You are on page 1of 13

Progress

in

Pediatric
Cardiology
ELSEVIER Progress in Pediatric Cardiology 7 (1997) 19-31

Pathogenesis of HIV infection in children

Grace M. Aldrovandi*
Universi@ ofAlabama at Birmingham 256 Bevill Biomedical Research Building 845 19th Street South,
Birmingham, AL 35294-2170, UK

Abstract

The increasing numbers of children infected with human immunodeficiency virus (HIV), the etiologic agent of the Acquired
Immunodeficiency Syndrome (AIDS), affects widely diverse socially and geographical populations. This review is intended to
highlight key aspects of HIV pathogenesis especially as it relates to children. Topics include aspects of the virus itself, the cells
it infects, the in vivo dynamics of infection and the host immune response to the virus. Discussion of the problems of diagnosis
and management of children with HIV-l infection will also be briefly considered. 0 1997 Elsevier Science Ireland Ltd.

Keywords: Human immunodeficiency virus; Acquired immunodeficiency syndrome; Pediatrics; Pathogenesis; Natural history

1. Introduction are only 40-50% identical at the nucleotide level


however, some serological reagents cross react
[lo-141. This paper will focus on HIV-l infection
In recent years, there has been an evolving under-
since it is by far the more prevalent and more
standing of AIDS as a dynamic viral infection. An
thoroughly characterized.
appreciation of the complex nature of this infection is
essential to comprehending the challenges involved in
the treatment of HIV-l infected persons. This review 2. Structure and life cycle of HIV
will focus on our current understanding of the dy-
namic nature of the mechanisms involved in the es-
tablishment and progression of this disease. Consider- HIV is a member of the retrovirus family. Retro-
ation will also be given to the unique problems in- viruses are a distinct subset of RNA viruses in which
volved in the diagnosis and management of children the viral RNA genome is transcribed to DNA by an
infected with HIV. RNA dependent DNA polymerase (reverse transcrip-
There are two related but distinct subtypes of HIV: tase) in the host cell. This DNA is then integrated
types 1 and 2. Both of these viruses are associated into the infected cell’s chromosomal DNA as a
with AIDS, however, there is some recent evidence provirus, and subsequently behaves like a cellular
that HIV-2 is less pathogenic [l] and less likely to be gene. Thus, under appropriate conditions the proviral
transmitted [2-81. Although sporadic cases have been genes are expressed and the progeny virus made.
reported in the US [9] and elsewhere, HIV-2 is cur- Structurally HIV has a cylindrical eccentric core or
rently only endemic in West Africa. HIV-l and HIV-2 nucleoid containing the diploid RNA surrounded by
reverse transcriptase and nucleocapsid (NC) proteins
(~9, p6) (see Fig. 1). These nucleoid components are
*Tel.: + 44 205 9342456; fax: +44 205 9341640; email: enclosed by the capsid antigen p24 forming the nucle-
gracea@uab.edu ocapsid. The matrix antigen p17 surrounds the nucleo-

1058-9813/97/$17.00 0 1997 Elsevier Science Ireland Ltd. All rights reserved.


PZZSlOSS-9813(97)00198-7
20 GM Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31

capsid and lines the inner surface of the envelope of chemokine receptor family serve as co-receptors for
the virus. The surface of the virion contains 72 knobs HIV-l infection [17-221.. Chemokines are proteins
of the envelope glycoprotein gp120. The gp120 is involved in inflammatory responses and have been
non-covalently anchored to the viral envelope by the found in vitro to suppress HIV replication 1231. Some
transmembrane protein gp41. This weak interaction individuals who have been able to escape infection
allows the gp120 to be easily stripped off the virion despite extremely high risk multiple exposures to HIV
and may in part account for the relatively poor infec- have defects in these receptors 124-261.
tivity of this virus. The viral envelope is a lipid bilayer, After entry into the cell, HIV-l undergoes reverse
derived from the host cell plasma membrane and transcription and then integration into the host
consequently contains various host cell antigens. genome. Initiation of RNA transcription of the provi-
HIV can infect T-lymphocytes, monocytes/macro- ral DNA is due to cellular factors and appears to
phages, dendritic cells and microglia in the central depend on the overall state of activation of the cell.
nervous system. Infection begins with binding of the Gene expression is regulated by various virus-specific
gp120 on the viral envelope to a cellular receptor. proteins and can be influenced by cytokines and co-
The CD4 molecule is the principle receptor, however, infection with other viruses. Translation of singly and
there is some evidence that HIV can enter cells via multiply spliced mRNAs results in the production of
the complement receptor, Fc or the Gal/C receptor structural, replicative, regulatory and other proteins,
[15,16]. This attachment is believed to result in con- which co-assemble at the inner surface of the cell
formational changes exposing the gp41 transmem- membrane. The proteins insert themselves into the
brane component permitting virus-cell membrane fu- cell membrane where the virion is assembled and
sion and subsequent internalization of the virion. Re- then released.
cently, it has been demonstrated that’members of the The viremia in HIV-l infection is sustained by

Legend
1 ss RNA
IQ p66RTiRNAase H

l P6
a Int egrase Host proteins

gP 120
gP 41

P24

Fig. 1. Schematic representation of the morphologic structure of HIV-l.


G.M. Akirovandi /Progress in Pediatric Cardiology 7 (1997) 19-31 21

rapid, high level viral replication, requiring continu- in persons with lower baseline CD4+. This suggests
ous reinfection and destruction of CD4+ T cells. that the decline in CD4+ cell number that precedes
Recent elegant mathematical modeling studies have the ultimate immune system collapse may largely re-
provided insight into the kinetics of viral replication flect increased HIV replication and not immune ex-
which are summarized in Fig. 2. Analysis of adults haustion [28].
with moderate to advanced disease have revealed a Clearance rates do not appear to vary substantially
very dynamic process of viral production and clear- between persons with different CD4+ counts or viral
ance [27-291. On average greater than 10n virions load, suggesting that viral clearance may not be de-
per day are released into the extracellular space; the pendent on specific host immune responses. Thus, the
half-life of these vii-ions is only approximately 6 h [29]. extent of the viremia depends on virus production.
The average life span of a productively infected cell is The factor(s) that mediate this clearance (e.g. inher-
about 2 days [27,28]. The time from release of a virion ent instability of virions, the reticuloendothelial sys-
from one cell until it infects another cell and results tem, etc.) are as yet unidentified. Thus, HIV infection
in the release of a new generation of k-ions is re- involves a dynamic, continuous process of de-novo
ferred to as the generation time. For HIV the genera- infection of target cells that produce the virus for a
tion time is approximately 2.6 days [29]. It is estimated very short time before being consumed in the process
that about 5% of the total body CD4* cell population of viral replication. Studies of viral and cellular dy-
are being destroyed daily [27,28]. Thus the immune namics in children have yet to be performed. There
system must constantly be replenishing its losses. In- are several important differences between young chil-
terestingly, the rates of T cell recovery in persons dren and adults which may have a profound effect on
treated with potent anti-retrovirals, tend to be higher viral and cellular dynamics. Among these are: much

ELLS INFECTED
WITH DEFECTIVE VIRUS
LYMPHOCYTES
ONG -LIVED POPULATIONS

< 1% OF VIRUS PRODUCTION

PRODUCTIVELY INFECTED ACTIVATED


LYMPHOCYTES UNINFECTED
AVERAGE t ,,2 1.6 days LYMPHOCYTES

Fig. 2. Schematic representation of the dynamics of HIV-l infection in vivo.


22 GM Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31

Table 1
Conditions associated with categories of disease

Category A Category B Category C

Have two or more of the Examples of conditions in clinical Multiple or recurrent serious bacterial infections
conditions listed below but category B (septicemia, pneumonia, meningitis, bone or joint
no categories B and C infection, or abscess of an internal organ)
conditions Anemia ( < 8 g/dl), neutropenia Candidiasis, esophageal or pulmonary
( < 1000/mm3), or thrombocytopenia Coccidiomycosis, disseminated
Lymphadenopathy (> 0.5 cm ( < 1000 O00/mm3) persisting > 30 days Cryptococcosis, extrapuhnonary
at more than two sites) Bacterial meningitis, pneumonia or Cryptosporidiosis or isosporiasis with diarrhea
Hepatomegaly sepsis (single episode) persisting for > 1 month
Splenomegaly Candidiasis, oropharyngeal (thrush), Cytomegalovirus disease with onset of symptoms prior
Dermatitis persistent (> 2 month) in a child > 6 month to 1 month of age
Parotitis Cardiomyopathy Cytomegalovirus retinitis (with loss of vision)
Recurrent or persistent Cytomegalovirus infection, with onset Encephalopathy in the absence of a concurrent illness
upper respiratory infection, > 1 month of age other than HIV infection that could explain the findings
sinusitis, or otitis media Diarrhea, recurrent or chronic, HSV infection causing a mucocutaneous ulcer
Hepatitis, persisting for > 1 month; or as the etiology of
HSV stomatitis, recurrent( > 2 episodes bronchitis, pneumonitis or esophagitis
within 1 year) Histoplasmosis, disseminated
HSV bronchitis, pneumonitis, or Kaposi’s sarcoma
esophagitis within < 1 month of age Primary CNS lymphoma
Herpes Zoster involving > 2 distinct Lymphoma, Burkitt’s, large cell, or immunoblastic
episodes or > 1 dermatome M. Tuberculosis, disseminated or extrapulmonary
Leiomyosarcoma Mycobacterium infections other than tuberculosis,
LIP or pulmonary Iymphoid disseminated
hyperplasia PCP
Nephropathy Progressive multifocal leukoencephalopathy
Nocardiosis Toxoplasmosis of the brain with onset at > 1 month of age
Persistent fever (lasting > 1 month) Wasting syndrome in the absence of a concurrent
Toxoplasmosis, onset < 1 month of age illness other than HIV infection
Varicella, disseminated

higher CD4+ cell counts (see Table 11, infection replication rate also means that even subtle selection
during the immunological development, an immature pressures will have dramatic effects on the population
immune system, and greater immune activation. of viruses. This also explains the virtually inevitable
and rapid emergence (within a few weeks) of drug
3. Genetic variation resistance. It also accounts for the ability of this virus
to escape what appears to be a very strong immune
The genetic homogeneity of initial infection with
response and eventually leads to the death of the
HIV-l rapidly gives way to tremendous diversity
infected individual. Why certain individuals are able
[30-321. As a result of this rapid genetic drift, HIV
to avoid and perhaps even escape this fate is an area
infection in an individual is not due to a homoge-
of considerable interest (see references [34-371).
neous virus but a population of viruses - a quasi-
species or a distribution of mutants within which each 4. Transmission of HIV
daughter virion differs from that of its parent. Diver-
sity is present not only between individuals but within HIV is not an easily transmitted virus; direct con-
an individual at any given timepoint. Moreover, this tact with infected bodily fluids is necessary. The risk
heterogeneity is manifested at the molecular, biologic of transmission after percutaneous injury is much less
and serologic levels. These quasispecies differ not with HIV than Hepatitis B (0.2-0.5% vs. 2-40%)
only in their genetic structure but their replication [38,39]. The administration of zidovudine shortly after
kinetics, level of virus production, cytopathicity, cellu- exposure to HIV can decrease this risk by almost 80%
lar tropism and their sensitivity to immune responses [38]. Most children acquire HIV perinatally, but even
(see reference [33] for an excellent discussion); in no by this route, only about 12-30% of children born of
small measure this accounts for HIV-l’s remarkable HIV-infected women are themselves infected [40-431.
adaptability. This diversity has in large part been By administering zidovudine to the pregnant woman
attributed to the intrinsic infidelity of reverse tran- this transmission rate can be decreased by two thirds
scription. It has been estimated that every possible [44]. This ability to decrease transmission makes the
single point mutation would occur between lo4 and case for prenatal testing even more compelling [45,46].
10’ times per day in an infected person 1331. High There is evidence that transmission can occur in utero
GM Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31 23

[47-491, intrapartum 1491 and post-partum via breast interventions such as virucidal vaginal lavage during
feeding [50-521. The risk of transmission via breast labor, active and/or passive immunization are also
milk appears to increase if the woman acquires the being studied.
infection while lactating (approximately 26%), rather
than if she is infected prior to pregnancy (8-18%) 5. Factors associated with perinatal transmission
[50]. In most of the developed nations HIV positive
women are counseled to avoid breast feeding how- There have been numerous attempts to discern
ever, in less developed countries where malnutrition factor(s) which increase the risk of vertical HIV tran-
and other infectious diseases are a major cause of smission. Maternal, obstetrical, virologic and fetal fac-
infant mortality the risks of HIV transmission are tors have all been proposed as variables in determin-
outweighed by its benefits. ing perinatal transmission (see references [75-781 for
The proportion of transmission events that occur at an excellent reviews). Discordant twins and the obser-
these different times is still uncertain, A working vation that women could have one infected child and
definition of the timing of infection has been pro- then subsequently deliver an uninfected child support
posed in non-breastfeeding population [53]. In utero a multifactorial mechanism. Several studies have sug-
infection is believed to have occurred if HIV is cul- gested an association with maternal factors and an
tured from peripheral blood or detected by PCR increased rate of HIV transmission from mother to
within 48 h of birth. Infants are regarded as being child. Among these factors are: high virus load [79,80];
infected intrapartum if peripheral blood samples ob- low CD4+ cell count [42,81]; and advanced clinical
tained in the first week of life are negative for HIV disease state [66,67]. Other factors such as p24 anti-
culture, DNA PCR or both, and subsequently posi- genemia [42,82-841 viral phenotype 1841 and de-
tive. Using this definition it is estimated more than creased maternal neutralizing antibody to autologous
50% of children are infected intrapartum [47,48,54,55]. virus are controversial [85-901. As discussed above,
Preliminary studies suggest that the timing of infec- placental breaches and/or events occurring at the
tion has an influence on clinical course (see below). time of delivery such as infant exposure to maternal
HIV has been detected in amniotic fluid and in blood, prolonged duration of ruptured membranes
fetal tissues as early as 8 weeks gestation [56-591. It (> 4 hours) and increased quantity of HIV in mater-
has also been detected in the placental trophoblast nal blood at delivery have also been associated with
particularly in Hoefbauer cells which are of an elevated risk of transmission. There is also some
macrophage lineage [60]; all these cells can be readily evidence that HLA type may influence the susceptibil-
infected with HIV in vitro 161-631. Nevertheless, the ity of an individual to infection [91,92]. Nevertheless,
presence of HIV in the placenta is not directly related none of these factors alone or in combination can
to the risk of transmission to the infant [64,65]. There reliably predict the outcome in an individual case.
is evidence that placental inflammation or damage
increases the risk of transmission [60,66,67]. The 6. Diagnosis of HIV infection
mechanisms by which an HIV infected placenta serves
as an effective barrier against the virus are unknown. In children older than 18 months and in adults the
During birth, the infants’ mucosal surfaces and diagnosis of HIV infection is made by detecting anti-
potentially traumatized skin are exposed to HIV in- bodies to HIV. In adults, antibodies are usually de-
fected blood and cervical secretions. Evidence for tected within 6-12 weeks post-infection. The period
intrapartum transmission comes from studies of twins between infection with the virus and the detection of
born to HIV-infected women. The first born twin has antibodies is known as the ‘window period’. Although
greater than a twofold higher risk (35% vs. 15%) of there are a few case reports of persons infected with
contracting the virus [68]. Presumably the presenting HIV not having antibodies against the virus these
twin has a longer exposure to infected blood and cases are exceedingly rare [931. The determination of
cervical secretions or has an increased incidence of HIV infection in children younger than 18 months is
trauma during labor and delivery. However, this in- complicated by the presence of maternal antibody.
creased risk was observed even in a comparison of The transplacentally acquired maternal IgG against
twins delivered by cesarean section (16% vs. 8%) [68]. HIV is indistinguishable from antibody produced by
Several other reports have agreed that cesarean sec- the infant. Thus, virtually all infants born to HIV-in-
tion can reduce the frequency of transmission [69-741, fected women will have detectable antibodies to HIV.
further supporting the importance of events close to These antibodies are usually lost by 7-9 months of
delivery. Elective cesarean section is not routinely age but can persist for as long as 18 months of age
advocated since it is estimated that 16 infants would [42]. Therefore, the alternative methods discussed
have to be delivered by cesarean section in order to below have been employed for the diagnosis of HIV
prevent one child from being infected [73]. Other infection in young children. Only three methods, i.e.
24 G.M. Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31

virus culture, polymerase chain reaction (PCR) and and after this timepoint, it is of moderate sensitivity
detection of p24 antigen are accepted by the Centers but high specificity [97-1001.
for Disease Control and Prevention (CDC) for the Other tests which have been used are HIV p24
diagnosis of HIV infection in children younger than antigen (Ag) and in vitro antibody production assays.
18 months [94]. Detection of p24 Ag provides definitive proof of infec-
tion. The sensitivity of this test can be enhanced by
immune complex dissociation (ICD) of Ag-Ab com-
7. Serologic tests for HIV diagnosis
plexes in the sera. This easy and relatively inexpensive
test is only moderately sensitive; after 6 months of
The standard test for screening for HIV infection is age the sensitivity is approximately 80-100%
the ELISA (enzyme-linked immunosorbent assay). [101-1041. Since it does not rely on antibodies it is
This tests uses antigens derived from disrupted extra- useful for diagnosis of infants who are hypogamma-
cellular virions that are purified and immobilized on globulinemic. Detection of HIV-specific antibody pro-
beads or wells. Test serum is incubated with these ducing B-lymphocytes has been used for diagnosis of
antigens and then treated to detect a spectrophoto- HIV infection; however, false positives and negatives
metric color change. This test is only considered posi- remain important limitations [105-1081.
tive if it is positive on two or more repetitions. Most
of the kits will detect both HIV-l and HIV-2. There 8. HIV culture and PCR
was a recent report that an unusual serotype found in
Cameroon, Africa was not detected by some manufac-
The ‘gold standard’ for the diagnosis of HIV infec-
turers’ kits in the US, however, in conjunction with
tion in infants is viral culture. Unfortunately, the
the CDC this problem has been rectified [95].
requirement for special laboratory facilities and highly
Once an ELISA is repeatedly positive, a confirma-
trained personnel limit its use outside of research
tory test such as a Western blot must be performed to
settings. In addition, this technique takes 2-4 weeks
validate the results. The Western blot assay also uses
to obtain results and is very expensive. The sensitivity
HIV antigen derived from purified, disrupted whole
of polymerase chain reaction (PCR) is comparable to
virus. In this assay antigens are electrophoretically
viral culture and has the advantage of being extremely
separated into bands, transferred onto test strips of
rapid (l-2 days) [49,54,109-1121. The development of
nitrocellulose paper and then incubated with patient’s
commercial kits and methods to decrease the likeli-
serum. There are several different criteria for what
hood of contamination have made PCR amenable for
constitutes a positive Western blot. The CDC has
use in the clinical laboratory [113]. Both viral culture
recommended that this assay be considered positive
and DNA-PCR are of comparable but limited sensi-
on the identification of two of the following HIV gene
tivity in identifying HIV infected neonates at the time
products ~24, gp41 or gp120/160 [13,96]. If fewer
of birth or shortly thereafter [103,110,114]. Under 1
bands are present the test is considered indetermi-
week of age only 20-30% of infected infants can be
nate; if no bands are present the test is interpreted as
identified by these techniques, however, at 4 weeks of
negative. Individuals with an indeterminate Western
age this increases to approximately 90%. There is
blot should be referred to specialists for careful clini-
excellent correlation between PCR and culture results
cal follow-up, repeat testing or testing by alternative
in the first 7 days of life, suggesting that the limiting
methods. Most laboratories in the US will only issue a
factor may be viral load. This would support the
final report of HIV seropositivity if both the ELISA
prevailing hypothesis that in utero infected infants
and Western blot are positive. This combination of
would have more detectable virus early on compared
currently licensed tests have a sensitivity and specific-
to those infants infected intrapartum.
ity exceeding 99% [13,96]. These tests can rarely be
falsely negative especially if the person is hypogam-
maglobulinemic. 9. Natural history of HIV infection
Since IgM and IgA antibodies do not cross the
placenta, their presence could be used to diagnose Infection of adults with HIV is classically followed
infection of infants. Unfortunately, the sensitivity and by three distinct virologic stages: primary infection;
specificity of these tests, particularly in the first 6 clinical latency; and finally progression to AIDS (see
months of life have been disappointing. Detection of Fig. 3). The time interval between primary infection
HIV-specific IgM has been neither sensitive or speci- and the development of AIDS is variable. For adults
fic presumably because of low levels, transient pro- in developed countries, it is on average lo-11 years
duction and/or non-specific reactions. HIV-IgA can [115], however, about 20% of individuals will progress
be detected in sera, salvia and urine, however, prior in less than 5 years, while a few (< 5%) will remain
to 6 months of age it is neither sensitive or specific immunologically normal for over 10 years [1161. The
GM Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31 2.5

biological basis for this variability is unclear but un- in response to anti-retroviral therapy are predictive of
doubtedly reflects differences in viral strains, host improved prognosis [123].
immune responses and exposure to other cofactors Our understanding of the natural history of pedi-
(microbial or environmental) especially those leading atric HIV infection is still evolving. Viral dynamics
to immune activation. The role of persistent virus and cell turnover studies are in progress. Clinically, it
replication throughout the entire course of infection would appear that there are at least two patterns of
has been established as a fundamental driving force disease progression. About lo-20% of infected chil-
for disease pathogenesis [27-29,117]. During primary dren will develop profound immunosuppression, se-
infection, extremely high levels of plasma viremia vere encephalopathy, organomegaly, and multiple op-
have been observed with levels of plasma RNA greater portunistic infections in the first few months of life
than lo7 copies/ml [ 118-1201. These concentrations [124,125]. Without treatment few of these children
will usually precipitously drop by 2 logs or greater survive more than 2 years. These children are far
after several weeks. In 50-90% of cases this viremia more likely to have had HIV detected during the first
is symptomatic. After a period fluctuation of several 7 days of life [124] and have higher proviral loads
months, the level of plasma RNA stabilizes around a [126]. These rapid progressors are believed to have
so called ‘set point’ usually between lo3 and lo5 been infected in utero. Presumably their fulminant
copies/ml (range < 200 copies to > 106). This level of course is a consequence of uncontrolled viral replica-
plasma viral RNA is a critical CD4 independent pre- tion in the face of a developing immune system.
dictor of rapid progression to AIDS [121]. While there However, all perinatally infected children have a worse
is an inverse correlation between CD4+ cell count prognosis compared to adults (see Fig. 4).
and plasma RNA levels, at any CD4+ cell level there Pediatric HIV infection has been classified by the
is a wide inter-individual variation in plasma RNA CDC as comprising four categories: stage N, no signs
concentration [117,122]. The determinants of this set- or symptoms; stage A, mild signs or symptoms; stage
point are not yet understood but probably include the B, moderate signs or symptoms; stage C, severe signs
effectiveness of host immune responses, the number or symptoms [94]. All the AIDS-defining illnesses in
of available target cells, the degree of immune activa- the 1987 CDC definition of pediatric AIDS are in-
tion and the intrinsic pathogenicity of the infecting cluded in stage C with the exception of LIP (lymphoid
strain. Irrespective of this initial set point, increasing interstitial pneumonitis) which is a B illness (see
concentrations of plasma HIV RNA herald the devel- Table 1). A summary of a recent analysis of the
opment of advancing immunodeficiency and AIDS clinical progression of over 2000 perinatally infected
[122]. Likewise reduced concentrations of HIV-l RNA children is presented in Fig. 4 [127]. Although this

Primary Infection Asympt omat ic Symptomatic AIDS

seroconvet sion
infect ion
c

I Plasma Viremia
CD4 count

HIV antibodies

[fi HIV specific CTL 1 ,A< )

< >
4-6 weeks lo-12 years 2-3 years
Fig. 3. The natural history of HIV infection in adults.
GM Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31

0 A: Mildly symptomatic

I- ~~ B: Moderately symptomatic

C: Severely symptomatic

I I I
I
0 10 14 79(6.6) 113(9.4)
Birth Death
Time in months (years)
Fig. 4. Mean age for clinical stages of perinatal HIV infection.

data has a number of potential biases toward en- cell counts or the percentage of CD4+ lymphocytes
rolling more symptomatic children, it suggests that on (see Table 2). If cell count and percentage place a
average children, progress to moderate symptoms in child in different categories it is recommended that
the second year of life. the more severe category be used.
HIV infected children are also categorized on the Although the number of studies are still small, just
basis of the degree of immunosuppression [127]. In as in adult infection viral load appears to be the
adults, CD4+ cell level has proven to be a reliable critical determinant of pediatric disease progression.
marker of the severity of immunosuppression, as well One recent study [126] found that patterns of plasma
as, determining the risk of developing certain oppor- viral RNA kinetics in the first few months of life
tunistic infections. For example, the risk of developing correlated with disease course. Rapid progressors had
Pneumocystis car&i pneumonia (PCP) increases dra- rapid increase in viral RNA which did not decline
matically at a CD4+ cell counts of 200 or less [128]. In while in more typical progressors the levels of RNA
children the use of CD4+ cell counts is complicated declined [ 1261.
by the fact that they vary with age. In the first year of A small subset of persons infected with HIV ( < 5%)
life CD4+ cell counts are more than three times the are able to maintain normal CD4+ cell counts and
adult number and then gradually decline [129,130]. In low viral loads for periods of greater than 12-15 years
addition, children may develop opportunistic infec- without anti-retroviral therapy. They have been called
tions at higher CD4+ levels than adults [131]. The long-term survivors or long-term non-progressors
current CDC classification system is based on age- [132-1351. In adults, this appellation is usually defined
specific CD4+ counts and either the absolute CD4+ as those who have intact immune systems after more

Table 2
Age related changes in CD4 cell values

Age of child No evidence of immune Evidence of moderate Evidence of severe immune


suppression immune suppression suppression
CD4 no. %CD4 CD4 no. %CD4 CD4 no. %CD4
< 1 year 2 1500 r 25 750-1499 15-24 < 750 < 15
l-5 years 2 1000 225 500-999 15-24 < 500 < 15
6-12 years 2 500 2 25 200-499 15-24 < 200 < 15
GM Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31 27

than 12-15 years. A few of these individuals appear host immune response or develop resistance to anti-
to be infected with less pathogenic variants of the retroviral agents. Effective reduction of viral load
virus [37,136]. However, in most cases non-progres- early on, could also reset the viral set point and
sion is believed to be due to immunogenetic factors thereby improve the subsequent clinical course. Pre-
(see for a review [137]). The definition of a long-term liminary studies in the SIV system support this ap-
pediatric survivor includes those who have survived proach [144-1461. Studies are underway in adults to
longer than 8 years and unlike the adult definition, test this hypothesis, however, perinatal HIV infection
permits inclusion of persons who have received anti- is a near ideal situation in which to study early
retrovirals and are mildly symptomatic [138]. therapeutic intervention. The source and timing of
Pediatric centers that prospectively follow HIV ex- infection can be readily identified and children have a
posed children have described seroreverting infants much greater capacity to regenerate T cells [147].
who had unexplained positive results [42,139], how- Armed with greater understanding of HIV pathogen-
ever, such cases were usually dismissed as laboratory esis and the an arsenal of potent anti-retrovirals,
mistakes. One controversial case report claimed to there is now tremendous optimism that we will be
describe an infant who transiently had positive HIV able to fundamentally alter the course of this fatal
cultures and a positive PCR [140]. disease and even cure it.
Studies of macaques infected with Simian Im-
munodeficiency Virus (SIV) would suggest that tran- Acknowledgements
sient infection is possible. Infection with extremely
low levels of SIV or very attenuated strains can result
in transient infection [141]. How often this occurs in Grace Aldrovandi is a Scholar of the Pediatric Aids
humans and the virologic and immunologic factors Foundation.
that govern this phenomena, have yet to be defined.
References
10. Current management of pediatric HIV infection
[ll Markovitz DM. Infection with the human immunodeficiency
The management of children with HIV involves virus type 2. Ann Intern Med 1993; 118(3):211-218.
treatment of not only the virus but the consequences La Adjorlolo-Johnson G, De Cock KM, Ekpini E et al. Prospec-
tive comparison of mother-to-child transmission of HIV-l
of immune dysregulation and destruction. Regular and HIV-2 in Abidjan, Ivory Coast. Jama 1994;272(6):
monitoring of multiple organ systems, virological and 462-426.
immunological parameters is necessary. The clinical 131 Comparison of vertical human immunodeficiency virus type
manifestations of HIV are protean, potentially af- 2 and human immunodeficiency virus type 1 transmission in
flicting every organ system. Our knowledge of how the French prospective cohort. The HIV Infection in New-
borns French Collaborative Study Group. Pediatr Infect Dis
best to treat infected children is rapidly evolving; J 1994;13(6):502-506.
increasingly new antivirals, prophylactic regimens and [41 De Cock KM, Adjorlolo G, Ekpini E et al. Epidemiology
immunomodulatory agents are being tested and re- and transmission of HIV-2. Why there is no HIV-2 pan-
leased. Given the complex nature of the disease and demic. Jama 1993;270(17):2083-2086.
the rapid advances in treatment, it is therefore not [51 De Cock KM, Zadi F, Adjorlolo G et al. Retrospective study
of maternal HIV-l and HIV-Z infections and child survival
surprising that a recent study of HIV infected adults in Abidjan, Cote d’Ivoire. Bmj 1994;308(6926):441-443.
found a direct correlation between mortality and the k51 Poulsen AG, Kvinesdal BB, Aaby P et al. Lack of evidence
HIV experience of their treating physician [142]. of vertical transmission of human immunodeficiency virus
Hence, the care of these children is best provided by type 2 in a sample of the general population in Bissau. J Acq
a specialized multi-disciplinary team. Immune Defic Syndr 1992;5(1):25-30.
t71 Paulsen AG, Aaby P, Gottschau A et al. HIV-2 infection in
Our greater understanding of the dynamics of HIV Bissau, West Africa, 1987-1989: incidence, prevalences, and
infection coupled with availability of new anti-retro- routes of transmission. J Acquir Immune Defic Syndr
virals which decrease viral load by 2-3 logs, has led 1993;6(8):941-948.
many to advocate early aggressive treatment [143]. Bl Prazuck T, Yameogo JM, Heylinck B et al. Mother-to-child
Viral diversity and high replication rates appears to transmission of human immunodeficiency virus type 1 and
type 2 and dual infection: a cohort study in Banfora, Burk-
be fundamental to HIVs ability to escape the immune ina Faso. Pediatr Infect Dis J 1995;14(11):940-947.
system and anti-retrovirals. The relative homogeneity 191 Update: HIV-2 infection among blood and plasma donors
of the viral quasispecies in early infection make it - United States, June 1992-June 1995. MMWR Morb
more likely to respond to treatment since it would be Mortal Wkly Rep 1995;44(32):603-606.
less likely to contain pre-existing drug resistant vari- [lOI Imai M, Hayashi T, Kondo M et al. Differentiation of
human immunodeficiency virus type 1 (HIV-l) infections
ants. An early decrease in the number of rounds of with HIV-2-cross-reacting antibody from mixed infections
replication would also markedly decrease viral diver- with HIV-l and HIV-2 by serological absorption test. J Clin
sity. In turn, this could decrease the ability to escape Microbial 1995;33(7):1727-1729.
28 GM. Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31

WI Leads from the MMWR. Update: serologic testing for anti- cell life-span, and viral generation time. Science
body to human immunodeficiency virus. Jama 1988;259(5): 1996;271(5255):1582-1586.
653,656,662-664. 1301 Pang S, Shlesinger Y, Daar ES, Moudgil T, Ho DD, Chen
[121 Update: serologic testing for antibody to humanimmunod- IS. Rapid generation of sequence variation during primary
eficiency virus. Morb Mortal Wkly Rep 1988;36(52):833-840, HIV-l infection. AIDS 1992;6(5):453-460.
845. [311 Wolinsb SM, Wike CM, Korber BT et al. Selective tran-
I131 From the Centers for Disease Control. Interpretation and smission of human immunodeficiency virus type-l variants
use of the western blot assay for serodiagnosis of human from mothers to infants. Science 1992;255(5048>1134-1137.
immunodeficiency virus type 1 infections. Jama WI Zhu T, MO H, Wang N et al. Genotypic and phenotypic
1989;262(24):3395-3397. characterization of HIV-1 patients with primary infection.
[141 From the Centers for Disease Control and Prevention. Re- Science 1993;261(5125):1179-1181.
commendations for HIV testing services for inpatients and t331 Coffin JM. HIV population dynamics in vivo: implications
outpatients in acute-care hospital settings. Jama for genetic variation, pathogenesis, and therapy. Science
1993;269(16):2071-2072. 1995;267(5197):483-489.
WI Harouse JM, Kunsch C, Hartle HT, et al. CD4 independent 1341 Baltimore D. Lessons from people with nonprogressive HIV
infection of human neural cells by human immunodeficiency infection. N Engl J Med 1995;332(4):259-260.
virus type 1. J Virol 1989;63:2527-2533. Pantaleo G, Menzo S, Vaccarezza M et al. Studies in sub-
[351
[161 Homsy J, Meyer M, Tateno M, Clarkson S, Levy JA. The Fc jects with long-term nonprogressive human immunodefi-
and not CD4 receptor mediates antibody enhancement of ciency virus infection. N Engl J Med 1995;332(4):209-216.
HIV infection in human cells. Science 1989;244: 1357-1360.
[361 Cao Y, Gin L, Zhang L, Safrit J, Ho DD. Virologic and
[I71 Alkhatib G, Combadiere C, Broder CC et al. CC CKRS: a immunologic characterization of long-term survivors of hu-
RANTES, MIP-1 alpha, MIP-1 beta receptor as a fusion man immunodeficiency virus type 1 infection. N Engl J Med
cofactor for macrophage-tropic HIV-l. Science 1995;332(4):201-208.
1996;272(5270): 1955-1958.
[371 Kirchhoff F, Greenough TC, Brettler DB, Sullivan JL,
1181 Choe H, Farzan M, Sun Y et al. The beta-chemokine Desrosiers RC. Brief report: absence of intact nef sequences
receptors CCR3 and CCRS facilitate infection by primary in a long-term survivor with nonprogressive HIV-l infection.
HIV-l isolates. Cell 1996;85(7): 1135-1148. N Engl J Med 1995;332(4):228-232.
1191 Deng H, Liu R, Ellmeier W et al. Identification of a major
[381 Case-control study of HIV seroconversion in health-care
co-receptor for primary isolates of HIV-l. Nature
workers after percutaneous exposure to HIV-infected
1996;381(6584):661-666.
blood-France, United Kingdom, and United States, January
DOI Doranz BJ, Rucker .I, Yi Y et al. A dual-tropic primary
1988-August 1994. Morb Mortal Wkly Rep
HIV-1 isolate that uses fusin and the beta-chemokine recep-
1995;44(50):929-933.
tors CKR-5, CKR3, and CKR-2b as fusion cofactors. Cell
1996;85(7): 1149-1158. WI Gerberding JL. Prophylaxis for occupational exposure to
HIV. Ann Intern Med 1996;125(6):497-501.
D11 Feng Y, Broder CC, Kennedy PE, Berger EA. HIV-1 entry
cofactor: functional cDNA cloning of a seven-transmem- 1401 Rates of mother-to-child transmission of HIV-l in Africa,
brane, G protein-coupled receptor. Science 1996;272(5263): America, and Europe: results from 13 perinatal studies. The
872-877. working group on mother-to-child transmission of HIV. J
Acq Immune Defic Syndr Hum Retrovirol 1995;8(5):
[221 Dragic T, Litwin V, Allaway GP et al. HIV-l entry into
CD4+ cells is mediated by the chemokine receptor CC- 506-510.
CKR5. Nature 1996;381(6584):667-673. 1411 Report of a Consensus Workshop, Siena, Italy, January
17-18, 1992. Maternal factors involved in mother-to-child
Dl Cocchi F, DeVico AL, Garzino-Demo A,Arya SK, Gal10
RC, Lusso P. Identification of RANTES, MIP-1 alpha, and transmission of HIV-l. J Acq Immune Defic Syndr
MIP-1 beta as the major HIV-suppressive factors produced 1992;5(10):1019-1029.
by CD8+ T cells. Science 1995;270(5243):1811-1815. 1421 Risk factors for mother-to-child transmission of HIV-l. Eu-
D41 Dean M, Carrington M, Wiier C et al. Genetic restriction ropean collaborative study. Lancet 1992;339(8800):
of HIV-l infection and progression to AIDS by a deletion 1007-1012
allele of the CKR5 structural gene. Hemophilia Growth and [431 Gabiano C, Tovo PA, de Martin0 M et al. Mother-to-child
Development Study, Multicenter AIDS Cohort Study, Multi- transmission of human immunodeficiency virus type 1: risk
center Hemophilia Cohort Study, San Francisco City Cohort, of infection and correlates of transmission. Pediatrics
ALIVE Study. Science 1996;273(5283):1856-1862. 1992;90(3):369-374.
[251 Liu R, Paxton WA, Choe S et al. Homozygous defect in WI Connor EM, Sperling RS, Gelber R et al. Reduction of
HIV-l coreceptor accounts for resistance of some multiply- maternal-infant transmission of human immunodeficiency
exposed individuals to HIV-l infection. Cell virus type 1 with zidovudine treatment. Pediatric AIDS
1996;86(3):367-377. Clinical Trials Group Protocol 076 Study Group. N Engl J
WI Samson M, Libert F, Doranz BJ et al. Resistance to HIV-l Med 1994;331(18):1173-1180.
infection in Caucasian individuals bearing mutant alleles of [451 U.S. Public Health Service recommendations for human
the CCR-5 chemokine receptor gene. Nature 1996; immunodeficiency virus counseling and voluntary testing for
382(6593):722-725. pregnant women. Morb Mortal Wkly Rep 1995;44(RR-
WI Wei X, Ghosh SK, Taylor ME et al. Viral dynamics in 7):1-15.
human immunodeficiency virus type 1 infection. Nature [461 Perinatal human immunodeficiency virus testing. Provisional
1995;373(6510):117-122. Committee on Pediatric AIDS, American Academy of Pedi-
D81 Ho D, Neumann AU, Perelson AS, Chen W, Leonard JM, atrics. Pediatrics 1995;95(2):303-307.
Markowitz M. Rapid turnover of plasma virions and CD4 [471 Rouzioux C, Costagliola D, Burgard M et al. Timing of
lymphocytes in HIV-1 infection. Nature 1995;373:123-126. mother-to-child HIV-l transmission depends on maternal
WI Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho status. The HIV infection in newborns French collaborative
DD. HIV-1 dynamics in vivo: virion clearance rate, infected study group. AIDS 1993;7(Suppl. 2):S49-S52.
GM Aldrouandi /Progress in Pediatric Cardiology 7 (1997) 19-31 29

[48] Rouzioux C, Costagliola D, Burgard M et al. Estimated core antigen in term human placentas. Pediatrics
timing of mother-to-child human immunodeficiency virus 1992;89(2):207-209.
type 1 (HIV-l) transmission by use of a Markov model. The b51 St Louis ME, Kamenga M, Brown C et al. Risk for perinatal
HIV Infection in Newborns French Collaborative Study HIV-l transmission according to maternal immunologic, vi-
Group. Am J Epidemiol 1995;142(12):1330-1337. rologic, and placental factors. Jama 1993;269(22):2853-2859.
[49] Burgard M, Mayaux MJ, Blanche S et al. The use of viral WI Ryder RW, Nsa W, Hassig SE et al. Perinatal transmission
culture and p24 antigen testing to diagnose human im- of the human immunodeficiency virus type 1 to infants of
munodeficiency virus infection in neonates. The HIV infec- seropositive women in Zaire. N Engl J Med
tion in newborns French collaborative study group. N Engl J 1989;320(25):1637-1642.
Med 1992;327(17):1192-1197. 1681 Duliege AM, Amos CI, Felton S, Biggar RJ, Goedert JJ.
[50] Datta P, Embree JE, Kreiss JK et al. Mother-to-child tran- Birth order, delivery route, and concordance in the tran-
smission of human immunodeficiency virus type 1: report smission of human immunodeficiency virus type 1 from
from the Nairobi Study. J Infect Dis 1994;170(5):1134-1140. mothers to twins. International Registry of HIV-Exposed
[Sl] Datta P, Embree JE, Kreiss JK et al. Resumption of breast- Twins. J Pediatr 1995;126(4):625-632.
feeding in later childhood: a risk factor for mother to child [@I Kind C. Mother-to-child transmission of human immuno-
human immunodeficiency virus type 1 transmission. Pediatr deficiency virus type 1: influence of parity and mode of
Infect Dis J 1992;11(11):974-976. delivery. Paediatric AIDS Group of Switzerland. Eur J Pedi-
[52] Van de Perre P. Postnatal transmission of human immunod- atr 1995;154(7):542-545.
eficiency virus type 1: the breast-feeding dilemma. Am J [701 Caesarean section and risk of vertical transmission of HIV-l
Obstet Gynecol 1995;173(2):483-487. infection. The European Collaborative Study. Lancet
[53] Bryson YJ, Luzuriaga K, Sullivan JL, Wara DW. Proposed 1994;343(8911):1464-1467.
definitions for in utero versus intrapartum transmission of [711 Thomas PA, Weedon J, Krasinski K et al. Maternal predic-
HIV-l [letter]. N Engl J Med 1992;327(17):1246-1247. tors of perinatal human immunodeficiency virus transmis-
[54] Kline MW, Lewis DE, Hollinger FB et al. A comparative sion. The New York City Perinatal HIV Transmission Col-
study of human immunodeficiency virus culture, polymerase laborative Study Group. Pediatr Infect Dis J
chain reaction and anti-human immunodeficiency virus im- 1994;13(6):489-495.
munoglobulin A antibody detection in the diagnosis during [721 Tovo PA, de Martino M, Gabiano C et al. Mode of delivery
early infancy of vertically acquired human immunodeficiency and gestational age influence perinatal HIV-l transmission.
virus infection. Pediatr Infect Dis J 1994;13(2):90-94. Italian Register for HIV Infection in Children. J Acq Im-
[55] Comeau AM, Hsu HW, Schwerzler M et al. Identifying mune Defic Syndr Hum Retrovirol 1995;11(1):88-94.
human immunodeficiency virus infection at birth: applica- [731 Villari P, Spino C, Chalmers TC, Lau J, Sacks HS. Cesarean
tion of polymerase chain reaction to Guthrie cards. J Pedi- section to reduce perinatal transmission of human im-
atr 1993;123(2):252-258. munodeficiency virus. A meta-analysis. Online J Curr Clin
[56] Backe E, Unger M, Jimenez E, Siegel G, Schafer A, Vogel Trials 1993;Doc No 74
M. Fetal organs infected by HIV-l [letter]. AIDS [741 Kuhn L, Bobat R, Coutsoudis A et al. Cesarean deliveries
1993;7(6):896-897. and maternal-infant HIV transmission: results from a
[57] Viscarello RR, Cullen MT, DeGermaro NJ, Hobbins JC. prospective study in South Africa. J Acq Immune Defic
Fetal blood sampling in human immunodeficiency virus- Syndr Hum Retrovirol 1996;11(5):478-483.
seropositive women before elective midtrimester termina- 1751 Douglas GC, King BF. Maternal-fetal transmission of hu-
tion of pregnancy. Am J Obstet Gynecol 1992;167(4 Pt man immunodeficiency virus: a review of possible routes and
1):1075-1079. cellular mechanisms of infection. Clin Infect Dis
[58] Tanaka KE, Hatch WC, Kress Y et al. HIV-l infection of 1992;15(4):678-691.
human fetal thymocytes. J Acq Immune Defic Syndr 1761 Landesman SH, Kalish LA, Bums DN et al. Obstetrical
1992;5(1):94-101. factors and the transmission of human immunodeficiency
[59] Mundy D, Schinzai RF, Gerber AR, Nahmias AI, Randal virus type 1 from mother to child. The Women and Infants
HWJ. HIV isolated from amniotic fluid. Lancet Transmission Study. N Engl J Med 1996;334(25):1617-1623.
1987;ii:459-460. [771 von Seidlein L, Bryson Y. Maternal-Fetal Transmission of
[60] Chandwani S, Greco MA, Krasinski K, Borkowsky W. the Human Immunodeficiency Virus Type 1. Seminars in
Pathology of the placenta in HIV-l infection. Prog AIDS Pediatric Infectious Diseases 1994;5(1):78-86.
Path01 1992;3(1):65-81. L’81 Ukwu HN, Graham BS, Lambert JS, Wright PF. Perinatal
1611 David FJ, Autran B, Tran HC et al. Human trophoblast cells transmission of human immunodeficiency virus-l infection
express CD4 and are permissive for productive infection and maternal immunization strategies for prevention. Obstet
with HIV-l. Clin Exp Immunol 1992,88(1):10-16. Gynecol 1992;80(3 Pt 0458-468.
[62] Fazely F, Fry GN, Thirkill TL, Hakim H, King BF, Douglas [791 Dickover RE, Garratty EM, Herman SA et al. Identification
GC. Kinetics of HIV infection of human placental syncy- of levels of maternal HIV-l RNA associated with risk of
tiotrophoblast cultures: an ultrastructural and immuno- perinatal transmission. Effect of maternal zidovudine treat-
cytochemical study. AIDS Res Hum Retrovir ment on viral load. Jama 1996;275(8):599-605.
1995;11(9):1023-1030. WI Borkowsky W, Krasinski K, Cao Y et al. Correlation of
[63] Kesson AM, Fear WR, Kazazi F et al. Human immunodefi- perinatal transmission of human immunodeficiency virus
ciency virus type 1 infection of human placental macrophages type 1 with maternal viremia and lymphocyte phenotypes. J
in vitro. J Infect Dis 1993;168(3):571-579. Pediatr 1994;125(3):345-351.
1641 Backe E, Jimenez E, Unger M et al. Vertical human im- [811 Bredberg-Raden U, Urassa W, Urassa E et al. Predictive
munodeficiency virus transmission: a study of placental markers for mother-to-child transmission of HIV-l in Dar
pathology in relation to maternal risk factors. Am J Perina- es Salaam, Tanzania. J Acquir Immune Defic Syndr Hum
to1 1994;11(5):326-330. Retrovirol 1995;8(2):182-187.
[65] Mattem CF, Murray K, Jensen A, Farzadegan H, Pang J, [821 Boyer PJ, Dillon M, Navaie M et al. Factors predictive of
Modlin JF. Localization of human immunodeficiency virus maternal-fetal transmission of HIV-l. Preliminary analysis
30 GM Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31

of zidovudine given during pregnancy and/or deliver. J Am [991 Parekh BS, Shaffer N, Coughhn R et al. Human immunod-
Med Assoc 1994;271(24):1925-1930. eficiency virus l-specific IgA capture enzyme immunoassay
D331 Erb P, Krauchi S, Burgin D, Biedermann K, Camli C, Rudin for early diagnosis of human immunodeficiency virus 1 in-
C. Quantitative anti-p24 determinations can predict the risk fection in infants. NYC Perinatal HIV Transmission Study
of vertical transmission. Swiss HIV and pregnancy collabora- Group. Pediatr Infect Dis J 1993;12(11):908-9.
tive study group. J Acq Immune Defic Syndr mI George JR, Parekh BS, Shaffer N et al. Detection of HIV-1
1994;7(3):261-264. IgA by an IgA capture enzyme immunoassay for early diag-
WI Husson RN, Lan Y, Kojima E, Venzon D, Mitsuya H, nosis in infants. NYC Perinatal HIV Transmission Col-
McIntosh K. Vertical transmission of human immunodefi- laborative Study Group. Ann NY Acad Sci 1993;693:272-274.
ciency virus type 1: autologous neutralizing antibody, virus ml Lewis DE, Adu-Oppong A, Hollinger FB et al. Sensitivity of
load, and virus phenotype. J Pediatr 1995;126(6):865-871. immune complex-dissociated p24 antigen testing for early
k351 Papaevangelou V, Moore T, Nagaraj V, Krasinski K, detection of human immunodeficiency virus in infants. Clin
Borkowsb W. Lack of predictive value of maternal human Diagn Lab Immunol 1995;2(1):87-90.
immunodeficiency virus p24 antigen for transmission of in-
m21 Miles SA, Balden E, Magpantay L et al. Rapid serologic
fection to their children. Pediatr Infect Dis J
testing with immune-complex-dissociated HIV p24 antigen
1992;11(10):851-855. for early detection of HIV infection in neonates. Southern
B61 Halsey NA, Markham R, Wahren B, Boulos R, Rossi P, California Pediatric AIDS Consortium. N Engl J Med
Wigzell H. Lack of association between maternal antibodies
1993;328(5):297-302.
to V3 loop peptides and maternal-infant HIV-1 transmis-
sion. J Acq Immune Defic Syndr 1992;5(2):153-157. m31 Borkowsky W, Krasinski K, Pollack H, Hoover W, Kaul A,
Ilmet-Moore T. Early diagnosis of human immunodeficiency
[871 Scarlatti G, Leitner T, Hodara V et al. Neutralizing anti-
bodies and viral characteristics in mother-to-child transmis- virus infection in children less than 6 months of age: com-
sion of HIV-l. AIDS 1993;7(Suppl 2):S45-48. parison of polymerase chain reaction, culture, and plasma
antigen capture techniques. J Infect Dis 1992;166(3):616-619.
k381 Rossi P, Moschese V, Broliden PA et al. Presence of mater-
nal antibodies to human immunodeficiency virus 1 envelope DO41 Nielsen K, Santos E, Rubini N et al. Immune complex-disso-
glycoprotein gp120 epitopes correlates with the uninfected ciated p24 antigenemia in the diagnosis of human immunod-
status of children born to seropositive mothers. Proc Nat1 eficiency virus infection in vertically infected Brazilian chil-
Acad Sci USA 1989;86(20):8055-58. dren. Pediatr infect Dis J 1995;14(1):67-69.
WI Devash Y, Calvelli T, Wood D, Reagan K, Rubinstein A. HO51 Wang XP, Pa51 M, Tetali S et al. Improved specificity of in
Vertical transmission of human immunodeficiency virus is vitro anti-HIV antibody production: implications for diag-
correlated with the absence of high-affinity/avidity maternal nosis and timing of transmission in infants born to
antibodies to the gp 1 20 principal neutralizing domain. Proc HIV-seropositive mothers. AIDS Res Hum Retrovir
Nat1 Acad Sci USA 1990;87:3445-3449. 1994;10(6):691-699.
[901 Jean-Pierre A, Matthews T, Coombs R et al. Antibody to V3 DO61 Amadori A, de Rossi A, Giaquinto C, Faulkner-Valle G,
loop peptides does not predict vertical transmission of HIV. Zacchello F, Chieco-Bianchi L. In-vitro production of HIV-
VII International Conference on AIDS. Florence, Italy, specific antibody in children at risk of AIDS. Lancet
1991. 1988;1(8590):852-854.
[911 Just JJ, Abrams E, Louie LG et al. Influence of host DO71 Pahwa S, Chirmule N, Leombruno C et al. In vitro synthesis
genotype on progression to acquired immunodeficiency syn- of human immunodeficiency virus-specific antibodies in pe-
drome among children infected with human immunodefi- ripheral blood lymphocytes of infants. Proc Nat1 Acad Sci
ciency virus type 1. J Pediatr 1995;127(4):544-549. USA 1989;86(19):7532-7536.
WI Kaslow RA, Carrington M, Apple R et al. Influence of [lo81 Lee FK, Nahmias AJ, Lowery S et al. ELISPOT: a new
combinations of human major histocompatibility complex approach to studying the dynamics of virus-immune system
genes on the course of HIV-l infection. Nat Med interaction for diagnosis and monitoring of HIV infection.
1996;2(4):405-411. AIDS Res Hum Retrovir 1989;5(5):517-523.
[931 Persistent lack of detectable HIV-l antibody in a person DO91 Paul MO, Tetali S, Lesser ML et al. Laboratory diagnosis of
with HIV infection-Utah, 1995. Morb Mortal Wkly Rep infection status in infants perinatally exposed to human
1996;45(9):181-185. immunodeficiency virus type 1. J Infect Dis 1996;173(1):
[941 CDC 1994 revised classification system for human immuno- 68-76.
deficiency virus infection in children less than 13 years.
[1101 Krivine A, Firtion G, Cao L, Francoual C, Henrion R,
Morb Mortal Wkly Report 1994;43:1-19. Lebon P. HIV replication during the first weeks of life.
[951 Identification of HIV-l group 0 infection-1996. From the Lancet 1992;339(8803):1187-1189.
Centers for Disease Control and Prevention. J Am Med
Assoc 1996;276(7):521-522. [1111 Comeau AM, Harris JA, McIntosh K, Weiblen BJ, Hoff R,
Grady GF. Polymerase chain reaction in detecting HIV
Ml CDC. Interpretive criteria used to report Western blot re- infection among seropositive infants: relation to clinical
sults for HIV-l antiboy testing - United States. Morb Mor-
tal Wkly Report 1991;40(2):692-695. status and age and to results of other assays.J Acq Immune
Defic Syndr 1992;5(3):271-278.
[971 McIntosh K, Comeau AM, Wara D et al. The utility of IgA
antibody to human immunodeficiency virus type 1 in early [1121 Rogers MF, Ou CY, Ray leld M et al. Use of the po-
diagnosis of vertically transmitted infection. National Insti- lymerase chain reaction for early detection of the proviral
tute of Allergy and Infectious Diseases and National Insti- sequences of human immunodeficiency virus in infants born
tute of Child Health and Human Development Women and to seropositive mothers. New York City Collaborative Study
Infants Transmission Study Group. Arch Pediatr Adolesc of Maternal HIV Transmission and Montefiore Medical
Med 1996;150(6):598-602. Center HIV Perinatal Transmission Study Group. N Engl J
[981 Livingston RA, Hutton N, Halsey NA, Kline RL, Joyner M, Med 1989;320(25):1649-1654.
Quinn TC. Human immunodeficiency virus-specific IgA in ill31 Bremer JW, Lew JF, Cooper E et al. Diagnosis of infection
infants born to human immunodeficiency virus-seropositive with human immunodeficiency virus type 1 by a DNA po-
women. Arch Pediatr Adolesc Med 1995;149(5):503-507. lymerase chain reaction assay among infants enrolled in the
G.M. Aldrovandi /Progress in Pediatric Cardiology 7 (1997) 19-31 31

Women and Infants’ Transmission Study. J Pediatr I1311 Kovacs A, Frederick T, Church J, Eller A, Qxtoby M,
1996;129(21:198-207. Mascola L. CD4 lymphocyte counts and Pneumocyhs carinii
[1141 McIntosh K, Pitt J, Brambilla D et al. Blood culture in the pnewumonia in pediatric HIV infection. JAMA
first 6 months of lifefor the diagnosis of vertically transmit- 1991;265:1698-1703.
ted human immunodeficiency virus infection. The Women D321 Buchbinder SP, Katz MI-I, Hess01 NA, Q’Malley PM, Holm-
and Infants Transmission Study Group. J Infect Dis berg SD. Long-term HIV-l infection without immunologic
1994;170(4):996-1000. progression. Aids 1994;8(8):1123-1128.
ill51 Munoz A, Wang MC, Bass S et al. Acquired immuno- t1331 Sheppard HW, Lang W, Ascher MS, Vittinghoff E, Winkel-
deficiency syndrome (AIDS)-free time after human im- stein W. The characterization of non-progressors: long-term
munodeficiency virus type 1 (HIV-l) seroconversion in ho- HIV-1 infection with stable CD4+ T-cell levels. Aids
mosexual men. Multicenter AIDS Cohort Study Group. Am 1993;7(9):1159-1166.
J Epidemiol 1989;130(3):530-539. [1341 Keet IP, Krol A, Klein MR et al. Characteristics of long-term
WI Munoz A, Kirby AJ, He YD et al. Long-term survivors with asymptomatic infection with human immunodeficiency virus
HIV-l infection: incubation period and longitudinal pat- type 1 in men with normal and low CD4+ cell counts. J
terns of CD4+ lymphocytes. J Acq Immune Defic Syndr infect Dis 1994;169(6):1236-1243.
Hum Retrovirol 1995;8(5):496-505.
I1351 Learmont J, Cook L, Dunckley H, Sullivan JS. Update on
D171 Piatak M, Jr., Saag MS, Yang LC et al. High levels of HIV-l
long-term symptomless HIV type 1 infection in recipients of
in plasma during all stages of infection determined by com-
blood products from a single donor [letter]. AIDS Res Hum
petitive PCR. Science 1993;259(5102):1749-1754.
Retrovir 1995;11(1):1.
I1181 Piatak M, Jr., Saag MS, Yang LC et al. Determination of
plasma viral load in HIV-l infection by quantitative compet- [I361 Deacon NJ, Tsykin A, Solomon A et al. Genomic structure
itive polymerase chain reaction. AIDS 1993;7 Suppl of an attenuated quasispecies of HIV-1 from a blood trans-
2:S65-71. fusion donor and recipients. Science 1995;270(5238):
Clark SJ, Shaw GM. The acute retroviral syndrome and the 988-991.
t1191
pathogenesis of HIV-1 infection. Semin Immunol I1371 Haynes BF, Pantaleo G, Fauci AS. Toward an understand-
1993;5(3):149-155. ing of the correlates of protective immunity to HIV infec-
WOI Daar E, TM, RD M, DD H. Transient high levels of viremia tion. Science 1996;271(5247):324-328.
in patients with primary human immunodeficiency virus type [1381 Schrager L, Young J, Fowler M, Mathieson B, S. V. Long-
1 infection. N Engl J Med 1991;324(14):961-964. term survivors of HIV-l infection: definitions and research
K31 Mellors JW, Kingsley LA, Rinaldo CR, Jr. et al. Quantita- challenges. AIDS 1994;8:S95-S108.
tion of HIV-l RNA in plasma predicts outcome after sero- D391 Borkowsky W, Krasinski K, Paul D et al. Human immuno-
conversion. Ann Intern Med 1995;122(8):573-579. deficiency virus type 1 antigenemia in children. J Pediatr
K4 Mellors JW, Rinaldo CR, Jr., Gupta P, White RM, Todd JA, 1989;114(6):940-945.
Kingsley LA. Prognosis in HIV-l infection predicted by the [1401 Bryson YJ, Pang S, Wei LS, Dickover R, Diagne A, Chen IS.
quantity of virus in plasma. Science 1996;272(5265): Clearance of HIV infection in a perinatally infected infant.
1167-1170. N Engl J Med 1995;332(13):833-838.
WA O’Brien WA, Hartigan PM, Martin D et al. Changes in u411 Miller CJ, Marthas M, Torten J et al. Intravaginal inocula-
plasma HIV-l RNA and CD4+ lymphocyte counts and the tion of rhesus macaques with cell-free simian immunodefi-
risk of progression to AIDS. Veterans Affairs Cooperative ciency virus results in persistent or transient viremia. J Virol
Study Group on AIDS. N Engl J Med 1996;334(7):426-431. 1994;68(10):6391-6400.
w41 Mayauz MJ, Burgard M, Teglas JP et al. Neonatal charac- [1421 Guerra-Romero L, Parras Vazquez F. Physicians’ experi-
teristics in rapidly progressive perinatally acquired HIV-l ence and survival inpatients with AIDS [letter]. N Engl J
disease. The French Pediatric HIV Infection Study Group. Med 1996;335(5):350-351.
Jama 1996;275(8):606-610.
[1431 Ho DD. Time to hit HIV, early and hard. N Engl J Med
w51 Auger I, Thomas P, DeGruttola V et al. Incubation periods 1995;333(7):450-451.
for pedatric AIDS patients. Nature 1988;336:575-577.
De Rossi A, Masiero S, Giaquinto C et al. Dynamics of viral I1441 Rausch DM, Heyes MP, Murray EA, Eiden LE. Zidovudine
[1261 treatment prolongs survival and decreases virus load in the
replication in infants with vertically acquired human im-
munodeficiency virus type 1 infection. J Clin Invest central nervous system of rhesus macaques infected perina-
1996;97(2):323-330. tally with simian immunodeficiency virus. J infect Dis
1995;172(1):59-69.
I1271 Bamhart Hx, Caldwell MB, Thomas P et al. Natural history
of human immunodeficiency virus disease in perinatally in- D451 Van Rompay KK, Otsyula MG, Marthas ML, Miller CJ,
fected children: an analysis from the Pediatric Spectrum of McChesney MB, Pedersen NC. Immediate zidovudine treat-
Disease Project. Pediatr 1996;97(5):710-716. ment protects simian immunodeficiency virus-infected new-
[I281 Munoz A, Schrager LK, Bacellar H et al. Trends in the born macaques against rapid onset of AIDS. Antimicrob
incidence of outcomes defining acquired immunodeficiency Agents Chemother 1995;39(1):125-131.
syndrome (AIDS) in the Multicenter AIDS Cohort Study: W61 I-e Grand R, Clayette P, Noack 0 et al. An animal model
1985-1991. Am J Epidemiol 1993;137(4):423-438. for antilentiviral therapy: effect of zidovudine on viral load
WI Denny T, Yogev R, Gelman R et al. Lymphocyte subsets in during acute infection after exposure of macaquesto simian
healthy children during the first 5 years of life. Jama immunodeficiency virus. AIDS Res Hum Retrovir
1992;267(11):1484-1488. 1994;10(10):1279-1287.
t1301 Erkeller-Yuksel FM, Deneys V, Yuksel B et al. Age-related [1471 Ma&all CL, Fleisher TA, Brown MR et al. Age, thy-
changes in human blood lymphocyte subpopulations. J Pedi- mopoiesis, and CD4+ T-lymphocyte regeneration after in-
atr 1992;120(2 Pt 0216-222. tensive chemotherapy. Engl J Med 1995;332(3):143-149.

You might also like