Buscar en
Enfermedades Infecciosas y Microbiología Clínica
Toda la web
Inicio Enfermedades Infecciosas y Microbiología Clínica Immunotherapy and therapeutic vaccines in HIV infection
Información de la revista
Vol. 23. Núm. S2.
Páginas 95-104 (Julio 2005)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Vol. 23. Núm. S2.
Páginas 95-104 (Julio 2005)
Acceso a texto completo
Immunotherapy and therapeutic vaccines in HIV infection
Inmunoterapia y vacunas terapéuticas en la infección por VIH
Visitas
185191
Felipe Garcíaa, Lidia Ruizb, Juan Carlos López-Bernaldo de Quirósc, Santiago Morenod, Pere Domingoe
a Servicio de Enfermedades Infecciosas. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universidad de Barcelona.
b Unitat VIH i Laboratori de Retrovirologia IrsiCaixa. Hospital Germans Trias i Pujol. Badalona.
c Hospital Universitario Gregorio Marañón. Madrid.
d Servicio de Enfermedades Infecciosas. Hospital Ramón y Cajal. Madrid.
e Servicio de Medicina Interna. Hospital de Sant Pau. Barcelona. Spain.
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Tablas (1)
TABLE 1. Immune-mediated therapies used in HIV-1 infection
Resistance to medication, adverse effects in the medium-long term, and cost are important limitations to lifelong adherence to highly active antiretroviral therapy (HAART). The combination of HAART with immune therapy to restore and/or boost immune-specific responses to HIV has been proposed, with the ultimate aim of controlling viral replication in the absence of HAART over long periods. The functional defects of the cellular and humoral responses would explain the lack of control of the immune system over viral replication. Different types of immune-mediated therapy have been investigated to solve these problems, including passive immune therapy, cytokines, structured treatment interruptions, immunosuppressors and therapeutic vaccines. Our still limited knowledge of immune mechanisms which can control HIV viral replication and of the causes of the deterioration of cellular and humoral immunity have produced only modest benefits in immune-mediated therapy, and are therefore confined to research for the time being. The availability of an optimal therapeutic vaccine would be an important scientific advance which could be compared with the arrival of protease inhibitors in clinical practice. Therefore, priority should be given to research in this field.
Keywords:
Inmunoterapia
VIH
Vacunas terapéuticas
El desarrollo de resistencia a la medicación, la aparición de efectos adversos a medio y largo plazo y el elevado coste económico constituyen importantes limitaciones para el cumplimiento de por vida de la terapia antirretroviral (TARGA). Se ha propuesto la combinación de TARGA con inmunoterapias para restaurar y/o potenciar las respuestas inmuno-específicas frente al VIH, con el último propósito de controlar la replicación vírica en ausencia de TARGA durante períodos prolongados de tiempo. Los defectos funcionales de las respuestas celulares y humorales, así como, la interrelación entre ellas explicarían la falta de control por parte del sistema inmunológico de la replicación viral. Se han investigado distintos tipos de terapias inmunomediadas para solucionar los problemas antes mencionados, entre ellas la inmunoterapia pasiva, la utilización de citocinas, las interrupciones estructuradas de tratamiento, la utilización de inmunosupresores y las vacunas terapéuticas. Los todavía limitados conocimientos que disponemos acerca de los mecanismos inmunológicos capaces de controlar la replicación viral del VIH y de las causas del deterioro de la inmunidad celular y humoral, han producido modestos beneficios de las terapias inmunomedidas obtenidos hasta la actualidad, y en una escasa o ninguna aplicabilidad en la clínica diaria, quedando, hoy por hoy, confinados al campo de la investigación. La disponibilidad de una óptima vacuna terapéutica sería un gran avance científico, comparable a la llegada de los inhibidores de la proteasa a la clínica, por lo que actualmente debe ser una línea prioritaria de investigación.
Palabras clave:
Immunotherapy
HIV
Therapeutic vaccines
Texto completo

Introduction

The advent of highly active antiretroviral therapy (HAART) has significantly reduced the morbidity and mortality of HIV-1 infection, even in those patients affected by aids-defining conditions1,2. This benefit is obtained due to an increase in the absolute number of circulating naive CD4 T lymphocytes, a concomitant reduction in the number of T lymphocytes with activation markers, and restoration of the response to memory antigens3. Nevertheless, despite the clinical efficacy of HAART4,5, this treatment by itself is unable to eradicate the infection, even if it were administered for more than 60 years6,7. This limitation is mainly because therapy cannot eliminate latent HIV-1 in the form of integrated ≤ proviral DNA, in addition to the existence of low levels of viral replication, which makes possible even cell-to-cell infection8,9. Furthermore, HAART is incapable of restoring the immune-specific response to HIV10,11, and, in fact, leads to a fall in the specific CTL response due to the lack of antigenic exposure12. Recent reports have shown that the helper proliferative response to HIV p24 Ag presented by some HAART patients does not reflect an improvement in the immune phenotype or function of CD4 or CD8 cells, but is secondary to the small increases in viremia typically observed in patients taking HAART13. This would explain the rapid "rebound" of viral load after suspending HAART, in a question of days or weeks, even after several years of effective therapy14,15. This rebound occurs even if HAART is initiated in very early-stage HIV-infected patients, in whom the immune system is theoretically still well preserved (circulating CD4+ T lymphocytes > 500 cells/mL; viral load in plasma (PVL): 5,000-10,000 copies/ml). Similarly, these viral dynamics occur even when immune restoration is practically complete in terms of the homeostasis of T lymphocytes and their subpopulations, and in terms of the capacity for response to polyclonal stimuli and memory antigens with HAART14,16.

These findings reinforce the need for suitable long-term treatment. Resistance, adverse effects in the medium-long term, and cost are important limitations for lifelong adherence to this therapy17. These concerns mean that new therapeutic strategies must be evaluated. The two possibilities being investigated at present are simplification of therapy18 and the combination of HAART with immune therapy to restore and/or boost such immune responses with the primary objective of controlling viral replication in the absence of HAART10. The idea is that HAART-free periods could be longer if we used pre-HAART withdrawal strategies aimed at stimulating the immune system to partially control viral replication after withdrawal10.

Pathogenic basis for the design of immune-mediated strategies

The main question to be answered is whether the immune system can contain viral replication without HAART, even if only for limited periods. This hypothesis arises from the following facts:

1. Although in most infected patients replication leads to the progressive destruction of the immune system and evolves inevitably towards aids, a small number of immunologically "privileged" individuals, or "Long Term Non-Progressors" (LTNP), have a potent and sustained response of anti-HIV-1 CTL, Th cells, and neutralizing HIV-1 antibodies. This is associated with a control of viral replication and the presence of very low or undetectable viral concentrations in plasma in the absence of HAART19.

2. The anti-HIV-1 cytotoxic response (CTL) is detected in all cases studied during the acute phase of the infection, and it is believed to reduce the peak of PVL which characterizes it to the stabilization level, or "setpoint", of PVL, which is established at the end of the acute phase. Direct data on the critical role of the CTL response in the control of viral replication have been obtained both in the infection model with macaques devoid of CD8+ T lymphocytes20,21, and in the immunodeficient murine model22.

3. There is clear evidence that a specific helper T response against HIV is crucial in obtaining an optimal specific CTL response which can control viral replication both in human10,23 and in animal models24. This concept is consistent with other recently reported data on chronic viral infections in murine models25.

4. Studies in primate and murine models show that high levels of neutralizing antibodies can block infection regardless of the route of exposure to the virus26.

Despite the importance of the immune response in infection, it cannot contain viral replication. Alterations of the immune system may allow us to explain this inability or dysfunction. Even though CD4+ and CD8+ cells capable of secreting interferon gamma (IFN-gamma) can be found, in most HIV-infected patients, the proliferative CD4 responses are normally absent11,23,27, and the CD8+ cells are defective with regard to their cytolytic activity28-30. One explanation of these functional deficits of CD4 and CD8 responses would be that the antigen-presenting functions of the dendritic cells could be deteriorated in these patients, and this could contribute to the functional defects observed in the Th1 and CTL cellular responses31,32. The absence of a correct proliferation and expansion of the CD4+ responses may in turn influence the lack of cytolytic activity of the CD8+ cells27,33. In animal models, there is a clear deficit in the secretion of cytokines by CD4+ cells which starts when PVL peaks in primary infection34. Lastly, the selective infection of HIV-specific CD4+ cells in infected patients would explain why these responses are quickly lost in HIV infection35.

Apparently, therefore, cellular and immune responses, and the relationship between them, are vital for a correct functioning of the immune system. The defects would be more from alterations of these responses than from viral escape. Different types of immune-mediated therapies have been examined to solve these problems, including passive immune therapy, cytokines, structured treatment interruptions, immunosuppressors and therapeutic vaccines (table 1). This review will try to summarize the different approaches.

Passive immune therapy

Two types of passive immune therapy have been investigated in HIV-infected patients. The first type is based on infusion, of both CD4+ and CD8+ cells, and the second on plasma or neutralizing antibody infusion.

Passive immune therapy by cell infusion

Several studies on the infusion of specific CTL cells have been performed, although the results to date are not very promising36-38. Brodie et al39,40 investigated the functional activity of HIV-specific CTL cells and the capacity of these effector cells to migrate in vivo to areas of infection. Briefly, these authors expanded HIV-specific autologous CTL gag clones in vivo and injected them into HIV patients. The transferred cells retained their lytic activity in vivo, accumulate in territories close to where HIV-infected cells are found in the lymph nodes and reduce transitorily the circulating levels of HIV-infected CD4+ cells. Apart from expanding and infusing CTL, other groups have tried to infuse expanded CD4+ cells in vitro by a strategy which allows only virus-free cells to be conserved41. After infusion, a moderate improvement is observed in the CD4+ lymphocyte figure with a reduction in the CCR5 co-receptor, which implies a relative reduction in the infective capacity of these cells. In summary, passive therapies based on the transfer of cells are still very experimental, and have provided us with a better knowledge of the immune pathogenesis of the disease, although with no immediate clinical application.

Passive immune therapy by infusion of plasma or neutralizing antibodies

Plasma passive immune therapy (PIT) and monoclonal neutralizing antibodies appeared many years ago (although they are no longer used) for use in daily clinical practice. PIT as therapy for aids patients was investigated during the first half of the 1990s. It was proposed and initiated by Abraham Karpas, a virologist from the University of Cambridge (UK), who published his first results in 1988. This technique involved the intravenous infusion in advanced aids patients of plasma from asymptomatic HIV+ patients. The first study used a monthly infusion of 500 ml of plasma for 3 months in 10 advanced patients (7 with aids and 3 with ARC "aids-related complex", according to the classification criteria from this period). The plasma was previously inactivated with propiolactone to eliminate the infectivity of the donor virus. The results showed that PIT produced negative p24 antigenemia values and increased neutralizing activity with no adverse effects42. Shortly afterwards (1990), Karpas demonstrated the efficacy of the PIT protocol by observing reductions in circulating viral load43. In this second study, Karpas also showed that the monthly plasma infusion increased and maintained a high level of neutralizing activity in serum. The hypothesis which resulted from these studies was that asymptomatic HIV-infected patients had serum containing high titers of viral neutralizing activity, and that this activity disappeared as patients progressed to aids. Karpas concluded that this neutralizing activity was correlated with non-progression to aids and therefore, passive transfer of these antibodies in the plasma of asymptomatic patients would help recipients to control and slow down the progression of their immunodeficiency44. Given that there was no treatment for the infection at this time, these studies aroused great interest in PIT as a possible therapy, with the result that PIT study groups soon began to be formed. Double-blind and placebo-controlled randomized clinical trials were started with a large number of cases led by two independent groups, one in California45 and the other in Paris46. The California trial45 studied 220 aids patients with CD4+ T cell counts of between 50 and 200 cells/microL, which were randomized into three groups to receive over 12 months a monthly infusion of (i) 500 ml of hyperimmune plasma; (ii) half the dose of plasma (250 ml of plasma diluted to 500 ml with 5% human seroalbumen), and (iii) 500 ml of 5% seroalbumen, as a placebo. Mortality fell and the number of CD4 cells grew compared with the placebo group, and with the group which received half the plasma dose. The French trial included 86 aids patients who were randomized to two groups to receive over 12 months a fortnightly infusion of: (i) 300 ml of hyperimmune plasma, and (ii) 300 ml of plasma from healthy donors (non-immune) as placebo. The group treated showed a smaller incidence of aids-defining events (p = 0.009), a smaller accumulation of these events (3 times smaller), and a lower mortality rate (p = 0.009). The conclusion of both studies was clear, in the sense that the infusion of plasma from non-advanced patients to advanced patients had no adverse effects and was clinically useful in "curbing" progression of the disease. Later, systematic and controlled PIT clinical trials were practically abandoned, given the efficacy of the antiretroviral agents which appeared.

There has been renewed interest in passive immune therapy, but with specific monoclonal antibodies, based on studies of the macaque model, which show that passive transfer of antibodies prevents infection by oral, vaginal or intravenous inoculation of the virus47-50. This current interest in antibodies as a potential therapy or prophylaxis is beginning to be seen in human clinical care. Phase I trials have begun in infected patients to evaluate the pharmacokinetics and safety of human monoclonal antibodies (known as 2F5 and 2G12), which were taken some years back from two non-progressors. Unlike other human monoclonal antibodies, these can inhibit in vitro infection by R5 strains and X4 strains51. A recent study has shown that the administration of a neutralizing antibody called TNX-355 produces some antiviral efficacy (a fall of 0.5-1 log10) and an increase in the number of CD4+ lymphocytes. This effect persisted in patients for up to four weeks after infusion of the antibody52. If the efficacy of these monoclonal antibodies is confirmed in humans, in vitro and experimental animal models, they may soon be clinically useful, although for now they are only a promising possibility.

Cytokines

Several studies and clinical trials have used cytokines (table 1), all with the aim of restoring the cytokine imbalance caused by HIV infection, and suitably controlling infection using neutralizing antibodies, especially that caused by specific CTL cells. The best options for human medicine are IL-2, IL-12, IL-15, growth hormone and GM-CSF.

IL-2 infusion with different strategies, doses and routes leads to a clear increase in the CD4+ lymphocyte count53,54. The most widely recommended dose at present is 4.5 M IU/q 12 h x 5 days. In general, an induction phase is with 6 X 5-day cycles every 8 weeks, followed by a maintenance phase with a number of variable cycles if there is a new fall in the CD4+ count. Toxicity is dose-dependent with a frequency of grade 3-4 adverse effects in < 10% of cases. The most common local adverse effects are nodules and blisters at the injection site. The most common systemic adverse events are pseudoflu syndrome (90% of patients), skin lesions (60%), gastrointestinal disorders, edema, disorders of the central nervous, respiratory and endocrine systems. Other uncommon effects (< 10%) include cytopenia, electrolytic alterations, and cardiovascular disorders (arrhythmias, congestive cardiac insufficiency, ischemic cardiopathy, hypotension). It is the most widely studied and clinically advanced drug used in immune therapy. Nevertheless, after years of research, it remains unclear whether the increase in CD4+ T cells affects clinical progression positively, although there are studies in progress which try to answer these questions.

Apart from increasing total CD4+ lymphocytes, IL-2 has been used with at least three other objectives:

1. The first is as a cytosine which tries to restore the T cell repertoire by increasing the total CD4+ lymphocyte count. Progression of HIV infection is known to be associated with a more rapid loss of naïve cells than of memory cells. Immune control of viral infections depends on the immunocompetent cells having a wide repertoire, and HIV infection leads to the loss of important parts of this repertoire. Administration of IL-2 is associated with polyclonal increases both of naive cells and of memory cells in HIV-infected patients, but analysis of repertoire has shown that defects are not corrected by the administration of IL-255.

2. Therefore, the combination of IL-2 and other immune-mediated therapies has been proposed to restore the dysfunction of the helper response (perhaps due to a lack of sufficient endogenous IL-2) in HIV-infected patients. Nevertheless, several pilot clinical trials have failed to show the usefulness of IL-2, at least when it is combined with structured treatment interruptions56,57, or a canarypox vaccine (ALVAC-HIV vCP 1433)58.

3. Some years ago, an attempt was made to eliminate the virus from the reservoirs by stimulating the IL-2 of quiescent HIV-infected cells which, when stimulated, produce viruses, which would be inactivated by HAART. In one clinical trial, patients who received HAART and IL-2 showed a lower quantity of detectable infectious viruses than the control group, which only received HAART59. Nevertheless, on withdrawing therapy in both groups, the viral rebound displayed similar dynamics, which would suggest that IL-2 had little effect on the viral reservoir60.

Other cytokines have been proposed in human medicine (table 1). The most important are IL-12 and IL-15, which lead to an increase in the specific CTL response in vitro61-63. Both are produced mainly in activated antigen-presenting cells and are thought to promote the development of TH-1 type cellular responses. This type of response is essential for stimulating CTL responses. Other effects of these cytokines are the increase in lytic activity by the natural killer cells and the increase in the HIV-specific proliferative capacity62-64. Petrovas et al65 recently reported that IL-15, administered twice a week for four weeks to SIV-infected cynomulgus macaques, increased the proliferation and expansion of CD8+ cells without affecting viral replication.

The growth hormone (GH) has been suggested in clinical practice for HIV-infected patients to promote the T cell response and generate a lymphopoietic effect and induce effects in peripheral T cells. Therefore, it seems to be an appropriate immunomodulating agent in states of immunodeficiency. In HIV+ patients treated with GH a marked increase in thymic mass has also been observed. This increase was accompanied by a significant increase in the number of naïve CD4 T cells, an increase in the HIV-specific CD4 and CD8 responses, and induction in the differentiation of T cells from functional memory effector cells66,67. These results suggest that GH produces an increase in thymopoiesis and therefore that it has important effects on the human immune system, including reversion of thymic atrophy in HIV-1-infected adults.

Finally, GM-CSF has been combined with treatment interruptions, and a slight improvement in virological control and a smaller fall in CD4+ lymphocytes after definitive interruption of treatment have been observed. Nevertheless, the results are currently unacceptable in terms of toxicity, given that more than 80% of patients had local and general reactions68.

Structured treatment interruption (STI)

Since the description of the anecdotal cases presented by Franco Lori and Douglas Nixon69,70, the concept of antiretroviral therapy interruption as a therapeutic strategy has been investigated with interest by several groups. Initially, this strategy was considered as "autovaccination" with an attenuated autologous virus, in which the attenuation came from the gradual reintroduction of antiretroviral therapy. With time, other objectives of STI, which were not important initially (e.g. savings in medication, reduction of secondary effects, etc.), have come to the fore and are currently among the most widely investigated strategies71. Nevertheless, it must be stressed that one of the most important concerns in the application of STI in HIV+ patients receiving treatment is the risk of selecting resistance72. This review will examine neither the latter type of interruptions nor the so-called "therapy vacations" used in patients experiencing therapeutic failure73, rather we shall concentrate on STI as an immune-mediated strategy.

Several studies show that STI in patients who started antiretroviral therapy during the acute phase of HIV infection allow viral replication to be controlled transitorily, and stress the intrinsic potential of the immune system to adequately control the disease74. However, these data have not been confirmed by other groups56, and it has been observed that the virological efficacy of STI started during acute infection is lost over time, therefore more research is necessary into whether starting therapy (with or without STI) during the acute phase is beneficial for long-term patients75.

Re-exposure to viral antigens boosts and stimulates virus-specific immune responses, although only 20% of chronic patients who use this strategy manage to effectively control viral replication in the short-medium term76-82. It is important to understand why there is a lack of control of viral replication despite the induction of CTL and helper responses in chronic HIV-infected patients. These conclusions serve to design other immune-mediated strategies which allow more effective control of viral replication for a longer period.

1. First, during the interruption, we can observe very high peaks of viral load in some patients and, given that the CD4+ cells with an HIV-specific response are more infected after viral rebound35,83, clonal elimination of these cells could occur, which may explain the lack of response84,85. Plana et al recently studied a group of 40 patients with intermittent therapy. In these patients, the helper response was shown to be induced weakly during the interruption cycles, and it is lost during definitive interruption of therapy86. This would explain the inability of the CTL response to control viral replication87-89. Contrary to the helper response, the CTL response is induced considerably (both in magnitude and in amplitude) after definitive interruption of antiretroviral therapy77,78, but it is incapable of controlling viral replication. Some authors report that CTLs induced after discontinuation of therapy would not be functional (they would be in a pre-terminal stage and would produce fewer perforins), and they attribute the inability of these strategies to stimulate an efficacious CTL response to the loss of a specific T helper response90. Strategies aimed at avoiding clonal detection of T cells with the capacity of an HIV-specific response caused by STI, could be predicted to improve the control of viral replication by inducing a functional CTL specific response.

2. Second, many authors have contributed data from sequencing and cloning of the env gene. They suggest that, in a viral rebound, the virus which appears may be very different from that observed in other rebounds or that which appears at baseline91-93, and that the specific immunity recovered is no more than an increase in the existing immunity77,94, probably due to the fact that they are repeatedly exposed to the same antigen95. This mobilization of viruses from different reservoirs, and the inability to create a new specific response may be what prevents efficacious control of viral replication. Therefore, we should use immunogens capable of inducing new efficacious HIV-specific responses, both functionally, since this would prevent the clonal disappearance of helper T cells and improve the CTL response, and in amplitude, so that they could control replication of ancestral viruses present in memory cells, which would finish by replicating once HAART was interrupted.

Immunosuppressors

Parallel to the fall in the CD4 count, HIV infection is characterized by an intense and sustained state of immune activation manifested by a high number of T and B lymphocytes, natural-killer cells, and a marked release of pro-inflammatory cytokines such as IL-7 and TNF-α10. A constant element of this process is the high count of activated T-CD8 clones which express surface receptors, DR+/CD38+, a phenomenon which is today considered as a true marker of disease progression96,97. This sustained activation process can lead to an exhaustion of the immune system, similar to an increase in cellular infectivity, thus allowing dissemination. This phenomenon of immune activation lays the foundations for the use of immunosuppressors such as corticoids, hydroxyurea (HU), mycophenolate mophetil (MPM), thalidomide and cyclosporine A, as adjuvants to antiretrovirals.

The results obtained on the control of viremia in macaques, treated since acute infection by SIV, using HU as an adjuvant and treatment interruption cycles, lead us to consider this drug as a clinically useful immunuosuppressor in the future98. We can, therefore, formulate the hypothesis that HU inhibits activation of T lymphocytes during interruption cycles, thus preventing infection in the target cells and the production of high peaks of viral replication without destroying the specific immune response. Although it is well known and clinically proven that HU inhibits the ribonucleotide-reductase enzyme98,99, it also induces a cytostatic effect by halting the cellular cycle at the beginning of the S phase, and a reduction in cellular activity. It was this property that led García et al78 to evaluate the usefulness of the drug in patients who were programmed to undergo intermittent interruptions of HAART. Five interruption cycles of 2 weeks' duration were scheduled, but continuing with HU only for the last two cycles of interruption. This schedule made it possible to evaluate the effect of HU on viral dynamics between interruption cycles with and without the drug. Thus, if HU manages to reduce the initial phase of viral rebound, which starts from the reservoirs, resting lymphocytes, macrophages, and dendritic cells, where it has been shown that HU is excellent as monotherapy100, the effect of HU was observed even when this ceased to be administered. Secondly, HU can slow down later phases of viral replication from activated T lymphocytes, mainly due to its cytostatic effects. In this case, the control of viral replication is only obtained by maintaining HU when HAART is withdrawn. There were no differences in viral rebound after three interruption cycles, although when HU was maintained after interruption, the viral load was on average 1 log lower than that obtained during previous interruption cycles and lower than in the control group, treated with HAART only. This phenomenon shows the cytostatic activity of HU and its potential use against the intracellular viral reservoir. From a clinical viewpoint, the use of HU significantly increased the number of patients who achieved sustained viral replication (8/9 patients with viral load < 5,000 copies RNA/mL) for 48 weeks after 5 HAART interruption cycles, regardless of baseline viral load (4.6 log RNA HIV).

Other groups have studied the capacity of other immunosuppressors such as mycophenolic acid as an adjuvant to HAART. Chapuis et al101 studied in vitro and in vivo the mechanisms by which mycophenolic acid (MPA) and its sterilic derivative mycophenolate mophetil (MPM) suppressed infection by HIV. MPA selectively inhibits the synthesis of guanosine nucleotides by competitively inhibiting the dehydrogenase inosin-monophosphate enzyme. Given that there are no alternative enzymatic pathways for the synthesis of guanosine nucleotides in lymphocytes, MPA produces a profound cytostatic effect by depletion of this substrate. Furthermore, in vitro results show that MPA inhibits the proliferation of activated T cells, especially in those with low or intermediate expression of the CD4 receptor, by leading them to apoptosis even in the presence of IL-2. These data were confirmed in a clinical trial involving patients treated with abacavir and amprenavir, who were randomized to receive or not receive MMF101. In the MMF group, a reduction in the actively dividing CD4 and CD8 "pool" (Ki67+) was observed. Furthermore, the authors suggest that MMF can have an effect on the "pool" of latently infected CD4 cells, as they observed that, in patients treated with MMF, the ability to isolate viruses from the total population of T-CD4 was reduced. Even though MMF does not affect resting cells and therefore does not affect their number, once they have been activated in the presence of MMF, apoptosis and cell death are induced101. Other authors have investigated the role of MMF on viral load in plasma and in lymph tissue during and after intermittent interruptions of HAART102. Patients treated for at least one year with an abacavir-containing regimen were randomly assigned to receive or not receive MMF with HAART for four months before the interruption cycles. In those treated with MMF, the "pool" of dividing T-CD4 cells was reduced. The same occurred with the set-point of viral load after interrupting HAART. In this trial, lymphoproliferation was used to evaluate the capacity of serum in patients to reduce the response of a T cell line in vitro, using sequential samples at different time points after the dose of MMF. The changes obtained in the dynamics of viral load, especially those observed in the set-point of PVL after HAART interruption, were only observed in those patients who reduced lymphocyte proliferation to below 40% in the T cell line (CEM) for more than four hours after administration of MMF. In another context, MMF was used as part of a rescue schedule when added as an isolated drug to a HAART rescue regimen containing abacavir in patients with multiple resistance to antiretrovirals103. A significant reduction in viral load (> 0.5 log) was observed in those patients who increased their quotient: Carbovir (the antiviral active metabolite of abacavir) and deoxyguanosine triphosphate, owing to inhibition of the dehydrogenase ionosine monophosphate enzyme and subsequent depletion of guanosine nucleotides.

New therapeutic alternatives were proposed after the publication of the results of a pilot study in patients with acute HIV infection treated with short-term cyclosporine A (CyA) and HAART104. This trial involved reducing the high level of cellular activation which thus generated massive viral replication. This high level viral replication led to clonal exhaustion of HIV-specific CD4 lymphocytes. Despite the fact that CyA interferes with the synthesis of the viral gag proteins, its main effect is by inhibition of proliferation and differentiation of T cells. The CD4 count of patients treated with CyA was restored both in percentage terms and in absolute numbers, by maintaining the number of secreting CD4-HIV-?IFN. These data suggest that the use of CyA or any other drug with immunosuppressor properties during primary infection could reduce the number of active CD4 cells which sustain massive viral replication, and prevent the hijacking of these clones in lymph tissue, where antigens are presented and HIV infection is perpetuated. This process may have an impact on the clone of the resting T cells which shelter viruses with replicative capacity. Nevertheless, we do not know whether establishing a new immunological set-point to slow down the rate of progression offers a clinical benefit in long-term infection.

As several studies have shown, there are many doubts about the use of immunosuppressors as immune-mediated therapy in HIV infection. Knowing which patients are most suited, which drug to use, how long therapy should last and the ideal time to start therapy are questions which must be answered by clinical trials with a large number of patients and long-term follow-up. There are also concerns over long-term safety, such as the influence on the development of opportunistic infections or lymphoproliferative diseases, which limit extensive use of this type of drugs in the long term. Nevertheless, pharmacological strategies which interfere with the HIV life cycle by acting at the level of viral target cells, rather than by inhibiting viral enzymes, are still attractive. This has the advantage of avoiding the development of genomic mutations against antiviral drugs. The use of immunosuppressor drugs as an adjuvant to HAART must be evaluated with caution until we have more information on long-term efficacy and safety.

Therapeutic vaccines

Immune recovery of the HIV response has also been tried using therapeutic vaccination. In general, the capacity of the vaccines used to increase the CTL-specific response has been very limited and study results have been discouraging, as immunogenicity has not been demonstrated and there has been no clear impact on viral load105-109.

The Remune vaccine has received most attention. This is a vaccine of an inactivated complete vaccine in which the envelope protein has been removed during the process of inactivation which is carried out for synthesis. This vaccine stems from a virus originally obtained in Zaire and contains a type-A envelope and type-G gag. It has been administered to more than 3,000 people with an antiviral-controlled virus. The results showed that it was capable of inducing gag-specific helper responses which are sometimes very potent. Nevertheless, these studies did not observe a capacity for immunological control of viral replication110-112. The study which best demonstrates the capacity of a therapeutic vaccine to efficaciously increase specific immunity for the control of viral replication used dendritic cell (DC) vaccine in the SIV animal infection model (SIVmac251). In this study, four immunizations with cells pulsed by the same virus were made every two weeks for a period of eight weeks. In most of the inoculated animals (7/10), there was a significant reduction in viral load in plasma after the third immunization. This was sustained for the 34 weeks of the study. There was a 50 and 1,000-fold reduction in the SIV DNA and SIV RNA loads in peripheral blood, respectively. The analysis of lymph tissue revealed a correlation between the reduction in SIV DNA and RNA levels and the increase in the SIV-specific T cell response. The fall in viral load and subsequent increase in CD4 count was accompanied by an increase in circulating antibody levels113. Very similar results were obtained by two independent groups in a murine model with preventive vaccination114,115. Despite these incredible results, a clinical trial involving 12 patients with chronic infection receiving antiretroviral therapy from early stages of the infection, using a dendritic cell vaccine pulsed with heat-inactivated autologous viruses has offered much more moderate results116. In this study, in a first treatment interruption 18 months before receiving the first dose of vaccine, three plasmaphereses were performed in which 1800 cc of plasma was extracted. The median viral load of patients during plasmapheresis was 27,000 copies/ml. The virus was then inactivated by heat and concentrated using ultracentrifuge in 1 cc, all under conditions of good clinical practice (GCP). A schedule of five subcutaneous doses was administered every six weeks. The first dose was a control with non-pulsed DC. Every pulsed vaccine contained 2 x 106 DC, pulsed with five virions per cell during the first immunization and three virions/DC in the remaining vaccinations. In general, the results showed that a vaccine did not cause important adverse events, as in only 3 of the 60 doses administered (5%) was there an adverse reaction (only 1 mild local reaction and 2 episodes of flu symptoms 24 hours after the dose). This vaccine was able to control viral replication partially and transitorily, and was associated with a transitory, but not significant increase in the lymphoproliferative response to HIV P24 Ag, and with the changes in the CTL-specific response for peripheral HIV and in the CTL cells of lymph tissue. In lymph tissue, there was also a trend towards greater control of viral replication associated with an increase in CD4 and CTL cells in this tissue116. Furthermore, there was no significant increase in the neutralizing activity of the serum of these patients. Despite these moderate results, we must remember that the dose of antigen used in the human trial was 1,000 times lower that that used in macaques, therefore, new trials with a greater dose of antigen are necessary. If these results are confirmed, they would suggest that the defect in immunological control of HIV could be due to alterations in the induction phase of the immune response, which is consistent with recent studies on the induction of the immune response in the absence of a helper response117,118, and with data which suggest that antigen-presenting functions are altered in HIV-infected patients. This could contribute to functional defects in HIV-specific CTL and helper responses31,32,119.

Other vaccine trials have examined ALVAC, whose vector is a recombinant canarypox. Kinloch et al120 recently presented long-awaited results from the QUEST study. This international study was carried out on patients who started treatment during the acute phase. After a mean of two years of virological control, 79 were randomized to receive immunization with ALVAC VcP1452, ALVAC plus Remune, or placebo. After 24 weeks' immunization, HAART was interrupted. There was no difference between the groups in terms of viral rebound dynamics or in viral load figures.

Another therapeutic vaccination study has recently been presented by Cooper et al using patients with primary infection121. After a mean of four years of HAART, 35 patients with controlled viral replication were randomized to be vaccinated with a fowlpox vector free of HIV sequences, a vector containing gag/pol sequences, or a vector containing gag/pol sequences and a gene which encodes human interferon gamma. Surprisingly, there were few differences between the groups in terms of presence of CDL cells measured by ELISPOT or in cytolytic responses after vaccination and before interruption of treatment. Treatment was not interrupted in 10 patients. There were no differences in the control of viral replication between the placebo group and the group vaccinated with gag/pol. However, patients immunized with gag/pol and interferon gamma had better control of viral replication, with a mean viral load of 0.8 log10 less than the other two groups. The absence of immune responses in the two vaccinated groups is disappointing and the response in the interferon group is surprising.

Other vaccines with the potential to be used in therapy are those based on DNA which includes the proteins env-tat-nef. These have been tried as a preventive vaccine with promising results by inducing a primary response with DNA and a booster with the Ankara virus122. The vaccine tried as therapy presents the whole HIV genome minus the integrase gene and has shown promising results after intradermic administration in monkey models with STI123.

Conclusions

We have a limited knowledge of the immunological control of HIV viral replication, the causes of cellular and humoral immune deterioration, and a lack of clear immunological methods to correlate with an efficacious immune control of HIV in vivo. The efficacy of immune therapy and therapeutic vaccines has been modest in the best of cases. We must redouble our efforts to understand better the mechanisms of protection, virological control and immune deterioration. Without this knowledge, an efficacious therapeutic vaccine is a long way off. Nevertheless, given the toxicity and long-term efficacy problems with current drugs, this remains a priority line of investigation.

Acknowledgments

Some of the studies and data have received support from: Ministry of Health, Abbott Laboratories, Boehringer Ingelheim, Bristol Myers Squibb, GlaxoSmithKline, Merck Sharp and Dohme and Roche (FIPSE) 01/36259, SAF 01/2591, Red Temática Cooperativa de Grupos de Investigación en Sida (RIS) del Fondo de Investigación Sanitaria (FIS), marató de TV3, Objectif recherche vaccin sida (ORVACS). We are grateful to Dr. Teresa Gallart, Dr. Emilio Fumero and Dr. Margarita Bofill for their comments and help in preparing this manuscript.

Bibliography
[1]
Palella FJ, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, et al..
Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection..
N Engl J Med, 338 (1998), pp. 853-60
[2]
Palella FJ, Deloria-Knoll M, Chmiel J, Moorman AC, Wood K, Greenberg AE..
Survival Benefit of Initiating Antiretroviral Therapy in HIV-Infected Persons in Different CD4+ Cell Strata..
Ann Intern Med, 138 (2003), pp. 620-6
[3]
Autran B, Carcelain G, Li TS, Blanc C, Mathez D, Tubiana R, et al..
Positive effects of combined antiretroviral therapy on CD4+ T cell homeostasis and function in advanced HIV disease..
Science, 277 (1997), pp. 112-6
[4]
Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho DD..
HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span and viral generation time..
Science, 271 (1996), pp. 1582-6
[5]
Perelson AS, Essunger P, Cao Y..
Decay characteristics of HIV-1 infected compartments during combination therapy..
Nature, 387 (1997), pp. 188-91
[6]
Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, et al..
Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy..
Nat Med, 5 (1999), pp. 512-7
[7]
The challenge of viral reservoirs in HIV-1 infection. Annu Rev Med. 2002;2002;53:557-93.
[8]
Zhang L, Ramratnam B, Tenner-Racz K, He Y, Vesanen M, Lewin S, et al..
Quantifying residual HIV-1 replication in patients receiving combination antiretroviral therapy..
N Engl J Med, 340 (1999), pp. 1605-13
[9]
Furtado MR, Callaway DS, Phair JP, Kunstman KJ, Stanton JL, Macken CA, et al..
Persistence of HIV-1 transcription in peripheral-blood mononuclear cells in patients receiving potent antiretroviral therapy..
N Engl J Med, 340 (1999), pp. 1614-22
[10]
Letvin NL, Walker BD..
Immunopathogenesis and immunotherapy in AIDS virus infections..
Nat Med, 9 (2003), pp. 861-6
[11]
Plana M, García F, Gallart MT, Miró JM, Gatell JM..
Lack of T-cell proliferative response to HIV-1 antigens after one year of HAART in very early HIV-1 disease..
Lancet, 352 (1998), pp. 1194-5
[12]
Gamberg J, Barret L, Bowmer M, Howley C, Grant M..
Factors related to loss of HIV-specific cytotoxic T lymphocyte activity..
AIDS, 18 (2004), pp. 597-604
[13]
Lange C, Xu Z, Patterson BK, Medvik K, Harnisch B, Asaad R, et al..
Proliferation responses to HIVp24 during antiretroviral therapy do not reflect improved immune phenotype or function..
AIDS, 18 (2004), pp. 605-13
[14]
García F, Plana M, Vidal C, Cruceta A, O'Brien WA, Pantaleo G, et al..
Dynamics of viral load rebound and immunological changes after stopping effective antiretroviral therapy..
AIDS, 13 (1999), pp. F79-F86
[15]
Ruiz L, Martínez-Picado J, Romeu J..
Structured treatment interruption in chronically HIV-1 infected patients after long-term viral suppression..
AIDS, 14 (2000), pp. 397-403
[16]
Plana M, García F, Gallart MT, Tortajada C, Soriano A, Palou E, et al..
Immunological benefits of antiretroviral therapy in very early stages of asymtomatic chronic HIV-1 infection..
AIDS, 14 (2000), pp. 1921-33
[17]
Martínez E, Mocroft A, García-Viejo M, Pérez-Cuevas J, Blanco J, Mallolas J, et al..
A prospective cohort study on the risk for lipodystrophy in HIV-1 infected patients treated with protease inhibitor-containing regimens..
[18]
Martinez E, Arnaiz J, Podzamcer D, Dalmau D, Ribera E, Domingo P, et al..
Substitution of Nevirapine, Efavirenz, or Abacavir for Protease Inhibitors in Patients with Human Immunodeficiency Virus Infection..
N Engl J Med, 349 (2003), pp. 1036-46
[19]
Rosenberg ES, Billingsley JM, Caliendo AM, Boswell SL, Sax PE, Kalams SA, et al..
Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia [see comments]..
Science, 278 (1997), pp. 1447-50
[20]
Schmitz JE, Kuroda MJ, Santra S..
Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes..
Science, 283 (1999), pp. 857-60
[21]
Metzner K, Jin X, Gettie A, Bauer DE, Di Mascio M, et al..
Effects of in vivo CD8(+) T cell depletion on virus replication in rhesus macaques immunized with a live, attenuated simian immunodeficiency virus vaccine..
J Exp Med, 191 (2000), pp. 1921-31
[22]
De Quiros JC, Shupert WL, McNeil AC, Gea-Banacloche JC, Flanigan M, Savage A, et al..
Resistance to replication of human immunodeficiency virus challenge in SCID-Hu mice engrafted with peripheral blood mononuclear cells of nonprogressors is mediated by CD8(+) T cells and associated with a proliferative response to p24 antigen..
J Virol, 74 (2000), pp. 2023-8
[23]
Day C, Walker BD..
Progress in defining CD4 helper cell responses in chronic viral infections..
J Exp Med, 198 (2003), pp. 1773-7
[24]
Hel Z, Nacsa J, Tryniszewska E, Tsai WP, Parks RW, Montefiori DC, et al..
Containment of simian immunodeficiency virus infection in vaccinated macaques: correlation with the magnitude of virus-specific pre- and postchallenge CD4+ and CD8+ T cell responses..
J Immunol, 169 (2002), pp. 4778-87
[25]
Kaech SM, Ahmed R..
Immunology. CD8 T cells remember with a little help..
Science, 300 (2003), pp. 263-5
[26]
Nishimura Y, Igarashi T, Haigwood NL, Sadjadpour R, Donau OK, Buckler C, et al..
Transfer of neutralizing IgG to macaques 6 h but not 24 h after SHIV infection confers sterilizing protection: implications for HIV-1 vaccine development..
Proc Natl Acad Sci U S A, 100 (2003), pp. 15131-6
[27]
Pitcher C, Quittner C, Peterson D, Connors M, Koup RA, Maino VC, et al..
HIV-1 specific CD4+ T cells are detectable in most individuals with active HIV-1 infection, but decline with prolonged viral suppression..
Nat Med, 5 (1999), pp. 518-25
[28]
Yokomaku Y, Miura H, Tomiyama H, Kawana-Tachikawa A, Takiguchi M, Kojima A, et al..
Impaired processing and presentation of CTL epitopes are major escape mechanisms from CTL immune pressure in Human Immunodeficiency Virus type 1 infection..
J Virol, 78 (2004), pp. 1324-32
[29]
Seder R, Ahmed R..
Similarities and differences in CD4+ and CD8+ effector and memory T cell generation..
Nat Immunol, 4 (2003), pp. 835-42
[30]
Yang O, Sarkis P, Trocha A, Kalams SA, Johnson RP, Walker BD..
Impacts of avidity and specificity on the antiviral efficiency of HIV-1-specific CTL..
J Immunol, 171 (2003), pp. 3718-24
[31]
Donaghy H, Gazzard B, Gotch F, Patterson S..
Dysfunction and infection of freshly isolated blood myeloid and plasmacytoid dendritic cells in patients infected with HIV-1..
Blood, 101 (2003), pp. 4505-11
[32]
Chehimi J, Campbell DE, Azzoni L, Bacheller D, Papasavvas E, Jerandi G, et al..
Persistent decreases in blood plasmacytoid dendritic cell number and function despite effective highly active antiretroviral therapy and increased blood myeloid dendritic cells in HIV-infected individuals..
J Immunol, 168 (2002), pp. 4796-801
[33]
McNeil AC, Shupert WL, Iyasere CA, Hallahan CW, Mican JA, Davey RT Jr, et al..
High-level HIV-1 viremia suppresses viral antigen-specific CD4(+) T cell proliferation..
Proc Natl Acad Sci U S A, 98 (2001), pp. 13878-83
[34]
McKay PF, Barouch DH, Schmitz JE, Veazey RS, Gorgone DA, Lifton MA, et al..
Global dysfunction of CD4 T-lymphocyte cytokine expression in simian-human immunodeficiency virus/SIV-infected monkeys is prevented by vaccination..
J Virol, 77 (2003), pp. 4695-702
[35]
Douek DC, Brenchley J, Betts M, Ambrozak DR, Hill BJ, Okamoto Y, et al..
HIV preferentially infects HIV-specific CD4+ T cells..
Nature, 417 (2002), pp. 95-8
[36]
Koenig S, Conley AJ, Brewah YA, Jones GM, Leath S, Boots LJ, et al..
Transfer of HIV-1-specific cytotoxic T lymphocytes to an AIDS patient leads to selection for mutant HIV variants and subsequent disease progression..
Nat Med, 1 (1995), pp. 330-6
[37]
Lieberman J, Fabry JA, Shankar P, Beckett L, Skolnik PR..
Ex vivo expansion of HIV type 1-specific cytolytic T cells from HIV type 1-seropositive subjects..
AIDS Res Hum Retroviruses, 11 (1995), pp. 257-71
[38]
Tan R, Xu X, Ogg GS, Hansasuta P, Dong T, Rostron T, et al..
Rapid death of adoptively transferred T cells in acquired immunodeficiency syndrome..
Blood, 93 (1999), pp. 1506-10
[39]
Brodie SJ, Lewinsohn DA, Patterson BK, Jiyamapa D, Krieger J, Corey L, et al..
In vivo migration and function of transferred HIV-1-specific cytotoxic T cells [see comments]..
Nat Med, 5 (1999), pp. 34-41
[40]
Brodie SJ, Patterson BK, Lewinsohn DA, Diem K, Spach D, Greenberg PD, et al..
HIV-specific cytotoxic T lymphocytes traffic to lymph nodes and localize at sites of HIV replication and cell death..
J Clin Invest, 105 (2000), pp. 1407-17
[41]
Levine BL, Bernstein WB, Aronson NE, Schlienger K, Cotte J, Perfetto S, et al..
Adoptive transfer of costimulated CD4+ T cells induces expansion of peripheral T cells and decreased CCR5 expression in HIV infection..
Nat Med, 8 (2002), pp. 47-53
[42]
Karpas A, Hill F, Youle M, Cullen V, Gray J, Byron N, et al..
Effects of passive immunization in patients with the acquired immunodeficiency syndrome-related complex and acquired immunodeficiency syndrome..
Proc Natl Acad Sci U S A, 85 (1988), pp. 9234-7
[43]
Karpas A, Hewlett IK, Hill F, Gray J, Byron N, Gilgen D, et al..
Polymerase chain reaction evidence for human immunodeficiency virus 1 neutralization by passive immunization in patients with AIDS and AIDS-related complex..
Proc Natl Acad Sci U S A, 87 (1990), pp. 7613-7
[44]
Karpas A, Gillson W, Bevan PC, Oates JK..
Lytic infection by British AIDS virus and development of rapid cell test for antiviral antibodies..
Lancet, 2 (1985), pp. 695-7
[45]
Levy J, Youvan T, Lee ML..
Passive hyperimmune plasma therapy in the treatment of acquired immunodeficiency syndrome: results of a 12-month multicenter double-blind controlled trial..
Blood, 84 (1994), pp. 2130-5
[46]
Vittecoq D, Chevret S, Morand-Joubert L, Heshmati F, Audat F, Bary M, et al..
Passive immunotherapy in AIDS: a double-blind randomized study based on transfusions of plasma rich in anti-human immunodeficiency virus 1 antibodies vs transfusions of seronegative plasma..
Proc Natl Acad Sci U S A, 1994 (1995), pp. 1195-9
[47]
Parren PW, Marx PA, Hessell AJ, Luckay A, Harouse J, Cheng-Mayer C, et al..
Antibody protects macaques against vaginal challenge with a pathogenic R5 simian/human immunodeficiency virus at serum levels giving complete neutralization in vitro..
J Virol, 75 (2001), pp. 8340-7
[48]
Hofmann-Lehmann R, Vlasak J, Rasmussen RA, Smith BA, Baba TW, Liska V, et al..
Postnatal passive immunization of neonatal macaques with a triple combination of human monoclonal antibodies against oral simian-human immunodeficiency virus challenge..
[49]
Hofmann-Lehmann R, Rasmussen RA, Vlasak J, Smith BA, Baba TW, Liska V, et al..
transmission? J Med Primatol, 30 (2001), pp. 190-6
[50]
Mascola JR..
Passive transfer studies to elucidate the role of antibody-mediated protection against HIV-1..
Vaccine, 20 (2002), pp. 1922-5
[51]
Armbruster C, Stiegler GM, Vcelar BA, Jager W, Michael NL, Vetter N, et al..
A phase I trial with two human monoclonal antibodies (hMAb 2F5, 2G12) against HIV-1..
AIDS, 16 (2002), pp. 227-33
[52]
Kuritzkes DR, Jacobson J, Powderly WG, Godofsky E, De Jesús E, Haas F, et al..
Antiretroviral activity of the anti-CD4 monoclonal antibody TNX-355 in patients infected with HIV type 1..
J Infect Dis, 189 (2004), pp. 286-91
[53]
Kovacs JA, Vogel S, Albert JM, Falloon J, Davey RT.J, Walker RE, et al..
Controlled trial of interleukin-2 infusions in patients infected with the human immunodeficiency virus [see comments]..
N Engl J Med, 335 (1996), pp. 1350-6
[54]
Arno A, Ruiz L, Juan M, Jou A, Balagué M, Zayat MK, et al..
Efficacy of low-dose subcutaneous interleukin-2 to treat advanced human immunodeficiency virus type 1 in persons with < 250 microL CD4 T cells and undetectable plasma virus load..
J Infect Dis, 180 (1999), pp. 56-60
[55]
Connors M, Kovacs JA, Krevat S, Gea-Banacloche JC, Sneller MC, Flanigan M, et al..
HIV infection induces changes in CD4+ T-cell phenotype and depletions within the CD4+ T-cell repertoire that are not immediately restored by antiviral or immune-based therapies..
Nat Med, 3 (1997), pp. 533-40
[56]
Low-Dose Daily IL-2 combined with structured treatment interruptions (STI) did not increase the HIV-1-Specific T-cell responses in patients receiving HAART within 90 days after onset primary HIV-1 Infection (PHI) Symptoms. XV International AIDS Conference; Bangkok Thailand, 11-16 July, 2004.
[57]
Interleukin-2 prior to stopping effective antiretroviral therapy prolongs time off treatment: initial results of a pilot study utilizing CD4+ T-cell count < 350 cells/mm3 as the threshold for restarting ART (ACTG A5102). Abstract 510. 11st Conference on Retroviruses and Opportunistic Infections; San Francisco, February, 8-11, 2004.
[58]
Immunological and Virological Efficacy of ALVAC-VIH 1433 and HIV Lipopeptides (Lipo-6T) Combined with SC IL-2 in Chronically HIV-infected Patients-Results of the ANRS 093 Randomized Study. Abstract 62. 10th conference on retroviruses and opportunistic infections, Boston, MA. February 2003. p. 10-14.
[59]
Chun TW, Engel D, Mizell SB, Hallahan CW, Fischette M, Park S, et al..
Effect of interleukin-2 on the pool of latently infected resting CD4+ cells in HIV-1-infected patients receiving highly active anti-retroviral therapy..
Nat Med, 5 (1999), pp. 651-5
[60]
Chun TW, Davey RT.J, Ostrowski MA, Justement SJ, Engel D, Mullins JI, et al..
Relationship between pre-existing viral reservoirs and the re-emergence of plasma viremia after discontinuation of highly active anti-retroviral therapy..
Nat Med, 6 (2000), pp. 757-61
[61]
McFarland EJ, Harding PA, MaWhinney S, Schooley RT, Kuritzkes DR..
In vitro effects of IL-12 on HIV-1-specific CTL lines from HIV-1-infected children..
J Immunol, 161 (1998), pp. 513-9
[62]
Forcina G, D'Ettorre G, Mastroianni CM, Carnevalini M, Scorzolini L, Ceccarelli G, et al..
Interleukin-15 modulates interferon-gamma and beta-chemokine production in patients with HIV infection: implications for immune-based therapy..
Cytokine, 25 (2004), pp. 283-90
[63]
Mastroianni CM, D'Ettorre G, Forcina G, Vullo V..
for immunotherapy? Trends Immunol, 25 (2004), pp. 121-5
[64]
Landay AL, Clerici M, Hashemi F, Kessler H, Berzofsky JA, Shearer GM..
In vitro restoration of T cell immune function in human immunodeficiency virus-positive persons: effects of interleukin (IL)-12 and anti-IL-10..
J Infect Dis, 173 (1996), pp. 1085-91
[65]
IL-15 treatment of SIV-infected non-human primates (Abstract 512). 11st Conference on Retroviruses and Opportunistic Infections; San Francisco, February, 2004. p. 8-11.
[66]
Napolitano LA, Lo JC, Gotway MB, Mulligan K, Barbour JD, Schmidt D, et al..
Increased thymic mass and circulating naive CD4 T cells in HIV-1-infected adults treated with growth hormone..
AIDS, 16 (2002), pp. 1103-11
[67]
Nguyen BY, Clerici M, Venzon DJ, Bauza S, Murphy WJ, Longo DL, et al..
Pilot study of the immunologic effects of recombinant human growth hormone and recombinant insulin-like growth factor in HIV-infected patients..
AIDS, 12 (1998), pp. 895-904
[68]
Fagard C, Le Braz M, Gunthard H, Hirsch HH, Egger M, Vernazza P, et al..
A controlled trial of granulocyte macrophage-colony stimulating factor during interruption of HAART..
[69]
Lisziewicz J, Rosenberg E, Lieberman J, Jessen H, Lopalco L, Siliciano R, et al..
Control of HIV despite the discontinuation of antiretroviral therapy [letter]..
N Engl J Med, 340 (1999), pp. 1683-4
[70]
Ortiz GM, Nixon DF, Trkola A, Binley J, Jin X, Bonhoeffer S, et al..
HIV-1 specific immune response in subjects who temporarely contain virus replication after discontinuation of HAART..
J Clin Invest, 104 (1999), pp. R13-R8
[71]
A Multi-center, randomized controlled clinical trial of continuous vs Intermittent HAART Guided by CD4+ T-cell counts and plasma HIV-1 RNA Levels. Abstract 65. 10th conference on retroviruses and opportunistic infections, Boston; MA. February 10-14, 2003.
[72]
Martínez-Picado J, Morales-Lopetegui K, Wrinb T, Prado J, Frost SD.W, Petropoulos CJ, et al..
Selection of drug-resistant HIV-1 mutants in response to repeated structured treatment interruptions..
AIDS, 16 (2002), pp. 895-9
[73]
Ruiz L, Ribera E, Bonjoch A, Romeu J, Martínez-Picado J, Paredes R, et al..
Role of structured treatment interruption before a 5-drug salvage antiretroviral regimen: the Retrogene Study..
J Infect Dis, 188 (2003), pp. 977-85
[74]
Rosenberg ES, Altfeld M, Poon S, Phillips M, Wilkes B, Eldridge R, et al..
Immune control of HIV-1 after early treatment of acute infection..
Nature, 407 (2000), pp. 523-6
[75]
Smith DE, Walker BD, Cooper DA, Rosenberg ES, Kaldor JM..
Is antiretroviral treatment of primary HIV infection clinically justified on the basis of current evidence? AIDS, 18 (2004), pp. 709-18
[76]
Fagard C, Oxenius A, Gunthard H, García F, Mestre G, Le Braz M, et al..
A prospective trial of structured treatment interruptions in HIV infection..
Arch Intern Med, 163 (2003), pp. 1220-6
[77]
Oxenius A, Price D, Gunthard HF, Dawson S, Fagard C, Perrin L, et al..
Stimulation of HIV specific cellular immunity by structured treatment interruption fails to enhance viral control in chronic HIV-infection..
Proc Natl Acad Sci U S A, 99 (2002), pp. 13752
[78]
García F, Plana M, Arnedo M, Ortiz GM, Miró JM, Lopalco L, et al..
A Cytostatic Drug Improves Control of HIV-1 Replication During Structured Treatment Interruptions. A Randomized Study..
[79]
García F, Plana M, Ortiz GM, Bonhoeffer S, Soriano A, Vidal C, et al..
The virological and immunological consequences of structured treatment interruptions in chronic HIV-1 infection..
AIDS, 15 (2001), pp. F29-F40
[80]
Ortiz GM, Wellons M, Brancato J, Vo H, Zinn R, Clarkson D, et al..
Structured antiretroviral treatment interruptions in chronically HIV-1-infected subjects..
Proc Natl Acad Sci U S A, 98 (2001), pp. 13288-93
[81]
Carcelain G, Tubiana R, Samri A, Calvez V, Delaugerre C, Agut H, et al..
Transient mobilization of human immunodeciency virus (HIV)-specific CD4 T-helper cells fails to control virus rebounds during intermittent antiretroviral therapy in chronic HIV type 1 infection..
[82]
Ruiz L, Carcelain G, Martínez-Picado J, Frost SD, Marcia S, Paredes R, et al..
HIV dynamics and T-cell immunity after three structured treatment interruptions in chronic HIV-1 infection..
AIDS, 15 (2001), pp. F19-F27
[83]
Brenchley JM, Hill BJ, Ambrozak DR, Price DA, Guenaga FJ, Casazza JP, et al..
T-cell subsets that harbor human immunodeficiency virus (HIV) in vivo: implications for HIV pathogenesis..
J Virol, 78 (2004), pp. 1160-8
[84]
Younes SA, Yassine-Diab B, Dumont AR, Boulassel MR, Grossman Z, Routy JP, et al..
HIV-1 viremia prevents the establishment of interleukin 2-producing HIV-specific memory CD4+ T cells endowed with proliferative capacity..
J Exp Med, 198 (2003), pp. 1909-22
[85]
Palmer BE, Boritz E, Wilson CC..
Effects of sustained HIV-1 plasma viremia on HIV-1 gag-specific CD4+ T cell maturation and function..
J Immunol, 172 (2004), pp. 3337-47
[86]
Relevance of HIV-1-specific CD4+ T helper cell responses during structured treatment interruption in patients with a nadir CD4 T cells above 400/mm3. J Acquir Immune Defic Syndr. En prensa 2004.
[87]
Salkowitz JR, Sieg SF, Harding CV, Lederman MM..
In vitro human memory CD8 T cell expansion in response to cytomegalovirus requires CD4+ T cell help..
J Infect Dis, 189 (2004), pp. 971-83
[88]
Day CL, Walker BD..
Progress in defining CD4 helper cell responses in chronic viral infections..
J Exp Med, 198 (2003), pp. 1773-7
[89]
Oxenius A, Price DA, Hersberger M, Schlaepfer E, Weber R, Weber M, et al..
HIV-Specific Cellular Immune Response Is Inversely Correlated with Disease Progression as Defined by Decline of CD4+ T Cells in Relation to HIV RNA Load..
J Infect Dis, 189 (2004), pp. 1199-208
[90]
D'Offizi G, Montesano C, Agrati C, Gioia C, Amicosante M, Topino S, et al..
Expansion of pre-terminally differentiated CD8 T cells in chronic HIV-positive patients presenting a rapid viral rebound during structured treatment interruption..
AIDS, 16 (2002), pp. 2431-38
[91]
Frost SD.W, Martinez-Picado J, Ruiz L, Clotet B, Brown AJ..
Viral dynamics during structured treatment interruptions of chronic human immunodeficiency virus type 1 infection..
J Virol, 76 (2002), pp. 968-79
[92]
Martinez-Picado J, Frost SD, Izquierdo N, Morales-Lopetegi K, Marfil S, Puig T, et al..
Viral evolution during structured treatment interruptions in chronically human immunodeficiency virus-infected individuals..
J Virol, 76 (2002), pp. 12344-8
[93]
Dybul M, Daucher M, Jensen MA, Hallahan CW, Chun TW, Belson M, et al..
Genetic characterization of rebounding human immunodeficiency virus type 1 in plasma during multiple interruptions of highly active antiretroviral therapy..
J Virol, 77 (2003), pp. 3229-37
[94]
García F, Plana M, Mestre G, Arnedo M, Gil C, Miró JM, et al..
Immunologic and virologic factors at baseline may predict response to structured therapy interruption in early stage chronic HIV-1 infection..
AIDS, 16 (2002), pp. 1761-5
[95]
Altfeld M, Lunzen J, Frahm N, Yu XG, Schneider C, Eldridge R, et al..
Expansion of pre-existing, lymph node-localized CD8+ T cells during supervised treatment interruptions in chronic HIV-1 infection..
J Clin Invest, 109 (2002), pp. 837-43
[96]
Giorgi JV, Hultin LE, McKeating JA, Johnson TD, Owens B, Jacobson LP, et al..
Shorter survival in advanced human immunodeficiency virus type 1 infection is more closely associated with T lymphocyte activation than with plasma virus burden or virus chemokine coreceptor usage..
J Infect Dis, 179 (1999), pp. 859-70
[97]
Chun TW, Justement JS, Sanford C, Hallahan CW, Planta MA, Loutfy M, et al..
Relationship between the frequency of HIV-specific CD8+ T cells and the level of CD38+ CD8+ T cells in untreated HIV-infected individuals..
Proc Natl Acad Sci U S A, 101 (2004), pp. 2464-9
[98]
Lori F, Lewis M, Xu J, Varga G, Zinn D, Crabbs C, et al..
Control of SIV rebound through structured treatment interruptions during early infection..
Science, 290 (2000), pp. 1591-3
[99]
Hellinger J, Iwane M, Smith J, Fleishman A, Torres R, Schrader S, et al..
A randomized study of the safety and antiretroviral activity of hydroxyurea combined with didanosine in persons infected with human immunodeficiency virus type 1. American Foundation for AIDS Research Community-Based Clinical Trials Network..
J Infect Dis, 181 (2000), pp. 540-7
[100]
Piccinini G, Foli A, Comolli G, Lisziewicz J, Lori F..
Complementary antiviral efficacy of hydroxyurea and protease inhibitors in human immunodeficiency virus-infected dendritic cells and lymphocytes..
J Virol, 76 (2002), pp. 2274-8
[101]
Chapuis A, Rizzardi P, D'Agostini C, Attinger A, Knabenhans C, Fleury S, et al..
Effects of mycophenolic acid on human immunodeficiency virus infection in vitro and in vivo..
Nat Med, 6 (2000), pp. 762-8
[102]
Effect of mycophenolate mophetil on immune response and on plasma and lymphatic tissue viral load during and after interruption of HAART in chronic HIV-1 infected patients. A pilot randomized study. J Acquir Immune Defic Syndr. En prensa 2004.
[103]
Margolis D, Kewn S, Coull JJ, Ylisastigui L, Turner D, Wise H, et al..
The addition of mycophenolate mofetil to antiretroviral tharapy including abacavir is associated with depletion of intracellular deoxyguanosine triphosphate and a decrease in plasma HIV-1 RNA..
J Acquir Immune Defic Syndr, 31 (2002), pp. 45-9
[104]
Rizzardi GP, Harari A, Capiluppi B, Tambussi G, Ellefsen K, Ciuffreda D, et al..
Treatment of primary HIV-1 infection with cyclosporin A coupled with highly active antiretroviral therapy..
J Clin Invest, 109 (2002), pp. 681-8
[105]
Markowitz M, Jin X, Hurley A, Simon V, Ramratnam B, Louie M, et al..
Discontinuation of antiretroviral therapy commenced early during the course of human immunodeficiency virus type 1 infection, with or without adjunctive vaccination..
J Infect Dis, 186 (2002), pp. 634-43
[106]
MacGregor RR, Ginsberg R, Ugen KE, Baine Y, Kang CU, Tu XM, et al..
T-cell responses induced in normal volunteers immunized with a DNA-based vaccine containing HIV-1 env and rev..
AIDS, 16 (2002), pp. 2137-43
[107]
MacGregor RR, Boyer JD, Ciccarelli RB, Ginsberg RS, Weiner DB..
Safety and immune responses to a DNA-based human immunodeficiency virus (HIV) type I env/rev vaccine in HIV-infected recipients: follow-up data..
J Infect Dis, 181 (2000), pp. 406
[108]
Schooley RT, Spino C, Kuritzkes D, Walker BD, Valentine FA, Hirsch MS, et al..
Two double-blinded, randomized, comparative trials of 4 human immunodeficiency virus type 1 (HIV-1) envelope vaccines in HIV-1-infected individuals across a spectrum of disease severity: AIDS Clinical Trials Groups 209 and 214..
J Infect Dis, 182 (2000), pp. 1357-64
[109]
Smith D, Gow I, Colebunders R, Weller I, Tchamouroff S, Weber J, et al..
Therapeutic vaccination (p24-VLP) of patients with advanced HIV-1 infection in the pre-HAART era does not alter CD4 cell decline..
HIV Med, 2 (2001), pp. 272-5
[110]
Robbins GK, Addo MM, Troung H, Rathod A, Habeeb K, Davis B, et al..
Augmentation of HIV-1-specific T helper cell responses in chronic HIV-1 infection by therapeutic immunization..
[111]
Lederman MM, Douek DC..
Sometimes help may not be enough..
[112]
Kahn JO, Cherng DW, Mayer K, Murray H, Lagakos S..
Evaluation of HIV-1 immunogen, an immunologic modifier, administered to patients infected with HIV having 300 to 549 x 10(6)/L CD4 cell counts: A randomized controlled trial..
JAMA, 284 (2000), pp. 2193-202
[113]
Lu W, Wu X, Lu Y, Guo W, Andrieu JM..
Therapeutic dendritic-cell vaccine for simian AIDS..
Nat Med, 9 (2003), pp. 27-32
[114]
Lapenta C, Santini S, Logozzi M, Spada M, Andreotti M, Pucchio T, et al..
Potent Immune responses against HIV-1 and protection from virus challenge in hu-PBL-SCID mice immunized with inactivated virus-pulsed dendritic cells generated in the presence of IFN-alpha..
J Exp Med, 198 (2003), pp. 361-7
[115]
Yoshida A, Tanaka R, Murakami T, Takahashi Y, Koyanagi Y, Nakamura M, et al..
Induction of protective immune responses against R5 Human Immunodeficiency virus type 1 (HIV-1) infection in hu-PBL-SCID mice by intraesplenic immunization with HIV-1 pulsed dendritic cells: possible involvement of a novel factor of human CD4+ T-cell origin..
J Virol, 77 (2003), pp. 8719-28
[116]
Final results of a phase I study of a therapeutic vaccine using monocyte-derived autologous dendritic cells primed with autologous virus in patients with chronic HIV infection and nadir of CD4 T cells above 400 mm3. 11st Conference on Retroviruses and Opportunistic Infections. San Francisco; February, 2004. p. 8-11.
[117]
Shedlock DJ, Shen H..
Requirement for CD4 T cell help in generating functional CD8 T cell memory..
Science, 300 (2003), pp. 337-9
[118]
Sun JC, Bevan MJ..
Defective CD8 T cell memory following acute infection without CD4 T cell help..
Science, 300 (2003), pp. 339-42
[119]
Steinman RM, Pope M..
Exploiting dendritic cells to improve vaccine efficacy..
J Clin Invest, 109 (2002), pp. 1519-26
[120]
Evaluation of 2 therapeutic HIV vaccination regimens in HAART-treated primary HIV infection subjects following analytical treatment interruption: QUEST PROB3005, a randomized, placebo-controlled study. Abstract 168. 11st Conference on Retroviruses and Opportunistic Infections; San Francisco, February, 8-11 2004.
[121]
Randomized, placebo-controlled, phase1/2a evaluation of the safety, biological activity and antiretroviral properties of an avipox virus vaccine expressing HIV gag-pol and interferon-gamma in HIV-1 infected subjects. Abstract 169. 11st Conference on Retroviruses and Opportunistic Infections; San Francisco, February, 8-11 2004.
[122]
McConkey SJ, Reece WH, Moorthy VS, Webster D, Dunachie S, Butcher G, et al..
Enhanced T-cell immunogenicity of plasmid DNA vaccines boosted by recombinant modified vaccinia virus Ankara in humans..
Nat Med, 9 (2003), pp. 729-35
[123]
Lisziewicz J, Bakare N, Lori F..
Therapeutic vaccination for future management of HIV/AIDS..
Vaccine, 21 (2003), pp. 620-3
[124]
Mitsuyasu RT, Anton P, Deeks SG, Scadden DT, Connick ED, Owns M, et al..
Prolonged survival and tissue trafficking following adoptive transfer of CD4 zeta gene modified autologous CD4 and CD8 T cells in human immuno-deficiency virus-infected subjects..
Blood, 96 (2000), pp. 785-93
[125]
Jacobson E, Smith KA, Smith KA..
Rational interleukin 2 therapy for HIV-positive individuals: daily low doses enhance immune function without toxicity..
Proc Natl Acad Sci U S A, 93 (1996), pp. 10405-10
[126]
Aladdin H, Ullum H, Dam Nielsen S, Espersen C, Mathiesen L, Katzenstein TL, et al..
Granulocyte colony-stimulating factor increases CD4 T cell counts of human immu-nodeficiency virus-infected patients receiving stable, highly active antiretroviral therapy: results from a randomized, placebo-controlled trial..
J Infect Dis, 181 (2000), pp. 1148-52
[127]
Brites C, Gilbert MJ, Pedral-Sampaio D, Bahia F, Pedroso C, Alcantara A, et al..
A randomized, placebo-controlled trial of granulocyte-macrophage colony-stimulating factor and nucleoside analogue therapy in AIDS..
J Infect Dis, 182 (2000), pp. 1531-5
[128]
Alston B, Ellenberg JH, Standiford H, Muth K, Martínez A, Greaves W, et al..
A multicenter, randomized, controlled trial of three preparations of low-dose oral alpha-interferon in HIV-infected patients with CD4 counts between 50 and 350 cells/mm3. Division of AIDS Treatment Research Initiative (DATRI)..
J Acquir Immune Defic Syndr, 22 (1999), pp. 348-57
[129]
Angel J, Jacobson MA, Skolnik PR, Giordano M, Shapiro L, LeBeaut A, et al..
A multicenter, randomized, double-blind, placebo-controlled trial of recombinant human IL-10 in HIV-infected subjects..
AIDS, 14 (2000), pp. 2503-8
[130]
Jacobson MA, Hardy D, Connick E, Watson JS, Debruin M..
Phase 1trial of a single dose of recombinant human interleukin-12 in human immunodeficiency virus-infected patients with 100-500 CD4 cells/mL..
J Infect Dis, 182 (2000), pp. 1070-6
[131]
Kanai T, Thomas E, Yasutomi Y, Letvin NL..
IL-15 stimulates the expansion of AIDS virus-specific CTL..
J Immunol, 157 (1996), pp. 3681-7
[132]
Truong M, Darcissac E, Hermann E, Dewulf J, Capron A, Bahr J..
Interleukin-16 inhibits human immunodeficiency virus type 1 entry and replication in macrophages and in dendritic cells..
J Virol, 73 (1999), pp. 7008-13
[133]
Napolitano L, Grant RM, Deeks S, Schmidt D, De Rosa S, Herzenberg LA, et al..
Increased production of IL-7 accompanies HIV-1-mediated T-cell depletion: implications for T-cell homeostasis..
Nat Med, 7 (2001), pp. 73-9
[134]
Andrieu JM, Lu W, Levy R..
Sustained increases in CD4 cell counts in asymptomatic human immunodeficiency virus type 1-seropositive patients treated with prednisolone for 1 year..
J Infect Dis, 171 (1995), pp. 523-30
[135]
Rizzardi GP, Vaccarezza M, Capiluppi B, Tambussi G, Lazzarin A, Pantaleo G..
Cyclosporin A in combination with HAART in primary HIV-1 infection..
J Biol Regul Homeost Agents, 14 (2000), pp. 79-81
[136]
Jacobson JM, Greenspan JS, Spritzler J, Ketter N, Fahey JL, Jackson JB, et al..
Thalidomide for the treatment of oral aphthous ulcers in patients with human immu-nodeficiency virus infection. National Institute of Allergy and Infectious Diseases AIDS Clinical Trials Group..
N Engl J Med, 336 (1997), pp. 1487-93
[137]
Patterson BK, Carlo D, Kaplan MH, Marecki M, Pawha S, Moss R..
Cell-associated HIV-1 messenger RNA and DNA in T-helper cell and monocytes in asymp-tomatic HIV-1-infected subjects on HAART plus an inactivated HIV-1 immunogen..
AIDS, 13 (1999), pp. 1607-11
[138]
Amara R, Villinger F, Altman J, Lydy S, O'Neil S, Staprans SI, et al..
Control of a Mucosal Challenge and Prevention of AIDS by a Multiprotein DNA/MVA Vaccine..
Science, 292 (001), pp. 69-74
[139]
Buge S, Murty L, Arora K, Kalyanaraman VS, Markham PD, Richardson E, et al..
Factors associated with slow disease progression in macaques immunized with an adenovirus simian im-munodeficiency virus (SIV) envelope priming-gp120 boosting regimen and challenged vaginally with SIVmac 251..
J Virol, 73 (1999), pp. 7430-40
Opciones de artículo
Herramientas
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos

Quizás le interese:
10.1016/j.eimc.2024.03.006
No mostrar más