Você está na página 1de 20

Vaccine 24 (2006) 4062–4081

Review

A review of vaccine research and development: The human


immunodeficiency virus (HIV)夽
Marc P. Girard a,∗ , Saladin K. Osmanov b,1 , Marie Paule Kieny b,1
a University Paris 7, 39 rue Seignemartin, FR 69008 Lyon, France
b Initiative for Vaccine Research, World Health Organization, 20 Avenue Appia, CH-1211Geneva 27, Switzerland

Received 19 November 2005; received in revised form 10 February 2006; accepted 13 February 2006
Available online 28 February 2006

Abstract

Since the discovery of AIDS in 1981, the global spread of HIV has reached pandemic proportions, representing a global developmental and
public health threat. The development of a safe, globally effective and affordable HIV vaccine offers the best hope for the future control of
the pandemic. Significant progress has been made over the past years in the areas of basic virology, immunology, pathogenesis of HIV/AIDS
and the development of antiretroviral drugs. However, the development of an HIV vaccine faces formidable scientific challenges related to
the high genetic variability of the virus, the lack of immune correlates of protection, limitations with the existing animal models and logistical
problems associated with the conduct of multiple clinical trials. More than 35 vaccine candidates have been tested in Phase I/II clinical
trials, involving more than 10,000 volunteers, and two Phase III trials have been completed, themselves involving more than 7500 volunteers.
Multiple vaccine concepts and vaccination strategies have been tested, including DNA vaccines, subunit vaccines, live vectored recombinant
vaccines and various prime-boost vaccine combinations. This article reviews the state of the art in HIV vaccine development, summarizes the
results obtained so far and discusses the challenges to be met in the development of the various vaccine candidates.
© 2006 World Health Organization. Published by Elsevier Ltd. All rights reserved.

Keywords: Acquired immunodeficiency syndrome; AIDS; Human immunodeficiency virus; HIV-1; Simian immunodeficiency virus; SIV; Vaccines

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4063
2. Disease burden . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4063
3. Virology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4064
4. HIV vaccine reseach: challenges and difficulties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4065
4.1. Why is it so difficult to develop an HIV vaccine? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4065
4.2. Animal models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4066
5. Vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4066
5.1. Live attenuated vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4066
5.2. Inactivated vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4066
5.3. Virus-like particles (VLP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4067
5.4. Subunit vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4067

夽 The authors alone are responsible for the views expressed in this publication, which does not necessarily reflect the views of the World Health

Organization.
∗ Corresponding author. Tel.: +33 478 748 531.

E-mail addresses: marc.girard36@wanadoo.fr (M.P. Girard), osmanovs@who.int (S.K. Osmanov), kienym@who.int (M.P. Kieny).
1 Tel.: +41 22 791 35 91.

0264-410X/$ – see front matter © 2006 World Health Organization. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.vaccine.2006.02.031
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4063

5.4.1. Envelope-based subunit vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4067


5.4.2. Non-structural protein subunit vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4068
5.5. Naked DNA and live recombinant vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4068
5.6. The development of prime-boost combinations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4071
5.7. Fusion proteins and peptides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4071
6. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4071
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4073
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4073

1. Introduction 2. Disease burden

The Acquired Immunodeficiency Syndrome (AIDS) At the end of 2005, the global number of adults
emerged in the human population in the summer of 1981. and children living with HIV/AIDS was estimated by
There now is convincing evidence that AIDS is a zoonosis WHO/UNAIDS to have reached 40.3 million with an esti-
and that its etiological agent, the human immunodeficiency mated 2.9–3.5 million HIV-infected persons dying every year
virus (HIV), crossed the simian-human species barrier before from the disease [3].
the middle of the 19th century [1,2] and probably arose and Sub-Saharan Africa remains the hardest-hit region in the
spread among humans as a result of increased unsafe injec- world, with at least 25 million infected people, accounting
tion and transfusion practices in postcolonial Africa, enabling for 60% of the people living with HIV/AIDS and 77% of
the virus to adapt to humans through serial passages. Today, AIDS deaths in the world. The overwhelming majority of
HIV/AIDS is the leading cause of death in sub-Saharan HIV infections in the region stems from heterosexual trans-
Africa and the fourth biggest killer in the world. An esti- mission [18]. In many African countries, the overall HIV
mated 14,000 people/day (5 million persons/year, including prevalence in the adult population can be greater than 10%,
600,000 children less than 15 years of age) become infected with figures reaching up to 38.8% in some areas. Among
with HIV, with more than 95% of them living in underdevel- the most severely hit countries are South Africa, with more
oped regions of the world [3]. The number of HIV infections than 5.6 million infected people and a prevalence of 30%
is equally distributed between men and women, but infection among 21–29-year-old adults, especially women, together
rates in young women in today’s Africa are close to three with Botswana, Mozambique, Zimbabwe, and Tanzania.
times higher than those among young men, reflecting the The highest infection rates are found among commercial
degree to which gender inequities are driving the epidemic, sex workers, truck drivers and seasonal migrant workers.
as many women in developing countries lack socio-economic Sub-Saharan Africa also is home to an estimated 500,000
independence, education and access to health information HIV-infected infants who became infected prior to the intro-
and services, and have difficulty avoiding exposure to the duction of mother-to-child prevention programmes based
virus. on the use of antiretroviral drugs. In addition, sub-Saharan
The development of a safe, effective, easy to administer Africa faces numerous wars and civil conflicts, producing
and affordable AIDS vaccine is urgently needed. The first large numbers of refugees who are at heightened risk of con-
Phase I trial of an HIV vaccine was conducted in the USA tracting HIV. Life expectancy in South African countries like
in 1987. Since then, more than 35 candidate vaccines have Botswana, Swaziland or Zimbabwe, where a quarter to more
been tested in over 65 Phase I/II clinical trials, involving than a third of the general population is infected with HIV-1,
more than 10,000 healthy human volunteers in more than 10 has decreased from 65 years in 1985–1990 to 37 years in
countries [4,270]. Two Phase III trials have been carried to 2000–2005.
completion [5] and a third one is in progress [6]. However, A remarkable success story in the fight against AIDS was
we still are years away from an effective HIV vaccine, due achieved in Uganda, which faced the onset of a severe HIV
to multiple hurdles and challenges. The development of a epidemic in the mid 1980s. Through voluntary HIV coun-
safe and effective vaccine is hampered by the high genetic selling, expanded treatment of STDs, awareness campaigns
variability of the virus [7,8], the lack of knowledge of immune and community mobilization encouraging delayed initiation
correlates of protection [9,10], the difficulty of generating of sexual activity, monogamy and use of condoms, the level of
broadly neutralizing antibodies [11], the absence of relevant infection has declined significantly since 1992—from nearly
and predictive animal models and the complexities related to 30 to 11.2% in prenatal clinic settings in Kampala and from
the preparation and conduct of multiple large-scale clinical 13 to 5.9% in clinics outside major urban areas. Another
trials, especially in developing countries [12,13]. important HIV prevention strategy could be adult male cir-
This article reviews the current progress in HIV/AIDS cumcision, which was found in a recent study in South Africa
vaccine research and development and the many challenges to offer close to 65% protection rate from HIV infection. Two
that still need to be met in the field [14–17]. other randomized controlled trials are underway in Kenya and
4064 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

Uganda to obtain more data on the potential protective effect virus (EIAV), belongs to the genus Lentivirus in the family
of circumcision on female-to-male transmission. Retroviridae. These enveloped RNA viruses produce char-
The estimated number of people living today with HIV acteristically slow, progressive infections. Lentivirus replica-
in Asia and the Pacific region is more than 8.3 million, but tion depends on the presence of an active reverse transcriptase
the accuracy of the figure is questionable in view of the fast responsible for the transformation of the viral RNA genome
pace at which the epidemic is expanding. It has been projected into a proviral DNA copy that integrates into the host cell
that if current trends continue, the region will contribute 40% chromosome. The provirus is eventually transcribed into a
of all new infections worldwide by the end of the decade. set of mRNAs that encode the viral proteins and progeny
Increasing sex trade, use of illicit drugs, and high rates of genomic RNA.
sexually transmitted infections contribute to an increased vul- Two types of HIV have been described: HIV-1 and HIV-2,
nerability of the populations in the region. Gender inequities the latter being less virulent, less transmissible and geograph-
also play an important role in the epidemic, as young girls ically limited to West Africa. HIV-1 is phylogenetically close
are frequently steered towards commercial sex work by their to SIVcpz, a commensal virus in chimpanzees, and probably
families. Substantial transmission also occurs among men arose as the result of a single transmission event from chim-
who have sex with men, with prevalence rates of 14–20% panzees to humans [1], whereas HIV-2 is closely related to
reported in certain male homosexual communities. SIVmac, the etiological agent of simian AIDS, and to SIVsm,
The estimated number of adults and children living with a commensal virus in sooty mangabey monkeys. HIV-1 is
HIV in Latin America and the Caribbean at the end of 2005 further subdivided into three groups, M (“major”), O (“out-
was 2.1 million. While in some countries HIV infections lier”), and N (“non M/non O”). The vast majority of the HIV-1
remain concentrated mainly among men who have sex with strains responsible for the global AIDS pandemic belong to
men and injecting drug users, other countries are experienc- group M, which has evolved in humans to form at least 10
ing increasing rates of heterosexual transmission. genetic subtypes, also known as clades, designated by let-
The Eastern European countries continue to experience ters from A to K [20]. HIV-1 subtype B predominates in
one of the sharpest increases in the number of new HIV infec- industrialized countries as well as in Latin America and the
tions, most of which occur among injecting drug users. The Caribbean. Subtypes A and D are more common in Central
number of people living with HIV/AIDS in the region is esti- Africa. Subtype C accounts for the majority of infections in
mated to be above 1.6 million, 70–80% of whom are less than southern Africa, eastern Africa and India. Genetic subtypes,
30 years of age. More than 300,000 HIV seropositive persons in turn, have diversified further. Even within a given subtype,
are living in Russia alone, where commercial sex workers, antibodies that are specific for isolates from one patient typ-
injection drug users and prisoners are major victims of the ically do not recognize isolates from other patients. Over the
tuberculosis and HIV epidemics [19]. course of the epidemic and as a result of frequent dual and
The introduction of the Highly Active Antiretroviral Treat- superinfections, HIV strains can recombine and form mosaic
ment (HAART) in industrialized countries has considerably viruses, some of which, called “Circulating Recombinant
reduced disease progression to AIDS and transformed HIV Forms” (CRF), can gain epidemiological dominance [20–23].
infection from a lethal disease to an effectively manageable The major CRFs are CRF02 AG, prevalent in western Africa,
chronic disease. AIDS among infants, which a decade ago CRF01 AE, which predominates in south-eastern Asia, and
took the lives of thousands of babies per year may be on CRF07 BC and 08 BC, which are prevalent in China. Amino
the verge of elimination. Thus, in New York city, the num- acid sequence of the viral envelope glycoprotein shows up
ber of babies born with HIV fell from 321 in 1990 to 5 to 25–35% divergence between different subtypes and up to
in 2003, mostly due to education, counselling, testing and 20% divergence within any given subtype, which constitutes
antiviral treatment of the mother during pregnancy. How- a formidable challenge to vaccine development.
ever, successes achieved in treatment and care are not being The genome of HIV is a single-stranded positive sense
matched by progress in prevention, as some 75,000 individu- RNA molecule, about 9.5 kb in length, which encodes the typ-
als become infected with HIV every year in industrialized ical retrovirus proteins Gag, further cleaved into M (matrix),
countries, where an estimated 1.6 million people are liv- C (capsid) and N (nucleocapsid), Pol, cleaved into protease,
ing with HIV/AIDS, and where a new wave of rising HIV reverse transcriptase and integrase, and Env, a 160 kD glyco-
infection rates is emerging, particularly in the young and protein eventually cleaved into an external gp120 subunit and
marginalized communities. a transmembrane gp41 subunit that together form trimeric
spikes on the surface of the virion. In addition, the HIV
genome encodes a variety of non-structural proteins, such as
3. Virology the transactivator protein Tat [24], the splice regulator pro-
tein Rev [25] and accessory proteins such as Nef [26,27], Vif
The HIV, together with the simian, the feline, and the [28], Vpr [29] and Vpu.
bovine immunodeficiency viruses (SIV, FIV, and BIV, respec- The mature virion contains a cone-shaped protein cap-
tively), the Visna virus of sheep, the caprine arthritis- sid, which encapsidates two strands of the genomic RNA,
encephalitis virus (CAEV) and the equine infectious anemia the replication enzymes, tRNAs and cellular proteins such as
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4065

cyclophilin A and Tsg101. The capsid is enclosed within a infection, before the virus can seed the lymphoid organs in
lipid envelope, which is studded with the viral glycoprotein mucosal tissues [63,64].
spikes and cellular proteins, including MHC class II anti- HIV also has developed multiple mechanisms to circum-
gens and ICAM-1 [30], and which lies on top of a layer of vent the host immune responses including its ability to down-
matrix protein attached to the inside of the membrane through regulate the major histocompatibility complex (MHC) class
embedded myristyl groups. However, the actual structure of I molecules and by doing so to minimize its recognition by
the virion still remains speculative [31]. CTL, as well as its high genetic evolution rates, which allows
The envelope glycoprotein comprises two subunits, the it to evade immune responses through the emergence of viral
external gp120 subunit which binds to the CD4 receptor and CTL [3,65–68] and neutralizing antibody [69,70] escape vari-
CCR-5 or CXCR-4 coreceptors on the surface of target cells ants.
(for a review, see [32]), and the gp41 transmembrane subunit, Another difficulty with the development of an effective
which anchors the spikes in the viral envelope, maintains their HIV vaccine stems from the fact that the virus envelope
trimeric organization, and plays a major role in fusion of the glycoprotein conceals its conserved receptor- and coreceptor-
virus and host cell membranes through its hydrophobic N- binding sites in crypts that are masked by the hypervariable
terminal fusion peptide and a fusion active hairpin structure loops of the molecule and by glycan residues [45,70,71].
involving two heptad repeats that can fold into a six-helix Neutralizing antibodies induced in response to gp120 are pri-
coiled-coil bundle [33–36]. Neutralizing human monoclonal marily targeted to the hypervariable loops of the molecule and
antibodies, such as IgG b12 and 2G12, have allowed the iden- only rarely do they recognize the receptor binding sites, which
tification of several neutralization epitopes on gp120 [37], makes it hard to generate broadly cross-reactive neutraliz-
some of which are carbohydrate-specific [38,39] and oth- ing antibodies against primary virus isolates from patients
ers overlap the CD4 receptor- or coreceptor-binding sites [11,15,72–75]. The complete lack of efficacy of antibody
[40–43], but all of them appear to be little accessible to the responses raised by monomeric gp120 vaccines in protection
cognate antibodies due to hindrance by the many glycosy- against HIV infection has been proven beyond any doubt in
lation motifs on the molecule [44] as well as by the hyper- the world’s first two Phase III clinical trials of AIDS vaccines
variable loops [45,46], which act as antigenic decoys [47]. [5,76].
Two monoclonal fusion-blocking antibodies (2F5, 4E10) also Although neutralizing antibodies administered passively
have been described, with corresponding epitopes located at to non-human primates can provide protection against exper-
the base of the gp41 ectodomain [48–53]. A note of caution imental SHIV infection [77–85], proof of their protective role
has been raised by the results of one study, which showed that in infected humans remains circumstantial [86]. The best evi-
these monoclonal antibodies were reactive with the human dence for a role of HIV-1 neutralizing antibodies in vivo is the
phospholipid cardiolipin [54,55]. rapid and constant selection of neutralizing antibody-escape
variants during the course of infection [69,70]. However, con-
trary to laboratory-adapted virus strains, which use CXCR-4
4. HIV vaccine reseach: challenges and difficulties as a coreceptor (“X4” strains), and against which protection in
chimpanzees could readily be achieved by inducing neutraliz-
4.1. Why is it so difficult to develop an HIV vaccine? ing antibodies targeted to the hypervariable V3 loop [87,88],
primary virus isolates, which use CCR-5 as a coreceptor
Natural infection with HIV does not result in virus clear- (“R5” strains), are difficult to neutralize, which casts doubt
ance by the host immune system and the development of on the possibility for a vaccine to elicit protection against
natural immunity to re-infection. In spite of intense and infection by the induction of neutralizing antibodies alone.
sustained immune responses by both the humoral and cell- In view of all these problems, recent vaccine approaches
mediated defences, HIV is able to resist eradication and have focused on the induction of cellular immune responses
continues depleting CD4+ T cells, which eventually leads [14,15,23,89–91]. Evidence for the role of CD8+ T cells in
to clinical progression to AIDS. There even is evidence that the control of virus replication includes temporal correlation
superinfection with a second HIV isolate can readily occur between the appearance of HIV-specific CD8+ T cells and the
in HIV-infected persons, leading to the emergence of recom- decline of primary viremia [92,93], the fact that several HLA
binant virus variants and generating increased virus diversity class I alleles (HLA-B57, HLA B-27, HLA-B63) are associ-
[56–58]. Similarly, recombination was shown to readily occur ated with slow disease progression [94], the early selection of
between different SIV strains in SIV-infected monkeys [59]. CTL escape viral mutants during primary infection [95–98]
HIV-1 integrates as a latent proviral DNA into the genome and the rapid increase of viral loads in macaques infected with
of long-lived memory CD4+ T cells, which provide a persis- SIV after experimental depletion of CD8+ T cells [99–101].
tent reservoir of the virus that escapes immune surveillance The induction of a cellular immune response against HIV,
[60–62]. It has been calculated that it would take up to 60 especially a CD8+ CTL response, although not being able to
years to eradicate a reservoir of as few as 1× 10E5 latently provide sterilizing immunity and protection from infection,
infected cells. The window of opportunity for an HIV vac- should hopefully enable vaccinees to control virus replica-
cine is therefore narrowly limited to the very early stages of tion following infection, reduce their virus load, slow down
4066 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

their progression towards disease, and reduce the probability vaccinated animals with a huge dose of virus, usually in
of secondary transmission of the virus. the range of 103 –105 TCID50, equivalent to up to 5 × 107
SIV RNA copies/ml. Such a high dose of virus is required
4.2. Animal models to achieve100% infection of control animals in the placebo
group after a single exposure. However, high doses of virus
A significant obstacle in HIV vaccine research has been do not correspond to the natural exposures to HIV in humans,
the difficulty in developing an appropriate animal model. where concentrations of less than 103 to a few 105 HIV RNA
The only animals susceptible to experimental infection with copies/ml of seminal plasma have been reported [126–128].
HIV are chimpanzees, Pan troglodytes, and pigtail macaques, It has recently been observed that repeated low dose (10–30
Macaca nemestrina. Both maintain low levels of persis- TCID50) mucosal challenge of monkeys with SIV results
tent virus load and do not develop clinical manifestations in the same viral and immunological kinetics of infection as
of AIDS. African monkeys are the natural hosts to a vari- high dose challenges [129]. Such a new type of mucosal chal-
ety of SIVs (SIVagm, SIVsm, SIVsyk, SIVcol, etc.), but lenge might change the results of preclinical vaccine efficacy
do not appear to develop a clinical disease following infec- studies in the future (see as an example, [271]).
tion with these viruses [102]. In contrast, Asian monkeys, Of note is the fact that the challenge virus used in animal
especially rhesus monkeys from India, are highly susceptible model experiments is nearly always homologous to the vac-
to SIVmac infection and progressively develop an immun- cine, giving good chances of success that are unrealistic in
odeficiency syndrome, which fully mimics human AIDS humans.
[103]. Plasma virus levels during primary and chronic SIV-
mac251 infection in macaques parallel those observed in
humans, some animals containing viremia spontaneously and 5. Vaccines
progressing to disease slowly, as in HIV-1 infected human
long-term non-progressors, and others maintaining high virus 5.1. Live attenuated vaccines
loads and behaving as fast progressors [104,105]. As in
HIV-1-infected humans, the cellular immune responses to The observation that nef-deleted mutants of SIV could
SIVmac during primary and chronic infection differ sig- confer protection against challenge with pathogenic SIV in
nificantly [106], and evidence of immune escape is readily rhesus macaques [130–132] served as a model in favor of
documented [96–98]. Perhaps most importantly, the mucosal a live attenuated HIV vaccine approach. However, the SIV
immune system, especially the memory CD4+ CCR-5+ T nef mutant establishes a life-long, persistent low-grade viral
cells in the gut-associated lymphoid tissue (GALT), is the infection. It does not protect the vaccinated monkeys against
major site of viral replication and of CD4+ T-cell depletion superinfection with wild-type virus, although it does protect
in SIV-infected macaques [107–110] as in HIV-1-infected the animals from progression to AIDS. SIV nef still may
individuals [111–114]. The SIV/macaque model is currently, cause AIDS, especially when administered orally to infant
therefore, considered to be the most appropriate animal model monkeys [133,134]. Additional deletions or mutations of the
for studies on potential protective immune responses against virus can result in further attenuation but at the expense of
HIV [115,116]. its protective efficacy [135–137]. In view of such safety con-
Another animal model used in HIV vaccine research is cerns, this approach has not been actively pursued [138,139],
based on the use of SIV/HIV hybrid viruses (SHIVs) that but efforts are currently being made by IAVI, among others,
were engineered to carry the env gene from an HIV-1 iso- to try and explore systematically the nature of the protec-
late in the context of an SIV genome. These viruses can tive immune responses generated with live attenuated SIV
replicate in rhesus macaques, and after serial passages in the vaccines in macaques [140].
animal eventually lead to the emergence of highly pathogenic
SHIV variants that are capable of wiping out the circulating 5.2. Inactivated vaccines
CD4+ T-cell population of the animal within a few weeks
after infection and generating a lethal immunodeficiency syn- The difficulty of inactivating HIV-1 with formalin with-
drome within a year. However, the relevance to HIV of these out losing or destroying the antigenicity of the viral envelope
pathogenic SHIVs, such as SHIV 89.6P [117–119], is being has been a deterrent to the development of whole inacti-
questioned [120,121]. They exhibit a “X4” cellular tropism vated HIV vaccines. The obstacle was circumvented by using
and, paradoxically, turn out to be much easier to contain by sublethal doses of formalin followed by heat inactivation
vaccine-induced immune responses than “R5” viruses such as at 62 ◦ C [141]. The resulting killed virus preparation was
SIV [122,123]. SHIV strains with an “R5” tropism [124,125] shown to induce in mice and non-human primates modest
probably are a more appropriate and predictive model than but significant titers of antibodies capable of neutralizing het-
“X4” SHIVs, but they have not yet been widely used in vac- erologous primary isolates of HIV in a variety of infectivity
cine protection experiments. assays [142]. Another way to inactivate HIV or SIV infectiv-
The monkey models suffer from the fact that experi- ity has been to target the two nucleocapsid protein zinc finger
ments to evaluate vaccine efficacy require challenging the domains, which bind Zn ions in tetrahedral coordination com-
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4067

plexes that are essential for virus infectivity. These complexes homologous vaccine strain, but not to heterologous primary
can be disrupted by mild oxidation with 2,2 -dithiodipyridine virus isolates [76,152,153]. These vaccines elicited steriliz-
(aldithriol-2) [143] or by alkylation with N-ethyleneimine ing immunity against homologous virus challenge in animal
(NEM). Both compounds have been found to completely models [87,88,154]. Two gp120 subunit vaccines were fur-
inactivate the infectivity of HIV-1 and SIV, while keeping ther evaluated in a Phase II trial in the USA [155], and one
the envelope glycoprotein spikes intact and functional [144]. such vaccine, based on monomeric gp120 added with alum
The immunogenicity of the resulting preparations was stud- (VaxGen), was tested in two double-blind, placebo-controlled
ied in the SIV/macaque model [145]: vaccinated monkeys Phase III efficacy trials. The first trial involved 5000 volun-
were not protected against infection with wild-type virus but teers at risk (mostly men who had sex with men) in the USA,
experienced decreased viremia after challenge and showed Canada and the Netherlands, who were immunized every 6
no significant depletion of CD4+ T cells. months with a 300 ␮g mixture of two subtype B gp120s. The
second trial involved 2500 volunteers in Thailand (mostly
5.3. Virus-like particles (VLP) injection drug users), who were immunized with a mixture
of a subtype E (CRF AE) and a subtype B gp120s. None
When coexpressed in cells, for example using either a of these trials showed a statistically significant reduction of
baculovirus or a vaccinia virus expression system, the Gag HIV infection in the vaccinees in spite of continuous booster
and Env proteins of HIV or SIV spontaneously assemble immunizations during the 36 months of the study. A reduc-
to form pseudo-virions, i.e. virus-like particles (VLPs) that tion of the number of HIV infections was observed in certain
only contain envelope and core proteins. SIV VLPs have ethnic subgroups in the first trial, correlating with a higher
been tested as immunogens in non-human primate models level of anti-gp120 antibody, but the numbers were too small
either after priming with vaccinia virus recombinants or by to provide statistical confidence [5].
the nasal route using the cholera toxin B subunit (CTB) as The subtype E/B gp120 vaccine from the trial in Thai-
a mucosal adjuvant [146,147]. The administration of VLP land is presently being used for booster immunizations in
vaccines via mucosal surfaces might be a promising strategy a prime-boost Phase III trial, which was launched in late
to elicit mucosal and systemic anti-HIV immune responses 2003 in Thailand in collaboration between the Ministry of
[148]. Gag–Env pseudovirions were also tested in guinea pigs Health of Thailand, WRAIR, Sanofi-Pasteur and VaxGen.
and found to elicit low but significant titers of neutralizing This community-based trial, which includes 16,000 volun-
antibodies against both homologous and heterologous pri- teers and is meant to last for 4 years, involves a priming
mary HIV-1 isolates when administered in the presence of immunization with a recombinant canarypox virus (ALVAC)
a block copolymer adjuvant or of alum combined with CpG vaccine that expresses CRF AE gp120 and subtype B Gag,
oligonucleotides [149]. Pol and Nef antigens followed by boosts with the gp120 sub-
Heterologous VLPs have also been used as a delivery vehi- unit vaccine [6].
cle for HIV genes. Expression of the structural proteins of Other envelope-based subunit vaccine approaches aimed
Kunjin virus, an arbovirus, was used to prepare VLPs and at eliciting HIV neutralizing antibodies are at an early
encapsidate recombinant Kunjin RNA replicons encoding the clinical stage of evaluation. These include trimeric gp140
HIV-1 gag gene [150]. The resulting VLPs were found to molecules stabilized by the addition of heterologous trimer-
elicit strong CTL and antibody responses to HIV Gag. ization domains at the C-terminus of the gp41 ectodomain
Still another approach has been to induce systemic and [156,157], and similar trimers in which the gp120 moiety
mucosal neutralizing antibodies against HIV by immuniza- was deleted of the variable V2 loop, in order to expose neu-
tion with bovine papillomavirus (BPV)-HIV-1 gp41 chimeric tralization epitopes overlapping the CD4-binding site. This
VLPs. The ELDKWA amino acid sequence (2F5 epitope) approach is developed by Chiron [158–160]. Immunization
from HIV-1 gp41 was fused to the N-terminus of the BPV L1 of rabbits and macaques with a DNA vaccine encoding a V2-
capsid protein and the resulting chimeric protein was used to deleted gp140 from a South African subtype C HIV-1 strain,
generate chimeric VLPs. The latter elicited gp41-specific IgG followed by boosting with oligomeric V2-deleted gp140 in
after intra-muscular immunization and both systemic IgG and MF59 adjuvant elicited high titers of Env-binding antibod-
intestinal secretory IgA after oral immunization in mice. HIV ies and low level heterologous neutralizing antibodies [161].
neutralization activity could be detected in some of the mouse Gp140-GCN4 trimeric immunogens were found to induce
sera [151]. a modest but significant heterologous neutralization activ-
ity, which could be efficiently increased by emulsification in
5.4. Subunit vaccines adjuvants AS01B, AS02A or AS03 from GSK [162].
Gp140 trimers internally stabilized by an intermolecu-
5.4.1. Envelope-based subunit vaccines lar disulfide bond between gp120 and gp41 (SOS gp140),
Initial trials of HIV-1 Env-based subunit vaccines showed have been developed with an expectation to induce both
that soluble recombinant envelope glycoproteins gp120 or neutralizing and fusion-blocking antibodies [163,164]. Prim-
gp140 (gp120 prolonged with the ectodomain of gp41) were ing with DNA encoding a membrane-bound form of the
well-tolerated and elicited neutralizing antibodies to the SOS gp140 protein followed by repeated immunization with
4068 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

the soluble trimers resulted in high titer antibodies active sequences, which have been expressed in an immunogenic
against neutralization-sensitive HIV-1 strains, but even the form [177–179]. The problem still, however, remains basi-
most potent of the sera had a quite limited ability to cross- cally unsolved [11].
neutralize primary heterologous HIV-1 strains [165].
Still another approach has been to covalently couple 5.4.2. Non-structural protein subunit vaccines
monomeric gp120 or oligomeric gp140 molecules to solu- The hope that immune responses directed against antigens
ble CD4 or to synthetic mimetics of the CD4 receptor in that are expressed very early in the virus life cycle might
order to induce the conformational changes that are sup- lead to rapid elimination of infected cells has prompted the
posed to take place in the glycoprotein spike at the time of development of subunit vaccines based on the viral trans-
virus entry, resulting in the exposure of neutralization epi- activator Tat [180–182]. Immunization with Tat protected
topes that overlap the coreceptor-binding site. This approach macaques against SHIV infection [183–185] or resulted in
has been developed by Merck and by the University of Mary- attenuated virus replication in the animals [186]. However,
land [166], which also engineered single chain derivatives of immunization with different forms of the Tat protein did not
gp120 covalently linked to the first two domains of the human demonstrate any efficacy against virus challenge [187,188]
CD4 molecule [167]. The single chain fusion proteins were and a recent study using an adenovirus type 5 (Ad5)-HIV Tat
shown to elicit a broadly neutralizing antibody response in recombinant vaccine elicited no protection in rhesus mon-
guinea pigs, but this property may have primarily been due to keys against a SHIV challenge, in spite of demonstrable
the induction of anti-CD4 antibodies. In contrast, covalently Tat-specific T cell and antibody responses [189]. A detoxified
linked gp120-CD4 complexes were reported to elicit broadly Tat subunit vaccine developed by Neovax in collaboration
neutralizing antibodies in macaques that were not related to with Sanofi Pasteur, has been evaluated in a Phase I trial and
anti-CD4 antibodies [168]. However, this observation was should eventually enter Phase II trials. A Phase I clinical trial
not confirmed in later experiments (Sadoff J, personal com- of another subunit Tat vaccine was carried out in Italy on
munication). Replacing the CD4 binding sequences by the HIV seropositive and HIV negative volunteers [190], show-
appropriate synthetic mimetic might be key to this problem ing that the vaccine was well-tolerated and immunogenic.
but higher affinity mimetics need to be developed [64]. Phase II trials are planned to begin presently in seronegative
Engineering hyperglycosylated derivatives of gp120 was volunteers at risk as well as in HIV-infected volunteers. At
attempted in order to target the immune response to conserved the same time, the AIDS Vaccine Integrated Project (AVIP), a
epitopes on the molecule that form part of the CD4-binding European consortium, is developing a novel vaccine approach
site [169]. This approach is under development. based on the use of a trimeric gp140 molecule deleted of the
Induction of fusion-blocking antibodies by immuniza- V2 loop (gp140 V2 SF162) that will be mixed with either
tion with recombinant oligomeric gp41 molecules is another Tat or Nef [191].
promising new approach that still is at an early preclinical Different vaccine approaches using the Tat antigen have
stage of development [170,171]. also been developed such as a Tat-adenyl cyclase fusion pro-
Finally, IAVI’s Neutralizing Antibody Consortium has tein [192] or a Tat–Nef fusion protein that was combined
undertaken to solve the crystal structure of the complexes together with a recombinant gp120 subunit vaccine in the
formed between a broadly HIV-neutralizing human mono- AS02A adjuvant (an oil-in-water emulsion with monophos-
clonal antibody and the viral envelope glycoprotein spikes to phoryl lipid A and the saponin derivative QS21). This com-
try to unravel new clues for vaccine design [172]. bined vaccine was able to prevent the development of AIDS in
It has been generally recognized that the extremely high rhesus monkeys following challenge with a pathogenic SHIV
level of genetic variability of HIV and its continuous evo- strain [193]. The Tat–Nef fusion protein, which is developed
lution over time within one single individual, as well as by GSK, was recently tested in human volunteers in a Phase I
between different population groups and diverse geographi- clinical trial using MVA as a vector and was found to induce
cal regions represents a major challenge for the development strong CD4+ T cell and antibody responses.
of HIV vaccines capable of eliciting broadly reactive neu-
tralizing antibodies against globally prevalent HIV strains 5.5. Naked DNA and live recombinant vaccines
[173]. To address this challenge, multiple parallel vaccine
strategies are being explored, such as the use of cocktails of The majority of recent HIV vaccine studies have aimed
envelope immunogens derived from globally prevalent HIV to develop T-cell-stimulating vaccines that induce a HIV-
strains [55,174], that of multivalent DNA or live vectored specific CD8+ CTL response, whose role in control of virus
vaccines incorporating envelope genes from various clades, load and evolution of disease has been well-documented in
as well as combinations of live-vectored vaccines as a prime the monkey model (see above). Although vaccines that stimu-
followed by vaccination with a subunit recombinant vaccine late the cellular arm of the immune response are not expected
as a boost [175,176]. In addition, novel avenues to circumvent to provide protection against infection, they should allow vac-
the problem of the broad genetic diversity of HIV have been cinees who get infected to control virus replication and reduce
opened by the development of synthetic genes derived from viral loads, thus resulting in lower probability of virus trans-
theoretically defined consensus or ancestral HIV envelope mission to seronegative partners. A variety of vaccines were
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4069

developed based on this strategy, including naked DNA vac- pilot, monovalent Ad5-gag recombinant that was shown to be
cines and live vectored recombinant vaccines, some of which highly immunogenic in mice, nonhuman primates and human
already are in advanced stages of clinical trials (For recent volunteers. More than 50% of the volunteers showed sig-
reviews, see [17,194]). nificant, long-lasting HIV-1-specific CD8+ T-cell responses
Results from preclinical studies on plasmid DNA vac- to HIV-1 peptides, including secretions of IFN-␥, IL-2, and
cines in non-human primates were rather disappointing, even TNF-␣. A trivalent recombinant Ad5-gag/pol/nef vaccine
though successful protection against a SHIV challenge could has now been engineered and tested in human volunteers. A
be observed in macaques [195,196] as well as in the HIV- multi-center Phase II trial of this trivalent candidate vaccine
1/chimpanzee model [197]. Immunogenicity of the vaccines has been launched, involving some 1200 men and 400 women
could be improved by modification of the promoter, the use of at high-risk who will be followed for 3 years after three immu-
synthetic genes with optimized codons, insertion of the trans- nizations at 0, 4 and 26 weeks (NIAID and Merck). The trial
gene into an alphavirus replicon, or coexpression of cytokines is taking place in several centers in North America, Peru,
IL-2 or GM-CSF [198,199]. Naked DNA vaccines express- Brazil, the Caribbean Islands and Australia, with final results
ing the HIV-1 gag gene and either IL-12 or IL-15, which expected in 2008.
were developed by Wyeth, are being tested in Phase I trials in Another non-replicative adenovirus vector is being devel-
the USA, Brazil and Thailand. DNA vaccines were actually oped by the NIH Vaccine Research Center (VRC), together
found to be most useful as priming vaccines in prime-boost with a DNA-based vaccine. These are multicomponent vac-
strategies, using live recombinant vaccines for booster immu- cines, which express the Env glycoprotein from clades A,
nization (see below). These strategies were tested in monkeys B and C and the Gag, Pol and Nef proteins from clade B,
against pathogenic SHIV challenges and shown to result in and are designed for use in a prime-boost regimen strategy
reduction in virus load and protection against disease and [175]. The DNA vaccine was initially tested in Phase I trials
death in the vaccinated animals [200]. The same approach in the USA, where it showed good immunogenicity, and is
was, however, definitely less successful in protection against undergoing a Phase I trial in Uganda in collaboration with the
SIV challenge [122,201,202]. Makarere University and WRAIR. The VRC Ad5 recombi-
The first live recombinant HIV vaccines that were devel- nant candidate vaccine also has undergone testing in a Phase I
oped were based on the use of vaccinia virus as a vec- study in the USA and is now being tested on those volunteers
tor. Safety considerations prompted the search for other, who were earlier primed with the DNA vaccine. A Phase I
non-replicative poxvirus vectors. A canarypox virus vector DNA-Ad5 prime-boost trial was recently started in the USA,
(ALVAC) was developed by Sanofi-Pasteur and intensively Brazil and South Africa and will presently be extended to
evaluated in multiple Phase I/II trials [203–205]. It now is East Africa in collaboration with IAVI.
being tested in a Phase III trial in Thailand (see above). The There is little doubt that the best results so far, in terms
modified vaccinia virus Ankara (MVA) vector was tested in of percent human responders and levels and duration of T-
multiple prime-boost vaccine protection studies in macaques cell responses to HIV-1 Gag in human volunteers, have been
[206] and is currently still undergoing evaluation in Phase I/II obtained with the Ad5 recombinant vaccines, either delivered
trials in human volunteers in the USA and in several African alone or as boosting immunogens after plasmid DNA priming
countries, most often as a boost in combination with a DNA [212]. However, these vaccines are confronted with the prob-
vaccine priming [23,207]. A subtype C multigenic env, gag, lem of frequently pre-existing anti-vector immunity in the
tat–rev, nef MVA recombinant developed by Therion in col- human population, especially in developing countries, which
laboration with IAVI is thus undergoing trial in India, whereas may dampen the immune response to the HIV transgenes
another MVA recombinant that expresses subtype A,E env, [213]. This has prompted the development by Merck, Cru-
gag, pol and was developed by the NIH is in Phase I trial in cell and Transgene, in collaboration with IAVI, of candidate
the USA, soon to be followed by Thailand. The fowlpox virus vaccines based on less prevalent human adenovirus serotypes
(FPV) also has been used as a vector for HIV vaccines and has (Ad6, Ad35, Ad11, or Ad24) to replace the Ad5 vector in
been tested in Phase I trials in Australia [208]. It is currently future HIV vaccine trials [214,215]. Like Ad5, these vectors
being evaluated in combination with MVA in the USA. These readily multiply to large yields in PRC-6 cells. Nonreplica-
three vectors were used to express a variety of HIV antigens, tive chimpanzee adenoviruses (AdC68, AdC6 and AdC7),
such as Gag, Env, Pol and Nef. However, the immunogenicity against which humans do not have neutralizing antibodies,
of the poxvirus-based HIV vaccines in humans has usually also are being explored as novel vectors by GSK and IAVI.
been relatively modest, with less than 35% of the vaccinees In addition, as the major adenovirus neutralization epitopes
scoring positive for T-cell responses at any point in time, as are carried by the penton spike on the adenovirus virion, the
determined by IFN-␥ ELISPOT assays. This was emphasized development of adenovirus chimeras that escape anti-Ad5
by the disappointing results of recent IAVI-sponsored DNA pre-existing immunity has been achieved by replacing the
prime-MVA boost studies in the UK and in Africa. fiber gene of an Ad5 vector by that from a rare Ad subtype.
Replication-defective adenovirus type 5 (Ad5) represents Adenovirus chimeras such as Ad5/Ad11 or Ad5/Ad35 have
one of the most promising live viral vectors for HIV vaccines been constructed and successfully tested in animal models
[209–211]. It was first used by Merck for the development of a [216].
4070 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

A different vaccine strategy using adenovirus vectors The alphavirus replicon particle vectors have been devel-
also has been developed by the NIH, employing replication- oped using Sindbis virus, Semliki Forest virus (SFV), and
competent Ad recombinants in which the Ad vector is deleted Venezuelan equine encephalitis virus (VEEV). The vec-
only in the E3 region [217]. Ad4hr-, Ad5hr-, or Ad7hr-HIV tor RNA codes for its replicase and contains cis-active
recombinants together with boosts of subunit envelope glyco- replication sequences, which allow its self-amplification in
protein were found to protect chimpanzees against challenge the cytoplasm of infected host cells. VEEV targets imma-
with a primary, heterologous HIV-1 isolate [218,219]. Pro- ture dendritic cells, which play a major role in present-
tective efficacy was also demonstrated in the SIV/macaque ing antigens to the immune system, and has a natural
model, using a replication-competent Ad5hr-SIV env/rev and lymphotropism that may be ideal for vaccine strategies.
Ad5hr-SIV gag priming followed by SIV gp120 boosting The VEE replicon particles were shown to induce potent
[220]. and protective immune responses in primates [227,228].
The list of other virus vectors used to construct live recom- A replication-incompetent vector that was engineered to
binant vector-based HIV vaccines is long [17,194]. Some of encode the subtype C HIV gag gene was tested in a
the most promising vector candidates include the measles Phase I trial in the USA, South Africa and Botswana. This
virus (MV), vesicular stomatitis virus (VSV), Sendai virus approach is developed by Alphavax in the USA. Recombinant
(SeV) and Venezuelan equine encephalitis virus (VEEV). SFV particles expressing HIV-1 gp140 efficiently primed
The Schwarz attenuated MV strain, which has a long stand- immune responses as measured after a single boost with
ing safety and efficacy record as a live attenuated measles purified trimeric gp140 protein, resulting in a Th1-biased
vaccine, was engineered to express the HIV gp160 molecule immune response and neutralizing antibodies against HIV-
deleted of the V3 loop. The recombinant was found to 1 [157].
elicit neutralizing antibodies with broad specificity after a A number of other vectors has also been used to prepare
single immunization in mice [221]. Recombinant MV-HIV live recombinant HIV vaccines, including, among others:
vaccines were also successfully tested in a SHIV/macaque
model. Their development is currently planned in collabora- - Adenovirus-associated virus [229,230], which is devel-
tion between the Pasteur Institute and GSK. oped by Targeted Genetics and IAVI and was tested in
AIDS vaccines based on an attenuated VSV vector Phase I trials in Germany, Belgium and India. This can-
expressing the Gag and Env proteins were found to provide didate vaccine recently entered Phase II evaluation in
complete protection against T-cell loss and disease progres- South Africa, later to be followed by trials in Uganda and
sion in the SHIV/macaque model, with all the vaccinated Zambia.
animals having low or undetectable virus load levels for up - BCG, which was developed as a vector for HIV by the
to 14 months after challenge [222]. VSV, which is developed Japanese NIID and already underwent a Phase I trial in
as a vector for HIV by Wyeth, is a particularly attractive vec- Thailand with an early construct [231]. Recombinant BCG-
tor as it can efficiently be administered by a mucosal route, based vectors may have potential as an HIV vaccine when
for example as a single intranasal vaccination [223]. Potential administered in combination with a recombinant poxvirus
neurovirulence of VSV was circumvented in a new genera- vector-based vaccine in a “prime-boost” vaccine strategy
tion vector by reshuffling the viral N gene in the VSV genome [232].
and truncating the G protein cytoplasmic tail [272]. Not only - Attenuated Salmonella, that are used as vectors for the
is this new vector devoid of detectable neurovirulence by the development of oral vaccines at the University of Mary-
intracerebral route in mice, ferrets and monkeys, but it also land [233].
appears to be more immunogenic that its wild-type parent
strain. A VSV vector capable of only a single cycle of repli- Poliovirus attenuated Sabin strains from the oral polio
cation was recently generated and found to be equivalent to vaccine (OPV) also have been engineered to express short
a replication-competent VSV vector in generating high-level sequences of the HIV genome and could be used as an oral
primary and memory CD8+ T-cell responses as well as anti- vaccine [234], but the future of this approach remains uncer-
body responses to Env in mice [224]. tain in view of the probable adverse impact of pre-existing
Another vector of interest for the construction of live immunity in the human population and the fact that OPV is
recombinant HIV vaccines is SeV, a paramyxovirus from progressively abandoned, whenever possible, in favor of the
mice which is devoid of pathogenicity for primates [225]. inactivated polio vaccine (IPV) in the poliomyelitis eradica-
A second-generation vector was engineered by deletion of tion program.
the fusion glycoprotein (F) gene, making the virus non- Another picornavirus, rhinovirus, recently has been engi-
replicative. Macaques which were primed with a DNA-Gag neered to express the 2F5 epitope on its surface. Intranasal
vaccine then boosted with a single immunization with a administration of the recombinant virus induced broadly neu-
SeVF-HIV-1 Gag recombinant fully controlled virus repli- tralizing antibodies, which, if confirmed, would represent a
cation and CD4+ T-cell loss after challenge with pathogenic remarkable achievement [273].
SHIV 89.6P and remained with undetectable virus load levels It is too early at this time to evaluate the chances of success
during 1 year follow-up [226,274]. of these various vector-based vaccine approaches.
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4071

5.6. The development of prime-boost combinations fashion have been explored as a potential strategy to gener-
ate diverse CTL responses and bypass the natural hierarchy
The experience gained so far with the first generation of epitope bias [238,239]. Multi-epitope DNA immunogens
of candidate HIV vaccines has been that many were mod- can efficiently prime for broadly reactive CTL responses
estly immunogenic and only induced short-lived immune [89,240,241], but do not necessarily overcome hierarchies
responses. One of the strategies experienced over the last of epitope dominance [242]. A multiepitope DNA vaccine
decade to increase their immunogenicity was to combine developed by Epimmune is currently undergoing a Phase I
these vaccines in “prime-boost” vaccination regimens, ini- trial in the USA and Peru and will be followed by booster
tially using DNA vaccine candidates for priming followed immunization with a multiepitope recombinant fusion pro-
by live viral vectored vaccines for boosting [23,175,235]. tein.
Another type of prime-boost vaccine regimen was developed Induction of persistent HIV Gag-specific CD8+ CTL
using two different live recombinant vectors expressing the responses was evaluated in a Phase I trial involving immu-
same antigens, such as an Ad5 vector followed by a poxvirus nization with a fusion protein comprising the HIV p24Gag
vector [236], or two successive adenovirus vectors, such as protein and detoxified Bacillus anthracis lethal factor to
Ad11 and Ad35, or two succcessive poxviruses, such as MVA target antigen-presenting cells (Avant Therapeutics and
and FPV, whose combination has been tested in Phase I trials WRAIR) [243].
in the USA and Brazil by Therion in collaboration with the Synthetic lipopeptides containing MHC class I-restricted
NIAID. Heterologous adenovirus prime-boost vaccine reg- T-cell epitopes were found to induce strong CD8+ T-cell
imens using successively Ad11 and Ad35 vectors, or the responses against HIV in mice, non-human primates and
same vectors in reverse order, were found to elicit in mice humans without additional adjuvant [244–246]. Lipopeptides
higher frequency immune responses than homologous regi- with sequences corresponding to that of CTL epitope-rich
mens [237], emphasizing the advantage of multiple vector- regions in the HIV-1 Gag and Nef proteins were tested in
based vaccines. In rhesus monkeys, responses arising from an Phase I trials and were shown to induce strong, multiepitopic
Ad5-poxvirus (MVA or ALVAC) prime-boost regimen were CD4+ and CD8+ T-cell responses. Parallel Phase II trials
significantly greater than those elicited by homologous reg- were started in 2004 in the USA and in France under the
imens with the individual vectors. However, the increased sponsorship of NIAID and ANRS, respectively, to study the
immunogenicity observed in monkeys was not confirmed in efficiency of lipopeptides as priming or boosting immuno-
human volunteers. Combinations of different vectors in het- gens, but the trials had to be interrupted due to the occurrence
erologous prime-boost regimens are likely to be developed of a severe neurological side effect in one of the US volun-
in the future to circumvent the problem of anti-vector immu- teers. The trial now has resumed in France.
nity, which follows immunization with any live recombinant
vaccine.
Another prime-boost strategy has been developed in the 6. Concluding remarks
hope to strengthen both the humoral and cellular immune
responses to vaccination, by combining a T-cell stimulating The history of the HIV pandemic is now well into its
vaccine such as recombinant plasmid DNA or a live recombi- third decade. Tremendous progress has been made in our
nant vectored vaccine with a subunit vaccine, especially one understanding of the complex interaction between HIV and
based on envelope proteins (gp120, gp140 or gp41) [203]. the host immune system that has laid ground for the devel-
There is no strong evidence, however, that such a dual vacci- opment of new, potent antiviral drugs able to control virus
nation regimen resulted in a significantly higher antibody or replication. However, many basic questions that bear on the
T-cell response. feasibility of developing an HIV vaccine still remain unan-
For any prime-boost strategy to be commercially feasi- swered, including identification of protective immune mech-
ble, the combined regimen needs to demonstrate significantly anisms, addressing the high variability of the virus and its
greater efficacy over single modality vaccines in order to bal- ability to evade immune responses and eliciting potent virus-
ance the increased costs and complexities associated with neutralizing antibodies with broad specificity against primary
developing two vaccines, including potential regulatory and HIV-1 isolates [10,11,14–16,247,248].
licensing problems, as well as logistical hurdles with the The experience with current viral vaccines suggests that
delivery of the vaccines in the field. an effective vaccine or vaccine regimen against HIV infec-
tion might need to induce both neutralizing antibodies and
5.7. Fusion proteins and peptides cell-mediated immunity. One of the challenges for HIV vac-
cine research, therefore, is to understand how to design
Multiepitopic combinations of peptides, fusion proteins envelope immunogens to effectively elicit long-lasting high
and long lipopeptides are at an early stage of clinical devel- titer broadly neutralizing antibodies. Meanwhile, the atten-
opment, either alone or in prime-boost combinations with live tion of the AIDS vaccine field has focused on the induc-
vector-based recombinant vaccines. Vaccine constructs that tion of HIV-specific cellular immune responses, including
express a series of minimal epitopes arranged in a string-like CTL, based on the hypothesis that such a response would
4072 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

hopefully enable the vaccinated persons to better control Last, but not least, it would seem of paramount importance
their virus load following infection, slow down or even pre- to develop HIV vaccines that stimulate the mucosal immune
clude their progression to AIDS and decrease the probability system so as to block the major virus transmission route. The
of secondary transmission. This hypothesis has been con- first cellular targets of HIV-1 are CD4+ CCR-5+ memory T
firmed in the SHIV/macaque model using a variety of T- cells that are mostly found in the mucosal lymphoid tissue
cell-stimulating vaccines, but not in the SIV/macaque model, of the GALT, where the early phase of HIV infection and
where virus control following vaccination and challenge replication take place [110–114,255]. The need, therefore, is
often appears to be short-lived, ultimately leading to vaccine to develop vaccines capable of raising an immune barrier at
failure [122,123,201,202,274]. the site of the genital, rectal and intestinal mucosa to effi-
These vaccines, therefore, urgently need to be tested in ciently prevent HIV-1 infection. At this time, however, the
clinical trials on human volunteers at risk [15]. Of note is the design of such vaccines remains a major challenge [256].
technical challenge associated with the design and follow- HIV-1-specific mucosal CTLs could be efficiently induced
up of large Phase III trials of vaccines that are not meant to in the intestinal mucosa by intrarectal administration of a
induce protection against infection. Indeed, although the cor- synthetic peptide vaccine incorporating a detoxified E. coli
relation between viral loads at early times after infection and heat-labile toxin, LT(R192G). Following a SHIV challenge,
speed of evolution of the disease are well documented in nat- the mucosally immunized monkeys readily cleared the virus
ural infection, the translation of this correlation in vaccinated to undetectable levels both in blood and in the intestine
populations remains to be established. In addition, a number and did not experience any CD4+ T-cell depletion [257].
of questions remains unanswered at present, like the nature Mucosal HIV vaccine delivery should be considered among
of the best end points for long-term follow-up of volunteers the most effective immunization strategies for the induc-
in Phase III clinical trials, and the time during which volun- tion of mucosal CTL that could provide early protection
teers will need to be monitored. These technical questions against HIV replication and amplification [258,259]. Immu-
are accompanied by a number of ethical questions relating to nization of macaque monkeys by the nasal or oral route using
standard of care and duration of the sponsors’ responsibility a VSV vectored vaccine expressing SIV antigens Gag and
for providing this care. The population-wide effects of vac- Env resulted in significant protection against SIV infection
cines that do not prevent infection but only reduce viral load [222]. Alphavirus replicons represent another type of recom-
levels in vaccinees are largely unknown, but a few studies binant vaccine that can be administered efficiently by the
based upon mathematical models suggest that even a 1-log mucosal route [260]. The administration of a HIV-1 VLP
reduction in viral load, which is a reasonable expectation vaccine by the mucosal route is under study [147,148]. The
with the currently available candidate vaccines, would have tonsillar route of immunization has been explored in macaque
a major impact and significantly reduce HIV mortality within monkeys using tonsillar sprays of SIV vaccines [261,262].
20 years after introduction of the vaccine [249]. Generally speaking, the nasal route of immunization is prob-
Among the great many hurdles that are met in the develop- ably the most advantageous one, both from the point of view
ment of HIV vaccines is the lack of scientific knowledge on of public acceptance and for its potential efficacy, but the use
the nature and level of the immune responses that would be of nonliving vaccines by the nasal route still raises a few prob-
required to achieve protection against HIV infection and/or lems including that of adjuvants [263–265] The production of
development of the disease. The most sensible strategies mucosally targeted immunogens also has been attempted in
adopted todate in HIV vaccine research therefore aim at elicit- plants. A chimeric protein was expressed in plants that com-
ing an effective response of both arms of the immune system. bines the GM1 ganglioside-binding B subunit of the cholera
One way to increase the immune response to HIV would be to toxin (CTB) fused to a peptide (P1) which spans the 2F5
help target the antigens to the antigen-presenting cells, espe- and 4E10 epitopes and the galactosyl ceramide-binding site
cially dendritic cells (DC). Coupling the antigen of choice to a at the C-terminus of the ectodomain of HIV-1 gp41 [266].
DC-specific monoclonal antibody [250] has yielded impres- The CTB-P1 fusion protein was found to induce vaginal and
sive results in mice, enhancing the efficiency and kinetics of intestinal antibody responses after intranasal immunization
the immune response as compared with soluble antigen. This in mice. This might pave the way to a new generation of
approach is now being studied in nonhuman primates. mucosally administered HIV vaccines.
Another player in the immune response, which proba- Most of the efforts to develop and evaluate HIV vaccines
bly has not received enough attention so far, is the CD4+ are borne by the National Institute of Allergy and Infectious
effector T cells, which seem to play a critical role in the pro- Diseases (NIAID), the Centers for Disease Control (CDC)
tection provided by the live attenuated SIVnef vaccine in and the Walter Reed Army Institute for Research (WRAIR)
macaques (Johnson P, personal communication), and which in the USA, as well as by the French National AIDS Research
also were implicated as a correlate of protection in long- Agency (ANRS) in France, the International AIDS Vaccine
term non-progressors [251–254]. The CD4+ T-cell response Initiative (IAVI) in New York, the European Union, initia-
to vaccines might be an important correlate of potential vac- tives at WHO and UNAIDS, and the African AIDS Vaccine
cine efficacy that should more systemically be taken into Programme (AAVP). The recent commitment of the Bill and
acount. Melinda Gates Foundation has resulted in the foundation of
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4073

a Global HIV Vaccine Enterprise [267]. The creation of a [11] Burton DR, Desrosiers RC, Doms RW, Koff WC, Kwong PD,
Center for HIV/AIDS Immunology (CHAVI), which could Moore JP, et al. HIV vaccine design and the neutralizing antibody
problem. Nat Immunol 2004;5(3):233–6.
receive more than US$300 million from the NIAID over
[12] Excler JL, Beyrer C. Human immunodeficiency virus vaccine
the next 6 years, is part of this novel effort to speed up and development in developing countries: are efficacy trials feasible? J
coordinate the search for an HIV vaccine [268]. The HIV Vac- Hum Virol 2000;3(4):193–214.
cine Trial Network (HVTN) established by NIAID in 2000, [13] Esparza J, Osmanov S, Pattou-Markovic C, Toure C, Chang ML,
with 25 clinical sites on four continents, represents a major Nixon S. Past, present and future of HIV vaccine trials in devel-
resource for clinical HIV vaccine research. The European oping countries. Vaccine 2002;20(15):1897–8.
[14] Emini EA, Koff WC. AIDS/HIV. Developing an AIDS vaccine:
Union has similarly created the European and Developing need, uncertainty, hope. Science 2004;304(5679):1913–4.
Countries Clinical Trials Partnership (EDCTP) with the aim [15] Garber DA, Silvestri G, Feinberg MB. Prospects for an AIDS
to help developing countries strengthen their capacity in test- vaccine: three big questions, no easy answers. Lancet Infect Dis
ing the efficacy of new drugs, microbicides, and vaccines. 2004;4(7):397–413.
Strong efforts are indeed needed to harmonize vaccine trials [16] Lemckert AA, Goudsmit J, Barouch DH. Challenges in the search
for an HIV vaccine. Eur J Epidemiol 2004;19(6):513–6.
and help to define the next clinical trial step in a ‘learning [17] Excler JL. AIDS vaccine development: perspectives, challenges and
by doing’ procedure based on continuous vaccine improve- hopes. Ind J Med Res 2005;121(4):568–81.
ment in iterative clinical trial processes [269]. Developing [18] Buve A, Bishikwabo-Nsarhaza K, Mutangadura G. The spread
countries are essential partners in this international effort. and effect of HIV-1 infection in sub-Saharan Africa. Lancet
There is little doubt that the development of a safe, effec- 2002;359(9322):2011–7.
[19] Drobniewski FA, Balabanova YM, Ruddy MC, Graham C,
tive, and affordable HIV vaccine constitutes a top priority for Kuznetzov SI, Gusarova GI, et al. Tuberculosis, HIV seropreva-
HIV/AIDS research and a formidable scientific and public lence and intravenous drug abuse in prisoners. Eur Respir J
health challenge at the dawn of this century. 2005;26(2):298–304.
[20] Tatt ID, Barlow KL, Nicoll A, Clewley JP. The public health sig-
nificance of HIV-1 subtypes. AIDS 2001;15(Suppl. 5):S59–71.
[21] McCutchan FE. In: Thompson RCA, editor. The Molecular Epi-
Acknowledgement demiology of Infectious Diseases. London: Holder, A; 2000. p.
143–67.
The efficient editorial help of Olga Assossou is gratefully [22] McCutchan FE. Understanding the genetic diversity of HIV-1.
acknowledged. AIDS 2000;14(Suppl. 3):S31–44.
[23] Robinson HL. New hope for an AIDS vaccine. Nat Rev Immunol
2002;2(4):239–50.
[24] Goldstein G. HIV-1 Tat protein as a potential AIDS vaccine. Nat
References Med 1996;2(9):960–4.
[25] Pomerantz RJ, Seshamma T, Trono D. Efficient replication of
[1] Gao F, Bailes E, Robertson DL, Chen Y, Rodenburg CM, Michael human immunodeficiency virus type 1 requires a threshold level of
SF, et al. Origin of HIV-1 in the chimpanzee Pan troglodytes Rev: potential implications for latency. J Virol 1992;66(3):1809–13.
troglodytes. Nature 1999;397(6718):436–41. [26] Renkema GH, Manninen A, Saksela K. Human immunodefi-
[2] Korber B, Muldoon M, Theiler J, Gao F, Gupta R, Lapedes A, ciency virus type 1 Nef selectively associates with a catalytically
et al. Timing the ancestor of the HIV-1 pandemic strains. Science active subpopulation of p21-activated kinase 2 (PAK2) indepen-
2000;288(5472):1789–96. dently of PAK2 binding to Nck or beta-PIX. J Virol 2001;75(5):
[3] Joint United Nations Programme on HIV/AIDS (UNAIDS). Report 2154–60.
on the global AIDS epidemic, Geneva Switzerland, December [27] James CO, Huang MB, Khan M, Garcia-Barrio M, Powell MD,
2005. Bond VC. Extracellular Nef protein targets CD4+ T cells for
[4] Girard M, Mastro TD, Koff WC. Human Immunodeficiency Virus. apoptosis by interacting with CXCR4 surface receptors. J Virol
In: Plotkin SA, Orenstein WA, editors. Vaccines. Philadelphia: 2004;78(6):3099–109.
Saunders; 2004. p. 1219–58. [28] Yu X, Yu Y, Liu B, Luo K, Kong W, Mao P, et al. Induc-
[5] Cohen J. Public health. AIDS vaccine trial produces disappointment tion of APOBEC3G ubiquitination and degradation by an HIV-1
and confusion. Science 2003;299(5611):1290–1. Vif–Cul5–SCF complex. Science 2003;302(5647):1056–60.
[6] McNeil JG, Johnston MI, Birx DL, Tramont EC. Policy rebuttal. [29] Lahti AL, Manninen A, Saksela K. Regulation of T cell activation
HIV vaccine trial justified. Science 2004;303(5660):961. by HIV-1 accessory proteins: Vpr acts via distinct mechanisms
[7] McCutchan FE, Viputtigul K, de Souza MS, Carr JK, Markowitz to cooperate with Nef in NFAT-directed gene expression and to
LE, Buapunth P, et al. Diversity of envelope glycoprotein from promote transactivation by CREB. Virology 2003;310(1):190–6.
human immunodeficiency virus type 1 of recent seroconverters in [30] Tardif MR, Tremblay MJ. Presence of host ICAM-1 in human
Thailand. AIDS Res Hum Retroviruses 2000;16(8): 801–5. immunodeficiency virus type 1 virions increases productive infec-
[8] Korber B, Gaschen B, Yusim K, Thakallapally R, Kesmir C, tion of CD4+ T lymphocytes by favoring cytosolic delivery of viral
Detours V. Evolutionary and immunological implications of con- material. J Virol 2003;77(22):12299–309.
temporary HIV-1 variation. Br Med Bull 2001;58:19–42. [31] Kuznetsov YG, Victoria JG, Robinson Jr WE, McPherson A.
[9] Lifson JD, Rossio JL, Arnaout R, Li L, Parks TL, Schneider DK, Atomic force microscopy investigation of human immunode-
et al. Containment of simian immunodeficiency virus infection: ficiency virus (HIV) and HIV-infected lymphocytes. J Virol
cellular immune responses and protection from rechallenge fol- 2003;77(22):11896–909.
lowing transient postinoculation antiretroviral treatment. J Virol [32] Brelot A, Alizon M. HIV-1 entry and how to block it. AIDS
2000;74(6):2584–93. 2001;15(Suppl. 5):S3–11.
[10] Desrosiers RC. Prospects for an AIDS vaccine. Nat Med 2004; [33] Chan DC, Kim PS. HIV entry and its inhibition. Cell 1998;93(5):
10(3):221–3. 681–4.
4074 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

[34] Eckert DM, Kim PS. Mechanisms of viral membrane fusion and [51] Barbato G, Bianchi E, Ingallinella P, Hurni WH, Miller MD, Cilib-
its inhibition. Annu Rev Biochem 2001;70:777–810. erto G, et al. Structural analysis of the epitope of the anti-HIV
[35] Markovic I, Clouse KA. Recent advances in understanding the antibody 2F5 sheds light into its mechanism of neutralization and
molecular mechanisms of HIV-1 entry and fusion: revisiting current HIV fusion. J Mol Biol 2003;330(5):1101–15.
targets and considering new options for therapeutic intervention. [52] Ofek G, Tang M, Sambor A, Katinger H, Mascola JR, Wyatt R, et
Curr HIV Res 2004;2(3):223–34. al. Structure and mechanistic analysis of the anti-human immun-
[36] Desmezieres E, Gupta N, Vassell R, He Y, Peden K, Sirota L, et al. odeficiency virus type 1 antibody 2F5 in complex with its gp41
Human immunodeficiency virus (HIV) gp41 escape mutants: cross- epitope. J Virol 2004;78(19):10724–37.
resistance to peptide inhibitors of HIV fusion and altered receptor [53] Cardoso RM, Zwick MB, Stanfield RL, Kunert R, Binley JM,
activation of gp120. J Virol 2005;79(8):4774–81. Katinger H, et al. Broadly neutralizing anti-HIV antibody 4E10
[37] Zolla-Pazner S. Identifying epitopes of HIV-1 that induce protective recognizes a helical conformation of a highly conserved fusion-
antibodies. Nat Rev Immunol 2004;4(3):199–210. associated motif in gp41. Immunity 2005;22(2):163–73.
[38] Kessler II JA, McKenna PM, Emini EA, Chan CP, Patel MD, Gupta [54] Haynes BF, Fleming J, St Clair EW, Katinger H, Stiegler G, Kunert
SK, et al. Recombinant human monoclonal antibody IgG1b12 neu- R, et al. Cardiolipin polyspecific autoreactivity in two broadly neu-
tralizes diverse human immunodeficiency virus type 1 primary tralizing HIV-1 antibodies. Science 2005;308(5730):1906–8.
isolates. AIDS Res Hum Retroviruses 1997;13(7):575–82. [55] Nabel GJ. Immunology. Close to the edge: neutralizing the HIV-1
[39] Scanlan CN, Pantophlet R, Wormald MR, Ollmann Saphire E, Stan- envelope. Science 2005;308(5730):1878–9.
field R, Wilson IA, et al. The broadly neutralizing anti-human [56] Wang B, Lal RB, Dwyer DE, Miranda-Saksena M, Boadle R, Cun-
immunodeficiency virus type 1 antibody 2G12 recognizes a cluster ningham AL, et al. Molecular and biological interactions between
of alpha1→2 mannose residues on the outer face of gp120. J Virol two HIV-1 strains from a coinfected patient reveal the first evidence
2002;76(14):7306–21. in favor of viral synergism. Virology 2000;274(1):105–19.
[40] D’Souza MP, Milman G, Bradac JA, McPhee D, Hanson CV, [57] Altfeld M, Allen TM, Yu XG, Johnston MN, Agrawal D, Kor-
Hendry RM. Neutralization of primary HIV-1 isolates by anti- ber BT, et al. HIV-1 superinfection despite broad CD8+ T-cell
envelope monoclonal antibodies. AIDS 1995;9(8):867–74. responses containing replication of the primary virus. Nature
[41] Ugolini S, Mondor I, Parren PW, Burton DR, Tilley SA, Klasse 2002;420(6914):434–9.
PJ, et al. Inhibition of virus attachment to CD4+ target cells is [58] McCutchan FE, Hoelscher M, Tovanabutra S, Piyasirisilp S,
a major mechanism of T cell line-adapted HIV-1 neutralization. J Sanders-Buell E, Ramos G, et al. In-depth analysis of a heterosexu-
Exp Med 1997;186(8):1287–98. ally acquired human immunodeficiency virus type 1 superinfection:
[42] Zwick MB, Bonnycastle LL, Menendez A, Irving MB, Barbas evolution, temporal fluctuation, and intercompartment dynamics
III CF, Parren PW, et al. Identification and characterization of from the seronegative window period through 30 months postin-
a peptide that specifically binds the human, broadly neutralizing fection. J Virol 2005;79(18):11693–704.
anti-human immunodeficiency virus type 1 antibody b12. J Virol [59] Kim EY, Busch M, Abel K, Fritts L, Bustamante P, Stanton J,
2001;75(14):6692–9. et al. Retroviral recombination in vivo: viral replication patterns
[43] Xiang SH, Doka N, Choudhary RK, Sodroski J, Robinson and genetic structure of simian immunodeficiency virus (SIV)
JE. Characterization of CD4-induced epitopes on the HIV populations in rhesus macaques after simultaneous or sequen-
type 1 gp120 envelope glycoprotein recognized by neutraliz- tial intravaginal inoculation with SIVmac239Deltavpx/Deltavpr and
ing human monoclonal antibodies. AIDS Res Hum Retroviruses SIVmac239Deltanef. J Virol 2005;79(8):4886–95.
2002;18(16):1207–17. [60] Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K,
[44] Koch M, Pancera M, Kwong PD, Kolchinsky P, Grundner C, Wang Pierson T, et al. Latent infection of CD4+ T cells provides a mecha-
L, et al. Structure-based, targeted deglycosylation of HIV-1 gp120 nism for lifelong persistence of HIV-1, even in patients on effective
and effects on neutralization sensitivity and antibody recognition. combination therapy. Nat Med 1999;5(5):512–7.
Virology 2003;313(2):387–400. [61] Blankson JN, Persaud D, Siliciano RF. The challenge of viral reser-
[45] Kwong PD, Doyle ML, Casper DJ, Cicala C, Leavitt SA, voirs in HIV-1 infection. Annu Rev Med 2002;53:557–93.
Majeed S, et al. HIV-1 evades antibody-mediated neutralization [62] Peterlin BM, Trono D. Hide, shield and strike back: how
through conformational masking of receptor-binding sites. Nature HIV-infected cells avoid immune eradication. Nat Rev Immunol
2002;420(6916):678–82. 2003;3(2):97–107.
[46] Labrijn AF, Poignard P, Raja A, Zwick MB, Delgado K, Franti [63] Miller CJ, Li Q, Abel K, Kim EY, Ma ZM, Wietgrefe S, et al.
M, et al. Access of antibody molecules to the conserved core- Propagation and dissemination of infection after vaginal transmis-
ceptor binding site on glycoprotein gp120 is sterically restricted sion of simian immunodeficiency virus. J Virol 2005;79(14):9217–
on primary human immunodeficiency virus type 1. J Virol 27.
2003;77(19):10557–65. [64] Gallo RC. The end or the beginning of the drive to an HIV-
[47] Huang CC, Tang M, Zhang MY, Majeed S, Montabana E, Stanfield preventive vaccine: a view from over 20 years. Lancet 2005;
RL, et al. Structure of a V3-containing HIV-1 gp120 core. Science 366(9500):1894–8.
2005;310(5750):1025–8. [65] Evans DT, Desrosiers RC. Immune evasion strategies of the primate
[48] Trkola A, Pomales AB, Yuan H, Korber B, Maddon PJ, Allaway lentiviruses. Immunol Rev 2001;183:141–58.
GP, et al. Cross-clade neutralization of primary isolates of human [66] O’Connor D, Friedrich T, Hughes A, Allen TM, Watkins D. Under-
immunodeficiency virus type 1 by human monoclonal antibodies standing cytotoxic T-lymphocyte escape during simian immunode-
and tetrameric CD4-IgG. J Virol 1995;69(11):6609–17. ficiency virus infection. Immunol Rev 2001;183:115–26.
[49] Parker CE, Deterding LJ, Hager-Braun C, Binley JM, Schulke [67] Goulder PJ, Watkins DI. HIV and SIV CTL escape: implications
N, Katinger H, et al. Fine definition of the epitope on the for vaccine design. Nat Rev Immunol 2004;4(8):630–40.
gp41 glycoprotein of human immunodeficiency virus type 1 for [68] Feeney ME, Tang Y, Pfafferott K, Roosevelt KA, Draenert
the neutralizing monoclonal antibody 2F5. J Virol 2001;75(22): R, Trocha A, et al. HIV-1 viral escape in infancy followed
10906–11. by emergence of a variant-specific CTL response. J Immunol
[50] Zwick MB, Labrijn AF, Wang M, Spenlehauer C, Saphire EO, 2005;174(12):7524–30.
Binley JM, et al. Broadly neutralizing antibodies targeted to the [69] Richman DD, Wrin T, Little SJ, Petropoulos CJ. Rapid evolution
membrane-proximal external region of human immunodeficiency of the neutralizing antibody response to HIV type 1 infection. Proc
virus type 1 glycoprotein gp41. J Virol 2001;75(22):10892–905. Natl Acad Sci U S A 2003;100(7):4144–9.
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4075

[70] Wei X, Decker JM, Wang S, Hui H, Kappes JC, Wu X, through passive transfer of human neutralizing antibodies. Nat Med
et al. Antibody neutralization and escape by HIV-1. Nature 2005;11(6):615–22.
2003;422(6929):307–12. [87] Berman PW, Gregory TJ, Riddle L, Nakamura GR, Champe MA,
[71] Wyatt R, Sodroski J. The HIV-1 envelope glycoproteins: fusogens, Porter JP, et al. Protection of chimpanzees from infection by HIV-
antigens, and immunogens. Science 1998;280(5371):1884–8. 1 after vaccination with recombinant glycoprotein gp120 but not
[72] Parren PW, Moore JP, Burton DR, Sattentau QJ. The neutralizing gp160. Nature 1990;345(6276):622–5.
antibody response to HIV-1: viral evasion and escape from humoral [88] Girard M, Meignier B, Barre-Sinoussi F, Kieny MP, Matthews T,
immunity. AIDS 1999;13(Suppl. A):S137–62. Muchmore E, et al. Vaccine-induced protection of chimpanzees
[73] Calarota SA, Weiner DB. Present status of human HIV vaccine against infection by a heterologous human immunodeficiency virus
development. AIDS 2003;17(Suppl. 4):S73–84. type 1. J Virol 1995;69(10):6239–48.
[74] Gorny MK, Revesz K, Williams C, Volsky B, Louder MK, [89] McMichael A, Hanke T. The quest for an AIDS vaccine: is
Anyangwe CA, et al. The v3 loop is accessible on the surface of the CD8+ T-cell approach feasible? Nat Rev Immunol 2002;2(4):
most human immunodeficiency virus type 1 primary isolates and 283–91.
serves as a neutralization epitope. J Virol 2004;78(5):2394–404. [90] McMichael AJ, Rowland-Jones SL. Cellular immune responses to
[75] Yang X, Tomov V, Kurteva S, Wang L, Ren X, Gorny MK, et HIV. Nature 2001;410(6831):980–7.
al. Characterization of the outer domain of the gp120 glycoprotein [91] Letvin NL. Strategies for an HIV vaccine. J Clin Invest 2002;
from human immunodeficiency virus type 1. J Virol 2004;78(23): 110(1):15–20.
12975–86. [92] Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky
[76] Mascola JR, Snyder SW, Weislow OS, Belay SM, Belshe RB, W, et al. Temporal association of cellular immune responses with
Schwartz DH, et al. Immunization with envelope subunit vaccine the initial control of viremia in primary human immunodeficiency
products elicits neutralizing antibodies against laboratory-adapted virus type 1 syndrome. J Virol 1994;68(7):4650–5.
but not primary isolates of human immunodeficiency virus type 1. [93] Kuroda MJ, Schmitz JE, Charini WA, Nickerson CE, Lifton MA,
The National Institute of Allergy and Infectious Diseases AIDS Lord CI, et al. Emergence of CTL coincides with clearance of virus
Vaccine Evaluation Group. J Infect Dis 1996;173(2):340–8. during primary simian immunodeficiency virus infection in rhesus
[77] Shibata R, Igarashi T, Haigwood N, Buckler-White A, Ogert R, monkeys. J Immunol 1999;162(9):5127–33.
Ross W, et al. Neutralizing antibody directed against the HIV-1 [94] Frahm N, Adams S, Kiepiela P, Linde CH, Hewitt HS, Lichter-
envelope glycoprotein can completely block HIV-1/SIV chimeric feld M, et al. HLA-B63 presents HLA-B57/B58-restricted cytotoxic
virus infections of macaque monkeys. Nat Med 1999;5(2):204–10. T-lymphocyte epitopes and is associated with low human immun-
[78] Mascola JR, Lewis MG, Stiegler G, Harris D, VanCott TC, Hayes odeficiency virus load. J Virol 2005;79(16):10218–25.
D, et al. Protection of Macaques against pathogenic simian/human [95] Borrow P, Lewicki H, Wei X, Horwitz MS, Peffer N, Meyers
immunodeficiency virus 89.6PD by passive transfer of neutralizing H, et al. Antiviral pressure exerted by HIV-1-specific cytotoxic
antibodies. J Virol 1999;73(5):4009–18. T lymphocytes (CTLs) during primary infection demonstrated by
[79] Mascola JR, Stiegler G, VanCott TC, Katinger H, Carpenter CB, rapid selection of CTL escape virus. Nat Med 1997;3(2):205–
Hanson CE, et al. Protection of macaques against vaginal transmis- 11.
sion of a pathogenic HIV-1/SIV chimeric virus by passive infusion [96] Evans DT, O’Connor DH, Jing P, Dzuris JL, Sidney J, da Silva
of neutralizing antibodies. Nat Med 2000;6(2):207–10. J, et al. Virus-specific cytotoxic T-lymphocyte responses select for
[80] Baba TW, Liska V, Hofmann-Lehmann R, Vlasak J, Xu W, Ayehu- amino-acid variation in simian immunodeficiency virus Env and
nie S, et al. Human neutralizing monoclonal antibodies of the IgG1 Nef. Nat Med 1999;5(11):1270–6.
subtype protect against mucosal simian-human immunodeficiency [97] Allen TM, O’Connor DH, Jing P, Dzuris JL, Mothe BR, Vogel
virus infection. Nat Med 2000;6(2):200–6. TU, et al. Tat-specific cytotoxic T lymphocytes select for SIV
[81] Parren PW, Marx PA, Hessell AJ, Luckay A, Harouse J, Cheng- escape variants during resolution of primary viraemia. Nature
Mayer C, et al. Antibody protects macaques against vaginal chal- 2000;407(6802):386–90.
lenge with a pathogenic R5 simian/human immunodeficiency virus [98] Barouch DH, Kunstman J, Kuroda MJ, Schmitz JE, Santra S,
at serum levels giving complete neutralization in vitro. J Virol Peyerl FW, et al. Eventual AIDS vaccine failure in a rhesus
2001;75(17):8340–7. monkey by viral escape from cytotoxic T lymphocytes. Nature
[82] Letvin NL, Robinson S, Rohne D, Axthelm MK, Fanton JW, Bilska 2002;415(6869):335–9.
M, et al. Vaccine-elicited V3 loop-specific antibodies in rhesus [99] Matano T, Shibata R, Siemon C, Connors M, Lane HC, Martin
monkeys and control of a simian-human immunodeficiency virus MA. Administration of an anti-CD8 monoclonal antibody inter-
expressing a primary patient human immunodeficiency virus type feres with the clearance of chimeric simian/human immunodefi-
1 isolate envelope. J Virol 2001;75(9):4165–75. ciency virus during primary infections of rhesus macaques. J Virol
[83] Nishimura Y, Igarashi T, Haigwood NL, Sadjadpour R, Donau OK, 1998;72(1):164–9.
Buckler C, et al. Transfer of neutralizing IgG to macaques 6 h [100] Schmitz JE, Kuroda MJ, Santra S, Sasseville VG, Simon
but not 24 h after SHIV infection confers sterilizing protection: MA, Lifton MA, et al. Control of viremia in simian immun-
implications for HIV-1 vaccine development. Proc Natl Acad Sci odeficiency virus infection by CD8+ lymphocytes. Science
U S A 2003;100(25):15131–6. 1999;283(5403):857–60.
[84] Nishimura Y, Igarashi T, Haigwood N, Sadjadpour R, Plishka [101] Lifson JD, Rossio JL, Piatak Jr M, Parks T, Li L, Kiser R, et
RJ, Buckler-White A, et al. Determination of a statistically valid al. Role of CD8(+) lymphocytes in control of simian immunodefi-
neutralization titer in plasma that confers protection against simian- ciency virus infection and resistance to rechallenge after transient
human immunodeficiency virus challenge following passive trans- early antiretroviral treatment. J Virol 2001;75(21):10187–99.
fer of high-titered neutralizing antibodies. J Virol 2002;76(5): [102] Peeters M, Courgnaud V, Abela B, Auzel P, Pourrut X, Bibollet-
2123–30. Ruche F, et al. Risk to human health from a plethora of simian
[85] Ferrantelli F, Rasmussen RA, Hofmann-Lehmann R, Xu W, immunodeficiency viruses in primate bushmeat. Emerg Infect Dis
McClure HM, Ruprecht RM. Do not underestimate the power of 2002;8(5):451–7.
antibodies—lessons from adoptive transfer of antibodies against [103] Heeney JL. Primate models for AIDS vaccine development. AIDS
HIV. Vaccine 2002;20(Suppl. 4):A61–5. 1996;10(Suppl. A):S115–22.
[86] Trkola A, Kuster H, Rusert P, Joos B, Fischer M, Leemann C, et [104] Hel Z, Venzon D, Poudyal M, Tsai WP, Giuliani L, Woodward R,
al. Delay of HIV-1 rebound after cessation of antiretroviral therapy et al. Viremia control following antiretroviral treatment and thera-
4076 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

peutic immunization during primary SIV251 infection of macaques. [121] Staprans SI, Feinberg MB. The roles of nonhuman primates in
Nat Med 2000;6(10):1140–6. the preclinical evaluation of candidate AIDS vaccines. Expert Rev
[105] Parker RA, Regan MM, Reimann KA. Variability of viral load Vaccines 2004;3(Suppl. 4):S5–32.
in plasma of rhesus monkeys inoculated with simian immunodefi- [122] Horton H, Vogel TU, Carter DK, Vielhuber K, Fuller DH, Ship-
ciency virus or simian-human immunodeficiency virus: implications ley T, et al. Immunization of rhesus macaques with a DNA
for using nonhuman primate AIDS models to test vaccines and prime/modified vaccinia virus Ankara boost regimen induces broad
therapeutics. J Virol 2001;75(22):11234–8. simian immunodeficiency virus (SIV)-specific T-cell responses and
[106] Goulder PJ, Altfeld MA, Rosenberg ES, Nguyen T, Tang Y, reduces initial viral replication but does not prevent disease pro-
Eldridge RL, et al. Substantial differences in specificity of HIV- gression following challenge with pathogenic SIVmac239. J Virol
specific cytotoxic T cells in acute and chronic HIV infection. J 2002;76(14):7187–202.
Exp Med 2001;193(2):181–94. [123] Vogel TU, Reynolds MR, Fuller DH, Vielhuber K, Shipley T,
[107] Veazey RS, DeMaria M, Chalifoux LV, Shvetz DE, Pauley DR, Fuller JT, et al. Multispecific vaccine-induced mucosal cytotoxic T
Knight HL, et al. Gastrointestinal tract as a major site of CD4+ lymphocytes reduce acute-phase viral replication but fail in long-
T cell depletion and viral replication in SIV infection. Science term control of simian immunodeficiency virus SIVmac239. J Virol
1998;280(5362):427–31. 2003;77(24):13348–60.
[108] Veazey RS, Tham IC, Mansfield KG, DeMaria M, Forand AE, [124] Harouse JM, Gettie A, Eshetu T, Tan RC, Bohm R, Blanchard J, et
Shvetz DE, et al. Identifying the target cell in primary simian al. Mucosal transmission and induction of simian AIDS by CCR5-
immunodeficiency virus (SIV) infection: highly activated memory specific simian/human immunodeficiency virus SHIV(SF162P3). J
CD4(+) T cells are rapidly eliminated in early SIV infection in Virol 2001;75(4):1990–5.
vivo. J Virol 2000;74(1):57–64. [125] Hsu M, Harouse JM, Gettie A, Buckner C, Blanchard
[109] Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, Roederer J, Cheng-Mayer C. Increased mucosal transmission but not
M. Massive infection and loss of memory CD4+ T cells in multiple enhanced pathogenicity of the CCR5-tropic, simian AIDS-inducing
tissues during acute SIV infection. Nature 2005;434(7037):1093–7. simian/human immunodeficiency virus SHIV(SF162P3) maps to
[110] Li Q, Duan L, Estes JD, Ma ZM, Rourke T, Wang Y, et al. Peak envelope gp120. J Virol 2003;77(2):989–98.
SIV replication in resting memory CD4+ T cells depletes gut lam- [126] Vernazza PL, Eron JJ, Fiscus SA, Cohen MS. Sexual transmission
ina propria CD4+ T cells. Nature 2005;434(7037):1148–52. of HIV: infectiousness and prevention. AIDS 1999;13(2):155–66.
[111] Guadalupe M, Reay E, Sankaran S, Prindiville T, Flamm J, McNeil [127] Chakraborty H, Sen PK, Helms RW, Vernazza PL, Fiscus SA,
A, et al. Severe CD4+ T-cell depletion in gut lymphoid tissue Eron JJ, et al. Viral burden in genital secretions determines male-
during primary human immunodeficiency virus type 1 infection and to-female sexual transmission of HIV-1: a probabilistic empiric
substantial delay in restoration following highly active antiretroviral model. AIDS 2001;15(5):621–7.
therapy. J Virol 2003;77(21):11708–17. [128] Gray RH, Wawer MJ, Brookmeyer R, Sewankambo NK, Serwadda
[112] Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beil- D, Wabwire-Mangen F, et al. Probability of HIV-1 transmission per
man GJ, et al. CD4+ T cell depletion during all stages of HIV coital act in monogamous, heterosexual, HIV-1-discordant couples
disease occurs predominantly in the gastrointestinal tract. J Exp in Rakai, Uganda. Lancet 2001;357(9263):1149–53.
Med 2004;200(6):749–59. [129] McDermott AB, Mitchen J, Piaskowski S, De Souza I, Yant LJ,
[113] Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A, Stephany J, et al. Repeated low-dose mucosal simian immunod-
Hogan C, et al. Primary HIV-1 infection is associated with prefer- eficiency virus SIVmac239 challenge results in the same viral
ential depletion of CD4+ T lymphocytes from effector sites in the and immunological kinetics as high-dose challenge: a model for
gastrointestinal tract. J Exp Med 2004;200(6):761–70. the evaluation of vaccine efficacy in nonhuman primates. J Virol
[114] Veazey RS, Lackner AA. HIV swiftly guts the immune system. 2004;78(6):3140–4.
Nat Med 2005;11(5):469–70. [130] Daniel MD, Kirchhoff F, Czajak SC, Sehgal PK, Desrosiers RC.
[115] Hel Z, Nacsa J, Tryniszewska E, Tsai WP, Parks RW, Montefiori Protective effects of a live attenuated SIV vaccine with a deletion
DC, et al. Containment of simian immunodeficiency virus infection in the nef gene. Science 1992;258(5090):1938–41.
in vaccinated macaques: correlation with the magnitude of virus- [131] Whatmore AM, Cook N, Hall GA, Sharpe S, Rud EW, Cranage
specific pre- and postchallenge CD4+ and CD8+ T cell responses. MP. Repair and evolution of nef in vivo modulates simian immun-
J Immunol 2002;169(9):4778–87. odeficiency virus virulence. J Virol 1995;69(8):5117–23.
[116] Franchini G, Nacsa J, Hel Z, Tryniszewska E. Immune interven- [132] Wyand MS, Manson K, Montefiori DC, Lifson JD, John-
tion strategies for HIV-1 infection of humans in the SIV macaque son RP, Desrosiers RC. Protection by live, attenuated simian
model. Vaccine 2002;20(Suppl. 4):A52–60. immunodeficiency virus against heterologous challenge. J Virol
[117] Reimann KA, Li JT, Veazey R, Halloran M, Park IW, Karlsson GB, 1999;73(10):8356–63.
et al. A chimeric simian/human immunodeficiency virus expressing [133] Baba TW, Liska V, Khimani AH, Ray NB, Dailey PJ, Penninck
a primary patient human immunodeficiency virus type 1 isolate D, et al. Live attenuated, multiply deleted simian immunodefi-
env causes an AIDS-like disease after in vivo passage in rhesus ciency virus causes AIDS in infant and adult macaques. Nat Med
monkeys. J Virol 1996;70(10):6922–8. 1999;5(2):194–203.
[118] Reimann KA, Li JT, Voss G, Lekutis C, Tenner-Racz K, Racz P, et [134] Hofmann-Lehmann R, Vlasak J, Williams AL, Chenine AL,
al. An env gene derived from a primary human immunodeficiency McClure HM, Anderson DC, et al. Live attenuated, nef-deleted
virus type 1 isolate confers high in vivo replicative capacity to a SIV is pathogenic in most adult macaques after prolonged obser-
chimeric simian/human immunodeficiency virus in rhesus monkeys. vation. AIDS 2003;17(2):157–66.
J Virol 1996;70(5):3198–206. [135] Johnson RP, Lifson JD, Czajak SC, Cole KS, Manson KH, Glick-
[119] Joag SV, Adany I, Li Z, Foresman L, Pinson DM, Wang C, et man R, et al. Highly attenuated vaccine strains of simian immun-
al. Animal model of mucosally transmitted human immunodefi- odeficiency virus protect against vaginal challenge: inverse rela-
ciency virus type 1 disease: intravaginal and oral deposition of tionship of degree of protection with level of attenuation. J Virol
simian/human immunodeficiency virus in macaques results in sys- 1999;73(6):4952–61.
temic infection, elimination of CD4+ T cells, and AIDS. J Virol [136] Guan Y, Whitney JB, Detorio M, Wainberg MA. Construction
1997;71(5):4016–23. and in vitro properties of a series of attenuated simian immun-
[120] Feinberg MB, Moore JP. AIDS vaccine models: challenging chal- odeficiency viruses with all accessory genes deleted. J Virol
lenge viruses. Nat Med 2002;8(3):207–10. 2001;75(9):4056–67.
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4077

[137] Blancou P, Chenciner N, Ho Tsong Fang R, Monceaux V, Cumont odeficiency virus-HIV-1 chimeric virus. J Gen Virol 1998;79(Part
MC, Guetard D, et al. Simian immunodeficiency virus promoter 3):423–32.
exchange results in a highly attenuated strain that protects against [155] Graham BS, McElrath MJ, Connor RI, Schwartz DH, Gorse
uncloned challenge virus. J Virol 2004;78(3):1080–92. GJ, Keefer MC, et al. Analysis of intercurrent human immun-
[138] Jekle A, Keppler OT, De Clercq E, Schols D, Weinstein M, Gold- odeficiency virus type 1 infections in phase I and II trials of
smith MA. In vivo evolution of human immunodeficiency virus candidate AIDS vaccines. AIDS Vaccine Evaluation Group, and
type 1 toward increased pathogenicity through CXCR4-mediated the Correlates of HIV Immune Protection Group. J Infect Dis
killing of uninfected CD4 T cells. J Virol 2003;77(10):5846–54. 1998;177(2):310–9.
[139] Whitney JB, Ruprecht RM. Live attenuated HIV vaccines: pitfalls [156] Yang X, Lee J, Mahony EM, Kwong PD, Wyatt R, Sodroski J.
and prospects. Curr Opin Infect Dis 2004;17(1):17–26. Highly stable trimers formed by human immunodeficiency virus
[140] Koff WC, Johnson PR, Watkins DI, Burton DR, Lifson JD, type 1 envelope glycoproteins fused with the trimeric motif of T4
Hasenkrug KJ, et al. HIV vaccine design: insights from live atten- bacteriophage fibritin. J Virol 2002;76(9):4634–42.
uated SIV vaccines. Nat Immunol 2006;7(1):19–23. [157] Forsell MN, Li Y, Sundback M, Svehla K, Liljestrom P, Mas-
[141] Poon B, Hsu JF, Gudeman V, Chen IS, Grovit-Ferbas K. cola JR, et al. Biochemical and immunogenic characterization
Formaldehyde-treated, heat-inactivated virions with increased of soluble human immunodeficiency virus type 1 envelope gly-
human immunodeficiency virus type 1 env can be used to induce coprotein trimers expressed by semliki forest virus. J Virol
high-titer neutralizing antibody responses. J Virol 2005;79(16): 2005;79(17):10902–14.
10210–7. [158] Barnett AL, Cunningham JM. Receptor binding transforms the
[142] Poon B, Safrit JT, McClure H, Kitchen C, Hsu JF, Gude- surface subunit of the mammalian C-type retrovirus envelope
man V, et al. Induction of humoral immune responses following protein from an inhibitor to an activator of fusion. J Virol
vaccination with envelope-containing, formaldehyde-treated, ther- 2001;75(19):9096–105.
mally inactivated human immunodeficiency virus type 1. J Virol [159] Srivastava IK, Stamatatos L, Kan E, Vajdy M, Lian Y, Hilt S, et
2005;79(8):4927–35. al. Purification, characterization, and immunogenicity of a soluble
[143] Arthur LO, Bess Jr JW, Chertova EN, Rossio JL, Esser MT, Ben- trimeric envelope protein containing a partial deletion of the V2
veniste RE, et al. Chemical inactivation of retroviral infectivity by loop derived from SF162, an R5-tropic human immunodeficiency
targeting nucleocapsid protein zinc fingers: a candidate SIV vac- virus type 1 isolate. J Virol 2003;77(20):11244–59.
cine. AIDS Res Hum Retroviruses 1998;14(Suppl. 3):S311–9. [160] Srivastava IK, VanDorsten K, Vojtech L, Barnett SW, Stamatatos
[144] Rossio JL, Esser MT, Suryanarayana K, Schneider DK, Bess L. Changes in the immunogenic properties of soluble gp140
Jr JW, Vasquez GM, et al. Inactivation of human immunode- human immunodeficiency virus envelope constructs upon partial
ficiency virus type 1 infectivity with preservation of conforma- deletion of the second hypervariable region. J Virol 2003;77(4):
tional and functional integrity of virion surface proteins. J Virol 2310–20.
1998;72(10):7992–8001. [161] Lian Y, Srivastava I, Gomez-Roman VR, Zur Megede J, Sun Y,
[145] Lifson JD, Rossio JL, Piatak Jr M, Bess Jr J, Chertova E, Schneider Kan E, et al. Evaluation of envelope vaccines derived from the
DK, et al. Evaluation of the safety, immunogenicity, and protective South African subtype C human immunodeficiency virus type 1
efficacy of whole inactivated simian immunodeficiency virus (SIV) TV1 strain. J Virol 2005;79(21):13338–49.
vaccines with conformationally and functionally intact envelope [162] Li Y, Svehla K, Mathy NL, Voss G, Mascola JR, Wyatt
glycoproteins. AIDS Res Hum Retroviruses 2004;20(7):772–87. R. Characterization of antibody responses elicited by human
[146] Kang SM, Compans RW. Enhancement of mucosal immunization immunodeficiency virus type 1 primary isolate trimeric and
with virus-like particles of simian immunodeficiency virus. J Virol monomeric envelope glycoproteins in selected adjuvants. J Virol
2003;77(6):3615–23. 2006;80(3):1414–26.
[147] Kang SM, Yao Q, Guo L, Compans RW. Mucosal immunization [163] Binley JM, Sanders RW, Clas B, Schuelke N, Master A, Guo Y, et
with virus-like particles of simian immunodeficiency virus conju- al. A recombinant human immunodeficiency virus type 1 envelope
gated with cholera toxin subunit B. J Virol 2003;77(18):9823–30. glycoprotein complex stabilized by an intermolecular disulfide bond
[148] Doan LX, Li M, Chen C, Yao Q. Virus-like particles as HIV-1 between the gp120 and gp41 subunits is an antigenic mimic of the
vaccines. Rev Med Virol 2005;15(2):75–88. trimeric virion-associated structure. J Virol 2000;74(2):627–43.
[149] Hammonds J, Chen X, Fouts T, DeVico A, Montefiori D, Spearman [164] Sanders RW, Vesanen M, Schuelke N, Master A, Schiffner L,
P. Induction of neutralizing antibodies against human immunode- Kalyanaraman R, et al. Stabilization of the soluble, cleaved,
ficiency virus type 1 primary isolates by Gag-Env pseudovirion trimeric form of the envelope glycoprotein complex of human
immunization. J Virol 2005;79(23):14804–14. immunodeficiency virus type 1. J Virol 2002;76(17):8875–89.
[150] Harvey TJ, Anraku I, Linedale R, Harrich D, Mackenzie J, Suhrbier [165] Beddows S, Schulke N, Kirschner M, Barnes K, Franti M,
A, et al. Kunjin virus replicon vectors for human immunodeficiency Michael E, et al. Evaluating the immunogenicity of a disulfide-
virus vaccine development. J Virol 2003;77(14):7796–803. stabilized, cleaved, trimeric form of the envelope glycoprotein
[151] Zhang H, Huang Y, Fayad R, Spear GT, Qiao L. Induction complex of human immunodeficiency virus type 1. J Virol
of mucosal and systemic neutralizing antibodies against human 2005;79(14):8812–27.
immunodeficiency virus type 1 (HIV-1) by oral immunization with [166] Devico A, Silver A, Thronton AM, Sarngadharan MG, Pal R. Cova-
bovine Papillomavirus-HIV-1 gp41 chimeric virus-like particles. J lently crosslinked complexes of human immunodeficiency virus
Virol 2004;78(15):8342–8. type 1 (HIV-1) gp120 and CD4 receptor elicit a neutralizing
[152] Graham BS, Wright PF. Candidate AIDS vaccines. N Engl J Med immune response that includes antibodies selective for primary
1995;333(20):1331–9. virus isolates. Virology 1996;218(1):258–63.
[153] Jeffs SA, Goriup S, Kebble B, Crane D, Bolgiano B, Sattentau Q, [167] Fouts TR, Tuskan R, Godfrey K, Reitz M, Hone D, Lewis GK, et
et al. Expression and characterisation of recombinant oligomeric al. Expression and characterization of a single-chain polypeptide
envelope glycoproteins derived from primary isolates of HIV-1. analogue of the human immunodeficiency virus type 1 gp120-CD4
Vaccine 2004;22(8):1032–46. receptor complex. J Virol 2000;74(24):11427–36.
[154] Stott EJ, Almond N, Kent K, Walker B, Hull R, Rose J, et al. [168] Fouts T, Godfrey K, Bobb K, Montefiori D, Hanson CV, Kalya-
Evaluation of a candidate human immunodeficiency virus type 1 naraman VS, et al. Crosslinked HIV-1 envelope-CD4 receptor com-
(HIV-1) vaccine in macaques: effect of vaccination with HIV-1 plexes elicit broadly cross-reactive neutralizing antibodies in rhesus
gp120 on subsequent challenge with heterologous simian immun- macaques. Proc Natl Acad Sci U S A 2002;99(18):11842–7.
4078 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

[169] Pantophlet R, Wilson IA, Burton DR. Hyperglycosylated mutants [187] Silvera P, Richardson MW, Greenhouse J, Yalley-Ogunro J, Shaw
of human immunodeficiency virus (HIV) type 1 monomeric N, Mirchandani J, et al. Outcome of simian-human immunodefi-
gp120 as novel antigens for HIV vaccine design. J Virol ciency virus strain 89.6p challenge following vaccination of rhesus
2003;77(10):5889–901. macaques with human immunodeficiency virus Tat protein. J Virol
[170] McGaughey GB, Barbato G, Bianchi E, Freidinger RM, Garsky 2002;76(8):3800–9.
VM, Hurni WM, et al. Progress towards the development of a [188] Allen TM, Mortara L, Mothe BR, Liebl M, Jing P, Calore B, et al.
HIV-1 gp41-directed vaccine. Curr HIV Res 2004;2(2):193–204. Tat-vaccinated macaques do not control simian immunodeficiency
[171] Lenz O, Dittmar MT, Wagner A, Ferko B, Vorauer-Uhl K, Stiegler virus SIVmac239 replication. J Virol 2002;76(8):4108–12.
G, et al. Trimeric membrane-anchored gp41 inhibits HIV mem- [189] Liang X, Casimiro DR, Schleif WA, Wang F, Davies ME, Zhang
brane fusion. J Biol Chem 2005;280(6):4095–101. ZQ, et al. Vectored Gag and Env but not Tat show efficacy against
[172] Pantophlet R, Burton DR. GP120: target for neutralizing HIV-1 simian-human immunodeficiency virus 89.6P challenge in Mamu-
antibodies. Annu Rev Immunol 2006. A*01-negative rhesus monkeys. J Virol 2005;79(19):12321–31.
[173] Kuiken CL, Foley B, Freed E, Hahn BH, McCutchan FE, Mellors [190] Caputo A, Gavioli R, Ensoli B. Recent advances in the development
JW, et al. HIV Sequence Compendium 2002, Theoretical Biology of HIV-1 Tat-based vaccines. Curr HIV Res 2004;2(4):357–76.
and Biophysics Group. Los Alamos, NM: Los Alamos Laboratory; [191] Ensoli B. Rational vaccine strategies against AIDS: background
2002. and rationale. Microbes Infect 2005;7(14):1445–52.
[174] Hurwitz JL, Slobod KS, Lockey TD, Wang S, Chou TH, Lu S. [192] Mascarell L, Fayolle C, Bauche C, Ladant D, Leclerc C. Induction
Application of the polyvalent approach to HIV-1 vaccine develop- of neutralizing antibodies and Th1-polarized and CD4-independent
ment. Curr Drug Targets Infect Disord 2005;5(2):143–56. CD8+ T-cell responses following delivery of human immunodefi-
[175] Mascola JR, Sambor A, Beaudry K, Santra S, Welcher B, Louder ciency virus type 1 Tat protein by recombinant adenylate cyclase
MK, et al. Neutralizing antibodies elicited by immunization of of Bordetella pertussis. J Virol 2005;79(15):9872–84.
monkeys with DNA plasmids and recombinant adenoviral vectors [193] Voss G, Manson K, Montefiori D, Watkins DI, Heeney J, Wyand
expressing human immunodeficiency virus type 1 proteins. J Virol M, et al. Prevention of disease induced by a partially heterologous
2005;79(2):771–9. AIDS virus in rhesus monkeys by using an adjuvanted multicom-
[176] Seaman MS, Xu L, Beaudry K, Martin KL, Beddall MH, Miura A, ponent protein vaccine. J Virol 2003;77(2):1049–58.
et al. Multiclade human immunodeficiency virus type 1 envelope [194] Sauter SL, Rahman A, Muralidhar G. Non-replicating viral vector-
immunogens elicit broad cellular and humoral immunity in rhesus based AIDS vaccines: interplay between viral vectors and the
monkeys. J Virol 2005;79(5):2956–63. immune system. Curr HIV Res 2005;3(2):157–81.
[177] Gao F, Weaver EA, Lu Z, Li Y, Liao HX, Ma B, et al. Antigenic- [195] Letvin NL, Montefiori DC, Yasutomi Y, Perry HC, Davies ME,
ity and immunogenicity of a synthetic human immunodeficiency Lekutis C, et al. Potent, protective anti-HIV immune responses
virus type 1 group m consensus envelope glycoprotein. J Virol generated by bimodal HIV envelope DNA plus protein vaccination.
2005;79(2):1154–63. Proc Natl Acad Sci U S A 1997;94(17):9378–83.
[178] Doria-Rose NA, Learn GH, Rodrigo AG, Nickle DC, Li F, Maha- [196] Barouch DH, Craiu A, Kuroda MJ, Schmitz JE, Zheng XX,
lanabis M, et al. Human immunodeficiency virus type 1 subtype B Santra S, et al. Augmentation of immune responses to HIV-1 and
ancestral envelope protein is functional and elicits neutralizing anti- simian immunodeficiency virus DNA vaccines by IL-2/Ig plas-
bodies in rabbits similar to those elicited by a circulating subtype mid administration in rhesus monkeys. Proc Natl Acad Sci U S
B envelope. J Virol 2005;79(17):11214–24. A 2000;97(8):4192–7.
[179] Thomson SA, Jaramillo AB, Shoobridge M, Dunstan KJ, Everett [197] Boyer JD, Ugen KE, Wang B, Agadjanyan M, Gilbert L, Bagarazzi
B, Ranasinghe C, et al. Development of a synthetic consensus ML, et al. Protection of chimpanzees from high-dose heterolo-
sequence scrambled antigen HIV-1 vaccine designed for global use. gous HIV-1 challenge by DNA vaccination. Nat Med 1997;3(5):
Vaccine 2005;23(38):4647–57. 526–32.
[180] Gallo RC. Tat as one key to HIV-induced immune pathogenesis [198] Calarota SA, Weiner DB. Approaches for the design and evalua-
and Tat (correction of Pat) toxoid as an important component of a tion of HIV-1 DNA vaccines. Expert Rev Vaccines 2004;3(Suppl.
vaccine. Proc Natl Acad Sci U S A 1999;96(15):8324–6. 4):S135–49.
[181] Ferrantelli F, Cafaro A, Ensoli B. Nonstructural HIV proteins as [199] Giri M, Ugen KE, Weiner DB. DNA vaccines against human
targets for prophylactic or therapeutic vaccines. Curr Opin Biotech- immunodeficiency virus type 1 in the past decade. Clin Microbiol
nol 2004;15(6):543–56. Rev 2004;17(2):370–89.
[182] Rezza G, Fiorelli V, Dorrucci M, Ciccozzi M, Tripiciano A, [200] Nabel GJ. HIV vaccine strategies. Vaccine 2002;20(15):1945–7.
Scoglio A, et al. The presence of anti-Tat antibodies is predic- [201] Casimiro DR, Wang F, Schleif WA, Liang X, Zhang ZQ, Tobery
tive of long-term nonprogression to AIDS or severe immunodefi- TW, et al. Attenuation of simian immunodeficiency virus SIV-
ciency: findings in a cohort of HIV-1 seroconverters. J Infect Dis mac239 infection by prophylactic immunization with dna and
2005;191(8):1321–4. recombinant adenoviral vaccine vectors expressing Gag. J Virol
[183] Cafaro A, Caputo A, Fracasso C, Maggiorella MT, Goletti D, 2005;79(24):15547–55.
Baroncelli S, et al. Control of SHIV-89.6P-infection of cynomol- [202] McDermott AB, O’Connor DH, Fuenger S, Piaskowski S, Martin
gus monkeys by HIV-1 Tat protein vaccine. Nat Med 1999;5(6): S, Loffredo J, et al. Cytotoxic T-lymphocyte escape does not always
643–50. explain the transient control of simian immunodeficiency virus SIV-
[184] Osterhaus AD, van Baalen CA, Gruters RA, Schutten M, Siebelink mac239 viremia in adenovirus-boosted and DNA-primed Mamu-
CH, Hulskotte EG, et al. Vaccination with Rev and Tat against A*01-positive rhesus macaques. J Virol 2005;79(24):15556–66.
AIDS. Vaccine 1999;17(20–21):2713–4. [203] Pialoux G, Excler JL, Riviere Y, Gonzalez-Canali G, Feuillie V,
[185] Maggiorella MT, Baroncelli S, Michelini Z, Fanales-Belasio E, Coulaud P, et al. A prime-boost approach to HIV preventive vaccine
Moretti S, Sernicola L, et al. Long-term protection against using a recombinant canarypox virus expressing glycoprotein 160
SHIV89.6P replication in HIV-1 Tat vaccinated cynomolgus mon- (MN) followed by a recombinant glycoprotein 160 (MN/LAI). The
keys. Vaccine 2004;22(25–26):3258–69. AGIS Group, and l’Agence Nationale de Recherche sur le SIDA.
[186] Pauza CD, Trivedi P, Wallace M, Ruckwardt TJ, Le Buanec H, AIDS Res Hum Retroviruses 1995;11(3):373–81.
Lu W, et al. Vaccination with tat toxoid attenuates disease in [204] Belshe RB, Stevens C, Gorse GJ, Buchbinder S, Weinhold K, Shep-
simian/HIV-challenged macaques. Proc Natl Acad Sci U S A pard H, et al. Safety and immunogenicity of a canarypox-vectored
2000;97(7):3515–9. human immunodeficiency virus Type 1 vaccine with or without
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4079

gp120: a phase 2 study in higher- and lower-risk volunteers. J macaques against intrarectal simian immunodeficiency virus
Infect Dis 2001;183(9):1343–52. SIV(mac251) challenge by a replication-competent Ad5hr-SIVenv/
[205] Paris R, Bejrachandra S, Karnasuta C, Chandanayingyong D, rev and Ad5hr-SIVgag recombinant priming/gp120 boosting regi-
Kunachiwa W, Leetrakool N, et al. HLA class I serotypes and men. J Virol 2003;77(15):8354–65.
cytotoxic T-lymphocyte responses among human immunodeficiency [221] Lorin C, Mollet L, Delebecque F, Combredet C, Hurtrel B,
virus-1-uninfected Thai volunteers immunized with ALVAC-HIV in Charneau P, et al. A single injection of recombinant measles virus
combination with monomeric gp120 or oligomeric gp160 protein vaccines expressing human immunodeficiency virus (HIV) type 1
boosting. Tissue Antigens 2004;64(3):251–6. clade B envelope glycoproteins induces neutralizing antibodies and
[206] Amara RR, Villinger F, Altman JD, Lydy SL, O’Neil SP, Staprans cellular immune responses to HIV. J Virol 2004;78(1):146–57.
SI, et al. Control of a mucosal challenge and prevention of AIDS by [222] Rose NF, Marx PA, Luckay A, Nixon DF, Moretto WJ, Donahoe
a multiprotein DNA/MVA vaccine. Science 2001;292(5514):69–74. SM, et al. An effective AIDS vaccine based on live attenuated
[207] Im EJ, Hanke T. MVA as a vector for vaccines against HIV-1. vesicular stomatitis virus recombinants. Cell 2001;106(5):539–49.
Expert Rev Vaccines 2004;3(Suppl. 4):S89–97. [223] Roberts A, Kretzschmar E, Perkins AS, Forman J, Price R,
[208] Kent SJ, Zhao A, Best SJ, Chandler JD, Boyle DB, Ramshaw Buonocore L, et al. Vaccination with a recombinant vesicular
IA. Enhanced T-cell immunogenicity and protective efficacy of a stomatitis virus expressing an influenza virus hemagglutinin pro-
human immunodeficiency virus type 1 vaccine regimen consisting vides complete protection from influenza virus challenge. J Virol
of consecutive priming with DNA and boosting with recombinant 1998;72(6):4704–11.
fowlpox virus. J Virol 1998;72(12):10180–8. [224] Publicover J, Ramsburg E, Rose JK. A single-cycle vaccine vector
[209] Shiver JW, Fu TM, Chen L, Casimiro DR, Davies ME, Evans based on vesicular stomatitis virus can induce immune responses
RK, et al. Replication-incompetent adenoviral vaccine vector comparable to those generated by a replication-competent vector.
elicits effective anti-immunodeficiency-virus immunity. Nature J Virol 2005;79(21):13231–8.
2002;415(6869):331–5. [225] Matano T, Kano M, Nakamura H, Takeda A, Nagai Y. Rapid
[210] Casimiro DR, Chen L, Fu TM, Evans RK, Caulfield MJ, Davies appearance of secondary immune responses and protection from
ME, et al. Comparative immunogenicity in rhesus monkeys of DNA acute CD4 depletion after a highly pathogenic immunodeficiency
plasmid, recombinant vaccinia virus, and replication-defective ade- virus challenge in macaques vaccinated with a DNA prime/Sendai
novirus vectors expressing a human immunodeficiency virus type virus vector boost regimen. J Virol 2001;75(23):11891–6.
1 gag gene. J Virol 2003;77(11):6305–13. [226] Takeda A, Igarashi H, Nakamura H, Kano M, Iida A, Hirata T,
[211] Shiver JW, Emini EA. Recent advances in the development of HIV- et al. Protective efficacy of an AIDS vaccine, a single DNA prim-
1 vaccines using replication-incompetent adenovirus vectors. Annu ing followed by a single booster with a recombinant replication-
Rev Med 2004;55:355–72. defective Sendai virus vector, in a macaque AIDS model. J Virol
[212] Santra S, Seaman MS, Xu L, Barouch DH, Lord CI, Lifton MA, 2003;77(17):9710–5.
et al. Replication-defective adenovirus serotype 5 vectors elicit [227] Perri S, Greer CE, Thudium K, Doe B, Legg H, Liu H, et al.
durable cellular and humoral immune responses in nonhuman pri- An alphavirus replicon particle chimera derived from venezuelan
mates. J Virol 2005;79(10):6516–22. equine encephalitis and sindbis viruses is a potent gene-based vac-
[213] Kostense S, Koudstaal W, Sprangers M, Weverling GJ, Penders cine delivery vector. J Virol 2003;77(19):10394–403.
G, Helmus N, et al. Adenovirus types 5 and 35 seroprevalence [228] Davis NL, Caley IJ, Brown KW, Betts MR, Irlbeck DM, McGrath
in AIDS risk groups supports type 35 as a vaccine vector. AIDS KM, et al. Vaccination of macaques against pathogenic simian
2004;18(8):1213–6. immunodeficiency virus with Venezuelan equine encephalitis virus
[214] Vogels R, Zuijdgeest D, van Rijnsoever R, Hartkoorn E, Damen I, replicon particles. J Virol 2000;74(1):371–8.
de Bethune MP, et al. Replication-deficient human adenovirus type [229] Liu XL, Clark KR, Johnson PR. Production of recombinant adeno-
35 vectors for gene transfer and vaccination: efficient human cell associated virus vectors using a packaging cell line and a hybrid
infection and bypass of preexisting adenovirus immunity. J Virol recombinant adenovirus. Gene Ther 1999;6(2):293–9.
2003;77(15):8263–71. [230] Johnson PR, Schnepp BC, Connell MJ, Rohne D, Robinson S,
[215] Barouch DH, Pau MG, Custers JH, Koudstaal W, Kostense S, Krivulka GR, et al. Novel adeno-associated virus vector vaccine
Havenga MJ, et al. Immunogenicity of recombinant adenovirus restricts replication of simian immunodeficiency virus in macaques.
serotype 35 vaccine in the presence of pre-existing anti-Ad5 immu- J Virol 2005;79(2):955–65.
nity. J Immunol 2004;172(10):6290–7. [231] Someya K, Cecilia D, Ami Y, Nakasone T, Matsuo K, Burda S,
[216] Nanda A, Lynch DM, Goudsmit J, Lemckert AA, Ewald BA, et al. Vaccination of rhesus macaques with recombinant Mycobac-
Sumida SM, et al. Immunogenicity of recombinant fiber-chimeric terium bovis bacillus Calmette-Guerin Env V3 elicits neutralizing
adenovirus serotype 35 vector-based vaccines in mice and rhesus antibody-mediated protection against simian-human immunodefi-
monkeys. J Virol 2005;79(22):14161–8. ciency virus with a homologous but not a heterologous V3 motif.
[217] Patterson LJ, Malkevitch N, Pinczewski J, Venzon D, Lou Y, Peng J Virol 2005;79(3):1452–62.
B, et al. Potent, persistent induction and modulation of cellular [232] Ami Y, Izumi Y, Matsuo K, Someya K, Kanekiyo M, Horibata S, et
immune responses in rhesus macaques primed with Ad5hr-simian al. Priming-boosting vaccination with recombinant Mycobacterium
immunodeficiency virus (SIV) env/rev, gag, and/or nef vaccines bovis bacillus Calmette-Guerin and a nonreplicating vaccinia virus
and boosted with SIV gp120. J Virol 2003;77(16):8607–20. recombinant leads to long-lasting and effective immunity. J Virol
[218] Robert-Guroff M, Kaur H, Patterson LJ, Leno M, Conley AJ, 2005;79(20):12871–9.
McKenna PM, et al. Vaccine protection against a heterologous, [233] Devico AL, Fouts TR, Shata MT, Kamin-Lewis R, Lewis GK,
non-syncytium-inducing, primary human immunodeficiency virus. Hone DM. Development of an oral prime-boost strategy to
J Virol 1998;72(12):10275–80. elicit broadly neutralizing antibodies against HIV-1. Vaccine
[219] Peng B, Wang LR, Gomez-Roman VR, Davis-Warren A, Mon- 2002;20(15):1968–74.
tefiori DC, Kalyanaraman VS, et al. Replicating rather than non- [234] Crotty S, Miller CJ, Lohman BL, Neagu MR, Compton L,
replicating adenovirus-human immunodeficiency virus recombinant Lu D, et al. Protection against simian immunodeficiency virus
vaccines are better at eliciting potent cellular immunity and priming vaginal challenge by using Sabin poliovirus vectors. J Virol
high-titer antibodies. J Virol 2005;79(16):10200–9. 2001;75(16):7435–52.
[220] Zhao J, Pinczewski J, Gomez-Roman VR, Venzon D, Kalyanara- [235] Wu L, Kong WP, Nabel GJ. Enhanced breadth of CD4 T-cell
man VS, Markham PD, et al. Improved protection of rhesus immunity by DNA prime and adenovirus boost immunization to
4080 M.P. Girard et al. / Vaccine 24 (2006) 4062–4081

human immunodeficiency virus Env and Gag immunogens. J Virol progressive human immunodeficiency virus type 1 infection. J Virol
2005;79(13):8024–31. 2002;76(18):9011–23.
[236] Casimiro DR, Bett AJ, Fu TM, Davies ME, Tang A, Wilson [253] Younes SA, Yassine-Diab B, Dumont AR, Boulassel MR,
KA, et al. Heterologous human immunodeficiency virus type Grossman Z, Routy JP, et al. HIV-1 viremia prevents the
1 priming-boosting immunization strategies involving replication- establishment of interleukin 2-producing HIV-specific memory
defective adenovirus and poxvirus vaccine vectors. J Virol CD4+ T cells endowed with proliferative capacity. J Exp Med
2004;78(20):11434–8. 2003;198(12):1909–22.
[237] Lemckert AA, Sumida SM, Holterman L, Vogels R, Truitt DM, [254] Imami N, Pires A, Burton C. The challenge of developing
Lynch DM, et al. Immunogenicity of heterologous prime-boost reg- an effective HIV-I vaccine. Drug Dis Today:Ther Strategies
imens involving recombinant adenovirus serotype 11 (Ad11) and 2004;1(4):461–7.
Ad35 vaccine vectors in the presence of anti-ad5 immunity. J Virol [255] Haase AT. Perils at mucosal front lines for HIV and SIV and their
2005;79(15):9694–701. hosts. Nat Rev Immunol 2005;5(10):783–92.
[238] Yewdell JW, Bennink JR. Immunodominance in major histocom- [256] Holmgren J, Czerkinsky C. Mucosal immunity and vaccines. Nat
patibility complex class I-restricted T lymphocyte responses. Annu Med 2005;11(Suppl. 4):S45–53.
Rev Immunol 1999;17:51–88. [257] Belyakov IM, Hel Z, Kelsall B, Kuznetsov VA, Ahlers JD, Nacsa
[239] Ishioka GY, Fikes J, Hermanson G, Livingston B, Crimi C, Qin M, J, et al. Mucosal AIDS vaccine reduces disease and viral load in
et al. Utilization of MHC class I transgenic mice for development gut reservoir and blood after mucosal infection of macaques. Nat
of minigene DNA vaccines encoding multiple HLA-restricted CTL Med 2001;7(12):1320–6.
epitopes. J Immunol 1999;162(7):3915–25. [258] Belyakov IM, Ahlers JD, Berzofsky JA. Mucosal AIDS vac-
[240] Hanke T, Samuel RV, Blanchard TJ, Neumann VC, Allen TM, cines: current status and future directions. Expert Rev Vaccines
Boyson JE, et al. Effective induction of simian immunodeficiency 2004;3(Suppl. 4):S65–73.
virus-specific cytotoxic T lymphocytes in macaques by using a [259] Kuznetsov VA, Stepanov VS, Berzofsky JA, Belyakov IM. Assess-
multiepitope gene and DNA prime-modified vaccinia virus Ankara ment of the relative therapeutic effects of vaccines on virus load
boost vaccination regimen. J Virol 1999;73(9):7524–32. and immune responses in small groups at several time points:
[241] Allen TM, Vogel TU, Fuller DH, Mothe BR, Steffen S, Boyson efficacy of mucosal and subcutaneous polypeptide vaccines in rhe-
JE, et al. Induction of AIDS virus-specific CTL activity in fresh, sus macaques exposed to SHIV. J Clin Virol 2004;31(Suppl. 1):
unstimulated peripheral blood lymphocytes from rhesus macaques S69–82.
vaccinated with a DNA prime/modified vaccinia virus Ankara boost [260] Gupta S, Janani R, Bin Q, Luciw P, Greer C, Perri S, et
regimen. J Immunol 2000;164(9):4968–78. al. Characterization of human immunodeficiency virus Gag-
[242] Subbramanian RA, Kuroda MJ, Charini WA, Barouch DH, specific gamma interferon-expressing cells following protective
Costantino C, Santra S, et al. Magnitude and diversity of cytotoxic- mucosal immunization with alphavirus replicon particles. J Virol
T-lymphocyte responses elicited by multiepitope DNA vaccination 2005;79(11):7135–45.
in rhesus monkeys. J Virol 2003;77(18):10113–8. [261] Stahl-Hennig C, Steinman RM, Ten Haaft P, Uberla K, Stolte N,
[243] McEvers K, Elrefaei M, Norris P, Deeks S, Martin J, Lu Y, et Saeland S, et al. The simian immunodeficiency virus deltaNef vac-
al. Modified anthrax fusion proteins deliver HIV antigens through cine, after application to the tonsils of Rhesus macaques, replicates
MHC Class I and II pathways. Vaccine 2005;23(32):4128–35. primarily within CD4(+) T cells and elicits a local perforin-positive
[244] Mortara L, Gras-Masse H, Rommens C, Venet A, Guillet JG, CD8(+) T-cell response. J Virol 2002;76(2):688–96.
Bourgault-Villada I. Type 1 CD4(+) T-cell help is required [262] Tenner-Racz K, Stahl Hennig C, Uberla K, Stoiber H, Ignatius
for induction of antipeptide multispecific cytotoxic T lympho- R, Heeney J, et al. Early protection against pathogenic virus
cytes by a lipopeptidic vaccine in rhesus macaques. J Virol infection at a mucosal challenge site after vaccination with atten-
1999;73(5):4447–51. uated simian immunodeficiency virus. Proc Natl Acad Sci U S A
[245] Gahery-Segard H, Pialoux G, Charmeteau B, Sermet S, Poncelet 2004;101(9):3017–22.
H, Raux M, et al. Multiepitopic B- and T-cell responses induced [263] Jiang JQ, Patrick A, Moss RB, Rosenthal KL. CD8+ T-cell-
in humans by a human immunodeficiency virus type 1 lipopeptide mediated cross-clade protection in the genital tract following
vaccine. J Virol 2000;74(4):1694–703. intranasal immunization with inactivated human immunodefi-
[246] Pialoux G, Gahery-Segard H, Sermet S, Poncelet H, Fournier ciency virus antigen plus CpG oligodeoxynucleotides. J Virol
S, Gerard L, et al. Lipopeptides induce cell-mediated anti- 2005;79(1):393–400.
HIV immune responses in seronegative volunteers. AIDS [264] Berzofsky JA, Ahlers JD, Janik J, Morris J, Oh S, Terabe M, et al.
2001;15(10):1239–49. Progress on new vaccine strategies against chronic viral infections.
[247] Tonini T, Barnett S, Donnelly J, Rappuoli R. Current approaches to J Clin Invest 2004;114(4):450–62.
developing a preventative HIV vaccine. Curr Opin Investig Drugs [265] Mitragotri S. Immunization without needles. Nat Rev Immunol
2005;6(2):155–62. 2005;5(12):905–16.
[248] Srivastava IK, Ulmer JB, Barnett SW. Neutralizing antibody [266] Matoba N, Magerus A, Geyer BC, Zhang Y, Muralidharan M,
responses to HIV: role in protective immunity and challenges for Alfsen A, et al. A mucosally targeted subunit vaccine candidate
vaccine design. Expert Rev Vaccines 2004;3(Suppl. 4):S33–52. eliciting HIV-1 transcytosis-blocking Abs. Proc Natl Acad Sci U
[249] Anderson R, Hanson M. Potential public health impact of imperfect S A 2004;101(37):13584–9.
HIV type 1 vaccines. J Infect Dis 2005;191(Suppl. 1):S85–96. [267] Klausner RD, Fauci AS, Corey L, Nabel GJ, Gayle H, Berkley
[250] Bonifaz LC, Bonnyay DP, Charalambous A, Darguste DI, Fujii S, S, et al. Medicine. The need for a global HIV vaccine enterprise.
Soares H, et al. In vivo targeting of antigens to maturing dendritic Science 2003;300(5628):2036–9.
cells via the DEC-205 receptor improves T cell vaccination. J Exp [268] Cohen J. Immunology. New virtual center aims to speed AIDS
Med 2004;199(6):815–24. vaccine progress. Science 2005;309(5734):541.
[251] Rosenberg ES, Billingsley JM, Caliendo AM, Boswell SL, [269] Kaufmann SH, McMichael AJ. Annulling a dangerous liaison:
Sax PE, Kalams SA, et al. Vigorous HIV-1-specific CD4+ vaccination strategies against AIDS and tuberculosis. Nat Med
T cell responses associated with control of viremia. Science 2005;11(Suppl. 4):S33–44.
1997;278(5342):1447–50. [270] Flores JE. The HIV-1 pipeline and global expansion of clini-
[252] Imami N, Pires A, Hardy G, Wilson J, Gazzard B, Gotch F. A cal trials. Keystone meeting, Banff, Canada, 2005 [Abstract no
balanced type 1/type 2 response is associated with long-term non- 043].
M.P. Girard et al. / Vaccine 24 (2006) 4062–4081 4081

[271] Ellenberger D, Otten R, Li B, Rodriguez IV, Sariol C, Martinez [273] Arnold G, Velasco P, Wrin T, Hughes S, Elsasser D, Ma X, et al.
M, et al. Evidence of protection after repeated mucosal SHIV chal- Recombinant human rhinovirus displaying the HIV-1 gp41 ELD-
lenges to HIV-1 vaccinated rhesus monkeys. AIDS Vaccine 2005 KWA epitope can elicit broad neutralization of HIV-1 primary iso-
meeting, Montreal, Quebec, Canada, September 6–9, 2005 [abstract lates. AIDS Vaccine 2005, Montreal, Canada, 2005 [abstract 500].
110]. [274] Kawada M, Igarashi H, Takeda A, Tsukamoto T, Yamamoto H,
[272] Clarke D, Johnson E, Nasar F, Coleman J, Coorer D, Calderon Dohki S, et al. Involvement of multiple epitope-specific cyto-
P, et al. Assessment of rVSV/HIV vaccine vector neurovirulence toxic T-lymphocyte responses in vaccine-based control of simian
in murine, ferret and non-human primate models. AIDS Vaccine immunodeficiency virus replication in rhesus macaques. J Virol
2005, Montreal, Canada, 2005 [abstract no 13]. 2006;80(4):1949–58.

Você também pode gostar