Human Immunodeficiency Virus/Acquired Immunodeficiency Syndrome in the Infant


The human immunodeficiency virus type 1 (HIV-1) pandemic remains one of the greatest public health challenges in the 21st century. More than 3 decades after the initial descriptions of acquired immunodeficiency syndrome (AIDS) in adults and children, the epidemiology of HIV has evolved significantly in high-income as well as low- and middle-income countries (LMIC) because of extraordinary biomedical advances in prevention, coupled with programmatic scale-up of antiretroviral treatment (ART). Mother-to child transmission (MTCT) of HIV is the most common source of HIV infection among infants and children. Although HIV infection in children has been acquired in the past by the transfusion of contaminated blood or coagulation products, this mode of transmission has been virtually eliminated in the United States. In the United States and other high-income countries, widespread implementation of universal antenatal HIV testing, combination antiretroviral treatment (cART) during pregnancy, elective cesarean section, and avoidance of breastfeeding has led to virtual elimination of MTCT. Dramatic declines in the number of HIV-infected children who acquired the infection perinatally have been reported, and new pediatric infections are exceedingly rare. Rates of MTCT in the United States are less than 2%, and most new infections occur because of missed prevention opportunities. Availability of cART has led to improved survival of HIV-infected children into adolescence and adulthood, changing most HIV infections into a treatable chronic illness rather than a fatal disease.

In contrast, prevention of mother-to-child transmission of HIV (PMTCT) remains a major public health challenge in LMIC. It is estimated that more than 900 infections occur daily worldwide in children younger than 15 years, of which 90% occur via MTCT. More than 90% of these affected children reside in sub-Saharan Africa, where prolonged breastfeeding is the cultural norm. In these settings, postnatal transmission through breastfeeding accounts for 30% to 40% of new pediatric HIV infections. In recent years, remarkable advances have been made in the development of effective antiretroviral (ARV) interventions to reduce the risk of postnatal transmission of HIV during breastfeeding. In response to new evidence from clinical trials, the World Health Organization (WHO) has issued new HIV and PMTCT guidelines. In addition, progress has been made in increasing access to ART for people living with HIV/AIDS in LMIC. The possibility that success of PMTCT interventions in high-income countries could be extended to LMIC has led to an ambitious global plan toward virtual elimination of MTCT worldwide by 2015, developed by the Joint United Nations Programme on HIV/AIDS (UNAIDS).

This chapter reviews the epidemiology and prevention of HIV infection in infants; discusses evaluation and management of HIV-exposed infants; and highlights certain unique features of HIV infection in infants, with a focus on early diagnosis and treatment.

Epidemiology

Global Scope of the Problem

The HIV pandemic is a major public health problem, with cases reported to the WHO from virtually every country. At the end of December 2011, an estimated 34 million people (31.4-35.9 million) were living with HIV (an increase from 29.4 million in 2001), including 15.7 million women and 3.3 million children younger than 15 years. Nearly 97% of people living with HIV live in LMICs, and the adult HIV prevalence worldwide is estimated at 0.8%. Sub-Saharan Africa bears the brunt of the epidemic, accounting for 69% of HIV infections; approximately 23.5 million HIV-infected people live in sub-Saharan Africa, including more than 3 million children younger than 15 years, representing more than 90% of all children with HIV. South and Southeast Asia account for 4 million HIV-infected people, followed by 3 million people living with HIV in the Americas, including the Caribbean.

The complex and diverse epidemiology of HIV worldwide has evolved significantly after the peak of HIV incidence around 1997. Recent data from UNAIDS indicate an encouraging trend, with rates of new HIV infections continuing to decline among general populations. Overall, a greater than 20% decline in new HIV infections was noted in 2011 (when compared with 2001 incidence rates), with the most significant declines in the Caribbean and sub-Saharan Africa. In 2011, an estimated 2.5 million people became newly infected with HIV, including 330,000 children younger than 15 years. Although HIV testing centers have increased in many countries, approximately 50% of people living with HIV do not know their HIV status. In 2011, an estimated 1.7 million people died because of AIDS-related illness (24% fewer than in 2005), including 230,000 children younger than 15 years (20% less than in 2005).

Primary HIV infection among women of childbearing age fuels the perinatal HIV epidemic. Women constitute nearly one half of all HIV/AIDS infections worldwide, and more than one half of these (58%) living in sub-Saharan Africa. Young women and adolescent females are at highest risk of HIV acquisition because of a myriad of complex biologic, behavioral, and structural factors. In LMIC, antenatal HIV prevalence rates can vary considerably, ranging from less than 5% to greater than 35% in some high–HIV-burden countries. Although HIV prevalence among young pregnant women has declined in Kenya, Malawi, and Zimbabwe, most countries in Southern Africa have antenatal prevalence rates between 20% to 30%; rates can reach 35% to 40% in some antenatal clinics in South Africa, Swaziland, Lesotho, and Botswana. In 2009, an estimated 1.4 million HIV-infected women became pregnant, 90% of whom lived in 22 countries in sub-Saharan Africa and India. Approximately 900 HIV-infected infants are born each day.

In LMIC, children constitute 14% of new HIV infections worldwide and nearly a fifth of annual HIV deaths. Without access to combination ART, greater than 50% of HIV-infected children in sub-Saharan Africa die by their second birthday. In the absence of rapid identification and early access to ART, 26% of postnatal and 52% of perinatal HIV infections result in child mortality 1 year after acquiring HIV. Based on UNAIDS estimates, 17.3 million children have been orphaned by the AIDS epidemic.

United States

Human Immunodeficiency Virus in Women

Currently, approximately 1.2 million individuals in the United States are living with HIV/AIDS, including approximately 280,000 women. In 2010, an estimated 47,500 people became newly infected with HIV. Since the beginning of the HIV epidemic in the United States, men accounted for the majority of new HIV infections and AIDS diagnoses. However, the incidence of HIV infection among women increased gradually until the late 1980s but then declined in the early 1990s and has remained relatively stable over the past decade. Of reported AIDS cases in adults, women accounted for 8% in 1985, 13% in 1993, 20% in 1995, 23% in 1999, and 27% in 2000; at the end of 2010, an estimated 25% of adults and adolescent females aged 13 years or older and living with HIV were women; 15% of HIV-positive women are unaware of their infection. In 2010, there were 9500 new HIV infections among women, acquired mostly (84%) via heterosexual contact with an individual known to have or at high risk for HIV infection. Young women (25-44 years of age) accounted for the majority of new HIV infections among women.

Women of color, especially black/African-American, and Hispanic/Latina women are disproportionately affected by HIV infection compared with women of other races or ethnicities. Of the total number of new HIV diagnosis among women in 2010, 64% occurred in black/African-American females (representing 12% of the U.S. female population), followed by 15% in Hispanic/Latina females (representing ≈14% of the population). In contrast, white females (representing 68% of the female adult and adolescent population) accounted for only 18% of new HIV diagnosis among females. In 2010, the rate of new HIV infections (per 100,000 population) is estimated to be 20 times higher among black/African-American women compared with rate of white women, whereas the rate among Hispanic/Latina women was four times higher compared with white women; however, recent data from the Centers for Disease Control and Prevention (CDC) indicate a decrease in new HIV infections among black/African-American women ( n = 6100; 21% decrease between 2008 and 2010). Greater than 75% of women with AIDS are in the reproductive age group at the time of diagnosis. In 2009, 4693 deaths occurred among women with AIDS. In 2010, HIV infection was among the top 10 leading causes of death for black/African-American females aged 10 to 64 years and Hispanic/Latina females aged 25 to 44 years.

Regional HIV seroprevalence rates vary, with the highest rates found among women residing in the Northeast and southern states. In 2009, the top 10 states with the highest number of women/girls living with AIDS were New York ( n = 23,859), followed by Florida ( n = 15,081), California ( n = 7817), Texas ( n = 6795), New Jersey ( n = 6312), Maryland ( n = 6080), Pennsylvania ( n = 4961), Georgia ( n = 4679), Illinois ( n = 3638) and Puerto Rico ( n = 3326). During 2010, the highest rates of new AIDS diagnoses (per 100,000 population) among adult and adolescent females were in the District of Columbia (79.9), Maryland (18.5), and Louisiana (15.5), followed by Florida (14.7), the U.S. Virgin Islands (14.3), New York (14.1), New Jersey (11.6), Delaware (10.9), and Mississippi (10.3).

For women living with HIV, the most common mode of transmission is high-risk heterosexual exposure and intravenous drug use (IDU). Among black and Latina women, heterosexual transmission accounts for 85% and 82% of new HIV/AIDS diagnosis, respectively, compared with white women (72%). Injection drug use accounts for a greater proportion (28%) of new infections among white women. A number of social factors, such as poverty, tight social networks, and assortative mixing as well as lack of prenatal care, antenatal HIV testing, and early access to combination ART, contribute to the high rates of HIV infection among black women in the United States.

Human Immunodeficiency Virus/Acquired Immunodeficiency Syndrome in Infants and Children

At the end of 2009, using a confidential name-based HIV infection reporting mechanism, an estimated 10,834 children younger than 13 years were diagnosed with HIV in 46 states in the United States; of these, 9522 (88%) were infected via MTCT. In the United States, almost all new diagnosis of HIV/AIDS in children younger than 13 years resulted from MTCT. In 2006, approximately 8700 pregnant women with HIV gave birth, compared with 6000 to 7000 births in 2000, representing an increase of 30%. However, since the mid-1990s, the annual number of diagnoses of perinatally acquired HIV/AIDS cases has declined by greater than 90% in the United States as a result of routine antenatal HIV testing in conjunction with implementation of effective interventions to prevent transmission. In 1991, the annual number of perinatally acquired HIV infections peaked at 1650 infants but significantly declined to an estimated 215 to 370 cases in 2005, 182 cases in 2008, and 131 cases in 2009 . In 2010, an estimated 217 children younger than 13 years were diagnosed with HIV in the 46 states using a confidential name-based HIV infection reporting mechanism; of these, 162 (75%) acquired HIV perinatally.

Cases of AIDS in children have accounted for less than 1% of all reported AIDS cases in the United States. An estimated 9898 cumulative AIDS diagnoses have been reported among children; 91% of those children acquired HIV perinatally. Since the beginning of the HIV/AIDS epidemic through 2009, an estimated 5626 children younger than 13 years who were diagnosed with AIDS have died in the 50 states and the District of Columbia; of these, 4986 (89%) were infected via the perinatal route.

The racial and ethnic and geographic distribution of AIDS cases in children parallels that of women with AIDS. Racial/ethnic disparities in perinatal HIV/AIDS incidence have persisted since the early part of the epidemic; 78% of children with AIDS were black or Hispanic in 1981 to 1986. Of the 9522 perinatal HIV cases at the end of 2009, 63% were in black/African Americans, 22% were in Hispanics/Latinos, and 13% were in whites; these figures include persons of all ages who acquired HIV via MTCT. Of the perinatally infected children born during 2007 through 2009, the CDC estimated that black/African-American children had the highest HIV rate per 100,000 live births annually; however, the annual rate had declined from 15.2 in 2007 to 9.9 in 2009. Among Hispanic/Latino children, HIV rates had remained stable from 2007 through 2009.

Although significant declines in the rate of perinatally acquired HIV infection among infants have been documented in the United States, the rate of acquisition of HIV/AIDS among adolescents and young adults continues to increase, occurring primarily among populations of minority race or ethnicity. Young black men who have sex with men (MSM) are at highest risk; new HIV cases increased by 48% in young MSM from 2006 to 2009. Recent data from the CDC indicated a continued increase (22%) in new infections among young gay and bisexual men from 2007 to 2010. Infection among adolescent women is acquired primarily through heterosexual contact. In 2007, 31% of adolescents and youth aged 13 to 19 years diagnosed with HIV were females, compared with 23% of individuals aged 20 to 24 years and 26% of adults aged 25 years or older. In 2007, cases of AIDS in adolescents and young adults aged 13 to 24 years accounted for 4% of people living with HIV infection in the United States.

Although a dramatic decline (<2%) in the rate of perinatal HIV transmission has been documented, because of the relatively stable incidence of HIV among women of childbearing age, perinatal HIV infection will continue to occur each year, primarily among young black women, warranting continued surveillance. In addition to MTCT, transmission of HIV to infants, children, and adolescents can occur via sexual contact (vaginal, anal, or orogenital), mucous membrane exposure to contaminated blood or other body fluids, IDU, and percutaneous blood exposure from contaminated needles or other sharp objects, such as tattooing. Probable HIV transmission from HIV-infected caregivers to their infants via feeding blood-tinged prechewed food has been reported, but no transmission of HIV infection to household contacts through casual contact has occurred in the United States. Other modes of transmission of HIV, such as exposure to contaminated blood products, have declined significantly and almost eliminated because of effective screening methods.

Transmission

Mother-to-Child HIV Transmission: Rates, Timing, Risk Factors, and Mechanisms

Mother-to-child transmission of HIV can occur in utero, during labor and delivery, or postnatally through breastfeeding. In the United States, the transmission rate without PMTCT interventions is estimated to be 25% to 30%; in European studies, it was found to be lower, at 15% to 20%. A higher transmission rate of 25% to 45% has been observed among breastfeeding populations in sub-Saharan Africa. These variations in transmission rates likely reflect differences in infant feeding patterns, maternal and obstetric risk factors, viral factors, and methodologic differences among studies. Data suggest that most children are infected during the intrapartum period. Studies based on cord blood or newborn HIV polymerase chain reaction (PCR) testing indicate that 50% to 60% of the perinatal HIV transmission occurs around the time of labor and delivery.

An infant is considered to have been infected in utero if the HIV-1 genome can be detected by PCR or cultured from blood within 48 hours of birth. In contrast, a child is considered to have intrapartum infection if diagnostic assays, such as culture, PCR, and serum p24 antigen, are negative in blood samples obtained during the first week of life but became positive during the period from day 7 to 90 of life and the infant has not been breastfed. In the breastfed infant, 20% to 25% of HIV transmission occurs during pregnancy, 35% to 50% during labor and delivery, and another 25% to 35% during lactation.

Intrauterine Transmission

In utero transmission may occur through HIV infection in the placenta or fetal exposure to cell-free or cell-associated HIV in the amniotic fluid. In early studies using PCR and in situ hybridization technology, the virus was detected in some aborted fetuses of 8 to 20 weeks of gestational age and in amniotic fluid. Maternal decidual leukocytes, placental villous macrophages (Hofbauer cells), and endothelial cells stain positive for gp41 antigen and HIV nucleic acids. The placenta can be infected through CD4 + trophoblasts or through the occasional occurrence of chorioamnionitis. However, subsequent studies in animals and human fetuses reported almost no transmission during the first and second trimester of pregnancy. There is no clear predictive value for the identification of HIV in the placenta and the infection of the fetus or newborn, and important technical limitations exist in studies conducted on fetal or placenta tissues, particularly because of the difficulty in excluding contamination with maternal blood. Based on viral detection during the first 48 hours of birth, intrauterine transmission occurs in about 20% to 25% of infections. Statistical modeling data also suggest that most in utero HIV transmission occurs during the last few weeks before delivery when the vascular integrity of the placenta is disrupted.

Intrapartum Infection

Intrapartum transmission may occur because of direct exposure of the fetus or infant with infected maternal secretions during birth or maternal-to-fetal microtransfusions during uterine contractions. Intrapartum transmission is supported by studies failing to detect HIV in the first month of life in infants born to HIV-infected women but with subsequent detection of virus after 1 to 3 months of life. In a study by the French Collaborative Study Group, timing of transmission was estimated with a mathematical model. Data for 95 infected infants (infants seropositive at 18 months and infants who died of HIV disease before this age and who were exclusively bottle-fed) were used in the model, which indicated that one third of the infants were infected in utero less than 2 months before delivery (95th percentile). In the remaining 65% of cases (95% confidence interval [CI], 22% to 92%), the date of infection was estimated as the day of birth. The estimated median period between birth and the positivity of viral markers (HIV PCR or HIV culture) was 10 days (95% CI, 6% to 14%), and the 95th percentile was estimated at 56 days.

Additional evidence to support mucosal exposure to maternal virus during delivery as a likely route of transmission includes findings of increased HIV infection rates in first-born twins, increased risk associated with prolonged rupture of membranes, and the protective effect, although incomplete, of cesarean delivery before onset of labor. Some reports have documented an increased risk of MTCT associated with placental malaria infection, but not others.

Although numerous maternal, obstetric, infant, host genetic, and viral factors may modify perinatal HIV transmission risk, maternal plasma HIV RNA level is the strongest predictor of intrauterine and intrapartum transmission ( Box 22-1 ). Transmission can occur rarely, however, among pregnant women with low or undetectable serum levels of HIV around the time of labor and delivery. Other maternal risk factors associated with higher rates of perinatal HIV infection include women with advanced clinical disease, acute HIV infection during pregnancy, and low CD4 + counts. HIV viral load in cervicovaginal secretions is an independent risk factor for perinatal HIV transmission. Maternal genital ulcer diseases, especially herpes simplex virus (HSV) and syphilis and other coinfections (such as hepatitis C virus, hepatitis B virus, malaria, tuberculosis), may increase the risk. Behavioral risk factors including maternal substance abuse, cigarette smoking during pregnancy, and noncompliance to ART may also increase the risk of transmission.

Box 22-1
Risk Factors Contributing to Perinatal Human Immunodeficiency Virus (HIV) Transmission
ART, Antiretroviral therapy; HBV, hepatitis B virus; HCV, hepatitis C virus; HLA, human leukocyte antigen; HSV, herpes simplex virus; STIs, sexually transmitted infections; TB, tuberculosis; VL, viral load.

Maternal

  • High maternal viral load in plasma (and breast milk in breastfeeding populations)

  • Low maternal CD4 T-lymphocyte count

  • High vaginal/cervical shedding of HIV

  • Advanced maternal clinical disease

  • Concurrent genital ulcer disease resulting from STIs (syphilis, genital HSV)

  • Coinfections (TB, malaria, HBV, HCV)

  • Behavioral (cigarette smoking, substance abuse, poor adherence to combination ART)

Obstetric

  • Instrumentation (amniocentesis, invasive monitoring)

  • Chorioamnionitis

  • Prolonged rupture of membranes

  • Vaginal delivery (VL > 1000 copies/mL)

Fetal/Infant

  • Prematurity

  • Twin gestation (higher infection rate in first born twin)

  • Low birth weight

  • Infant feeding choice (breastfeeding duration, mixed infant feeding)

  • Maternal breast disease (mastitis, abscess, cracked or bleeding nipples)

Genetic/Immunologic

  • Maternal-infant HLA concordance

  • Upregulation of CCR5 receptor expression in placenta

  • Toll-like receptor 9 polymorphisms

  • β-Defensin polymorphisms

  • Mannose-binding lectin polymorphisms

Obstetric risk factors associated with increased risk of transmission include vaginal delivery, prolonged rupture of membranes, chorioamnionitis, and invasive obstetric procedures. Premature infants born to HIV-infected women have a higher rate of perinatal HIV infection than full-term infants. Data from a large international meta-analysis of 15 prospective cohort studies and a randomized controlled trial from Europe have shown that cesarean section performed before labor and rupture of membranes reduces perinatal transmission of HIV-1 by 50% to 87%, independent of the use of ART or zidovudine (ZDV) prophylaxis.

Viral and host genetic factors may also influence MTCT. Besides maternal viral load, other factors affecting transmission include viral subtype, circulating recombinant forms, resistance viral strains and replication kinetics, and fitness. Genetic and phylogenetic studies indicate that infant quasi-species are highly homogeneous and generally represent minor maternal variants, confirming that vertical transmission of clade B HIV occurs across a selective bottleneck. In one study, infant clones did not differ from the maternal clones in env length or glycosylation, and all infant variants used the CCR5 co-receptor but were not macrophage tropic. Preferential in utero transmission of HIV subtype C, compared with subtype A or D, has been reported.

Host genetic factors including maternal-infant human leukocyte antigen (HLA) concordance and maternal HLA class I homozygosity have been associated with increased risks of MTCT, whereas genetic variants of chemokine and chemokine receptors have yielded conflicting results. Genetic variants of Toll-like receptors (TLRs) and defensins (e.g., TLR9 and β-defensin 1), crucial determinants of innate immune response, may also influence host-virus interactions and MTCT.

Postnatal Infection

In LMIC, where prolonged breastfeeding (>6 months) is the cultural norm and safe replacement feeding is not affordable, feasible, sustainable, or safe, postnatal transmission of HIV through breast milk remains a significant challenge. In the absence of interventions, postnatal transmission of HIV through breastfeeding can account for one third to one half of all HIV infections globally and carries an estimated transmission risk of about 15% when breastfeeding is prolonged and continued into the second year of life.

Studies show that the highest risk of breast-milk HIV transmission occurs during the first 4 to 6 weeks of life, varying from 0.7% to 1% per week, with a lower but continued risk thereafter. In studies from sub-Saharan Africa, the risk of late postnatal HIV transmission after 4 to 6 weeks of age was 8.9 infections per 100 child-years of breastfeeding and relatively constant at approximately 0.7% per month of breastfeeding. Acute HIV infection in the mother during breastfeeding confers a higher risk of MTCT because of high levels of viral replication in maternal plasma and breast milk. In one report, the cumulative risk of breast-milk HIV transmission was 14% for chronic maternal infection compared with 25% to 30% for maternal infections acquired during late pregnancy or lactation.

Risk factors for breast-milk HIV transmission include women seroconverting during lactation, high HIV DNA or RNA level in maternal plasma and breast milk, low maternal CD4 + cell count, maternal symptomatic disease or AIDS, prolonged duration of breastfeeding, mixed infant feeding (breast milk plus formula milk) in the initial few months of life, oral lesions in the infant (e.g., candidiasis), abrupt weaning, and maternal breast problems (e.g., bleeding or cracked nipples, subclinical and clinical mastitis, and breast abscesses).

HIV has been isolated from cell-associated (DNA) and cell-free fractions of human breast milk from HIV-infected women and has been detected by culture or PCR in varying frequencies (39%-89%) in many studies. Breast-milk viral load levels are very high soon after birth. However, viral shedding may be intermittent, and variation in breast-milk viral load between left and right breasts during the first 3 months of lactation has been reported.

Mechanisms of Transmission

The mechanisms of MTCT are complex, requiring the virus to breach a mucosal barrier (placenta, the oral mucosa, or gastrointestinal [GI] tract). Mucosal transmission may be enhanced via the interaction of HIV with several C-type lectins, including dendritic cell–specific intercellular adhesion molecule-3–grabbing nonintegrin (DC-SIGN), expressed on dendritic cells located beneath the mucosal epithelium. DC-SIGN serves as an HIV-1 receptor, mediates virus adhesion to DCs, and recognizes pathogen-associated molecular patterns (PAMPs). DC-SIGN heightens HIV capture at mucosal entry sites, allowing its transport to CD4 T lymphocytes through transinfection. DC-SIGN is expressed on placental macrophages (Hofbauer cells), and genetic polymorphisms of this molecule have been associated with risk of MTCT.

In general, most infants escape infection despite an immature immune system and continued repetitive exposure to the virus (e.g., breastfeeding). Of the various routes of MTCT, transplacental HIV transmission is the least efficient. Some experts have hypothesized that fetal T-regulatory (Treg) cells may curb intrauterine transmission, given the ability of these cells to suppress inflammatory responses. Treg cells have been shown to suppress immune responses when exposed to maternal antigens in utero. Exposure to certain antigens (e.g., placental malarial infection) in the intrauterine period has been associated with increased Treg cells at birth. In addition, robust HIV-specific T-cell responses have been demonstrated in HIV-exposed, uninfected infants after depletion of Treg cells in vitro. Further, placental inflammation is associated with in utero. The gut of newborn infants has an abundance of memory CD4 + CCR5 + T cells, which are targets for HIV infection and facilitate MTCT. Likewise, fetal tissues and cells are more susceptible to HIV infection than adult cells. Thus other amniotic and/or placental factors may play a role in suppression of fetal gut inflammatory responses in utero.

Most intrapartum MTCT likely occurs via mucosal exposure. This is supported by studies demonstrating the protective effect of cesarean section, presence of HIV in infant gastric aspirates, and presence of CD4 + CCR5 + T cells in the neonatal gut. In addition, inflammatory cytokines and chemokines found in placentas of transmitting mothers are absent in placentas of mothers who transmit during intrapartum period. MTCT generally occurs in the presence of HIV-specific antibody transferred across the placenta during the third trimester of pregnancy. The role of maternal neutralizing and/or nonneutralizing antibodies in influencing MTCT risk is unclear, although in simian immunodeficiency models, broadly neutralizing antibodies can prevent infection and modify immune responses in infected neonatal macaques.

The mechanisms of breast-milk HIV transmission are poorly understood. A complex interplay of factors, including the nature and size of HIV reservoirs in breast milk, host susceptibility, various immune and nonimmune breast-milk factors, may be responsible for postnatal transmission during breastfeeding. Only a minority of breastfed infants born to HIV-infected mothers acquire postnatal infection by unknown mechanisms, despite prolonged and repeated daily exposure to HIV in breast milk. The reported probability of breast-milk transmission of HIV is extremely low, at 0.00064/L ingested and 0.00028/day of breastfeeding. Prolonged breastfeeding over a period of several months to years can result in postnatal transmission rates of greater than 15%.

The primary HIV reservoirs in human milk are RNA (as cell-free virus), DNA (as cell-associated virus integrated in latent T cells, and intracellular RNA (as cell-associated virus in activated T cells). HIV can gain entry into breast milk via passive transfer from the vascular compartment or through local replication in mammary gland tissues and breast milk. The mammary epithelium serves as an effective barrier to HIV entry. Compared with plasma, the concentration of cell-free HIV RNA in breast milk is approximately 2 logarithms lower. Cell-free, as well as cell-associated, viruses are associated with breast-milk HIV transmission. However, recent studies suggest that cell-associated virus (either latently infected or activated virus-producing T cells) in breast milk may be a stronger predictor for transmission of HIV to the infant than cell-free virus. cART administered during pregnancy or postpartum can suppress cell-free HIV RNA but not cell-associated HIV DNA load in breast milk. Another recent report found that cell-associated virus level (per mL) is more important for early postpartum HIV transmission (at 6 weeks) than cell-free virus.

Inflammatory breast conditions (e.g., mastitis or abscess) can result in increased mammary epithelial cell (MEC) permeability (rise in breast-milk sodium levels), viral shedding, and transmission. Subclinical mastitis (defined by elevated breast-milk sodium levels in asymptomatic women) can occur in mothers who do not breastfeed exclusively or during weaning. In such instances, infrequent breast emptying can lead to ductal inflammation and increased MEC permeability. Subclinical mastitis has been associated with increased breast-milk HIV loads and a higher risk of postnatal HIV transmission through breast milk. However, a study from Zimbabwe found that laboratory indicators of mastitis (e.g., sodium levels and cell counts) were not predictive of increased breast-milk HIV RNA levels.

The origin and evolution of HIV in breast milk is an area of ongoing research. HIV variants found in breast milk are genetically indistinguishable from those present in plasma. Studies indicate that viral evolution is limited in breast milk, reflecting restricted replication. A recent report demonstrated that breast milk may be initially seeded by an early HIV variant differing from the viral variants present in maternal plasma. Thereafter exchange of distinct viral sequences between plasma and breast milk results in multiple independent lineages of HIV without compartmentalization.

Although the virus enters through the infant’s mouth during breastfeeding, the precise portal of entry of HIV at various mucosal targets (e.g., oral mucosa, tonsils, GI tract) is unclear. HIV in breast milk may enter the submucosal layer of the infant gut because of breaks in the intestinal epithelial cell layer, which may occur from inflammation caused by nonexclusive breastfeeding or other coexisting pathogens. Low gastric acidity in neonates may facilitate entry of cell-free or cell-associated virus into intestinal epithelial cells. Mixed infant feeding may result in mucosal inflammation and immune activation in the infant GI tract, possibly facilitating HIV transfer across the gut lumen.

Although mucosal breaches facilitate viral entry, transmission of HIV may also occur through intact fetal oral and intestinal mucosal membranes. Transmission of HIV across the intestinal mucosal barrier is believed to occur after the virus attaches to immature dendritic cells of the infant gut, which then transport antigen to Peyer patches of the intestinal mucosa. Once the mucosal barrier is breached, HIV infects CD4 + T cells and disseminates to the draining lymph nodes and the lymphoid system.

Breast milk contains myriad factors with immunologic, antimicrobial, and antiinflammatory properties that may reduce the risk of transmission of HIV during breastfeeding among infants, despite repeated daily exposure to the virus. Some innate factors (e.g., mucin, polyanionic proteins, lysozyme, lactoferrin, bile salt–stimulated lipase, and secretory leucocyte protease inhibitor) in human milk exhibit strong HIV inhibitory activity in vitro. For example, bile salt–stimulated lipase can bind DC-SIGN and differentially inhibit viral capture and infection of CD4 T lymphocytes. Increased concentration of long-chain n-6 polyunsaturated fatty acids, human milk oligosaccharides, α-defensins, and erythropoietin in breast milk has been correlated with decreased risk of postnatal transmission of HIV during breastfeeding.

Breast milk also contains soluble/humoral and cellular factors that mediate innate and adaptive immune effects, including cytokines (interferon-γ [IFN-γ]), chemokines (regulated on activation, normal T-cell expressed and secreted [RANTES]), HIV-specific antibodies, CD8 + HIV-specific cytotoxic T lymphocytes, CD4 + HIV-specific helper T lymphocytes, natural killer (NK) cells, and macrophages. The source of HIV in breast milk is likely infected T cells or macrophages derived from gut-associated lymphoid tissue (GALT) via the enteromammary axis. CCR5-expressing memory CD4 T cells represent the majority of lymphocytes in breast milk. Compared with peripheral blood CD4 T + cells, breast-milk T lymphocytes exhibit primarily an effector memory phenotype (CD45RO + ), express more activation markers, display stronger expression of HIV co-receptors, and demonstrate mucosal homing receptors, identical to B cells located in GALT. The presence of breast-milk cellular immune responses against HIV has also been studied, but their significance in decreasing transmission is unclear. In recent studies, HIV–Gag-specific IFN-γ responses were detected among HIV-exposed breastfeeding infants and have been associated with early protection from MTCT.

Activated latently infected CD4 T + cells favor viral replication and release from the persistent stable HIV reservoirs in breast milk. Recent studies indicate that latently infected CD4 cells and spontaneously activated CD4 T + cells are not affected by maternal ART and may play an important role in residual breast-milk HIV transmission. Breast-milk cellular reservoirs and possible cell-to-cell HIV transfer from breast milk to the infant’s intestinal mucosae remain a concern regarding the feasibility of elimination of breastfeeding transmission of HIV. The biologic relevance of HIV-specific T cells in breast milk needs future research. Although breast milk contains maternal HIV-specific antibodies (secretory immunoglobulin A [IgA], secretory IgM, and IgG), their role in protection against postnatal HIV infection is uncertain. Likewise, the role of functionally effective neutralizing antibodies in preventing breast-milk HIV transmission is unknown and warrants future studies.

Molecular Biology

Significant progress has been made in understanding the structural and molecular biology of HIV. Based on the sequence homology of the envelope proteins, two types of HIV viruses, HIV-1 and HIV-2, can cause AIDS in humans. HIV-1 and HIV-2 are lentiviruses belonging to the Retroviridae family and have a complex genomic structure. The HIV viruses are closely related to the simian immunodeficiency viruses (SIVs), which are agents in African green monkeys and sooty mangabey monkeys (Cercocebus atys) . In 1989, the virus closely related to HIV-1 (SIVcpz) was found in chimpanzees (Pan troglodytes), whose natural range is in West-Central Africa (Cameroon, Equatorial Guinea, Congo, and Central African Republic), whereas the virus closely related to HIV-2 (SIVsm) was found in sooty mangabey monkeys, whose natural range is in West Africa. Although HIV-1 and HIV-2 share similar transmission routes, cellular targets, and AIDS-defining manifestations, HIV-2 infection is characterized by lower transmission rates, slower disease progression, and a lower mortality rate compared with HIV-1.

HIV-1 variants are classified into four distinct groups: group M (main), group O (outlier), group N (new), and a recently described group P, each representing an independent cross-species transmission event from chimpanzees and/or gorillas to humans. Group M accounts for most HIV-1 infections globally and is divided further into 10 subtypes or clades (A to K), sub-subtypes, and inter-subtype circulating recombinant forms (CRFs). The HIV-1 subtypes and CRFs vary considerably from region to region. Subtype B is the predominant HIV-1 subtype in North America, the Caribbean, Latin America, and Australia, whereas subtypes C predominate in Southern Africa, Ethiopia, and India. There is considerable genetic diversity in Central Africa, Western and Central Europe, where all subtypes and many CRFs are implicated. In Western and Central Europe, all major subtypes and many CRFs and unique recombinant forms (URFs) are detected. In Eastern Europe and Central Asia, the HIV-1 epidemic is dominated by subtype A and subtype B. The majority of infections in South and Southeast Asia are caused by CRF01_AE subtype.

HIV-1 is an enveloped cytopathic virus with a diameter of approximately 110 to 150 nm. The outer coat of the virus is composed of a lipid bilayer membrane envelope surrounding a cylindric, electrodense core of structural proteins. The HIV core includes the replication enzymes, viral genomic RNA, and is encased by a cone-shaped shell consisting of the viral capsid protein. Embedded throughout the outer coat of the virus are host cellular proteins, as well as trimeric envelope (Env) spikes that protrude through the surface of the HIV virion. Each Env spike is composed of three surface glycoproteins gp120, each of which is noncovalently anchored by the transmembrane glycoprotein gp41 molecule. The outer subunits of gp120 and gp41 are derived from a common precursor, gp160, and remain primary targets for vaccine development.

The bullet-shaped retroviral core or capsid consists of 2000 copies of the viral protein p24 and contains two copies of the single-stranded HIV RNA, each of which possesses a copy of viral genes. Both ends of the HIV RNA strands contain an RNA sequence called the long terminal repeat (LTR). Viral or host cellular proteins can activate certain regions of the LTR, which act as molecular switches to control production of new viruses. Similar to all retroviruses, HIV-1 contains structural and regulatory genes and accessory proteins. The structural genes include gag, which encodes the core nucleocapsid polypeptides (gp24, p17, p9); env, which encodes for the surface-coated proteins of the virus (gp120 and gp41); and pol, which encodes for the viral replication enzymes (reverse transcriptase, integrase, and protease) involved in the later steps of the virus life cycle. In addition, there are two regulatory (Tat and Rev) and four accessory proteins (Vif, Vpr, Vpu, and Nef) that are essential for viral replication and pathogenicity.

The regulatory proteins Tat and Rev are expressed early during cellular infection, whereas virion-associated proteins, such as Env, Pol, and Gag are expressed later. The accessory proteins often play an important role in modulating the host immune response and are major determinants of HIV-1 species tropism. For example, Vif, a cytoplasmic phosphoprotein encoded by the vif gene during the late stage of the HIV life cycle, acts by preventing viral encapsidation of a potent restriction factor in host cells (apolipoprotein B messenger RNA [mRNA]-editing enzyme-catalytic APOBEC3G/F protein), resulting in inactivation of the antiviral effect and enhancing HIV replication. Besides Vif-APOBEC proteins, other major viral and cellular determinants for HIV-1 species tropism include Gag-CA/CypA, Gag-CA/tripartite motif (TRIM) proteins, and Vpu-Tetherin/BST-2. More recently, two studies showed that the SIV Vpx protein can counteract inhibition of HIV-1 infection of macrophages mediated by the sterile alpha motif and histidine-aspartic (HD) domain–containing protein-1 (SAMHD1), a myeloid-cell–specific HIV-1 restriction factor.

The life cycle of HIV-1 is characterized by several distinct steps, beginning with viral entry into host cells engaging cell surface receptors, uncoating and integration, viral mRNA biogenesis and transport, and ending with viral egress and maturation of nascent particles into infectious virions. A wide array of host proteins are involved in the replication cycle, whereas certain host restriction factors counteract to suppress viral replication. HIV-1 primarily infects CD4 T-lymphocyte cells, macrophages, and other host cells (e.g., dendritic cells). During sexual transmission of HIV, dendritic cells located at or near mucosal sites are the first target cells for initiation of HIV infection.

Members of the chemokine receptor family are coreceptors for HIV-1 entry into target cells. The main HIV coreceptors are CC chemokine receptor 5 (CCR5) or CXC chemokine receptor 4 (CXCR4). Early reports described two classes of HIV-1 based on their ability to infect CD4 + T cells (T tropic) or macrophages (M tropic). Subsequently, it was discovered that T-tropic viruses use the CXCR4 chemokine coreceptor (called X4 viruses), whereas M-tropic viruses use the CCR5 chemokine co-receptor (called R5 viruses). HIV-1 may use CCR5 (R5) or CXCR4 (X4) co-receptors either individually or in combination (R5X4). HIV-1 phenotype (co-receptor usage and tropism) is determined by genetic sequences within gp120 molecule, especially in the highly variable envelope V3 residues.

The first step in the entry process of HIV into a cell is the binding of virion Env glycoprotein gp120 to the host cell-surface receptor CD4, followed by interactions with a co-receptor CC chemokine receptor (CCR5) or CXC chemokine receptor type 4 (CXCR4). This binding results in a cascade of conformational change in the Env that exposes a fusion domain in gp41, resulting in fusion of the viral and host cell membranes, allowing the viral genome to gain entry into the cell. HIV virions enter the cell and are partially uncoated, facilitating the process of reverse transcription, an essential step in retroviral replication. The viral reverse-transcriptase enzyme transforms the single-stranded viral RNA into linear double-stranded DNA, whereas the less specific ribonuclease H (RNase H) degrades and removes the viral genomic RNA template. This linear viral DNA molecule is circularized, yielding the preintegration complex (PIC). After nuclear import, the retroviral PIC is inserted into a host cell chromosome by the viral integrase at random sites as an integrated provirus, aided by the host chromatin-binding protein lens epithelium-derived growth factor. Proviral transcription by host RNA polymerase II, aided by the viral transactivator protein (Tat) and positive transcription elongation factor b (P-TEFb) results in production of viral mRNAs of varying sizes. The smaller viral mRNAs are exported easily from the nucleus into the cytoplasm, whereas the export of full length (unspliced) and partially spliced mRNAs is regulated by the viral Rev protein and aided by the host nuclear export factor CRM1 protein. mRNA templates serve as templates for Gag, Pol, and Env proteins, and genome-length RNA is packaged into viral particles with protein components.

HIV-1 becomes enveloped while budding through the plasma membrane. During or after retroviral budding, the precursor polypeptide Gag and Gag-Pol is cleaved by the viral protease, resulting in synthesis of retroviral structural proteins capsid (CA), matrix (MA), and nucleocapsid (NC) proteins and the enzymes reverse transcriptase, integrase, and protease, leading to the formation of the mature conical core. Virus maturation is crucial for particle infectivity. Proteins of the ESCRT (endosomal sorting complex required for transport) complex and ALG-2-interacting proteinX (ALIX) mediate viral particle budding, separation of the viral envelope from the plasma membrane, and release of infectious virions.

Although integration is a central event in the replication cycle of retroviruses, it is also a common feature of all retroviruses to accumulate large amounts of unintegrated viral DNA in cells that are fully competent templates for HIV-1 core and envelope antigen production. The latent proviral DNA represents a viral reservoir that is unaffected by ART and can be activated by host-cell responses to antigens, mitogens, or cytokines. Latently infected cells, primarily resting memory CD4 T cells, remain a major hurdle to achieving a sterilizing or a functional HIV-1 cure.

Immunopathogenesis

Research conducted over the past 2 decades has yielded significant insights into understanding of the viral and immunopathogenesis of HIV infection. Dendritic cells or macrophages are initially targeted by HIV, an interaction that transmits the virus to CD4 + T cells. The activation and direct infection of CD4 + T cells result in high rates of viral production and eventually severe immune deficiency resulting from CD4 + T-cell depletion. CD4 + T-cell loss in HIV infection occurs because of direct viral-induced cell killing as well as immune activation–induced cell death.

It is well established that the mucosal immune system plays a vital role in the transmission and pathogenesis of AIDS. After an early burst of viremia and dissemination, rapid and profound loss of “activated” memory CD4 + T cells (mostly Th17 phenotype) in the GALT, particularly in the intestine, is a hallmark of early HIV infection in adults. In acute infection, CD4 depletion and impairment of the intestinal barrier from enteropathy and subsequent microbial translocation result in chronic immune activation. Recent evidence suggests that HIV-induced chronic immune activation via activation of the innate and adaptive immune system plays a central role in the immunopathogenesis of HIV/AIDS. Although progress has been made in the immunology of pediatric HIV infection, the true correlates of immune protection and immune failure and mechanisms of infection in neonatal target cells in the context of extreme viral and HLA diversity are poorly understood.

Viral Replication in Early Infant Infection

Human immunodeficiency virus strains transmitted from mother to child are typically minor maternal variants, predominantly macrophage-tropic and nonsyncytium-inducing R5 strains, that use CCR5 co-receptor to infect CD4 + T cells, although use of CXCR4 (X4) has been reported. Compared with adult cells, HIV replicates more readily in neonatal T lymphocytes and monocytes or macrophages, and this increased susceptibility is influenced by differential mRNA expression of several host genes associated with the virus life cycle. In vitro studies suggest that integration of HIV into highly conserved host genes in neonatal (cord blood) cells may contribute to increased gene expression and viral replication compared with adult cells. Analysis of many HIV structural, regulatory ( tat and rev ), and accessory ( vif, vpr, vpu, nef ) genes, has revealed conservation of functional domains of these genes during vertical transmission. In addition, the vif and vpr sequences of transmitting mothers were more heterogeneous and more functional than sequences of nontransmitting mothers.

Other HIV genes may also play a crucial role in virus transmission and pathogenesis. Functional domains in the HIV-1 functional LTR are conserved during vertical transmission, suggesting that a functional LTR sequence is crucial in viral gene expression, transmission, and pathogenesis. Other properties exhibited by vertically transmitted viruses include enhanced replication kinetics and fitness compared with nontransmitted viruses.

A recent study showed that fetal and neonatal intestine have a large subset of memory CD4 + CCR5 + T cells with predominantly a Th1 and Th17 phenotype, suggesting that the neonatal gut may be a primary target for HIV infection and replication, as observed in adults. A study in neonatal rhesus macaques demonstrated a rapid and profound loss of intestinal CD4 + T cells after acute SIV infection. Compared with adult macaques, intestinal CD4 + T-cell turnover and proliferation are markedly increased in infant macaques and may result in a more sustained pool of selective target cells for HIV replication. Increased viral replication may lead to a more rapid exhaustion of a finite precursor CD4 + T-cell pool, resulting in faster disease progression in infants.

Perinatal HIV infection is characterized by plasma RNA levels that rapidly reach very high levels, often exceeding 10 5 to 10 7 copies/mL during the first year of life, decreasing only slowly with age to a “setpoint” by approximately 5 years of age. In a study of 106 HIV-infected infants, the median plasma HIV RNA value at 1 month of age was 318,000 copies/mL, and it was common to see viral levels that exceeded 10 6 copies/mL. In the absence of ART, HIV replication is very efficient, and plasma HIV RNA levels exceeding 1 million copies/mL are not unusual, with levels declining gradually over the first 24 to 36 months of life. A continued decrease in plasma HIV-1 RNA levels (mean, −0.2 to −0.3 logarithm decline/year) has been noted in vertically infected children through 5 to 6 years of age. Compared with adults, peak and setpoint viral load are usually greater than 1 logarithm greater in perinatal HIV infection. Viral loads are higher among infants who acquire HIV infection during the peripartum period, compared with those infected postnatally through breast milk. In contrast, the viral setpoint in many adults is attained within weeks of acute infection because of HIV-specific T-cell responses, remains relatively steady for several years during clinical latency, and is an important predictor for subsequent disease progression.

As in adults, higher infant viral loads correlate with a more rapid disease progression and high risk of mortality in the absence of ART, whereas lower levels of plasma HIV RNA with combination ART are associated with clinical benefit. These data indicate that high viral load is a critical determinant of pediatric disease progression and provide a strong argument for early and aggressive intervention with ART. In one study, significant age-related differences in HIV disease progression were noted in vertically infected children younger than 5 years. Younger children were at higher risk for disease progression or mortality compared with older children, irrespective of plasma viral load or CD4 lymphocyte count. In children older than 5 years, the risk of disease progression and mortality was similar to that noted in young adults; viral load and CD4 lymphocyte level were important predictors of HIV progression. In contrast, the clinical course of HIV infection in adolescents and adults is relatively slow, with progression to AIDS or death within 10 years among untreated patients.

The very high plasma HIV RNA levels in infants with HIV infection may be related to many factors, including the increased susceptibility of neonatal target cells to HIV-1 infection, a large and renewable CD4 T-cell pool size, presence of an active thymus, and delayed or ineffective HIV-specific immune responses.

Human Immunodeficiency Virus–Specific Immune Control

Vertically infected infants face many challenges in mounting a specific immune response to HIV. Perinatal HIV transmission occurs before the immune system is fully developed in an infant, allowing for more efficient viral replication and less efficient immunologic containment of the virus. The immunologic milieu in the neonate is tolerogenic, characterized by enhanced regulatory T-cell activity and increased interleukin 10 (IL-10) production. The adaptive immune response in early months of life is characterized by the preferential induction of Th17- and Th2-type immunity, which facilitates clearance of extracellular pathogens rather than intracellular pathogens (e.g., viruses). In addition, there is decreased production of type I IFNs and Th1 decrease in cytokines, such as IL-12 and IFN-γ .

In adults with acute HIV infection, HLA class I–restricted CD8 + (cytotoxic) T lymphocytes (CTL) play a crucial role in generation of HIV-specific immune responses to reduce viral load. CTL lyse HIV-infected cells through the recognition of virus-derived peptides (or “epitopes”) presented on the surface of the infected cells by HLA class I molecules. HLA-restricted CTL responses drive the evolution of HIV through selection of viral sequence polymorphisms (“immune escape mutations”).

Although HIV-specific CTL activity can be shown at a very early age, even in the fetus, the response is weak and less broad to optimally reduce viral load in infants. In addition, vertically transmitted maternal virus may be preadapted to the infant’s HLA alleles, affecting the infant’s ability to mount an early CTL response restricted by shared HLA alleles. HIV-specific CTL responses become more frequent and broad in infected infants with age. In one study, the Gag-specific CD8 T-cell response at 3 months of age correlated with clinical outcome and survival at 12 months. Another study found that robust functional CD8 T-cell responses are not detected until 3 years of age in vertically acquired children, primarily among those with adequate CD4 T-cell compartment. Polyfunctional HIV-specific CD8 T-cell responses are associated with slower disease progression in children as in adult HIV infection. Immune activation and the upregulation of programmed cell-death protein 1 (PD1), an inhibitory molecule produced by CD8 T cells, have been noted during the course of HIV infection in children. The narrow CTL responses in infants can result in viral escape mutants within the first year of life. In addition, persistent viral replication may lead to the selection of compensatory mutations that increase viral fitness. Certain HLA alleles (HLA-B 27, HLA-B 57, Cw-2, or DQB1-2) are associated with slower disease progression, indicating that CTL responses can have an important role in suppression of viral replication in pediatric HIV infection.

Compared with adults, HIV-specific CD4 T-cell responses are not detected initially in most vertically infected infants; subsequent responses remain narrow and low magnitude but increase with age. Studies in infant SIV-macaque model suggest that increased regulatory T-cell activity in early life may contribute to the suppression of viral specific CD4 T-cell responses. HIV-specific CD4 T-cell responses may also be important for T-cell–mediated viral control and enhance the capacity of HIV-specific CD8 T cells to suppress viral replication. In one study, the frequency of Gag-specific CD4 T cells in infected infants at 3 and 6 months of age correlated inversely with viral load. In another study, the magnitude of Gag-specific CD4 T-cell responses was associated with control of viral replication in ARV-naïve children.

The passive transfer of maternal, nonneutralizing antibodies could inhibit development of HIV-specific immune responses. Detection of antibodies mediating HIV-specific antibody-dependent, cell-mediated cytotoxicity (ADCC) is delayed in most vertically infected infants until after 1 year of age. Defective ADCC activity against HIV-infected target cells in neonates may also result in poor control of HIV. After the decline of passively transferred maternal antibody-dependent, cell-mediated cytotoxicity antibodies, the production of HIV-envelope cytotoxic antibodies is delayed in vertically infected infants. Although neutralizing antibodies can be generated during early infection, the precise role of neutralizing antibodies in limiting mother-to-child transmission of HIV is unclear and warrants further investigation.

The viral load in perinatally acquired HIV infection eventually decreases over time, although the precise mechanisms remain unknown. In the absence of ART, less than 10% of vertically infected children are well by 8 years of age, referred to as HIV long-term nonprogressors (LTNPs). There are fewer reports of LTNPs or “slow progressors” in children. In one study, lower levels of immune activation were noted among LTNP in the first weeks of perinatally acquired HIV infection. In adult HIV infection, immune activation results in CD4 + T-cell loss and is an important prognosis marker for disease progression than viral load. In contrast, CD8 T-cell activity may not be significant in the neonate given the tolerogenic regulatory T-cell environment and low-magnitude Th-cell responses. In pediatric slow progressors, the decline of HIV viremia eventually occurs because of increase in the breadth and magnitude of HIV-specific adaptive immune responses over time.

Immune Consequences in Human Immunodeficiency Virus Infection

Untreated HIV infection results in profound deficiencies in cell-mediated and humoral immunity caused by quantitative and qualitative defects. Progressive dysfunction of the immune system with selective depletion of CD4 + T cells occurs because of direct infection and immune activation. Increased immune activation can occur in HIV-infected children in association with decreased naïve T-cell numbers, increased apoptosis, and accelerated T-cell differentiation. In infants, thymic dysfunction associated with HIV infection may also have a significant impact on the developing immune system, leading to progressive depletion of thymic CD4 T-lymphocyte cells, dramatic decrease in cortical CD4/CD8 double-positive cells, and an increased percentage of CD8 cells.

Compared with adults, CD4 + T-cell depletion may be less striking in children because of their relative lymphocytosis. Flow cytometric analysis of lymphocyte subpopulations in healthy children revealed age-related changes in many of the different subgroups. Comparison of lymphocyte subsets in HIV-infected versus noninfected children younger than 2 years showed no difference for absolute CD8 + counts but clearly decreased levels of CD4 + cells. HIV-specific CD4 T-cell responses are crucial for B-cell antibody formation in addition to generation of HIV-specific CD8 T-cell responses to control viremia.

Abnormalities of the humoral immune system can precede or accompany abnormalities of the cellular immune system in HIV infection and consist of polyclonal B lymphocyte activation, resulting in hypergammaglobulinemia (especially IgG and IgA), abnormal primary or secondary antibody responses (T-lymphocyte–dependent and –independent antigens), decreased lymphocyte proliferation in response to an antigen; in addition, impaired function of other immune cells (monocytes/macrophages, NK lymphocytes, dendritic cells, and neutrophils) has been reported. Panhypogammaglobulinemia is noted in less than 10% of patients and is associated with poor prognosis.

Several studies indicate that HIV-infected children have reduced antibody responses to certain childhood vaccines (e.g., diphtheria, acellular pertussis vaccine). Reduced antibody responses after immunization and vaccine failures in HIV-infected infants may result from a poor primary immune response, failure to generate memory responses, or loss of memory cells. Most vertically infected infants who receive cART before 3 months of age develop antibody and lymphoproliferative responses to routine infant vaccines, although persistent HIV-specific immune responses are not detected. More recent studies have reported a sustained increase in peripheral blood CD5 cell counts and robust immune reconstitution among HIV-infected children after prolonged cART, permitting discontinuation of prophylactic therapy for opportunistic infections (OIs).

Human Immunodeficiency Virus Reservoirs

In individuals receiving suppressive ART, HIV can establish a state of latent infection in resting memory CD4 + T cells during development of immunologic memory responses. The size and distribution of the viral reservoir may be affected by chronic immune activation, inflammation, and immune dysfunction even in patients receiving potent ART. Although early initiation of ART in HIV-infected individuals can significantly reduce the size of the replication-competent resting CD4 + T-cell latent HIV reservoir, a stable pool of long-lived latently infected CD4 + T cells persists, representing a formidable challenge to HIV cure. The CD4 + T-cell latent HIV reservoir is established in vertically infected infants despite initiation of ART by 8 weeks of age. However, the size of the replication-competent resting CD4 + T cell in infants is associated with time to first undetectable viral load, suggesting that starting ART very early in infants may prevent establishment of a long-lived latent HIV reservoir. The recent finding of detectable viral load (relapse) after an earlier report of a “functional” cure in a perinatally infected infant who received cART within hours of birth is disappointing and warrants further studies.

Diagnosis

Early Infant Diagnosis

Many advances have been made in the area of laboratory diagnosis of HIV infection. Routine HIV antibody testing (e.g., enzyme immunoassays, Western blot) cannot be used in infants for the diagnosis of HIV infection because of transplacental passage of maternal IgG antibodies to the virus that are present in infants up to 18 months of age. The diagnosis of HIV infection in infants warrants the use of PCR-based DNA or RNA assays (referred to as HIV nucleic acid amplification tests [NAATs]) that are highly sensitive and specific and now widely available in developed countries.

The HIV DNA PCR assay detects cell-associated proviral DNA and is the preferred test for early infant HIV diagnosis in the United States; approximately 30% to 40% of HIV-infected neonates will test positive by HIV DNA PCR assay by 48 hours of life, whereas 93% of infected infants will have a positive HIV DNA PCR assay result by 2 weeks of age. By 1 month of age, the sensitivity and specificity of HIV DNA PCR assays for detection of HIV subtype B are 90% to 100% and 95% to 100%, respectively. However, the HIV DNA PCR assay is less sensitive for identifying non–B subtype virus and has been associated with false-negative tests in patients with non–B subtype HIV infection.

HIV RNA PCR assay detects plasma viral RNA and can also be used for early infant diagnosis. The newer HIV RNA assay is as sensitive or more sensitive and as specific for detection of HIV subtype B compared with HIV DNA PCR assay. False-positive HIV RNA results can occur in HIV-exposed neonates, warranting the need to repeat any positive test for confirmation. In addition, a false-negative result can occur in neonates receiving ARV prophylaxis. In contrast, HIV DNA PCR assay can detect proviral DNA in peripheral blood mononuclear cells and will be positive in HIV-infected patients on ART with undetectable viral loads. The choice of NAATs also depends on the HIV subtype because of the genetic variability of HIV globally. HIV RNA PCR may be more sensitive than HIV DNA PCR test for detection of non–B subtype virus, such as subtype C, which is highly prevalent in sub-Saharan Africa. Therefore it is prudent to use HIV RNA PCR for diagnosis of infants born to women known or suspected to have non–B subtype HIV infection. In many studies, the sensitivity of HIV-1 RNA PCR is not affected by the presence of maternal or infant ZDV or nevirapine (NVP) prophylaxis.

HIV DNA or RNA PCR testing is recommended by the U.S. Public Health Service (PHS) at 14 to 21 days of age, and if test results are negative, repeat testing at 1 to 2 months of age and again at 4 to 6 months of age. Virologic testing is recommended at birth by some experts to diagnose in utero infection if mothers did not receive ART or prophylaxis during pregnancy or in other high-risk scenarios, but cord blood specimen should not be used because of possible contamination with maternal blood. It is assumed that children who have a positive HIV PCR result within the first 48 hours after birth were infected in utero, whereas those who are infected during the intrapartum period might become positive 2 to 6 weeks after birth. An infant is diagnosed with HIV infection if two separate blood samples test positive for HIV DNA or RNA PCR. If infection is confirmed, the infant should be promptly referred to a pediatric HIV specialist for consideration of cART and care to prevent rapid disease progression noted in some vertically infected infants.

HIV infection can be presumptively excluded in nonbreastfeeding HIV-exposed children younger than 18 months if (1) two negative HIV DNA or RNA PCR tests result from separate specimens, both of which were obtained at or after 2 weeks of age and one of which was obtained at or after 4 weeks of age; or (2) one negative HIV RNA or DNA PCR test results from a specimen obtained at or after 8 weeks of age; or (3) one negative HIV antibody test is obtained at or after 6 months of age; and no other laboratory (e.g., no subsequent positive PCR test results, if performed) or clinical (e.g., no AIDS-defining illness) evidence of HIV infection is obtained.

HIV infection can be definitively excluded in nonbreastfeeding HIV-exposed children younger than 18 months if (1) two negative HIV DNA or RNA PCR tests result from separate specimens, both of which were obtained at or after 1 month of age and one of which was obtained at or after 4 months of age; (2) two negative HIV antibody tests from separate specimens, both of which were obtained at or after 6 months of age; and no other laboratory (e.g., no subsequent positive PCR test results, if performed) or clinical (e.g., no AIDS-defining illness) evidence of HIV infection is obtained.

In children with two negative HIV DNA PCR test results, many physicians confirm the absence of HIV infection by documenting a negative HIV antibody test result at 12 to 18 months of age (“seroreversion”). A nonbreastfed infant is considered HIV negative if two antibody test samples drawn at least 1 month apart, and both obtained after 6 months of age, are negative. If HIV antibody testing is performed at 12 months of age in an HIV-exposed infant not known to be infected, and if the infant is still antibody positive, repeat testing at 18 months of age is recommended. Detection of HIV antibody in a child 18 months of age or older is diagnostic of HIV infection. Documentation of seroreversion may be more important when non–subtype B HIV is possible or present. In breastfed infants, definite exclusion of HIV infection is based on negative diagnostic test obtained at greater than 6 weeks after cessation of breastfeeding. Table 22-1 provides a diagram outlining the initial evaluation and clinical care of the HIV-exposed infant, as recommended by the American Academy of Pediatrics (AAP).

Table 22-1
Evaluation and Management of Infants Exposed to Human Immunodeficiency Virus (HIV)
Infant Age
Test Birth 2 wk 3 wk 4 wk 6 wk 2 mo 4 mo 6 mo 9 mo 12 mo 15 mo 18 mo
History, physical examination (including weight, height, head circumference) + + + + + + + + + + + +
Assess risk of other infections +
Antiretroviral prophylaxis + +
Complete blood cell count + + +
HIV DNA or RNA PCR § + + + +
PCP prophylaxis (with TMP-SMX) + щ
HIV antibody testing + #
Immunizations HIV-exposed infants should receive all routine childhood immunizations, including hepatitis B vaccine, inactivated polio vaccine, diphtheria–tetanus toxoids–acellular pertussis vaccine, Haemophilus influenzae type b vaccine, pneumococcal conjugate vaccine, and rotavirus vaccine. If infant HIV infection is confirmed, then recommendations for the HIV-infected must be followed. Readers are referred to the 2012 edition of the American Academy of Pediatrics’ Red Book for guidance regarding immunization practices in HIV-infected children.
ARV, Antiretroviral; cART, combination antiretroviral therapy ; PCP, Pneumocystis jirovecii (formerly carinii ) pneumonia; PCR, polymerase chain reaction; TMP-SMX, trimethoprim-sulfamethoxazole; ZDV, zidovudine.

Review maternal history for possible exposure to coinfections (e.g., tuberculosis, syphilis, herpes simplex virus, cytomegalovirus, or hepatitis B virus).

ZDV to prevent perinatal transmission should be started soon after birth but certainly within 12 hr. ZDV is discontinued at 6 wk of age; combination ARV prophylaxis is recommended for infants whose mothers have not received antepartum ARVs; cART should be started in a child who is proved to be infected according to pediatric treatment guidelines, with close laboratory monitoring.

Complete blood cell count is measured at baseline before starting ZDV, at 4 wk, and rechecked at 8 wk if severe anemia was noted at the 4-wk visit.

§ Repeat PCR immediately if positive to confirm infection. If the initial test at 2-3 wk of age is negative, repeat test at 4 wk-2 mo and at 4-6 mo of age to identify or exclude HIV-1 infection as early as possible.

щ PCP prophylaxis can be avoided if or discontinued if HIV infection is presumptively or definitely excluded on the basis of negative HIV virologic tests at 2 and 4-6 wk; if infant is diagnosed with HIV, PCP prophylaxis should be continued for 1 yr.

Some experts recommend obtaining an HIV PCR test at birth to exclude intrauterine HIV infection if the mother did not receive cART during pregnancy or in other high-risk situations.

# HIV-1 antibody testing may be performed to confirm loss of passively transferred maternal antibody in infants already determined to be definitely HIV uninfected on the basis of serial negative virologic tests.

HIV isolation by culture is not recommended for routine diagnosis because culture is less sensitive, more expensive, needs a specialized laboratory, and results are not available for up to 28 days. Use of HIV-1 p24 antigen detection is not recommended for diagnosis of infant HIV-1 because of its poor sensitivity compared with HIV DNA PCR or culture.

Nonspecific laboratory parameters may also suggest HIV infection. Hypergammaglobulinemia, a nonspecific but early finding of HIV infection, is noted in up to 90% of perinatally infected infants by 6 months of age. CD4 counts must be interpreted within the bounds of the age-dependent normal range, and changes in counts may result in a decrease in the normal CD4 to CD8 T-lymphocyte count ratio of greater than or equal to 1.0 in vertically infected infants by 1 to 2 months of age, compared with HIV-exposed, uninfected infants.

Classification of Human Immunodeficiency Virus Infection in Children

Human immunodeficiency virus infection in children and adolescents causes a wide range of clinical manifestations, from asymptomatic infection to AIDS resulting from progressive immunodeficiency. The CDC has classified HIV infection in children to determine the severity of disease. The AIDS case definitions developed by the CDC in 1987, and subsequently revised in 1993, 1994, and 2008, are intended primarily for public health surveillance and reporting purposes for monitoring the HIV epidemic.

The revised 1994 CDC classification system for HIV infection in children younger than 13 years is based on (1) HIV infection status, (2) clinical disease, and (3) immunologic status ( Table 22-2 ). Clinical categories are stratified from N, indicating no signs or symptoms, through A, B, and C, for mild, moderate, and severe (AIDS-defining) symptoms, respectively. The revised 2008 pediatric AIDS case definitions did not make any changes in the HIV infection classification system, the 24 AIDS-defining conditions for children younger than 13 years, or the AIDS case definition for children younger than 18 months. Diagnosis is established if an AIDS-defining disease occurs ( Table 22-3 ).

Table 22-2
1994 Centers for Disease Control and Prevention Revised Classification System for Human Immunodeficiency Virus Infection in Children Younger Than 13 Years
From Centers for Disease Control and Prevention: Recommendations of the U.S. Public Health Service Task Force on the use of zidovudine to reduce perinatal transmission of human immunodeficiency virus, MMWR Morb Mortal Wkly Rep 43:1-20, 1994.
Using this system, children are classified according to three parameters: infection status, clinical status, and immunologic status. The categories are mutually exclusive. When classified in a more severe category, a child is not reclassified in a less severe category, even if the clinical or immunologic status improves.
Pediatric HIV Classification
Clinical Categories
Immune Categories No Symptoms (N) Mild Symptoms (A) Moderate Symptoms (B) Severe Symptoms (C)
No suppression (1) N1 A1 B1 C1
Moderate suppression (2) N2 A2 B2 C2
Severe suppression (3) N3 A3 B3 C3
Immunologic Categories Based on Age-Specific CD4 + T-Lymphocyte Counts and Percentage of Total Lymphocytes
The immunologic category classification is based on age-specific CD4 + T-lymphocyte count or percentage of total lymphocytes and is designed to determine severity of immunosuppression attributable to HIV for age. If either CD4 + count or percent results in classification into a different category, the child should be classified into the more severe category. A value should be confirmed before the child is reclassified into a less severe category.
Age Groups
Immunologic Category 0-11 mo 1-5 yr >6 yr
No suppression (1) >1500 cells/μL (>25%) >1000 cells/μL (>25%) >500 cells/μL (>25%)
Moderate suppression (2) 750-1499 cells/μL (15%-24%) 500-999 cells/μL (15%-24%) 200-499 cells/μL (15%-24%)
Severe suppression (3) <750 cells/μL (<15%) <500 cells/μL (<15%) <200 cells/μL (<15%)
Clinical Categories for Children with HIV Infection
Category N: Not Symptomatic
Children who have no signs or symptoms considered to be the result of HIV infection or who have only one of the conditions listed in category A
Category A: Mildly Symptomatic
Children with two or more of the following conditions but none of the conditions listed in categories B and C:
  • Lymphadenopathy (>0.5 cm at more than two sites; bilateral = one site)

  • Hepatomegaly

  • Splenomegaly

  • Dermatitis

  • Parotitis

  • Recurrent or persistent respiratory infection, sinusitis, or otitis media

Category B: Moderately Symptomatic
Children who have symptomatic conditions other than those listed for category A or C that are attributed to HIV infection
Examples of conditions in clinical category B include but are not limited to:
  • Anemia (<8 g/dL), neutropenia (<1000/mm 3 ), or thrombocytopenia (<100,000/mm 3 ) persisting >30 days

  • Bacterial meningitis, pneumonia, or sepsis (single episode)

  • Candidiasis, oropharyngeal thrush, persisting for >2 mo in children >6 mo of age

  • Cardiomyopathy

  • Cytomegalovirus infection, with onset before 1 mo of age

  • Diarrhea, recurrent or chronic

  • Hepatitis

  • Herpes simplex virus stomatitis, recurrent (more than two episodes within 1 yr)

  • Herpes simplex virus bronchitis, pneumonitis, or esophagitis with onset before 1 mo of age

  • Herpes zoster (shingles) involving at least two distinct episodes or more than one dermatome

  • Leiomyosarcoma

  • Lymphoid interstitial pneumonia or pulmonary lymphoid hyperplasia complex

  • Nephropathy

  • Nocardiosis

  • Persistent fever (lasting >1 mo)

  • Toxoplasmosis, onset before 1 mo of age

  • Varicella, disseminated (complicated chickenpox)

Category C: Severely Symptomatic
Children who have any condition listed in the 1987 surveillance case definition for AIDS, with the exception of lymphoid interstitial pneumonia
Serious bacterial infections, multiple or recurrent (i.e., any combination of at least two culture-confirmed infections within a 2-yr period of the following types: septicemia, pneumonia, meningitis, bone or joint infection, or abscess of an internal body organ or body cavity [excluding otitis media], superficial skin or mucosal abscesses, and indwelling catheter-related infections)
Candidiasis, esophageal or pulmonary (bronchi, trachea, lungs)
Coccidioidomycosis, disseminated (at site other than or in addition to lungs or cervical or hilar nodes)
Cryptosporidiosis or isosporidiosis with diarrhea persisting >1 mo
Cytomegalovirus disease with onset of symptoms at age >1 mo (other than liver, spleen, or lymph nodes)
Encephalopathy (at least one of the following progressive findings present for at least 2 mo in the absence of a concurrent illness, other than HIV infection, that could explain the findings): (1) failure to attain or loss of developmental milestones or loss of intellectual ability verified by standard developmental scale or neuropsychological tests; (2) impaired brain growth or acquired microcephaly shown by head circumference measurements or brain atrophy shown by CT or MRI (serial imaging is required for children <2 yr of age); (3) acquired symmetrical motor deficit manifested by two or more of the following: paresis, pathologic reflexes, ataxia, or gait disturbance
  • Herpes simplex virus infection causing a mucocutaneous ulcer that persists for more than 1 mo or bronchitis, pneumonitis, or esophagitis for any duration affecting a child >1 mo of age

  • Histoplasmosis, disseminated (other than or in addition to lungs or cervical lymph nodes)

  • Kaposi sarcoma

  • Lymphoma, primary, in brain

  • Lymphoma, small, noncleaved cell (Burkitt), or immunoblastic or large cell lymphoma of B cell or unknown immunologic phenotype

  • Mycobacterium tuberculosis, disseminated or extrapulmonary

  • Mycobacterium, other species or unidentified species, disseminated (other than or in addition to lungs, skin, or cervical or hilar lymph nodes)

  • Mycobacterium avium-intracellulare complex or Mycobacterium kansasii, disseminated (other than or in addition to lungs, skin, or cervical or hilar lymph nodes)

  • Pneumocystis jirovecii (formerly carinii ) pneumonia

  • Progressive multifocal leukoencephalopathy

  • Salmonella (nontyphoid) septicemia, recurrent

  • Toxoplasmosis of the brain with onset >1 mo of age

  • Wasting syndrome in the absence of a concurrent illness, other than HIV infection, that could explain the following findings: persistent weight loss >10% of baseline or downward crossing of at least two of the following percentiles on weight-for-height chart on two consecutive measurements >30 days apart plus chronic diarrhea (i.e., at least two loose stools per day for >30 days) or documented fever (for >30 days, intermittent or constant)

AIDS, Acquired immunodeficiency syndrome; HIV, human immunodeficiency virus.

Children whose HIV infection status is not confirmed are classified by using the grid with a letter E (for vertically exposed) placed before the appropriate classification code (e.g., EN2 ).

Category C and lymphoid interstitial pneumonitis in category B are reportable to state and local health departments as AIDS.

Table 22-3
AIDS Indicator Diseases Diagnosed in 8086 Children Younger Than 13 Years Reported to the Centers for Disease Control and Prevention Through 1997
From Centers for Disease Control and Prevention: U.S. HIV and AIDS cases reported through December 1997. HIV/AIDS Surveillance report: year-end edition, MMWR Morb Mortal Wkly Rep 9:1-44, 1997.
Disease No. Children Diagnosed Percent of Total
Pneumocystis jirovecii (formerly carinii ) pneumonia 2700 33
Lymphocytic interstitial pneumonitis 1942 24
Recurrent bacterial infections 1619 20
Wasting syndrome 1419 18
Encephalopathy 1322 16
Candida esophagitis 1266 16
Cytomegalovirus disease 658 8
Mycobacterium avium infection 639 8
Severe herpes simplex infection 370 5
Pulmonary candidiasis 307 4
Cryptosporidiosis 291 4
Cancer 162 2
AIDS, Acquired immunodeficiency syndrome.

The sum of percentages is greater than 100 because some patients have more than one disease.

In the pre–highly active antiretroviral treatment (HAART)-era, Pneumocystis jirovecii (formerly carinii ) pneumonia (PCP) was the leading AIDS-defining illness diagnosed during the first year of life and associated with a high mortality rate. Other common AIDS-defining conditions in U.S. children with vertically acquired infection include lymphoid interstitial pneumonitis (LIP), multiple or recurrent serious bacterial infections, HIV encephalopathy, wasting syndrome, Candida esophagitis, cytomegalovirus (CMV) disease, and Mycobacterium avium-intracellulare complex (MAC) infection (see Table 22-3 ). In the post-HAART period, a significant decrease in the number of OIs was noted in HIV-infected children. Likewise, significant decline in mortality was noted between 1994 and 2000, from 7.2 to 0.8 deaths/100 person-years.

The case definitions are similar for adults and children, with some important exceptions. LIP and multiple or recurrent serious bacterial infections are AIDS-defining illness only for children. Also, certain herpes virus infections (cytomegalovirus, herpes simplex virus) and toxoplasmosis of the central nervous system (CNS) are AIDS-defining conditions only for adults and children older than 1 month.

The immunologic categories place emphasis on the CD4 T-cell lymphocyte count and percentages for age and include stage 1, no evidence of immunosuppression; stage 2, moderate immunosuppression; and stage 3, indicating severe immunosuppression. Once classified, a child cannot be reclassified into a less severe category, even if the child’s clinical status or immune function improves in response to ART or resolution of clinical events. HIV-exposed infants whose HIV infection status is indeterminate (unconfirmed) are classified by placing a prefix E (for perinatally exposed) before the appropriate classification code (e.g., EN2).

HIV infection in infants and children has a different presentation from that in adults. Children are more likely than adults to have serious bacterial infections, and LIP is almost entirely restricted to the pediatric age group. OIs such as PCP often manifest as primary diseases with a more aggressive course because of lack of prior immunity. Toxoplasmosis, cryptococcal infection, and the occurrence of cancer, especially Kaposi sarcoma, are less common in HIV-infected children.

Clinical Manifestations and Pathology

The early clinical manifestations of HIV infection in infants are highly variable and often nonspecific. Infants with perinatally acquired HIV infection are often asymptomatic, and physical examination is usually normal in the neonatal period. Although a distinctive craniofacial dysmorphism characterized by microcephaly, hypertelorism, flattened nasal bridge, triangular philtrum, and patulous lips was suggested as a possible congenital HIV syndrome, these findings reported during the early AIDS epidemic have not been confirmed in subsequent reports. In a prospective cohort study of 200 perinatally acquired HIV-1 infections, the median age of onset of any HIV-related symptom or sign was 5.2 months; the probability of remaining asymptomatic was 19% at 1 year and 6.1% at 5 years. In another large prospective cohort study, AIDS-defining conditions developed in approximately 23% and 40% of perinatally infected infants by 1 and 4 years.

Growth delay is an early and frequent finding of untreated perinatal HIV infection, and linear growth is most severely affected in children with high viral loads. Other features of infection in early infancy could include unexplained persistent or recurrent fevers, generalized lymphadenopathy, often associated with hepatosplenomegaly and recurrent or persistent otitis media. Also commonly encountered are oral or diaper candidiasis, developmental delay, parotitis, and nonspecific dermatitis.

Before the widespread use of pediatric ART and use of prophylaxis, PCP was a common AIDS-defining illness in infancy, accounting for approximately 50% of all AIDS-defining conditions. Other clinical manifestations noted during the course of HIV infection in infants and children who remain undiagnosed or receive ineffective ART include encephalopathy, recurrent and chronic otitis media and sinusitis, recurrent invasive bacterial infections, other OIs, chronic diarrhea, cardiomyopathy, and nephropathy.

Infectious Complications

Infections in HIV-infected infants not receiving combination ART can be serious or life threatening. The difficulty in treating these infectious episodes, their chronicity, and their tendency to recur distinguish them from the normal infections of early infancy. It is helpful to document each episode and to evaluate the course and frequency of their recurrences. In the pre-HAART era, the frequency of OIs varied by age, immune status, prior history of OI, and pathogen. With early infant HIV diagnosis and linkage to care and an ART program, the frequency of historically reported AIDS-defining illness and OIs has dramatically decreased among children living with HIV in the United States and other resource-rich countries. During the pre-ART era, the most common OIs (event rate, > 1.0/100 child-years) reported among U.S. children with vertically acquired infection included serious bacterial infections (e.g., pneumonia, bacteremia), PCP, disseminated MAC, herpes zoster, esophageal and tracheobronchial candidiasis, and CMV disease. Other rare OIs (event rate, <1.0/100 child-years) included CMV disease, cryptosporidiosis, tuberculosis (TB), systemic fungal infections, and toxoplasmosis. In the post-ART era, the rate of OI decreased from 12.5 to 0.8 cases per 100,000 person-years pre-ART and post-ART, respectively. Despite these impressive declines, HIV-infected children continue to experience the same types of OIs, even in the era of combination ART, highlighting the need for early recognition, treatment, and prevention.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here