University of California, San Francisco Logo

University of California, San Francisco | About UCSF | Search UCSF | UCSF Medical Center

Clinical Overview
Clinical Overview of HIV Disease
transparent image
transparent image
transparent image
transparent image
Introduction and History
transparent image
transparent image
Basics of HIV Virology and Immunology
transparent image
transparent image
transparent imageVirology
transparent image
transparent imageImmunology
transparent image
Epidemiology
transparent image
transparent image
Transmission and Risk Factors
transparent image
transparent image
Classification of HIV Disease
transparent image
transparent image
Natural History of Untreated HIV Infection
transparent image
transparent image
transparent imagePrimary HIV Infection
transparent image
transparent imageChronic HIV Infection
transparent image
transparent imageClinical AIDS
transparent image
transparent imageSpecial Considerations in Disease Progression
transparent image
transparent image
transparent imageHost Factors
transparent image
transparent imageViral Factors
transparent image
transparent imageCoinfections
transparent image
transparent imageLong-Term Nonprogressors
transparent image
transparent image
Laboratory Testing
transparent image
transparent image
transparent imageHIV Antibody Testing
transparent image
transparent image
transparent image"Detuned" Antibody Testing
transparent image
transparent image
transparent imageCD4 Testing
transparent image
transparent imageHIV Viral Load Testing
transparent image
transparent imageHIV Antigen Testing
transparent image
transparent imageHIV Resistance Testing
transparent image
transparent imageTherapeutic Drug Monitoring
transparent image
transparent imageOther HIV Testing Techniques
transparent image
Treatment of HIV Infection
transparent image
transparent image
transparent imageOverview of Antiretroviral Medications
transparent image
transparent imageInitiating Treatment
transparent image
transparent imageMonitoring ART
transparent image
transparent imageChanging ART
transparent image
transparent imageTreatment Interruption
transparent image
transparent imageImmune Modulators for HIV Treatment
transparent image
transparent imageProphylaxis Against OIs
transparent image
Routine Health Care Maintenance in HIV Infection
transparent image
transparent image
Summary and Future Directions
transparent image
transparent image
transparent image
References
transparent image
transparent image
Tables
Table 1.The 1993 AIDS Surveillance Case Definition of the U.S. Centers for Disease Control and Prevention
transparent image
Table 2.CDC Categorization of HIV/AIDS
transparent image
Table 3.WHO Staging System for HIV Infection and Disease in Adults and Adolescents
transparent image
Table 4.Selected adverse reactions associated with antiretrovirals
transparent image
Table 5.Relationship between CD4, viral load and AIDS progression
transparent image
transparent image
Figures
Figure 1.Prevalence of HIV and distribution of major clades
transparent image
Figure 2.Typical course of untreated HIV infection
transparent image
transparent image
transparent image
transparent image
Introduction and History
transparent image

Widespread awareness of HIV disease began with a brief report in 1981, published in the Morbidity and Mortality Weekly Report, of a rare pneumonia caused by Pneumocystis carinii (now known as P jiroveci) as well as other unusual infections in 5 young homosexual men in Los Angeles.(1) Awareness that a significant epidemic was developing grew as case reports mounted and similar immune deficiency syndromes were described in New York, California,(2,3) and elsewhere among homosexual men, intravenous drug users, Haitians,(4) hemophiliacs,(5) recipients of blood transfusions,(6) infants,(7) female sexual partners of infected men,(8,9) prisoners,(10) and Africans.(11) As researchers began to describe the epidemiology and risk factors in a systematic way, many theories emerged regarding the cause of the mysterious disease. An infectious agent was postulated, and, in 1983, a novel human retrovirus was isolated as the putative etiologic agent.(12-14) That virus was eventually named human immunodeficiency virus, or HIV.(15) Despite dramatic advances in basic virology and clinical management, HIV infection has developed into a worldwide pandemic, with tens of millions of individuals infected by the virus and many millions more affected by it. Clinicians treating HIV are challenged by a clinically complex illness with relatively limited resources for treatment in most settings.

By 1985, serologic assays had been developed to test for HIV infection in asymptomatic persons, to identify new infections by seroconversion, and to screen blood donations.(16) Early trials of antiviral treatments for HIV and immune modulators were fraught with disappointment.(17-22) In 1987, zidovudine (AZT, or azidothymidine) became the first drug approved by the U.S. Food and Drug Administration (FDA) for the treatment of AIDS.(23) Early excitement over the life-extending effects of the drug soon waned, as patients treated with this single-drug therapy began to experience disease progression leading in most cases, to death. However, understanding of the epidemiology, treatment, and prophylaxis of opportunistic infections (OIs) associated with HIV-induced immune deficiency led to significant life-saving advances, particularly in the areas of infection with Pneumocystis jiroveci and Mycobacterium avium complex (MAC).(24,25)

The introduction of protease inhibitors (PIs) in the mid-1990s revolutionized the treatment of HIV.(26) Effective combination antiretroviral therapy (ART) became the standard of care in the United States and Western Europe. Very soon thereafter, countries in which effective ART was available began to note sharply declining morbidity and mortality associated with HIV infection.(27)

Studies of patients receiving the new therapies shed light on HIV pathogenesis. Patients treated with potent ART showed precipitous decreases in the amount of HIV RNA circulating in their serum, indicating interference with HIV replication (which, unimpeded, can produce more than 10 billion viral particles per day). Additionally, after successful inhibition of viral replication, CD4 T-cell counts began to increase in treated individuals, demonstrating the regenerative capacity of the damaged immune system.(28) Corroborating this understanding of the dynamic interaction between viral replication and the host immune system, studies began to show the value of HIV RNA measurement (viral load) as both a predictor of disease progression and a measure of treatment success.(29-33)

Potent therapy was not without complications, however; and the dogma of the late 1990s, "hit early, hit hard," (34) became balanced by realization that long-term medication toxicity was likely among individuals who were now living longer, healthier lives with HIV infection. Once again, the paradigm of HIV treatment underwent revision, and treatment was now recommended primarily for individuals with more advanced disease.(35)

This chapter provides a general overview of issues relevant to clinical practitioners. More extensive discussions on the topics covered here are available in other chapters of the HIV InSite Knowledge Base.

transparent image
Basics of HIV Virology and Immunology
transparent image

(See chapters "Molecular Insights Into HIV Biology" and "Structure, Expression, and Regulation of the HIV Genome")

transparent image
Virology
transparent image

HIV-1 and the less common HIV-2 belong to the family of retroviruses. HIV-1 contains a single-stranded RNA genome that is 9 kilobases in length and contains 9 genes that encode 15 different proteins.(36,37) The major viral proteins (some of which contain >1 protein subunit) are classified as structural proteins (Gag, Pol, and Env), regulatory proteins (Tat and Rev), and accessory proteins (Vpu, Vpr, Vif, and Nef).

Three major classes of HIV-1 have emerged: M (main), N (new), and O (outlier). Among M group viruses, which account for >90% of HIV infections worldwide, there are 9 subtypes, called clades, designated by the letters A-D, F-H, J, and K, as well as many recombinant forms.(38,39) Variation between one clade and another in the amino acid sequences of the envelope protein may exceed 30%. Clade B, the most common subtype in the Americas and Western Europe, differs considerably from those clades found in Asia and Africa, where the majority of HIV-infected individuals reside (see Figure 1). Viral diversity is greatest in sub-Saharan Africa. To date, most HIV drug development has targeted clade B. As HIV treatment is extended into regions where non-B clades predominate, issues of differential drug response, drug mutation patterns, and reliability of viral testing (ie, viral loads and resistance testing) may emerge.(40,41) Sequence diversity among various clades also needs to be considered in vaccine development, as most HIV-specific neutralizing antibodies (42) and some cytotoxic T-lymphocyte (CTL) responses (43) are type specific.

HIV infection of a host cell begins with the binding of the virus particle (virion) to the host cell. This process is initiated when the surface envelope protein (Env, which consists of 3 copies each of the 2 subunit proteins gp120 and gp41) engages its primary receptor, the CD4 molecule on the surface of the target cell.(44) Initial binding to CD4 exposes another portion of the Env trimer, which then binds to a coreceptor, usually the chemokine receptor CXCR4 (in the case of T-cell-tropic, or syncytium-inducing strains of HIV) or the chemokine receptor CCR5 (in the case of macrophage-tropic, or nonsyncytium-inducing strains).(45) This coreceptor binding causes the gp41 trimer portion of the envelope molecule to spring open and "harpoon" the lipid bilayer of the target cell membrane. The "hairpin" domains of gp41 then fold together to pull the virus and host cell membranes together, allowing fusion to occur.(46) The viral contents, including copies of the viral genetic material and the Pol protein (reverse transcriptase, or RT) thus enter the cytoplasm of the host cell. Reverse transcription, that is, the copying of the viral genetic material from RNA into DNA can then occur.

The preintegration complex (PIC), composed of the copied DNA (cDNA) and a number of viral and host proteins, then enters the cell nucleus, where the viral enzyme integrase mediates the insertion of the viral cDNA into the host chromosomal DNA.(47) The resulting integrated DNA virus (also called a provirus, to distinguish it from the virion form) may remain latent for hours to years before becoming active through transcription (copying of DNA into RNA).(48) Transcription of the viral genome is under complex control of a number of proteins, including Tat and cellular DNA transcription factors.(49) Transport of the transcribed viral RNA out of the nucleus also depends on a number of host and viral factors, including Rev.(50) The transcribed viral RNA may be transported out of the nucleus in its full-length form to serve as genetic material for new virions, or it may be partially or fully spliced. The unspliced, partially spliced, and fully spliced versions of viral RNA direct the synthesis of different viral proteins by the cell ribosomes. New viral particles are assembled at the plasma membrane and incorporate Gag subunits, Pol, Nef, Env, Vpr, and viral genomic RNA.(51) The HIV viral protease enzyme acts following virion assembly to cleave viral proteins into functional structural and enzymatic components. Gag then functions in the budding of mature virions from the plasma membrane.(52) The Nef protein acts on the cellular environment to promote replication by inhibiting the host immunologic response to HIV (53-55) and inhibiting death of infected cells by apoptosis.(56)

Current HIV therapies inhibit the viral replication process at the binding and entry stage (fusion inhibitors), the reverse transcription stage (nucleoside and nonnucleoside reverse transcriptase inhibitors [NRTIs and NNRTIs, respectively]), or the protein cleavage stage (PIs). Inhibitors of coreceptor binding, integration, and maturation are in clinical trials.

transparent image
Immunology
transparent image

Individuals infected with HIV show both cellular and humoral (antibody) immune responses to the virus, but these responses are unable to prevent the ultimate progression of disease in the great majority of infected individuals. Cellular responses are mediated by CTLs (CD8 cells) and helper T lymphocytes (CD4 cells). CTLs inhibit HIV replication both directly, by recognizing and killing infected cells, and indirectly, by producing soluble chemokine antiviral factors.(57,58)

CTL-mediated killing of virally infected host cells occurs through direct contact, whereby the T-cell receptor on the surface of the CTL recognizes a fragment (epitope) of an HIV protein bound to a major histocompatibility complex (MHC) class I molecule on the surface of the infected host cell. After this interaction, the CTL releases enzymes that kill the infected cell. CTL responses directed against certain epitopes of the Gag protein have been associated with slower HIV disease progression than CTL responses against other epitopes.(59) CTLs also exert effects through soluble factors such as RANTES, macrophage inflammatory protein (MIP)-1-alpha, and MIP-1-beta, which inhibit HIV from infecting new cells by blocking HIV coreceptors.(60)

CD4 responses to HIV are important in viral control, and strong HIV-specific CD4 responses are associated with lower HIV viral loads.(61) CD4 cells respond to HIV antigens presented in conjunction with MHC class II molecules on the surface of infected cells. The fact that HIV infects CD4 cells themselves is an evolutionary strategy with a number of consequences. Because productive HIV infection occurs in activated CD4 cells, infection and killing of CD4 cells that are responding to HIV infection itself may cause a selective decrease in the number of HIV-specific CD4 cells. (HIV can also exist in nonactivated CD4 cells in a preintegrated form, which can become integrated if activation occurs within a few days.[62]) Additionally, as some of the activated, infected CD4 cells differentiate into resting memory CD4 cells, they may carry copies of the HIV genome in a postintegrated form that can persist for decades.(63) Current antiretroviral medications cannot efficiently eliminate the virus from cells in the resting state, leading to persistence of infection even in the presence of suppressive therapy.(63) Moreover, HIV continues to evolve under the selection pressure of the immune response that occurs in each infected individual, and mutations in the viral epitopes recognized by the immune system may enable the virus to escape the control of even broad and robust CD4 and CD8 HIV-specific responses.(64)

Depletion of CD4 lymphocytes is the hallmark of HIV infection, and predicts an individual's risk for infection with opportunistic pathogens as well as other complications of HIV disease. Evidence has shown that both increased peripheral destruction and decreased production of CD4 cells likely play a role in this decline.(65-69)

Humoral immunity appears to be less effective in controlling viremia than cellular responses, as HIV is remarkably effective at evading host antibody responses, and broadly neutralizing antibodies are rare.(70,71) The difficulty in eliciting broadly neutralizing antibody responses against HIV has posed a particularly difficult challenge to the development of a protective HIV vaccine.

transparent image
Epidemiology
transparent image

There are currently approximately 900,000 persons living with HIV/AIDS in the United States, 180,000 (20%) of whom are women and 10,000 (1.1%) of whom are children.(72) Young men who have sex with men (MSM) remain heavily affected. In a sample of 15- to 22-year-old MSM in 7 urban areas, 7% were already infected with HIV. Within this group, there were higher percentages of HIV infection among African Americans (14%) and Hispanics (7%) than whites (3%). In 2000 in the United States, over half of reported HIV infections among males aged 13-24 years were attributed to male-to-male sexual contact. Ethnic and racial minorities make up a disproportionate number of new AIDS cases in the United States. In 2000, the incidence rate among African Americans was 58.1 per 100,000 population, among Hispanics, 22.5, and among whites, 6.6.(73)

Worldwide, UNAIDS estimates that 40,000,000 persons are living with HIV/AIDS, 18,500,000 (44%) of whom are women, and 3,000,000 (7.1%) of whom are children (72,74). The most heavily affected area of the world is sub-Saharan Africa, with almost 30,000,000 people infected with HIV.(75) In countries with the highest prevalence such as Botswana and Zimbabwe, rates of HIV infection may exceed 30% in the general population and may be >50% among selected groups, including pregnant women, male patients at sexually transmitted infection clinics, and female sex workers.(75) Southeast Asia is estimated to have 6,000,000 infections, and China and the former Soviet Union are estimated to have more than 1,000,000 infections each. Infection rates are increasing in China, India, and Eastern Europe, fueled by high rates of intravenous drug use, increasing prevalence of other sexually transmitted diseases, and public health systems that are poorly prepared to prevent the spread of HIV.(76,77)

There were 2,400,000 deaths from AIDS in 2001, and some 14,000,000 children orphaned by the disease are living in the world.(78)

transparent image
Transmission and Risk Factors
transparent image

The primary method of spread of HIV infection worldwide is through sexual exposure. In the United States and Europe, acquisition of the virus through homosexual contact remains important, and there is some evidence of increasing incidence of infection among young gay men and ethnic minorities.(79) MSM, however, now account for <50% of new infections in the United States.(79) In the areas of highest HIV prevalence globally, heterosexual intercourse is the primary mode of transmission, accounting for approximately 70% of the overall sexual transmission.(80)

HIV has been isolated from blood, seminal fluid, pre-ejaculate, vaginal secretions, cerebrospinal fluid, saliva, tears, and breast milk of infected individuals.(81-84) HIV-1 DNA sequences have also been detected in pre-ejaculatory fluid.(85) In genital fluids, HIV may be found in both cell-free and cell-associated compartments, but it is unknown which is responsible for productive infection.(86) Viral concentrations in tears and saliva are comparatively low, and there are substances in saliva that appear to inhibit infectivity. No cases of HIV infection have been documented to arise from contact with nonbloody saliva or tears.

Transmission of HIV occurs more frequently through penile-anal intercourse and penile-vaginal intercourse than through fellatio, although clear cases of transmission through oral sex exist.(87) Female-to-female HIV transmission has been reported, but is rare.(88) In a meta-analysis, the overall efficacy of condoms in reducing HIV transmission was 69%.(89)

Sexual activity that is associated with exposure to infected blood increases the risk of transmission, as does the presence of genital ulcers.(90-92) Serum HIV viral load is strongly associated with heterosexual transmission between HIV-serodiscordant African sexual partners, where transmission was noted to be rare at viral loads <1,500 copies/mL.(93) The effect of viral load reduction with ART on HIV transmission is being investigated. Intervention with antiretroviral medications soon after high-risk sexual exposures has been proven to be safe and may be effective in preventing transmission of HIV (as discussed in the chapter "Prophylaxis Following Nonoccupational Exposure to HIV").(94)

Nonsexual HIV transmission can occur through transfusion with contaminated blood products, injection drug use, occupational exposure, or accidental needlesticks. The risk from occupational needlesticks to health care workers from known HIV-positive source patients in case series performed prior to the availability of potent ART was found to be 0.33-0.5%.(95,96) Factors increasing the risk of HIV acquisition from an occupational needlestick include deep injury, injury with a visibly bloody device, or injury with a device that had been previously used in the source patient's vein or artery.(96) Postexposure prophylaxis (PEP) has been associated with a reduction of HIV transmission after occupational needlestick events of approximately 80%.(96,97)

HIV transmission through transfusion of contaminated blood products was recognized early in the epidemic.(6) With current testing methods, the risk of acquiring HIV from a unit of transfused blood in the United States is 1 in 676,000,(98) but is significantly higher in many developing countries.

In the absence of interventions, mother-to-child transmission occurs in approximately 25% of live births to HIV-infected mothers.(99) Various regimens of antiretrovirals can reduce the rate of perinatal transmission by 50% or more.(99-102,103). Breast-feeding is also a risk factor for HIV transmission. Approximately one-third of cases of mother-to-child transmission result from breast-feeding, and the risk increases with the duration of breast-feeding.(104) Thus, interventions to prevent mother-to-child transmission at delivery may be largely negated if mothers are not provided with safe alternatives to breast-feeding.

transparent image
Classification of HIV Disease
transparent image

HIV damages the immune system, leaving the infected person vulnerable to a variety of infections (called "opportunistic" infections to indicate that they arise in the setting of immune impairment). The effect of HIV on the immune system is monitored by measuring the CD4 (helper) lymphocyte count in the blood. A normal CD4 count (between approximately 600 and 1,200 cells/µL) indicates that the immune system has not undergone sufficient damage to put the individual at risk for opportunistic illness. However, recent studies have demonstrated that even those with CD4 counts above 350-500 cells/µL are at elevated risk for a number of conditions that were not previously recognized as related to HIV infection. These include cardiovascular disease, kidney and liver disease, malignancies, and neurocognitive decline. As a result, experts have increasingly recommended initiation of treatment at higher CD4 counts. CD4 counts <500 cells/µL indicate that impairment of immune function is present, and are an indication for ART. CD4 counts <200 cells/µL indicate imminent risk of serious OIs or other complications of HIV disease, and prompt treatment is recommended.(35) [Editor's note: This paragraph was updated on December 8, 2009, to reflect new guidelines]

Untreated HIV disease is chronic and progressive. Primary HIV infection, often marked by a mononucleosis-like acute viral syndrome, is followed by a period of clinical latency typically lasting several years, during which high levels of viral replication and CD4 cell turnover lead to progressive immune dysfunction, eventually resulting in clinical disease progression. The distinction between "HIV infection" and "AIDS" is important, as it has clinical and prognostic implications, as well as utility in research.

The U.S. Centers for Disease Control and Prevention (CDC) definition of AIDS, initially published in 1986 and revised in 1993, is based on certain clinical conditions, infections, and malignancies associated with HIV infection (see Table 1). Additionally, AIDS may be defined by a CD4 count of <200 cells/µL or <14% of all lymphocytes, even in the absence of the listed conditions (see Table 2).(105)

The World Health Organization has developed a clinical staging system for HIV infection (see Table 3).(106) This system relies more heavily on clinical rather than laboratory evaluation, and has been used widely in resource-constrained areas where laboratory testing is not widely available.

transparent image
Natural History of Untreated HIV Infection
transparent image
transparent image
Primary HIV Infection
transparent image

Primary HIV infection is defined as the time period from initial infection with HIV to the development of an antibody response detectable by standard tests. Data from careful prospective evaluations of populations at risk for HIV infection demonstrate that up to 87% of individuals who acquire HIV may experience some symptoms of primary HIV infection.(107) The acute viral syndrome of primary HIV infection (sometimes referred to as "seroconversion illness") was first defined in 1985, with symptoms resembling those of mononucleosis appearing within days to weeks following exposure to HIV.(108,109). Symptoms may be mild or severe and may last from a few days to several weeks, with the average duration being 14 days. The most common presenting symptom is fever, seen in over 75% of patients.(110) Other commonly reported symptoms include fatigue, lymphadenopathy, headache, and rash. The rash, which is present in 40-80% of cases, may be evanescent, is typically maculopapular in character, and typically involves the trunk.(110) Evaluation of cohorts from Kenya (111) and India (112) found more frequent reports of joint pains, night sweats, and mucosal candidiasis and less frequent rash and pharyngitis in these study populations. A more severe clinical syndrome in primary HIV infection has been associated with a more rapid subsequent clinical course of HIV disease.(113)

The nonspecific symptoms of primary HIV infection may make diagnosis a challenge. In a study of high-risk individuals presenting with symptoms consistent with primary HIV infection, only 25% were diagnosed during their initial presentation.(107)

Diagnosis of HIV during the acute seroconversion phase requires not only high clinical suspicion but also an understanding of appropriate testing strategies. Routine HIV antibody testing may be negative for several weeks or even months after exposure in the so-called "window period."(114) During primary infection with HIV, plasma viral load often reaches very high levels in the range of millions of RNA copies/mL.(115,116) Thus, for individuals in whom primary HIV infection is clinically suspected, HIV RNA assays, which have a sensitivity approaching 100% and specificity of 97.4% in this setting,(117) should be included in the diagnostic evaluation. HIV RNA tests are not licensed for the diagnosis of HIV infection, and positive RNA tests during acute infection should be confirmed by documentation of subsequent HIV antibody conversion. The high levels of viremia seen in primary infection do not persist, however,(116) providing evidence of a host immune response capable of bringing the infection under some degree of control, at least in the short term.

During primary HIV infection, HIV-specific CD8 cells undergo a marked clonal expansion and express high levels of activation markers such as CD38 and human leukocyte antigen (HLA)-DR.(118) The breadth and strength of this CTL response correlate positively with the degree of viral control and inversely with the rapidity of clinical progression.(119-122)

CD4 counts and CD4 function may decline during primary HIV infection, occasionally to levels that allow OIs to develop.(123-125) Absolute CD4 count often rebounds after the primary infection, but may not return to a normal baseline. In patients with clinical progression of HIV disease, CD4 responses against HIV itself appear to remain particularly impaired following primary infection.(126)

After the initial reduction of viremia, a viral "set-point" is established in each infected individual. The magnitude of this set-point correlates with the rate of progression of HIV disease (see Table 5).(31,127,128) Studies of individuals during primary HIV infection have raised the question of whether the set-point might be reduced by early treatment.(129) Although early antiretroviral therapy may preserve immune function,(130) rapid control of viremia may also inhibit the full development of a mature immunologic response.(131) Carefully supervised interruptions of antiretroviral treatment after initial control during acute infection may permit the development of an effective immune response in the short term,(129) but long-term follow-up suggests that increases in viral load and emergence of drug resistance may occur in such patients.(132) The best strategy for treatment of acute HIV infection remains a matter of investigation.

transparent image
Chronic HIV Infection
transparent image

After the period of acute HIV infection--during which CD4 counts and viral load change dramatically--a relative equilibrium between viral replication and the host immune response is reached, and individuals may have little or no clinical manifestations of HIV infection. This time between initial infection and the development of AIDS may be long, averaging 10 years, even in the absence of treatment.(133)

Despite the relative clinical latency of this stage of HIV infection, viral replication and CD4 cell turnover remain active, with millions of CD4 cells and billions of virions produced and destroyed each day.(28) During this period, most infected individuals will have progressive loss of CD4 lymphocytes and perturbation of immune function.(134-137) On average, CD4 counts will drop by 50-90 cells/µL per year in asymptomatic individuals, usually with an acceleration of this rate over time.(138)

The rate of progression of infection may vary considerably. In adults, progression from infection to clinical AIDS is rare in the first 2 years of infection; however, reports describe rapid disease progression in infants infected by blood transfusion.(139) In a well-characterized cohort of HIV seroconverters who were identified in a retrospective analysis of stored serum samples from hepatitis B vaccine trials in the 1970s, 87% of infected individuals had developed AIDS by 17 years postseroconversion. Twelve percent maintained a CD4 count >500 cells/µL at 10 years, but only 3% maintained a CD4 count >500 cells/µL at 16 years after seroconversion.(140)

During chronic HIV infection, HIV RNA levels in plasma correlate with the rate of CD4 decline, with higher plasma viral loads predicting more rapid progression to AIDS and death.(141,142) An undetectable HIV RNA level in peripheral blood is associated with stable CD4 lymphocyte counts, and increases in HIV RNA correlate with more rapid rates of CD4 cell decline.(143,144)

The analogy of a train on a track (attributed to John Coffin of Tufts University, circa 1996) has been helpful in illustrating the independent contributions of CD4 count and HIV viral load in an individual person. If the infected individual is imagined as being on that train traveling toward a clinical event--such as acquiring an OI or dying from AIDS--the CD4 count provides information on the distance of the train from that destination, whereas the viral load provides information on the speed of the train in reaching the destination (see Figure 2).

transparent image
Clinical AIDS
transparent image

According to CDC criteria (see Table 1 and Table 2), AIDS is defined by either diagnosis of one of the AIDS-defining conditions, or by measurement of CD4 levels <200 cells/µL. Progression to AIDS from time of infection occurs, on average, 2 years earlier when defined by laboratory criteria (CD4 levels <200 cells/µL) compared to clinical criteria (development of an opportunistic illness).(145,146) Survival time from the development of AIDS varies according to the AIDS-defining event. In the Multicenter Hemophilia Cohort Study, median survival after a single AIDS-defining condition ranged from 3 to 51 months for the 10 most common conditions.(147) The mean survival time after diagnosis of AIDS in the United States prior to the availability of antiretroviral treatment was 10-12 months.(147)

transparent image
Special Considerations in Disease Progression
transparent image
transparent image
Host Factors
transparent image

A number of host factors influence HIV disease progression. Individuals who acquire HIV at an older age tend to have more rapid disease progression (134) and shorter survival times.(148) Variation in HIV coreceptor molecules, notably CCR5, influences both HIV susceptibility and disease progression. A mutant allele of CCR5 with a 32-base-pair deletion, CCR5-delta-32, is frequent in populations of European origin (10-15% of Caucasians are heterozygous, and 1% are homozygous), and encodes a nonfunctional truncated protein that is not transported to the cell surface. Homozygotes for the delta-32 allele exhibit a strong, although not complete, resistance to HIV infection, whereas heterozygotes display nearly normal rates of infection, but delayed progression to AIDS.(149)

Genetic differences in HLA alleles have also been shown to influence HIV disease susceptibility (150,151) and disease progression.(152-157) The class I alleles B35 and Cw4 have been associated with accelerated progression of disease,(158-160) as has general HLA homozygosity.(161) Because HLA class I alleles determine which viral epitopes can be presented to CD8 cells, greater diversity of HLA (heterozygosity) in an individual may reflect greater options for effective cell-mediated immunity to HIV. Conversely, HLA B27 and B57 have been associated with long-term nonprogression of HIV disease.(153) In particular, HLA B*5701 has been found to be highly overrepresented in long-term nonprogressors.(157)

Behavioral or psychological host factors may also influence HIV disease progression. More rapid HIV disease progression has been reported with unprotected anal intercourse,(162) smoking,(163) poor nutrition,(164) and depression (165); however, not all studies confirm these findings. Drug use might be expected to influence HIV disease progression, but studies of that question have produced mixed results.(162,166) Additionally, differences in disease course based on the route of HIV transmission have been difficult to prove.(167,168)

transparent image
Viral Factors
transparent image

HIV virions infect human cells by first binding to the CD4 receptor on the cell surface. This alone is not sufficient for the virus to enter the host cell; binding to an additional coreceptor is also required. Macrophage- or M-tropic viruses preferentially infect monocytes and macrophages, using the cell surface protein CCR5 (R5) as the preferred coreceptor to enter cells, and produce a nonsyncytium-inducing (NSI) phenotype in cell culture. Conversely, thymocyte- or T-tropic viruses preferentially infect T cells, use CXCR4 (X4) as the preferred coreceptor to enter cells, and produce a syncytium-inducing (SI) phenotype in cell culture.(169) Dual-tropic viruses, which may use either CCR5 or CXCR4 coreceptors, also exist. M-tropic viruses are frequently found in early HIV infection, and a switch to T-tropic strains in the course of disease is associated with rapid CD4 cell depletion.(170-172)

The concept of viral "fitness" refers to the pathogenicity of certain strains of HIV. HIV replicative capacity (RC) has been studied as a component of viral fitness. RC is a measure of the ability of a given virus to replicate successfully in a given environment.(173-177) During the course of drug treatment, mutations arise in the HIV reverse transcriptase and protease enzymes that make the virus resistant to particular drugs, thus conferring a selective advantage to that subpopulation that arises from a resistant variant.(178-180) Several of these mutations have been shown to cause a reduction in RC in the absence of drug when compared to wild-type virus.(173,174,176) Further accumulation of mutations over time under drug selection pressure may increase the "fitness" of the drug-resistant variant by further increasing phenotypic resistance,(178,181,182) or by increasing RC of the resistant virus.(174,183) The role of viral fitness on individual disease progression is just beginning to be understood.

Other viral factors may be important as well. For example, faster rates of disease progression have been observed in Ugandan individuals infected with subtype D compared with subtype A isolates.(184) Additionally, rare individuals who are infected with variant HIV strains, particularly those with a defective nef gene product, may experience slower disease progression.(185)

transparent image
Coinfections
transparent image

The development of OIs during HIV disease not only indicates the degree of immunosuppression, but may also influence disease progression itself. When stratified by CD4 counts, patients with prior histories of OIs have higher mortality rates than those without prior histories of OIs.(186)

Hepatitis C coinfection is common in HIV-infected patients, present in up to 40-50% of all patients in urban setting and in 90% of intravenous drug users.(187) HIV clearly leads to more rapid HCV disease progression; however, the effect of HCV infection on HIV progression is less clear. In a study of the Swiss HIV Cohort, HCV coinfection was associated with poorer CD4 responses to ART, development of new AIDS-defining events, and increased mortality (188); however, other authors have not found these associations.(189)

transparent image
Long-Term Nonprogressors
transparent image

A small subset of individuals infected with HIV--probably <5%--remain free of symptoms, achieve good control of HIV viral replication, and maintain high CD4 counts in the absence of antiretroviral medications over many years of infection, although some individuals initially identified as long-term nonprogressors (LTNPs) have experienced disease progression over time.(140) In general, LTNPs appear to have strong cellular immune responses to a variety of HIV antigens.(126,190,191)

transparent image
Laboratory Testing
transparent image
transparent image
HIV Antibody Testing
transparent image

(See chapter "HIV Antibody Assays")

HIV infection is usually diagnosed by testing serum for antibodies to HIV using a commercially available enzyme-linked immunosorbent assay (ELISA or EIA). Because the ELISA test is not entirely specific, positive results are confirmed with a Western blot assay, which identifies antibodies to specific components of HIV.(192,193). The 2-step process may mean that a patient must wait for a week or more to receive test results.

ELISA is quite sensitive in chronic HIV infection (although decline in antibody responses have been reported in advanced AIDS), but because antibody production does not occur immediately upon infection, an infected individual may test ELISA negative during a "window period" that varies in length from a few weeks to a few months after infection, depending on the individual case and assay used. Despite negative antibody testing during this window period, an individual may have high viral load and be at high risk of transmitting infection.

Newer methodologies allow antibody testing on saliva (194,195) and urine (195,196) specimens, although positive results should be confirmed with serologic testing. Home testing methods are also available.(197) Rapid HIV serum testing, with results available in 3-30 minutes, has shown 99-100% sensitivity and specificity compared to ELISA when tested in clinical settings,(198) including in resource-poor settings (199-201) and in pooled specimens.(202) In recent years, with the availability of rapid tests such as OraQuick (Abbott Laboratories, Abbott Park, IL) and Reveal (MedMira, Halifax, Nova Scotia, Canada), rapid testing protocols are being implemented in many countries, and will likely become commonplace.

transparent image
"Detuned" Antibody Testing
transparent image

By decreasing the sensitivity of ELISA assays, relatively recent infection (in which antibodies are present in lower concentrations and bind to HIV less effectively) can be distinguished from established infection (in which antibodies reach stable levels and have been selected for more avid binding to HIV). Soon after infection (but after the window period) an individual will test positive on the standard ELISA, but negative on the less-sensitive ("detuned") test. After maturation of the antibody response, both tests will give a positive result. Such "sensitive/less sensitive" or detuned ELISA testing strategies can be used to identify individuals who are in the early months of HIV infection and can help to identify incident infections in epidemiologic studies.(203,204)

transparent image
CD4 Testing
transparent image

The CD4 cell count in blood correlates with the risk of OIs in HIV disease,(205,206) and is therefore a useful marker for HIV disease staging. CD4 count is the main criterion for clinical decision making in guidelines developed in the United States for the prophylaxis of OIs (24,207) and for HIV treatment.(208)

The CDC recommends CD4 testing every 3-6 months in all HIV-infected persons,(209) but different intervals may be appropriate to the individual case. More than 1.6 million CD4 cell measurements are performed annually by approximately 600 testing laboratories in the United States.(210)

Because CD4 cells are a subset of all T lymphocytes, which are in turn a subset of all white blood cells, variations in CD4 count can occur in response to a variety of variables including concurrent infection, medications, stress, malnutrition, vitamin deficiencies, and normal diurnal variation. Often, these variables affect many subsets of lymphocytes and not exclusively CD4 cells; thus, the percentage of T lymphocytes that are CD4 positive will remain relatively stable. On the contrary, the depletion of T lymphocytes in HIV disease primarily affects CD4 cells, causing a relative CD4 cytopenia and a drop in the CD4 cell percentage. Additionally, an inversion of the normal CD4/CD8 cell ratio, which is usually >1 in non-HIV-infected individuals, may be seen with progressive CD4 cell depletion due to HIV. Thus, the CD4 percent and the CD4/CD8 ratio may help the clinician determine if a change in absolute CD4 count is due to the effects of HIV disease or to some other factor.

Until recently, most absolute CD4 cell counts were determined using 2 instruments, a hematology analyzer and a flow cytometer (dual-platform technology [DPT]). The CD4 count produced from DPT is the product of 3 laboratory measurements: the white blood cell count, the percentage of white blood cells that are lymphocytes (differential), and the percentage of lymphocytes that are CD positive (determined by flow cytometry). Single-platform technology (SPT) is designed to enable determinations of both absolute and percentage lymphocyte subset values using a single tube.(211) SPT, introduced for clinical application in 1996 is becoming the preferred method of CD4 count determination in a number of laboratories.(212)

Both SPT and DPT flow cytometry technology for CD4 count determination require specialized equipment and technician training. In resource-limited settings where CD4 count may be unavailable, the total lymphocyte count (TLC), which can be determined simply and cheaply, may be used as a surrogate for CD4 in determining stage of HIV infection.(213-215) For example, in a cohort of HIV-positive people in south India, a TLC of <1,400 cells/µL has been shown to be a good predictor of a CD4 count <200 cells/µL and thus an appropriate surrogate marker for initiating cotrimoxazole prophylaxis.(215) TLC may also have applications in monitoring response to antiretroviral therapy in place of or in conjunction with CD4 count. In an analysis of patients initiating a triple antiretroviral drug regimen, an increase in TLC was associated with an increase in CD4 count and a decrease in plasma viral load.(216)

transparent image
HIV Viral Load Testing
transparent image

(See chapter "HIV Viral Load Assays")

Three technologies exist to measure HIV viral load in serum: reverse transcription polymerase chain reaction (RT-PCR), branched DNA (bDNA), and nucleic acid sequence-based amplification assay (NASBA). The basic principles underlying these assays are similar--HIV is detected using DNA sequences that bind specifically to those in the virus--but results may vary between tests. Whereas early versions of the bDNA and RT-PCR techniques showed 2- to 2.5-fold differences in results, the version 3.0 bDNA assay and version 1.5 RT-PCR test yield values that are highly correlated (r = 0.96) and in good agreement (92.7%).(217,218) Testing of non-clade B HIV-1 viruses using these methods may not yield such highly correlated results.(219) Thus, it is advised that clinicians consistently use the same test when possible to compare results over time.

Because viral load may vary by orders of magnitude, results of viral load testing are often expressed in log units, where each increase of 1 log corresponds to a factor of 10. Thus, a viral load of 1,000 would be 3 log units, and the difference between a viral load of 1,000 and 10,000 would be 1 log unit. A change in viral load of >0.5 log copies/mL (approximately 3-fold) exceeds assay and diurnal variations, and may be considered to represent a true biological event, whereas a change of <0.5 log copies/mL cannot be distinguished from random variability. Diurnal variation in stable HIV viral loads is approximately 0.4 log copies/mL.(220) Acute intercurrent infection (221) or immunization (222,223) may also transiently increase viral load.

transparent image
HIV Antigen Testing
transparent image

Assays for HIV antigens, notably the p24 antigen encoded by the gag gene, can be used to screen donated blood products.(224) Measurement of p24 antigen may also be a less expensive yet effective alternative to HIV RNA testing in monitoring response to treatment.(225) Testing for p24 may also be used to diagnose early HIV infection, because this viral antigen can be detected in the blood of infected individuals prior to the development of antibodies (seroconversion) detectable by ELISA or Western blot tests. In identifying primary HIV infection, p24 is more specific (99% vs. 95-97%) but less sensitive (79% vs. 100%) than HIV RNA determinations (either PCR or bDNA).(117)

transparent image
HIV Resistance Testing
transparent image

(See chapters "Assays for Antiretroviral Resistance" and "Genotypic Testing for HIV")

Resistance to antiretroviral drugs is unfortunately common in treated populations. Resistance testing can be useful in determining which drugs not to use in a treatment-experienced patient whose viral load is increasing despite therapy, or in a previously untreated individual who may have been infected with HIV resistant to one or more drugs.

Two types of HIV resistance testing are available. Genotypic assays detect genetic mutations in the coding regions of the protease and reverse transcriptase enzymes in HIV isolated from the patient. Using the results, standardized algorithms are applied to predict resistance to various antiretrovirals. Phenotypic assays are more similar to standard bacteriologic sensitivity assays in that they are performed by culturing a fixed inoculum of HIV genetic material isolated from the patient with serial dilutions of individual antiretroviral drugs.

Prospective trials of genotypic (226-228) and phenotypic (229) assays have shown benefit with each of these assays in achieving virologic control in patients failing antiretroviral regimens. Additionally, with increasing incidence of drug resistance in individuals recently infected with HIV,(230) resistance testing during acute or early HIV infection may have important long-term clinical relevance. Resistance testing in chronically HIV-infected individuals provides information only on resistance to the medications being taken at the time of the test. In individuals who have changed or interrupted antiretroviral treatment, HIV harboring resistance mutations selected by prior treatment may be "archived" as proviral DNA in long-lived resting lymphocytes or macrophages, and may not be detected by resistance tests. Archived strains, however, can be expected to return to dominance if selected by the drugs to which they possess resistance. Results of resistance testing are therefore not a substitute for the patient's clinical antiretroviral history, which must also be taken into account. Further, because many resistance mutations tend to become outgrown by wild-type virus when the drug in question is no longer present to select for the resistance mutation, resistance testing in chronically infected individuals who are not on ART at the time of testing is unlikely to be of use and may provide misleading information.

In general, genotypic assays are more easily available, cheaper, and more rapidly performed than phenotypic assays. A genotype result is more likely than a phenotype to detect resistance from a minor variant or population mixture. Genotype testing identifies only the dominant strains representing >10-20% of virus circulating in blood at the time of testing.(231,232). Genotypic assays require an HIV viral load of >1,000 copies/mL to be reliable. Different laboratories use different algorithms for determining drug resistance from a given genotype result, and those different algorithms often produce discordant results, particularly for NRTIs.(233)

A phenotypic resistance test has the advantage that results are generated in a minimal inhibitory concentration (MIC) format that is more familiar to many clinicians and also emphasizes the gradation of resistance that often exists; however, the phenotypic threshold value that correlates with clinical resistance is still debated for certain antiretrovirals. A phenotype gives results for one drug at a time and is unable to predict the effects of combinations of drugs. Phenotypic resistance assays may be particularly useful when combined with monitoring of drug levels in treating individuals with highly resistant virus.

The "virtual phenotype" approach combines databases of matched genotypes and phenotypes from the same viral isolates to predict the phenotypic susceptibility of viruses with known genotypic sequences.

Current guidelines in the United States recommend resistance testing in cases of acute or recent HIV infection, for certain patients who have been infected as long as 2 years or more prior to initiating therapy, in cases of antiretroviral failure, and during pregnancy.(234)

It is worth reemphasizing that, given the limitations of all currently available resistance assays to detect archived mutations and minor variants, results of resistance testing must always be interpreted in the context of prior antiretroviral history and previous resistance testing.

transparent image
Therapeutic Drug Monitoring
transparent image

The measurement of antiretroviral drug concentrations in patient serum may be used to predict toxicity,(235) maximize efficacy,(236) assess effects of drug-drug interactions,(237,238) and provide evidence regarding medication adherence.(239)

Therapeutic drug monitoring (TDM) requires proper timing of sampling relative to dosing and meals, and sampling the appropriate body compartment. For PIs and NNRTIs, drug concentrations are measured in the plasma compartment, whereas for nucleoside and nucleotide reverse transcriptase inhibitors, measurement of intracellular metabolites is necessary.

In general, data on the efficacy of TDM in clinical practice are mixed. In certain circumstances, such as in pregnant or pediatric patients, TDM may provide data on drug concentrations that have not otherwise been well characterized, but data from large studies are lacking to support its routine use in clinical care.(240)

transparent image
Other HIV Testing Techniques
transparent image

ELISA or HIV viral load testing of fluids other than blood (seminal and vaginal fluid, cerebrospinal fluid, urine, and saliva) are currently available or under investigation. The clinical applications of some of these methods are well proven (ie, diagnosis of HIV infection with saliva or urine ELISA testing [241]), whereas for others it is less so (ie, serum viral load as a predictor of infectivity [242] or testing of semen for use in in vitro fertilization [243]).

Various culture techniques are available to isolate HIV from patient specimens.(244) HIV can be quantitated by determination of proviral DNA in peripheral blood mononuclear cells.(245) Proviral DNA has been used to test babies for HIV infection who were born to HIV-positive mothers.(246)

Assays of HIV antigens, notably p24 antigen, can be used to screen donated blood products.(224) It may also be an effective, inexpensive alternative to HIV RNA testing in monitoring response to treatment.(225) Additionally, because it is a direct viral antigen, p24 can be detected in the blood of infected individuals prior to the seroconversion detected by ELISA or Western blot tests. When comparing p24 antigenemia to HIV RNA determinations (either PCR or bDNA) in identifying primary HIV infection, p24 is equally specific (99% vs. 95-97%) but less sensitive (79% vs. 100%).(117)

transparent image
Treatment of HIV Infection
transparent image
transparent image
Overview of Antiretroviral Medications
transparent image

(See "Overview of Antiretroviral Drugs" )

Currently, all FDA-approved antiretroviral medications work by inhibiting 1 of 3 steps in the life cycle of HIV:

  1. Blocking the reverse transcriptase (RT) enzyme. The RT enzyme is used by the virus to convert its RNA into DNA after the virus enters the cell but before it enters the nucleus. All nucleoside and nucleotide analogues as well as NNRTIs function by interfering with the activity of this enzyme.

  2. Blocking the protease enzyme. Protease inhibitors, as their name indicates, inhibit the action of the HIV protease, namely, cleaving protein products of the viral structural genes into the functional subunits needed to create new infectious virions.

  3. Inhibiting fusion of the viral and host membranes. By attaching itself to the HIV envelope glycoprotein gp41, fusion inhibitors prevent formation of the "hairpin" structure (see "Virology", above) required for fusion of the HIV and host cell membranes, and thus prevent viral entry into the host cell.

New antiretroviral medications that are in development include improved formulations of currently approved drugs (to enhance bioavailability, increase half-life, or reduce adverse effects); new drugs in the same classes as currently approved drugs (such as PIs or NNRTIs with fewer adverse effects or unique resistance patterns); and drugs with novel mechanisms of action (eg, integrase inhibitors, entry inhibitors, and HIV coreceptor blockers).

transparent image
Initiating Treatment
transparent image

Not all HIV-infected individuals require ART at a given point in time. For those whose CD4 count and clinical assessment indicate a low risk of imminent disease progression, the potential adverse effects of immediate treatment would be expected to outweigh any benefit. Others may have psychosocial barriers to adherence that preclude effective ART, at least in the short term, until conditions related to these issues can be improved (eg, through stable housing, substance abuse counseling, or treatment of medical or psychiatric conditions). Because incomplete adherence can rapidly lead to lasting resistance against available antiretrovirals, it is usually worthwhile to address issues that are likely to impair adherence before starting ART. Still other individuals may opt for complimentary/alternative medicine (CAM) for management of their disease. In one large population-based study, 3% of individuals with HIV chose to use CAM as a substitute for standard therapy.(247) Unfortunately, there is no convincing evidence that CAM is effective in improving clinical status or survival in HIV infection.

Individuals for whom initiation of ART is not indicated must be monitored closely for changes in immune status (eg, CD4 counts) that might signal increased risk of OIs and thus trigger initiation of ART, OI prophylaxis, or other intervention.(24,207) In general, determination of CD4 counts and viral loads should be made every 3-6 months in individuals with stable infection. Measurements of CD4 count may be transiently decreased and measurements of HIV viral load transiently increased during acute infection with another pathogen (248-251) or immediately after vaccination,(252-255) and providers should avoid measurement of these parameters in such situations.

The decision to initiate (or reinitiate) ART should be made with clear goals that address the concerns of the patient as well as the provider. Active viral replication in the presence of antiretroviral drugs can be expected to result in selection of drug-resistant virus leading ultimately to treatment failure. Therefore, any antiretroviral regimen should ideally be prescribed with the intention of complete viral suppression. When complete suppression is not possible, secondary goals of treatment may include partial virologic suppression (which might be the best attainable goal if the virus is highly resistant to antiretrovirals), immunologic reconstitution, or alleviation of symptoms (such as fever, night sweats, or weight loss). Sufficient patient education on the antiretroviral regimen chosen--dosing schedule, meal restrictions, anticipated adverse effects and adverse effect management--is also crucial to the success of any treatment regimen. Individual adherence is one of the most challenging and most important aspects of treatment success.(256,257)

In initiating or changing an antiretroviral regimen, patients and clinicians must be aware of the development of potential drug adverse effects or toxicities--in both the short and long term (see Table 4).

transparent image
Monitoring ART
transparent image

The efficacy of an antiretroviral regimen should be monitored by regular determinations of HIV viral load and CD4 count. The first indication of successful treatment is a decline in viral load. The decline in viral load is biphasic.(258) The first (fast) decay phase occurs during the first 2 weeks of treatment for most patients; a second (slow) phase of decay is evident thereafter. The first phase may result primarily from antiviral effects of medications, whereas immune factors such as CTL antiviral activity may contribute to the elimination of virally infected cells in the second phase.(259) The kinetics of response are variable among individuals, and do not appear to be entirely determined by adherence or drug levels. Moreover, although viral decay rates are not entirely predictive of subsequent virologic failure,(259) virologic response at 4 weeks of treatment has been shown to correlate with response at 48 weeks.(260) As a rule of thumb, in a successful regimen the viral load should decline by at least 1 log by 4 weeks after treatment initiation; slower rates should prompt closer assessment of patient adherence, viral resistance, and possible drug-drug interactions. Even a successful regimen, however, may take 4-6 months or even longer to attain undetectable viral loads.

As viral load declines, the number of circulating CD4 cells begins to increase. Initial increases in CD4 count during the first 1-3 months of therapy are believed to be caused primarily by a redistribution of cells trapped from lymphoid tissue.(261) The subsequent rate of improvement in CD4 levels is variable among individuals, and gradual CD4 count increases may continue for many years with therapy that effectively suppresses HIV replication.(262-264) Individuals with lower nadir CD4 counts may have a slower and less complete CD4 recovery, whereas those who start therapy with higher CD4 counts and maintain continued viral suppression with antiretroviral medications may reach CD4 counts similar to those of HIV-uninfected individuals.(264)

As the immune system begins to reconstitute itself in the early phases of ART, it may mount vigorous inflammatory responses against certain pathogens. The restoration of immune function may therefore cause paradoxical worsening of certain coexisting OIs, a phenomenon known as "immune reconstitution syndrome." Infections for which immune reconstitution syndrome has been reported include pneumocystosis,(265) cryptococcosis,(266) cytomegalovirus (CMV) retinitis,(267) Mycobacterium avium complex infection,(268) and tuberculosis.(269) Immune reconstitution syndrome generally presents as inflammation of tissues involved in the infection (eg, marked lymphadenopathy in tuberculosis, potentially sight-threatening inflammation of the vitreous in CMV retinitis), presumably reflecting localized antigen load. The syndrome appears to occur most frequently in individuals with low CD4 counts and develops in the initial weeks of ART. It may present a diagnostic challenge to the clinician, and should be evaluated thoroughly to exclude other etiologies such as failure of therapy or other infections. Exact diagnostic criteria and guidelines for management of immune reconstitution syndrome are being developed. No clear guidelines for the management of immune reconstitution syndrome exist; however some approaches include using systemic corticosteroid treatment, or stopping antiretrovirals in the most severe cases. For more information on immune reconstitution inflammatory syndromes, see "Clinical Implications of Immune Reconstitution in AIDS".

transparent image
Changing ART
transparent image

Antiretroviral regimens may fail to suppress viral replication for a number of reasons, including incomplete adherence, poor absorption, toxicity leading to missed or lowered doses, pharmacokinetic interaction, suboptimal antiviral potency, and preexisting drug resistance. Virologic failure has been reported in as many as 63% of patients in population-based studies.(270,271) However, virologic outcomes may be improving over time; in a recent large cohort study, 72% of subjects on therapy attained HIV RNA <500 copies/mL at 6 months.(272)

At the time of changing antiretroviral regimens, careful assessment of the reasons for changing should be undertaken. Understanding the factors of adherence, toxicity, and resistance that prompted the switch will aid in designing subsequent regimens and optimizing patient outcomes. Changes to regimens for reasons of toxicity may be as simple as exchanging a new drug for the one that was causing the toxicity, provided that virus resistant to the other drugs in the regimen has not emerged. Patients who have failed multiple regimens or who have highly drug-resistant virus may require a more complicated "salvage" regimen. Such regimens may contain more drugs, may involve more complicated dosing schedules and may include experimental agents. Complete viral suppression may not be an achievable goal in some patients, decisions to change or continue treatment should take into account immunologic and clinical stability,(217) as well as the observation that selective pressure by antiretrovirals can maintain drug-resistant mutations that impair viral RC.(273,274)

transparent image
Treatment Interruption
transparent image

Situations arise when interruption of therapy is necessary in a given patient--toxicity, severe illness, and other circumstances making adherence impossible. In these situations, clinicians and patients should be aware of the recommendations to stop all antiretrovirals at once (or possibly stagger according to each drug's half-life) to minimize the possible emergence of resistance.(35)

Intentional supervised (or structured) treatment interruption (STI) presents several theoretical benefits: minimizing drug exposure, possibly decreasing short- and long-term adverse effects; "autovaccinating" individuals with their own virus in hopes of boosting the host immune control of HIV; allowing reversion of resistant virus to wild type, potentially creating a more drug-sensitive target for salvage therapy; and reducing the cost of therapy.

Studies conducted thus far demonstrate that STI does not increase HIV-specific immune response by acting as "autoimmunizations," and does not lead to persistent control of viremia.(275-281) A possible exception is in the unique situation of treatment during acute infection.(129,282) Studies of STI in conjunction with immune modulators such as interleukin (IL)-2 or therapeutic vaccination are underway.

A trial of intermittent therapy (2 months on, 1 month off) demonstrated no significant difference in virologic or immunologic markers compared to continuous therapy, but did show increased development of drug resistance in the intermittent therapy arm (3 of 8 patients on efavirenz-based regimens developed NNRTI resistance).(283) Using STIs composed of shorter cycles (eg, 1 week on, 1 week off) or guided by CD4 count or viral load parameters, other investigators have shown that these types of intermittent therapy may be able to reduce drug toxicity, reduce time on drug, reduce cost, and improve quality of life without sacrificing clinical control of HIV.(284,285)

Two large trials have examined STI as a part of salvage therapy. In 1 study, highly treatment-experienced patients with advanced disease and drug-resistant virus were randomized to immediate salvage therapy versus salvage therapy following a 4-month treatment interruption.(286) That trial showed no benefit to the STI group, which in fact demonstrated a higher rate of AIDS-associated events, causing accrual to be closed early. In a second study, highly treatment-experienced patients were randomized to a 2-month STI versus no STI prior to initiating a 7- to 8-antiretroviral drug + hydroxyurea salvage regimen.(287) In this study, STI patients showed a benefit in viral load and CD4 cell response through 48 weeks. The disparate results in these trials are not easily reconciled. Differences in the patient population and duration of STI exist, but still fail to completely explain the differences in outcomes.

In general, studies of the safety and efficacy of STIs do not support their use in routine care, and STI cannot be recommended outside of a research setting.(35)

In a modification of this concept, the idea of partial treatment interruptions has emerged as a possibility for treating drug-resistant HIV.(288) In a small, selected cohort of patients with persistent viremia on a stable PI- and NRTI-containing antiretroviral regimen, discontinuation of the PI component of the regimen resulted in stable viremia, reduced toxicity, and halted accumulation of new PI mutations. Discontinuation of the NRTI component of the regimen, however, was associated with rapid rises in viral load.(274) This concept, too, must still be considered experimental and cannot be recommended for routine practice.

transparent image
Immune Modulators for HIV Treatment
transparent image

Control of HIV viremia depends on host immune response as well as exogenous intervention with antiretroviral medications. In an effort to enhance host immune response to HIV, several techniques of immune modulation have been studied. Early attempts at immune modulation through bone marrow transplantation, donor lymphocyte infusions, and cytokine infusions demonstrated no consistent, durable clinical benefit. Immunomodulatory approaches may prove more effective in combination with effective ART.

In the context of established infection, therapeutic immunization has been studied as a means to enhance HIV-specific immune responses. No study to date has demonstrated sustained clinical benefit to such "therapeutic vaccination." Some studies have demonstrated improved HIV-specific immune responses after therapeutic vaccination,(289) but the clinical significance of these responses remains unclear.

IL-2, a cytokine released by activated CD4 cells that regulates T-cell proliferation and maturation, has been studied as an immune modulator in HIV-infected patients. Administration of IL-2 to individuals with controlled HIV viremia leads to increased CD4 counts,(290-292) expansion of both memory and naive CD4 pools,(292) and decreased T-cell activation.(292) IL-2 administration is accompanied by numerous adverse effects and toxicities. No clinical benefits of IL-2 have yet been demonstrated. Large clinical trials of IL-2 are underway. Other cytokines, such as IL-12 (293) and IL-4 (294) are being studied, as is IL-2 in combination with therapeutic vaccination.

Techniques of immune manipulation through infusion of antigen-presenting cells that have been activated in vitro,(295) infusion of expanded (296) or activated (297) CD4 cells, and genetic manipulation of cells to induce anti-HIV CTL activity (298) are also under investigation, but even if such resource-intensive, individualized approaches are shown to be effective, it is doubtful that they will become accessible to most people infected with HIV.

Administration of human growth hormone (hGH) as a method of enhancing CD4 recovery in HIV is another technique being studied, spurred by the observations of increased circulating naive CD4 cells and increased thymic mass, suggesting increased thymopoeisis, during hGH treatment.(299)

transparent image
Prophylaxis Against OIs
transparent image

Many commonly encountered OIs may be prevented by administering prophylactic antibiotics to those at risk, based on pathogen-specific CD4 cell count thresholds.(207) In geographic areas where local epidemiology of opportunistic pathogens varies from that of the United States, different recommendations for primary prophylaxis (treatment to prevent an initial episode of an OI) may be appropriate. In general, secondary prophylaxis (treatment to prevent recurrence of an OI) should be provided as long as immune impairment persists.

Among individuals who experience reconstitution of immunologic function with ART, as assessed by sustained increases in CD4 count to levels above those associated with OI, discontinuation of primary prophylaxis or secondary prophylaxis has been shown to be safe.(300)

transparent image
Routine Health Care Maintenance in HIV Infection
transparent image

All individuals, whether on ART or not, will have other health care needs, some related to HIV infection and some not. Individuals with HIV infection must be considered to be at risk for other blood-borne pathogens and sexually transmitted infections. All HIV-infected individuals should be screened for viral hepatitis A, B, and C, and immunized (against A and/or B) or treated as appropriate. Routine screening for syphilis, chlamydia, gonorrhea, and other sexually transmitted diseases should be done according to the individual's risk behavior. Age- and gender-appropriate cancer screening should also be done in HIV-infected individuals, with special recommendations for increased screening for cervical and anal dysplasia associated with human papillomavirus (HPV) disease.(301-303) Annual screening for tuberculosis with purified protein derivative (PPD) testing is indicated, particularly in high incidence areas.

Hyperlipidemia, glucose intolerance, and insulin resistance are common consequences of antiretroviral therapy.(304-306) With indications of potential increases in cardiac events among HIV-infected individuals, careful attention should be paid to modifiable cardiac risk factors.(307) Routine monitoring of fasting lipid panels, particularly among patients on antiretrovirals, and lipid management that follows the guidelines of the National Cholesterol Education Program (NCEP) is recommended.(308)

Routine immunizations for HIV-infected individuals should follow standard guidelines with a few exceptions. Oral polio vaccine and smallpox vaccine are contraindicated in HIV-infected individuals and in those with whom they have significant contact. The risks of other live vaccines such as measles, mumps, and rubella must be weighed against the potential benefits of vaccination. Data on varicella vaccination in HIV-infected adults are lacking. Pneumococcal vaccination is recommended every 5 years for those with HIV infection, as is annual influenza vaccine. Vaccination may be more effective in individuals with relatively intact immune systems (CD4 count >200 cells/µL), and for individuals beginning ART, may be delayed until such a goal is reached.

Discussions of sexual health should involve education to reduce risk of transmitting HIV to uninfected partners, as well as preventing other sexually transmitted diseases. Contraceptive options and issues of family planning should be addressed regularly with individuals of reproductive potential.

Other routine health measures such as blood pressure determination, depression and domestic violence screening, smoking cessation interventions, drug and alcohol counseling, and dental and ophthalmologic evaluation should be used with HIV-infected individuals, just as with HIV-uninfected individuals.

transparent image
Summary and Future Directions
transparent image

Clinical care of patients with HIV requires familiarity with a wide range of medical, social, economic, and scientific issues. Despite the limitations of current forms of ART, it is clear that HIV replication can be effectively suppressed. Current challenges include improving upon existing antiretroviral therapies, developing new therapies to control and eliminate virus and enhance the immune system, extending current and future therapies to all individuals in need of treatment worldwide, and developing effective strategies to prevent HIV infection.

transparent image
transparent image

References

transparent image
1.   Pneumocystis pneumonia--Los Angeles. MMWR Morb Mortal Wkly Rep 1981; 30:250-2.
transparent image
2.   Kaposi's sarcoma and Pneumocystis pneumonia among homosexual men--New York City and California. MMWR Morb Mortal Wkly Rep 1981; 30:305-8.
transparent image
3.   A cluster of Kaposi's sarcoma and Pneumocystis carinii pneumonia among homosexual male residents of Los Angeles and Orange Counties, California. MMWR Morb Mortal Wkly Rep 1982; 31:305-7.
transparent image
4.   Opportunistic infections and Kaposi's sarcoma among Haitians in the United States. MMWR Morb Mortal Wkly Rep 1982; 31:353-4, 360-1.
transparent image
5.   Pneumocystis carinii pneumonia among persons with hemophilia A. MMWR Morb Mortal Wkly Rep 1982; 31:365-7.
transparent image
6.   Possible transfusion-associated acquired immune deficiency syndrome (AIDS) - California. MMWR Morb Mortal Wkly Rep 1982; 31:652-4.
transparent image
7.   Unexplained immunodeficiency and opportunistic infections in infants--New York, New Jersey, California. MMWR Morb Mortal Wkly Rep 1982; 31:665-7.
transparent image
8.   Masur H, Michelis MA, Wormser GP, Lewin S, Gold J, Tapper ML, Giron J, Lerner CW, Armstrong D, Setia U, Sender JA, Siebken RS, Nicholas P, Arlen Z, Maayan S, Ernst JA, Siegal FP, Cunningham-Rundles S. Opportunistic infection in previously healthy women. Initial manifestations of a community-acquired cellular immunodeficiency. Ann Intern Med 1982; 97:533-9.
transparent image
9.   Immunodeficiency among female sexual partners of males with acquired immune deficiency syndrome (AIDS) - New York. MMWR Morb Mortal Wkly Rep 1983; 31:697-8.
transparent image
10.   Acquired immune deficiency syndrome (AIDS) in prison inmates--New York, New Jersey. MMWR Morb Mortal Wkly Rep 1983; 31:700-1.
transparent image
11.   Clumeck N, Mascart-Lemone F, de Maubeuge J, Brenez D, Marcelis L. Acquired immune deficiency syndrome in Black Africans. Lancet 1983; 1:642.
transparent image
12.   Barre-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, Gruest J, Dauguet C, Axler-Blin C, Vezinet-Brun F, Rouzioux C, Rozenbaum W, Montagnier L. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science 1983; 220:868-71.
transparent image
13.   Gallo RC, Salahuddin SZ, Popovic M, Shearer GM, Kaplan M, Haynes BF, Palker TJ, Redfield R, Oleske J, Safai B, et al. Frequent detection and isolation of cytopathic retroviruses (HTLV-III) from patients with AIDS and at risk for AIDS. Science 1984; 224:500-3.
transparent image
14.   Levy JA, Hoffman AD, Kramer SM, Landis JA, Shimabukuro JM, Oshiro LS. Isolation of lymphocytopathic retroviruses from San Francisco patients with AIDS. Science 1984; 225:840-2.
transparent image
15.   Coffin J, Haase A, Levy JA, Montagnier L, Oroszlan S, Teich N, Temin H, Toyoshima K, Varmus H, Vogt P, et al. Human immunodeficiency viruses. Science 1986; 232:697.
transparent image
16.   Antibodies to a retrovirus etiologically associated with acquired immunodeficiency syndrome (AIDS) in populations with increased incidences of the syndrome. MMWR Morb Mortal Wkly Rep 1984; 33:377-9.
transparent image
17.   Byers VS, Levin AS, Malvino A, Waites L, Robins RA, Baldwin RW. A phase II study of effect of addition of trichosanthin to zidovudine in patients with HIV disease and failing antiretroviral agents. AIDS Res Hum Retroviruses 1994; 10:413-20.
transparent image
18.   Pert CB, Hill JM, Ruff MR, Berman RM, Robey WG, Arthur LO, Ruscetti FW, Farrar WL. Octapeptides deduced from the neuropeptide receptor-like pattern of antigen T4 in brain potently inhibit human immunodeficiency virus receptor binding and T-cell infectivity. Proc Natl Acad Sci U S A 1986; 83:9254-8.
transparent image
19.   Mildvan D, Buzas J, Armstrong D, Antoniskis D, Sacks HS, Rhame FS, Mosbach EW, Pettinelli C. An open-label, dose-ranging trial of AL 721 in patients with persistent generalized lymphadenopathy and AIDS-related complex. J Acquir Immune Defic Syndr 1991; 4:945-51.
transparent image
20.   Schooley RT, Merigan TC, Gaut P, Hirsch MS, Holodniy M, Flynn T, Liu S, Byington RE, Henochowicz S, Gubish E, et al. Recombinant soluble CD4 therapy in patients with the acquired immunodeficiency syndrome (AIDS) and AIDS-related complex. A phase I-II escalating dosage trial. Ann Intern Med 1990; 112:247-53.
transparent image
21.   Abrams DI, Kuno S, Wong R, Jeffords K, Nash M, Molaghan JB, Gorter R, Ueno R. Oral dextran sulfate (UA001) in the treatment of the acquired immunodeficiency syndrome (AIDS) and AIDS-related complex. Ann Intern Med 1989; 110:183-8.
transparent image
22.   Pedersen C, Sandstrom E, Petersen CS, Norkrans G, Gerstoft J, Karlsson A, Christensen KC, Hakansson C, Pehrson PO, Nielsen JO, et al. The efficacy of inosine pranobex in preventing the acquired immunodeficiency syndrome in patients with human immunodeficiency virus infection. The Scandinavian Isoprinosine Study Group. N Engl J Med 1990; 322:1757-63.
transparent image
23.   Fischl MA, Richman DD, Grieco MH, Gottlieb MS, Volberding PA, Laskin OL, Leedom JM, Groopman JE, Mildvan D, Schooley RT, et al. The efficacy of azidothymidine (AZT) in the treatment of patients with AIDS and AIDS-related complex. A double-blind, placebo-controlled trial. N Engl J Med 1987; 317:185-91.
transparent image
24.   1999 USPHS/IDSA guidelines for the prevention of opportunistic infections in persons infected with human immunodeficiency virus. Clin Infect Dis 2000; 30 Suppl 1:S29-65.
transparent image
25.   Shafer RW, Seitzman PA, Tapper ML. Successful prophylaxis of Pneumocystis carinii pneumonia with trimethoprim-sulfamethoxazole in AIDS patients with previous allergic reactions. J Acquir Immune Defic Syndr 1989; 2:389-93.
transparent image
26.   Hammer SM, Squires KE, Hughes MD, Grimes JM, Demeter LM, Currier JS, Eron JJ, Jr., Feinberg JE, Balfour HH, Jr., Deyton LR, Chodakewitz JA, Fischl MA. A controlled trial of two nucleoside analogues plus indinavir in persons with human immunodeficiency virus infection and CD4 cell counts of 200 per cubic millimeter or less. AIDS Clinical Trials Group 320 Study Team. N Engl J Med 1997; 337:725-33.
transparent image
27.   Palella FJ, Jr., Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, Aschman DJ, Holmberg SD. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998; 338:853-60.
transparent image
28.   Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, Markowitz M. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 1995; 373:123-6.
transparent image
29.   Mellors JW, Rinaldo CR, Jr., Gupta P, White RM, Todd JA, Kingsley LA. Prognosis in HIV-1 infection predicted by the quantity of virus in plasma. Science 1996; 272:1167-70.
transparent image
30.   Mellors JW, Munoz A, Giorgi JV, Margolick JB, Tassoni CJ, Gupta P, Kingsley LA, Todd JA, Saah AJ, Detels R, Phair JP, Rinaldo CR, Jr. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med 1997; 126:946-54.
transparent image
31.   Hughes MD, Johnson VA, Hirsch MS, Bremer JW, Elbeik T, Erice A, Kuritzkes DR, Scott WA, Spector SA, Basgoz N, Fischl MA, D'Aquila RT. Monitoring plasma HIV-1 RNA levels in addition to CD4+ lymphocyte count improves assessment of antiretroviral therapeutic response. ACTG 241 Protocol Virology Substudy Team. Ann Intern Med 1997; 126:929-38.
transparent image
32.   O'Brien WA, Hartigan PM, Martin D, Esinhart J, Hill A, Benoit S, Rubin M, Simberkoff MS, Hamilton JD. Changes in plasma HIV-1 RNA and CD4+ lymphocyte counts and the risk of progression to AIDS. Veterans Affairs Cooperative Study Group on AIDS. N Engl J Med 1996; 334:426-31.
transparent image
33.   O'Brien WA, Hartigan PM, Daar ES, Simberkoff MS, Hamilton JD. Changes in plasma HIV RNA levels and CD4+ lymphocyte counts predict both response to antiretroviral therapy and therapeutic failure. VA Cooperative Study Group on AIDS. Ann Intern Med 1997; 126:939-45.
transparent image
34.   Guidelines for the use of antiretroviral agents in HIV-infected adults and adolescents. Department of Health and Human Services and Henry J. Kaiser Family Foundation. MMWR Recomm Rep 1998; 47:43-82.
transparent image
35.   Dybul M, Fauci AS, Bartlett JG, Kaplan JE, Pau AK. Guidelines for using antiretroviral agents among HIV-infected adults and adolescents. Ann Intern Med 2002; 137:381-433.
transparent image
36.   Muesing MA, Smith DH, Cabradilla CD, Benton CV, Lasky LA, Capon DJ. Nucleic acid structure and expression of the human AIDS/lymphadenopathy retrovirus. Nature 1985; 313:450-8.
transparent image
37.   Gallo R, Wong-Staal F, Montagnier L, Haseltine WA, Yoshida M. HIV/HTLV gene nomenclature. Nature 1988; 333:504.
transparent image
38.   Hu DJ, Dondero TJ, Rayfield MA, George JR, Schochetman G, Jaffe HW, Luo CC, Kalish ML, Weniger BG, Pau CP, Schable CA, Curran JW. The emerging genetic diversity of HIV. The importance of global surveillance for diagnostics, research, and prevention. Jama 1996; 275:210-6.
transparent image
39.   Spira J, Wainberg MA, Loemba H. Impact of clade diversity on HIV-1 virulence, antiretroviral drug sensitivity and drug resistance. J Antimicrob Chemother. 2003 Feb;51(2):229-40.
transparent image
40.  Caride E, Hertogs K, Larder B. Genotyping and phenotyping analysis of B and non-B HIV-1 subtypes from Brazilian patients under HAART (abstract 164). Antiviral Therapy. 2000:5, Suppl. 3:128.
transparent image
41.  Pillay D, Sinka K, Rice P. Impact of HIV-1 subtype on NNRTI resistance mutations (abstract 163). Antiviral Therapy. 2000: 5, Suppl 3:128.
transparent image
42.   Goudsmit J, Back NK, Nara PL. Genomic diversity and antigenic variation of HIV-1: links between pathogenesis, epidemiology and vaccine development. Faseb J 1991; 5:2427-36.
transparent image
43.   Carmichael A, Jin X, Sissons P. Analysis of the human env-specific cytotoxic T-lymphocyte (CTL) response in natural human immunodeficiency virus type 1 infection: low prevalence of broadly cross-reactive env-specific CTL. J Virol 1996; 70:8468-76.
transparent image
44.   Kwong PD, Wyatt R, Robinson J, Sweet RW, Sodroski J, Hendrickson WA. Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody. Nature 1998; 393:648-59.
transparent image
45.   Scarlatti G, Tresoldi E, Bjorndal A, Fredriksson R, Colognesi C, Deng HK, Malnati MS, Plebani A, Siccardi AG, Littman DR, Fenyo EM, Lusso P. In vivo evolution of HIV-1 co-receptor usage and sensitivity to chemokine-mediated suppression. Nat Med 1997; 3:1259-65.
transparent image
46.   Chan DC, Kim PS. HIV entry and its inhibition. Cell 1998; 93:681-4.
transparent image
47.   Miller MD, Farnet CM, Bushman FD. Human immunodeficiency virus type 1 preintegration complexes: studies of organization and composition. J Virol 1997; 71:5382-90.
transparent image
48.   Adams M, Sharmeen L, Kimpton J, Romeo JM, Garcia JV, Peterlin BM, Groudine M, Emerman M. Cellular latency in human immunodeficiency virus-infected individuals with high CD4 levels can be detected by the presence of promoter-proximal transcripts. Proc Natl Acad Sci U S A 1994; 91:3862-6.
transparent image
49.   Wei P, Garber ME, Fang SM, Fischer WH, Jones KA. A novel CDK9-associated C-type cyclin interacts directly with HIV-1 Tat and mediates its high-affinity, loop-specific binding to TAR RNA. Cell 1998; 92:451-62.
transparent image
50.   Cullen BR. Retroviruses as model systems for the study of nuclear RNA export pathways. Virology 1998; 249:203-10.
transparent image
51.   Zimmerman C, Klein KC, Kiser PK, Singh AR, Firestein BL, Riba SC, Lingappa JR. Identification of a host protein essential for assembly of immature HIV-1 capsids. Nature 2002; 415:88-92.
transparent image
52.   Garnier L, Parent LJ, Rovinski B, Cao SX, Wills JW. Identification of retroviral late domains as determinants of particle size. J Virol 1999; 73:2309-20.
transparent image
53.   Le Gall S, Erdtmann L, Benichou S, Berlioz-Torrent C, Liu L, Benarous R, Heard JM, Schwartz O. Nef interacts with the mu subunit of clathrin adaptor complexes and reveals a cryptic sorting signal in MHC I molecules. Immunity 1998; 8:483-95.
transparent image
54.   Collins KL, Chen BK, Kalams SA, Walker BD, Baltimore D. HIV-1 Nef protein protects infected primary cells against killing by cytotoxic T lymphocytes. Nature 1998; 391:397-401.
transparent image
55.   Xu XN, Laffert B, Screaton GR, Kraft M, Wolf D, Kolanus W, Mongkolsapay J, McMichael AJ, Baur AS. Induction of Fas ligand expression by HIV involves the interaction of Nef with the T cell receptor zeta chain. J Exp Med 1999; 189:1489-96.
transparent image
56.   Greenway AL, McPhee DA, Allen K, Johnstone R, Holloway G, Mills J, Azad A, Sankovich S, Lambert P. Human immunodeficiency virus type 1 Nef binds to tumor suppressor p53 and protects cells against p53-mediated apoptosis. J Virol 2002; 76:2692-702.
transparent image
57.   Haynes BF, Pantaleo G, Fauci AS. Toward an understanding of the correlates of protective immunity to HIV infection. Science 1996; 271:324-8.
transparent image
58.   Walker CM, Moody DJ, Stites DP, Levy JA. CD8+ lymphocytes can control HIV infection in vitro by suppressing virus replication. Science 1986; 234:1563-6.
transparent image
59.   Pontesilli O, Klein MR, Kerkhof-Garde SR, Pakker NG, de Wolf F, Schuitemaker H, Miedema F. Longitudinal analysis of human immunodeficiency virus type 1-specific cytotoxic T lymphocyte responses: a predominant gag-specific response is associated with nonprogressive infection. J Infect Dis 1998; 178:1008-18.
transparent image
60.   Malnati MS, Tambussi G, Fischetti L, Algeri M, Veglia F, Capiluppi B, Lazzarin A, Lusso P. Analysis of serum and plasma beta chemokines in primary HIV infection (PHI). J Biol Regul Homeost Agents 2000; 14:75-8.
transparent image
61.   Rosenberg ES, Walker BD. HIV type 1-specific helper T cells: a critical host defense. AIDS Res Hum Retroviruses 1998; 14 Suppl 2:S143-7.
transparent image
62.   Perelson AS, Essunger P, Cao Y, Vesanen M, Hurley A, Saksela K, Markowitz M, Ho DD. Decay characteristics of HIV-1-infected compartments during combination therapy. Nature 1997; 387:188-91.
transparent image
63.   Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, Spina CA, Richman DD. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 1997; 278:1291-5.
transparent image
64.   McMichael AJ, Rowland-Jones SL. Cellular immune responses to HIV. Nature 2001; 410:980-7.
transparent image
65.   Wei X, Ghosh SK, Taylor ME, Johnson VA, Emini EA, Deutsch P, Lifson JD, Bonhoeffer S, Nowak MA, Hahn BH, et al. Viral dynamics in human immunodeficiency virus type 1 infection. Nature 1995; 373:117-22.
transparent image
66.   Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, Markowitz M. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 1995; 373:123-6.
transparent image
67.   Hellerstein M, Hanley MB, Cesar D, Siler S, Papageorgopoulos C, Wieder E, Schmidt D, Hoh R, Neese R, Macallan D, Deeks S, McCune JM. Directly measured kinetics of circulating T lymphocytes in normal and HIV-1-infected humans. Nat Med 1999; 5:83-9.
transparent image
68.   Rowland-Jones S. HIV infection: where have all the T cells gone? Lancet 1999; 354:5-7.
transparent image
69.   Mohri H, Perelson AS, Tung K, Ribeiro RM, Ramratnam B, Markowitz M, Kost R, Hurley A, Weinberger L, Cesar D, Hellerstein MK, Ho DD. Increased turnover of T lymphocytes in HIV-1 infection and its reduction by antiretroviral therapy. J Exp Med 2001; 194:1277-87.
transparent image
70.   Peterlin BM, Trono D. Hide, shield and strike back: how HIV-infected cells avoid immune eradication. Nat Rev Immunol 2003; 3:97-107.
transparent image
71.   Sattentau QJ. Neutralization of HIV-1 by antibody. Curr Opin Immunol 1996; 8:540-5.
transparent image
72.  UNAIDS AIDS Epidemic Update, December 2003. http://data.unaids.org/publications/IRC-pub06/JC943-EpiUpdate2003_en.pdf
transparent image
73.  CDC data. http://www.cdc.gov/hiv/pubs/facts/hispanic.htm.
transparent image
74.  UNAIDS Report on the global HIV/AIDS epidemic. http://hivinsite.ucsf.edu/pdf/UNAIDS/barcelona_table.pdf
transparent image
75.  CDC data. http://www.cdc.gov/hiv/pubs/facts/msm.htm.
transparent image
76.   Hamers FF, Downs AM. HIV in central and eastern Europe. Lancet 2003; 361:1035-44.
transparent image
77.   Kaufman J, Jing J. China and AIDS--the time to act is now. Science 2002; 296:2339-40.
transparent image
78.  WHO data from the end of 2002. http://www.who.int/hiv/pub/epidemiology/pubepidemic2002/en.
transparent image
79.   Holmberg SD. The estimated prevalence and incidence of HIV in 96 large US metropolitan areas. Am J Public Health 1996; 86:642-54.
transparent image
80.   Gayle H. An overview of the global HIV/AIDS epidemic, with a focus on the United States. Aids 2000; 14 Suppl 2:S8-17.
transparent image
81.   Ho DD, Byington RE, Schooley RT, Flynn T, Rota TR, Hirsch MS. Infrequency of isolation of HTLV-III virus from saliva in AIDS. N Engl J Med 1985; 313:1606.
transparent image
82.   Wofsy CB, Cohen JB, Hauer LB, Padian NS, Michaelis BA, Evans LA, Levy JA. Isolation of AIDS-associated retrovirus from genital secretions of women with antibodies to the virus. Lancet 1986; 1:527-9.
transparent image
83.   Hollander H, Levy JA. Neurologic abnormalities and recovery of human immunodeficiency virus from cerebrospinal fluid. Ann Intern Med 1987; 106:692-5.
transparent image
84.   Geier SA, Gurtler L, Klauss V, Mueller A, Bader L, Eberle J, Bogner JJ, Kronawitter U, Goebel FD, Lund OE. [Differences in detectability of human immunodeficiency virus type 1 in tears and blood lymphocytes]. Klin Monatsbl Augenheilkd 1992; 201:164-8.
transparent image
85.   Ilaria G, Jacobs JL, Polsky B, Koll B, Baron P, MacLow C, Armstrong D, Schlegel PN. Detection of HIV-1 DNA sequences in pre-ejaculatory fluid. Lancet 1992; 340:1469.
transparent image
86.   Alexander NJ. Sexual transmission of human immunodeficiency virus: virus entry into the male and female genital tract. World Health Organization, Global Programme on Acquired Immune Deficiency Syndrome. Fertil Steril 1990; 54:1-18.
transparent image
87.   Rothenberg RB, Scarlett M, del Rio C, Reznik D, O'Daniels C. Oral transmission of HIV. Aids 1998; 12:2095-105.
transparent image
88.   Monzon OT, Capellan JM. Female-to-female transmission of HIV. Lancet 1987; 2:40-1.
transparent image
89.   Weller SC. A meta-analysis of condom effectiveness in reducing sexually transmitted HIV. Soc Sci Med 1993; 36:1635-44.
transparent image
90.   Cameron DW, Simonsen JN, D'Costa LJ, Ronald AR, Maitha GM, Gakinya MN, Cheang M, Ndinya-Achola JO, Piot P, Brunham RC, et al. Female to male transmission of human immunodeficiency virus type 1: risk factors for seroconversion in men. Lancet 1989; 2:403-7.
transparent image
91.   Greenblatt RM, Lukehart SA, Plummer FA, Quinn TC, Critchlow CW, Ashley RL, D'Costa LJ, Ndinya-Achola JO, Corey L, Ronald AR, et al. Genital ulceration as a risk factor for human immunodeficiency virus infection. Aids 1988; 2:47-50.
transparent image
92.   Plummer FA, Simonsen JN, Cameron DW, Ndinya-Achola JO, Kreiss JK, Gakinya MN, Waiyaki P, Cheang M, Piot P, Ronald AR, et al. Cofactors in male-female sexual transmission of human immunodeficiency virus type 1. J Infect Dis 1991; 163:233-9.
transparent image
93.   Quinn TC, Wawer MJ, Sewankambo N, Serwadda D, Li C, Wabwire-Mangen F, Meehan MO, Lutalo T, Gray RH. Viral load and heterosexual transmission of human immunodeficiency virus type 1. Rakai Project Study Group. N Engl J Med 2000; 342:921-9.
transparent image
94.   Kahn JO, Martin JN, Roland ME, Bamberger JD, Chesney M, Chambers D, Franses K, Coates TJ, Katz MH. Feasibility of postexposure prophylaxis (PEP) against human immunodeficiency virus infection after sexual or injection drug use exposure: the San Francisco PEP Study. J Infect Dis 2001; 183:707-14.
transparent image
95.   Leentvaar-Kuijpers A, Dekker MM, Coutinho RA, Dekker EE, Keeman JN, Ansink-Schipper MC. Needlestick injuries, surgeons, and HIV risks. Lancet 1990; 335:546-7.
transparent image
96.   Cardo DM, Culver DH, Ciesielski CA, Srivastava PU, Marcus R, Abiteboul D, Heptonstall J, Ippolito G, Lot F, McKibben PS, Bell DM. A case-control study of HIV seroconversion in health care workers after percutaneous exposure. Centers for Disease Control and Prevention Needlestick Surveillance Group. N Engl J Med 1997; 337:1485-90.
transparent image
97.   From the Centers for Disease Control and Prevention. Case-control study of HIV seroconversion in health-care workers after percutaneous exposure to HIV-infected blood--France, United Kingdom, and United States, January 1988-August 1994. Jama 1996; 275:274-5.
transparent image
98.   Schreiber GB, Busch MP, Kleinman SH, Korelitz JJ. The risk of transfusion-transmitted viral infections. The Retrovirus Epidemiology Donor Study. N Engl J Med 1996; 334:1685-90.
transparent image
99.   Connor EM, Sperling RS, Gelber R, Kiselev P, Scott G, O'Sullivan MJ, VanDyke R, Bey M, Shearer W, Jacobson RL, et al. Reduction of maternal-infant transmission of human immunodeficiency virus type 1 with zidovudine treatment. Pediatric AIDS Clinical Trials Group Protocol 076 Study Group. N Engl J Med 1994; 331:1173-80.
transparent image
100.   Shaffer N, Chuachoowong R, Mock PA, Bhadrakom C, Siriwasin W, Young NL, Chotpitayasunondh T, Chearskul S, Roongpisuthipong A, Chinayon P, Karon J, Mastro TD, Simonds RJ. Short-course zidovudine for perinatal HIV-1 transmission in Bangkok, Thailand: a randomised controlled trial. Bangkok Collaborative Perinatal HIV Transmission Study Group. Lancet 1999; 353:773-80.
transparent image
101.   Guay LA, Musoke P, Fleming T, Bagenda D, Allen M, Nakabiito C, Sherman J, Bakaki P, Ducar C, Deseyve M, Emel L, Mirochnick M, Fowler MG, Mofenson L, Miotti P, Dransfield K, Bray D, Mmiro F, Jackson JB. Intrapartum and neonatal single-dose nevirapine compared with zidovudine for prevention of mother-to-child transmission of HIV-1 in Kampala, Uganda: HIVNET 012 randomised trial. Lancet 1999; 354:795-802.
transparent image
102.   Lallemant M, Jourdain G, Le Coeur S, Kim S, Koetsawang S, Comeau AM, Phoolcharoen W, Essex M, McIntosh K, Vithayasai V. A trial of shortened zidovudine regimens to prevent mother-to-child transmission of human immunodeficiency virus type 1. Perinatal HIV Prevention Trial (Thailand) Investigators. N Engl J Med 2000; 343:982-91.
transparent image
103.  Chalermchokcharoenkit A, Asavapiriyanont S, Teeraratkul A. Combination Short-course Zidovudine plus 2-Dose Nevirapine for Prevention of Mother-to-Child Transmission: Safety, Tolerance, Transmission, and Resistance Results. In: Program and abstracts of the 11th Conference on Retroviruses and Opportunistic Infections; February 8-11, 2004; San Francisco. Abstract 96.
transparent image
104.   Richardson BA, John-Stewart GC, Hughes JP, Nduati R, Mbori-Ngacha D, Overbaugh J, Kreiss JK. Breast-milk infectivity in human immunodeficiency virus type 1-infected mothers. J Infect Dis 2003; 187:736-40.
transparent image
105.   Centers for Disease Control. 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.
transparent image
106.   Proposed 'World Health Organization staging system for HIV infection and disease': preliminary testing by an international collaborative cross-sectional study. The WHO International Collaborating Group for the Study of the WHO Staging System. Aids 1993; 7:711-8.
transparent image
107.   Schacker T, Collier AC, Hughes J, Shea T, Corey L. Clinical and epidemiologic features of primary HIV infection. Ann Intern Med 1996; 125:257-64.
transparent image
108.   Cooper DA, Gold J, Maclean P, Donovan B, Finlayson R, Barnes TG, Michelmore HM, Brooke P, Penny R. Acute AIDS retrovirus infection. Definition of a clinical illness associated with seroconversion. Lancet 1985; 1:537-40.
transparent image
109.   Kahn JO, Walker BD. Acute human immunodeficiency virus type 1 infection. N Engl J Med 1998; 339:33-9.
transparent image
110.   Vanhems P, Dassa C, Lambert J, Cooper DA, Perrin L, Vizzard J, Hirschel B, Kinloch-de Loes S, Carr A, Allard R. Comprehensive classification of symptoms and signs reported among 218 patients with acute HIV-1 infection. J Acquir Immune Defic Syndr 1999; 21:99-106.
transparent image
111.   Lavreys L, Thompson ML, Martin HL, Jr., Mandaliya K, Ndinya-Achola JO, Bwayo JJ, Kreiss J. Primary human immunodeficiency virus type 1 infection: clinical manifestations among women in Mombasa, Kenya. Clin Infect Dis 2000; 30:486-90.
transparent image
112.   Bollinger RC, Brookmeyer RS, Mehendale SM, Paranjape RS, Shepherd ME, Gadkari DA, Quinn TC. Risk factors and clinical presentation of acute primary HIV infection in India. Jama 1997; 278:2085-9.
transparent image
113.   Pedersen C, Lindhardt BO, Jensen BL, Lauritzen E, Gerstoft J, Dickmeiss E, Gaub J, Scheibel E, Karlsmark T. Clinical course of primary HIV infection: consequences for subsequent course of infection. Bmj 1989; 299:154-7.
transparent image
114.   Busch MP, Lee LL, Satten GA, Henrard DR, Farzadegan H, Nelson KE, Read S, Dodd RY, Petersen LR. Time course of detection of viral and serologic markers preceding human immunodeficiency virus type 1 seroconversion: implications for screening of blood and tissue donors. Transfusion 1995; 35:91-7.
transparent image
115.   Piatak M, Jr., Saag MS, Yang LC, Clark SJ, Kappes JC, Luk KC, Hahn BH, Shaw GM, Lifson JD. High levels of HIV-1 in plasma during all stages of infection determined by competitive PCR. Science 1993; 259:1749-54.
transparent image
116.   Daar ES, Moudgil T, Meyer RD, Ho DD. Transient high levels of viremia in patients with primary human immunodeficiency virus type 1 infection. N Engl J Med 1991; 324:961-4.
transparent image
117.   Hecht FM, Busch MP, Rawal B, Webb M, Rosenberg E, Swanson M, Chesney M, Anderson J, Levy J, Kahn JO. Use of laboratory tests and clinical symptoms for identification of primary HIV infection. Aids 2002; 16:1119-29.
transparent image
118.   Roos MT, de Leeuw NA, Claessen FA, Huisman HG, Kootstra NA, Meyaard L, Schellekens PT, Schuitemaker H, Miedema F. Viro-immunological studies in acute HIV-1 infection. Aids 1994; 8:1533-8.
transparent image
119.   Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, Farthing C, Ho DD. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol 1994; 68:4650-5.
transparent image
120.   Borrow P, Lewicki H, Hahn BH, Shaw GM, Oldstone MB. Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J Virol 1994; 68:6103-10.
transparent image
121.   Musey L, Hughes J, Schacker T, Shea T, Corey L, McElrath MJ. Cytotoxic-T-cell responses, viral load, and disease progression in early human immunodeficiency virus type 1 infection. N Engl J Med 1997; 337:1267-74.
transparent image
122.   Pantaleo G, Demarest JF, Schacker T, Vaccarezza M, Cohen OJ, Daucher M, Graziosi C, Schnittman SS, Quinn TC, Shaw GM, Perrin L, Tambussi G, Lazzarin A, Sekaly RP, Soudeyns H, Corey L, Fauci AS. The qualitative nature of the primary immune response to HIV infection is a prognosticator of disease progression independent of the initial level of plasma viremia. Proc Natl Acad Sci U S A 1997; 94:254-8.
transparent image
123.   Vento S, Di Perri G, Garofano T, Concia E, Bassetti D. Pneumocystis carinii pneumonia during primary HIV-1 infection. Lancet 1993; 342:24-5.
transparent image
124.   Gupta KK. Acute immunosuppression with HIV seroconversion. N Engl J Med 1993; 328:288-9.
transparent image
125.   Pedersen C, Dickmeiss E, Gaub J, Ryder LP, Platz P, Lindhardt BO, Lundgren JD. T-cell subset alterations and lymphocyte responsiveness to mitogens and antigen during severe primary infection with HIV: a case series of seven consecutive HIV seroconverters. Aids 1990; 4:523-6.
transparent image
126.   Rosenberg ES, Billingsley JM, Caliendo AM, Boswell SL, Sax PE, Kalams SA, Walker BD. Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia. Science 1997; 278:1447-50.
transparent image
127.   Mellors JW, Kingsley LA, Rinaldo CR, Jr., Todd JA, Hoo BS, Kokka RP, Gupta P. Quantitation of HIV-1 RNA in plasma predicts outcome after seroconversion. Ann Intern Med 1995; 122:573-9.
transparent image
128.   Mellors JW, Rinaldo CR, Jr., Gupta P, White RM, Todd JA, Kingsley LA. Prognosis in HIV-1 infection predicted by the quantity of virus in plasma. Science 1996; 272:1167-70.
transparent image
129.   Rosenberg ES, Altfeld M, Poon SH, Phillips MN, Wilkes BM, Eldridge RL, Robbins GK, D'Aquila RT, Goulder PJ, Walker BD. Immune control of HIV-1 after early treatment of acute infection. Nature 2000; 407:523-6.
transparent image
130.   Oxenius A, Price DA, Easterbrook PJ, O'Callaghan CA, Kelleher AD, Whelan JA, Sontag G, Sewell AK, Phillips RE. Early highly active antiretroviral therapy for acute HIV-1 infection preserves immune function of CD8+ and CD4+ T lymphocytes. Proc Natl Acad Sci U S A 2000; 97:3382-7.
transparent image
131.   Stranford SA, Ong JC, Martinez-Marino B, Busch M, Hecht FM, Kahn J, Levy JA. Reduction in CD8+ cell noncytotoxic anti-HIV activity in individuals receiving highly active antiretroviral therapy during primary infection. Proc Natl Acad Sci U S A 2001; 98:597-602.
transparent image
132.   Tremblay CL, Hicks JL, Sutton L, Giguel F, Flynn T, Johnston M, Sax PE, Walker BD, Hirsch MS, Rosenberg ES, D'Aquila RT. Antiretroviral resistance associated with supervised treatment interruptions in treated acute HIV infection. Aids 2003; 17:1086-9.
transparent image
133.   Bacchetti P, Moss AR. Incubation period of AIDS in San Francisco. Nature 1989; 338:251-3.
transparent image
134.   Moss AR, Bacchetti P, Osmond D, Krampf W, Chaisson RE, Stites D, Wilber J, Allain JP, Carlson J. Seropositivity for HIV and the development of AIDS or AIDS related condition: three year follow up of the San Francisco General Hospital cohort. Br Med J (Clin Res Ed) 1988; 296:745-50.
transparent image
135.   Lefrere JJ, Morand-Joubert L, Mariotti M, Bludau H, Burghoffer B, Petit JC, Roudot-Thoraval F. Even individuals considered as long-term nonprogressors show biological signs of progression after 10 years of human immunodeficiency virus infection. Blood 1997; 90:1133-40.
transparent image
136.   Balotta C, Bagnarelli P, Riva C, Valenza A, Antinori S, Colombo MC, Sampaolesi R, Violin M, de Pasquale MP, Moroni M, Clementi M, Galli M. Comparable biological and molecular determinants in HIV type 1-infected long-term nonprogressors and recently infected individuals. AIDS Res Hum Retroviruses 1997; 13:337-41.
transparent image
137.   Koblin BA, van Benthem BH, Buchbinder SP, Ren L, Vittinghoff E, Stevens CE, Coutinho RA, van Griensven GJ. Long-term survival after infection with human immunodeficiency virus type 1 (HIV-1) among homosexual men in hepatitis B vaccine trial cohorts in Amsterdam, New York City, and San Francisco, 1978-1995. Am J Epidemiol 1999; 150:1026-30.
transparent image
138.   Phillips AN. CD4 lymphocyte depletion prior to the development of AIDS. Aids 1992; 6:735-6.
transparent image
139.   van den Berg H, Gerritsen EJ, van Tol MJ, Dooren LJ, Vossen JM. Ten years after acquiring an HIV-1 infection: a study in a cohort of eleven neonates infected by aliquots from a single plasma donation. Acta Paediatr 1994; 83:173-8.
transparent image
140.  Buchbinder S VE, Kaslow R. HIV disease progression and associated genetic markers. In: Program and Abstracts of the 5th Conference on Retroviruses and Opportunistic Infections, February 1-5, 1998, Chicago.
transparent image
141.   O'Brien TR, Blattner WA, Waters D, Eyster E, Hilgartner MW, Cohen AR, Luban N, Hatzakis A, Aledort LM, Rosenberg PS, Miley WJ, Kroner BL, Goedert JJ. Serum HIV-1 RNA levels and time to development of AIDS in the Multicenter Hemophilia Cohort Study. Jama 1996; 276:105-10.
transparent image
142.   Mellors JW, Kingsley LA, Rinaldo CR, Jr., Todd JA, Hoo BS, Kokka RP, Gupta P. Quantitation of HIV-1 RNA in plasma predicts outcome after seroconversion. Ann Intern Med 1995; 122:573-9.
transparent image
143.   Mellors JW, Munoz A, Giorgi JV, Margolick JB, Tassoni CJ, Gupta P, Kingsley LA, Todd JA, Saah AJ, Detels R, Phair JP, Rinaldo CR, Jr. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med 1997; 126:946-54.
transparent image
144.   Iuliano R, Forastieri G, Brizzi M, Mecocci L, Mazzotta F, Ceccherini-Nelli L. Correlation between plasma HIV-1 RNA levels and the rate of immunologic decline. J Acquir Immune Defic Syndr Hum Retrovirol 1997; 14:408-14.
transparent image
145.   Osmond D, Charlebois E, Lang W, Shiboski S, Moss A. Changes in AIDS survival time in two San Francisco cohorts of homosexual men, 1983 to 1993. Jama 1994; 271:1083-7.
transparent image
146.   Longini IM, Jr., Clark WS, Gardner LI, Brundage JF. The dynamics of CD4+ T-lymphocyte decline in HIV-infected individuals: a Markov modeling approach. J Acquir Immune Defic Syndr 1991; 4:1141-7.
transparent image
147.   Gail MH, Tan WY, Pee D, Goedert JJ. Survival after AIDS diagnosis in a cohort of hemophilia patients. Multicenter Hemophilia Cohort Study. J Acquir Immune Defic Syndr Hum Retrovirol 1997; 15:363-9.
transparent image
148.   Bacchetti P, Osmond D, Chaisson RE, Dritz S, Rutherford GW, Swig L, Moss AR. Survival patterns of the first 500 patients with AIDS in San Francisco. J Infect Dis 1988; 157:1044-7.
transparent image
149.   Liu R, Paxton WA, Choe S, Ceradini D, Martin SR, Horuk R, MacDonald ME, Stuhlmann H, Koup RA, Landau NR. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 1996; 86:367-77.
transparent image
150.   Rowland-Jones SL, Dong T, Fowke KR, Kimani J, Krausa P, Newell H, Blanchard T, Ariyoshi K, Oyugi J, Ngugi E, Bwayo J, MacDonald KS, McMichael AJ, Plummer FA. Cytotoxic T cell responses to multiple conserved HIV epitopes in HIV-resistant prostitutes in Nairobi. J Clin Invest 1998; 102:1758-65.
transparent image
151.   Kaul R, Trabattoni D, Bwayo JJ, Arienti D, Zagliani A, Mwangi FM, Kariuki C, Ngugi EN, MacDonald KS, Ball TB, Clerici M, Plummer FA. HIV-1-specific mucosal IgA in a cohort of HIV-1-resistant Kenyan sex workers. Aids 1999; 13:23-9.
transparent image
152.   Kaslow RA, Duquesnoy R, VanRaden M, Kingsley L, Marrari M, Friedman H, Su S, Saah AJ, Detels R, Phair J, et al. A1, Cw7, B8, DR3 HLA antigen combination associated with rapid decline of T-helper lymphocytes in HIV-1 infection. A report from the Multicenter AIDS Cohort Study. Lancet 1990; 335:927-30.
transparent image
153.   Kaslow RA, Carrington M, Apple R, Park L, Munoz A, Saah AJ, Goedert JJ, Winkler C, O'Brien SJ, Rinaldo C, Detels R, Blattner W, Phair J, Erlich H, Mann DL. Influence of combinations of human major histocompatibility complex genes on the course of HIV-1 infection. Nat Med 1996; 2:405-11.
transparent image
154.   Keet IP, Tang J, Klein MR, LeBlanc S, Enger C, Rivers C, Apple RJ, Mann D, Goedert JJ, Miedema F, Kaslow RA. Consistent associations of HLA class I and II and transporter gene products with progression of human immunodeficiency virus type 1 infection in homosexual men. J Infect Dis 1999; 180:299-309.
transparent image
155.   Itescu S, Rose S, Dwyer E, Winchester R. Certain HLA-DR5 and -DR6 major histocompatibility complex class II alleles are associated with a CD8 lymphocytic host response to human immunodeficiency virus type 1 characterized by low lymphocyte viral strain heterogeneity and slow disease progression. Proc Natl Acad Sci U S A 1994; 91:11472-6.
transparent image
156.   Kroner BL, Goedert JJ, Blattner WA, Wilson SE, Carrington MN, Mann DL. Concordance of human leukocyte antigen haplotype-sharing, CD4 decline and AIDS in hemophilic siblings. Multicenter Hemophilia Cohort and Hemophilia Growth and Development Studies. Aids 1995; 9:275-80.
transparent image
157.   Migueles SA, Laborico AC, Imamichi H, Shupert WL, Royce C, McLaughlin M, Ehler L, Metcalf J, Liu S, Hallahan CW, Connors M. The differential ability of HLA B*5701+ long-term nonprogressors and progressors to restrict human immunodeficiency virus replication is not caused by loss of recognition of autologous viral gag sequences. J Virol 2003; 77:6889-98.
transparent image
158.   Tomiyama H, Miwa K, Shiga H, Moore YI, Oka S, Iwamoto A, Kaneko Y, Takiguchi M. Evidence of presentation of multiple HIV-1 cytotoxic T lymphocyte epitopes by HLA-B*3501 molecules that are associated with the accelerated progression of AIDS. J Immunol 1997; 158:5026-34.
transparent image
159.   Jeannet M, Sztajzel R, Carpentier N, Hirschel B, Tiercy JM. HLA antigens are risk factors for development of AIDS. J Acquir Immune Defic Syndr 1989; 2:28-32.
transparent image
160.   Itescu S, Mathur-Wagh U, Skovron ML, Brancato LJ, Marmor M, Zeleniuch-Jacquotte A, Winchester R. HLA-B35 is associated with accelerated progression to AIDS. J Acquir Immune Defic Syndr 1992; 5:37-45.
transparent image
161.   Tang J, Costello C, Keet IP, Rivers C, Leblanc S, Karita E, Allen S, Kaslow RA. HLA class I homozygosity accelerates disease progression in human immunodeficiency virus type 1 infection. AIDS Res Hum Retroviruses 1999; 15:317-24.
transparent image
162.   Vittinghoff E, Hessol NA, Bacchetti P, Fusaro RE, Holmberg SD, Buchbinder SP. Cofactors for HIV disease progression in a cohort of homosexual and bisexual men. J Acquir Immune Defic Syndr 2001; 27:308-14.
transparent image
163.   Royce RA, Winkelstein W, Jr. HIV infection, cigarette smoking and CD4+ T-lymphocyte counts: preliminary results from the San Francisco Men's Health Study. Aids 1990; 4:327-33.
transparent image
164.   Moseson M, Zeleniuch-Jacquotte A, Belsito DV, Shore RE, Marmor M, Pasternack B. The potential role of nutritional factors in the induction of immunologic abnormalities in HIV-positive homosexual men. J Acquir Immune Defic Syndr 1989; 2:235-47.
transparent image
165.   Burack JH, Barrett DC, Stall RD, Chesney MA, Ekstrand ML, Coates TJ. Depressive symptoms and CD4 lymphocyte decline among HIV-infected men. Jama 1993; 270:2568-73.
transparent image
166.   Veugelers PJ, Page KA, Tindall B, Schechter MT, Moss AR, Winkelstein WW, Jr., Cooper DA, Craib KJ, Charlebois E, Coutinho RA, et al. Determinants of HIV disease progression among homosexual men registered in the Tricontinental Seroconverter Study. Am J Epidemiol 1994; 140:747-58.
transparent image
167.   Prins M, Veugelers PJ. Comparison of progression and non-progression in injecting drug users and homosexual men with documented dates of HIV-1 seroconversion. European Seroconverter Study and the Tricontinental Seroconverter Study. Aids 1997; 11:621-31.
transparent image
168.   Operskalski EA, Stram DO, Lee H, Zhou Y, Donegan E, Busch MP, Stevens CE, Schiff ER, Dietrich SL, Mosley JW. Human immunodeficiency virus type 1 infection: relationship of risk group and age to rate of progression to AIDS. Transfusion Safety Study Group. J Infect Dis 1995; 172:648-55.
transparent image
169.   Scarlatti G, Tresoldi E, Bjorndal A, Fredriksson R, Colognesi C, Deng HK, Malnati MS, Plebani A, Siccardi AG, Littman DR, Fenyo EM, Lusso P. In vivo evolution of HIV-1 co-receptor usage and sensitivity to chemokine-mediated suppression. Nat Med 1997; 3:1259-65.
transparent image
170.   Tersmette M, de Goede RE, Al BJ, Winkel IN, Gruters RA, Cuypers HT, Huisman HG, Miedema F. Differential syncytium-inducing capacity of human immunodeficiency virus isolates: frequent detection of syncytium-inducing isolates in patients with acquired immunodeficiency syndrome (AIDS) and AIDS-related complex. J Virol 1988; 62:2026-32.
transparent image
171.   Connor RI, Sheridan KE, Ceradini D, Choe S, Landau NR. Change in coreceptor use coreceptor use correlates with disease progression in HIV-1--infected individuals. J Exp Med 1997; 185:621-8.
transparent image
172.   Shankarappa R, Margolick JB, Gange SJ, Rodrigo AG, Upchurch D, Farzadegan H, Gupta P, Rinaldo CR, Learn GH, He X, Huang XL, Mullins JI. Consistent viral evolutionary changes associated with the progression of human immunodeficiency virus type 1 infection. J Virol 1999; 73:10489-502.
transparent image
173.   Croteau G, Doyon L, Thibeault D, McKercher G, Pilote L, Lamarre D. Impaired fitness of human immunodeficiency virus type 1 variants with high-level resistance to protease inhibitors. J Virol 1997; 71:1089-96.
transparent image
174.   Ho DD, Toyoshima T, Mo H, Kempf DJ, Norbeck D, Chen CM, Wideburg NE, Burt SK, Erickson JW, Singh MK. Characterization of human immunodeficiency virus type 1 variants with increased resistance to a C2-symmetric protease inhibitor. J Virol 1994; 68:2016-20.
transparent image
175.   Sharma PL, Crumpacker CS. Attenuated replication of human immunodeficiency virus type 1 with a didanosine-selected reverse transcriptase mutation. J Virol 1997; 71:8846-51.
transparent image
176.   Harrigan PR, Bloor S, Larder BA. Relative replicative fitness of zidovudine-resistant human immunodeficiency virus type 1 isolates in vitro. J Virol 1998; 72:3773-8.
transparent image
177.   Caliendo AM, Savara A, An D, DeVore K, Kaplan JC, D'Aquila RT. Effects of zidovudine-selected human immunodeficiency virus type 1 reverse transcriptase amino acid substitutions on processive DNA synthesis and viral replication. J Virol 1996; 70:2146-53.
transparent image
178.   Condra JH, Schleif WA, Blahy OM, Gabryelski LJ, Graham DJ, Quintero JC, Rhodes A, Robbins HL, Roth E, Shivaprakash M, et al. In vivo emergence of HIV-1 variants resistant to multiple protease inhibitors. Nature 1995; 374:569-71.
transparent image
179.  Mellors J, Larder B, Schinazi R. Mutations in HIV-1 reverse transcriptase and protease associated with drug resistance. Int. Antiviral News 1995:3:8-13.
transparent image
180.   Roberts NA. Drug-resistance patterns of saquinavir and other HIV proteinase inhibitors. Aids 1995; 9 Suppl 2:27-S32.
transparent image
181.   Larder BA, Kemp SD, Harrigan PR. Potential mechanism for sustained antiretroviral efficacy of AZT-3TC combination therapy. Science 1995; 269:696-9.
transparent image
182.   Molla A, Korneyeva M, Gao Q, Vasavanonda S, Schipper PJ, Mo HM, Markowitz M, Chernyavskiy T, Niu P, Lyons N, Hsu A, Granneman GR, Ho DD, Boucher CA, Leonard JM, Norbeck DW, Kempf DJ. Ordered accumulation of mutations in HIV protease confers resistance to ritonavir. Nat Med 1996; 2:760-6.
transparent image
183.   Borman AM, Paulous S, Clavel F. Resistance of human immunodeficiency virus type 1 to protease inhibitors: selection of resistance mutations in the presence and absence of the drug. J Gen Virol 1996; 77 ( Pt 3):419-26.
transparent image
184.   Kaleebu P, French N, Mahe C, Yirrell D, Watera C, Lyagoba F, Nakiyingi J, Rutebemberwa A, Morgan D, Weber J, Gilks C, Whitworth J. Effect of human immunodeficiency virus (HIV) type 1 envelope subtypes A and D on disease progression in a large cohort of HIV-1-positive persons in Uganda. J Infect Dis 2002; 185:1244-50.
transparent image
185.   Kirchhoff F, Greenough TC, Brettler DB, Sullivan JL, Desrosiers RC. Brief report: absence of intact nef sequences in a long-term survivor with nonprogressive HIV-1 infection. N Engl J Med 1995; 332:228-32.
transparent image
186.   Saravolatz L, Neaton JD, Sacks L, Deyton L, Rhame F, Sherer R. CD4+ T lymphocyte counts and patterns of mortality among patients infected with human immunodeficiency virus who were enrolled in community programs for clinical research on AIDS. Clin Infect Dis 1996; 22:513-20.
transparent image
187.   Sulkowski MS, Mast EE, Seeff LB, Thomas DL. Hepatitis C virus infection as an opportunistic disease in persons infected with human immunodeficiency virus. Clin Infect Dis 2000; 30 Suppl 1:S77-84.
transparent image
188.   Greub G, Ledergerber B, Battegay M, Grob P, Perrin L, Furrer H, Burgisser P, Erb P, Boggian K, Piffaretti JC, Hirschel B, Janin P, Francioli P, Flepp M, Telenti A. Clinical progression, survival, and immune recovery during antiretroviral therapy in patients with HIV-1 and hepatitis C virus coinfection: the Swiss HIV Cohort Study. Lancet 2000; 356:1800-5.
transparent image
189.   Sulkowski MS, Moore RD, Mehta SH, Chaisson RE, Thomas DL. Hepatitis C and progression of HIV disease. Jama 2002; 288:199-206.
transparent image
190.   Kalams SA, Buchbinder SP, Rosenberg ES, Billingsley JM, Colbert DS, Jones NG, Shea AK, Trocha AK, Walker BD. Association between virus-specific cytotoxic T-lymphocyte and helper responses in human immunodeficiency virus type 1 infection. J Virol 1999; 73:6715-20.
transparent image
191.   Lisziewicz J, Rosenberg E, Lieberman J, Jessen H, Lopalco L, Siliciano R, Walker B, Lori F. Control of HIV despite the discontinuation of antiretroviral therapy. N Engl J Med 1999; 340:1683-4.
transparent image
192.   Update: serologic testing for antibody to human immunodeficiency virus. MMWR Morb Mortal Wkly Rep 1988; 36:833-40, 845.
transparent image
193.   Schwartz JS, Dans PE, Kinosian BP. Human immunodeficiency virus test evaluation, performance, and use. Proposals to make good tests better. Jama 1988; 259:2574-9.
transparent image
194.   Emmons WW, Paparello SF, Decker CF, Sheffield JM, Lowe-Bey FH. A modified ELISA and western blot accurately determine anti-human immunodeficiency virus type 1 antibodies in oral fluids obtained with a special collecting device. J Infect Dis 1995; 171:1406-10.
transparent image
195.   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.
transparent image
196.   Tiensiwakul P. Urinary HIV-1 antibody patterns by western blot assay. Clin Lab Sci 1998; 11:336-8.
transparent image
197.   Colfax GN, Lehman JS, Bindman AB, Vittinghoff E, Vranizan K, Fleming PL, Chesney M, Osmond D, Hecht FM. What happened to home HIV test collection kits? Intent to use kits, actual use, and barriers to use among persons at risk for HIV infection. AIDS Care 2002; 14:675-82.
transparent image
198.   Ketema F, Zeh C, Edelman DC, Saville R, Constantine NT. Assessment of the performance of a rapid, lateral flow assay for the detection of antibodies to HIV. J Acquir Immune Defic Syndr 2001; 27:63-70.
transparent image
199.   Phili R, Vardas E. Evaluation of a rapid human immunodeficiency virus test at two community clinics in Kwazulu-Natal. S Afr Med J 2002; 92:818-21.
transparent image
200.   Ramalingam S, Kannangai R, Raj AA, Jesudason MV, Sridharan G. Rapid particle agglutination test for human immunodeficiency virus: hospital-based evaluation. J Clin Microbiol 2002; 40:1553-4.
transparent image
201.   Respess RA, Rayfield MA, Dondero TJ. Laboratory testing and rapid HIV assays: applications for HIV surveillance in hard-to-reach populations. Aids 2001; 15 Suppl 3:S49-59.
transparent image
202.   Soroka SD, Granade TC, Phillips S, Parekh B. The use of simple, rapid tests to detect antibodies to human immunodeficiency virus types 1 and 2 in pooled serum specimens. J Clin Virol 2003; 27:90-6.
transparent image
203.   Janssen RS, Satten GA, Stramer SL, Rawal BD, O'Brien TR, Weiblen BJ, Hecht FM, Jack N, Cleghorn FR, Kahn JO, Chesney MA, Busch MP. New testing strategy to detect early HIV-1 infection for use in incidence estimates and for clinical and prevention purposes. Jama 1998; 280:42-8.
transparent image
204.   McFarland W, Busch MP, Kellogg TA, Rawal BD, Satten GA, Katz MH, Dilley J, Janssen RS. Detection of early HIV infection and estimation of incidence using a sensitive/less-sensitive enzyme immunoassay testing strategy at anonymous counseling and testing sites in San Francisco. J Acquir Immune Defic Syndr 1999; 22:484-9.
transparent image
205.   Crowe SM, Carlin JB, Stewart KI, Lucas CR, Hoy JF. Predictive value of CD4 lymphocyte numbers for the development of opportunistic infections and malignancies in HIV-infected persons. J Acquir Immune Defic Syndr 1991; 4:770-6.
transparent image
206.   Mellors JW, Munoz A, Giorgi JV, Margolick JB, Tassoni CJ, Gupta P, Kingsley LA, Todd JA, Saah AJ, Detels R, Phair JP, Rinaldo CR, Jr. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med 1997; 126:946-54.
transparent image
207.   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.
transparent image
208.   Dybul M, Fauci AS, Bartlett JG, Kaplan JE, Pau AK. Guidelines for using antiretroviral agents among HIV-infected adults and adolescents. Recommendations of the Panel on Clinical Practices for Treatment of HIV. MMWR Recomm Rep 2002; 51:1-55.
transparent image
209.   Recommendations for prophylaxis against Pneumocystis carinii pneumonia for adults and adolescents infected with human immunodeficiency virus. MMWR Recomm Rep 1992; 41:1-11.
transparent image
210.  Results of the 1996 T-lymphocyte immunophenotyping questionnaire survey mailed to laboratories participating in the Model Performance Evaluation Program. Atlanta, GA: US Department of Health and Human Services, Public Health Service, CDC, 1996:16.
transparent image
211.   Mandy FF, Nicholson JK, McDougal JS. Guidelines for performing single-platform absolute CD4+ T-cell determinations with CD45 gating for persons infected with human immunodeficiency virus. Centers for Disease Control and Prevention. MMWR Recomm Rep 2003; 52:1-13.
transparent image
212.   Mandy F, Nicholson J, Autran B, Janossy G. T-cell subset counting and the fight against AIDS: reflections over a 20-year struggle. Cytometry 2002; 50:39-45.
transparent image
213.   Schechter M, Zajdenverg R, Machado LL, Pinto ME, Lima LA, Perez MA. Predicting CD4 counts in HIV-infected Brazilian individuals: a model based on the World Health Organization staging system. J Acquir Immune Defic Syndr 1994; 7:163-8.
transparent image
214.   Badri M, Ehrlich R, Wood R, Maartens G. Initiating co-trimoxazole prophylaxis in HIV-infected patients in Africa: an evaluation of the provisional WHO/UNAIDS recommendations. Aids 2001; 15:1143-8.
transparent image
215.   Kumarasamy N, Mahajan AP, Flanigan TP, Hemalatha R, Mayer KH, Carpenter CC, Thyagarajan SP, Solomon S. Total lymphocyte count (TLC) is a useful tool for the timing of opportunistic infection prophylaxis in India and other resource-constrained countries. J Acquir Immune Defic Syndr 2002; 31:378-83.
transparent image
216.  Flanigan TP, Mahajan AP, Kumarasamy N. Total lymphocyte count (TLC) as a surrogate for CD4 count to initiate and monitor HAART in resource-limited countries (Abstract G160e).
transparent image
217.  Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents (November 10, 2003). http://aidsinfo.nih.gov/guidelines (accessed March 2, 2004).
transparent image
218.   Anastassopoulou CG, Touloumi G, Katsoulidou A, Hatzitheodorou H, Pappa M, Paraskevis D, Lazanas M, Gargalianos P, Hatzakis A. Comparative evaluation of the QUANTIPLEX HIV-1 RNA 2.0 and 3.0 (bDNA) assays and the AMPLICOR HIV-1 MONITOR v1.5 test for the quantitation of human immunodeficiency virus type 1 RNA in plasma. J Virol Methods 2001; 91:67-74.
transparent image
219.   Elbeik T, Alvord WG, Trichavaroj R, de Souza M, Dewar R, Brown A, Chernoff D, Michael NL, Nassos P, Hadley K, Ng VL. Comparative analysis of HIV-1 viral load assays on subtype quantification: Bayer Versant HIV-1 RNA 3.0 versus Roche Amplicor HIV-1 Monitor version 1.5. J Acquir Immune Defic Syndr 2002; 29:330-9.
transparent image
220.   Deeks SG, Coleman RL, White R, Pachl C, Schambelan M, Chernoff DN, Feinberg MB. Variance of plasma human immunodeficiency virus type 1 RNA levels measured by branched DNA within and between days. J Infect Dis 1997; 176:514-7.
transparent image
221.   Sulkowski MS, Chaisson RE, Karp CL, Moore RD, Margolick JB, Quinn TC. The effect of acute infectious illnesses on plasma human immunodeficiency virus (HIV) type 1 load and the expression of serologic markers of immune activation among HIV-infected adults. J Infect Dis 1998; 178:1642-8.
transparent image
222.   Stanley SK, Ostrowski MA, Justement JS, Gantt K, Hedayati S, Mannix M, Roche K, Schwartzentruber DJ, Fox CH, Fauci AS. Effect of immunization with a common recall antigen on viral expression in patients infected with human immunodeficiency virus type 1. N Engl J Med 1996; 334:1222-30.
transparent image
223.   Staprans SI, Hamilton BL, Follansbee SE, Elbeik T, Barbosa P, Grant RM, Feinberg MB. Activation of virus replication after vaccination of HIV-1-infected individuals. J Exp Med 1995; 182:1727-37.
transparent image
224.   Snyder EL, Dodd RY. Reducing the risk of blood transfusion. Hematology (Am Soc Hematol Educ Program) 2001:433-42.
transparent image
225.   Schupbach J, Boni J, Flepp M, Tomasik Z, Joller H, Opravil M. Antiretroviral treatment monitoring with an improved HIV-1 p24 antigen test: an inexpensive alternative to tests for viral RNA. J Med Virol 2001; 65:225-32.
transparent image
226.   Durant J, Clevenbergh P, Halfon P, Delgiudice P, Porsin S, Simonet P, Montagne N, Boucher CA, Schapiro JM, Dellamonica P. Drug-resistance genotyping in HIV-1 therapy: the VIRADAPT randomised controlled trial. Lancet 1999; 353:2195-9.
transparent image
227.   Baxter JD, Mayers DL, Wentworth DN, Neaton JD, Hoover ML, Winters MA, Mannheimer SB, Thompson MA, Abrams DI, Brizz BJ, Ioannidis JP, Merigan TC. A randomized study of antiretroviral management based on plasma genotypic antiretroviral resistance testing in patients failing therapy. CPCRA 046 Study Team for the Terry Beirn Community Programs for Clinical Research on AIDS. Aids 2000; 14:F83-93.
transparent image
228.   Tural C, Ruiz L, Holtzer C, Schapiro J, Viciana P, Gonzalez J, Domingo P, Boucher C, Rey-Joly C, Clotet B. Clinical utility of HIV-1 genotyping and expert advice: the Havana trial. Aids 2002; 16:209-18.
transparent image
229.   Cohen CJ, Hunt S, Sension M, Farthing C, Conant M, Jacobson S, Nadler J, Verbiest W, Hertogs K, Ames M, Rinehart AR, Graham NM. A randomized trial assessing the impact of phenotypic resistance testing on antiretroviral therapy. Aids 2002; 16:579-88.
transparent image
230.   Little SJ, Holte S, Routy JP, Daar ES, Markowitz M, Collier AC, Koup RA, Mellors JW, Connick E, Conway B, Kilby M, Wang L, Whitcomb JM, Hellmann NS, Richman DD. Antiretroviral-drug resistance among patients recently infected with HIV. N Engl J Med 2002; 347:385-94.
transparent image
231.   Larder BA, Kohli A, Kellam P, Kemp SD, Kronick M, Henfrey RD. Quantitative detection of HIV-1 drug resistance mutations by automated DNA sequencing. Nature 1993; 365:671-3.
transparent image
232.   Shafer RW, Warford A, Winters MA, Gonzales MJ. Reproducibility of human immunodeficiency virus type 1 (HIV-1) protease and reverse transcriptase sequencing of plasma samples from heavily treated HIV-1-infected individuals. J Virol Methods 2000; 86:143-53.
transparent image
233.   Kijak GH, Rubio AE, Pampuro SE, Zala C, Cahn P, Galli R, Montaner JS, Salomon H. Discrepant results in the interpretation of HIV-1 drug-resistance genotypic data among widely used algorithms. HIV Med 2003; 4:72-8.
transparent image
234.   Hirsch MS, Brun-Vezinet F, Clotet B, Conway B, Kuritzkes DR, D'Aquila RT, Demeter LM, Hammer SM, Johnson VA, Loveday C, Mellors JW, Jacobsen DM, Richman DD. Antiretroviral drug resistance testing in adults infected with human immunodeficiency virus type 1: 2003 recommendations of an International AIDS Society-USA Panel. Clin Infect Dis 2003; 37:113-28.
transparent image
235.   Nunez M, Gonzalez-Requena D, Gonzalez-Lahoz J, Soriano V. Short communication: interactions between nevirapine plasma levels, chronic hepatitis C, and the development of liver toxicity in HIV-infected patients. AIDS Res Hum Retroviruses 2003; 19:187-8.
transparent image
236.   Mallon PW, Ray J, Cooper DA. Effect of therapeutic drug monitoring on outcome in antiretroviral experienced HIV-infected individuals. J Clin Virol 2003; 26:223-7.
transparent image
237.   Dasgupta A, Okhuysen PC. Pharmacokinetic and other drug interactions in patients with AIDS. Ther Drug Monit 2001; 23:591-605.
transparent image
238.   Molla A, Mo H, Vasavanonda S, Han L, Lin CT, Hsu A, Kempf DJ. In vitro antiviral interaction of lopinavir with other protease inhibitors. Antimicrob Agents Chemother 2002; 46:2249-53.
transparent image
239.   Hugen PW, Burger DM, Aarnoutse RE, Baede PA, Nieuwkerk PT, Koopmans PP, Hekster YA. Therapeutic drug monitoring of HIV-protease inhibitors to assess noncompliance. Ther Drug Monit 2002; 24:579-87.
transparent image
240.   Acosta EP, Gerber JG. Position paper on therapeutic drug monitoring of antiretroviral agents. AIDS Res Hum Retroviruses 2002; 18:825-34.
transparent image
241.   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.
transparent image
242.   Kalichman SC, Cage M, Barnett T, Tharnish P, Rompa D, Austin J, Luke W, O'Mowrey J, Schinazi RF. Human immunodeficiency virus in semen and plasma: investigation of sexual transmission risk behavioral correlates. AIDS Res Hum Retroviruses 2001; 17:1695-703.
transparent image
243.   Dunne AL, Mitchell F, Allen KM, Baker HW, Garland S, Clarke GN, Mijch A, Crowe SM. Analysis of HIV-1 viral load in seminal plasma samples. J Clin Virol 2003; 26:239-45.
transparent image
244.   Anton PA, Mitsuyasu RT, Deeks SG, Scadden DT, Wagner B, Huang C, Macken C, Richman DD, Christopherson C, Borellini F, Lazar R, Hege KM. Multiple measures of HIV burden in blood and tissue are correlated with each other but not with clinical parameters in aviremic subjects. Aids 2003; 17:53-63.
transparent image
245.   Garrigue I, Pellegrin I, Hoen B, Dumon B, Harzic M, Schrive MH, Sereni D, Fleury H. Cell-associated HIV-1-DNA quantitation after highly active antiretroviral therapy-treated primary infection in patients with persistently undetectable plasma HIV-1 RNA. Aids 2000; 14:2851-5.
transparent image
246.   Brandt CD, Rakusan TA, Sison AV, Saxena ES, Ellaurie M, Sever JL. Human immunodeficiency virus infection in infants during the first 2 months of life. Reliable detection and evidence of in utero transmission. Arch Pediatr Adolesc Med 1994; 148:250-4.
transparent image
247.   Hsiao AF, Wong MD, Kanouse DE, Collins RL, Liu H, Andersen RM, Gifford AL, McCutchan A, Bozzette SA, Shapiro MF, Wenger NS. Complementary and alternative medicine use and substitution for conventional therapy by HIV-infected patients. J Acquir Immune Defic Syndr 2003; 33:157-65.
transparent image
248.   Mole L, Ripich S, Margolis D, Holodniy M. The impact of active herpes simplex virus infection on human immunodeficiency virus load. J Infect Dis 1997; 176:766-70.
transparent image
249.   Sulkowski MS, Chaisson RE, Karp CL, Moore RD, Margolick JB, Quinn TC. The effect of acute infectious illnesses on plasma human immunodeficiency virus (HIV) type 1 load and the expression of serologic markers of immune activation among HIV-infected adults. J Infect Dis 1998; 178:1642-8.
transparent image
250.   Donovan RM, Bush CE, Markowitz NP, Baxa DM, Saravolatz LD. Changes in virus load markers during AIDS-associated opportunistic diseases in human immunodeficiency virus-infected persons. J Infect Dis 1996; 174:401-3.
transparent image
251.   Bush CE, Donovan RM, Markowitz NP, Kvale P, Saravolatz LD. A study of HIV RNA viral load in AIDS patients with bacterial pneumonia. J Acquir Immune Defic Syndr Hum Retrovirol 1996; 13:23-6.
transparent image
252.   Ramilo O, Hicks PJ, Borvak J, Gross LM, Zhong D, Squires JE, Vitetta ES. T cell activation and human immunodeficiency virus replication after influenza immunization of infected children. Pediatr Infect Dis J 1996; 15:197-203.
transparent image
253.   Staprans SI, Hamilton BL, Follansbee SE, Elbeik T, Barbosa P, Grant RM, Feinberg MB. Activation of virus replication after vaccination of HIV-1-infected individuals. J Exp Med 1995; 182:1727-37.
transparent image
254.   Ho DD. HIV-1 viraemia and influenza. Lancet 1992; 339:1549.
transparent image
255.   Brichacek B, Swindells S, Janoff EN, Pirruccello S, Stevenson M. Increased plasma human immunodeficiency virus type 1 burden following antigenic challenge with pneumococcal vaccine. J Infect Dis 1996; 174:1191-9.
transparent image
256.   Wahl LM, Nowak MA. Adherence and drug resistance: predictions for therapy outcome. Proc R Soc Lond B Biol Sci 2000; 267:835-43.
transparent image
257.   Paterson DL, Swindells S, Mohr J, Brester M, Vergis EN, Squier C, Wagener MM, Singh N. Adherence to protease inhibitor therapy and outcomes in patients with HIV infection. Ann Intern Med 2000; 133:21-30.
transparent image
258.   Perelson AS, Essunger P, Cao Y, Vesanen M, Hurley A, Saksela K, Markowitz M, Ho DD. Decay characteristics of HIV-1-infected compartments during combination therapy. Nature 1997; 387:188-91.
transparent image
259.   Wu H, Kuritzkes DR, McClernon DR, Kessler H, Connick E, Landay A, Spear G, Heath-Chiozzi M, Rousseau F, Fox L, Spritzler J, Leonard JM, Lederman MM. Characterization of viral dynamics in human immunodeficiency virus type 1-infected patients treated with combination antiretroviral therapy: relationships to host factors, cellular restoration, and virologic end points. J Infect Dis 1999; 179:799-807.
transparent image
260.   Powderly WG, Saag MS, Chapman S, Yu G, Quart B, Clendeninn NJ. Predictors of optimal virological response to potent antiretroviral therapy. Aids 1999; 13:1873-80.
transparent image
261.   Pakker NG, Notermans DW, de Boer RJ, Roos MT, de Wolf F, Hill A, Leonard JM, Danner SA, Miedema F, Schellekens PT. Biphasic kinetics of peripheral blood T cells after triple combination therapy in HIV-1 infection: a composite of redistribution and proliferation. Nat Med 1998; 4:208-14.
transparent image
262.   Staszewski S, Miller V, Sabin C, Schlecht C, Gute P, Stamm S, Leder T, Berger A, Weidemann E, Hill A, Phillips A. Determinants of sustainable CD4 lymphocyte count increases in response to antiretroviral therapy. Aids 1999; 13:951-6.
transparent image
263.   Kaufmann GR, Bloch M, Zaunders JJ, Smith D, Cooper DA. Long-term immunological response in HIV-1-infected subjects receiving potent antiretroviral therapy. Aids 2000; 14:959-69.
transparent image
264.   Smith CJ, Sabin CA, Lampe FC, Kinloch-de-Loes S, Gumley H, Carroll A, Prinz B, Youle M, Johnson MA, Phillips AN. The potential for CD4 cell increases in HIV-positive individuals who control viraemia with highly active antiretroviral therapy. Aids 2003; 17:963-9.
transparent image
265.   Koval CE, Gigliotti F, Nevins D, Demeter LM. Immune reconstitution syndrome after successful treatment of Pneumocystis carinii pneumonia in a man with human immunodeficiency virus type 1 infection. Clin Infect Dis 2002; 35:491-3.
transparent image
266.   Jenny-Avital ER, Abadi M. Immune reconstitution cryptococcosis after initiation of successful highly active antiretroviral therapy. Clin Infect Dis 2002; 35:e128-33.
transparent image
267.   Mutimer HP, Akatsuka Y, Manley T, Chuang EL, Boeckh M, Harrington R, Jones T, Riddell SR. Association between immune recovery uveitis and a diverse intraocular cytomegalovirus-specific cytotoxic T cell response. J Infect Dis 2002; 186:701-5.
transparent image
268.   Jenny-Avital ER. A patient with refractory disseminated Mycobacterium avium after immune-reconstitution localized MAC. AIDS Clin Care 2003; 15:24-6.
transparent image
269.   Chien JW, Johnson JL. Paradoxical reactions in HIV and pulmonary TB. Chest 1998; 114:933-6.
transparent image
270.   Lucas GM, Chaisson RE, Moore RD. Highly active antiretroviral therapy in a large urban clinic: risk factors for virologic failure and adverse drug reactions. Ann Intern Med 1999; 131:81-7.
transparent image
271.   Ledergerber B, Egger M, Opravil M, Telenti A, Hirschel B, Battegay M, Vernazza P, Sudre P, Flepp M, Furrer H, Francioli P, Weber R. Clinical progression and virological failure on highly active antiretroviral therapy in HIV-1 patients: a prospective cohort study. Swiss HIV Cohort Study. Lancet 1999; 353:863-8.
transparent image
272.  Sterne JAC, May M, Costagliola D, et al. Does it matter where you came from? Prognosis of patients starting potent therapy, according to initial response. In: Program and Abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, MA. Abstract 181.
transparent image
273.   Nijhuis M, Deeks S, Boucher C. Implications of antiretroviral resistance on viral fitness. Curr Opin Infect Dis 2001; 14:23-8.
transparent image
274.  Deeks SG, Martin JN, Hoh R, et al. Continued reverse transcriptase inhibitor therapy is sufficient to maintain short-term partial suppression of multi-drug resistant viremia. Program and abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, Massachusetts. Abstract 640.
transparent image
275.  Fagard C, Lebraz M, Gunthard C. SSITT: a prospective trial of strategic treatment interruptions in 128 patients.
transparent image
276.   Lori F, Lewis MG, Xu J, Varga G, Zinn DE, Jr., Crabbs C, Wagner W, Greenhouse J, Silvera P, Yalley-Ogunro J, Tinelli C, Lisziewicz J. Control of SIV rebound through structured treatment interruptions during early infection. Science 2000; 290:1591-3.
transparent image
277.   Ruiz L, Martinez-Picado J, Romeu J, Paredes R, Zayat MK, Marfil S, Negredo E, Sirera G, Tural C, Clotet B. Structured treatment interruption in chronically HIV-1 infected patients after long-term viral suppression. Aids 2000; 14:397-403.
transparent image
278.   Garcia F, Plana M, Ortiz GM, Bonhoeffer S, Soriano A, Vidal C, Cruceta A, Arnedo M, Gil C, Pantaleo G, Pumarola T, Gallart T, Nixon DF, Miro JM, Gatell JM. The virological and immunological consequences of structured treatment interruptions in chronic HIV-1 infection. Aids 2001; 15:F29-40.
transparent image
279.   Ortiz GM, Wellons M, Brancato J, Vo HT, Zinn RL, Clarkson DE, Van Loon K, Bonhoeffer S, Miralles GD, Montefiori D, Bartlett JA, Nixon DF. Structured antiretroviral treatment interruptions in chronically HIV-1-infected subjects. Proc Natl Acad Sci U S A 2001; 98:13288-93.
transparent image
280.   Ruiz L, Carcelain G, Martinez-Picado J, Frost S, Marfil S, Paredes R, Romeu J, Ferrer E, Morales-Lopetegi K, Autran B, Clotet B. HIV dynamics and T-cell immunity after three structured treatment interruptions in chronic HIV-1 infection. Aids 2001; 15:F19-27.
transparent image
281.   Fischer M, Hafner R, Schneider C, Trkola A, Joos B, Joller H, Hirschel B, Weber R, Gunthard HF. HIV RNA in plasma rebounds within days during structured treatment interruptions. Aids 2003; 17:195-9.
transparent image
282.  Hecht FM, Wang L, Collier A. Is HAART for Primary/Early HIV Infection Associated with Improved Outcomes After Treatment Discontinuation? Program and abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, Massachusetts. Abstract 519.
transparent image
283.  Dybul M, Nies-Kraske E, Daucher M, et al. A randomized, controlled trial of long cycle structured intermittent versus continuous ARV therapy for chronic HIV infection. Program and abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, Massachusetts. Abstract 68lb.
transparent image
284.  Ananworanich J, Cardiello P, Srasuebkul P, et al. HIV-NAT 0014: A prospective randomized trial of structured treatment interruption in patients with chronic HIV infection. Program and abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, Massachusetts. Abstract 64.
transparent image
285.  Ruiz L, Gomez L, Domingo P, et al. A multi-center, randomized controlled clinical trial of continuous vs intermittent HAART guided by CD4+ T-cell counts and plasma HIV-1 RNA levels. Program and abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, Massachusetts. Abstract 65.
transparent image
286.  Lawrence J, Mayers D, Huppler Hullsiek K, et al, the CPCRA 064 Study Team of the Terry Beirn Community Programs for Clinical Research on AIDS. CPCRA 064: A randomized trial examining structured treatment interruption for patients failing therapy with multi-drug resistant HIV. Program and abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, Massachusetts. Abstract 67.
transparent image
287.  Katlama C, Dominguez S, Duvivier C, et al. Long-term benefit of treatment interruption in salvage therapy (GIGHAART ANRS 097). Program and abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, Massachusetts. Abstract 68.
transparent image
288.  Deeks SG. When to switch therapy. Symposium at the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003: Boston, Massachusetts.
transparent image
289.  Valentine F, DeGruttola V and the REMUNE 816 Study Team: immunological and virological evaluations of the effects of HAART compared to HAART plus an inactivated HIV immunogen after 32 weeks. Program and abstracts of the 6th Conference on Retroviruses and Opportunistic Infections; January 31-February 4, 1999; Chicago, Illinois. Abstract 346.
transparent image
290.   Kovacs JA, Vogel S, Albert JM, Falloon J, Davey RT, Jr., Walker RE, Polis MA, Spooner K, Metcalf JA, Baseler M, Fyfe G, Lane HC. Controlled trial of interleukin-2 infusions in patients infected with the human immunodeficiency virus. N Engl J Med 1996; 335:1350-6.
transparent image
291.   Davey RT, Jr., Chaitt DG, Albert JM, Piscitelli SC, Kovacs JA, Walker RE, Falloon J, Polis MA, Metcalf JA, Masur H, Dewar R, Baseler M, Fyfe G, Giedlin MA, Lane HC. A randomized trial of high- versus low-dose subcutaneous interleukin-2 outpatient therapy for early human immunodeficiency virus type 1 infection. J Infect Dis 1999; 179:849-58.
transparent image
292.  Hecht FM, Hare CB, McGrath MS. Interleukin-2 in Conjunction with HAART in Early HIV Infection Increases Naive and Memory CD4 Cells and Lowers Activation Markers. Program and abstracts of the 10th Conference on Retroviruses and Opportunistic Infections; February 10-14, 2003; Boston, Massachusetts. Abstract 649.
transparent image
293.   McFarland EJ, Harding PA, MaWhinney S, Schooley RT, Kuritzkes DR. In vitro effects of IL-12 on HIV-1-specific CTL lines from HIV-1-infected children. J Immunol 1998; 161:513-9.
transparent image
294.   Huang LR, Chen FL, Chen YT, Lin YM, Kung JT. Potent induction of long-term CD8+ T cell memory by short-term IL-4 exposure during T cell receptor stimulation. Proc Natl Acad Sci U S A 2000; 97:3406-11.
transparent image
295.   Lu W, Wu X, Lu Y, Guo W, Andrieu JM. Therapeutic dendritic-cell vaccine for simian AIDS. Nat Med 2003; 9:27-32.
transparent image
296.   Levine BL, Mosca JD, Riley JL, Carroll RG, Vahey MT, Jagodzinski LL, Wagner KF, Mayers DL, Burke DS, Weislow OS, St Louis DC, June CH. Antiviral effect and ex vivo CD4+ T cell proliferation in HIV-positive patients as a result of CD28 costimulation. Science 1996; 272:1939-43.
transparent image
297.   Levine BL, Bernstein WB, Aronson NE, Schlienger K, Cotte J, Perfetto S, Humphries MJ, Ratto-Kim S, Birx DL, Steffens C, Landay A, Carroll RG, June CH. Adoptive transfer of costimulated CD4+ T cells induces expansion of peripheral T cells and decreased CCR5 expression in HIV infection. Nat Med 2002; 8:47-53.
transparent image
298.   Roberts MR, Qin L, Zhang D, Smith DH, Tran AC, Dull TJ, Groopman JE, Capon DJ, Byrn RA, Finer MH. Targeting of human immunodeficiency virus-infected cells by CD8+ T lymphocytes armed with universal T-cell receptors. Blood 1994; 84:2878-89.
transparent image
299.   Napolitano LA, Lo JC, Gotway MB, Mulligan K, Barbour JD, Schmidt D, Grant RM, Halvorsen RA, Schambelan M, McCune JM. Increased thymic mass and circulating naive CD4 T cells in HIV-1-infected adults treated with growth hormone. Aids 2002; 16:1103-11.
transparent image
300.   Kirk O, Reiss P, Uberti-Foppa C, Bickel M, Gerstoft J, Pradier C, Wit FW, Ledergerber B, Lundgren JD, Furrer H. Safe interruption of maintenance therapy against previous infection with four common HIV-associated opportunistic pathogens during potent antiretroviral therapy. Ann Intern Med 2002; 137:239-50.
transparent image
301.   Palefsky J. Human papillomavirus infection among HIV-infected individuals. Implications for development of malignant tumors. Hematol Oncol Clin North Am 1991; 5:357-70.
transparent image
302.   Feingold AR, Vermund SH, Burk RD, Kelley KF, Schrager LK, Schreiber K, Munk G, Friedland GH, Klein RS. Cervical cytologic abnormalities and papillomavirus in women infected with human immunodeficiency virus. J Acquir Immune Defic Syndr 1990; 3:896-903.
transparent image
303.   Piketty C, Darragh TM, Da Costa M, Bruneval P, Heard I, Kazatchkine MD, Palefsky JM. High prevalence of anal human papillomavirus infection and anal cancer precursors among HIV-infected persons in the absence of anal intercourse. Ann Intern Med 2003; 138:453-9.
transparent image
304.   Vergis EN, Paterson DL, Wagener MM, Swindells S, Singh N. Dyslipidaemia in HIV-infected patients: association with adherence to potent antiretroviral therapy. Int J STD AIDS 2001; 12:463-8.
transparent image
305.   Carr A, Samaras K, Thorisdottir A, Kaufmann GR, Chisholm DJ, Cooper DA. Diagnosis, prediction, and natural course of HIV-1 protease-inhibitor-associated lipodystrophy, hyperlipidaemia, and diabetes mellitus: a cohort study. Lancet 1999; 353:2093-9.
transparent image
306.   Walli R, Herfort O, Michl GM, Demant T, Jager H, Dieterle C, Bogner JR, Landgraf R, Goebel FD. Treatment with protease inhibitors associated with peripheral insulin resistance and impaired oral glucose tolerance in HIV-1-infected patients. Aids 1998; 12:F167-73.
transparent image
307.   Friis-Moller N, Sabin CA, Weber R, d'Arminio Monforte A, El-Sadr WM, Reiss P, Thiebaut R, Morfeldt L, De Wit S, Pradier C, Calvo G, Law MG, Kirk O, Phillips AN, Lundgren JD. Combination antiretroviral therapy and the risk of myocardial infarction. N Engl J Med 2003; 349:1993-2003.
transparent image
308.   Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). Jama 2001; 285:2486-97.
transparent image
transparent image