Está en la página 1de 11

JOURNAL OF VIROLOGY, Oct. 2007, p. 1020910219 Vol. 81, No.

19
0022-538X/07/$08.000 doi:10.1128/JVI.00872-07
Copyright 2007, American Society for Microbiology. All Rights Reserved.

MINIREVIEW
Human Immunodeficiency Virus Type 1 Subtype Distribution in the
Worldwide Epidemic: Pathogenetic and Therapeutic Implications
L. Buonaguro,* M. L. Tornesello, and F. M. Buonaguro
Laboratory of Viral Oncogenesis and Immunotherapy and AIDS Reference Center, Ist. Naz. Tumori Fond. G. Pascale, Naples, Italy

Human immunodeficiency virus type 1 (HIV-1) is the caus- creasing number of viral isolates available worldwide and im-
ative agent of AIDS (10, 38, 105, 114). It is characterized by provements in sequencing methods, HIV-1 phylogenetic clas-
extensive and dynamic genetic diversity, generating variants sifications are currently based either on nucleotide sequences
falling into distinct molecular subtypes as well as recombinant derived from multiple subgenomic regions (gag, pol, and env)
forms; these forms display an uneven global distribution (55). of the same isolates or on full-length genome sequence analysis.
This diversity has implications for our understanding of viral This approach has revealed virus isolates in which phylogenetic
transmission, pathogenesis, and diagnosis and profoundly in- relations with different subtypes switch along their genomes.
fluences strategies for vaccine development. These intersubtype recombinant forms are thought to have orig-
Here we review selected aspects of the genetic diversity of inated in individuals multiply infected with viruses of two or more
HIV-1, with particular emphasis on its pathogenetic and ther- subtypes. When an identical recombinant virus is identified in at
apeutic implications. least three epidemiologically unlinked people and is characterized
by full-length genome sequencing, it can be designated as a cir-
HIV-1 GENETIC SUBTYPES culating recombinant form (CRF) (99, 110) (Fig. 1).
More than 20 CRFs whose origins can be tracked to areas
HIV-1 is characterized by extensive genetic heterogeneity where the parental strains are cocirculating have been reported
driven by several factors, such as the lack of proofreading (Fig. 2). Cocirculation of multiple subtypes and CRFs in the
ability of the reverse transcriptase (RT) (96, 108), the rapid same populations increases the probability that individuals will
turnover of HIV-1 in vivo (56), host selective immune pres- become superinfected with different HIV-1 genetic forms
sures (84), and recombination events during replication (122). which can swap parts of their genetic material, resulting in the
Due to this variability, HIV-1 variants are classified into three generation of several recombinants, called unique recombi-
major phylogenetic groups: group M (main), group O (outlier), nant forms, or URFs, which, if spread to other people, will
and group N (non-M/non-O) (6, 52, 116). Group M, which is lead to their classification as CRFs (79).
responsible for the majority of infections in the worldwide
HIV-1 epidemic, can be further subdivided into 10 recognized
phylogenetic subtypes, or clades (A to K), which are approxi- GEOGRAPHIC DISTRIBUTION OF HIV-1 SUBTYPES
mately equidistant from one another (Fig. 1). Within group M,
the average intersubtype genetic variability is 15% for the gag Molecular epidemiological studies show that, with the excep-
gene and 25% for the env gene (58, 64, 68, 70, 92, 109). tion of sub-Saharan Africa, where almost all subtypes, CRFs, and
Moreover, within a subtype, it is possible to identify groups several URFs have been detected, there is a specific geographic
of viral isolates forming genetically related sister clades, distribution pattern for HIV-1 subtypes (55, 97). This distribution
termed subsubtypes (109), which appear to be phylogenetically pattern seems to be the consequence of either accidental traffick-
more closely related to each other than to other subtypes. This ing (viral migration), with a resulting founder effect, or a prev-
is the case with the A and F clades, whose members are cur- alent route of transmission, which results in a strong advantage
rently classified into subsubtypes A1 to A2 and F1 to F2, for and local predominance of the prevalent subtype transmitted
respectively (42, 123). Clades B and D are more closely related in that population (17, 81, 91, 93, 98).
to each other than to other subtypes, and clade D is considered According to recent studies, on a global scale the most
the early clade B African variant, but their original designation prevalent HIV-1 genetic forms are subtypes A, B, and C, with
as subtypes is retained by authors for consistency with earlier subtype C accounting for almost 50% of all HIV-1 infections
published works (40, 74). worldwide (Fig. 3). Subtype A viruses are predominant in areas
Classification of HIV-1 subtypes was originally based on the of central and eastern Africa (Kenya, Uganda, Tanzania, and
subgenomic regions of individual genes. However, with an in- Rwanda) and in eastern European countries formerly consti-
tuting the Soviet Union. Subtype B is the main genetic form in
western and central Europe, the Americas, and Australia and
* Corresponding author. Mailing address: Lab. of Viral Oncogenesis is also common in several countries of Southeast Asia, north-
and Immunotherapy & AIDS Ref. Center, Ist. Naz. Tumori Fond. G.
Pascale, Via Mariano Semmola, 1, 80131 Naples, Italy. Phone: 39-
ern Africa, and the Middle East and among South African and
081-5903.609. Fax: 39-081-545.1276. E-mail: irccsvir@unina.it. Russian homosexual men. Subtype C viruses are predominant

Published ahead of print on 18 July 2007. in those countries with 80% of all global HIV-1 infections,

10209
10210 MINIREVIEW J. VIROL.

FIG. 1. Evolutionary relationships among nonrecombinant HIV-1 strains. The phylogenetic tree shows HIV-1 groups M, O, and N and subtypes
and CRFs within the M group. The phylogenetic analysis was performed on nearly full-length sequences and is based on the neighbor-joining
method. The internal branches defining a subtype have been estimated from 1,000 bootstrap replicates.

such as southern Africa and India. The relevance of CRFs in simian immunodeficiency virus SIVcpz from the chimpanzee
the global HIV-1 pandemic is increasingly recognized, ac- subspecies Pan troglodytes troglodytes to humans (39). This
counting for 18% of infections (55, 97) and representing the cross-species transmission could have occurred in West Equa-
predominant local form in Southeast Asia (CRF01-AE) (82, torial Africa from direct exposure to animal blood as a conse-
89, 104) and in West and West Central Africa (CRF02-AG) quence of hunting, butchering, and consumption of raw meat
(80, 87) (Fig. 4). (53). In fact, this region includes countries (Gabon, Equatorial
Guinea, Cameroon, and the Republic of Congo) where the
HIV-1 DIVERSITY AND ITS ORIGIN following conditions in support of this hypothesis are found: (i)
HIV-1 groups M, N, and O cocirculate in human populations;
Strong phylogenetic evidence suggests that HIV-1 originated (ii) HIV-1 group M viruses show the greatest diversity (28, 59,
by zoonotic cross-species transmission of the simian lentivirus 85, 86, 88, 101, 127); and (iii) chimpanzees (Pan troglodytes
VOL. 81, 2007 MINIREVIEW 10211

FIG. 2. Mosaic structure of HIV-1 CRFs. All CRFs that have been described to date are shown (modified from http://www.hiv.lanl.gov/content
/hiv-db/CRFs/CRFs.html). The letters and graphic patterns represent the different subtypes of HIV-1 involved in the recombination events. U,
unknown.

troglodytes) have been found to be infected with genetically tiple founders, and make zoonotic transmission from infected
closely related viruses (26, 39, 78, 102, 116). These conditions animals to humans plausible.
both support long-lasting circulation of the whole spectrum of Moreover, the interspersion of HIV-1 group M, N, and O
the known HIV-1 genetic forms, possibly resulting from mul- sequences among different SIVcpz (Pan troglodytes troglodytes)
lineages in phylogenetic trees requires that HIV-1 viruses from
the three groups originated from no fewer than three separate
SIVcpz transmission events (Fig. 5). However, while group M
viruses appear to have efficiently adapted to the new host
species, spreading all around the world and generating multi-
ple genetic subtypes, the other two groups show less efficient
adaptations to humans. HIV-1 group O seems to be endemic
to Cameroon and neighboring countries in West Central Af-
rica, where it represents only approximately 1 to 5% of HIV-
1-positive samples (102); likewise, group N viruses have been
identified only in a limited number of individuals from Cam-
eroon (6, 116). Moreover, group N viruses appear to be the
result of a recombination event between an SIVcpz-like and an
FIG. 3. Global prevalence of HIV-1 genetic forms. The global
HIV-1-like virus (39), and their high degree of similarity to
prevalence of each form, expressed as a percentage of the total number
of HIV-1 isolates identified worldwide, is shown. Isolates from HIV-1 chimpanzee viruses may indicate a significantly recent zoonotic
groups O and N were not included in this analysis. cross-species transmission. This evidence suggests that cross-
10212 MINIREVIEW J. VIROL.

FIG. 4. Global geographical distribution of HIV-1 genetic forms. Genetic forms predominant in different world regions are shown. The pie
charts show the prevalence of individual genetic forms in each region.

species zoonotic transmissions to humans of additional pri- Considering that African primates are the natural hosts of
mate lentiviruses and/or recombinations between HIV-1 vi- several different lentiviruses (100), it is quite reasonable that
ruses and primate lentiviruses may still occur, giving rise to such cross-species zoonotic lentivirus transmissions to humans
new HIV-1 groups with unpredictable virulence. have periodically occurred through the centuries in West

FIG. 5. Phylogenetic tree of primate lentivirus sequences. Phylogenetic tree analysis was performed by using the neighbor-joining method, based on
a 550-base pair pol fragment from SIV and HIV isolates. One isolate per each HIV-1 group M subtype was used. Branch lengths are drawn to scale (the
bar indicates 10% divergence). The branches were estimated from 1,000 bootstrap replicates, and only bootstrap values of 80% are shown.
VOL. 81, 2007 MINIREVIEW 10213

Equatorial Africa. Exposure to SIV in natural settings is com- infection. Nevertheless, a consistent demonstration of this as-
mon for individuals exposed to blood and body fluids of nat- sociation has not been given. There is clearly no predeter-
urally SIV-infected nonhuman primates. However, although mined linkage between a specific subtype and a unique mode
there is a significant percentage of seropositivity to SIV anti- of transmission. In fact, subtype A is transmitted among het-
gens in such high-risk groups (17.1%), no productive infection erosexuals in sub-Saharan Africa and IDUs in Eastern Europe;
has been detected (60). It has been proposed that the lack of similarly, subtype B is transmitted among all the historical risk
productive infections is a consequence of either exposure to groups in Western countries.
nonviable or defective SIV, a nonproductive cleared infection, Therefore, the apparent segregation of HIV-1 subtypes by
or sequestering of the virus in lymphoid tissues. The reason type of risk behavior rather than as a result of virologic factors
why HIV-1 gave rise to the AIDS pandemic only in the 20th (cell tropism, coreceptor specificity) could derive from genetic,
century has not yet been determined but could reasonably be
demographic, economic, and social factors that separate the
the sum of significant cultural and socio-behavioral changes,
different risk groups for HIV-1 infection. Moreover, the over-
the use of nonsterile needles for parenteral injections and
whelming predominance of the C subtypes in areas where
vaccinations, and the unwitting contamination of biological
unprotected heterosexual intercourse is the main transmission
products for medical treatments (e.g., oral polio vaccine) (46,
57). The earliest case of HIV-1 infection, dating from 1959, route could result from a founder effect with a fast-coloniza-
was identified in the Democratic Republic of Congo (134), and tion outcome.
using different methods of molecular clock analysis, it has been
estimated that HIV-1 group M began to radiate from its source
around the 1930s (63, 113). IMPACT OF HIV-1 GENETIC DIVERSITY ON
SPECIFICITY AND SENSITIVITY OF
DIAGNOSTIC TESTS
HIV-1 SUBTYPES AND TRANSMISSION
The genetic variability of HIV-1 may pose significant prob-
Diverse risk behaviors sustain HIV-1 transmission in differ-
lems for the specificity and/or sensitivity of serological and
ent regions of the world, and within the same region, multiple
transmission routes can be involved in spreading the epidemic. molecular diagnostic tests, which may represent a serious risk
For example, in Eastern Europe and Central Asia in 2005, 67% factor for the spreading of unidentified infections.
of HIV infections were due to needle sharing among intrave- Fourth-generation HIV immunoassays are designed to de-
nous drug users (IDUs). In South and Southeast Asia, not tect both the HIV p24 antigen and antibody in a single test in
including India, 49% of HIV infections reported were in com- order to reduce the seroconversion window (131). The sensi-
mercial sex workers and their clients, while 22% were in IDUs. tivity of these assays for the p24 antigen and seroconversion,
In Latin America, 26% of HIV infections were in men who however, can vary significantly (76, 130). Nevertheless, recent
have sex with men (MSM), and 19% were in IDUs (http://www comparative studies have shown that some of these immuno-
.unaids.org/en/HIV_data/2006GlobalReport/default.asp). In assays have levels of specificity and sensitivity comparable to
Western Europe, unprotected intercourse among heterosex- those of second- and third-generation assays designed to detect
uals accounted for 45%, and among MSM, 28%, of HIV in- either antigens or antibodies and can identify most of the known
fections (http://www.eurohiv.org/reports/report_73/pdf/report group M subtypes, the most diffused CRFs (CRF01_AE and
_eurohiv_73.pdf). CRF02_AG), and group O viruses (69).
As reported in the previous paragraph, the HIV-1 epidemic In parallel, a similar comparative study reported on molec-
in these regions is sustained by different subtypes, and within ular diagnostic tests, which might be even more affected by the
each region, segregation of subtypes to different risk groups high variability of the target HIV-1 nucleotide sequences.
has been reported. For example, the cocirculation of subtype B Analysis has shown that the widely used real-time RT-PCR for
among IDUs and CRF01_AE (originally defined as subtype E) viral load measurements is able to identify all the tested group
among heterosexuals was originally described in Thailand (41); M and group O isolates within a target concentration ranging
the segregation of subtype B to homosexuals and subtype C to
from approximately 316,230 copies/ml (5.5 log10) to below 50
heterosexuals was described in South Africa (126); more re-
copies/ml (1.6 log10), with a coefficient of correlation between
cently, two concurrent epidemics in Argentina have been re-
0.991 and 0.999 (120).
ported, one among MSM, sustained by subtype B, and the
Therefore, these reports indicate that the currently used
other among heterosexuals and IDUs, sustained by BF recom-
serological and molecular diagnostic tests for HIV-1 are still
binants (5). In Europe, where subtype B has sustained the
HIV-1 epidemic among the historical IDU and homosexual sufficiently specific and sensitive to detect the most prevalent
risk groups, non-B subtypes and CRFs are progressively being HIV-1 genetic forms worldwide. Nevertheless, anecdotal un-
introduced in association with increased heterosexual trans- identified HIV-1 isolates have been reported in primary HIV-1
mission of HIV-1 between migrants and/or immigrants from infection cohorts in monoclade HIV epidemics (21). There-
regions where HIV-1 is endemic and their European partners fore, the continuous genetic variation and intersubtype recom-
(19, 20, 121). bination events make frequent evaluation analyses of the di-
All these observations, reported at different phases of the agnostic tests and, possibly, national registries to report
HIV-1 epidemic around the world, may suggest different bio- undiagnosed cases necessary. These measures would help
logical properties for the subtypes, resulting in their segrega- greatly in keeping the diagnostic capacity of the tests up to
tion among individuals with different risk behaviors for HIV-1 date.
10214 MINIREVIEW J. VIROL.

CORRELATION BETWEEN HIV-1 GENETIC DIVERSITY (GTG to ATG), compared to two transitions in viruses of other
AND DRUG RESISTANCE subtypes (GTA to ATG). Moreover, natural resistance to
NNRTIs has been observed in patients carrying group O
The genetic variability characterizing the different HIV-1 HIV-1 viruses (29, 107), and it is believed to have a genetic
subtypes and CRFs also affects the protease and RT genes basis relating to the natural RT Y181C polymorphism, similar
coding for the viral enzymes that are the main targets of anti- to the Y181I divergence seen in naturally NNRTI-resistant
retroviral drugs. These polymorphisms, if conferring drug re- HIV-2 (117).
sistance, can be selected by drug-selective pressure and dra- All these in vitro genetic studies showing both the absence of
matically influence the therapeutic outcome. Alternatively, the nave drug resistance mutations in non-B subtypes and sub-
polymorphisms do not confer drug resistance but may change stantial similarity between B and non-B subtypes in the degree
the genetic barrier, which is defined as the number of viral of probability that they will develop drug resistance are con-
mutations required to develop escape mutations able to over- firmed by a generally conserved drug susceptibility in vivo. In
come the drug-selective pressure. In general, several mutations fact, HIV-1 subtypes do not appear to affect the efficacy of
are generally required for the virus to become resistant to first-line therapy and currently available protease and RT in-
protease inhibitors (high genetic barrier), whereas a single hibitors are equally active on all HIV-1 subtypes (3, 13, 37,
amino acid substitution can induce resistance to nonnucleoside 103). A global collaborative study based on non-B subtype
reverse transcriptase inhibitors (NNRTIs) (low genetic bar- sequences from 3,686 individuals showed (i) that most of the
rier) (12). protease and RT positions associated with drug resistance in
The frequency and pattern among HIV-1 subtypes of poly- subtype B viruses are selected by ART in one or more non-B
morphisms inducing resistance per se or leading to a faster subtypes as well and (ii) that no evidence is available that
emergence of drug resistance under pharmacological pressure non-B viruses develop resistance by mutations at positions that
have been evaluated in several recent studies. In particular, are not associated with resistance in subtype B viruses (61).
considering the lack of extensive information on non-B sub- However, a few exceptions to this general rule have been
types, which predominate in countries where the availability of reported in ART-treated patients. Subtype C viruses show a
antiretroviral therapy (ART) is very limited, great emphasis faster emergence of drug resistance to NNRTIs resulting from
has been given to comparative studies of B versus non-B sub- the appearance of the V106M mutation (73). A Ugandan study
types. In a large European study performed on approximately to prevent mother-to-child transmission of HIV-1 showed that
2,000 protease and RT gene sequences (600 non-B) from a single dose of nevirapine induced more frequent selection of
antiretroviral-nave patients, a genetic barrier similar for B and genotypic mutations associated with resistance to nevirapine in
non-B subtypes was found at almost all positions (125). women infected with subtype D than in women infected with
In addition, specific data are available for individual ART subtype A viruses (34).
drug classes. In regard to the few positions of the protease In contrast to the overall extensive genetic variability along
sequence where differences were found, such as the 82A and the HIV-1 genome, the overall observed genetic similarities in
82T polymorphism, the genetic barrier was either lower for the drug resistance-related codons among HIV-1 subtypes can
subtype C or higher for subtype G, suggesting that non-B be explained by the biological relevance of such sites. In fact,
subtypes may require even more mutations than B subtypes to sequences containing major drug resistance-associated substi-
develop drug resistance. Moreover, specific polymorphisms in tutions show reduced fitness, which results in less efficient
protease positions may confer greater susceptibility to protease replication and spread of these viral populations (75, 128).
inhibitors to other non-B subtypes. In particular, subtype C
and CRF02_AG recombinant viruses from drug-nave patients
HIV-1 GENETIC DIVERSITY AND
show an in vitro hypersusceptibility to protease inhibitors (1,
VACCINE DEVELOPMENT
48), a finding which needs to be confirmed by in vivo clinical
studies. Transmission of HIV in the general population can possibly
Extensive differences between the subtypes were found for be blocked by an effective, safe, and affordable anti-HIV-1
the minor protease substitutions, which do not impair drug vaccine, which should induce a strong humoral, as well as
susceptibility but may affect the genetic pathway of resistance cellular, immune response against the whole array of HIV-1
once the virus generates a relevant major substitution (47, 83, genetic forms.
106). This finding implies that a faster emergence of drug The significant intra- and intersubtype variability of the
resistance to a particular protease inhibitor could be expected HIV-1 envelope protein (58, 64, 68, 70, 92, 109), the main
in some non-B subtypes. target of a preventive and possibly sterilizing vaccine approach,
In regard to nucleoside reverse transcriptase inhibitor com- together with the undeniable fiasco of the first clinical trial of
pounds, non-B subtypes do not show a lower genetic barrier in a phase III vaccine based on a monomeric gp120 (35, 50), has
any resistance-associated substitutions, indicating evolution to drastically driven the HIV-1 vaccine field in recent years to-
the drug-resistance phenotype at a rate comparable to that of ward the development of cytotoxic T-lymphocyte (CTL)-induc-
subtype B isolates. For NNRTI resistance-related substitu- ing vaccine approaches. This approach is also based on early
tions, the most relevant finding is the reduced genetic barrier observations of the strong correlation between the anti-HIV-1
for the V106M substitution in subtype C, which has been ob- CTL response and disease progression (67, 90, 95). Moreover,
served in different studies and which confers high-level resis- CTL responses can be raised against Gag and Pol proteins,
tance to all NNRTIs (16, 124, 125). In particular, the V106M which are less variable proteins, possibly circumventing the
mutation in RT is facilitated in subtype C by a single transition variability of the virus as well as the necessity of using HIV
VOL. 81, 2007 MINIREVIEW 10215

vaccine candidates that correspond to the HIV-1 strains prev- influence their antigenicity and immunogenicity. For example,
alent in the target population. out of five envelope consensus and ancestral sequences gener-
In support of this strategy, promising broad cross-clade cel- ated (30, 43, 65, 66, 72, 129), only one envelope consensus of
lular immunity against specific HIV-1 epitopes has been de- the group M sequences has been shown to elicit high titers of
scribed (27, 45, 51, 133); however, only a few epitopes are neutralizing antibodies effective against primary isolates from
conserved across different subtypes, and single amino acid sub- three different group M clades (72).
stitutions are selected by the CTL immune pressure, allowing
the virus to escape with a dramatic deterioration of clinical CONCLUDING REMARKS
conditions (8, 9). Moreover, several studies have identified
sequence variability within, as well as proximal to, character- The genetic complexity of the worldwide HIV-1 epidemic is
ized optimal epitopes, which can either modulate binding to still growing, with continuous molecular evolution of existing
the HLA molecule, reduce the binding affinity to the cognate subtypes and identification of new CRFs, as well as URFs. The
T-cell receptor, or interfere with efficient antigen processing, B subtype is still predominant in Western countries, with a
resulting in escape from CTL surveillance (4, 14, 31, 71). progressive introduction of non-B subtypes from countries
As a consequence, it is now currently believed that an anti- with higher levels of epidemic disease, while subtype C repre-
HIV-1 vaccine should elicit efficient cellular, as well as hu- sents the most prevalent form globally. A founder effect with a
moral, immune responses and, in particular, broadly anti-en- fast-colonization outcome, along with genetic, demographic,
velope neutralizing antibodies able to target the largest number economic, and social factors, appears to explain this epidemic
of HIV-1 genetic forms. Most of the envelope immunogens distribution of different subtypes. The available serological and
developed, however, have failed to achieve this objective (11, molecular diagnostic tools show levels of specificity and sensi-
23, 77). tivity appropriate for identifying the broad spectrum of circu-
Broadly neutralizing monoclonal antibodies targeting differ- lating genetic variants. The antiretroviral drugs currently used
ent epitopes in gp120 and gp41 have been obtained from nat- are equally active on all HIV-1 subtypes (with the exception of
urally infected individuals (for a comprehensive review, see group O viruses, which are resistant to NNRTIs), although
Srivastava et al., [119]), suggesting that envelope immunogens subtype-related differences in the rate of drug resistance emer-
able to raise such a broad neutralizing response can be gener- gence strongly suggest the need for HIV-1 presubtyping to
ated. However, polyspecific reactivity of some of these mono- guide selection of the appropriate therapeutic strategy. The
clonal antibodies with autoantigens has recently been reported, development of an effective anti-HIV-1 preventive vaccine
raising serious doubts about the possibility of eliciting such with broad efficacy against the vast array of HIV-1 genetic
broad anti-HIV humoral responses in vaccinees (2, 54). Nev- forms is still a work in progress. However, it is extremely
ertheless, using this concept, a few laboratories are currently relevant that vaccines targeted to subtypes other than B are
focusing on designing an HIV vaccine by following a working now being developed, a model which did not have many fans
backwards strategy, consisting of isolating and characterizing until a few years ago. This modified attitude, together with the
human/animal antibodies that can neutralize a broad range of growing body of information and better technologies, should
HIV strains and then using these antibodies to identify the accelerate the process of developing vaccines with the desired
HIV target regions to be incorporated into vaccines. The ef- broad immunoprotection efficacy. Efforts to meet the unprec-
fectiveness of such vaccine candidates in developing protective edented challenge to develop such vaccines, which is perceived
immunity will be tested in the near future. as a difficult-to-achieve goal, may benefit from understanding
In the meantime, several strategies have been developed to the exceptional cross-species transmission of SIV to humans.
engineer envelope immunogens presenting epitopes able to In conclusion, continuing studies of HIV-1 molecular phy-
induce neutralizing antibodies with broad efficacy against T- logenetics are necessary to trace the genetic evolution of the
cell-adapted as well as primary HIV-1 isolates (7, 18, 22, 32, 36, virus and to keep the diagnostic, preventive (vaccine), and
118, 132). therapeutic tools necessary to control the spread of the global
Furthermore, the genetic diversity of HIV-1 and the distri- HIV-1 epidemic up to date.
bution of different subtypes in each population is currently
being addressed by vaccine approaches based on either multi- ACKNOWLEDGMENTS
valent formulations or consensus and ancestral sequences. The This study was supported by grants from the Ministero Italiano della
first approach is based on using envelope molecules from dif- Sanita (Ricerca Corrente and Progetto Finalizzato AIDS 2006) and
ferent HIV-1 subtypes in order to induce antibodies with a the ICSC-World Lab, Lausanne, Switzerland (project MCD-2/7).
broader breadth of neutralizing activities (15, 24, 25, 49, 111, We are grateful to Marv Reitz (Institute of Human Virology, Bal-
112, 115). The second approach is based on computer-derived timore, MD) for his critical reading of the manuscript.
sequences (consensus sequences) generated by aligning cir- REFERENCES
culating primary isolates and selecting the most common nu-
1. Abecasis, A. B., K. Deforche, L. T. Bacheler, P. McKenna, A. P. Carvalho,
cleotide at each position. The resulting inferred genes are in an P. Gomes, A. M. Vandamme, and R. J. Camacho. 2006. Investigation of
intermediate position within an evolutionary tree, being equi- baseline susceptibility to protease inhibitors in HIV-1 subtypes C, F, G and
CRF02_AG. Antivir. Ther. 11:581589.
distant from the currently circulating isolates (30, 33, 44, 62, 2. Alam, S. M., M. McAdams, D. Boren, M. Rak, R. M. Scearce, F. Gao, Z. T.
94). However, these presumed consensus sequences are sub- Camacho, D. Gewirth, G. Kelsoe, P. Chen, and B. F. Haynes. 2007. The role
ject to sampling bias and may include polymorphisms, post- of antibody polyspecificity and lipid reactivity in binding of broadly neu-
tralizing anti-HIV-1 envelope human monoclonal antibodies 2F5 and 4E10
translational modifications (e.g., glycosylation) that do not re- to glycoprotein 41 membrane proximal envelope epitopes. J. Immunol.
flect the naturally spreading viruses, which may severely 178:44244435.
10216 MINIREVIEW J. VIROL.

3. Alexander, C. S., V. Montessori, B. Wynhoven, W. Dong, K. Chan, M. V. 19. Buonaguro, L., M. Tagliamonte, M. L. Tornesello, and F. M. Buonaguro.
OShaughnessy, T. Mo, M. Piaseczny, J. S. Montaner, and P. R. Harrigan. 2007. Evolution of the HIV-1 V3 region in the Italian epidemic. New
2002. Prevalence and response to antiretroviral therapy of non-B subtypes Microbiol. 30:111.
of HIV in antiretroviral-naive individuals in British Columbia. Antivir. 20. Buonaguro, L., M. Tagliamonte, M. L. Tornesello, and F. M. Buonaguro.
Ther. 7:3135. 2007. Genetic and phylogenetic evolution of HIV-1 in a low subtype het-
4. Allen, T. M., M. Altfeld, X. G. Yu, K. M. OSullivan, M. Lichterfeld, G. S. erogeneity epidemic: the Italian example. Retrovirology 4:34.
Le, M. John, B. R. Mothe, P. K. Lee, E. T. Kalife, D. E. Cohen, K. A. 21. Buonaguro, L., M. Tagliamonte, M. L. Tornesello, E. Pilotti, C. Casoli, A.
Freedberg, D. A. Strick, M. N. Johnston, A. Sette, E. S. Rosenberg, S. A. Lazzarin, G. Tambussi, M. Ciccozzi, G. Rezza, F. M. Buonaguro, et al.
Mallal, P. J. Goulder, C. Brander, and B. D. Walker. 2004. Selection, 2004. Screening of HIV-1 isolates by reverse heteroduplex mobility assay
transmission, and reversion of an antigen-processing cytotoxic T-lympho- and identification of non-B subtypes in Italy. J. Acquir. Immune Defic.
cyte escape mutation in human immunodeficiency virus type 1 infection. Syndr. 37:12951306.
J. Virol. 78:70697078. 22. Buonaguro, L., M. L. Visciano, M. L. Tornesello, M. Tagliamonte, B.
5. Avila, M. M., M. A. Pando, G. Carrion, L. M. Peralta, H. Salomon, M. G. Biryahwaho, and F. M. Buonaguro. 2005. Induction of systemic and mu-
Carrillo, J. Sanchez, S. Maulen, J. Hierholzer, M. Marinello, M. Negrete, cosal cross-clade neutralizing antibodies in BALB/c mice immunized with
K. L. Russell, and J. K. Carr. 2002. Two HIV-1 epidemics in Argentina: human immunodeficiency virus type 1 clade A virus-like particles adminis-
different genetic subtypes associated with different risk groups. J. Acquir. tered by different routes of inoculation. J. Virol. 79:70597067.
Immune Defic. Syndr. 29:422426. 23. Bures, R., A. Gaitan, T. Zhu, C. Graziosi, K. M. McGrath, J. Tartaglia, P.
6. Ayouba, A., S. Souquieres, B. Njinku, P. M. Martin, M. C. Muller-Trutwin, Caudrelier, R. El Habib, M. Klein, A. Lazzarin, D. M. Stablein, M. Deers,
P. Roques, F. Barre-Sinoussi, P. Mauclere, F. Simon, and E. Nerrienet. L. Corey, M. L. Greenberg, D. H. Schwartz, and D. C. Montefiori. 2000.
2000. HIV-1 group N among HIV-1-seropositive individuals in Cameroon. Immunization with recombinant canarypox vectors expressing membrane-
AIDS 14:26232625. anchored glycoprotein 120 followed by glycoprotein 160 boosting fails to
7. Barnett, S. W., S. Lu, I. Srivastava, S. Cherpelis, A. Gettie, J. Blanchard, S. generate antibodies that neutralize R5 primary isolates of human immu-
Wang, I. Mboudjeka, L. Leung, Y. Lian, A. Fong, C. Buckner, A. Ly, S. Hilt, nodeficiency virus type 1. AIDS Res. Hum. Retrovir. 16:20192035.
J. Ulmer, C. T. Wild, J. R. Mascola, and L. Stamatatos. 2001. The ability of 24. Catanzaro, A. T., R. A. Koup, M. Roederer, R. T. Bailer, M. E. Enama, Z.
an oligomeric human immunodeficiency virus type 1 (HIV-1) envelope Moodie, L. Gu, J. E. Martin, L. Novik, B. K. Chakrabarti, B. T. Butman,
antigen to elicit neutralizing antibodies against primary HIV-1 isolates is J. G. Gall, C. R. King, C. A. Andrews, R. Sheets, P. L. Gomez, J. R. Mascola,
improved following partial deletion of the second hypervariable region. G. J. Nabel, and B. S. Graham. 2006. Phase 1 safety and immunogenicity
J. Virol. 75:55265540. evaluation of a multiclade HIV-1 candidate vaccine delivered by a replica-
8. Barouch, D. H., J. Kunstman, J. Glowczwskie, K. J. Kunstman, M. A. Egan, tion-defective recombinant adenovirus vector. J. Infect. Dis. 194:1638
F. W. Peyerl, S. Santra, M. J. Kuroda, J. E. Schmitz, K. Beaudry, G. R. 1649.
Krivulka, M. A. Lifton, D. A. Gorgone, S. M. Wolinsky, and N. L. Letvin. 25. Chakrabarti, B. K., X. Ling, Z. Y. Yang, D. C. Montefiori, A. Panet, W. P.
2003. Viral escape from dominant simian immunodeficiency virus epitope- Kong, B. Welcher, M. K. Louder, J. R. Mascola, and G. J. Nabel. 2005.
specific cytotoxic T lymphocytes in DNA-vaccinated rhesus monkeys. J. Vi- Expanded breadth of virus neutralization after immunization with a multi-
rol. 77:73677375. clade envelope HIV vaccine candidate. Vaccine 23:34343445.
9. Barouch, D. H., J. Kunstman, M. J. Kuroda, J. E. Schmitz, S. Santra, F. W. 26. Clewley, J. P. 2004. Enigmas and paradoxes: the genetic diversity and
Peyerl, G. R. Krivulka, K. Beaudry, M. A. Lifton, D. A. Gorgone, D. C. prevalence of the primate lentiviruses. Curr. HIV Res. 2:113125.
Montefiori, M. G. Lewis, S. M. Wolinsky, and N. L. Letvin. 2002. Eventual 27. Coplan, P. M., S. B. Gupta, S. A. Dubey, P. Pitisuttithum, A. Nikas, B.
AIDS vaccine failure in a rhesus monkey by viral escape from cytotoxic T Mbewe, E. Vardas, M. Schechter, E. G. Kallas, D. C. Freed, T. M. Fu, C. T.
lymphocytes. Nature 415:335339. Mast, P. Puthavathana, J. Kublin, C. K. Brown, J. Chisi, R. Pendame, S. J.
10. Barre-Sinoussi, F., J. C. Chermann, F. Rey, M. T. Nugeyre, S. Chamaret, J. Thaler, G. Gray, J. Mcintyre, W. L. Straus, J. H. Condra, D. V. Mehrotra,
Gruest, C. Dauguet, C. Axler-Blin, F. Vezinet-Brun, C. Rouzioux, W. H. A. Guess, E. A. Emini, and J. W. Shiver. 2005. Cross-reactivity of
Rozenbaum, and L. Montagnier. 1983. Isolation of a T-lymphotropic ret- anti-HIV-1 T cell immune responses among the major HIV-1 clades in
rovirus from a patient at risk for acquired immune deficiency syndrome HIV-1-positive individuals from 4 continents. J. Infect. Dis. 191:14271434.
(AIDS). Science 220:868871. 28. Delaporte, E., W. Janssens, M. Peeters, A. Buve, G. Dibanga, J. L. Perret,
11. Beddows, S., S. Lister, R. Cheingsong, C. Bruck, and J. Weber. 1999. V. Ditsambou, J. R. Mba, M. C. Courbot, A. Georges, A. Bourgeois, B.
Comparison of the antibody repertoire generated in healthy volunteers Samb, D. Henzel, L. Heyndrickx, K. Fransen, G. van der Groen, and B.
following immunization with a monomeric recombinant gp120 construct Larouze. 1996. Epidemiological and molecular characteristics of HIV in-
derived from a CCR5/CXCR4-using human immunodeficiency virus type 1 fection in Gabon, 19861994. AIDS 10:903910.
isolate with sera from naturally infected individuals. J. Virol. 73:17401745. 29. Descamps, D., G. Collin, F. Letourneur, C. Apetrei, F. Damond, I. Loussert-
12. Beerenwinkel, N., M. Daumer, T. Sing, J. Rahnenfuhrer, T. Lengauer, J. Ajaka, F. Simon, S. Saragosti, and F. Brun-Vezinet. 1997. Susceptibility of
Selbig, D. Hoffmann, and R. Kaiser. 2005. Estimating HIV evolutionary human immunodeficiency virus type 1 group O isolates to antiretroviral agents:
pathways and the genetic barrier to drug resistance. J. Infect. Dis. 191:1953 in vitro phenotypic and genotypic analyses. J. Virol. 71:88938898.
1960. 30. Doria-Rose, N. A., G. H. Learn, A. G. Rodrigo, D. C. Nickle, F. Li, M.
13. Bocket, L., A. Cheret, S. Deuffic-Burban, P. Choisy, Y. Gerard, X. de la Mahalanabis, M. T. Hensel, S. McLaughlin, P. F. Edmonson, D. Monte-
Tribonniere, N. Viget, F. Ajana, A. Goffard, F. Barin, Y. Mouton, and Y. fiori, S. W. Barnett, N. L. Haigwood, and J. I. Mullins. 2005. Human
Yazdanpanah. 2005. Impact of human immunodeficiency virus type 1 sub- immunodeficiency virus type 1 subtype B ancestral envelope protein is
type on first-line antiretroviral therapy effectiveness. Antivir. Ther. 10:247 functional and elicits neutralizing antibodies in rabbits similar to those
254. elicited by a circulating subtype B envelope. J. Virol. 79:1121411224.
14. Brander, C., K. E. Hartman, A. K. Trocha, N. G. Jones, R. P. Johnson, B. 31. Draenert, R., G. S. Le, K. J. Pfafferott, A. J. Leslie, P. Chetty, C. Brander,
Korber, P. Wentworth, S. P. Buchbinder, S. Wolinsky, B. D. Walker, and E. C. Holmes, S. C. Chang, M. E. Feeney, M. M. Addo, L. Ruiz, D. Ram-
S. A. Kalams. 1998. Lack of strong immune selection pressure by the duth, P. Jeena, M. Altfeld, S. Thomas, Y. Tang, C. L. Verrill, C. Dixon, J. G.
immunodominant, HLA-A*0201-restricted cytotoxic T lymphocyte re- Prado, P. Kiepiela, J. Martinez-Picado, B. D. Walker, and P. J. Goulder.
sponse in chronic human immunodeficiency virus-1 infection. J. Clin. In- 2004. Immune selection for altered antigen processing leads to cytotoxic T
vestig. 101:25592566. lymphocyte escape in chronic HIV-1 infection. J. Exp. Med. 199:905915.
15. Brave, A., K. Ljungberg, A. Boberg, E. Rollman, M. Isaguliants, B. 32. Earl, P. L., W. Sugiura, D. C. Montefiori, C. C. Broder, S. A. Lee, C. Wild,
Lundgren, P. Blomberg, J. Hinkula, and B. Wahren. 2005. Multigene/ J. Lifson, and B. Moss. 2001. Immunogenicity and protective efficacy of
multisubtype HIV-1 vaccine induces potent cellular and humoral immune oligomeric human immunodeficiency virus type 1 gp140. J. Virol. 75:645
responses by needle-free intradermal delivery. Mol. Ther. 12:11971205. 653.
16. Brenner, B., D. Turner, M. Oliveira, D. Moisi, M. Detorio, M. Carobene, 33. Ellenberger, D. L., B. Li, L. D. Lupo, S. M. Owen, J. Nkengasong, M. S.
R. G. Marlink, J. Schapiro, M. Roger, and M. A. Wainberg. 2003. A V106M Kadio-Morokro, J. Smith, H. Robinson, M. Ackers, A. Greenberg, T. Folks,
mutation in HIV-1 clade C viruses exposed to efavirenz confers cross- and S. Butera. 2002. Generation of a consensus sequence from prevalent
resistance to non-nucleoside reverse transcriptase inhibitors. AIDS 17: and incident HIV-1 infections in West Africa to guide AIDS vaccine de-
F1F5. velopment. Virology 302:155163.
17. Buonaguro, L., E. Del Gaudio, M. Monaco, D. Greco, P. Corti, E. Beth- 34. Eshleman, S. H., G. Becker-Pergola, M. Deseyve, L. A. Guay, M. Mracna,
Giraldo, F. M. Buonaguro, and G. Giraldo. 1995. Heteroduplex mobility T. Fleming, S. Cunningham, P. Musoke, F. Mmiro, and J. B. Jackson. 2001.
assay and phylogenetic analysis of V3 region sequences of HIV 1 isolates Impact of human immunodeficiency virus type 1 (HIV-1) subtype on
from Gulu, Northern Uganda. J. Virol. 69:79717981. women receiving single-dose nevirapine prophylaxis to prevent HIV-1 ver-
18. Buonaguro, L., L. Racioppi, M. L. Tornesello, C. Arra, M. L. Visciano, B. tical transmission. J. Infect. Dis. 184:914917.
Biryahwaho, S. D. K. Sempala, G. Giraldo, and F. M. Buonaguro. 2002. 35. Flynn, N. M., D. N. Forthal, C. D. Harro, F. N. Judson, K. H. Mayer, and
Induction of neutralizing antibodies and cytotoxic T lymphocytes in Balb/c M. F. Para. 2005. Placebo-controlled phase 3 trial of a recombinant glyco-
mice immunized with virus-like particles presenting a gp120 molecule from protein 120 vaccine to prevent HIV-1 infection. J. Infect. Dis. 191:654665.
a HIV-1 isolate of clade A. Antivir. Res. 54:189201. 36. Fouts, T., K. Godfrey, K. Bobb, D. Montefiori, C. V. Hanson, V. S. Kaly-
VOL. 81, 2007 MINIREVIEW 10217

anaraman, A. Devico, and R. Pal. 2002. Crosslinked HIV-1 envelope-CD4 Markowitz. 1995. Rapid turnover of plasma virions and CD4 lymphocytes
receptor complexes elicit broadly cross-reactive neutralizing antibodies in in HIV-1 infections. Nature 373:123126.
rhesus macaques. Proc. Natl. Acad. Sci. USA 99:1184211847. 57. Hooper, E. 2000. Search for the origin of HIV and AIDS. Science 289:
37. Frater, A. J., D. T. Dunn, A. J. Beardall, K. Ariyoshi, J. R. Clarke, M. O. 11401141.
McClure, and J. N. Weber. 2002. Comparative response of African HIV- 58. Janssens, W., L. Heyndrickx, K. Fransen, J. Motte, M. Peeters, J. N.
1-infected individuals to highly active antiretroviral therapy. AIDS 16:1139 Nkengasong, P. M. Ndumbe, E. Delaporte, J. L. Perret, C. Atende, P. Piot,
1146. and G. van der Groen. 1994. Genetic and phylogenetic analysis of env
38. Gallo, R. C., P. S. Sarin, E. P. Gelmann, M. Robert-Guroff, E. Richardson, subtypes G and H in Central Africa. AIDS Res. Hum. Retrovir. 10:877879.
V. S. Kalyanaraman, D. Mann, G. D. Sidhu, R. E. Stahl, S. Zolla-Pazner, 59. Janssens, W., J. N. Nkengasong, L. Heyndrickx, K. Fransen, P. M.
J. Leibowitch, and M. Popovic. 1983. Isolation of human T-cell leukemia Ndumbe, E. Delaporte, M. Peeters, J. L. Perret, A. Ndoumou, C. Atende, P.
virus in acquired immune deficiency syndrome (AIDS). Science 220:865 Piot, and G. van der Groen. 1994. Further evidence of the presence of
867. genetically very aberrant HIV-1 strains in Cameroon and Gabon. AIDS
39. Gao, F., E. Bailes, D. L. Robertson, Y. Chen, C. M. Rodenburg, S. F. 8:10121013.
Michael, L. B. Cummins, L. O. Arthur, M. Peeters, G. M. Shaw, P. M. 60. Kalish, M. L., N. D. Wolfe, C. B. Ndongmo, J. McNicholl, K. E. Robbins, M.
Sharp, and B. H. Hahn. 1999. Origin of HIV-1 in the chimpanzee Pan Aidoo, P. N. Fonjungo, G. Alemnji, C. Zeh, C. F. Djoko, E. Mpoudi-Ngole,
troglodytes troglodytes. Nature 397:436441. D. S. Burke, and T. M. Folks. 2005. Central African hunters exposed to
40. Gao, F., S. G. Morrison, D. L. Robertson, C. L. Thornton, S. Craig, G. simian immunodeficiency virus. Emerg. Infect. Dis. 11:19281930.
Karlsson, J. Sodroski, M. Morgado, B. Galvao-Castro, H. von Briesen, S. 61. Kantor, R., D. A. Katzenstein, B. Efron, A. P. Carvalho, B. Wynhoven, P.
Beddows, J. Weber, P. M. Sharp, G. M. Shaw, B. H. Hahn, et al. 1996. Cane, J. Clarke, S. Sirivichayakul, M. A. Soares, J. Snoeck, C. Pillay, H.
Molecular cloning and analysis of functional envelope genes from human Rudich, R. Rodrigues, A. Holguin, K. Ariyoshi, M. B. Bouzas, P. Cahn,
deficiency virus type 1 sequence subtypes A through G. J. Virol. 70:1651 W. Sugiura, V. Soriano, L. F. Brigido, Z. Grossman, L. Morris, A.-M.
1667. Vandamme, A. Tanuri, P. Phanuphak, J. N. Weber, D. Pillay, P. R. Harrigan,
41. Gao, F., D. L. Robertson, S. Morrison, H. Hui, S. Craig, J. M. Decker, P. N. R. Camacho, J. M. Schapiro, and R. W. Shafer. 2005. Impact of HIV-1
Fultz, M. Girard, G. M. Shaw, B. H. Hahn, and P. M. Sharp. 1996. The subtype and antiretroviral therapy on protease and reverse transcriptase
heterosexual human immunodeficiency virus type 1 epidemic in Thailand is genotype: results of a global collaboration. PLoS Med. 2:e112.
caused by an intersubtype (A/E) recombinant of African origin. J. Virol. 62. Korber, B., B. Gaschen, K. Yusim, R. Thakallapally, C. Kesmir, and V.
70:70137029. Detours. 2001. Evolutionary and immunological implications of contempo-
42. Gao, F., N. Vidal, Y. Li, S. A. Trask, Y. Chen, L. G. Kostrikis, D. D. Ho, J. rary HIV-1 variation. Br. Med. Bull. 58:1942.
Kim, M.-D. Oh, K. Choe, M. Salminen, D. L. Robertson, G. M. Shaw, B. H. 63. Korber, B., M. Muldoon, J. Theiler, F. Gao, R. Gupta, A. Lapedes, B. H.
Hahn, and M. Peeters. 2001. Evidence of two distinct subsubtypes within Hahn, S. Wolinsky, and T. Bhattacharya. 2000. Timing the ancestor of the
the HIV-1 subtype A radiation. AIDS Res. Hum. Retrovir. 17:675688. HIV-1 pandemic strains. Science 288:17891796.
43. Gao, F., E. A. Weaver, Z. Lu, Y. Li, H.-X. Liao, B. Ma, S. M. Alam, R. M.
64. Kostrikis, L. G., E. Bagdades, Y. Cao, L. Zhang, D. Dimitriou, and D. D.
Scearce, L. L. Sutherland, J.-S. Yu, J. M. Decker, G. M. Shaw, D. C.
Ho. 1995. Genetic analysis of human immunodeficiency virus type 1 strains
Montefiori, B. T. Korber, B. H. Hahn, and B. F. Haynes. 2005. Antigenicity
from patients in Cyprus: identification of a new subtype designated subtype
and immunogenicity of a synthetic human immunodeficiency virus type 1
I. J. Virol. 69:61226130.
group M consensus envelope glycoprotein. J. Virol. 79:11541163.
65. Kothe, D. L., J. M. Decker, Y. Li, Z. Weng, F. Bibollet-Ruche, K. P. Zammit,
44. Gaschen, B., J. Taylor, K. Yusim, B. Foley, F. Gao, D. Lang, V. Novitsky, B.
M. G. Salazar, Y. Chen, J. F. Salazar-Gonzalez, Z. Moldoveanu, J.
Haynes, B. H. Hahn, T. Bhattacharya, and B. Korber. 2002. Diversity
Mestecky, F. Gao, B. F. Haynes, G. M. Shaw, M. Muldoon, B. T. Korber,
considerations in HIV-1 vaccine selection. Science 296:23542360.
and B. H. Hahn. 2007. Antigenicity and immunogenicity of HIV-1 consen-
45. Geels, M. J., S. A. Dubey, K. Anderson, E. Baan, M. Bakker, G. Pollakis,
sus subtype B envelope glycoproteins. Virology 360:218234.
W. A. Paxton, J. W. Shiver, and J. Goudsmit. 2005. Broad cross-clade T-cell
responses to Gag in individuals infected with human immunodeficiency 66. Kothe, D. L., Y. Li, J. M. Decker, F. Bibollet-Ruche, K. P. Zammit, M. G.
virus type 1 non-B clades (A to G): importance of HLA anchor residue Salazar, Y. Chen, Z. Weng, E. A. Weaver, F. Gao, B. F. Haynes, G. M. Shaw,
conservation. J. Virol. 79:1124711258. B. T. Korber, and B. H. Hahn. 2006. Ancestral and consensus envelope
46. Goldberg, B., and R. B. Stricker. 2000. Bridging the gap: human diploid cell immunogens for HIV-1 subtype C. Virology 352:438449.
strains and the origin of AIDS. J. Theor. Biol. 204:497503. 67. Koup, R. A., J. T. Safrit, Y. Cao, C. A. Andrews, G. McLeod, W. Borkowsky,
47. Gonzalez, L. M. F., R. M. Brindeiro, R. S. Aguiar, H. S. Pereira, C. M. C. Farthing, and D. D. Ho. 1994. Temporal association of cellular immune
Abreu, M. A. Soares, and A. Tanuri. 2004. Impact of nelfinavir resistance responses with the initial control of viremia in primary human immunode-
mutations on in vitro phenotype, fitness, and replication capacity of human ficiency virus type 1 syndrome. J. Virol. 68:46504655.
immunodeficiency virus type 1 with subtype B and C proteases. Antimicrob. 68. Kuiken, C., B. Foley, B. H. Hahn, F. E. McCutchan, J. W. Mellors, J. I.
Agents Chemother. 48:35523555. Mullins, J. Sodroski, S. Wolinsky, and B. Korber. 2000. Human retrovi-
48. Gonzalez, L. M. F., R. M. Brindeiro, M. Tarin, A. Calazans, M. A. Soares, ruses and AIDS 2000: a compilation and analysis of nucleic acids and amino
S. Cassol, and A. Tanuri. 2003. In vitro hypersusceptibility of human im- acid sequences. Theoretical Biology and Biophysics Group, Los Alamos
munodeficiency virus type 1 subtype C protease to lopinavir. Antimicrob. National Laboratory, Los Alamos, New Mexico.
Agents Chemother. 47:28172822. 69. Kwon, J. A., S. Y. Yoon, C. K. Lee, C. S. Lim, K. N. Lee, H. J. Sung, C. A.
49. Graham, B. S., R. A. Koup, M. Roederer, R. T. Bailer, M. E. Enama, Z. Brennan, and S. G. Devare. 2006. Performance evaluation of three auto-
Moodie, J. E. Martin, M. M. McCluskey, B. K. Chakrabarti, L. Lamoreaux, mated human immunodeficiency virus antigen-antibody combination im-
C. A. Andrews, P. L. Gomez, J. R. Mascola, and G. J. Nabel. 2006. Phase 1 munoassays. J. Virol. Methods 133:2026.
safety and immunogenicity evaluation of a multiclade HIV-1 DNA candi- 70. Leitner, T., A. Alaeus, S. Marquina, E. Lilja, K. Lidman, and J. Albert.
date vaccine. J. Infect. Dis. 194:16501660. 1995. Yet another subtype of HIV type 1? AIDS Res. Hum. Retrovir.
50. Graham, B. S., and J. R. Mascola. 2005. Lessons from failurepreparing 11:995997.
for future HIV-1 vaccine efficacy trials. J. Infect. Dis. 191:647649. 71. Leslie, A. J., K. J. Pfafferott, P. Chetty, R. Draenert, M. M. Addo, M.
51. Gupta, S. B., C. T. Mast, N. D. Wolfe, V. Novitsky, S. A. Dubey, E. G. Kallas, Feeney, Y. Tang, E. C. Holmes, T. Allen, J. G. Prado, M. Altfeld, C.
M. Schechter, B. Mbewe, E. Vardas, P. Pitisuttithum, D. Burke, D. Freed, Brander, C. Dixon, D. Ramduth, P. Jeena, S. A. Thomas, A. St. John, T. A.
R. Mogg, P. M. Coplan, J. H. Condra, R. S. Long, K. Anderson, D. R. Roach, B. Kupfer, G. Luzzi, A. Edwards, G. Taylor, H. Lyall, G. Tudor-
Casimiro, J. W. Shiver, and W. L. Straus. 2006. Cross-clade reactivity of Williams, V. Novelli, J. Martinez-Picado, P. Kiepiela, B. D. Walker, and
HIV-1-specific T-cell responses in HIV-1-infected individuals from P. J. Goulder. 2004. HIV evolution: CTL escape mutation and reversion
Botswana and Cameroon. J. Acquir. Immune. Defic. Syndr. 42:135139. after transmission. Nat. Med. 10:282289.
52. Gurtler, L., J. Eberle, A. von Brunn, S. Knapp, H. P. Hauser, L. Zekeng, 72. Liao, H. X., L. L. Sutherland, S. M. Xia, M. E. Brock, R. M. Scearce, S.
J. M. Tsague, E. Selegny, and L. Kaptue. 1994. A new subtype of human Vanleeuwen, S. M. Alam, M. McAdams, E. A. Weaver, Z. Camacho, B. J.
immunodeficiency virus type 1 (MVP-5180) from Cameroon. J. Virol. 68: Ma, Y. Li, J. M. Decker, G. J. Nabel, D. C. Montefiori, B. H. Hahn, B. T.
15811585. Korber, F. Gao, and B. F. Haynes. 2006. A group M consensus envelope
53. Hahn, B. H., G. M. Shaw, K. M. De Cock, and P. M. Sharp. 2000. AIDS as glycoprotein induces antibodies that neutralize subsets of subtype B and C
a zoonosis: scientific and public health implications. Science 287:607614. HIV-1 primary viruses. Virology 353:268282.
54. Haynes, B. F., J. Fleming, E. W. St. Clair, H. Katinger, G. Stiegler, R. 73. Loemba, H., B. Brenner, M. A. Parniak, S. Maayan, B. Spira, D. Moisi, M.
Kunert, J. Robinson, R. M. Scearce, K. Plonk, H. F. Staats, T. L. Ortel, Oliveira, M. Detorio, and M. A. Wainberg. 2002. Genetic divergence of
H. X. Liao, and S. M. Alam. 2005. Cardiolipin polyspecific autoreactivity in human immunodeficiency virus type 1 Ethiopian clade C reverse transcrip-
two broadly neutralizing HIV-1 antibodies. Science 308:19061908. tase (RT) and rapid development of resistance against nonnucleoside in-
55. Hemelaar, J., E. Gouws, P. D. Ghys, and S. Osmanov. 2006. Global and hibitors of RT. Antimicrob. Agents Chemother. 46:20872094.
regional distribution of HIV-1 genetic subtypes and recombinants in 2004. 74. Louwagie, J., F. E. McCutchan, M. Peeters, T. P. Brennan, E. Sanders-
AIDS 20:W13W23. Buell, G. A. Eddy, G. van der Groen, K. Fransen, G. M. Gershy-Damet, R.
56. Ho, D. D., A. U. Neumann, A. S. Perelson, J. Chen, J. M. Leonard, and M. De Leys, and D. S. Burke. 1993. Phylogenetic analysis of gag genes from 70
10218 MINIREVIEW J. VIROL.

international HIV-1 isolates provides evidence for multiple genotypes. of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA.
AIDS 7:769780. Science 279:21032106.
75. Lucas, G. M. 2005. Antiretroviral adherence, drug resistance, viral fitness 96. Op de Coul, E. L. M., M. Prins, M. Cornelissen, A. van der Schoot, F.
and HIV disease progression: a tangled web is woven. J. Antimicrob. Che- Boufassa, R. P. Brettle, I. Hernandez-Aguado, V. Schiffer, J. McMenamin,
mother. 55:413416. G. Rezza, R. Robertson, R. Zangerle, J. Goudsmit, R. A. Coutinho, and V.
76. Ly, T. D., S. Laperche, C. Brennan, A. Vallari, A. Ebel, J. Hunt, L. Martin, Lukashov. 2001. Using phylogenetic analysis to trace HIV-1 migration
D. Daghfal, G. Schochetman, and S. Devare. 2004. Evaluation of the sen- among western European injecting drug users seroconverting from 1984 to
sitivity and specificity of six HIV combined p24 antigen and antibody assays. 1997. AIDS 15:257266.
J. Virol. Methods 122:185194. 97. Osmanov, S., C. Pattou, N. Walker, B. Schwardlander, J. Esparza, et al.
77. Mascola, J. R., S. W. Snyder, O. S. Weislow, S. M. Belay, R. B. Belshe, D. H. 2002. Estimated global distribution and regional spread of HIV-1 genetic
Schwartz, M. L. Clements, R. Dolin, B. S. Graham, G. J. Gorse, M. C. subtypes in the year 2000. J. Acquir. Immune Defic. Syndr. 29:184190.
Keefer, M. J. McElrath, M. C. Walker, K. F. Wagner, J. G. McNeil, F. E. 98. Ou, C. Y., Y. Takebe, C. C. Luo, M. Kalish, W. Auwanit, C. Bandea, N. de
McCutchan, and D. S. Burke. 1996. Immunization with envelope subunit la Torre, J. L. Moore, G. Schochetman, and S. Yamazaki. 1992. Wide
vaccine products elicits neutralizing antibodies against laboratory-adapted distribution of two subtypes of HIV-1 in Thailand. AIDS Res. Hum. Ret-
but not primary isolates of human immunodeficiency virus type 1. J. Infect. rovir. 8:14711472.
Dis. 173:340348. 99. Peeters, M. 2001. Recombinant HIV sequences: their role in the global
78. Mauclere, P., I. Loussert-Ajaka, F. Damond, P. Fagot, S. Souquieres, L. M. epidemic, p. 5472. In C. Kuiken, B. Foley, B. Hahn, F. E. McCutchan,
Monny, F. X. Mbopi Keou, F. Barre-Sinoussi, S. Saragosti, F. Brun-Vezinet, J. W. Mellors, J. I. Mullins, J. Sodroski, S. Wolinsky, and B. Korber (ed.),
and F. Simon. 1997. Serological and virological characterization of HIV-1 HIV sequence compendium 2000. Theoretical Biology and Biophysics
group O infection in Cameroon. AIDS 11:445453. Group, Los Alamos National Laboratory, Los Alamos, NM.
79. McCutchan, F. E. 2006. Global epidemiology of HIV. J. Med. Virol. 100. Peeters, M., and V. Courgnaud. 2002. Overview of primate lentiviruses and
78(Suppl. 1):S7S12. their evolution in non-human primates in Africa, p. 223. In C. Kuiken, B.
80. McCutchan, F. E., J. K. Carr, M. Bajani, E. Sanders-Buell, T. O. Harry, Foley, E. Freed, B. Hahn, B. Korber, P. A. Marx, F. E. McCutchan, J. W.
T. C. Stoeckli, K. E. Robbins, W. Gashau, A. Nasidi, W. Janssens, and M. L. Mellors, and S. Wolinsky (ed.), HIV sequence compendium 2002. Theo-
Kalish. 1999. Subtype G and multiple forms of A/G intersubtype recombi- retical Biology and Biophysics Group, Los Alamos National Laboratory,
nant human immunodeficiency virus type 1 in Nigeria. Virology 254:226 Los Alamos, NM.
234. 101. Peeters, M., A. Gaye, S. Mboup, W. Badombena, K. Bassabi, M. Prince-
81. McCutchan, F. E., P. A. Hegerick, T. P. Brennan, P. Phanupak, P. Sing- David, M. Develoux, F. Liegeois, G. van der Groen, E. Saman, and E.
haraj, A. Jugsude, P. Berman, A. M. Gray, A. K. Fowler, and D. S. Burke. Delaporte. 1996. Presence of HIV-1 group O infection in West Africa.
1992. Genetic variants of HIV-1 in Thailand. AIDS Res. Hum. Retrovir. AIDS 10:343344.
8:18871895. 102. Peeters, M., A. Gueye, S. Mboup, F. Bibollet-Ruche, E. Ekaza, C. Mulanga,
82. Menu, E., T. X. Truong, M. E. Lafon, T. H. Nguyen, M. C. Muller-Trutwin, R. Ouedrago, R. Gandji, P. Mpele, G. Dibanga, B. Koumare, M. Saidou, E.
T. T. Nguyen, A. Deslandres, G. Chaouat, Q. T. Duong, B. K. Ha, H. J. Esu-Williams, J. P. Lombart, W. Badombena, N. Luo, H. M. Vanden, and
Fleury, and F. Barre-Sinoussi. 1996. HIV type 1 Thai subtype E is pre- E. Delaporte. 1997. Geographical distribution of HIV-1 group O viruses in
dominant in South Vietnam. AIDS Res. Hum. Retrovir. 12:629633. Africa. AIDS 11:493498.
83. Menzo, S., A. Castagna, A. Monachetti, H. Hasson, A. Danise, E. Carini, P. 103. Pillay, D., A. S. Walker, D. M. Gibb, A. de Rossi, S. Kaye, M. Ait-Khaled,
Bagnarelli, A. Lazzarin, and M. Clementi. 2004. Resistance and replicative M. Munoz-Fernandez, and A. Babiker. 2002. Impact of human immunode-
capacity of HIV-1 strains selected in vivo by long-term enfuvirtide treat- ficiency virus type 1 subtypes on virologic response and emergence of drug
ment. New Microbiol. 27:5161. resistance among children in the Paediatric European Network for Treat-
84. Michael, N. L. 1999. Host genetic influences on HIV-1 pathogenesis. Curr. ment of AIDS (PENTA) 5 trial. J. Infect. Dis. 186:617625.
Opin. Immunol. 11:466474. 104. Piyasirisilp, S., F. E. McCutchan, J. K. Carr, E. Sanders-Buell, W. Liu,
85. Mokili, J. L., M. Rogers, J. K. Carr, P. Simmonds, J. M. Bopopi, B. T. J. Chen, R. Wagner, H. Wolf, Y. Shao, S. Lai, C. Beyrer, and X.-F. Yu. 2000.
Foley, B. T. Korber, D. L. Birx, and F. E. McCutchan. 2002. Identification A recent outbreak of human immunodeficiency virus type 1 infection in
of a novel clade of human immunodeficiency virus type 1 in Democratic southern China was initiated by two highly homogeneous, geographically
Republic of Congo. AIDS Res. Hum. Retrovir. 18:817823. separated strains, circulating recombinant form AE and a novel BC recom-
86. Mokili, J. L., C. M. Wade, S. M. Burns, W. A. Cutting, J. M. Bopopi, S. D. binant. J. Virol. 74:1128611295.
Green, J. F. Peutherer, and P. Simmonds. 1999. Genetic heterogeneity of 105. Popovic, M., M. G. Sarngadharan, E. Read, and R. C. Gallo. 1984. Detec-
HIV type 1 subtypes in Kimpese, rural Democratic Republic of Congo. tion, isolation, and continuous production of cytopathic retroviruses
AIDS Res. Hum. Retrovir. 15:655664. (HTLV-III) from patients with AIDS and pre-AIDS. Science 224:497500.
87. Montavon, C., C. Toure-Kane, F. Lieggeois, E. Mpoudi, A. Bourgeois, L. 106. Prado, J. G., S. Franco, T. Matamoros, L. Ruiz, B. Clotet, L. Menendez-
Vergne, J. L. Perret, A. Boumah, E. Saman, S. Mboup, E. Delaporte, and Arias, M. A. Martinez, and J. Martinez-Picado. 2004. Relative replication
M. Peeters. 2000. Most env and gag subtype A HIV-1 viruses circulating in fitness of multi-nucleoside analogue-resistant HIV-1 strains bearing a
West and West Central Africa are similar to the prototype AG recombinant dipeptide insertion in the fingers subdomain of the reverse transcriptase
virus IBNG. J. Acquir. Immune Defic. Syndr. 23:363374. and mutations at codons 67 and 215. Virology 326:103112.
88. Montavon, C., C. Toure-Kane, J. N. Nkengasong, L. Vergne, K. Hertogs, S. 107. Quinones-Mateu, M. E., J. L. Albright, A. Mas, V. Soriano, and E. J. Arts.
Mboup, E. Delaporte, and M. Peeters. 2002. CRF06-cpx: a new circulating 1998. Analysis of pol gene heterogeneity, viral quasispecies, and drug re-
recombinant form of HIV-1 in West Africa involving subtypes A, G, K, and sistance in individuals infected with group O strains of human immunode-
J. J. Acquir. Immune. Defic. Syndr. 29:522530. ficiency virus type 1. J. Virol. 72:90029015.
89. Motomura, K., S. Kusagawa, K. Kato, K. Nohtomi, H. H. Lwin, K. M. Tun, 108. Roberts, J. D., K. Bebenek, and T. A. Kunkel. 1988. The accuracy of reverse
M. Thwe, K. Y. Oo, S. Lwin, O. Kyaw, M. Zaw, Y. Nagai, and Y. Takebe. transcriptase from HIV-1. Science 242:11711173.
2000. Emergence of new forms of human immunodeficiency virus type 1 109. Robertson, D. L., J. P. Anderson, J. A. Bradac, J. K. Carr, B. Foley, R. K.
intersubtype recombinants in central Myanmar. AIDS Res. Hum. Retrovir. Funkhouser, F. Gao, B. H. Hahn, M. Kalish, C. Kuiken, G. H. Learn, T.
17:18311843. Leitner, F. E. McCutchan, S. Osmanov, M. Peeters, D. Pieniazek, M.
90. Musey, L., J. Hughes, T. Schacker, T. Shea, L. Corey, and M. J. McElrath. Salminen, P. M. Sharp, S. Wolinsky, and B. Korber. 2000. HIV-1 nomen-
1997. Cytotoxic-T-cell responses, viral load, and disease progression in early clature proposal. Science 288:5556.
human immunodeficiency virus type 1 infection. N. Engl. J. Med. 337:1267 110. Robertson, D. L., J. P. Anderson, J. A. Bradac, J. K. Carr, B. Foley, R. K.
1274. Funkhouser, F. Gao, B. H. Hahn, M. L. Kalish, C. Kuiken, G. H. Learn, T.
91. Myers, G. 1994. Tenth anniversary perspectives on AIDS. HIV: between Leitner, F. E. McCutchan, S. Osmanov, M. Peeters, D. Pieniazek, M.
past and future. AIDS Res. Hum. Retrovir. 10:13171324. Salminen, P. M. Sharp, S. Wolinsky, and B. Korber. 2000. HIV-1 nomen-
92. Myers, G., K. MacInnes, and B. Korber. 1992. The emergence of simian/ clature proposal, p. 492505. In C. Kuiken, B. Foley, B. H. Hahn, B. Korber,
human immunodeficiency viruses. AIDS Res. Hum. Retrovir. 8:373386. F. E. McCutchan, P. A. Marx, J. W. Mellors, J. I. Mullins, J. Sodroski, and
93. Naderi, H. R., M. Tagliamonte, M. L. Tornesello, M. Ciccozzi, G. Rezza, R. S. Wolinsky (ed.), Human retroviruses and AIDS, 1999: a compilation and
Farid, F. M. Buonaguro, and L. Buonaguro. 2006. Molecular and phyloge- analysis of nucleic acid and amino acid sequences. Los Alamos National
netic analysis of HIV-1 variants circulating among injecting drug users in Laboratory, Los Alamos, NM.
Mashhad-Iran. Infect. Agents Cancer 1:4. 111. Rollman, E., A. Brve, A. Boberg, L. Gudmundsdotter, G. Engstrom, M.
94. Nickle, D. C., M. A. Jensen, G. S. Gottlieb, D. Shriner, G. H. Learn, A. G. Isaguliants, K. Ljungberg, B. Lundgren, P. Blomberg, J. Hinkula, B.
Rodrigo, and J. I. Mullins. 2003. Consensus and ancestral state HIV vac- Hejdeman, E. Sandstrom, M. Liu, and B. Wahren. 2005. The rationale
cines. Science 299:15151518. behind a vaccine based on multiple HIV antigens. Microbes Infect. 7:1414
95. Ogg, G. S., X. Jin, S. Bonhoeffer, P. R. Dunbar, M. A. Nowak, S. Monard, 1423.
J. P. Segal, Y. Cao, S. L. Rowland-Jones, V. Cerundolo, A. Hurley, M. 112. Rollman, E., J. Hinkula, J. Arteaga, B. Zuber, A. Kjerrstrom, M. Liu, B.
Markowitz, D. D. Ho, D. F. Nixon, and A. J. McMichael. 1998. Quantitation Wahren, and K. Ljungberg. 2004. Multi-subtype gp160 DNA immunization
VOL. 81, 2007 MINIREVIEW 10219

induces broadly neutralizing anti-HIV antibodies. Gene Ther. 11:1146 Mori, M. Essex, S. Maayan, and M. A. Wainberg. 2004. Nucleotide and
1154. amino acid polymorphisms at drug resistance sites in non-B-subtype vari-
113. Salemi, M., K. Strimmer, W. W. Hall, M. Duffy, E. Delaporte, S. Mboup, M. ants of human immunodeficiency virus type 1. Antimicrob. Agents Che-
Peeters, and A. M. Vandamme. 2001. Dating the common ancestor of mother. 48:29932998.
SIVcpz and HIV-1 group M and the origin of HIV-1 subtypes using a new 125. van de Vijver, D. A., A. M. Wensing, G. Angarano, B. Asjo, C. Balotta, E.
method to uncover clock-like molecular evolution. FASEB J. 15:276278. Boeri, R. Camacho, M. L. Chaix, D. Costagliola, A. De Luca, I. Derdelinckx,
114. Sarngadharan, M. G., M. Popovic, L. Bruch, J. Schupbach, and R. C. Gallo. Z. Grossman, O. Hamouda, A. Hatzakis, R. Hemmer, A. Hoepelman, A.
1984. Antibodies reactive with human T-lymphotropic retroviruses (HTLV- Horban, K. Korn, C. Kucherer, T. Leitner, C. Loveday, E. MacRae, I.
III) in the serum of patients with AIDS. Science 224:506508. Maljkovic, C. de Mendoza, L. Meyer, C. Nielsen, E. L. Op de Coul, V.
115. Seaman, M. S., L. Xu, K. Beaudry, K. L. Martin, M. H. Beddall, A. Miura, Ormaasen, D. Paraskevis, L. Perrin, E. Puchhammer-Stockl, L. Ruiz, M.
A. Sambor, B. K. Chakrabarti, Y. Huang, R. Bailer, R. A. Koup, J. R. Salminen, J. C. Schmit, F. Schneider, R. Schuurman, V. Soriano, G. Stanc-
Mascola, G. J. Nabel, and N. L. Letvin. 2005. Multiclade human immuno- zak, M. Stanojevic, A. M. Vandamme, K. Van Laethem, M. Violin, K. Wilbe,
deficiency virus type 1 envelope immunogens elicit broad cellular and hu- S. Yerly, M. Zazzi, and C. A. Boucher. 2006. The calculated genetic barrier
moral immunity in rhesus monkeys. J. Virol. 79:29562963. for antiretroviral drug resistance substitutions is largely similar for different
116. Simon, F., P. Mauclere, P. Roques, I. Loussert-Ajaka, M. C. Muller- HIV-1 subtypes. J. Acquir. Immune Defic. Syndr. 41:352360.
Trutwin, S. Saragosti, M. C. Georges-Courbot, F. Barre-Sinoussi, and F. 126. van Harmelen, J., R. Wood, M. Lambrick, E. P. Rybicki, A. L. Williamson,
Brun-Vezinet. 1998. Identification of a new human immunodeficiency virus and C. Williamson. 1997. An association between HIV-1 subtypes and
type 1 distinct from group M and group O. Nat. Med. 4:10321037. mode of transmission in Cape Town, South Africa. AIDS 11:8187.
117. Spira, S., M. A. Wainberg, H. Loemba, D. Turner, and B. G. Brenner. 2003. 127. Vidal, N., M. Peeters, C. Mulanga-Kabeya, N. Nzilambi, D. L. Robertson,
Impact of clade diversity on HIV-1 virulence, antiretroviral drug sensitivity W. Ilunga, H. Sema, K. Tshimanga, B. Bongo, and E. Delaporte. 2000.
and drug resistance. J. Antimicrob. Chemother. 51:229240. Unprecedented degree of human immunodeficiency virus type 1 (HIV-1)
118. Srivastava, I. K., L. Stamatatos, E. Kan, M. Vajdy, Y. Lian, S. Hilt, L. group M genetic diversity in the Democratic Republic of Congo suggests
Martin, C. Vita, P. Zhu, K. H. Roux, L. Vojtech, C. Montefiori, J. Donnelly, that the HIV-1 pandemic originated in central Africa. J. Virol. 74:10498
J. B. Ulmer, and S. W. Barnett. 2003. Purification, characterization, and 10507.
immunogenicity of a soluble trimeric envelope protein containing a partial 128. Wainberg, M. A. 2004. The impact of the M184V substitution on drug
deletion of the V2 loop derived from SF162, an R5-tropic human immu- resistance and viral fitness. Expert Rev. Anti Infect. Ther. 2:147151.
nodeficiency virus type 1 isolate. J. Virol. 77:1124411259. 129. Weaver, E. A., Z. Lu, Z. T. Camacho, F. Moukdar, H.-X. Liao, B.-J. Ma, M.
119. Srivastava, I. K., J. B. Ulmer, and S. W. Barnett. 2005. Role of neutralizing Muldoon, J. Theiler, G. J. Nabel, N. L. Letvin, B. T. Korber, B. H. Hahn,
antibodies in protective immunity against HIV. Hum. Vaccines 1:4560. B. F. Haynes, and F. Gao. 2006. Cross-subtype T-cell immune responses
120. Swanson, P., S. Huang, K. Abravaya, C. de Mendoza, V. Soriano, S. G. Devare, induced by a human immunodeficiency virus type 1 group M consensus Env
and J. Hackett, Jr. 2007. Evaluation of performance across the dynamic range immunogen. J. Virol. 80:67456756.
of the Abbott RealTime HIV-1 assay as compared to VERSANT HIV-1 RNA 130. Weber, B. 2002. Human immunodeficiency virus (HIV) antigen-antibody
3.0 and AMPLICOR HIV-1 MONITOR v1.5 using serial dilutions of 39 group combination assays: evaluation of HIV seroconversion sensitivity and sub-
M and O viruses. J. Virol. Methods 141:4957. type detection. J. Clin. Microbiol. 40:44024403.
121. Tagliamonte, M., N. Vidal, M. L. Tornesello, M. Peeters, F. M. Buonaguro, 131. Weber, B., E. H. Fall, A. Berger, and H. W. Doerr. 1998. Reduction of
and L. Buonaguro. 2006. Genetic and phylogenetic characterization of diagnostic window by new fourth-generation human immunodeficiency vi-
structural genes from non-B HIV-1 subtypes in Italy. AIDS Res. Hum. rus screening assays. J. Clin. Microbiol. 36:22352239.
Retrovir. 22:10451051. 132. Yang, X., R. Wyatt, and J. Sodroski. 2001. Improved elicitation of neutral-
122. Temin, H. M. 1993. Retrovirus variation and reverse transcription: abnor- izing antibodies against primary human immunodeficiency viruses by solu-
mal strand transfers result in retrovirus genetic variation. Proc. Natl. Acad. ble stabilized envelope glycoprotein trimers. J. Virol. 75:11651171.
Sci. USA 90:69006903. 133. Yu, X. G., M. Lichterfeld, B. Perkins, E. Kalife, S. Mui, J. Chen, M. Cheng,
123. Triques, K., A. Bourgeois, N. Vidal, E. Mpoudi-Ngole, C. Mulanga-Kabeya, W. Kang, G. Alter, C. Brander, B. D. Walker, and M. Altfeld. 2005. High
N. Nzilambi, N. Torimiro, E. Saman, E. Delaporte, and M. Peeters. 2000. degree of inter-clade cross-reactivity of HIV-1-specific T cell responses at
Near-full-length genome sequencing of divergent African HIV-1 subtype F the single peptide level. AIDS 19:14491456.
viruses leads to the identification of a new HIV-1 subtype designated K. 134. Zhu, T., B. T. Korber, A. J. Nahmias, E. Hooper, P. M. Sharp, and D. D. Ho.
AIDS Res. Hum. Retrovir. 16:139151. 1998. An African HIV-1 sequence from 1959 and implications for the origin
124. Turner, D., B. Brenner, D. Moisi, M. Detorio, R. Cesaire, T. Kurimura, H. of the epidemic. Nature 391:594597.

También podría gustarte