin prophylaxis against Mycobacterium avium complex infection in AIDS. N Engl J Med 1993;329:828-33.
204. Benson CA, Williams PL, Cohn DL, Becker S, Hojczyk P, Nevin T, et al. Clarithromycin or rifabutin alone or in combination for primary prophylaxis of Mycobacterium avium complex disease in patients with AIDS: A randomized, double-blind, placebo-controlled trial. The AIDS Clinical Trials Group 196/Terry Beirn Community Programs for Clinical Research on AIDS 009 Protocol Team. J Infect Dis 2000;181:1289-97.
205. Havlir DV, Dube MP, Sattler FR, Forthal DN, Kemper CA, Dunne MW, et al. Prophylaxis against disseminated Mycobacterium avium complex with weekly azithromycin, daily rifabutin, or both. California Collaborative Treatment Group. N Engl J Med 1996;335:392-8.
206. Brosgart CL, Louis TA, Hillman DW, Craig CP, Alston B, Fisher E, et al. A randomized, placebo-controlled trial of the safety and efficacy of oral ganciclovir for prophylaxis of cytomegalovirus disease in HIV-infected individuals. Terry Beirn Community Programs for Clinical Research on AIDS. AIDS 1998;12:269-7.
207. Spector SA, McKinley GF, Lalezari JP, Samo T, Andruczk R, Follansbee S, et al. Oral ganciclovir for the prevention of cytomegalovirus disease in persons with AIDS. Roche Cooperative Oral Ganciclovir Study Group. N Engl J Med 1996;334:1491-7.
208. Phillips AN, Lepri AC, Lampe F, Johnson M, Sabin CA. When should antiretroviral therapy be started for HIV infection? Interpreting the evidence from observational studies [Editorial]. AIDS 2003;17:1863-9.
209. Wood E, Hogg RS, Yip B, Harrigan PR, O'Shaughnessy MV, Montaner JS. Effect of medication adherence on survival of HIV-infected adults who start highly active antiretroviral therapy when the CD4+ cell count is 0.200 to 0.350 x 10(9) cells/L. Ann Intern Med 2003;139:810-6.
210. Palella FJ Jr, Deloria-Knoll M, Chmiel JS, Moorman AC, Wood KC, Greenberg AE, et al. Survival benefit of initiating antiretroviral therapy in HIV-infected persons in different CD4+ cell strata. Ann Intern Med 2003;138:620-6.
211. Opravil M, Ledergerber B, Furrer H, Hirschel B, Imhof A, Gallant S, et al. Clinical efficacy of early initiation of HAART in patients with asymptomatic HIV infection and CD4 cell count > 350 x 10(6)/l. AIDS 2002;16:1371-81.
212. Ahdieh-Grant L, Yamashita TE, Phair JP, Detels R, Wolinsky SM, Margolick JB, et al. When to initiate highly active antiretroviral therapy: a cohort approach. Am J Epidemiol 2003;157:738-46.
213. Sterling TR, Chaisson RE, Moore RD. Initiation of highly active antiretroviral therapy at CD4+ T lymphocyte counts of >350 cells/mm3: disease progression, treatment durability, and drug toxicity. Clin Infect Dis 2003;36:812-5.
214. Kaplan JE, Hanson DL, Cohn DL, Karon J, Buskin S, Thompson M, et al. When to begin highly active antiretroviral therapy? Evidence supporting initiation of therapy at CD4+ lymphocyte counts > 350 cells/microL. Clin Infect Dis 2003;37:951-8.
215. Anastos K, Barron Y, Miotti P, Weiser B, Young M, Hessol N, et al. Risk of progression to AIDS and death in women infected with HIV-1 initiating highly active antiretroviral treatment at different stages of disease. Arch Intern Med 2002;162:1973-80.
216. Sterling TR, Chaisson RE, Moore RD. HIV-1 RNA, CD4 T-lymphocytes, and clinical response to highly active antiretroviral therapy. AIDS 2001;15:2251-7.
217. Sterling TR, Chaisson RE, Keruly J, Moore RD. Improved outcomes with earlier initiation of highly active antiretroviral therapy among human immunodeficiency virus-infected patients who achieve durable virologic suppression: longer follow-up of an observational cohort study. J Infect Dis 2003;188:1659-65.
218. Wang C, Vlahov D, Galai N, Bareta J, Strathdee SA, Nelson KE, et al. Mortality in HIV-seropositive versus -seronegative persons in the era of highly active antiretroviral therapy: implications for when to initiate therapy. J Infect Dis 2004;190:1046-54.
219. Cozzi Lepri A, Phillips AN, d'Arminio Monforte A, Castelli F, Antinori A, de Luca A, et al. When to start highly active antiretroviral therapy in chronically HIV-infected patients: evidence from the ICONA study. AIDS 2001;15:983-90.
220. A comparison of two ways to manage anti-HIV treatment (the SMART study). Sponsored by National Institute of Allergy and Infectious Diseases (NIAID). June 2004. Accessed at www.clinicaltrials.gov/ct/show/NCT00027352?order = 1 on 16 July 2004.
221. The SMART Study—Strategies for Management of Anti-Retroviral Therapy. Accessed at www.smart-trial.com on 29 November 2004.
222. O'Brien ME, Clark RA, Besch CL, Myers L, Kissinger P. Patterns and correlates of discontinuation of the initial HAART regimen in an urban outpatient cohort. J Acquir Immune Defic Syndr 2003;34:407-14.
223. Chen RY, Westfall AO, Mugavero MJ, Cloud GA, Raper JL, Chatham AG, et al. Duration of highly active antiretroviral therapy regimens. Clin Infect Dis 2003;37:714-22.
224. Fellay J, Boubaker K, Ledergerber B, Bernasconi E, Furrer H, Battegay M, et al. Prevalence of adverse events associated with potent antiretroviral treatment: Swiss HIV Cohort Study. Lancet 2001;358:1322-7.
225. Schambelan M, Benson CA, Carr A, Currier JS, Dube MP, Gerber JG, et al. Management of metabolic complications associated with antiretroviral therapy for HIV-1 infection: recommendations of an International AIDS Society—USA panel. J Acquir Immune Defic Syndr 2002;31:257-75.
226. Dube MP, Sprecher D, Henry WK, Aberg JA, Torriani FJ, Hodis HN, et al. Preliminary guidelines for the evaluation and management of dyslipidemia in adults infected with human immunodeficiency virus and receiving antiretroviral therapy: Recommendations of the Adult AIDS Clinical Trial Group Cardiovascular Disease Focus Group. Clin Infect Dis 2000;31:1216-24.
227. Friis-Moller N, Sabin CA, Weber R, d'Arminio Monforte A, El-Sadr WM, Reiss P, et al. Combination antiretroviral therapy and the risk of myocardial infarction. N Engl J Med 2003;349:1993-2003.
228. d'Arminio A, Sabin CA, Phillips AN, Reiss P, Weber R, Kirk O, et al. Cardio- and cerebrovascular events in HIV-infected persons. AIDS 2004;18:1811-7.
229. Coplan PM, Nikas A, Japour A, Cormier K, Maradit-Kremers H, Lewis R, et al. Incidence of myocardial infarction in randomized clinical trials of protease inhibitor-based antiretroviral therapy: an analysis of four different protease inhibitors. AIDS Res Hum Retroviruses 2003;19:449-55.
230. Holmberg SD, Moorman AC, Greenberg AE. Trends in rates of myocardial infarction among patients with HIV [Letter]. N Engl J Med 2004;350:730-2;author reply 730-2.
231. Holmberg SD, Moorman AC, Williamson JM, Tong TC, Ward DJ, Wood KC, et al. Protease inhibitors and cardiovascular outcomes in patients with HIV-1. Lancet 2002;360:1747-8.
232. Barbaro G, Di Lorenzo G, Cirelli A, Grisorio B, Lucchini A, Hazra C, et al. An open-label, prospective, observational study of the incidence of coronary artery disease in patients with HIV infection receiving highly active antiretroviral therapy. Clin Ther 2003;25:2405-18.
233. Mary-Krause M, Cotte L, Simon A, Partisani M, Costagliola D. Increased risk of myocardial infarction with duration of protease inhibitor therapy in HIV-infected men. AIDS 2003;17:2479-86.
234.  Jutte A, Schwenk A, Franzen C, Romer K, Diet F, Diehl V, et al. Increasing morbidity from myocardial infarction during HIV protease inhibitor treatment? [Letter]. AIDS 1999;13:1796-7.
235. Leport C, Saves M, Ducimetiere P, Le Moal G, Amouyel P, Arveiler D, et al. Coronary heart disease risk (CHD) in French HIV-infected men started on a protease inhibitor (PI)-containing regimen compared to the general population [Abstract]. Ninth Conference on Retroviruses and Opportunistic Infections, Seattle, WA, February 2002: Abstract 697-T.
236. Klein D, Hurley LB, Quesenberry CP Jr, Sidney S. Do protease inhibitors increase the risk for coronary heart disease in patients with HIV-1 infection? J Acquir Immune Defic Syndr 2002;30:471-7.
237. Currier JS, Taylor A, Boyd F, Dezii CM, Kawabata H, Burtcel B, et al. Coronary heart disease in HIV-infected individuals. J Acquir Immune Defic Syndr 2003;33:506-12.
238. Torre D, Pugliese A, Orofino G. Effect of highly active antiretroviral therapy on ischemic cardiovascular disease in patients with HIV-1 infection [Letter]. Clin Infect Dis 2002;35:631-2.
239. Bozzette SA, Ake CF, Tam HK, Chang SW, Louis TA. Cardiovascular and cerebrovascular events in patients treated for human immunodeficiency virus infection. N Engl J Med 2003;348:702-10.
240. Rickerts V, Brodt H, Staszewski S, Stille W. Incidence of myocardial infarctions in HIV-infected patients between 1983 and 1998: the Frankfurt HIV-cohort study. Eur J Med Res 2000;5:329-33.
241. Braitstein P, Yip B, Heath KV, Levy AR, Montaner JS, Humphries K, et al. Interventional cardiovascular procedures among HIV-infected individuals on antiretroviral therapy 1995-2000. AIDS 2003;17:2071-5.
242. Sanders GD, Bayoumi AM, Sundaram V, Bilir SP, Neukermans CP, Rydzak CE, et al. Cost-effectiveness of screening for HIV in the era of highly active antiretroviral therapy. N Engl J Med 2005;352:570-85.
243. Paltiel AD, Weinstein MC, Kimmel AD, Seage GR 3rd, Losina E, Zhang H, et al. Expanded screening for HIV in the United States—an analysis of cost-effectiveness. N Engl J Med 2005;352:586-95.
244. Semba RD, Shah N, Klein RS, Mayer KH, Schuman P, Vlahov D, et al. Prevalence and cumulative incidence of and risk factors for anemia in a multicenter cohort study of human immunodeficiency virus-infected and -uninfected women. Clin Infect Dis 2002;34:260-6.
245. French N, Nakiyingi J, Carpenter LM et al. 23-valent pneumococcal polysaccharide vaccine in HIV-1-infected Ugandan adults: double-blind, randomized and placebo controlled trial. Lancet 2000;355:2106-11.
246. Watera C, Nakiyingi J, Miiro G, et al. 23-Valent pneumococcal polysaccharide vaccine in HIV-infected Ugandan adults: 6-year follow-up of a clinical trial cohort. AIDS 2004;18:1210-3.
247. Dworkin MS, Ward JW, Hanson DL, et al. Pneumococcal disease among human immunodeficiency virus-infected persons: incidence, risk factors, and impact of vaccination. Clinical Infectious Diseases 2001;32:794-800.
248. Lindenburg CE, Langendam MW, Benthem BH, Miedema F, Coutinho RA. No evidence that vaccination with a polysaccharide pneumococcal vaccine protects drug users against all-cause pneumonia. AIDS 2001;15:1315-7.
249. Gebo KA, Moore RD, Keruly JC, Chaisson RE. Risk factors for pneumococcal disease in human immunodeficiency virus-infected patients. Journal of Infectious Diseases 1996;173:857-62.
250. Breiman RF, Keller DW, Phelan MA, et al. Evaluation of effectiveness of the 23-valent pneumococcal capsular polysaccharide vaccine for HIV-infected patients. Archives of Internal Medicine 2000;160:2633-8.
251. Guerrero M, Kruger S, Saitoh A, et al. Pneumonia in HIV-infected patients: a case-control survey of factors involved in risk and prevention. AIDS 1999;13:1971-5.
252. Tasker SA, Treanor JJ, Paxton WB, Wallace MR. Efficacy of influenza vaccination in HIV-infected persons. A randomized, double-blind, placebo-controlled trial. Annals of Internal Medicine 1999;131:430-3.
253. Kellerman SE, Hanson DL, McNaghten AD, Fleming PL. Prevalence of chronic hepatitis B and incidence of acute hepatitis B infection in human immunodeficiency virus-infected subjects. Journal of Infectious Diseases 2003;188:571-7.
254. Bartlett JA, DeMasi R, Quinn J, Moxham C, Rousseau F. Overview of the effectiveness of triple combination therapy in antiretroviral-naive HIV-1 infected adults. AIDS 2001;15:1369-77.
255. Garcia F, de Lazzari E, Plana M, et al. Long-term CD4+ T-cell response to highly active antiretroviral therapy according to baseline CD4+ T-cell count. Journal of Acquired Immune Deficiency Syndromes 2004;36:702-13.
256. Kaufmann GR, Perrin L, Pantaleo G, et al. CD4 T-lymphocyte recovery in individuals with advanced HIV-1 infection receiving potent antiretroviral therapy for 4 years: the Swiss HIV Cohort Study. Archives of Internal Medicine 2003;163:2187-95.
257. Gulick RM, Meibohm A, Havlir D, et al. Six-year follow-up of HIV-1-infected adults in a clinical trial of antiretroviral therapy with indinavir, zidovudine, and lamivudine. AIDS 2003;17:2345-9.
258. Chene G, Sterne JA, May M, et al. Prognostic importance of initial response in HIV-1 infected patients starting potent antiretroviral therapy: analysis of prospective studies. Lancet 2003;362:679-86.
259. Quinn TC, Wawer MJ, Sewankambo N, et al. Viral load and heterosexual transmission of human immunodeficiency virus type 1. New England Journal of Medicine 2000;342:921-9.
260. Des Jarlais DC, Friedmann P, Hagan H, Friedman SR. The protective effect of AIDS-related behavioral change among injection drug users: a cross-national study. WHO Multi-Centre Study of AIDS and Injecting Drug Use. American Journal of Public Health 1996;86:1780-5.
261. Gibson DR, Flynn NM, Perales D. Effectiveness of syringe exchange programs in reducing HIV risk behavior and HIV seroconversion among injecting drug users. AIDS 2001;15:1329-41.
262. Gibson DR, Flynn NM, McCarthy JJ. Effectiveness of methadone treatment in reducing HIV risk behavior and HIV seroconversion among injecting drug users. AIDS 1999;13:1807-18.
263. Interpretation and use of the western blot assay for serodiagnosis of human immunodeficiency virus type 1 infections. MMWR Morb Mortal Wkly Rep 1989;38:1-7.
264. Recommended adult immunization schedule by age group and medical conditions—United States, 2003-2004. Atlanta: Centers for Disease Control and Prevention. Accessed at www.immunizenc.com/images/PDFs/03-04adultsched.pdf on 21 March 2005.
265. Hirsch MS, Brun-Vezinet F, D'Aquila RT, Hammer SM, Johnson VA, Kuritzkes DR, et al. Antiretroviral drug resistance testing in adult HIV-1 infection: recommendations of an International AIDS Society—USA Panel. JAMA 2000;283:2417-26.
266. Analysis of HIV-1 clinical trials: statistical magic? The AVANTI Steering Committee. Lancet 1999;353:2061-4.
267. Report of the NIH panel to define principles of therapy of HIV infection and guidelines for the use of antiretroviral agents in HIV-infected adults and adolescents. MMWR Morb Mortal Wkly Rep 1998:47(No. RR-5):1-82.
268. Bucher HC, Guyatt GH, Griffith LE, Walter SD. The results of direct and indirect treatment comparisons in meta-analysis of randomized controlled trials. J Clin Epidemiol 1997;50:683-91.
269. Song F, Altman DG, Glenny AM, Deeks JJ. Validity of indirect comparison for estimating efficacy of competing interventions: empirical evidence from published meta-analyses. BMJ 2003;326:472.
270. McQuillan GM, Khare M, Karon JM, Schable CA, Vlahov D. Update on the seroepidemiology of human immunodeficiency virus in the United States household population: NHANES III, 1988-1994. J Acquir Immune Defic Syndr Hum Retrovirol 1997;14:355-60.
271. Valleroy LA, MacKellar DA, Karon JM, Rosen DH, McFarland W, Shehan DA, et al. HIV prevalence and associated risks in young men who have sex with men. Young Men's Survey Study Group. JAMA 2000;284:198-204.
272. Holmberg SD. The estimated prevalence and incidence of HIV in 96 large US metropolitan areas. Am J Public Health 1996;86:642-54.
273. Macke BA, Maher JE. Partner notification in the United States: an evidence-based review. Am J Prev Med 1999;17:230-42.
274.  Partner counseling and referral services to identify persons with undiagnosed HIV—North Carolina, 2001. MMWR Morb Mortal Wkly Rep 2003;52:1181-4.
275.Weber B, Moshtaghi-Boronjeni M, Brunner M, Preiser W, Breiner M, Doerr HW. Evaluation of the reliability of 6 current anti-HIV-1/HIV-2 enzyme immunoassays. J Virol Methods 1995;55:97-104.
276. McAlpine L, Gandhi J, Parry JV, Mortimer PP. Thirteen current anti-HIV-1/HIV-2 enzyme immunoassays: how accurate are they? J Med Virol 1994;42:115-8.
277. Ihaka R, Gentleman R. R: A language for data analysis and graphics. J Comput Graph Stat. 1996;5:299-314.
278. Weinstein MC, Siegel JE, Gold MR, Kamlet MS, Russell LB. Recommendations of the Panel on Cost-effectiveness in Health and Medicine. JAMA 1996;276:1253-8.Ihaka R, Gentleman R. R: A language for data analysis and graphics. J Comput Graph Stat 1996;5:299-314.

Return to Table of Contents

The Figure is an analytic framework which begins at the left with "Screening" and "Risk Factor Assessment." From "Screening," the arrow proceeds to key question (KQ) 1: "Does screening for HIV in pregnant women reduce mother-to-child transmission or premature death and disability?" From there it proceeds to "Reduced premature death and disability for women and children and reduced mother-to-child transmission."  Under "Risk Factor Assessment" is KQ 2 with the note: "Asymptomatic Pregnant Women including Adolescents" KQ 2 is "Can clinical or demographic characteristics (including specific settings) identify subgroups of asymptomatic pregnant women at increased risk for HIV infection compared to the general population of pregnant women?" Two arrows proceed from KQ 2. One goes to "Low Risk" and the other to "High Risk."   The arrows both proceed to KQ 3. "What are the test characteristics of HIV antibody (HIV ab) test strategies in pregnant women?" One arrow proceeds from KQ 3 to KQ 4: What are the harms (including labeling and anxiety) associated with screening? Is screening acceptable to pregnant women?" Another arrow from KQ 3 splits and goes to "HIV Ab Positive" and "HIV Ab Negative."   From "HIV Ab Positive," an arrow proceeds to KQ 5: "How many HIV-infected pregnant women who meet criteria for interventions receive them?" Then the arrow splits in two. One prong goes to KQ 6: "What are the harms associated with the work-up for HIV infection in pregnant women?" The second prong splits in two and goes to "Low CD4 cell count or high viral load" and "High CD4 cell count and low viral load." The prongs reunite then split and proceed either to "Interventions," KQ 7a and KQ 7b, or to KQ 7c.  KQ 7a is "How effective are interventions (antiretroviral prophylaxis [to prevent mother-to-child transmission] or treatment [to improve maternal outcomes]; avoidance of breastfeeding, elective cesarean section [in selected patients], or other labor management practices; counseling on risky behaviors; immunizations; routine monitoring and follow-up; or prophylaxis against opportunistic infections) in reducing mother-to-child transmission rates or improving clinical outcomes (mortality, functional status, quality of life, symptoms, or opportunistic infections) in pregnant women with HIV infection?" KQ 7b is:"Does immediate antiretroviral treatment in HIV-infected pregnant women result in improvements in clinical outcomes compared to delayed treatment until the infected woman becomes symptomatic?" That arrow continues to "Reduced premature death and disability for women and children, and reduced mother-to-child transmission" and ends there.  KQ 7c is "How well do interventions reduce the rate of viremia, improve CD4 cell counts, or reduce risky behaviors? How does identification of HIV infection in pregnant women affect future reproductive choices?" The arrow splits from KQ 7a and one prong goes to KQ 8, "What are the harms (including adverse effects from in utero exposure) associated with antiretroviral drugs and elective cesarean section?" The other prong goes to KQ 9: "Have improvements in intermediate outcomes (CD4 cell counts, viremia, or risky behaviors) in HIV-infected pregnant women been shown to improve clinical outcomes or reduce mother-to-child transmission?" That arrow continues as a dotted line to "Reduced premature death and disability for women and children and reduced mother-to-child transmission."

Key Question (KQ) 1: Does screening for HIV infection in asymptomatic adolescents and adults reduce premature death and disability or spread of disease? KQ 2: Can clinical or demographic characteristics including specific settings) identify subgroups of asymptomatic adolescents and adults at increased risk for HIV compared to the general population? KQ 3: What are the test characteristics of HIV antibody test strategies? KQ 4: What are the harms (including labeling and anxiety) associated with screening? Is screening acceptable to patients? KQ 5: How many newly diagnosed HIV-positive patients meet criteria for antiretroviral treatment or prophylaxis against opportunistic infections? How many patients who meet criteria for interventions receive them? KQ 6: What are the harms associated with the work-up for HIV infection? KQ 7: a) How effective are interventions (antiretroviral treatment, counseling on risky behaviors, immunizations, routine monitoring and follow-up, more frequent Papanicolaou testing, or prophylaxis against opportunistic infections) in improving clinical outcomes (mortality, functional status, quality of life, symptoms, opportunistic infections, or transmission rates)? b) In asymptomatic patients with HIV infection, does immediate antiretroviral treatment result in improvements in clinical outcomes compared to delayed treatment until the patient is symptomatic? c) How well do interventions reduce the rate of viremia, improve CD4 counts, or reduce risky behaviors? KQ 8: What are the harms associated with antiretroviral therapy? KQ 9: Have improvements in intermediate outcomes (CD4 counts, viremia, risky behaviors) been shown to reduce premature death and disability or spread of disease? KQ 10: What is the cost-effectiveness of screening for HIV infection? *Excluding pregnant women, patients undergoing dialysis, and patients receiving transplants. A separate report13 reviews KQs 6, 7c, 9, and portions of 7a (immunizations, routine monitoring and follow-up, and more frequent Papanicolaou testing).

Return to Table of Contents

Persons seeking treatment for sexually transmitted diseases*
Homosexual or bisexual men*
Past or present injection drug users*
Persons who exchange sex for money or drugs, and their sex partners*
Women whose past or present sex partners were HIV-infected, bisexual, or injection drug users*
Persons with a history of transfusion between 1978 and 1985*
Persons having unprotected vaginal or anal intercourse with >1 sex partner

* Source: U.S. Preventive Services Task Force, 1996.11
Source: Centers for Disease Control and Prevention, 2001.21

Return to Table of Contents

Regimen Comparison Relative Risk (95% CI) Source, Year (Reference)
Pneumocystis carinii Pneumonia Cerebral Toxoplasmosis Mortality Outcomes
Systematic reviews
Any primary prophylaxis vs. placebo 0.39 (0.27-0.55) Not reported 0.87 (0.60-1.25) Ioannidis, et al., 1996*194
Trimethoprim-sulfamethoxazole vs. aerosolized pentamidine 0.59 (0.45-0.76) 0.78 (0.55-1.11) 0.88 (0.74-1.06) Bucher, et al., 1997195
0.58 (0.45-0.75) Not reported 0.99 (0.80-1.22) Ioannidis, et al., 1996*194
Dapsone-based regimen vs. aerosolized pentamidine 0.90 (0.71-1.15) 0.72 (0.54-0.97) 1.07 (0.90-1.27) Bucher, et al., 1997* 195
0.93 (0.72-1.19) Not reported 0.98 (0.86-1.12) Ioannidis, et al., 1996* [194]]
Trimethoprim-sulfamethoxazole vs. dapsone-based regimen 0.49 (0.26-0.92) 1.17 (0.68-2.18) 1.08 (0.88-1.25) Bucher, et al., 1997* 195
0.61 (0.34-1.10) Not reported 0.95 (0.82-1.11) Ioannidis, et al., 1996*194
Clinical trials
Atovaquone vs. dapsone 0.81 (0.58-1.12) 1.18 (0.26-5.30) 1.25 (0.98-1.59) El-Sadr, et al., 1998196
Dapsone vs. pyrimethamine-sulfadoxine 0.87 (P > 0.05) 1.07 (P > 0.05) 1.15 (not significant) Payen, et al., 1997197
Azithromycin vs. rifabutin in patients already receiving P. carinii prophylaxis 0.42 (0.24-0.76) Not reported Not reported Dunne, et al., 1999198

* Systematic review.
Includes studies of secondary prophylaxis.

Return to Table of Contents

Regimen Comparison Disseminated Mycobacterium avium intracellulare Infection (95% CI) Mortality (95% CI) Source, Year (Reference)
Azithromycin vs. placebo HR, 0.34 (P = 0.004) HR, 1.02 (P = 0.955) Oldfield, et al., 1998201
Clarithromycin vs. placebo HR, 0.31 (CI, 0.18-0.53) HR, 0.75 (0.58-0.97) Pierce, et al., 1996202
Rifabutin vs. placebo RR, 0.43 (0.26-0.70) RR, 0.68 (0.43-1.06) Nightingale, et al., 1993 (Studies 023 and 027)203
RR, 0.47 (0.29-0.77)
Clarithromycin vs. rifabutin RR, 0.56 (0.37-0.85) RR, 0.97 (0.78-1.20) Benson, et al., 2000204
Azithromycin vs. rifabutin HR, 0.53 (0.34-0.85) No differences Havlir, et al., 1996205
Clarithromycin + rifabutin vs. rifabutin RR, 0.43 (0.27-0.69) No differences Benson, et al., 2000204
Azithromycin + rifabutin vs. rifabutin HR, 0.28 (0.16-0.49) No differences Havlir, et al., 1996205
Azithromycin + rifabutin vs. azithromycin HR, 0.53 (0.29-0.95) No differences Havlir, et al., 1996205
Clarithromycin + rifabutin vs. clarithromycin RR, 0.79 (0.48-1.31) No differences Benson, et al., 2000204

* HR = hazard ratio; RR = relative risk.
For study 023.
For study 027.

Return to Table of Contents

CD4 Cell Count at Which HAART
Was Started, x 109 cells/L
Clinical Progression or Mortality Mortality (95% CI) Source, Year (Reference)
0.501-0.750 vs. <0.500 Not reported RR, 1.20 (0.17-8.53) Palella, et al., 2003210
0.351-0.500 vs. 0.200-0.350 Not reported RR, 0.61 (0.22-1.67) Palella, et al., 2003210
HR,0.95 (P = 0.897) Not reported Ahdieh-Grant, et al., 2003212
0.350-0.499 vs. <0.350 P = 0.21, log-rank test P = 0.10, log-rank test Sterling, et al., 2003213
0.350 vs. <0.350 HR, 0.28 (0.12-0.68) HR, 0.20 (0.07-0.52) Opravil, et al., 2002211
0.350-0.499 vs. <0.200 HR, 0.37 (P = 0.003) Not reported Ahdieh-Grant, et al., 2003212
0.201-0.350 vs. <200 Not reported RR, 0.27 (0.14-0.55) Palella, et al., 2003210
HR,0.39 (P < 0.001) Not reported Ahdieh-Grant, et al., 2003212

* All studies controlled for lead-time bias.
HAART = highly active antiretroviral therapy; HR = hazard ratio; RR = relative risk.

Return to Table of Contents

Results Prevalence, 0.3% Prevalence, 1% Prevalence, 5%-15% (High Risk)
Persons screened, n 10,000 10,000 10,000
Persons identified as HIV-positive, n 30 100 500-1500
Patients receiving test results, n 24-28 79-93 400-1400
Partners identified as HIV-positive, n 2-6 6-21 32-320
Total HIV-positive patients identified, n 26-34 85-114 426-1720
Patients with CD4 cell count < 0.200 X 109 cells/L, n 3-15 10-49 51-740
Cases of clinical progression or deaths prevented over 3 y with HAART, n 0.7-8.2 2-28 12-410
NNSB to prevent 1 clinical progression or death over 3 y 1210-13,800 360-4140 24-830
NNTB with HAART to prevent 1 clinical progression or death over 3 y 1.8 (1.5-2.2) 1.8 (1.5-2.2) 1.8 (1.5-2.2)
NNCB, NNSB, or NNTB to prevent 1 horizontal transmission over 3 y Unable to calculate Unable to calculate Unable to calculate
Cardiovascular or cerebrovascular events caused by HAART over 3 y, n 0.006-0.6 0.02-2 0.1-30
NNSH to cause 1 cardiovascular or cerebrovascular event over 3 y 16,900-1,580,500 5100-474,400 340-95,000
NNTH with HAART to cause 1 cardiovascular or cerebrovascular event over 3 y 69 (21-257) 69 (21-257) 69 (21-257)

* Values in parentheses are 95% CIs.
NNCB = number needed to counsel for benefit; NNSB = number needed to screen for benefit; NNSH = number needed to screen for harm; NNTB = number needed to treat for benefit; NNTH = number needed to screen for harm.

Return to Table of Contents

Question Number Key Question Level and Type of Evidence Overall Evidence for the Link Findings
1 Does screening for HIV in asymptomatic adolescents and adults reduce premature death and disability and spread of disease? None Not applicable No controlled studies or observational studies link screening directly to health outcomes.
2 Can clinical or demographic characteristics (including specific settings) identify a subgroup of asymptomatic adolescents and adults at increased risk for HIV compared to the general population? II-2. Cohort and cross-sectional studies Good The strongest risk factors for HIV infection from multiple large observational studies are intravenous drug use, male to male sex, and high-risk sexual behaviors. The largest U.S. study found that in federally funded testing sites, 20%-26% of HIV-positive patients reported no risk factors.28 In high-risk settings, several observational studies found that targeted screening based on broad criteria could increase the yield of screening but would still miss 7%-13% of positive patients while testing a much higher proportion.37-39
3 What are the test characteristics of HIV antibody test strategies? Studies of diagnostic test accuracy Good for standard and OraQuick rapid test; fair for other testing and collection methods Standard testing is associated with a sensitivity and specificity >99%.45-47 Initial studies indicate that FDA-approved rapid tests are associated with similar diagnostic test accuracy, but data from clinical settings are limited for rapid tests other than OraQuick on blood specimens.50-55 Home sampling and oral specimen sampling appear to have diagnostic accuracy similar to that of standard testing,58,59,64 but urine specimens may be associated with lower accuracy.60-63
4 What are the harms (including labeling and anxiety) associated with screening? Is screening acceptable to patients? Studies of diagnostic test accuracy II-2; cohort and cross-sectional studies for harms of screening and acceptability Good for false-positive rates and false-negative rates; fair to good for harms from screening and acceptability of testing False-positive results appear rare with standard testing, even in low-prevalence settings (1 of 250,000 blood donors).48 False-positive results from rapid tests could occur if results are given before confirmatory testing. False-negative results could occur during the window period before seroconversion and provide false reassurance. True-negative results could also provide false reassurance in patients practicing high-risk behaviors. True-positive results are associated with social consequences, anxiety, and labeling, but these harms are difficult to measure. Violence is very frequent in HIV-infected persons, but the impact of screening is not clear. Larger or more recent observational studies have not clearly shown that disclosure increases partner dissolution,87-89 intimate partner violence,84-86 or suicide risk.79 Acceptance rates vary widely even in similar settings (10%-97%) and may be improved by the availability of newer screening methods (rapid tests, noninvasive samples, home-based collection, on-site testing).94 An opt-out testing policy increased testing rates in 1 study.95
5 How many newly diagnosed HIV-positive patients meet criteria for antiretroviral treatment or prophylaxis against opportunistic infections? How many patients who meet criteria for interventions receive them? II-2. Cohort and cross-sectional studies Fair for proportion of patients qualifying for intervention at treatment (little information on initial viral load); good for proportion receiving interventions Seven U.S. studies found that 12%-43% of patients are diagnosed with CD4 cell counts below 0.200 × 109 cells/L, and 46%-80% with CD4 cell counts below 0.500 × 109 cells/L.26,41,112-116 No studies reported initial CD4 cell counts and viral loads in asymptomatic patients. No studies estimated the effects of screening on the proportion of patients qualifying for interventions or the effects of HAART on the proportion of patients qualifying for prophylaxis. A substantial proportion of HIV-positive patients do not receive or decline care. An estimated 36%-63% of infected patients were receiving care at least once every 6 months in 1996;124 38%-58% with positive test results do not return for initial post-test counseling (although about 90% are eventually located),30,117-120 and 53%-85% of infected patients who met guidelines for antiretroviral treatment were receiving them.129-132 Patients with lower CD4 cell counts and higher viral loads appear to have poorer response to antiretroviral therapy, but data on long-term outcomes are lacking.
6 What are the harms associated with the work-up for HIV infection? None Not applicable No evidence.
7a 1. How effective is antiretroviral treatment in improving clinical outcomes (mortality, functional status, quality of life, symptoms, opportunistic infections, or transmission rates)? I, II-2. Randomized, controlled trials; large cohort studies Good for clinical progression and death; fair for quality of life and spread of disease HAART is associated with improved clinical outcomes (clinical progression and death) compared to 2-drug therapy (OR, 0.62 [95% CI, 0.51-0.70]) and other less intense regimens.133 Quality-of-life outcomes from HAART have not been well studied. Beneficial effects of HAART on reducing horizontal transmission by reducing viral load may be offset by increases in risky behaviors,154,159 but there was insufficient evidence with which to estimate the effects of HAART on transmission rates.
2. How effective is counseling on risky behaviors in reducing transmission rates? II. Cohort studies Fair Few data address the effects of counseling and testing on HIV transmission rates in the United States. In Africa, uninfected women's knowledge of the HIV-positive status of their male partner was associated with a reduction in transmission by about 50%.184 Several observational studies indicate that sexually transmitted disease rates decline after an HIV diagnosis but may increase in persons testing negative.185,186 Interactive HIV counseling and testing was more effective than standard didactic counseling and testing in reducing sexually transmitted disease rates in 1 large, good-quality randomized trial, although there were too few cases to determine whether it was more effective at reducing new HIV infections.187 There is insufficient evidence with which to estimate effects of counseling on drug behaviors and transmission rates.
3. How effective are immunizations in improving clinical outcomes (mortality, functional status, quality of life, symptoms, opportunistic infections)? I, II-2. Randomized, controlled trials; large cohort studies Fair for pneumococcal, influenza, and hepatitis B vaccinations; poor for others In 1 randomized trial from Uganda, pneumococcal vaccination was associated with an increased risk for all-cause pneumonia (HR, 1.89 [95% CI, 1.1-3.2]),245 although long-term followup found an unexpected survival advantage (HR, 0.84 [CI, 0.7-1.0])246 (Observational studies mostly found a benefit from vaccination, particularly in patients with higher CD4 cell counts).247-251 Influenza vaccination was associated with a lower risk for symptomatic respiratory illness (49% vs. 29%; P = 0.04) in a clinical trial of HIV-infected patients in a military clinic.252 Hepatitis B vaccination was associated with a lower risk for acute hepatitis B in 1 observational study of HIV-infected persons.253 No studies had clinical outcomes of other immunizations in HIV-positive patients.
4. How effective is prophylaxis against opportunistic infections in improving clinical outcomes (mortality, functional status, quality of life, symptoms, opportunistic infections, or transmission rates)? I, II-2. Randomized, controlled trials; large cohort studies Good overall Good-quality systematic reviews found that preventive medication against PCP reduced the risk for PCP (RR, 0.39 [95% CI, 0.27-0.55]) and was associated with a nonsignificant mortality benefit (RR, 0.87 [CI, 0.60-1.25]).194,195 Some medications effective for PCP prophylaxis were also effective for toxoplasmosis prophylaxis. Two good-quality systematic reviews found that prophylaxis was effective at preventing active tuberculosis (risk reduced by 60%-86%) and death (risk reduced by 21%-23%) in patients with a positive skin test result.199,200 Multiple randomized, controlled trials found that preventive medication was effective for preventing disseminated Mycobacterium avium intracellulare infection, and may be associated with a mortality benefit (HR, ~0.75).201-206 In 2 randomized trials of ganciclovir, prophylaxis against CMV in patients who are positive for CMV antibody may prevent invasive CMV disease but does not appear associated with a significant mortality benefit.206,207
7b In asymptomatic patients with HIV infection, does immediate antiretroviral treatment result in reduced rates of premature death or disability compared to delayed treatment until symptomatic? II-2. Cohort studies Fair Large observational studies that controlled for lead-time bias consistently found that starting HAART at CD4 cell counts > 0.350 X 109 cells/L is associated with better clinical outcomes than starting at a count < 0.200 X 109 cells.210-213 The optimal CD4 cell count at which to start HAART in patients with counts between 0.200 and 0.350 X 109 cells is unclear. Observational studies that have controlled for lead-time bias did not control for other potentially important confounders (such as level of adherence or physician experience).
7c How well do interventions reduce the rate of viremia, improve CD4 cell counts, or reduce risky behaviors? I, II-2. Randomized, controlled trials; large cohort studies Good A fair-quality systematic review of HAART regimens found a rate of viral load suppression to <50 copies/mL at 48 wk of 47% overall (95% CI, 43%-51%).254 Observational studies found that 40%-50% of patients reached and maintained CD4 cell counts > 0.500 ×109 cells during HAART after 4-5 y,255,256 and 47% had a viral load < 50 copies/mL after 6 y.257 Two good-quality systematic reviews found that HIV counseling and testing are associated with decreases in risky sexually behaviors in persons testing positive, but the strength of the association varied according to the group studied.168-169 The strongest association was in heterosexual couples and in those testing positive. More intense or targeted counseling was more effective than standard counseling in several randomized trials.174-178
8 What are the harms associated with antiretroviral therapy? I, II-2. Randomized, controlled trials; large cohort studies Good In numerous clinical trials and observational studies, HAART regimens were associated with clinically significant short-term adverse events. Many patients can be switched to effective alternative regimens. Specific antiretroviral drugs and combinations are associated with specific adverse event profiles. A large, good-quality prospective cohort study found that the incidence of myocardial infarction and cardiac or cerebrovascular events increased with longer exposure to HAART (adjusted RR per year, 1.26 [95% CI, 1.12-1.41] and 1.26 [CI, 1.14-1.38], respectively) for the first 4 y, but the overall rate was low at 3.5 and 5.7 events, respectively, per 1000 person-years.228,244
9 Have improvements in intermediate outcomes (CD4 cell counts, viremia, risky behaviors) been shown to reduce premature death and disability or spread of disease? I, II-2. Randomized, controlled trials; large cohort studies Good for CD4 cell count or viral load and clinical progression and transmission risk; fair for behavior changes and transmission risk A large collaborative analysis of 13 cohort studies found that 6-mo CD4 cell count and viral load were strongly, independently associated with clinical outcomes in patients starting HAART.258 Observational studies found that low viral load was strongly correlated with decreased risk for HIV transmission in heterosexual couples,259 but data from patients treated with HAART are lacking. Condoms have been shown to be associated with decreased risk for transmission from HIV-infected persons.165,166 In mixed populations of infected and uninfected drug users, lower rates of HIV infection were associated with decreased risky drug use behaviors, participation in needle exchange programs, and participation in drug treatment programs.260-262
10 What is the cost-effectiveness of screening for HIV infection? Cost-effectiveness analyses Good Two good-quality cost-effectiveness analyses found that the cost-effectiveness of screening for HIV infections compared to no screening in settings with 1% prevalence was $38,000 to $42,000 per quality-adjusted life-year.242,243 One study found that when transmission benefits were incorporated into estimates, cost-effectiveness remained less than $50,000 per quality-adjusted life-year in settings with prevalences lower than that in the general population.242 Neither study evaluated the incremental cost-effectiveness of universal screening compared to targeted screening strategies in different populations.

* CMV = cytomegalovirus; FDA = U.S. Food and Drug Administration; HAART = highly active antiretroviral therapy; HR = hazard ratio; OR = odds ratio; PCP = Pneumocystis carinii pneumonia; RR = relative risk.
OraSure Technologies, Inc., Bethlehem, Pennsylvania.

Return to Table of Contents

Scope of Evidence Synthesis

The analytic framework in the Figure shows the target populations, interventions, and intermediate and health outcome measures we examined. The analytic framework was developed in consultation with the USPSTF and was refined after review by 6 content experts. We considered screening to be testing for HIV infection in asymptomatic persons or those with mild, nonspecific symptoms (such as fatigue) that are not predictive because they are so common. We excluded children (<13 years of age) because the prevalence of HIV in this population is low (9.3 per 100,000 population) and because most were infected vertically.3 We excluded other specific populations such as patients who had undergone transplantation, patients with known chronic viral hepatitis, and patients undergoing hemodialysis. In these groups, treatment considerations, adverse effects from treatment, and natural history may differ from those in the general population of HIV-infected persons; such patients are also usually excluded from clinical trials. We excluded patients with occupational exposures and blood donors because of consensus regarding testing for HIV infection in these situations. We excluded studies of HIV-2 infection because it is rare in the United States and its natural history differs substantially from that of HIV-1 infection.

Our review considered the standard screening strategy for HIV-1 infection to be an office-based venipuncture for anti-HIV enzyme-linked immunosorbent assay, followed by confirmatory Western blot for positive test results.46,263 We also considered rapid tests, home-based sampling, and tests using saliva or urine specimens. Viral load plus CD4 cell count testing was considered the standard work-up to determine the stage of infection and eligibility for interventions in infected patients.14,15,17,264

We evaluated recommended HAART regimens, prophylaxis against opportunistic infections, immunizations, Papanicolaou testing, counseling to reduce risky behaviors, and routine monitoring and followup. We excluded interventions not recommended for antiretroviral-naive patients or those not known to be effective. These include enfuvirtide; structured treatment interruptions; sequential initiation of therapy with antiretroviral drugs; induction-maintenance regimens; hydroxyurea; interleukin-2; acyclovir; and prophylaxis against candidiasis, histoplasmosis, coccidioidomycosis, herpes simplex virus infection, or cryptococcosis.14,17 We also did not consider resistance testing in antiretroviral-naive patients to be a routine intervention. Although the presence of primary antiretroviral drug resistance is increasing, resistance testing has mainly been studied in patients in whom a regimen has already failed. In patients with untreated chronic HIV infection, current U.S. guidelines either do not recommend routine resistance testing14 or do not give firm recommendations.265

For outcomes, we were particularly interested in reviewing literature on the benefit of early interventions in asymptomatic, treatment-naive patients. Clinical outcomes that we evaluated were mortality, AIDS-related opportunistic infections, spread of disease, and quality of life or functional status. For counseling, we included rates of sexually transmitted diseases as clinical markers of high-risk behaviors. Intermediate outcomes were loss of detectable viremia, improvement in CD4 cell counts, and changes in risky behaviors. We also reviewed harms from screening, work-up, and treatment. For harms from treatment, we focused on the long-term risk for cardiovascular complications and intolerable (causing discontinuation of therapy with the drug) side effects from HAART. Although interventions for chronic HIV infection, particularly HAART, are associated with many clinically significant short-term side effects, many are tolerable or patients can be switched to effective alternative regimens. In addition, intention-to-treat analyses of clinical outcomes incorporate the effects of intolerable or serious side effects.266 We did not include antiretroviral resistance as a separate outcome because its effects are seen in other intermediate (CD4 cell count, viral load) and clinical outcomes.

Methods

Literature Search and Strategy

We searched the topic of HIV in the MEDLINE® and Cochrane Library databases. Most searches were done from 1983 (the year that HIV was characterized) through 30 June 2004. For searches on antiretroviral therapy, we electronically searched these databases from 1998, the year that HAART was first recommended in U.S. guidelines;267 we supplemented these searches by an electronic search for systematic reviews of antiretroviral therapies from 1983. We performed a total of 13 searches covering the areas of risk factor assessment, screening tests, work-up, and interventions. Appendix B presents detailed electronic search strategies and results. Periodic hand searching of relevant medical journals, the Centers for Disease Control and Prevention Web site, and reviews of reference lists supplemented the electronic searches.

Content experts who reviewed the draft report identified additional citations. For rapid HIV tests, we included unpublished studies reported in manufacturer inserts. Other unpublished material was not included. Abstracts were not included in systematic searches, but major abstracts cited in reference lists or presented at recent conferences were included. We also obtained reviews, policy statements, and other papers with contextual value.

Inclusion and Exclusion Criteria

Papers were selected for full review if they were about HIV infection, were relevant to key questions, and met inclusion criteria. We also included cost-effectiveness analyses of HIV screening in outpatient settings in the HAART era. For all key questions, articles were limited to those that evaluated the general adult and adolescent population with chronic HIV infection. We excluded studies that included only overtly symptomatic patients or those with end-stage disease. Although the population of interest was persons with unsuspected HIV infection who would be identified by screening, we included studies of patients with a broad spectrum of chronic HIV disease to get a picture of the effects of screening and treatment in patients with different degrees of immune deficiency. We included studies performed in the United States, Australia, Canada, and countries of western Europe, in which the epidemiology and management of chronic HIV infection are similar. When important studies for a specific key question had been done only in other countries, we included these as well. We excluded studies of nonhuman subjects and those without original data. We considered non-English-language papers if they reported on clinical trials and an abstract was available in English. We searched for relevant systematic reviews for all key questions. A separate report lists additional key question-specific inclusion criteria.13

Data Extraction and Synthesis

We used predefined criteria from the USPSTF to assess the internal validity of included systematic reviews, trials, and observational studies, which we rated as "good," fair," or "poor." We also rated the applicability of each study to the population that would be identified by screening. The rating system was developed by the USPSTF and is described in detail elsewhere18 and summarized in Appendix C. For included trials and systematic reviews, we abstracted information about setting, patients, interventions, and outcomes. For intervention studies, when available we abstracted intention-to-treat results in which missing data were classified as treatment failures.266 We rated the overall body of evidence for each key question using the system developed by the USPSTF. We also rated studies evaluating cost-effectiveness of HIV screening in the HAART era using criteria developed by the USPSTF for evaluation of cost-effectiveness analyses (Appendix C).19

Methods for Outcomes Table

Table 5 estimates the outcomes after 3 years from one-time screening for HIV in 3 hypothetical cohorts of 10,000 adolescents or adults. We limited our time horizon to 3 years because longer studies on the clinical benefits from HAART are not yet available. We excluded areas from this table in which reliable data to estimate the clinical magnitude of benefit or harm were not available, such as harms from screening (anxiety, labeling, violence, suicide, partnership dissolution) and decreased transmission from counseling or other interventions. We also had insufficient data with which to estimate the impact of screening on earlier diagnosis of HIV and the proportion of patients qualifying for different interventions. Because short-term adverse events from HAART are usually self-limited, and effective alternative regimens are usually available, we focused on the long-term cardiovascular harms of HAART. We calculated numbers needed to screen and treat to prevent 1 case of clinical progression (new category B or C event) or death and to cause 1 cardiovascular event (myocardial infarction, invasive cardiovascular procedure, or stroke). Data from clinical trials were insufficient to separate clinical outcomes by severity.

Several assumptions made our estimates on the benefits of screening conservative. First, we focused on the effects of HAART. For some interventions (for example, most immunizations, more frequent Papanicolaou testing, routine monitoring and followup, and counseling), data were insufficient to estimate the magnitude of benefit. For others, such as prophylaxis against opportunistic infections, the magnitude of benefit from HAART substantially outweighs the benefit from other interventions, and successful treatment with HAART would also reduce the proportion of patients requiring prophylaxis by increasing CD4 counts. Second, we assumed that only asymptomatic patients with CD4 cell counts less than 0.200 X 109 cells/L would routinely receive HAART because they are at highest risk for clinical progression, evidence for clinical benefits of treatment is strongest in this group, and recommendations are less firm for asymptomatic patients with higher CD4 cell counts. Third, we estimated benefits only for the first 3 years after screening, although HAART is likely to be beneficial beyond that time period.

Methods for Calculating Relative Risk for Clinical Progression or Death during HAART Compared to No Treatment (used in Outcomes Table)

Because no clinical trials have directly evaluated the relative risk for clinical progression or death associated with HAART (antiretroviral therapy with 3 drugs) compared to no treatment in HIV-infected persons, we calculated this relative risk indirectly from data provided in a systematic review of clinical trials of 1-drug therapy versus no antiretroviral agents, 2-drug versus 1-drug therapy, and 3-drug versus 2-drug therapy in antiretroviral-naive persons.133 Bucher and colleagues268 proposed a method for indirect treatment comparisons to estimate odds ratios from 2 sets of clinical trials; we adapted this method to calculate the relative risk indirectly from the 3 sets of trials. Bucher and colleagues' method has been shown to usually agree with results of direct treatment comparisons.269 For this calculation, let RRMN, RRDM and RRTD denote relative risk for clinical progression or death on 1-drug therapy versus no antiretroviral drugs, 2-drug versus 1-drug therapy and 3-drug versus 2-drug therapy, respectively. The relative risk for clinical progression or death during 3-drug therapy versus no antiretroviral agents (RRTN) is given by:

RRTN = RRMN X RRDM X RRTD. (1)

To calculate the (1 -α)% Cl for RRTN, it is usual to use the natural log scale:

log(RRTN) = log(RRMN) + RRDM + log(RRTD) (2).

The variance of log relative risk is given as:

Var(log(RRTN)) = var(log(RRMN)) + var(log(RRDM)) + var(log(RRTD)). (3)

by assuming independence among log(RRMN), log(RRDM) and log(RRTD). Since log(RRTN) is approximately normally distributed, the (1 − α)% Cl for RRTN are


(RRTN exp(- Ζα/2sqrt(var(log(RRTN)))),  RRTN exp(Ζα/2sqrrt(var(log(RRTN))))). (4)

Jordan and colleagues133 reported the rates for clinical progression or death from clinical trials of 1-drug therapy vs. no antiretroviral agents (15 studies), 2-drug vs. 1-drug therapy (16 studies) and 3-drug versus 2-drug therapy (9 studies). In our analysis, we obtained estimates of RRMN and var(log(RRMN)) from a meta-analysis of the 15 trials comparing 1-drug therapy versus placebo. Similarly, we estimated RRDM and var(log(RRDM)) from a meta-analysis of the 16 trials comparing 2-drug versus 1-drug therapy; and we obtained estimates of RRTD and var(log(RRTD)) from a meta-analysis of the 9 studies of 3-drug versus 2-drug therapy. The assumption of independence between should be adequately satisfied because each value was estimated from different trials. We calculated an overall estimate of RRTN and its corresponding 95% CI by plugging these estimates into formulas (1) through (4). For each meta-analysis, tests for heterogeneity indicated statistically significant variation among studies, so we used a random-effects model to combine studies and calculate the estimates of RRMN, RRDM and RRTD. Estimates obtained by using a fixed-effects model, however, were similar to those from a random-effects model. Bucher and colleagues268 used a fixed-effects model to combine studies. Jordan and colleagues133 also used a fixed-effects approach to estimate odds ratios for 1-drug therapy versus placebo, 2-drug versus 1-drug therapy, and 3-drug versus 2-drug therapy.

Methods for Calculating 3-Year Risk for Cardiovascular Complications

The background rate (cases per 3 person-years) and relative risk for myocardial infarction and cardiovascular and cerebrovascular events (myocardial infarction, stroke, or invasive cardiovascular procedures) associated with combination antiretroviral therapy after 2 to 4 years compared to no exposure were calculated on the basis of raw data from the Data collection on Adverse events of anti-HIV Drugs (DAD) study (Figure; we used outcomes for no antiretroviral treatment and combined outcomes for 2 to 3 and 3 to 4 years of exposure) according to standard statistical methods.228,244

Methods To Calculate Numbers Needed To Screen and Treat

Calculations of numbers needed to screen for benefit (NNSB) and numbers needed to treat for benefit (NNTB) were based on estimates from different sources in the literature (Appendix Table). The indicated range of estimates and variation associated with estimates were incorporated in the calculations by using Monte Carlo simulations and are reflected by the ranges in the calculated NNSB and NNTB. The sampling distributions of the estimates used in the simulations were either the underlying distribution on which the calculation of 95% CI was based, or one that best approximated the point estimate and CI. For example, if the estimate was a rate or proportion, the logit of the rate or proportion was sampled assuming an approximately normal distribution; it was then transformed back to its original scale. For relative risks, we assumed that the log of relative risk was approximately normally distributed. The log of the relative risk was sampled from the normal distribution and then transformed back to relative risk. In each iteration of the Monte Carlo simulation, one sample of each proportion, relative risk, or other estimate was drawn to calculate the NNSB and NNTB. The point estimates and 95% CI of NNSB and NNTB were based on 1,000,000 samples. Similar calculations were performed to calculate numbers needed to screen for harm (NNSH) and numbers needed to treat for harm (NNTH). A simple program using R statistical language was written to perform simulations and calculate summary statistics.277

Return to Table of Contents

Immunization—Database: MEDLINE® (1996 to Present)

1. exp hiv infections/ or exp hiv/
2. exp Viral Hepatitis Vaccines/
3. exp Influenza Vaccine/
4. exp Bacterial Vaccines/
5. 2 or 3 or 4
6. 1 and 5
7. exp IMMUNIZATION/
8. exp Immunization Programs/
9. 7 or 8
10. exp HEPATITIS/
11. exp INFLUENZA/
12. exp PNEUMONIA/
13. 10 or 11 or 12
14. 1 and 9 and 13
15. 6 or 14
16. exp Evaluation Studies/
17. exp Epidemiologic Studies/
18. Comparative Study/
19. 16 or 17 or 18
20. 15 and 19
21. limit 15 to (clinical trial or guideline or meta-analysis or multicenter study or practice guideline)
22. 20 or 21
23. limit 22 to (human and english language)
24. from 23 keep 1-206

Prophylaxis—Database: MEDLINE® (1996 to Present)

1. exp AIDS-Related Opportunistic Infections/pc [Prevention & Control]
2. prophyla$.mp.
3. exp HIV Infections/co [Complications]
4. exp AIDS-Related Opportunistic Infections/
5. 2 and (3 or 4)
6. 1 or 5
7. limit 6 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
8. from 7 keep 1-396

Counseling—Database: MEDLINE® (1996 to Present)

1. exp HIV Infections/ or exp HIV/
2. exp COUNSELING/
3. 1 and 2
4. exp impulsive behavior/ or risk reduction behavior/ or risk-taking/
5. 1 and 4
6. 3 or 5
7. exp Evaluation Studies/
8. Comparative Study/
9. exp Epidemiologic Studies/
10. 7 or 8 or 9
11. 6 and 10
12. limit 6 to (clinical trial or guideline or meta-analysis or multicenter study or practice guideline)
13. 11 or 12
14. limit 13 to (human and english language)
15. from 14 keep 1-1272

Risk Factors—Database: MEDLINE® (1996 to Present)

1. exp RISK/
2. exp HIV Infections/mo, ep, eh, et, tm, pc [Mortality, Epidemiology, Ethnology, Etiology, Transmission, Prevention & Control]
3. 1 and 2
4. limit 3 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
5. exp HIV/
6. 1 and 5
7. limit 6 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
8. 4 or 7
9. exp Evaluation Studies/
10. Comparative Study/
11. exp Epidemiologic Studies/
12. 9 or 10 or 11
13. (3 or 6) and 12
14. limit 13 to (human and english language)
15. from 8 keep 1-573

Screening—Database: MEDLINE® (1996 to Present)

1. exp AIDS Serodiagnosis/
2. exp HIV SERONEGATIVITY/ or exp HIV ANTIGENS/ or exp HIV/ or exp HIV SEROPREVALENCE/ or exp HIV SEROPOSITIVITY/ or exp HIV ANTIBODIES/
3. exp Mass Screening/
4. 2 and 3
5. 1 or 4
6. exp "Sensitivity and Specificity"/
7. 5 and 6
8. ae.fs.
9. exp stress, psychological/
10. Life Change Events/
11. exp prejudice/ or prejudic$.mp.
12. 8 or 9 or 10 or 11
13. 5 and 12
14. exp diagnostic errors/
15. 5 and 14
16. 7 or 13 or 15
17. exp Evaluation Studies/
18. Comparative Study/
19. exp longitudinal studies/
20. 17 or 18 or 19
21. 16 and 20
22. limit 16 to (clinical trial or guideline or meta-analysis or multicenter study or practice guideline or review)
23. 22 or 21
24. limit 23 to (human and english language)
25. limit 23 to (human and abstracts)
26. 24 or 25
27. from 26 keep 1-247

Antiviral Drug—Database: MEDLINE® (1998 to Present)

1. exp HIV Infections/dt [Drug Therapy]
2. exp HIV/de [Drug Effects]
3. 1 or 2
4. exp Reverse Transcriptase Inhibitors/ad, tu
5. exp HIV Protease Inhibitors/ad, tu
6. exp antihiv agents/ad, tu
7. 4 or 5 or 6
8. 3 and 7
9. limit 8 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
10. exp Reverse Transcriptase Inhibitors/ae, ct, to, po
11. exp HIV Protease Inhibitors/ae, ct, to, po
12. exp antihiv agents/ae, ct, to, to
13. 10 or 11 or 12
14. 3 and 13
15. limit 14 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
16. 14 and exp epidemiologic studies/
17. 14 and (exp evaluation studies/ or exp comparative study/)
18. 16 or 17
19. limit 18 to (human and english language)
20. 15 or 19
21. limit 9 to yr = 1998-2003
22. from 21 keep 1-1157

Adverse Effects—Database: MEDLINE® (1998 to Present)

1. exp HIV Infections/dt [Drug Therapy]
2. exp HIV/de [Drug Effects]
3. 1 or 2
4. exp Reverse Transcriptase Inhibitors/ad, tu
5. exp HIV Protease Inhibitors/ad, tu
6. exp antihiv agents/ad, tu
7. 4 or 5 or 6
8. 3 and 7
9. limit 8 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
10. exp Reverse Transcriptase Inhibitors/ae, ct, to, po
11. exp HIV Protease Inhibitors/ae, ct, to, po
12. exp antihiv agents/ae, ct, to, to
13. 10 or 11 or 12
14. 3 and 13
15. limit 14 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
16. 14 and exp epidemiologic studies/
17. 14 and (exp evaluation studies/ or exp comparative study/)
18. 16 or 17
19. limit 18 to (human and english language)
20. 15 or 19
21. limit 9 to yr = 1998-2003
22. from 21 keep 1-1157
23. limit 20 to yr = 1998-2003
24. from 23 keep 1-732
25. from 24 keep 1-732

Work-up—Database: MEDLINE® (1998 to Present)

1. exp HIV/
2. viral load.mp. or Viral Load/
3. VIREMIA/
4. exp HIV Infections/
5. 1 or 4
6. 2 or 3
7. 5 and 6
8. (exp leukocyte count/ and cd4.mp.) or exp cd4 lymphocyte count/
9. exp "pathologic conditions, signs and symptoms"/ or disease progression/
10. 7 and 8 and 9
11. exp "sensitivity and specificity"/
12. 10 and 11
13. exp epidemiologic studies/
14. 10 and 13
15. limit 10 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
16. limit 14 to (human and english language)
17. 15 or 16
18. from 17 keep 1-232

Maternal—Database: MEDLINE® (1996 to Present)

1. exp HIV/ or exp HIV INFECTIONS/
2. exp Anti-HIV Agents/ad, ae, po, ct, tu, to [Administration & Dosage, Adverse Effects, Poisoning, Contraindications, Therapeutic Use, Toxicity]
3. exp Reverse Transcriptase Inhibitors/ad, ae, po, ct, tu, to [Administration & Dosage, Adverse Effects, Poisoning, Contraindications, Therapeutic Use, Toxicity]
4. exp HIV Protease Inhibitors/ad, ae, po, tu, ct, to [Administration & Dosage, Adverse Effects, Poisoning, Therapeutic Use, Contraindications, Toxicity]
5. 1 and (2 or 3 or 4)
6. exp Disease Transmission, Vertical/
7. exp HIV Infections/tm
8. pregnancy complications/ or exp pregnancy complications, infectious/
9. exp Pregnancy/
10. 6 or 7
11. 8 or 9
12. 10 and 11
13. 5 and 12
14. limit 13 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
15. exp Evaluation Studies/
16. Comparative Study/
17. exp Epidemiologic Studies/
18. 15 or 16 or 17
19. 13 and 18
20. limit 19 to (human and english language)
21. 14 or 20
22. from 21 keep 1-373

Cesarean—Database: MEDLINE® (1996 to Present)

1. exp HIV/ or exp HIV INFECTIONS/
2. exp Anti-HIV Agents/ad, ae, po, ct, tu, to [Administration & Dosage, Adverse Effects, Poisoning, Contraindications, Therapeutic Use, Toxicity]
3. exp Reverse Transcriptase Inhibitors/ad, ae, po, ct, tu, to [Administration & Dosage, Adverse Effects, Poisoning, Contraindications, Therapeutic Use, Toxicity]
4. exp HIV Protease Inhibitors/ad, ae, po, tu, ct, to [Administration & Dosage, Adverse Effects, Poisoning, Therapeutic Use, Contraindications, Toxicity]
5. exp cesarean section/
6. 1 and (2 or 3 or 4 or 5)
7. exp Disease Transmission, Vertical/
8. exp HIV Infections/tm
9. pregnancy complications/ or exp pregnancy complications, infectious/
10. exp Pregnancy/
11. 7 or 8
12. 9 or 10
13. 11 and 12
14. 6 and 13
15. limit 14 to (human and english language and (clinical trial or guideline or meta-analysis or multicenter study or practice guideline))
16. exp Evaluation Studies/
17. Comparative Study/
18. exp Epidemiologic Studies/
19. 16 or 17 or 18
20. 14 and 19
21. limit 20 to (human and english language)
22. 15 or 21

Cost of Screening—Database: MEDLINE® (1996 to Present)

1. exp HIV Infections/
2. exp HIV/
3. 1 or 2
4. exp "Costs and Cost Analysis"/
5. 3 and 4
6. Comparative Study/
7. exp Evaluation Studies/
8. exp epidemiologic study characteristics/
9. 5 and (6 or 7 or 8)
10. limit 9 to (human and english language)
11. exp Mass Screening/
12. 9 and 11
13. 5 and 11
14. limit 13 to (human and english language)
15. ec.fs.
16. 3 and 15
17. 16 and 11
18. limit 17 to (human and english language)
19. 14 or 18
20. from 19 keep 1-179

Systematic Reviews—Database: PubMED®

1. hiv/de [mh] OR hiv infections/dt [mh]
2. anti hiv agents[pa] OR reverse transcriptase inhibitors[pa] OR hiv protease inhibitors [pa]
3. #1 OR #2
4. evaluation studies[mh] OR epidemiologic studies[mh] OR comparative study [mh]
5. #3 AND #4
6. tu[sh] OR ad[sh] OR ae[sh] OR to[sh] OR po[sh] OR ct[sh]
7. #5 AND #6
8. #7 AND systematic [sb]
9. #8 AND Limits: Publication Date from 1989 to 1997, English, Human

Note: Systematic [sb] represents the following strategy as taken from the Clinical Queries search help page within PubMed®.

((systematic review$ OR systematic literature review$ OR meta-analysis.pt. OR meta-analysis.ti. OR meta-analysis.ti. OR meta-analyses.ti. OR evidence-based medicine OR (evidence-based AND (guideline.tw. OR guidelines.tw. OR recommendations)) OR (evidenced-based AND (guideline.tw. OR guidelines.tw. OR recommendation$)) OR consensus development conference.pt. OR health planning guidelines OR guideline.pt. OR cochrane database syst rev OR acp journal club OR health technol assess OR evid rep technol assess summ OR evid based nurs OR evid based ment health OR clin evid) OR ((systematic.tw. OR systematically OR critical.tw. OR (study.tw. AND selection.tw.) OR (predetermined OR inclusion AND criteri$.tw.) OR exclusion criteri$ OR main outcome measures OR standard of care) AND (survey.tw. OR surveys.tw. OR overview$ OR review.tw. OR reviews OR search$ OR handsearch OR analysis.tw. OR critique.tw. OR appraisal OR (reduction AND risk AND (death OR recurrence))) AND (literature.tw. OR articles OR publications.tw. OR publication.tw. OR bibliography.tw. OR bibliographies OR published OR unpublished OR citation OR citations OR database OR internet.tw. OR textbooks.tw. OR references OR trials OR meta-analysis.mh. OR (clinical.tw. AND studies) OR treatment outcome)) NOT (case report.ti. OR case report.mh. OR editorial.ti. OR editorial.pt. OR letter.pt. OR newspaper article.pt.))

Return to Table of Contents

Diagnostic Accuracy Studies

Criteria

  1. Screening test relevant, available for primary care, adequately described.
  2. Credible reference standard, performed regardless of test results.
  3. Reference standard interpreted independently of screening test.
  4. Indeterminate results handled in a reasonable manner.
  5. Spectrum of patients included in study.
  6. Sample size.
  7. Administration of reliable screening test.

Definition of Ratings Based on Above Criteria

Good: Evaluates relevant available screening test; uses a credible reference standard; interprets reference standard independently of screening test; assesses reliability of test; has few or handles indeterminate results in a reasonable manner; includes large number (>100) broad-spectrum patients with and without disease.

Fair: Evaluates relevant available screening test; uses reasonable although not best standard; interprets reference standard independently of screening test; has moderate sample size (50 to 100 participants), and includes a "medium" spectrum of patients.

Poor: Has important limitations, such as inappropriate reference standard, improperly administered screening test, biased ascertainment of reference standard, or very small sample size of very narrow selected spectrum of patients.

Randomized, Controlled Trials and Cohort Studies

Criteria

  1. Initial assembly of comparable groups: randomized, controlled trials—adequate randomization, including concealment and statement of whether potential confounders were distributed equally among groups; cohort studies—consideration of potential confounders with either restriction or measurement for adjustment in the analysis; consideration of inception cohorts.
  2. Maintenance of comparable groups (includes attrition, crossovers, adherence, and contamination).
  3. Important differential loss to followup or overall high loss to followup.
  4. Measurements: equal, reliable, and valid (includes masking of outcome assessment).
  5. Clear definition of interventions.
  6. Important outcomes considered.
  7. Analysis: adjustment for potential confounders for cohort studies, or intention-to-treat analysis for randomized, controlled trials.

Definition of Ratings Based on Above Criteria

Good: Meets all criteria—comparable groups are assembled initially and maintained throughout the study (followup > 80%), reliable and valid measurement instruments are used and applied equally to the groups, interventions are spelled out clearly, important outcomes are considered, and appropriate attention to confounders in analysis.

Fair: Studies will be graded "fair" if any or all of the following problems occur, without the important limitations noted in the "poor" category below: Generally comparable groups are assembled initially but some question remains as to whether some (although not major) differences occurred in followup, measurement instruments are acceptable (although not the best) and generally applied equally, some but not all important outcomes are considered, and some but not all potential confounders are accounted for.

Poor: Studies will be graded "poor" if any of the following major limitations exists: Groups assembled initially are not close to being comparable or maintained throughout the study, unreliable or invalid measurement instruments are used or not applied at all equally among groups (including failure to mask outcome assessment), and key confounders are given little or no attention.

Case-Control Studies

Criteria

  1. Accurate ascertainment of cases.
  2. Nonbiased selection of case-patients and controls, with exclusion criteria applied equally to both.
  3. Response rate.
  4. Diagnostic testing procedures applied equally to each group.
  5. Measurement of exposure accurate and applied equally to each group.
  6. Appropriate attention to potential confounding variable.

Definition of Ratings Based on Above Criteria

Good: Appropriate ascertainment of cases and nonbiased selection of case-patients and controls, exclusion criteria applied equally to case-patients and controls, response rate of 80% or greater, diagnostic procedures and measurements accurate and applied equally to case-patients and controls, and appropriate attention to confounding variables.

Fair: Recent, relevant, without major apparent selection or diagnostic work-up bias but with response rate less than 80% or attention to some but not all important confounding variables.

Poor: Major selection or diagnostic work-up biases, response rates less than 50%, or inattention to confounding variables.

Cost-Effectiveness Analyses: Criteria

Framing

  1. Are interventions and populations compared appropriate?
  2. Is the study conducted from the societal perspective?
  3. Is the time horizon clinically appropriate and relevant to the study question?

Effects

  1. Are all important drivers of effectiveness included?
  2. Are key harms included?
  3. Is the best available evidence used to estimate effectiveness?
  4. Are long-term outcomes used?
  5. Do effect measures capture preferences or utilities?

Costs

  1. Are all appropriate downstream costs included?
  2. Are charges converted to costs appropriately?
  3. Are the best available data used to estimate costs?

Results

  1. Are incremental cost-effectiveness ratios presented?
  2. Are appropriate sensitivity analyses performed? Quality criteria for cost-effectiveness analyses were based on those developed by the USPSTF,19 which, in turn, are based on recommendations of the Panel on Cost-Effectiveness in Health and Medicine.278 We used the criteria to guide our categorization of studies as good, fair, or poor. We assigned quality grades on the basis of a subjective assessment of study design and quality of data inputs.
Return to Table of Contents

Base-Case Assumptions Values Used in Outcomes Table Source, Year (Reference)
Prevalence of HIV infection Average-risk: 0.3%
High-risk: 5%-15%
CDC, 20033
McQuillan, et al., 1997269
Valleroy, et al., 2000270
Holmberg, 1996271
Yield of partner notification (newly diagnosed HIV infection per index patient) 0.08-0.23 Macke and Maher, 1999273
CDC, 2003274
Accuracy of standard testing ≥ 99% Weber, et al., 1995275
McAlpine, et al., 1994 276
CDC, 199045
CDC, 198846
Proportion of HIV-positive patients who receive test results 79%-93% Erickson, et al., 199030
Hightow, et al., 2003120
CDC, 200440
Molitor, et al., 1999119
Proportion of patients who would qualify for treatment (assuming only patients with CD4 count < 0.200 X 109 cells/L treated) 12%-43% Samet, et al., 2001112
Katz, et al., 1992113
Luby, et al., 1994114
Hutchinson, et al., 1991115
Klein, et al., 200326
Proportion of patients qualifying for antiretroviral therapy who would receive it 53%-85% Stall, et al., 2001129
Cunningham, et al., 2000130
Kaplan, et al., 1999131
McNaghten, et al., 2003132
3-y risk for clinical progression or death in untreated patients with CD4 count < 0.200 X 109 cells/L 86% (95% CI, 77%-93%) Mellors, et al., 19977
Relative risk for clinical progression or death with HAART compared to no treatment 0.35 (95% CI, 0.25-0.47) Calculated from Jordan, et al., 2002133
Background rate of myocardial infarction (cases per 3 person-years) 0.00158 (95% CI, 0.000508-0.00487) Calculated from Friis-Moller, 2003227
Relative risk for myocardial infarction with HAART after 2-4 y compared to no treatment 7.73 (95% CI, 2.42-24.71) Calculated from Friis-Moller, 2003227
Background rate of cardio- or cerebrovascular (myocardial infarction, stroke, or invasive cardiovascular procedure) events (cases per 3 person-years) 0.0037 (95% CI, 0.0018-0.00770) Calculated from Writing Group of the DAD Study, 2004228
Relative risk for cardiovascular or cerebrovascular events with HAART after 2-4 y compared to no treatment 5.00 (95% CI, 2.31-10.82) Calculated from Writing Group of the DAD Study, 2004228
Relative risk for spread of disease Unable to estimate  

* CDC = Centers for Disease Control and Prevention; DAD = Data collection of Adverse events of anti-HIV Drugs; HAART = highly active antiretroviral therapy.

Return to Table of Contents