<<

Drugs 50 (5) 792-804, 1995 REVIEW ARTICLE 0012-6667/95/0011-0792/$1300/0

© Ads Internatronall.Jmlted AU nghts reserved

HIV Preventive Progress to Date

Jose Esparza, Saladin Osmanov and William 1. Heyward Global Programme on AIDS, World Health Organization, Geneva, Switzerland

Contents Summary , , , , , , , , , , , , , , , . . . , , . , , . , , , . , , 792 1, Scientific Challenges in the Development of HIV Vaccines , 793 1.1 What Are the Immunological Correlates of Protection? 793 1.2 What Is the Significance of HIV Genetic Variability in Relation to Development? , . . . . . , , 794 2, Preclinical Vaccine Development ...... , ...... 795 3. Phase 1/11 Trials of HIV Preventive Vaccines .... , ...... 797 4, The Future: Prospects for the Conduct of Phase III Efficacy Trials 798

Summary The major conceptual problem for HIV vaccine development has been the lack of information on immune responses known to correlate with protection against HIV in humans. In this regard, studies on the natural history of HIV infection and AIDS, especially of people with apparent resistance to HIV infec­ tion and of patients with HIV infection who have long term survival without disease progression, may provide important information for vaccine develop­ ment. In addition, a major concern for the development of broadly effective vac­ cines has been the extensive genetic variability which is characteristic of HIY. In spite of these unknowns, the first generation of HIV candidate vaccines has been developed and evaluated. HIV candidate vaccines based on the subunit recombinant envelope concept (gp120 or gp160) have been shown to protect chimpanzees from HIV infection on challenge, and have now been evaluated in humans in phase I and phase II trials. These products are well tolerated, and capable of inducing neutralising antibodies, but not cytotoxic T lymphocytes. A second vaccine concept, currently in phase I trials, is based on live recombinant vectors, especially using poxvirus vectors followed by boosting with subunit recombinant envelope vaccines. This concept is theoretically very attractive be­ cause preliminary data suggest that these vaccines induce both humoral and cell­ mediated . However, no published information is available on the ability of live recombinant vector vaccines to protect chimpanzees from HIV infection. The next step in HIV vaccine development is to proceed carefully to expanded phase II and phase III trials to assess the protective efficacy of these candidate vaccines in humans. These trials will be extremely complex from the logistical, scientific and ethical points of view, and will require close collaboration between clinical, basic science and behavioural researchers, national and international organisations, and the pharmaceutical industry. HIV Preventive Vaccines 793

More than 16 million HIV are nevertheless do not control the infection or devel­ thought to have occurred worldwide.(1,2) If the opment of AIDS. present trend continues, by the year 2000 there will Therefore, at this stage of vaccine development, be a cumulative total of 30 to 40 million HIV in­ it might be wise to assume that both arms of the fections worldwide, with 90% of these infections may playa role in protective im­ occurring in developing countries. To bring this munity against HIV infection and disease, and that epidemic under control, a safe and effective pre­ preventive vaccines may need to induce both types ventive vaccine may be needed.(3) of immune responses. In this regard, studies on the Animal protection experiments with HIV and natural history of HIV infection and AIDS may simian virus (SIV) candidate provide important information to guide further de­ vaccines have provided information which has velopment of HIV vaccines.(16-18) brought cautious optimism about the possibility of HIV infection is usually chronic and, in most developing an HIV vaccine. However, these pro­ cases, eventually leads to the development of tection experiments were conducted under ideal AIDS. It has been recently observed, however, that conditions, and their relevance to protection in hu­ a very small proportion of individuals apparently mans is still unknown. Moreover, information on remain uninfected despite frequent and/or exten­ immune correlates of protection against HIV, and sive exposure to the viruS.(19) It has been hypoth­ on the significance ofthe genetic variability ofHIV esised that such 'resistance' could be due to genetic in relation to vaccine efficacy, is still lacking. factors [human leucocyte (HLA) haplo­ Since 1987 when the first HIV candidate vac­ type], alloimmunisation to cellular or nat­ cines entered phase I clinical trials, it has become urally acquired immunity as a result of frequent clear that the development of an effective preven­ exposure to subinfectious doses of the virus.[20-26) tive vaccine will be a long and difficult process.(4- Identification of specific immune responses re­ 12) This brief review discusses some of the scien­ sponsible for such 'resistance' would provide im­ tific challenges that have been identified, the portant clues as to the kind of immune responses different approaches used in the development of that an effective vaccine should induce. candidate HIV vaccines, the results from ongoing In the large majority of individuals who become phase IIII clinical trials and the prospects for the infected after exposure, the initial pathogenic event conduct of large-scale phase III efficacy trials. is characterised by high-level viraemia, with infec­ tion of peripheral CD4+ T lymphocytes, and early 1. Scientific Challenges in the dissemination of the virus to the lymphoid sys­ Development of HIV Vaccines tem.[16,27) This primary viraemia is rapidly control­ led, at the same time when HIV-specific CD8+ cytotoxic T lymphocytes (CTLs) are generated.[28-30) 1.1 What Are the Immunological Correlates of Protection? HIV-binding antibodies are also present early dur­ ing primary infection, although neutralising anti­ The major conceptual problem for HIV vaccine bodies are detectable only after the viraemia has development is the lack of information on immune been controlled.(28,3I) responses known to correlate with protection These observations point to the important role against HIV.(ll) There has been controversy about of cell-mediated immunity in the initial anti-HIV whether a vaccine must induce humoral and/or immune response. In addition, it has been sug­ cell-mediated immunity for effective protec­ gested that low levels of viraemia may be a prog­ tion.(13-15) This difficulty arises because individu­ nostic marker for increased survival,[32-34) a situa­ als naturally infected with HIV develop a ' broad tion which has also been observed in the range of viral-specific immune responses, which SIV/macaque modeU35 ,36) If confirmed, this find-

© Adis International Limited. All fights reserved. Drugs 50 (5) 1995 794 Esparza et al.

ing would have important implications for the se­ teins, gp120 and gp41, are both derived from the lection of end-points in the design of efficacy trials cleavage of gp160, an intracellular precursor mol­ of HIV candidate vaccines. It is conceivable that a ecule. Neutralising antibodies are mostly directed vaccine which is not fully protective against HIV to gp120, although neutralisation epitopes have infection (incapable of providing sterilising immu­ also been identified in gp41 and p 17 (the matrix nity) may still decrease the initial virus load, and ). thus delay or prevent the future course of HIV dis­ The principal neutralisation domain in gp120 is ease,!37-391 located within one of the 5 hypervariable regions Following the primary viraemia, the patient en­ of the molecule, known as the V3 loop, which in­ ters the asymptomatic or clinically latent period of duces the production of strain-specific neutralising infection. During this stage, the level of circulating antibodies. In addition, a number of conforma­ virus or virus-infected cells in peripheral blood tional epitopes are also present in gp 120, and the mononuclear cells is very low, but virus burden is neutralising antibodies which they induce are more 40 high in peripheral lymphoid tissues.l ,411 broadly reactive, some of them being capable of It is now known that HIV infection is a very preventing the binding of gp 120 to the CD4 recep­ dynamic process, with continuous synthesis and tor molecule. [4,5 1-561 destruction of circulating virus, accompanied by a Vaccine-induced protection may be affected by rapid turnover of CD4+ lymphocytes. There is a net the antigenic variability resulting from the genetic decrease in their number over time, and subsequent differences among envelope genes, especially if severe immunodeficiency. [42,431 this protection is dependent upon strain-specific The average duration of this asymptomatic pe­ neutralising antibodies. Phylogenetic analysis of riod is 8 to 10 years,[441although between 5 and 8% the nucleotide sequence of the envelope gene of of HIV-infected people may remain clinically numerous virus strains from different parts of the asymptomatic and immunologically competent for world has resulted in the identification of a number 45 up to 15 years after infection.[ 1 These 'long term of clades (or genetic subtypes) ofHIV-l,l57,581The nonprogressors' are being intensively studied in an majority of strains have been included within a ma­ attempt to identify possible immunological and vi­ jor group (group-M), which is subdivided into at rological correlates of survival, which may also be least 8 clades, designated A to H.[58-701 relevant for vaccine design. In 'addition, a number of highly divergent HIV-l More than one mechanism seems to be corre­ strains, which are significantly distant from group­ lated with long term survival, including low viral M viruses, have been identified and designated as load in blood and lymph nodes, potent and persist­ 'outliers', or group-O virusespl-751 Group-M vi­ ent cellular responses (CD8+ HIV-l-specific ruses are unequally distributed in the world, clade CTLs), persistent levels of broadly reactive neu­ tralising antibodies and/or infection with attenu­ B viruses being more prevalent in Europe, North ated strains of the virus,[32,33,45-481 which in one America and Australia; clades B, C and E in South case, but not in others, has been explained by a East Asia; and almost all clades being present in deletion in the nef gene.[49,501 different regions of sub-Saharan Africa. What is not fully understood at the present time 1.2 What Is the Significance of is the significance of genetic variability in relation HIV Genetic Variability in Relation to to the antigenic characteristics of viruses belonging Vaccine Development? to the different HIV clades.[76-781 It is interesting that most of the gp 120 conformational epitopes re­ HIV is characterised by extensive genetic vari­ lated to the CD4-binding site of clade B viruses ation, especially in the gene coding for the enve­ were found to be conserved among the gp120 mol­ lope glycoproteins (env). The 2 envelope glycopro- ecules from strains belonging to clades A to F, al-

© Adls International Limited All rights reserved. Drugs 50 (5) 1995 HIV Preventive Vaccines 795

though clade E strains were the least related to 2. Preclinical Vaccine Development clade B virusesP9] In this regard, it has been sug­ gested that clade Band E viruses from Thailand Two animal models have been extensively used may belong to 2 different neutralisation immuno­ in the development of HIV vaccines: SIV infection types.[80] On the other hand, preliminary informa­ in macaque monkeys, and HIV infection in chim­ tion obtained by investigators from the WHO Net­ panzees)86.87] Chimpanzees have been success­ work for HIV Isolation and Characterisation has fully protected with HIV experimental vaccines, suggested extensive in vitro cross-neutralisation whereas experimental SIV vaccines have been no­ between strains belonging to different genetic toriously poor in protecting macaque monkeys against SIV infection. This dichotomy in the ability clades (unpublished data). of different candidate vaccines to induce protective At this time, however, it is not possible to rule immunity in these 2 primate models should be kept out the possible existence of neutralisation immuno­ in mind when interpreting the relevance of these types, which may correspond to some of the clades experiments for humans. defined by genetic sequencing of the envelope The first protection experiments in the SIV / gene. However, a more basic question is the possi­ macaque model used whole inactivated virus vac­ ble role, if any, of neutralising antibodies in vac­ cines, although other studies later indicated that the cine-induced protection, and the ability of labora­ observed protection was due to immune responses tory tests to detect the relevant antibodies. against cellular incorporated into both the The genetic diversity of HIV-I strains has also immunising and challenge virus, rather than to a been correlated with growth characteristics in vitro specific immune response against the virus.[88.89] of the virus. VIruses isolated from recently infected In addition, many did not consider the whole inac­ persons are usually unable to replicate in estab­ tivated virus vaccine as a viable approach for the lished T cell lines, or to form syncytia in peripheral development of preventive HIV vaccines, because blood mononuclear cells. These non-syncytia­ of the possible associated risks. Thus, the initially inducing (NSI) viruses are different from isolates developed candidate vaccines for human use were obtained from infected persons with more ad­ based on the subunit recombinant envelope con­ vanced stages of the disease, which often have a cept, an approach considered to be safer than whole syncytia-inducing (SI) phenotype)81.82] inactivated virus. These findings suggest that HIV is usually Subunit recombinant envelope candidate vac­ transmitted as an NSIImacrophage tropic virus, cines use gp120 or gp160 as the immunogen, and and later evolves to an SI virus form as infection are produced by genetic engineering techniques in and disease progress.[83] Of potential importance yeast, insect cells or mammalian cells. Clade B vi­ for vaccine development is the observation that ruses have been used in most cases.[4.5] Even NSI and SI viruses differ in their susceptibility to though the immunological correlates of HIV vac­ neutralising antibodies, probably due,to a different cine protection are still unknown, these vaccines configuration of the outer envelope glycoprotein. were developed with the assumption that neutralis­ More specifically, the principal neutralisation do­ ing antibodies would be an importan,t component main of gp120 (the V3100p) seems to be relatively of the protective immune response against HIY. inaccessible to V3-specific neutralising antibodies Envelope subunit recombinant vaccines pro­ among NSI 'clinical' isolates, whereas it is more duced in mammalian cells have been shown to accessible among 'laboratory' strains which have protect chimpanzees against HIV infection. These been adapted to continuous culture in established experiments, however, were done under well con­ T celllines.[84.85] Therefore, vaccines may need to trolled conditions, which included intravenous induce immune responses capable of neutralising challenge with cell-free homologous virus at the NSI viruses. peak level of postvaccination antibodies.[4] Addi-

© Adis International limited, All rights reserved. Drugs 50 (5) 1995 796 Esparza et al.

tional experiments have shown that immunised non-envelope subunit candidate vaccines are still chimpanzees were also protected against a cell­ lacking. associated challenge.l90] Experiments are under Since subunit nonreplicating immunogens are way to determine if systemic immunisation with generally not efficient inducers of CD8+ CTLs, subunit recombinant envelope vaccines can protect much work has been invested in the development chimpanzees from mucosal (genital) exposure to of different approaches based on live-vectored vac­ HIV.l91] cines, especially using poxvirus vectors, which An important recent finding is that the neutralis­ would induce CTLs.l102-106] However, very few ing antibodies induced by subunit recombinant en­ data are available on the ability of these candidate velope vaccines in chimpanzees (as well as in hu­ vaccines to protect animals from HIV infection. In mans) are mostly directed against linear epitopes this regard, it has been disappointing that vaccina­ in the V3100p, and not conformational epitopes.[92] tion of rhesus monkeys with a recombinant vac­ As discussed above, these antibodies against V3 cinia virus expressing envelope and core antigens loop epitopes fail to neutralise 'clinical' isolates of was not more effective than subunit envelope pro­ HIV in vitro. Intriguingly, chimpanzees have also teins in conferring protection against SIV infec­ tion.l94] been protected from a challenge using a clinical isolate of HIV, even when the sera from the im­ In contrast to the relatively low protective effi­ cacy of SIV subunit recombinant envelope vac­ munised animals failed to neutralise the virus in cines, or of live vector SIV vaccines, nef-deleted vitro[91] (D. Francis, personal communication). This attenuated SIV has been reported to induce solid observation must bring some caution in the inter­ protection against virus challenge. [107-111] Al­ pretation of in vitro neutralisation tests as 'predic­ though the mechanisms of protection in macaque tors' of potential vaccine efficacy against clinical monkeys are not well understood, these observa­ isolates. tions open new possibilities for the development of Contrary to the HIV Ichimpanzee experiments, a new generation of live-attenuated vaccinesp12,1l3] vaccines based on SIV subunit envelope recombi­ However, since the administration of a live nant proteins have usually failed to protect mon­ retrovirus to an uninfected person might carry an keys against postvaccination virus challenge, al­ appreciable degree of risk, further research is though they have succeeded in reducing the initial needed to understand the potential risks and bene­ level of virus burden after infection. [36,93-95] On the fits, before decisions can be made regarding the other hand, macaques have been protected against initiation of human trials with live-attenuated HIV cell-free HIV-2 with an HIV-2 Iscom vaccine.[96] vaccines. [1 14-117] The significance of these experiments remains un­ Finally, one of the most exciting new develop­ clear, since neither the HIV/chimpanzee nor the ments in the area of vaccinology is the use of the SIVIHIV-2/macaque models have been validated direct injection of antigen-encoding free (naked) in terms of their relevance to protection in humans. DNA (nucleic acid vaccines) into the skeletal mus­ Since the V3 loop is the major neutralisation cle or dermal tissue, which results in protein ex­ domain in HIV-l, candidate vaccines based on pression and subsequent induction of antibodies multimeric V3 loop amino acid sequences have and CTLs against the encoded protein.[118-126] Re­ been developed.l97] Other subunit vaccines are also sults from ongoing primate protection experiments being developed based on internal proteins of the using different constructs of HIV and SIV nucleic virus, particularly the p24 core protein, which con­ acid vaccines are eagerly awaited, since this ap­ tain conserved T cell epitopes which induce cell­ proach may have many of the advantages of live mediated immune responses.[98-101] Animal protec­ attenuated vaccines, without many of its disadvan­ tion data with these synthetic peptides or with tages.

© Adls International Limited All rights reserved. Drugs 50 (5) 1995 HIV Preventive Vaccines 797

3. Phase 1/11 Trials of the homologous LAI strain, and to a lesser degree, HIV Preventive Vaccines against the heterologous (but also clade B virus) SF2 strain.D 28) A comparison between these im­ Since 1987, at least 17 candidate vaccines have mune responses, and those induced in chimpanzees been tested in small-scale phase I trials to assess which have been protected against HIV infection their safety and in HIV-negative by the same candidate vaccine, showed that the human volunteers; the early results ofthese candi­ half-life of the antibody response to gp120 was date vaccines have been reviewed elsewhere.[4,5,127) longer in humans (9 weeks) than in chimpanzees In summary, most of these candidate vaccines (2 weeks). On the other hand, immune responses have been based on the subunit recombinant enve­ to V3 loop epitopes, and virus neutralisation titres, lope concept, and they have been found to be gen­ were approximately lO-fold lower in humans than erally well tolerated in doses that are capable of in chimpanzees) 129) A similar subunit recombinant inducing HIV-specific immune responses, These envelope vaccine, but based on the more repre­ immune responses include binding antibodies sentative MN strain (gpI20/MN), also induced (measured by gp120- or pI60-enzyme-linked im­ antibodies that neutralised the homologous MN munosorbent assay, Western Blot or V3 peptide strain, as well as other clade B viruses (SF2 and binding assays), as well as production of functional LAI)) 130) antibodies with the ability to neutralise HIV infec­ A gp 120/SF2 candidate vaccine, combined with tivity, to mediate antibody-dependent cell cyto­ a novel adjuvant, MF59, and administered with or toxicity, to block gp 120-CD4 binding and to in­ without the immunomodulator muramyl tripeptide hibit fusion of HIV-infected cells. CD4+ CTLs, dipalmitoyl phosphatidylethanolamine (MTP-PE), and in some instances CD8+ CTLs, have been induced gpl20-binding antibodies that persisted identified in vaccine recipients, especially in those receiving live-vectored vaccines. for at least 24 weeks after a fourth injection. After 3 injections, all volunteers developed neutralising On the basis of this information, many of the candidate vaccines evaluated in the past have now antibodies against the homologous SF2 strain, been followed by newer generations that are based which in two-thirds of them also cross-neutralised on different virus strains (i.e. MN strain instead of the MN strain.[I3l) the originally used LAI strain, both of which be­ A fully glycosylated gp160ILAI candidate vac­ long to clade B), that use different manufacturing cine, combined with alum and deoxycholate adju­ systems (with a shift towards the use of mamma­ vant, also resulted in a dose-related induction of lian cells for the production of recombinant enve­ neutralising antibodies against the homologous lope glycoproteins), or that use newer adjuvants (in LAI strain)I32) The elicited a CTL re­ addition to the use of the standard aluminium hy­ sponse, which was not mediated by classic CD8+ droxide). Emphasis is also being put on vaccine cells, but rather by cells of the CD4+ pheno­ approaches which result in CTL responses. type)133) More recent results have been published from Finally, vaccination with a chimaeric gp160 phase I clinical trials of several subunit recom­ molecule, consisting of MN gp 120 and LAI gp41, binant envelope vaccines, all of them pro­ and boosting with a linear V3-MN synthetic pep­ duced in mammalian cells: gp120ILAI (Genentech), tide, resulted in high levels of neutralising antibod­ gp120/MN (Genentech), gp120/SF2 (Biocine), ies against the homologous MN strain. In about gp160/LAI (Immuno), and gp160ILAI/MN + V3 half of the volunteers, these antibodiies also neu­ peptides (Pasteur-Merieux). tralised the SF2 strain, but not the LAI isolate.[I34) Injection of a gp120ILAI candidate vaccine in Thus, a number of mammalian-derived subunit aluminium hydroxide adjuvant resulted in a dose­ recombinant envelope candidate vaccines have related induction of neutralising antibodies against been shown to be well tolerated and to induce neu-

© Adls International limited, All tights reserved Drugs 50 (5) 1995 798 Esparza et a/.

Table I. Progress in the develol'lment of HIV-1-preventive candidate vaccines (based on published Information only) Vaccine concept Candidate vaccine Preclinical Human clinical trials (developer) studies phase I phase II Subunit nonreplicating (envelope) gp120/LAI (Genentech) .r .r gp120/MN (Genentech) .r .r .r gp120/SF2 (Bioclne) .r .r .r gp160/LAI (Immuno) .r .r gp120/MN/LAI (Pasteur-Merieux) + V3/MN .r .r synthetic peptldes V3/MN synthetic peptldes (UBI) .r .r live-recombinant vector (envelope) VaCCInia vector gp160/LAI (Bnstol-Myers Squibb) + .r .r gp160/LAI (MicroGeneSys) Canary pox vector gp160/MN/LAI + gp160/MN/LAI .r .r (Pasteur-Meneux) Whole Inactivated .r (SIV) Live attenuated .r (SIV) Nucleic aCid .r (SIV) Abbreviation: SIV = simian immunodeficiency virus tralising antibodies efficiently against the homolo­ ing with the gp160 subunit preparation resulted in gous strains, which in a dose-dependent fashion increasing levels of antibodies and cellular re­ also neutralise other strains from the same B clade. sponses.[l36-138] It is important to indicate, however, that most neu­ Similar prime-boost trials are being conducted tralisation experiments have been performed using with a canarypox vector encoding gp I60/MN (Pas­ laboratory-adapted strains of HI V-I and, as discus­ teur-Merieux), as well as with other poxvirus vec­ sed above, the antibodies were not capable of neu­ tors, although full results of these trials have not tralising in vitro clinical isolates of HIV- I.l92,135] yet been published. [1 34] Two of the candidate vaccines described above Similarly, data from ongoing trials with a multi­ (gp120 HIVIMN and gp120/SF2) are presently be­ meric V3 (United Biomedical, ing evaluated in a phase II trial in the US among Inc.) have not been published.[l39] Candidate vac­ 300 healthy volunteers, some of whom are at high cines based on the nucleic acid immunisation con­ risk of infection.[l27] cept may enter phase I clinical trials in the near future. However, phase I testing of other vaccine Another vaccine concept which is beginning to concepts, such as using whole inactivated virus or be intensively explored is based on the use of live live-attenuated vaccines, may have to wait until ad­ recombinant vector vaccines, which are in most ditional preclinical data are obtained, especially in cases administered in combination with a subunit relation to their safety. recombinant envelope product. This approach aims Table I summarises the progress in the develop­ at the induction of both neutralising antibodies and ment of HIV candidate vaccines in relation to ani­ CD8+ CTLs. In fact, early clinical trials indicated mal protection experiments and phase IIII clinical that priming with a live recombinant vaccinia vec­ trials. tor encoding gp160 (Bristol-Myers Squibb), and boosting with a baculovirus-derived gp160 subunit 4. The Future: Prospects for the product (MicroGeneSys), resulted in stronger anti­ Conduct of Phase III Efficacy Trials body and cellular responses than those following either vaccine alone. These immune responses Although phase 1111 trials have shown that HIV waned after approximately 1 year, though reboost- candidate vaccines are capable of inducing HIV-l-

© Adis International limited. All rights reserved. Drugs 50 (5) 1995 HIV Preventive Vaccines 799

specific immune responses, they have not provided End-points for vaccine efficacy that are being information on the potential efficacy of the candi­ considered include prevention of infection, pre­ date vaccines because the immune correlates of vention of the establishment of chronic infection protection are not known. and the delay or prevention of progression to dis­ From the more than 1400 participants in phase ease. I and II trials sponsored by the National Institutes Preparation for the efficacy evaluation of HIV of Health of the US, at least 8 have acquired HIV vaccines in large-scale field trials will require ex­ infection during the studies.l140,141] Most of these tensive international collaboration and coordina­ infections occurred before the complete immunisa­ tion between host countries, research institutions, tion sequence was administered, and all infected pharmaceutical manufacturers, regulatory authori­ volunteers reported high-risk behaviour. HIV in­ ties, and national and international institu­ fections among vaccinees were expected, since the tions.[148,149] This preparation includes strengthen­ immunisations were given over several months, ing of research infrastructures, identification and and since no vaccine is 100% protective. Conse­ characterisation of potential populations for phase quently, conclusions about vaccine efficacy cannot III trials including epidemiological studies on HIV be drawn from the ongoing phase 1111 trials, but incidence,l150] social-behavioural studies to ensure large-scale phase III trials are required, involving proper counselling and informed consent,[151] viro­ thousands of volunteers at high risk of infec­ logical studies for the characterisation of prevalent tion.[142] strains in the trial population,[58] and the develop­ Decisions to initiate efficacy trials of HIV can­ ment of guidelines to ensure the conduct of the didate vaccines will be based on a careful analysis trials with the highest scientific and ethical stand­ of the scientific criteria, including: (i) animal pro­ ards,D52,153] ensuring the appropriate preparation tection data; (ii) safety and immunogenicity in hu­ and participation of the community. mans; (iii) information on potential correlates of It is likely that the eventual development of a protection; (iv) severity ofthe epidemic; (v) public well tolerated, highly effective and available HIV health considerations based on a r~sk/benefit anal­ vaccine will be a long and difficult process, requir­ ysis of proceeding with available candidate vac­ ing the development of a number of generations of cines or waiting for more data or for 'better' can­ vaccines over many years. In this process, it is im­ didate vaccines; (vi) feasibility of conducting a portant that.the initial efficacy trials are well de­ large-scale trial, including recruiting and retaining signed and conducted, and generate the informa­ the large number of volunteers needed to tion needed for the development of more demonstrate efficacy; and (vii) political and com­ promising vaccines in the future. munity support for the implementation of the triaUI43 ,144] References It is obvious that an analysis of the above cri­ 1. Merson MH. Slowing the spread ofHIV: agenda for the 1990s. teria may result in different decisions in different Science 1993; 260: 1266-8 countries.D44,1451 Some developing countries are 2. Mertens TE, Burton A, Stoneburner R, et al. Global estimates now actively preparing for phase III trials,[146,147] and of HIV infections and AIDS. AIDS 1994; 8 Suppl. 1: S361-72 which may be initiated within the next 2 years. 3. Esparza J, Osmanov S, Kallings LO. Planning for HIV vaccine These trials will be designed to test different vac­ trials: the World Health Organization perspectIve. AIDS 1991; 5 Suppl. 2: S159-63 cine concepts in their ability to protect: (a) against 4. Esparza J, Osmanov S. The development and evaluatIOn ofHIV homologous and/or heterologous virus subtypes; vaccmes. Curr Opm Infect Dis 1993: 6: 218-29 (b) against different routes of transmission; and (c) 5. Walker MC, Fast PE. Clmical trials of experimental AIDS vac­ cmes. AIDS 1994; 8 Suppl. I: S213-36 in the presence/absence of other co-factors (e.g. 6. Haynes BE Scienlific and socia1Issues of human Immunodefi­ sexually transmitted diseases). ciency VIrUS vaccme development. SCIence 1993; 260: 1279-86

© Adls International limited. All rights reserved Drugs 50 (5) 1995 800 Esparza et al.

7. Hilleman MR. Vaccinology, immunology, and comparative 31. Moore J, Cao Y, Ho DD, et al. Development of the anti-gpl20 pathogenesis of measles in the quest for a preventative against antibody response during seroconversion to human immuno­ AIDS. AIDS Res Hum Retroviruses 1994; 10: 3-12 deficiency virus type I. J Viro11994; 68: 5142-55 8. Hoth DF, Bolognesi DP, Corey L, et al. HIV vaccine develop­ 32. Baltimore D. Lessons from people with nonprogressive HIV ment: a progress report (NIH Conference). Ann Int Med 1994; infection. N Engl J Med 1995; 332: 259-60 8: 603-11 33. Pantaleo G, Menzo S, Vaccarezza M, et al. Studies in subjects 9. Letvin N. Vaccines against human immunodeficiency virus - with long-term nonprogressive human immunodeficiency vi­ progress and prospects. N Engl J Med 1993; 329: 1400-5 rus infection. N Engl J Med 1995; 332: 209-16 10. Norley SG, Vogel T, Kurth R. Anti-HIV vaccines: current status 34. Mellors JW, Kmgsley LA, Rinaldo CR, et al. Quantitation of and future development. Drugs 1993; 46: 947-60 HIV-I RNA in plasma predicts outcome after seroconversion. 11. Paul WE. Reexamining AIDS research priorities. SCience 1994; Ann Int Med 1995; 122: 573-9 267: 633-6 35. Chakrabarti L, Cumont M-C, Montagnier L, et al. Variable 12. Vermund SH, Schultz AM, Hoff R. Prevention of HIV/AIDS course of primary simian immunodeficiency virus infection with vaccines. Curr Opin Infect Dis 1994; 7: 82-94 in lymph nodes: relation to disease progression. J Virol 1994; 13. Clericl M. Cell-mediated immunity in HIV infection. AIDS 68: 6634-42 1993; 7 Suppl. I: S135-40 36. Hirsch VM, Goldstein S, Hynes NA, et al. Prolonged clinical 14. Cohen J. AIDS research shifts to immunity. Science 1992; 257: latency and survival of macaques given a whole inactivated 152-3 Simian immunodeficiency virus vaccine. J Infect Dis 1994; 15. Mosmann TR. Cytokine patterns during the progression to 170: 51-9 AIDS. Science 1994; 265: 193-4 37. Cohen J. A new goal: preventmg disease, not infection. Science 16. Pantaleo G, Graziosi C, Fauci AS. The immunopathogenesls of 1993; 262: 1820-1 human immunodeficiency virus infection. N Engl J Med 38. Miedema F, Meyaard L, Klem MR. Protection from HIV infec­ 1993;328: 327-35 tion or AIDS? Science 1993; 262: 1074-5 17. FaucI AS. Multifactorial nature of human immunodeficiency 39. Salk J, Bretscher PA, Salk PL, et al. A strategy for prophylactic virus disease: Implications for therapy. Science 1993; 262: vaccines against HIV. Science 1993; 260: 1270-2 1011-8 40. Embretson J, Zupancic M, Ribas JL, et al. Massive covert infection 18. Levy JA. HIV pathogenesis and long term survival. AIDS 1993; of helper T lymphocytes and macrophages by HIV during the 7: 1401-10 incubation period of AIDS. Nature 1993; 362: 359-62 19. Rowe P. Resistance to HIV infectIon. Lancet 1993; 341: 624 41. Pantaleo G, Graziosi C, Demarest JF, et al. HIV infection is 20. Cierici MJ, Giorgi JV, Chuo C-C, et al. Cell-mediated immune active and progressive in lymphoid tissue during the clinically response to human immunodeficiency virus (HIV) type I in latent stage of disease. Nature 1993; 362: 355-8 seronegative homosexual men with recent sexual exposure to 42. Ho DD, Neumann AU, Perelson AS, et al. Rapid turnover of HIV-1. J Infect Dis 1992; 165: 1012-9 plasma virions and CD4lymphocytes in HIV-l infection. Na­ 21 . Clerici MJ, Levme HA, Kessler A, et al. HIV-specific T-helper ture 1995; 373: 123-6 activity in seronegative health care workers exposed to con­ 43. Wei X, Ghosh SK, Taylor ME, et al. Vlfal dynamics in human taminated blood. JAMA 1994; 217: 42-6 Immunodeficiency virus type I infection. Nature 1995; 373: 22. De Maria A, Cinllo C, Moretta L. Occurrence of human immu­ 117-22 nodeficiency virus type I (HIV-I )-speciflc cytolytic T cell 44. Veugelers PJ, Page KA, Tindall B, et al. Determinants of HIV actIvity in apparently uninfected children born to HIV-I in­ disease progression among homosexual men registered in the fected mothers. J Infect Dis 1994; 170: 1296-9 tricontinental seroconverter study. Am J EpidemlOl 1994; 23. Kiprow DD, Sheppard HW, Hanson Cv. AliOimmunization to 140: 747-58 prevent AIDS? Science 1994; 263: 737-8 45. Easterbrook PJ. Non-progression in HIV mfection. AIDS 1994; 24. Rowland-Jones SL, Nixon DF, Aldhous MC, et al. HIV-specific 8: 1179-82 cytotoxic T-cell activity in an HIV-exposed but uninfected 46. Buchbinder SP, Katz MH, Hessol NA, et al. Long-term HIV-I mfant. Lancet 1993; 341: 860-1 mfection without immunological progressIOn. AIDS 1994; 8: 25. Shearer GM, Clericl M, Dalgleish A. Alloimmunization as an 1123-8 AIDS vaccine? Science 1993; 262: 161-2 47. Cao Y, Quin L, Zhang L, et al. Virologic and immunologic char­ 26. Willeford DM, Bwayo n, Hensel M, et al. Human immunode­ acterization of long-term survivors of human immunodefi­ ficiency virus infection among high-risk seronegative prosti­ ciency virus type I infectIon. NEnglJ Med 1995; 332: 201-8 tutes in Nairobi. lInfectDis 1993; 167: 1414-7 48. Wong MT, Warren RQ, Anderson SA, et al. Longitudinal anal­ 27. Niu MT, Stein DS, Schmttman SM. Primary human immuno­ ysis of the humoral response to human immunodeficiency deficiency virus type I infection: review of pathogenesis and ViruS type I (HIV-I) gp 160 epitopes in rapidly progressing early treatment intervention in human and animal retrovirus and nonprogressing HIV-I-infected subjects. J Infect Dis infections. J Infect Dis 1993; 168: 1490-501 1993; 168: 1523-7 28. Koup RA, Ho DD. Shutting down HIV. Nature 1994; 370: 416 49. Huang Y, Zhang L, Ho DD. Characterization of nefsequences 29. Koup RA, Safrit JT, Cao Y, et al. Temporal association of cel­ in long-term survivors of human Immunodeficiency virus lular immune responses with the initial control of viremia in type 1 infection. J Virol 1995; 69: 93-100 pnmary human immunodeficiency virus type I syndrome. J 50. KirchhoffF, Greenough TC, Brettler DB, etal. Absence of intact Virol 1994; 68: 4650-5 nef sequences in a long-term survivor With nonprogressive 30. Pantaleo G, Demarest JF, Soudeyns H, et al. Major expansion HIV-I infection. N Engl J Med 1995; 332: 228-32 of CD8+ T cells with a predominant V~ usage during the 51. Conley AJ, Kessler JA, Boots LJ, et al. Neutralization of diver­ primary immune response to HIV. Nature 1994; 370: 463-7 gent human Immunodeficiency virus type I variants and

© Adis International Limited. All rights reserved. Drugs 50 (5) 1995 HIV Preventive Vaccines 801

primary isolates by IAM-41-2F5, an anti-gp41 human 67. Louwagie J, Delwart EL, Mullins 11, et al. Genetic analysis of monoclonal antibody. Proc Nat! Acad Sci USA 1994; 91: HIV-I isolates from Brazil reveals the presence of two distinct 3348-52 genotypes. AIDS Res Hum Retroviruses 1994; 10: 561-7 52. Earl PL, Broder CC, Long D, et al. Native oligomenc human 68. Louwagie J, Janssens W, Mascola J, et al. Genetic diversity of immunodeficiency virus type I envelope glycoprotein elicits the envelope glycoprotein from human immunodeficiency vi­ diverse monoclonal antibody reactivities. J Virol 1994; 68: rus type 1 isolates of African origin. J Viro11995; 69: 263-71 3015-26 69. Sabino EC, Shpaer EG, Morgado MG, et al. Identification of 53. Kang C-Y, Hariharan K, Nara PL, et al. with a human immunodeficiency virus type I envelope genes re­ soluble CD4-gp120 complex preferentially induces neutral­ combinant between SUbtypes Band F in two epidemiological izing anti-human immunodeficiency virus type I antibodies linked individuals from Brazil. J Viro11994; 68: 6340-6 directed to conformation-dependent epitopes of gp 120. J 70. Sharp PM, Robertson DL, Gao F. Origins and diversity of human Virol 1994; 68: 5854-62 immunodeficiency viruses. AIDS 1994; 4 Suppl. I: S27-42 54. Laal S, Burda S, Gorny MK, et al. Synergistic neutralization of 71. Gurtler LG, Hauser PH, Eberle J, et al. A new subtype of human human immunodeficiency virus type I by combinations of immunodeficiency virus type I (MVP-5180) from Camer­ human monoclonal antibodies. J Viro11994; 68: 4001-8 oon. J Viro11994; 68: 1581-5 55. Moore JP, Ho DD. Antibodies to discontinuous or conforma­ 72. Nkengasong IN, Peeters M, Ndumbe P, et al. Cross-neutralizmg tionally sensitive epitopes on the gp 120 glycoprotein of hu­ antibodies to HIV-IANT7oand HIV-llIIbin sera of African and man immunodeficiency virus type I are highly prevalent in Belgian HIV-l-infected individuals. AIDS 1994; 8: 1089-96 sera of infected humans. J Virol 1993; 67: 863-75 73. Peeters M, Nkengasong J, Willems B, et al. Antibodies to V3 56. Muster T, Steindl F, Purtscher M, et al. A conserved neutralizing loop peptides derived from chimpanzee lentiviruses and the epitope on gp41 human immunodeficiency virus type I. J divergent HIV-IANT-7oisolate in human sera from different Virol 1993; 67: 6642-7 geographic regions. AIDS 1994; 8: 1657-61 57. Myers G, Korber BTM, Smith RF, et al. Human retroviruses 74. Vanden Hasevelde M, Decourt J-L, De Leys RI, et al. Genomic and AIDS. Los Alamos: Theoretical Biology and Biophysics cloning and complete sequence analysis of a highly divergent Group, Los Alamos National Laboratory, 1993 African human immunodeficiency virus isolate. J Viro11994; 58. WHO Network for HIV Isolation and Characterization. HIV­ 68: 1586-96 type 1 variation in World Health Organization-sponsored vac­ 75. Zekeng L, Gurtler L, Afane Ze E, et al. Prevalence of HIV-I cine evaluation sites: genetic screening, sequence analysis, SUbtype 0 infection in Cameroon: preliminary findings. and preliminary biological characterization of selected viral AIDS 1994; 8: 1626-7 strains. AIDS Res Human Retroviruses 1994; 10: 1327-43 76. Cheingsong-Popov R, Lister S, Callow D, et al. Serotyping HIV 59. Bachmann ME, Delwart EL, Shpaer EG, et al. Rapid genetic type I by antibody binding to the V3100p: relationship to viral characterization of HIV strains from four World Health Or­ genotype. AIDS Res Hum Retroviruses 1994; 10: 1379-86 ganization-sponsored vaccine evaluation sites using a heteroduplex mobility assay. AIDS Res Hum Retroviruses 77. Pau C-P, Kai M, Holloman-Candal DL, et al. Antigenic vana­ tion and serotyping of HIV type 1 from four World Health 1994; 10: 1345-53 60. Bobkov A, Cheingsong-Popov R, Garaev M, et al. Identification OrganizatIOn-sponsored HIV vaccine sites. AIDS Res Hum of an env G subtype and heterogeneity of HIV-l strains in the Retroviruses 1994; 10: 1369-77 Russian FederatIOn and Belarus. AIDS 1994; 8: 1649-55 78. Osmanov S,Heyward WL, Esparza J. The World Health Orga­ 61. Cernescu CE, Tardei G, Necula A, et al. The serologic signifi­ nization network for HIV isolation and characterization. cance of F viral genotype for human immunodeficiency virus AIDS Res Hum Re'troviruses 1994; 10: 1325-6 type I epidemic. J Infect Dis 1994; 170: 1043-4 79. Pau C-P, Kai M, Holloman-Candal DL, et al. Antigenic varia­ 62. Dumitrescu 0, Kalish ML, Kliks SC, et al. Characterization of tion and serotyping of HIV type 1 from four World Health human immunodeficiency virus type I isolates from chIldren Organization-sponsored HIV vaccine sites. AIDS Res Hum in Rumania: identification of a new envelope subtype. J Infect Retroviruses 1994;' 10: 1369-77 Dis 1994; 169: 281-8 79. Moore JP, McCutchan FE, Poon S-W, et al. Exploration of an­ 63. Gao F, Yue L, Craig S, et al. Genetic variation of HIV type I in tigenic variation in gp 120 from clades A through F of human four World Health Orgamzation-sponsored vaccine evalua­ immunodeficiency vi'rus type 1 by using monoclonal antibod­ tion sites: generation of functional envelope (glycoprotem ies. J Virol 1994; 68: 8350-64 160) clones representative of sequence subtypes A, B, C, and 80. Mascola JR, Louwagie J, McCutchan FE, et al. Two antigeni­ E. AIDS Res Hum Retroviruses 1994; 10: 1359-68 cally distinct SUbtypes of human immunodeficiency virus 64. Janssens W, Heydrickx L, Van de Peer Y, et al. Molecular phy­ type 1: viral genotype predicts neutralization serotype. J In­ logeny of part of the env gene of HIV-I strains isolated in fect Dis 1994; 169: 48-54 Cote d'Ivoire. AIDS 1994; 8: 21-6 81. De Wolf F, HogervorstlE, Goudsmit J, et al. Syncytium-induc­ 65. Korber BTM, Osmanov S, Esparza J, et al. The World Health ing and non-~yncytium inducing capacity of human immuno­ Orgamzation Global Programme on AIDS proposal for stand­ deficiency virus type '1 subtypes other than B: phenotypic and ardization of HIV sequence nomenclature. AIDS Res Hum genotypic characteristics. AIDS Res Hum Retroviruses 1994; Retroviruses 1994; 10: 1355-8 10: 1387-400 66. Louwagie J, McCutchan F, Peeters M, et al. Phylogenetic anal­ 82. Rubsamen-Waigmann H, Von Briesen H, Holmes H, et al. ysis of gag genes from seventy international HIV-J isolates Standard conditions of virus isolation reveal biological vari­ provided evidence for multiple genotypes. AIDS 1993; 7: ability of HI:V type 1 in different regions of the world. AIDS 769-80 Res Hum Retroviruses 1994; 10: 1401-8

© Adis International Limited. All rights reserved. Drugs 50 (5) 1995 802 Esparza et al.

83. Zhu T, Mo H, Wang N, et al. GenotypIc and phenotypic charac­ 101. Wolf HJ, Modrow S, Wagner R The HIV-I Pr55gag protem: terization of HI V-I m patIents with primary infection. Science understanding the mechamsm of virus assembly as a baSIS for 1993; 261: 1179-81 the development of novel therapeutIcs and vaccmes. In: Gi­ 84. Bou-Habib DC, Rodenquez G, Oravecz T, et al. Cryptic na­ rard M, Valette L, editors. Retroviruses of human AIDS and ture of envelope V3 regIOn epltopes protects primary related animal diseases. 8< Colloque des 'Cent Gardes' ; 1993 monocytotropic human immunodefIciency virus type I from Oct 25-27: Marnes-La-Coquette. Paris: Fondation Marcel antibody neutralization. J Virol 1994; 68: 6006-13 Merieux, 1994: 299-307 85. Sawyer LSW, Tern Wrin M, Crawford-Mlksza L, et al. Neutral­ 102. Adams SE, Paoletti E. Use of new vectors for the development ization sensItivity of human immunodeficiency VIruS type I of vaccines. AIDS 1993; 7 Suppl. I: S141-6 is determined in part by the cell in whIch the VIruS IS propa­ 103. Andino R, Silvera D, Suggett SD, et al. Engineering poliovirus gated. J Virol 1994; 68: 1342-9 as a vaccine vector for the expression of diverse antigens. 86. Fultz PN. Nonhuman pnmate models for AIDS. Clin Infect Dis Science 1994; 265: 1448-51 1993; 17 Suppl. I: S230-5 104. Natuk RJ, Lubeck MD, Chanda PK, et al. Immunogemclty of 87. Warren JT, Dolatshahi M. Annual update survey of worldwide Adeno-HIV vaccines in chImpanzees. In: Girard M, Valette HIV, SIV, and SHIV challenge studies m vaccmated nonhu­ L, editors. Retroviruses of human AIDS and related animal man primates. J Med Pnmatol 1994; 23: 184-225 dIseases. 8< Colloque des 'Cent Gardes'; 1993 Oct 25-27: 88. Stott EJ. Anti-cell antibodyin macaques. Nature 1992; 353' 393 Marnes-La-Coquette. Paris: Fondation Marcel Meneux, 89. Arthur LO, Bess JW, Sowder RC, et al. Cellular proteins bound 1994: 115-20 to Immunodeficiency viruses: Implications for pathogeneSIs 105. TartaglIa J, Franchini G, Robert-Guroff M, et al. Highly atten­ and vaccines. Science 1992; 258: 1935-8 uated poxvirus vector strains, NYVAC and ALVAC, in retro­ 90. Fultz PN, Nara P, Barre-Sinoussi F, et al. Vaccine protection of virus vaccine development. In: GIrard M, Valette L, editors. chimpanzees against challenge with HIV-I-infected periph­ Retroviruses of human AIDS and related animal diseases. 8< eral blood mononuclear cells. Science 1992; 256: 1687-90 Colloque des 'Cent Gardes'; 1993 Oct 25-27: Marnes-La-Co­ 91. GIrard M, Meignier B, Matthews T, et al. Protection of chim­ quette. Paris: FondatlOn Marcel Merieux, 1994: 293-8 panzees against heterologous HIV-I challenge. In: Guard M, 106. Vella C, Ferguson M, Dunn G, et al. Characterization and pri­ Valette L, editors. Retroviruses of human AIDS and related mary structure of a human Immunodeficiency virus type I animal diseases. 9< Colloque des 'Cent Gardes'; 1994 Oct (HIV-I) neutralizatIOn domam as presented by a poliovirus 24-26, Marnes-La-Coquette. Paris: Fondation Marcel type IIHIV-I chimera. J Gen Vlro11993; 74: 2601-7 Merieux, 1995 : 263-6 107. Daniel MD, Kirchoff F, CzaJak SC, et al. Protective effects of a 92. Graham BS. Serological responses to candidate AIDS vaccmes. live attenuated SIV vaccme with a deletion in the nef gene. AIDS Res Hum Retroviruses 1994; 10 Suppl. 10: S 145-8 Science 1992; 258: 1938-41 93 de Vnes P, Heeney JL, Boes J, et aI . ProtectIOn of rhesus ma­ 108. Desrosiers RC. HIV with multiple gene deletions as a lIve at­ caques from SIV infection by immunizatIOn with different tenuated vaccine for AIDS. AIDS Res Hum Retroviruses expenmental SIV vaccines. Vaccine 1994, 12. 1443-52 1992; 8: 411-21 94. Giavedoni LD, Planelles V, Haigwoqd NL, et al. Immune re­ 109. Kirchoff F, Kestler HW, DesrOSIers RC. Upstream U3 se­ sponse of rhesus macaques to recombinant simian immuno­ quences in SImian immunodeficIency virus are selectively de­ defICIency virus gp 130 does not protect from challenge infection. J Virol 1993; 67. 577-83 leted in vivo in the absence of an mtact nef gene. J Vlrol 1994, 68: 2031-7 95. Israel ZR, Edmonson PF, Maul DH, etal. Incomplete protection, 110. I1ymskil PO, Daniel MD, Simon MA, et al. The role of upstream but not suppression of virus burden, elicited by subunit simian immunodeficiency virus vaccines. J Virol 1994; 68: 1843-53 U3 sequences m the pathogenesis of silman immunodefi­ ciency virus-induced AIDS in rhesus monkeys. J Virol 1994; 96. Putkonen P, Bjorling E, Akerblom L, et al. Long-standing pro­ tection of macaques against cell-free HIV-2 with a HIV-2 68: 5933-44 iscom vaccine. J Acquir Immune Defic Syndr 1994; 7: 551-9 III. Kleney MP, Aubertm AM, Benavente A, et al. Protection of 97. Wang CY, Looney DJ, Li ML, et al. Long term hIgh titer neu- monkeys against SIV infectIOn with live attenuated viruses. tralizing activity induced by octameric synthetic HIV-I anti­ In' Girard M,,Valette L, editors. Retroviruses of human AIDS gen. SCIence 1991; 254: 285-8 and related animal dIseases. 8< Colloque des 'Cent Gardes'; 98. GriffIths JC, Harris SJ, Layton GT, et al. Hybrid human Immu­ 1993 Oct 25-27: Marnes-La-Coquette. Paris: Fondation Mar­ nodeficiency VIruS gag particles as an antigen carrier system: cel Merieux, 1994: 211-8 mduction of cytotoxic T-cell and humoral responses by a gag: 112. Desrosiers RC, Damel MD, Wyand MS, et al. A radical strategy V3 fusion. J Virol 1993; 67: 3191-8 for development of a vaccine against HIV-I. In: Girard M, 99. Martin SJ, Vyakarnam A, Chemgsong-Popov R, et al ­ Valette L, editors. Retroviruses of human AIDS and related zatIOn of human HIV-seronegative volunteers with recombI­ animal diseases. 8< Colloque des 'Cent Gardes' ; 1993 Oct nant p 17 /p24: Ty virus-like particles elicits HIV-I 25-27: Marnes-La-Coquette. Pans: FondatlOn Marcel p24-specific cellular and humoral immune responses. AIDS Merieux, 1994: 205-9 1993; 7: 1315-23 113 Norley S. Live attenuated immunodefIciency vIruses: Interest­ 100. Wagner R, Boltz T, Deml L, et al. InductIOn of cytolytic T lym­ ing sCIentifIc tools or vaccines of the future? In: Girard M, phocytes directed towards the V3 loop of the human Immu­ Valette L, editors. Retroviruses of human AIDS and related nodeficiency virus type I external glycoprotem gp 120 by animal diseases. 8< Colloque des 'Cent Gardes'; 1993 Oct p55gaW3 chimeric vaccinia viruses. J Gen Virol 1993; 74. 25-27: Marnes-La-Coquette. Paris: Fondation Marcel 1261-9 Merieux, 1994: 199-203

© Adrs International Limited. All rights reserved. Drugs 50 (5) 1995 HIV Preventive Vaccines 803

114. Bolognesl DP. Controversies in science: a live-virus AIDS vac­ 132. Belshe RE, Clements ML, Dolin R, et al. Safety and Immuno­ cine? Not yet, It is too early to consider use of a live-attenuated gemcity of a fully glycosylated recombinant gp160 human virus vaccine against HIV-1. J NIH Res 1994; 6: 55, 59-62 immunodeficiency virus type 1 vaccine in subjects at low risk 115. Cohen J. At conference, hope for success is further attenuated. of infection. IInfect Dis 1993; 168: 1387-95 SCience 1994; 266: 1154 133. Stanhope PE, Clements ML, Siliciano RF. Human CD4+ cyto­ 116. Desrosiers RC. Controversies m science: a live-virus AIDS vac­ lytiC T lymphocyte responses to a human immunodeficiency cine? Yes , It is time to consider use of a live-attenuated virus ViruS type 1 gp160 subumt vaccine. J Infect Dis 1993; 168: vaccme against HIV-1. J NIH Res 1994; 6: 54,56-59 92-100 117. World Health Organization Workmg Group. Feasibility of de­ 134. Excler JL, Salmon D, Plaloux G, et al. Combmed antigen veloping live attenuated HIV vaccmes: conclusIOns and rec­ regimes for vaccinatIOn against HIY. In' Girard M, Valette L, ommendatIOns. AIDS Res Hum Retroviruses 1994; 10: 221-2 editors. RetroViruses of human AIDS and related animal dis­ 118. Cichutek K. Nucleic acid immunization: a prophylactic gene eases. 8e Colloque des 'Cent Gardes'; 1993 Oct 25-27: therapy? Vaccine 1994; 12: 1520-5 Marnes-La-Coquette. Pans: Fondation Marcel Merieux, 119. Donnelly JJ, Ulmer JB, LIU MA. Immunization with poly­ 1994: 281-4 nucleotides: a novel approach to vaccmation. Immunologist 135. Bolognesi DP. Vaccine climcal trials: chairman's overview. In: 1994,2: 20-6 Girard M, VaIette L, editors. Retroviruses of human AIDS and 120. Fynan EF, Webster RG, Fuller DH, et al. DNA vaccmes: pro­ related ammal diseases. 8e Colloque des 'Cent Gardes'; 1993 tective by parenteral, mucosal, and gene-gun Oct 25-27: Marnes-La-Coquette. Paris: Fondation Marcel . Proc Natl Acad Sci USA 1993; 90: 11478-82 Merieux, 1994: 267-70 121. Raz E, Carson DA, Parker SE, et al. Intradermal gene Immuni­ 136. EI-Daher N, Keefer MC, Reichman RC, et al PefSlsting human zation: the possible role of DNA uptake in the mduction of immunodeficiency ViruS type 1 gpl60-speciflc human T lym­ cellular Immunity to viruses. Proc Natl Acad Sci USA 1994; phocyte responses including CD8+ cytotoxIc activity after 91: 9519-23 receipt of envelope vaccines. J Infect Dis 1993; 168: 306-13 122. Robertson JS. Safety consideratIOns for nucleic acid vaccmes. 137. Graham BS, Gorse GJ, Schwartz DH, et al. Determinants of Vaccme 1994; 12: 1526-8 antibody response after recombinant gp 160 boostmg m vac­ 123. Robinson JL, Fynan EF, Webster RG, et al. Gene vaccines, a cinia-naive volunteers pnmed with gp 160-recombmant vac­ new approach to immunization. In: Girard M, Valette L, edi­ cinia virus. J Infect Dis 1994; 170: 782-6 tors. Retroviruses of human AIDS and related animal dis­ 138. McElrath MJ, Corey L, Berger D, et al. Immune responses elic­ eases. 8e Colloque des 'Cent Gardes'; 1993 Oct 25-27: ited by recombmant vaccmia-human immunodeficiency VI­ Marnes-La-Coquette. Paris' Fondation Marcel Merieux, rus (HIV) envelope and HIV envelope protein: analysis of the 1994: 335-41 durability of responses and effect of repeated boosting. J In­ 124. Schoedel F, Aguado M-T, Lambert P-H. IntroductIOn: nucleic fect Dis 1994; 169: 41-7 acid vaccmes, WHO, Geneva, 17-18 May 1994. Vaccine 139. O'Hagan DT, McGee JP, Wang CY, et al. The UBI multi­ 1994; 12: 1491-2 component HIVR vaccme: the advantages of controlled re­ 125. Slllith HA. Regulatory considerations for nucleiC acid vaccines. lease mlcroparticles. In: Girard M, Valette L, edilors. Vaccme 1994; 12: 1515-9 Retrovlfuses of human AIDS and related animal diseases. 8e 126. Wang B, Ugen KE, Srikantan V, et aI Gene moculatlOn gener­ Colloque des 'Cent Gardes' ; 1993 Oct 25-27: Marnes-La-Co­ ates immune responses against human immunodeficiency VI­ quette. Paris: FondatlOn Marcel Meneux, 1994: 309-13 rus type I. Proc Nat! Acad Sci USA 1993; 90: 4156-60 140. Belshe RB, Bolognesl DP, Clements ML, et al. HIV mfeclion 127. Fast PE , Walker MC, Ketter N, et al. Evaluation of candidate in vaccinated volunteers. JAMA 1994; 272. 431 HIV-I prophylactic vaccmes m phase IIII trials. In: Girard M, 141. Cohen J. Will media reports KO upcoming real-life tnals? Sci­ Valette L, editors. RetrOViruses of human AIDS and related ence 1994;264: 1660 animal diseases. 8e Colloque des 'Cent Gardes'; 1993 Oct 142. Mascola JR. McNeil JG, Burke OS. AIDS vaccmes: are we 25-27: Marnes-La-Coquette. Paris: Fondalion Marcel ready for human efficacy trials? JAMA 1994; 272: 488-9 Merieux, 1994:271-9 143. Lawrence ON, Esparza J, Zoon KC. Considerations for com­ 128 Schwartz DH, Gorse G, Clements ML, et al. Induction of HI V­ mencement of HIV vaccine efficacy trials: a working group I-neutralismg and syncytium-inhibltmg anlibodies in unin­ summary. AIDS Res Hum Retroviruses 1993; 9 Suppl. I: fected recipients of HIV-1IIIB rgp120 subunit vaccine. Lancet S65-9 1993,342: 69-73 144. World Health Orgamzation. Scientific and public health ratio­ 129 Berman PW, Eastman DJ, Wilkes DM, et al. Comparison of the nale for HIV vaccine efficacy trials. AIDS 1995; 9: WHOI-4 immune response to recombmant gp 120 m humans and chim­ 145. Cohen J. US Panel votes to delay real-world vaccine trials. panzees. AIDS 1994; 8: 591-601 SCience 1994; 264: 1839 130. Belshe RB, Graham MS, Keefer MC, et al. Neutralizmg anti­ 146. Kunasol P. International HIV/ AIDS vaccme trials: expectations bodies to HIV-l in seronegative volunteers immunized with of host countries. AIDS Res Hum RetroViruses 1993; 9Suppl. recombinant gp120 from the MN strain of HIV-1. JAMA I:S135-6 1994; 272: 475-80 147. Katongole-Mbidde E. Expectations of host countries m HIV 131. Kahn JO, Sinangil F, Baenziger J, et al. Clinical and immuno­ vaccme tnals. AIDS Res Hum Retroviruses 9 Suppl. 1: S17-8 logical responses to human immunodeficiency VlfUS (HIV) 148. Esparza J. Development of WHO-sponsored sites for HIV vac­ type lsF2gp120 subumt vaccme combined with MF59 adju­ cme evaluation. AIDS Res Hum retroviruses 1993; 8 Suppl. vant with or without muramyl tripeplide dipalmiloyl phos­ 1: S133-4 phatidylethanolamine in non-HIV-infected human 149. Esparza J, Osmanov S, Clements ML, et aI. Preparation for volunteers. J Infect Dis 1994; 170: 1288-91 efficacy trials of prevenlive HIV candidate vaccines: the role

© Adis International Limited. All rights reserved. Drugs 50 (5) 1995 804 Esparza et al.

of the World Health Organization. In: Girard M, Valette L, nizations in Africa; 1994 March 13-16; Yamoussoukro, Cote editors. Retroviruses of human AIDS and related animal dis­ d'Ivoire. Paris: CollectIOn Fondation Marcel Merieux, 1994: eases. 7" Colloque des 'Cent Gardes'; 1992 Oct 26-28: 330-1 Marnes-La-Coquette. Paris: Fondation Marcel Merieux, 153. Lurie P, Bishaw M, Chesney MA, et a!. Ethical, behavioural, 1993: 217-22 and social aspects of HlV vaccine trials in developing coun­ l, 150. Heyward WL, Osmanov S, Saba et a!. Preparation for phase tries. lAMA 1994; 271 : 295-301 III HlV vaccine efficacy trials: methods for the determmation ofHlV incidence. AIDS 1994; 8: 1285-91 151. Temoshok LR. Behavioral research contributions to plannmg and conducting HIV vaccine efficacy trials. AIDS Res Hum Correspondence and reprints: Dr Vaccine De­ Retroviruses 1994; 10 Supp!. 2: S277-80 Jose Esparza, 152. Esparza l, Osmanov S, Heyward W, et a!. Ethical aspects of velopment Unit, Division of Research and Intervention De­ trials of candidate vaccines against the human immunodefi­ velopment, Global Programme on AIDS, World Health ciency virus (HIV). Fourth International Seminar on Immu- Organization, 1211 Geneva 27, Switzerland.

© Adis International limited. All fights reserved. Drugs 50 (5) 1995